inflammatory bowel disease monitor
Transcription
inflammatory bowel disease monitor
VOLUME 8 NUMBER 1 2007 INFLAMMATORY BOWEL DISEASE MONITOR Commentary and analysis on advances in the understanding and management of inflammatory bowel disease EDITORS-IN-CHIEF Stephen Hanauer, Chicago, IL, USA Jack Satsangi, Edinburgh, UK LEADING ARTICLES Classification of Inflammatory Bowel Disease: A Work in Progress Rupert Abdalian, MD and Mark S Silverberg, MD, PhD Antineoplastic Effects of 5-Aminosalicylic Acid in the Intestine: From Bench to Bedside Christel Rousseaux, PhD, Olivier Romano, MD, and Pierre Desreumaux, MD, PhD Orofacial Granulomatosis and Oral Crohn’s Disease Carlo Nunes, MD, Miranda Lomer, PhD, Michael Escudier, FDS, RCS, Stephen Challacombe, FMedSci, and Jeremy Sanderson, MD CLINICAL REVIEWS MEETING REPORTS ESPGHAN 2007 DDW 2007 www.ibdmonitor.com Jointly sponsored by the University of Kentucky Colleges of Pharmacy and Medicine and Remedica Medical Education and Publishing. The University of Kentucky is an equal opportunity university. This journal is supported by an educational grant from Shire Faculty Disclosures The following are relevant financial relationships declared by the journal’s Editors-in-Chief. Stephen Hanauer: Abbott, Amgen, Asahi, Bristol Myers Squibb, Centocor, Chemocentryx, Elan, Ferring, Genentech, GSK, Novartis, Otsuka, Protein Design Labs, Procter & Gamble, Prometheus, Salix, Shire, Targacept, Teva, Therakos, Millennium Pharmaceuticals, and UCB Pharma (Celltech). Jack Satsangi: Abbott, Elan, Schering-Plough, and UCB. Subscription Information Inflammatory Bowel Disease Monitor (ISSN 1466-7401) is published four times per year. Subscriptions are available at the following rates: Europe u150, USA, Canada and all other territories US$200 per year. Additional subscription information is available from the publisher. The following are relevant financial relationships declared by the journal’s Editors. Ian Arnott: None to declare. Federico Balzola: None to declare. Charles Bernstein: Abbot Canada, Axcan Pharma, Bristol Myers Squibb, Pfizer, Procter & Gamble, Shire Canada, and UCB Canada. Simon Murch: Not yet received. All rights reserved. No part of this publication may be reproduced, stored in a retrieval system or transmitted in any form or by any means, electronic, mechanical, photocopying, recording or otherwise without the prior permission of the copyright owners. While every effort is made by the publishers and editorial board to see that no inaccurate or misleading data, opinions, or statements appear in this journal, they wish to make it clear that the material contained in the publication represents a summary of the independent evaluations and opinions of the authors and contributors. As a consequence, the board, publishers and any sponsoring company accept no responsibility for the consequences of any such inaccurate or misleading data or statements. Neither do they endorse the content of the publication or the use of any drug or device in a way that lies outside its current licensed application in any territory. For detailed information on any drugs or devices discussed in this publication, readers are advised to consult the Physicians Circular issued by the manufacturer. The following are relevant financial relationships declared by the journal’s Editorial Advisory Board: Zane Cohen: Not yet received. Jean-Frédéric Colombel: Abbott, Centocor, ScheringPlough, and UCB Pharma. Anders Ekbom: AstraZeneca, Centocor, and Schering-Plough. Brian Feagan: Abbott, AstraZeneca, Berlex, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Centocor, CombinatoRx, Elan/Biogen, ISIS, Janssen-Ortho, Millennium, Napo Pharma, Novartis, Osiris, Otsuka, Procter & Gamble, Protein Design Labs, Santarus, Schering Canada, Schering Plough, Serono, Synta, Teva Pharmaceuticals, Tillotts, and UCB Pharma. Claudio Fiocchi: None to declare. John O’leary: Not yet received. Paul C Rutgeerts: Not yet received. Ernest G Seidman: Not yet received. Stephan Targan: Berlex, Bristol Myers Squibb, Elan, Gilead Science, Procter & Gamble, Prometheus, Salix, Santarus, and UCB Pharma. William J Tremaine: AstraZeneca, Procter & Gamble, and NPS Pharmaceuticals. Editorial Policy Inflammatory Bowel Disease Monitor is an independent journal published by Remedica Medical Education and Publishing Ltd. The aim is to provide an up-to-date overview of the recent literature compiled by an international team of practising physicians. Leading articles commissioned by the Editors-in-Chief review new therapeutic techniques and emerging technologies, and news from major international meetings is reported. Editorial control is vested entirely in the Editors-in-Chief, Editors, and Editorial Advisory Board. Publisher’s Statement © Remedica Medical Education and Publishing 2007. Editorial Team: Rhian Phillips, Amy Loader Editorial Manager: Scott Millar Design and Artwork: AS&K Skylight Creative Services Publishers: Ian Ackland-Snow, Simon Kirsch Any queries regarding the content of the journal should be addressed to: Remedica Medical Education and Publishing Ltd, Commonwealth House, 1 New Oxford Street, London, WC1A 1NU, UK. Tel:+44 (0) 20 7759 2999, Fax: +44 (0)20 7759 2951 Remedica Medical Education and Publishing Inc, 20 N. Wacker Drive, Suite 1642, Chicago, IL 60606, USA. Tel: +1 (312) 372 4020, Fax: +1 (312) 372 0217. Email: [email protected] ISSN 1466-7401 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 1 Editors-in-Chief Stephen Hanauer, MD University of Chicago, Chicago, IL, USA Jack Satsangi, MBBS, DPhil Western General Hospital, Edinburgh, UK Editors Ian Arnott, MD Western General Hospital, Edinburgh, UK Federico Balzola, MD Azienda Ospedaliera San Giovanni Battista di Torino, Turin, Italy Charles Bernstein, MD University of Manitoba, Winnipeg, MB, Canada Simon Murch, MD University of Warwick, Coventry, UK Editorial Advisory Board Zane Cohen, PhD Mount Sinai Hospital, Toronto, ON, Canada Jean-Frédéric Colombel, MD Hôpital Huriez, Lille, France Anders Ekbom, PhD Karolinska Institute, Stockholm, Sweden Brian Feagan, MD University of Western Ontario, London, ON, Canada Claudio Fiocchi, MD Case Western Reserve University, Cleveland, OH, USA John O’Leary, MD Coombe Women’s Hospital, Dublin, Ireland Paul C Rutgeerts, MD Universitaire Ziekenhuizen Leuven, UZ Gasthuisberg, Leuven, Belgium Ernest G Seidman, MD Hôpital Sainte-Justine, Université de Montréal, Montreal, QC, Canada Stephan Targan, MD Cedars-Sinai Medical Center, Los Angeles, CA, USA William J Tremaine, MD Mayo Clinic, Rochester, MN, USA Contents Leading Articles Classification of Inflammatory Bowel Disease: A Work in Progress Rupert Abdalian and Mark S Silverberg Antineoplastic Effects of 5-Aminosalicylic Acid in the Intestine: From Bench to Bedside Christel Rousseaux, Olivier Romano, and Pierre Desreumaux Orofacial Granulomatosis and Oral Crohn’s Disease Carlo Nunes, Miranda Lomer, Michael Escudier, Stephen Challacombe, and Jeremy Sanderson 2 8 18 Clinical Reviews Clinical Observations 23 Genetics 29 Epidemiology 31 Pathogenesis 32 Meeting Reports 40th Annual Meeting of the European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) Barcelona, Spain, 9–12 May, 2007 35 Digestive Diseases Week 2007 (DDW 2007) Washington, DC, USA, 20–23 May, 2007 38 LEADING ARTICLE RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 2 Classification of Inflammatory Bowel Disease: A Work in Progress Rupert Abdalian, MD1 and Mark S Silverberg, MD, PhD2 1 Department of Medicine, Division of Gastroenterology, University of Toronto, and 2Mount Sinai Hospital Inflammatory Bowel Disease Centre, Division of Gastroenterology, Toronto, ON, Canada Crohn’s disease and ulcerative colitis (UC) represent idiopathic forms of IBD with significant clinical heterogeneity. A major need in their investigation is the ability to identify patient subgroups with distinctive and, perhaps, unique characteristics. Definition of patient subtypes with specific prognoses and disease behavior could add power to research in therapeutics and genetics. It may also offer the clinician an invaluable tool to better assess disease prognosis, optimize patient counseling, and select the most optimal therapeutic strategy. The recent and ongoing discovery of serological biomarkers and genetic associations may soon revolutionize our approach and understanding of IBD. The current article is a comprehensive review of recently proposed classification systems for Crohn’s disease and UC, and aims to highlight their relevance to clinical practice and research endeavors. Inflamm Bowel Dis Monit 2007;8(1):2–7. Over the past few decades, investigators and clinicians have attempted to classify IBD using recognizable clinical, epidemiological, and endoscopic features. The recent discovery of novel genetic determinants and serological markers associated with IBD susceptibility and phenotype has made the prospect of developing a more integrated and useful classification system achievable. Evaluation of these markers is the subject of intense investigation, although their relative importance to the diagnosis, natural history, and phenotypic differentiation of IBD remains largely speculative. Several classification schemes have been proposed since 1991. Most recently, a Working Party with an interest in IBD classification was formed with the aim of summarizing recent developments in disease classification and establishing an integrated classification of IBD [1]. Their recommendations were presented at the 2005 World Congress of Gastroenterology in Montreal, QC, Canada. The following is a comprehensive review of existing and recently proposed classification systems for IBD, and highlights their relevance to clinical practice and research endeavors. Clinical classification of Crohn’s disease Greenstein et al. first distinguished patients with Crohn’s disease who require surgery into those with an aggressive, perforating phenotype from those with a more indolent, nonperforating disease [2]. Patients with perforation were found Address for correspondence: Mark S Silverberg, Mount Sinai Hospital Inflammatory Bowel Disease Centre, Division of Gastroenterology, Toronto, ON, Canada. Email: [email protected] 2 to have earlier disease recurrence following surgery, regardless of disease location. Indications for subsequent surgery were found to be generally the same as those for the first or previous operation. This categorization of Crohn’s disease into perforating and non-perforating subtypes was later integrated in the International Working Party’s report issued in Rome, Italy, in 1991, which proposed a classification based on anatomical distribution, operative history, and clinical behavior (inflammatory, fistulizing, or stenotic disease). However, this classification was felt to be impractical in the clinical setting, and was later revised at the World Congress of Gastroenterology in Vienna, Austria, in 1998 [3]. The ensuing classification scheme integrated age of onset (below or above 40 years), disease location (ileal, colonic, ileocolonic, or upper GI), and disease behavior (non-stricturing, non-penetrating, stricturing, or penetrating) as the key phenotypic determinants. Although the Vienna classification has not been widely applied to the clinical setting, its utility in this environment has been scrutinized for its potential applicability [4]. Difficulties with the Vienna classification included a failure to evaluate particular aspects of disease course and prognosis. For instance, severity of disease and rate of progression of disease behavior were difficult to reliably and objectively define, although they are important clinical parameters that have a probable impact on natural history, outcome of therapeutic trials, and the need for surgery. Additional features such as presence of extraintestinal manifestations, ethnicity, country of birth, smoking history, and family history are omitted as well. All of these are related to factors that may affect Crohn’s disease phenotype, course, and behavior. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 3 CLASSIFICATION OF IBD Recently, the Vienna classification has been employed as a categorization tool allowing the study of correlates between specific Crohn’s disease phenotypes and other aspects of disease presentation, course, and genetic susceptibility. An association between certain subtypes and perinuclear antineutrophil cytoplasmic antibody (pANCA) or anti-Saccharomyces cerevisiae antibody (ASCA) profiles has been described [5]. Additionally, the carriage of at least one nucleotide-binding oligomerization domain 2/caspase recruitment domain-containing protein 15 (NOD2/CARD15) gene mutation was significantly associated with ileal involvement, stricturing evolution, and presence of granulomas [6,7]. The Vienna classification also helped predict the need for immunosuppressant therapy and surgery in a retrospective review of 480 patients [8]. The Montreal revision of the original Vienna classification has not altered the three predominant parameters of age at diagnosis, location, and behavior of disease, but modifications within each of these categories have been made in light of evidence highlighting intriguing new correlates (Table 1). With regard to age at Crohn’s disease diagnosis, the Montreal classification allows for an earlier onset of disease (i.e. aged 16 years or younger) to be categorized separately as a novel A1 category, whereas A2 and A3 account for age of diagnosis between 17 and 40 years and >40 years, respectively. There is indeed evidence that disease phenotype varies according to age at Crohn’s disease diagnosis [9,10]. For instance, disease distribution seems to vary based on age at diagnosis, with small intestinal and upper GI involvement (L4) observed more commonly in individuals diagnosed before the age of 20 years and colonic disease seen more frequently in patients aged >60 years [11]. The male-to-female ratio decreases with increasing age of onset of Crohn’s disease, and a family history of IBD is more likely to be noted in patients with an earlier age at diagnosis [12]. Given the differing clinical characteristics of early-onset disease, the introduction of a separate age category was timely. This modification will allow the categorization of pediatric-onset disease as a clinically distinct entity both in practice and in the research setting. The major anatomical sites of Crohn’s disease distribution that are agreed upon by most experts are the ileum (L1), the colon (L2), and the ileocolonic region (L3). These were recognized and featured within the Vienna classification; however, upper GI involvement (L4) in this scheme implied an absence of disease activity distal to the jejunum. As modalities to investigate the upper GI tract have become more refined and accessible, especially with the widespread introduction of wireless capsule endoscopy, it is now apparent that foregut involvement in Crohn’s disease is not uncommon, and in fact may coexist with ileal (L1) and/or Table 1. Montreal classification of Crohn’s disease. Age at Diagnosis A1 ≤16 years A2 17–40 years A3 >40 years Location Upper GI Modifier (L4) L1 Terminal ileum L1 + L4 Terminal ileum + upper GI L2 Colon L2 + L4 Colon + upper GI L3 Ileocolon L3 + L4 Ileocolon + upper GI L4 Upper GI – – Behavior Perianal disease modifier (p) B1* Non-stricturing B1p non-penetrating Non-stricuring non-penetrating + perianal B2 Stricturing B2p Stricturing + perianal B3 Penetrating B3p Penetrating + perianal *B1 category should be considered “definitive” only after a pre-specified time period has elapsed (≥5 years of follow-up recommended). Otherwise, the identified disease behavior should be considered only “interim”. GI: gastrointestinal. Table redrawn with permission from [1]. colonic disease (L2 and/or L3). Therefore, in the revised Montreal classification, these parameters of disease distribution are no longer mutually exclusive. As for disease behavior, the Montreal revision of the Vienna classification denotes perianal disease alone as a separate subclassification (p), and no longer includes it in the penetrating disease category. It also recognizes that disease behavior is dynamic over time and that classifying it should incorporate some aspect of time course or at least a minimum elapsed period of follow-up before assigning a definite disease behavior designation. The arbitrary period of 5 years was selected, based on evidence from several studies that documented progression of disease during follow-up periods ranging from 5 to 10 years [13,14]. It is noteworthy to mention that this 5-year observation period is only the minimum, and that the preferred assessment point should be the latest observation preceding any surgery. In contrast, disease localization in Crohn’s seems surprisingly stable. This observation may stem from the role of defensins in the pathogenesis of chronic IBD. Defensins are endogenous antimicrobial peptides that mediate innate immunity. Expressed within the intestinal epithelium, they contribute to the maintenance of mucosal barrier integrity INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 3 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 4 RUPERT ABDALIAN AND MARK S SILVERBERG [15]. In fact, Crohn’s disease of the terminal ileum, especially if associated with a NOD2 mutation, is characterized by a diminished α-defensin (human defensin 5 and 6) expression, and in Crohn’s colitis, the β-defensin (human β-defensins 2 and 3) response is reduced [16]. NOD2 and defensin deficiency at the level of the epithelial barrier and gutassociated lymphoid tissue may favor Crohn’s disease by disrupting the barrier of protection from enteropathogens and interrupting adaptive immune responses in the gut micro-environment. Table 2. Montreal classification of UC. Clinical classification of ulcerative colitis S0 Clinical remission Asymptomatic Unlike Crohn’s disease, neither the Rome nor the Vienna working parties formally addressed a classification system for ulcerative colitis (UC). A systematic classification scheme for UC would be invaluable as it could help unravel intricacies of pathogenesis if certain subtypes are found to correlate with specific genetic or serological biomarkers. It may facilitate the implementation of optimal treatment strategies and help the challenging task of prognostication (e.g. predicting risk of surgery, colorectal cancer risk, or response to therapy). Expert opinion suggests that an adequate classification system for UC should take into account extent of colonic activity and overall disease severity. The Montreal classification of UC defines three subgroups of disease extent (E1: ulcerative proctitis, E2: left-sided UC, E3: extensive UC; Table 2). This three-tiered classification system is biologically plausible as it serves several clinical purposes. Firstly, it dictates choice of medical therapy, and helps differentiate those who are more likely to respond to topical therapy. Hydrocortisone and mesalamine suppositories are mostly useful as primary therapy in patients with ulcerative proctitis (E1), although alone they are probably insufficient for the management of extensive UC (E3) [17]. Secondly, classification by extent of disease helps predict subsequent course and severity of disease. Higher rates of medication usage and hospitalizations, as well as an increased and earlier need for colectomy, have been correlated with increased disease burden [18]. In addition, the differential risk of colorectal malignancy further strengthens this subclassification [19]. The predominant flaw in an extent-based classification system for UC is that it assumes that disease characteristics remain constant over time. However, the dynamic nature of IBD as demonstrated by disease progression or regression imposes a challenging dilemma. The actual risk of proximal extension of proctitis over a cumulative 10-year period is estimated to range between 41% and 54%, whereas the rate of progression of left-sided colitis may be even higher [1]. Regression of disease has also been well-described, from a crude rate of 1.6% to an age- and sex-adjusted rate of S1 Mild UC Passage of four or fewer stools per day No systemic illness Normal inflammatory markers (ESR) S2 Moderate UC Passage of more than four stools per day Signs of systemic toxicity S3 Severe UC Passage of at least six bloody stools per day Heart rate ≥90 beats/min Temperature >37.5°C Hemoglobin level <105 g/L ESR >30 mm/h 4 Disease extent E1 Ulcerative proctitis Involvement limited to rectum E2 Left-sided UC (distal UC) Inflammation limited to colorectum distal to the splenic flexure E3 Extensive UC Involvement proximal to splenic flexure Disease severity ESR: erythrocyte sedimentation rate; UC: ulcerative colitis. 71% after 10 years [20]. The Montreal classification thus proposes the maximal extent of involvement as the most useful and prognostic parameter. However, it fails to recognize a minimum disease extent within a given time period. Since extensive UC may present with a “prodrome” of up to 4–6 months of proctitis, it is probably premature to classify a UC case as proctitis prior to 4–6 months of observation. The Montreal classification also addressed UC disease severity with a categorization into four groups (S0: clinical remission, S1: mild UC, S2: moderate UC, S3: severe UC; Table 2). One of the limitations of such a subclassification attempt is that it predicts clinical course only in the short term and does not necessarily reflect a longitudinal outlook. The classification also proposes to omit the term “fulminant colitis”, a designation with debatable clinical utility or prognostic significance. As for the need for a separate classification system for patients with UC and primary sclerosing cholangitis (PSC) overlap, demonstrable differences in clinicopathological features (rectal sparing and backwash ileitis), and prognosis (increased incidence of pouchitis and colorectal neoplasia), suggest that PSC-IBD INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 5 CLASSIFICATION OF IBD may indeed be an IBD phenotype distinct from both UC and Crohn’s disease, warranting its own recognition as a separate subclass [21]. Indeterminate colitis: a distinct entity? In 1978, Price first introduced the term “indeterminate colitis” (IC) to describe patients following colectomy in whom specific clinicopathological features were not sufficient to diagnose either Crohn’s disease or UC but were sufficient to allow a diagnosis of IBD affecting the colon [22]. Today, there is marked confusion about the appropriate use of this term. This diagnosis is now routinely made in patients with obvious clinical and endoscopic features of colonic inflammation, but in whom the histological and all other clinical parameters do not permit a clear diagnosis of either Crohn’s disease or UC. The Montreal Working Party recommended that the term IC be reserved only for cases where colectomy has been performed and pathologists are unable to certifiably confirm one diagnosis after full examination of the gross specimen. An alternative designation of “inflammatory bowel disease, type unclassified” (IBDU) was suggested for patients with colonic disease and sparing of the terminal ileum, without definitive histological corollary to favor either Crohn’s disease or UC. Specific serological and genetic markers may help refine the classification of IC and IBDU further. Both established serological markers, ASCA and pANCA, in conjunction with novel markers currently under study (anti-OmpC, antiCBir1/flagellin, and anti-I2) have the potential to further the understanding of the classification of these entities. There is a need for more studies investigating which combination of serological and genetic markers provides the best specificity and positive predictive value for different forms of IBD, including IBDU. Recognition of IBDU as a distinct disease subgroup may be important, as its clinical course and prognosis seem worse than that of UC [23]. Post-surgical outcomes and pouchitis risk seem unfavorable as well [24,25]. An integrated molecular diagnosis of IBD: a way of the future? Various serological and genetic marker tests integrating clinical data are currently under investigation and may, in the future, have an impact on classification schemes in IBD. Thus far, the two most widely recognized serological markers have been pANCA and ASCA. The utility of routine testing of these in the diagnosis of IBD or in patients with illdefined GI symptoms is limited because of their lack of specificity and low sensitivity. For instance, pANCA positivity has been observed in other inflammatory diseases of the colon, such as eosinophilic and collagenous colitis [26]. Several independent studies found that a combination of both had positive predictive values ranging from 77–96% for differentiating Crohn’s disease from UC [27]. These markers may also have a role in disease stratification. In Crohn’s disease, pANCA has been linked to a UC-like phenotype, late-onset disease, and inflammatory diseasetype according to the Vienna classification [28]. ASCApositive status on the other hand is associated with more proximal (gastroduodenal and small bowel) involvement rather than purely colonic disease and with a more severe disease phenotype and requirement for surgery [29]. As for a role in disease monitoring and predicting treatment responsiveness, thus far the published data are more disparate. The presence of ASCA in Crohn’s disease is generally stable over time and is independent of Crohn’s disease activity or duration [30]. Studies assessing the power of these markers to predict the response to infliximab in Crohn’s disease or the risk of pouchitis in UC have yielded contradictory results and have been plagued with methodological flaws [31–33]. In light of the uncertainties and lack of diagnostic acumen of both pANCA and ASCA, there is a need to widen the autoantibody panel with serological markers of higher sensitivity and specificity. Studies of immune responsiveness to several specific microbial antigens in patients with Crohn’s disease and UC have yielded potential candidates. Most of the progress thus far has been recorded in the stratification of Crohn’s disease, with the identification of novel antibodies (anti-OmpC and anti-I2, and most recently anti-CBir1/flagellin) [34]. These have been associated with more complicated and severe Crohn’s disease behavior, including the need for surgery [35]. However, relying exclusively on serum indicators for diagnosis and classification of IBD is not yet justified. The available serological markers are not sensitive enough to be used for IBD screening in the general population. The addition of novel markers may improve overall sensitivity; however, more studies are needed to further delineate their role in clinical decision-making. Similar to serological indicators, integration of genetic markers in the proposed classification system cannot yet be justified. The most definite and well-replicated gene found to be associated with Crohn’s disease is NOD2/CARD15 [36]. The odds ratio for developing Crohn’s disease in simple heterozygotes in a meta-analysis was 2.4, and odds ratio for persons with two mutant chromosomes was estimated to be 17.1 [37]. The differences in risks associated with individual alleles were relatively minor. Despite widespread replication of the NOD2/CARD15 associations, a marked ethnic and geographical heterogeneity was noted in epidemiological surveys [38–40]. These mutations are scarce in Asians (Japanese, Chinese, Koreans), Arabs, Africans, and AfricanAmericans. However, even in Europe, the degree of disease INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 5 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 6 RUPERT ABDALIAN AND MARK S SILVERBERG susceptibility conveyed by different NOD2/CARD15 allelic variants differs in specific regions. The importance of these variants in Northern Europe (Scandinavia, Ireland, and Scotland) is less than in the index studies from central Europe, perhaps reflecting significant genetic founder effects [38]. This complexity between populations is further challenged by heterogeneity within populations and genotype–phenotype relationships. From a phenotypic perspective, NOD2/CARD15 mutations are associated with ileal rather than colonic Crohn’s disease with some studies supporting a correlation with more complicated internal fistulizing or stenosing disease [37]. NOD2/CARD15 variations have also been correlated with more severe disease, lower weight and earlier age at diagnosis, acute intestinal obstruction, and increased risk for surgery [41,42]. Conversely, they are uncommon in patients with perianal complications of Crohn’s disease [42]. No consistent association between NOD2/CARD15 variations and the presence of granulomas or extraintestinal manifestations of IBD has been found. Also, genetic variants have not been shown to predict treatment responsiveness, although only infliximab has been evaluated thus far [43]. The identification of the NOD2/CARD15 gene has been a significant discovery, shedding light on the complex pathogenesis of IBD. Moreover, the contribution of numerous additional genetic markers identified in genomewide association studies (autophagy-related 16-like 1 [ATG16L1], IL-23 receptor [IL23R], organic cation transporter [OCTN], disc, large homologue gene 5 [DLG5], multi-drug resistance 1 [MDR1], Toll-like receptors [TLRs], and pregnane X receptor [PXR] genes) [44–48], as well as other replicated candidate regions such as those within the major histocompatibility complex or the IBD5 locus, will eventually be integrated into a complex model of markers that aid in classifying IBD. However, to successfully achieve this, very large studies with well-phenotyped patients will be required and, ultimately, prospective validation studies conducted over many years will be needed. Disclosures Dr Silverberg has received funding for research from Prometheus Laboratories. Dr Abdalian has no relevant financial interests to disclose. References 1. Silverberg MS, Satsangi J, Ahmad T et al. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: Report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Can J Gastroenterol 2005;19(Suppl A):5–36. 2. Greenstein AJ, Lachman P, Sachar DB et al. Perforating and non-perforating indications for repeated operations in Crohn’s disease: evidence for two clinical forms. Gut 1988;29:588–92. 3. Gasche C, Scholmerich J, Brynskov J et al. A simple classification of Crohn’s disease: report of the Working Party for the World Congresses of Gastroenterology, Vienna 1998. Inflamm Bowel Dis 2000;6:8–15. 4. Freeman HJ. Application of the Vienna Classification for Crohn’s disease to a single clinician database of 877 patients. Can J Gastroenterol 2001;15:89–93. 5. Klebl FH, Bataille F, Bertea CR et al. Association of perinuclear antineutrophil cytoplasmic antibodies and anti-Saccharomyces cerevisiae antibodies with Vienna classification subtypes of Crohn’s disease. Inflamm Bowel Dis 2003;9:302–7. 6. Newman B, Silverberg MS, Gu X et al. CARD15 and HLA DRB1 alleles influence susceptibility and disease localization in Crohn’s disease. Am J Gastroenterol 2004;99:306–15. 7. Heresbach D, Gicquel-Douabin V, Birebent B et al. NOD2/CARD15 gene polymorphisms in Crohn’s disease: a genotype-phenotype analysis. Eur J Gastroenterol Hepatol 2004;16:55–62. 8. Veloso FT, Ferreira JT, Barros L et al. Clinical outcome of Crohn’s disease: analysis according to the Vienna classification and clinical activity. Inflamm Bowel Dis 2001;7:306–13. 9. Levine A, Karban A, Eliakim R et al. A polymorphism in the TNF-alpha promoter gene is associated with pediatric onset and colonic location of Crohn’s disease. Am J Gastroenterol 2005;100:407–13. 10. Idestrom M, Rubio C, Granath F et al. CARD15 mutations are rare in Swedish pediatric Crohn disease. J Pediatr Gastroenterol Nutr 2005;40:456–60. 11. Heyman MB, Kirschner BS, Gold BD et al. Children with early-onset inflammatory bowel disease (IBD): analysis of a pediatric IBD consortium registry. J Pediatr 2005;146:35–40. 12. Polito JM 2nd, Childs B, Mellits ED et al. Crohn’s disease: influence of age at diagnosis on site and clinical type of disease. Gastroenterology 1996;111:580–6. 13. Louis E, Collard A, Oger AF et al. Behaviour of Crohn’s disease according to the Vienna classification: changing pattern over the course of the disease. Gut 2001;49:777–82. 14. Schwartz DA, Loftus EV Jr, Tremaine WJ et al. The natural history of fistulizing Crohn’s disease in Olmsted County, Minnesota. Gastroenterology 2002;122:875–80. 15. Peyrin-Biroulet L, Chamaillard M. NOD2 and defensins: translating innate to adaptive immunity in Crohn’s disease. J Endotoxin Res 2007;13:135–9. Conclusion The classification of IBD is a formidable task in light of the significant heterogeneity of clinical presentation and disease course. An adequate classification scheme should be practical, easily remembered, and simple enough for clinical applicability, yet comprehensive enough to serve as categorization tool for research studies. Such a classification system for Crohn’s disease, UC, or both will be further strengthened by the identification and integration of novel genetic, serological, and future biomarkers with strong pathophysiological links to various disease phenotypes. 6 However, data available to date do not support their use in clinical practice and decision making at this time. The adoption of a standardized minimal dataset for research protocols may serve as an invaluable tool for the rigorous exploration of genotype–phenotype relationships and better delineate the sensitivity and specificity of potential biomarkers. The Montreal classification system aims to initiate that process. Hopefully, in 5–10 years from now, an integrated classification system for IBD will be more than simply a dream. 16. Wehkamp J, Harder J, Weichenthal M et al. NOD2 (CARD15) mutations in Crohn’s disease are associated with diminished mucosal alpha-defensin expression. Gut 2004:53:1658–64. 17. Cohen RD, Woseth DM, Thisted RA et al. A meta-analysis and overview of the literature on treatment options for left-sided ulcerative colitis and ulcerative proctitis. Am J Gastroenterol 2000;95:1263–76. 18. Ritchie JK, Powell-Tuck J, Lennard-Jones JE. Clinical outcome of the first ten years of ulcerative colitis and proctitis. Lancet 1978;1:1140–3. 19. Langholz E, Munkholm P, Davidsen M et al. Colorectal cancer risk and mortality in patients with ulcerative colitis. Gastroenterology 1992;103:1444–51. 20. Langholz E, Munkholm P, Davidsen M et al. Changes in extent of ulcerative colitis: a study on the course and prognostic factors. Scand J Gastroenterol 1996;31:260–6. 21. 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. 22. Price AB. Overlap in the spectrum of non-specific inflammatory bowel disease. J Clin Pathol 1978;31:567–77. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 7 CLASSIFICATION OF IBD 23. Brown CJ, Maclean AR, Cohen Z et al. Crohn’s disease and indeterminate colitis and the ileal pouch-anal anastomosis: outcomes and patterns of failure. Dis Colon Rectum 2005;48:1542–9. 24. Stewénius J, Adnerhill I, Ekelund GR et al. Operations in unselected patients with ulcerative colitis and indeterminate colitis. A long-term follow-up study. Eur J Surg 1996;162:131–7. 25. Koltun WA, Schoetz DJ Jr, Roberts PL et al. Indeterminate colitis predisposes to perineal complications after ileal pouch-anal anastomosis. Dis Colon Rectum 1991;34:857–60. 26. Holstein A, Burmeister J, Plaschke A et al. Autoantibody profiles in microscopic colitis. J Gastroenterol Hepatol 2006;21:1016–20. 27. Reumaux D, Sendid B, Poulain D et al. Serological markers in inflammatory bowel diseases. Best Pract Res Clin Gastroenterol 2003;17:19–35. 28. Klebl FH, Bataille F, Bertea CR et al. Association of perinuclear antineutrophil cytoplasmic antibodies and anti-Saccharomyces cerevisiae antibodies with Vienna classification subtypes of Crohn’s disease. Inflamm Bowel Dis 2003;9:302–7. 29. Walker LJ, Aldhous MC, Drummond HE et al. Anti-Saccharomyces cerevisiae antibodies (ASCA) in Crohn’s disease are associated with disease severity but not NOD2/CARD15 mutations. Clin Exp Immunol 2004;135:490–6. 30. Teml A, Kratzer V, Schneider B et al. Anti-Saccharomyces cerevisiae antibodies: a stable marker for Crohn’s disease during steroid and 5-aminosalicylic acid treatment. Am J Gastroenterol 2003;98:2226–31. 31. Esters N, Vermeire S, Joossens S et al. Serological markers for prediction of response to anti-tumor necrosis factor treatment in Crohn’s disease. Am J Gastroenterol 2002;97:1458–62. 32. Papp M, Altorjay I, Norman GL et al. Seroreactivity to microbial components in Crohn’s disease is associated with ileal involvement, noninflammatory disease behavior and NOD2/CARD15 genotype, but not with risk for surgery in a Hungarian cohort of IBD patients. Inflamm Bowel Dis 2007;13:984–92. 33. Aisenberg J, Legnani PE, Nilubol N et al. Are pANCA, ASCA, or cytokine gene polymorphisms associated with pouchitis? Long-term follow-up in 102 ulcerative colitis patients. Am J Gastroenterol 2004;99:432–41. 34. Targan SR, Landers CJ, Yang H et al. Antibodies to CBir1 flagellin define a unique response that is associated independently with complicated Crohn’s disease. Gastroenterology 2005;128:2020–8. 35. Mow WS, Vasiliauskas EA, Lin YC et al. Association of antibody responses to microbial antigens and complications of small bowel Crohn’s disease. Gastroenterology 2004;126:414–24. 36. Brant SR, Okazaki T. Inflammatory bowel disease genetics. In: Bernstein CM, ed. Inflammatory Bowel Disease Yearbook. London, UK; Remedica Publishing, 2004:79–128. 37. Economou M, Trikalinos TA, Loizou KT et al. Differential effects of NOD2 variants on Crohn’s disease risk and phenotype in diverse populations: a metaanalysis. Am J Gastroenterol 2004;99:2393–404. 38. Arnott ID, Nimmo ER, Drummond HE et al. NOD2/CARD15, TLR4 and CD14 mutations in Scottish and Irish Crohn’s disease patients: evidence for genetic heterogeneity within Europe? Genes Immun 2004;5:417–25. 39. Guo QS, Xia B, Jiang Yet al. NOD2 3020insC frameshift mutation is not associated with inflammatory bowel disease in Chinese patients of Han nationality. World J Gastroenterol 2004;10:1069–71. 40. Sugimura K, Taylor KD, Lin YC et al. A novel NOD2/CARD15 haplotype conferring risk for Crohn disease in Ashkenazi Jews. Am J Hum Genet 2003;72:509–18. 41. Brant SR, Picco MF, Achkar JP et al. Defining complex contributions of NOD2/CARD15 gene mutations, age at onset, and tobacco use on Crohn’s disease phenotypes. Inflamm Bowel Dis 2003;9:281–9. 42. Cukovic-Cavka S, Vermeire S, Hrstic I et al. NOD2/CARD15 mutations in Croatian patients with Crohn’s disease: prevalence and genotype-phenotype relationship. Eur J Gastroenterol Hepatol 2006;18:895–9. 43. Vermeire S, Louis E, Rutgeerts P et al.; Belgian Group of Infliximab Expanded Access Program and Fondation Jean Dausset CEPH, Paris, France. NOD2/CARD15 does not influence response to infliximab in Crohn’s disease. Gastroenterology 2002;123:106–11. 44. Hampe J, Franke A, Rosenstiel P et al. A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat Genet 2007;39:207–11. 45. Duerr RH, Taylor KD, Brant SR et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 2006;314:1461–3. 46. Yamazaki K, Onouchi Y, Takazoe M et al. Association analysis of genetic variants in IL23R, ATG16L1 and 5p13.1 loci with Crohn’s disease in Japanese patients. J Hum Genet 2007;52:575–83. 47. Martinez A, Marquez A, Mendoza J et al. Role of the PXR gene locus in inflammatory bowel diseases. Inflamm Bowel Dis 2007; [Epub ahead of print]. 48. Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature 2007;448:427–34. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 7 LEADING ARTICLE RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 8 Antineoplastic Effects of 5-Aminosalicylic Acid in the Intestine: From Bench to Bedside Christel Rousseaux, PhD, Olivier Romano, MD, Pierre Desreumaux, MD, PhD Institut National de la Santé et de la Recherche Médicale (INSERM) U795, Physiopathologie des Maladies Inflammatoires Intestinales, CHU Lille, Lille, France Colorectal cancer is a serious complication of IBD, affecting a significantly greater number of IBD sufferers compared with the general population. In patients with ulcerative colitis (UC), the risk of colorectal cancer is reported to increase from 2% at 10 years after diagnosis to 18% at 30 years. Thus, colorectal cancer represents a major challenge in the ongoing surveillance and therapeutic strategy in IBD patients. 5-Aminosalicylic acid (5-ASA) is a long-standing anti-inflammatory agent used in the treatment of IBD, which, in addition to its anti-inflammatory activity, has demonstrated antineoplastic effects in the gut. Recent studies have begun to elucidate the molecular mechanisms of these 5-ASA-mediated effects, and κB, Wnt/β β-catenin a variety of important pathways including arachidonic acid metabolism, inhibition of nuclear factor-κ signaling, epidermal growth factor receptor signaling, control of DNA replication, and peroxisome proliferator-activated receptor γ activation have been implicated. The present authors review the current evidence demonstrating 5-ASA-mediated modulation of colorectal cancer mechanisms. Inflamm Bowel Dis Monit 2007;8(1):8–17. The most serious complication of IBD affecting the colon is colorectal cancer (CRC). Globally, a diagnosis of IBD is associated with a six- to seven-fold increased risk of CRC [1], and for patients with ulcerative colitis (UC), the risk increases from 2% at 10 years after diagnosis to 18% at 30 years [2]. The duration of colitis, extent of the disease in the colon, personal history of primary sclerosing cholangitis, and familial history of colon cancer are well-established risk factors for the development of CRC in IBD patients [3]. The prevention strategy usually relies upon regular surveillance colonoscopy with random biopsies [4]. However, several disadvantages of this costly strategy exist, such as the difficulty in identification of high-risk patients and the performance and interpretation of mucosal biopsies. A promising option in the primary prevention of CRC among IBD patients is mesalazine (also known as mesalamine or 5-aminosalicylic acid [5-ASA]) therapy. 5-ASA is among the oldest anti-inflammatory agents in use today for IBD treatment [5]. New evidence, discussed herein, has emerged related to its potential therapeutic effect in the prevention of CRC occurrence. The mechanisms sustaining the putative antineoplastic effects of 5-ASA are multiple and not fully understood. Owing to its structural similarities to aspirin, it is assumed that 5-ASA has Address for correspondence: Pierre Desreumaux, Service de Gastroentérologie, Hôpital Huriez, CHU Lille, Lille 59037, France. Email: [email protected] 8 similar molecular targets to nonsteroidal anti-inflammatory drugs (NSAIDs). In fact, the therapeutic effect of 5-ASA is obtained through a local effect on colonic epithelial cells rather than via a systemic effect. In this review, the authors detail the preclinical data demonstrating – in vitro, in vivo, and ex vivo in humans – the antineoplastic effect of 5-ASA mediated through “inflammatory”-dependent and “non-inflammatory”-dependent mechanisms. References were selected from the “PubMed” database using combinations of the search words “IBD”, “Crohn”, “ulcerative colitis”, “cancer”, “mesalamine”, “5aminosalicylic acid” and “sulphasalazine”. Only in vitro studies reporting a specific role of 5-ASA (or sulfasalazine), used at clinically relevant concentrations (between 5 and 100 mM), in the modulation of CRC mechanisms are discussed in this review. In vitro studies Inflammatory-dependent antineoplastic mechanisms of 5-ASA Chronic inflammation and cancer are closely associated in the intestine. NSAIDs reduce intestinal neoplasia, while CRC incidence is increased in patients with UC. The main inflammatory-dependent antineoplastic mechanisms of 5-ASA involve inhibition of the nuclear factor-κB (NF-κB) pathway, cyclooxygenase (COX), and prostaglandin (PG) expression, and regulation of cell proliferation/apoptosis. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 9 ANTINEOPLASTIC EFFECTS OF 5-ASA IN THE INTESTINE Inhibition of the NF-κB pathway by 5-ASA Although the molecular mechanisms linking UC with CRC are not well understood, recent results obtained in preclinical models point to the transcription factor NF-κB as a central player. On the one hand, NF-κB regulates the expression of various cytokines and modulates the inflammatory processes in IBD. On the other, NF-κB stimulates the proliferation of tumor cells and enhances their survival through the regulation of anti-apoptotic genes. Furthermore, it has been clearly established that most carcinogens and tumor promoters activate NF-κB, while chemopreventive agents generally suppress this transcription factor. In fact, several lines of evidence suggest that activation of NF-κB may cause cancer [6]. These include the finding that NF-κB genes can act as oncogenes, and that this transcription factor controls apoptosis, cell-cycle progression and cell proliferation, and possibly also cell differentiation. To date, many in vitro studies have been published in the literature showing an inhibition of the NF-κB pathway by 5-ASA or sulphasalazine [7–10]. These studies, performed using rodent and human colonic epithelial cells, demonstrate that 5-ASA or sulphasalazine may inhibit NF-κB via distinct mechanisms: Inhibition of COX and PGs by 5-ASA In inflamed tissue, much of the mucosal PG seems to be generated by the action of inducible COX2 on its substrate, arachidonic acid. COX2 activity is associated with diverse antineoplastic effects such as inhibition of apoptosis [15], regulation of epidermal growth factor receptor (EGFR) and βcatenin signaling, and enhancement of angiogenesis [16–18], effects that are favorable to cancer development. Specific inhibitors of this enzyme, such as celecoxib, have been shown to reduce polyp numbers in individuals with familial adenomatous polyposis [19]. The mechanism underlying this effect still requires clarification. It is not clear which cell type is the predominant source of PGs in the diseased colon, but it is apparent that cancerous epithelial cells demonstrate a marked overexpression of COX2 [20]. There has been remarkably little study of mucosal PG metabolism in colitisassociated dysplasia and cancer, although there is evidence for increased COX2 expression [21]. 5-ASA inhibits lipoxygenase [22], and can be predicted to cause reduced mucosal metabolism of unesterified arachidonic acid. Thus, it is possible that 5-ASA may have a therapeutic effect in the prevention of dysplasia and CRC occurrence in IBD patients through, at least in part, inhibition of COX2 and PGE2 production in colitis-associated colon cancer. • Inhibition of inducible interleukin-1 (IL-1)-stimulated RelA phosphorylation without prevention of inhibitor of NF-κB-α (IκBα) degradation [10]. • Inhibition of NF-κB nuclear translocation and inhibition of the degradation of IκBα [9]. • Inhibition of tumor necrosis factor-α (TNF-α)-induced NF-κB and mitogen-activated protein kinase (MAPK) activation (Fig. 1) [7]. Regulation of cell proliferation and apoptosis by 5-ASA Inflammation mediated through NF-κB activation, production of cytokines or growth factors, and inhibition of anti-apoptotic genes has been shown to induce epithelial cell proliferation and decrease physiological apoptosis, two main abnormalities sustaining CRC formation. The putative chemopreventive actions of 5-ASA include effects on components involved in cell growth and proliferation, for example COX1 and COX2, which regulate cell proliferation through the formation of PGs, lipoxygenase, NF-κB, MAPKs, and the Bcl-2 protein, as well as the activation of apoptotic processes [23]. Many in vitro studies have demonstrated the antiproliferative effect of 5-ASA in colonic adenocarcinoma Caco-2 cells [24]. Reinacher-Schick et al. showed that 5ASA caused a dose- and time-dependent inhibition of the proliferation of colon cancer cells through a mitotic arrest. 5-ASA also induced apoptosis through partial activation of the caspase cascade, probably acting on caspase-3 [25]. These results were confirmed in animal models of carcinogenesis induced by chemical compounds such as azoxymethane (AOM) [26] and 1,2-dimethylhydrazine (DMH) [27], and in a genetically induced tumor model (B6Min/+ mice: B6 mice heterozygous for the multiple intestinal neoplasia nonsense mutation) [26]. Indeed, in these different models, 5-ASA reduced tumor load and The inhibitory roles of 5-ASA on the NF-κB pathway were not detected in lymphocytic leukemia and hepatic stellate cells, suggesting that the 5-ASA-induced NF-κB inhibition may be specific to intestinal epithelial cells [11]. These in vitro data were confirmed in vivo in animals and in humans, thus demonstrating that 5-ASA administration inhibits intestinal expression or activation of NF-κB. In an animal model of colitis induced by trinitrobenzene sulfate (TNBS) in rats, 5-ASA was effective for relieving and repairing colonic inflammation, and the effects were related – at least in part – to a downregulation of IL-1β production and NF-κB expression [12]. In 2006, Kim et al. also demonstrated that 5-ASA inhibited TNF-α-mediated NF-κB activation in a TNBS-induced experimental model of colitis [13]. Similar data were obtained by Bantel and colleagues, providing further evidence that 5-ASA downregulates the activation of NF-κB in actively inflamed mucosa of UC patients [14]. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 9 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 10 CHRISTEL ROUSSEAUX, OLIVIER ROMANO, AND PIERRE DESREUMAUX Figure 1. 5-ASA may inhibit the NF-κB pathway through (i) inhibition of inducible IL-1 stimulated RelA phosphorylation, (ii) inhibition of NF-κB nuclear translocation and degradation of IκBα, and (iii) inhibition of TNF-α induced NF-κB and MAPK activation. TNF IL-1 5-ASA 5-ASA IL-1R TNFR MyD88 MAPKKKs IRAK TRAF6 NEMO TRADD MAPKKs MEK P IKK1 IKK2 P TRAF2 MAPKs (p38, JNKs, ERKs) P Ub P IκB AP1 5-ASA p50 p50 p65 p65 Jun Jun Fos Fos AP1 5-ASA: 5-aminosalicylic acid; AP1: activator protein 1; ERK: extracellular-signal regulated kinase; IκBα: inhibitor of NF-κB subunit α; IL-1: interleukin-1; IL-1R: IL-1 receptor; IKK: IκB kinase; IRAK: IL-1 receptor-associated kinase; JNK: Jun N-terminal kinase; MAPK: mitogen-activated protein kinase; MAPKK: MAPK kinase; MAPKKK: MAPK kinase kinase; MEK: MAP/ERK kinase; NEMO: NF-κB essential modulator; NF-κB: nuclear factor-κB; P: phosphate; RelA: p65 subunit of NF-κB; TNF-α: tumor necrosis factor-α; TNFR: TNF receptor; TRADD: TNF-receptor 1-associated death domain; TRAF: TNF-receptor associated factor; Ub: ubiquitination. number, increased the rate of tumor apoptosis, and reduced the rate of tumor cell proliferation. Similarly, ex vivo studies using biopsies from patients with CRC or sporadic polyps of the large bowel showed an induction of apoptosis and a decrease of cell proliferation after treatment with 5-ASA (1 g/day for 14 days) [25,28]. Non-inflammatory dependent antineoplastic mechanisms of 5-ASA Modulation of the Wnt/β-catenin pathway An important role for β-catenin in colorectal carcinogenesis was implied by its association with the tumor suppressor protein, adenomatous polyposis coli (APC) [29], and by 10 evidence of dysregulation of β-catenin protein expression at all stages of the adenocarcinoma progression. Indeed, APC mutation leads to nuclear accumulation of β-catenin [30]. The degradation of β-catenin involves binding of the protein to a complex involving APC protein, axin, and glycogen synthase kinase-3β (GSK-3β), which serves to phosphorylate serine and threonine residues on β-catenin, a crucial step that is required to target the protein for ubiquitination and proteosomal degradation. An important regulator of GSK-3β activity is the Wnt pathway (for review, see [31]). Free pools of β-catenin must be regulated via a second pathway involving p53-inducible Siah-1. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 11 ANTINEOPLASTIC EFFECTS OF 5-ASA IN THE INTESTINE A direct effect of 5-ASA on the Wnt/β-catenin pathway is described in one recent study using CRC cell lines [32]. These data demonstrated that mesalazine affects the Wnt/βcatenin pathway in APC-mutated cells with intact β-catenin, as assessed by luciferase reporter gene assays. Furthermore, 5-ASA treatment reduced the expression of nuclear β-catenin and Wnt/β-catenin target genes, and increased β-catenin phosphorylation. This effect on the Wnt/β-catenin pathway is mediated via protein phosphatase 2A (PP2A): increased phosphorylation of PP2A is observed after 5-ASA treatment, which coincides with decreased PP2A enzymatic activity. The inhibition of PP2A enzymatic activity by 5-ASA is essential for its effect on the Wnt/β-catenin pathway, as shown by transient transfection with small interfering PP2A RNA and mutant PP2A. This study suggests that by using concentrations of 5-ASA identical to those that are clinically relevant in patients with IBD, the drug inhibits the Wnt/βcatenin pathway via inhibition of PP2A (Fig. 2) [32]. Other studies are needed to confirm this interesting finding. Figure 2. 5-ASA decreases the nuclear translocation of β-catenin through an activation of PPARγ and phosphorylation of PP2A. 5-ASA Conductin/Axin P P PP2A P APC Degradation Disruption of EGFR signaling The EGFR is thought to play a key role in the pathogenesis or maintenance of a number of human cancers of epithelial origin, including CRC. This supposition is based on the demonstration that EGFR itself is overexpressed in CRC cells [33], and that within the neoplastic microenvironment, there is secretion of factors that activate EGFR in an autocrine/paracrine manner [34,35]. Evidence also indicates that the aberrant activation of EGFR contributes to sustain the growth of CRC cells [36]. Indeed, EGFR expression seems to represent an independent predictor of decreased survival in CRC patients. Consistent with this, the EGFR has become a biochemical target for chemoprevention and chemotherapeutic agents against CRC [37,38]. Data from a study by Monteleone et al., who used four different CRC cell lines (HT-29, HT-115, T84, and Caco-2) and ex vivo organ cultures of CRC explants, indicate that clinically relevant concentrations of 5-ASA (from 10 to 50 mM) inhibit EGFR activation by specifically enhancing a phosphorylated EGFR-targeting phosphatase (PTP), namely SH-PTP2 (Fig. 3) [39]. Together, these data suggest that, in vitro, 5-ASA inhibits the activation of EGFR, a transmembrane tyrosine kinase involved in the mitogenic signaling of CRC cells. Control of activating checkpoint responses Another possible target for 5-ASA is the improvement of accuracy of DNA replication. The fidelity of DNA replication is a product of polymerase accuracy, its proofreading activity, and the proficiency of the postreplicational mismatch repair system [40]. Inefficiency of one of these processes can be a Nuclear importation Nucleus APC: adenomatous polyposis coli; GSK-3β: glycogen synthase kinase-3β; PPARγ: peroxisome proliferator-activated receptor γ; PP2A: protein phosphatase 2A. key to the development of human cancer, best illustrated by the familial cancer syndrome, hereditary nonpolyposis CRC (also called Lynch syndrome). Data from Gasche et al. suggest an effect of 5-ASA on the occurrence of frameshift mutations at a (CA)13 microsatellite, which is quite different from the chemopreventive effects of aspirin, despite their structural similarities [41]. The observed effects of 5-ASA on replication fidelity were seen at concentrations >1.25 mmol/L (HCT116-A1.3 cells) or 2.5 mmol/L (HCT116-A2.1 cells) and were greatest at the highest dose tested (5.0 mmol/L). At the 5.0 mmol/L level, a 19% reduction of the mutation rate was observed in mismatch repair-deficient HCT116 colon cancer cells. Keeping in mind that the rate of the emergence of mutations defines the speed of tumor progression (as predicted in the mutator phenotype hypothesis), INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 11 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 12 CHRISTEL ROUSSEAUX, OLIVIER ROMANO, AND PIERRE DESREUMAUX Figure 3. 5-ASA inhibits the activation of EGFR by enhancing SH-PTP2. 5-ASA Ligands of EGFR PGE2 EGFR dephosphorylation SH-PTP2 EGFR EP Amphiregulin PI3K Ras TyrP TyrP TyrP TyrP cAMP Src PKA CREB AKT MAPK Cyclin D1 TCF 5-ASA: 5-aminosalicylic acid; cAMP: cyclic adenosine monophosphate; CREB: cAMP response element-binding protein; EGFR: epidermal growth factor receptor; EP: PGE2 receptor 1; GSK-3β: glycogen synthase kinase-3β; MAPK: mitogen-activated protein kinase; PGE2: prostaglandin E2; PI3K: phosphatidyl inositol 3-kinase; PKA: protein kinase A; SH-PTP2: protein-tyrosine phosphatase containing two Src homology domains; TCF: tumor complement factor; TyrP: phosphorylated tyrosine. 5-ASA therapy could introduce a significantly delay in the clinical manifestation of a tumor. Thus, the authors of the study rightly state that if 5-ASA increases the number of years required for cells to accumulate the requisite number of mutations necessary for invasiveness or metastases by 19%, the drug would significantly reduce the life-threatening manifestations of cancer and diminish cancer deaths even in the absence of reducing cancer incidence. In this respect, 5ASA is potentially useful for prevention of CRC independent of its anti-inflammatory properties. Induction of peroxisome proliferator-activated receptor γ (PPARγ) expression and activation PPARγ is a nuclear receptor mainly expressed by colonic epithelial cells and involved in the regulation of gut homeostasis. Using a computerized medical literature search 12 of all English language articles in the PubMed online database, >100 articles were found that reported a role for PPARγ in the prevention of colon cancer and intestinal inflammation by distinct mechanisms [42,43]. Several molecular mechanisms that could explain the anticarcinogenic functions of PPARγ have been described. The data obtained in studies using various cancer cells that express PPARγ indicate that activation of this receptor by an agonist can: • Inhibit cell proliferation and S phase entry through increased degradation of cyclin D1. • Promote cell death by induction of the anti-apoptotic protein Bcl-2 and inhibition of the survival gene NF-κB. • Stimulate cell differentiation. • Activate the degradation of β-catenin. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 13 ANTINEOPLASTIC EFFECTS OF 5-ASA IN THE INTESTINE Figure 4. 5-ASA is a specific ligand of PPARγ. Binding of 5-ASA to PPARγ induces a coordinated cascade of reactions leading to the translocation of PPARγ to the cell nucleus, a modification of its conformation permitting the recruitment of coactivators (e.g. DRIP) and expression or repression of PPARγ target genes via the activation of the PPRE. Figure 5. In vivo in SCID mice, treatment with 5-ASA (50 mM) for 21 days reduces by 80% the human colonic tumor weight and volume compared with untreated animals. Anti-inflammatory effects Antineoplasic effects 5-ASA Cell membrane 5-ASA: 5-aminosalicylic acid; SCID: severe combined immunodeficient. 5-ASA PPARγ Cytoplasm DRIP 5-ASA RXR PPARγ Nucleus nucleus, to promote a PPARγ conformational change, and to recruit a coactivator named vitamin D receptor-interacting protein (DRIP). Docking simulations showed a binding mode of 5-ASA very similar to the crystal orientation of the known ligand of PPARγ. 5-ASA fitted tightly with the PPARγ ligandbinding domain, interacting with key determinants required for molecular recognition and PPARγ activation (Fig. 4). In the HT-29 colonic epithelial cell line, 5-ASA induces PPARγ expression, inhibits colonic epithelial cell proliferation, and induces epithelial cell apoptosis through binding to PPARγ [42]. In an in vivo model of severe combined immunodeficient (SCID) mice engrafted with human colon cancer, treatment with 5-ASA decreased the volume and weight of a human colonic tumor by 80%, in a PPARγdependent manner (Fig. 5). PPRE 5-ASA: 5-aminosalicylic acid; DRIP: vitamin D receptor-interacting protein; PPARγ: peroxisome proliferator-activated receptor γ, PPRE: PPARγ response element; RXR: retinoid X receptor. Redrawn from [49] with permission. • Induce expression of the transcriptional repressor transforming growth factor β-stimulated protein TSC22 and of the tumor-suppressor gene phosphatase and tensin homologue (PTEN). • Inhibit angiogenesis. Taken together, PPARγ acts as a tumor-suppressor gene at an early step in the development of colonic tumors before damage of the APC/β-catenin pathway. Recently, 5-ASA was described – at clinically relevant concentrations – to be a new synthetic ligand of PPARγ, able to induce its expression and activation in colonic epithelial cells (Fig. 2). 5-ASA is also able to bind PPARγ inducing its translocation from the cytosol of epithelial cells to the Summary 5-ASA regulates several mechanisms, related and unrelated to inflammation, associated with the enhanced risk of CRC occurrence in patients with IBD located in the colon. Regarding the role of 5-ASA during inflammation, it is only recently that an integrated understanding of the mechanisms of action of aminosalicylates has been proposed involving the nuclear receptor PPARγ, as previously described in this journal [44]. 5-ASA interacts with PPARγ, inducing its activation in epithelial cells, explaining, at least in part, the reduction of many key elements of inflammation such as the NF-κB signaling pathway, the production of cytokines and chemokines, the synthesis of COX enzymes and PGs, and the expression of adhesion molecules. The antineoplastic effect of 5-ASA is not solely due to PPARγ binding but involves also a reduced expression of nuclear β-catenin and Wnt/β-catenin target genes, increased β-catenin phosphorylation, disruption of EGFR signaling, and an enhanced fidelity of epithelial cell replication. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 13 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 14 CHRISTEL ROUSSEAUX, OLIVIER ROMANO, AND PIERRE DESREUMAUX Figure 6. Antineoplastic effects of 5-ASA can be divided into (i) inflammatory dependent mechanisms including an inhibition of the NF-κB pathway and COX2/PGE2 production, and (ii) non-inflammatory dependent mechanisms involving the Wnt/β-catenin pathway, disruption of EGFR signaling, and control of DNA replication. Inflammatory dependent mechanisms Non-inflammatory dependent mechanisms Wnt Wnt IKKγ P P P P P Axin IKKγ Axin APC Aspirin Salicylate Sulindac P RelA p50 RelA p50 P P APC P P RelA p50 P Ub P Ub Ub Ub Proteasomal Ub degradation Ub P 26S proteasome Groucho TCF RelA p50 Inflammation (cytokines, COX) Cell proliferation Cell growth Cell differentiation Apoptosis PGI2 TXA2 PGE2 PGF1α PGD2 Apoptosis PGH2 COX2 Arachidonic acid sPLA2 cPLA2 Repression 15d-PGJ2 CBP PYG Activation BCL9 TCF LBD Activation of signaltransduction cascades Apoptosis Invasion and metastasis Cell proliferation Angiogenesis 5-ASA Control of activating checkpoint response DNA replication P TK TK P TK TK EGFR COX/PGE2 5-ASA: 5-aminosalicylic acid; APC: adenomatous polyposis coli; β-cat: β-catenin; β-TRCP: β-transducin repeat containing protein; BCL9: B-cell lymphoma protein 9; CBP: cyclic adenosine monophosphate response element-binding protein; CK1α: casein kinase 1α; COX2: cyclooxygenase 2; cPLA2: cytosolic phospholipase A2; EGFR: epidermal growth factor receptor; GSK-3β: glycogen synthase kinase-3β; IκB: inhibitor of NF-κB; IKKα: IκB kinase α subunit; IKKβ: IκB kinase β subunit; LBD: ligand-binding domain; NF-κB: nuclear factor-κB; P: phosphate; PGD2: prostaglandin D2; PGE2: prostaglandin E2; PGF1α: prostaglandin F1α; PGH2: prostaglandin H2; PGI2: prostaglandin I2; PGJ2: prostaglandin J2; 15d-PGJ2: 15-deoxy-prostaglandin J2; PYG: pygopus; sPLA2: secretory phospholipase A2; TCF: tumor complement factor; TK: tyrosine kinase; TXA2: thromboxane A2; Ub: ubiquitination. 14 INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 15 ANTINEOPLASTIC EFFECTS OF 5-ASA IN THE INTESTINE Taken together, these in vitro data suggest that the antineoplastic effects of 5-ASA are multistep processes (summarized in Fig. 6). 5-ASA acts as a tumor-suppressor at an early stage in the development of colonic tumors, before damage of the APC/β-catenin pathway, but also at a later stage, delaying tumor progression. In vivo studies in mice This latter section of the review will focus on studies investigating the roles of 5-ASA in different experimental models of CRC in rodents including mice and rats. Briefly, there are two main groups of validated experimental models of CRC. The first are chemical-induced CRC models in mice and rats, using: • AOM, leading to aberrant crypt foci followed by invasive cancer. • DMH, resulting in Ras mutation and leading to invasive carcinomas. • N-methyl-N-nitrosurea (NMNU), leading to colon cancer within 10 days. To better mimic IBD-associated colon cancer, some studies have associated the administration of chemical carcinogen with inflammatory compounds, for example, AOM and dextran sodium sulphate (DSS). The second group of validated experimental models of CRC is characterized by the spontaneous occurrence of small bowel and colon cancer in mice carrying a nonsense mutation in the murine homologue of the APC gene, named APCMin mice. In all of these experimental models, 5-ASA has been demonstrated to prevent or reduce colon cancer occurrence. Presentation of studies in animals The antineoplastic effect of 5-ASA has been well characterized in chemically induced CRC in mice and rats. In 1992, Davies et al. evaluated the effect of therapeutic drugs used for the treatment of IBD in a model of colonic cancer induced by DMH (40 mg/kg) in Wistar rats [45]. They found that 5-ASA (60 mg/kg/day) inhibited tumor size by >60% compared with untreated animals. In the study by MacGregor et al., balsalazide reduced aberrant crypt foci induced by AOM in a dose-dependent manner, by 60% [26]. Similarly, 5-ASA treatment in rats receiving DMH reduced tumor number and load through an increased rate of tumor apoptosis and a reduced tumor cell proliferation rate [27]. The preventive anticarcinogenic effect of 5-ASA was also confirmed by Narisawa et al. in 2003, using a model of colon cancer induced by NMNU in F344 rats. They demonstrated that intrarectal administration of 5-ASA strongly suppressed the initial stage of chemically induced colon carcinogenesis (by 70%) [46]. In Min mice, which develop intestinal neoplasia, two studies report controversial results depending on the genetic strain of animals. Ritland et al. failed to observe the effect of several formulations of 5-ASA (free acid, sulfasalazine, and Pentasa® [Shire, Wayne, PA, USA]) at multiple dosage levels on tumor number in APCMin mice [47]. In contrast, MacGregor et al. treated another strain: B6-Min/+ mice, and found a dose-dependent reduction of intestinal tumor number, reaching 80% inhibition in the distal small intestine and colon [26]. Summary Most preclinical studies performed in experimental models of CRC in mice and rats demonstrated that 5-ASA, at clinically relevant concentrations, prevented and inhibited the progression of colonic tumors by 60–70%. These results confirm and extend data obtained in vitro using intestinal epithelial cell lines. Controversial results are only observed in one experimental model of CRC occurring in Min mice [47]. If we consider that 5-ASA acts at an early stage in the development of colonic tumors, before damage of the APC/βcatenin pathway, it is not surprising that the antineoplastic effect of 5-ASA was reduced in this model, which is characterized by mutation of the APC/β-catenin pathway. Ex vivo studies in human Endoscopic studies in IBD patients Inflammation-related antineoplastic mechanisms of 5-ASA involving an inhibition of the NF-κB pathway were investigated in inflammatory colonic endoscopic biopsies of 20 UC patients treated with 5-ASA [14]. In this study, Bantel and colleagues showed that 8 weeks of treatment with 5-ASA resulted in a strong abrogation of NF-κB activation in situ, evaluated by immunostaining using a specific antibody directed against activated NF-κB. Regarding the therapeutic properties of 5-ASA on cell apoptosis and proliferation, Reinacher-Schick and colleagues investigated the effects of 5-ASA ex vivo in 21 patients with sporadic colorectal polyps (≥5 mm) [48]. Among these patients, 17 underwent polypectomy and biopsy of uninvolved mucosa before and after treatment with 5-ASA 1 g/day (for 1, 3, 7, or 14 days); four patients served as untreated controls. Apoptotic and proliferation rates were measured by terminal transferase dUTP nick end labeling (TUNEL) assay and immunostaining using an anti-Ki67 antibody, respectively. The authors found that apoptosis was significantly increased 1 and 3 days after initiation of treatment with 5-ASA compared with control subjects (p=0.0107 for the 1-day treatment group and p=0.0142 for the 3-day treatment group), and inversely that proliferation appeared to be decreased by mesalazine in all treatment INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 15 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 16 CHRISTEL ROUSSEAUX, OLIVIER ROMANO, AND PIERRE DESREUMAUX groups, while proliferation in controls did not change. Similarly, in 10 patients with CRC, mesalazine enemas for 14 days were associated with an increased apoptotic score and a decreased cell proliferation in malignant tissue, according to the Ki67 antibody staining [28]. Disclosures Drs Rousseaux and Romano have no relevant financial interests to disclose. Dr Desreumaux has received consulting fees from, or served on advisory boards for, Biofortis, Danisco France SAS, Danone France, Ferring, Giuliani S.p.A., Roquette, TxCell, and UCB Pharma. Dr Desreumaux has also received lecture fees from CME events indirectly Short-term culture of intestinal biopsies with 5-ASA supported by commercial sponsors (Ferring, Procter & Gamble, As mentioned earlier, an inhibitory role of 5-ASA on EGFR signaling was demonstrated in ex vivo organ cultures of CRC explants in which clinically relevant concentrations of 5-ASA (10–50 mM) inhibit EGFR activation by specifically enhancing a phosphorylated EGFR-targeting phosphatase (PTP), namely SH-PTP2 [39]. These data suggest that, in vivo, 5-ASA inhibits the activation of EGFR, a transmembrane tyrosine kinase involved in the mitogenic signaling of CRC cells. Confirmation of functional binding of 5-ASA to PPARγ, a nuclear receptor with antineoplastic effects was obtained ex vivo in colonic biopsies of control subjects and patients with IBD (both Crohn’s disease and UC patients). In this study, 5ASA at clinically relevant concentrations (30 and 50 mM) induced PPARγ expression and activation, inducing particularly the expression of the PPARγ-activation related gene NGal [43]. Schering Plough, Shire, and UCB Pharma), and grant support from Astra Summary These ex vivo studies performed using colon specimens of different patients with UC, Crohn’s disease, sporadic polyps, and CRC confirm the preclinical results demonstrating that 5-ASA in human colon decreases NF-κB activation, epithelial cell proliferation, and EGFR signaling, increases epithelial cell apoptosis and induces PPARγ expression and activation. Conclusion 5-ASA has been a cornerstone in the treatment of UC patients for many years to control disease activity and, in the last decade, to reduce the risk of development of CRC. Large epidemiological studies have shown that between 48% and 65% of patients with UC receive continuous 5-ASA treatment. As ethical considerations mean that it will never be possible to carry out a randomized, placebocontrolled clinical trial to assess the efficacy of 5-ASA in reducing the risk of CRC in patients with UC, a better understanding of the mechanisms sustaining the putative antineoplastic effects of 5-ASA is crucial. The present in vitro data, in vivo studies in mice and rats, and the ex vivo experiments performed in patients provide a mechanistic foundation for the possibility that long-term treatment with 5-ASA can reduce the risk of CRC in patients with IBD located in the colon. This statement would imply that these patients should be put on continuous 5-ASA treatment to reduce the risk of neoplasia development. 16 Zeneca, Danisco France SAS, Danone France, Ferring, Giuliani S.p.A., Lesaffre, Ocera Therapeutics, Roquette, Sanofi-Synthelabo, UCB Pharma, and Yoplait. References 1. Terdiman JP, Steinbuch M, Blumentals WA et al. 5-Aminosalicylic acid therapy and the risk of colorectal cancer among patients with inflammatory bowel disease. Inflamm Bowel Dis 2007;13:367–71. 2. Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in ulcerative colitis: a meta-analysis. Gut 2001;48:526–35. 3. Eaden J. Review article: colorectal carcinoma and inflammatory bowel disease. Aliment Pharmacol Ther 2004;20(Suppl 4):24–30. 4. Ahmadi AA, Polyak S. Endoscopy/surveillance in inflammatory bowel disease. Surg Clin North Am 2007;87:743–62. 5. Moody GA, Jayanthi V, Probert CS et al. Long-term therapy with sulphasalazine protects against colorectal cancer in ulcerative colitis: a retrospective study of colorectal cancer risk and compliance with treatment in Leicestershire. Eur J Gastroenterol Hepatol 1996;8:1179–83. 6. Schottelius AJ, Dinter H. Cytokines, NF-kappaB, microenvironment, intestinal inflammation and cancer. Cancer Treat Res 2006;130:67–87. 7. Kaiser GC, Yan F, Polk DB. Mesalamine blocks tumor necrosis factor growth inhibition and nuclear factor kappaB activation in mouse colonocytes. Gastroenterology 1999;116:602–9. 8. Liptay S, Bachem M, Hacker G et al. Inhibition of nuclear factor kappa B and induction of apoptosis in T-lymphocytes by sulfasalazine. Br J Pharmacol 1999;128:1361–9. 9. Wahl C, Liptay S, Adler G et al. Sulfasalazine: a potent and specific inhibitor of nuclear factor kappa B. J Clin Invest 1998;101:1163–74. 10. Egan LJ, Mays DC, Huntoon CJ et al. Inhibition of interleukin-1-stimulated NF-kappaB RelA/p65 phosphorylation by mesalamine is accompanied by decreased transcriptional activity. J Biol Chem 1999;274:26448–53. 11. Habens F, Srinivasan N, Oakley F et al. Novel sulfasalazine analogues with enhanced NF-κB inhibitory and apoptosis promoting activity. Apoptosis 2005;10:481–91. 12. Song M, Xia B, Li J. Effects of topical treatment of sodium butyrate and 5-aminosalicylic acid on expression of trefoil factor 3, interleukin 1beta, and nuclear factor kappaB in trinitrobenzene sulphonic acid induced colitis in rats. Postgrad Med J 2006;82:130–5. 13. Kim H, Jeon H, Kong H et al. A molecular mechanism for the anti-inflammatory effect of taurine-conjugated 5-aminosalicylic acid in inflamed colon. Mol Pharmacol 2006;69:1405–12. 14. Bantel H, Berg C, Vieth M et al. Mesalazine inhibits activation of transcription factor NF-kappaB in inflamed mucosa of patients with ulcerative colitis. Am J Gastroenterol 2000;95:3452–7. 15. Gasparini G, Longo R, Sarmiento R et al. Inhibitors of cyclo-oxygenase 2: a new class of anticancer agents? Lancet Oncol 2003;4:605–15. 16. Cianchi F, Cortesini C, Fantappie O et al. Cyclooxygenase-2 activation mediates the proangiogenic effect of nitric oxide in colorectal cancer. Clin Cancer Res 2004;10:2694–704. 17. Gately S, Li WW. Multiple roles of COX-2 in tumor angiogenesis: a target for antiangiogenic therapy. Semin Oncol 2004;31(2 Suppl 7):2–11. 18. Kuwano T, Nakao S, Yamamoto H et al. Cyclooxygenase 2 is a key enzyme for inflammatory cytokine-induced angiogenesis. FASEB J 2004;18:300–10. 19. Steinbach G, Lynch PM, Phillips RK et al. The effect of celecoxib, a cyclooxygenase-2 inhibitor, in familial adenomatous polyposis. N Engl J Med 2000;342:1946–52. 20. Mauritz I, Westermayer S, Marian B et al. Prostaglandin E(2) stimulates progression-related gene expression in early colorectal adenoma cells. Br J Cancer 2006;94:1718–25. 21. Khan KN, Masferrer JL, Woerner BM et al. Enhanced cyclooxygenase-2 expression in sporadic and familial adenomatous polyposis of the human colon. Scand J Gastroenterol 2001;36:865–9. 22. Lauritsen K, Laursen LS, Kjeldsen J et al. Effects of mesalazine on the formation of lipoxygenase and cyclooxygenase products. Adv Exp Med Biol 1995;371B:1301–6. 23. Allgayer H. Review article: mechanisms of action of mesalazine in preventing colorectal carcinoma in inflammatory bowel disease. Aliment Pharmacol Ther 2003; 18(Suppl 2):10–4. 24. Orchel A, Parfiniewicz B, Lodowska J et al. The effect of sulphasalazine and its metabolites on the colonic epithelial Caco-2 cells. Acta Pol Pharm 2003;60:106–8. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 17 ANTINEOPLASTIC EFFECTS OF 5-ASA IN THE INTESTINE 25. Reinacher-Schick A, Schoeneck A, Graeven U et al. Mesalazine causes a mitotic arrest and induces caspase-dependent apoptosis in colon carcinoma cells. Carcinogenesis 2003;24:443–51. 26. MacGregor DJ, Kim YS, Sleisenger MH et al. Chemoprevention of colon cancer carcinogenesis by balsalazide: inhibition of azoxymethane-induced aberrant crypt formation in the rat colon and intestinal tumor formation in the B6-Min/+ mouse. Int J Oncol 2000;17:173–9. 37. Bianco R, Gelardi T, Damiano V et al. Rational bases for the development of EGFR inhibitors for cancer treatment. Int J Biochem Cell Biol 2007;39:1416–31. 38. Iqbal S, Lenz HJ. Integration of novel agents in the treatment of colorectal cancer. Cancer Chemother Pharmacol 2004;54(Suppl 1):S32–9. 39. Monteleone G, Franchi L, Fina D et al. Silencing of SH-PTP2 defines a crucial role in the inactivation of epidermal growth factor receptor by 5-aminosalicylic acid in colon cancer cells. Cell Death Differ 2006;13:202–11. 27. Brown WA, Farmer KC, Skinner SA et al. 5-Aminosalicyclic acid and olsalazine inhibit tumor growth in a rodent model of colorectal cancer. Dig Dis Sci 2000;45:1578–84. 40. Plotz G, Zeuzem S, Raedle J. DNA mismatch repair and Lynch syndrome. J Mol Histol 2006;37:271–83. 28. Bus PJ, Nagtegaal ID, Verspaget HW et al. Mesalazine-induced apoptosis of colorectal cancer: on the verge of a new chemopreventive era? Aliment Pharmacol Ther 1999;13:1397–402. 41. Gasche C, Goel A, Natarajan L et al. Mesalazine improves replication fidelity in cultured colorectal cells. Cancer Res 2005;65:3993–7. 29. Su LK, Vogelstein B, Kinzler KW. Association of the APC tumor suppressor protein with catenins. Science 1993;262:1734–7. 30. Hulsken J, Birchmeier W, Behrens J. E-cadherin and APC compete for the interaction with beta-catenin and the cytoskeleton. J Cell Biol 1994;127:2061–9. 31. Wong NA, Pignatelli M. Beta-catenin – a linchpin in colorectal carcinogenesis? Am J Pathol 2002;160:389–401. 32. Bos CL, Diks SH, Hardwick JC et al. Protein phosphatase 2A is required for mesalazinedependent inhibition of Wnt/beta-catenin pathway activity. Carcinogenesis 2006;27:2371–82. 33. Spano JP, Fagard R, Soria JC et al. Epidermal growth factor receptor signaling in colorectal cancer: preclinical data and therapeutic perspectives. Ann Oncol 2005;16:189–94. 34. Bradley SJ, Garfinkle G, Walker E et al. Increased expression of the epidermal growth factor receptor on human colon carcinoma cells. Arch Surg 1986;121:1242–7. 35. Janmaat ML, Giaccone G. The epidermal growth factor receptor pathway and its inhibition as anticancer therapy. Drugs Today (Barc) 2003;39(Suppl C):61–80. 36. Pai R, Soreghan B, Szabo IL et al. Prostaglandin E2 transactivates EGF receptor: a novel mechanism for promoting colon cancer growth and gastrointestinal hypertrophy. Nat Med 2002;8:289–93. 42. Grommes C, Landreth GE, Heneka MT. Antineoplastic effects of peroxisome proliferatoractivated receptor gamma agonists. Lancet Oncol 2004;5:419–29. 43. Rousseaux C, Lefebvre B, Dubuquoy L et al. Intestinal antiinflammatory effect of 5-aminosalicylic acid is dependent on peroxisome proliferator-activated receptor-gamma. J Exp Med 2005;201:1205–15. 44. Vignal C, Rousseaux C, Peyrin-Biroulet L et al. Novel anti-inflammatory mechanisms of 5-ASAs in ulcerative colitis. Inflamm Bowel Dis Monit 2007;7:90–8. 45. Davis AE, Patterson F, Crouch R. The effect of therapeutic drugs used in inflammatory bowel disease on the incidence and growth of colonic cancer in the dimethylhydrazine rat model. Br J Cancer 1992;66:777–80. 46. Narisawa T, Fukaura Y. Prevention by intrarectal 5-aminosalicylic acid of N-methyl nitrosourea-induced colon cancer in F344 rats. Dis Colon Rectum 2003;46:900–3. 47. Ritland SR, Leighton JA, Hirsch RE et al. Evaluation of 5-aminosalicylic acid (5-ASA) for cancer chemoprevention: lack of efficacy against nascent adenomatous polyps in the Apc(Min) mouse. Clin Cancer Res 1999;5:855–63. 48. Reinacher-Schick A, Seidensticker F, Petrasch S et al. Mesalazine changes apoptosis and proliferation in normal mucosa of patients with sporadic polyps of the large bowel. Endoscopy 2000;32:245–54. 49. Dubuquoy L, Rousseaux C, Thuru X et al. PPAR gamma as a new therapeutic target in inflammatory bowel diseases. Gut 2006;55:1341–9. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 17 LEADING ARTICLE RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 18 Orofacial Granulomatosis and Oral Crohn’s Disease Carlo Nunes, MD1, Miranda Lomer, PhD1, Michael Escudier, FDS, RCS2, Stephen Challacombe, FMedSci2, and Jeremy Sanderson, MD1 1 Department of Gastroenterology, Guy’s & St Thomas’s NHS Foundation Trust, and 2Department of Oral Medicine, Kings College London Dental Institute, London, UK Orofacial granulomatosis (OFG) is a rare, chronic inflammatory disease of unknown etiology affecting the orofacial tissue, with no apparent systemic involvement. Lip swelling is the most common clinical presentation but several intra-oral sites may also be affected. OFG is used as an umbrella term and includes the Melkersson–Rosenthal syndrome, cheilitis granulomatosa, and systemic granulomatous diseases, of which, Crohn’s disease is the most common. Histological characteristics consist of epithelioid granulomas and lymphedema of the corium. No definitive etiological cause has been identified, but dietary antigens appear to play an important role in the pathogenesis of the disease. Dietary manipulation, in particular a cinnamon- and benzoate-free diet, is used with success as a first-line treatment. Failing dietary therapy, disease management largely follows a Crohn’s disease-like treatment regimen involving immunosuppressants, whilst lipreduction surgery is reserved for treatment-resistant, severe cases. Inflamm Bowel Dis Monit 2007;8(1):18–22. Orofacial granulomatosis (OFG) is an uncommon chronic inflammatory condition affecting the mouth presenting most often with lip swelling but also affecting multiple sites throughout the oral cavity. The term “OFG” is used as an umbrella to cover a range of conditions in which granulomatous inflammation may involve the oral cavity, with the commonest of these being Crohn’s disease [1]. More rarely, sarcoidosis and tuberculosis present in this way [2,3]. Importantly, however, it is clear that OFG can exist as a separate entity [4]. The Melkersson–Rosenthal syndrome (MRS) and cheilitis granulomatosa, which is regarded as an oligosymptomatic form of MRS, are also included under the umbrella term OFG. The true prevalence is unknown as no reliable epidemiological data exist [5]. In known Crohn’s disease, true OFG occurs in <1% of cases [6]. Conversely, the incidence of the various other oral manifestations of Crohn’s disease may be as high as 20% [7]. OFG is more common in the pediatric and young adult population and, anecdotally, a long-held view has been that cases of pediatric OFG would frequently evolve into classical gut Crohn’s disease in adolescence or adulthood. Certainly, a small number of cases with OFG may Address for correspondence: Jeremy Sanderson, Department of Gastroenterology, 1st Floor, College House, Guy’s & St Thomas’ NHS Foundation Trust, Lambeth Palace Road, London, SE1 7EH, UK. Email: [email protected] 18 precede gut Crohn’s disease by a number of years, but specialist experience suggests this is the exception rather than the rule [8–11]. Geographical variation in the prevalence of OFG has been observed but not documented in any formal study. The condition is reported most frequently in the UK literature and there is a remarkable Celtic predominance. The greatest numbers of cases are seen in Scotland (in particular, Glasgow) and in Ireland [5,12]. Importantly, the psychosocial impact of OFG is disproportionately high, particularly in teenagers and young adults where avoidance behavior and anxiety/depression scores match those of individuals with major facial burns (Sanderson J, unpublished data). Clinical features The onset of OFG is often with acute or subacute lip swelling, either upper or lower, or both, and whilst the first episode may subside completely in hours to days, subsequent attacks may not fully resolve leaving the lip chronically swollen [13]. Spontaneous resolution is rare but has been reported [14]. The majority follow a chronic relapsing course, not infrequently with chronic swelling and intervening bouts of acute swelling. The features of true OFG should not be confused with the more common oral manifestations of Crohn’s disease, which include aphthous-like ulceration, angular cheilitis, glossitis, and intra-oral candidiasis. These are often related to INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 19 OFG AND ORAL CROHN’S DISEASE active intestinal disease, to hematinic deficiency, or to drug treatment (corticosteroids in particular). The characteristic features of OFG consist of lip swelling (Fig. 1), often with fissuring, and erythema, edema, and nodularity affecting a variety of intra-oral sites. Buccal cobblestoning, mucosal tags, and deep, painful, linear sulcal ulceration are features that are very reminiscent of gut Crohn’s disease [1,15]. Gingival enlargement and erythema is also commonly seen and may be the presenting feature [16]. Involvement of the salivary duct orifices in the floor of the mouth with firm swelling and erythema is unique and often referred to as a “staghorn appearance” [17]. Fissuring of the dorsum of the tongue is less common, but when seen in association with lip swelling and a lower motor neurone VIIth nerve palsy, is referred to as MRS [18,19]. Facial swelling and erythema, most commonly spreading peri-orally, may also occur. Rarely, cervical lymphadenopathy may be present [20]. Etiology The etiology of OFG remains unknown. The association with Crohn’s disease inevitably suggests that the pathogenesis involves an interaction between genetic susceptibility and environmental trigger factors. However, unlike most forms of Crohn’s disease, OFG has clinical features in keeping with an allergic mechanism, and an increased prevalence of atopy (up to 60%) is well described [16,21–24]. Whilst earlier reports of cutaneous patch testing to agents such as cobalt, dental materials, and food additives revealed no consistent pattern, more recent evidence suggests that dietary cinnamon and benzoate may play an important role in the disease [17,25]. Cutaneous patch test studies at the Glasgow Dental School (Glasgow, UK) related cinnamaldehyde and benzoate strongly to OFG and a favorable response was reported when these substances were excluded from the diet [26]. Benzoates are a preservative in many foods, especially fizzy drinks, whilst cinnamaldehyde is used as flavoring in soft drinks, chewing gum, ice cream, cakes, toothpaste, and mouthwashes. Whilst anecdotal, the remarkable incidence of OFG in Glasgow may relate to the well-known high rates of fizzy drink consumption (The Independent [London], August 17, 1998). Immunologically, the inflammatory infiltrate is T cell driven, with the formation of non-caseating epithelioid granulomas that frequently are deep seated in the corium [27,28]. There are few studies that have addressed the pathogenesis and immunology of OFG. Gibson and Wray demonstrated specific human leukocyte antigen genotypes, A2/3, B7, and DR2/3/4, in 16 patients with OFG [29], whilst Lim et al. identified a restricted T cell receptor (TCR) V-β gene expression by lesional lymphocytes compared with normal peripheral blood lymphocytes in patients with OFG. In the latter study, there was also an identical, unique V-D-J Figure 1. Predominant lower lip swelling and erythema in a patient with OFG. OFG: orofacial granulomatosis. Available in color at www.ibdmonitor.com junctional sequence seen in >20% of the V-β 6 TCR transcripts, suggesting a local antigen-driven V-β 6 T cell clonal expansion within the lesional lymphocytes only [28]. Moreover, immunohistochemical studies suggest an inflammatory process driven by a type 1 helper T cell environment similar to that seen in gut Crohn’s disease [30]. Recent evidence also implicates B cells in the pathogenesis of OFG [31]. Diagnosis The diagnosis of OFG may be delayed, not least because the disease is uncommon and presentation may be limited to intra-oral disease. Moreover, patients often present to a variety of specialists, most frequently general dental practitioners and maxillofacial surgeons [32]. The diagnosis of OFG is confirmed by typical histological changes observed in biopsies of affected sites, most commonly lip or buccal mucosa. The characteristic findings are the presence of noncaseating epithelioid granulomas on the background of a chronic inflammatory cell infiltrate (Fig. 2). Lymphedema of the corium and dilated lymphatics are also seen and are a major contributor to the persistence of lip swelling [1,13]. Granulomas are frequently deepseated and deeper sections may be required for a definitive diagnosis [27]. The current authors’ group has demonstrated, using ileocolonoscopy, that minor macroscopic intestinal abnormalities with microscopic granulomatosis are common and present in up to 60% of patients with OFG who have no gastrointestinal symptoms [33]. These findings were more common in those aged <30 years and, importantly, most patients have not progressed to gut Crohn’s disease with up to 15 years of follow-up. The true significance of these findings is not clear but they suggest that, in most cases, OFG represents an individual disease entity. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 19 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 20 CARLO NUNES ET AL. Figure 2. H/E-stained lip histology from a patient with OFG showing a sub-epithelial epithelioid granuloma, with surrounding chronic inflammatory cell infiltrate. The inflammatory infiltrate has focally infiltrated the epithelium. Magnification: x20. H/E: hematoxylin/eosin; OFG: orofacial granulomatosis. Available in color at www.ibdmonitor.com Approximately one in five patients with OFG from the present authors’ institute have concurrent gut Crohn’s disease and, although a large number present with gut involvement at the time of diagnosis, only a small number subsequently progress to true gut Crohn’s disease. The ability to predict which patients are at risk of developing Crohn’s disease is important as early therapeutic intervention may improve the natural history of the disease. Clinically, buccal cobblestoning and mucosal tags are predictive of Crohn’s disease compared to lip swelling [32]. Likewise, those with elevated inflammatory markers and hematinic deficiencies are significantly more likely to suffer from concurrent gut Crohn’s disease [32]. A recent investigation measuring a specific Saccharomyces cerevisiae 200-kDa glycoprotein antigen in serum and saliva in patients with OFG suggested that raised serum immunoglobulin A (IgA) antibodies to the S cerevisiae 200-kDa antigen are predictive of gut inflammation, while elevated salivary IgA antibody levels were not [34]. Thus, high titers of serum IgA antibodies to the S cerevisiae 200-kDa antigen might identify a subgroup of patients with OFG who may benefit from early gastrointestinal investigation. Management of OFG The initial analysis of patients with suspected OFG should include a detailed oral examination, and referral to a specialist 20 oral medicine clinic is advisable. Hematological assessment should include standard inflammatory parameters as these may reflect systemic involvement. A chest X-ray and serum angiotensin-converting enzyme level should be considered during preliminary analysis, for exclusion of sarcoidosis. Swabs of lip fissures and angular cheilitis are important as secondary infection is common, and aggravates inflammation. Likewise, salivary Candida culture may identify a group benefiting from antifungal medication. As with dermatology, clinical photography is an important element of recording clinical signs at any visit and an oral activity score can also be used to quantify disease activity. The treatment of OFG remains a challenge and strategies used are largely based on small, non-randomized trials and anecdotal case reports. In general, the management of OFG follows a similar approach to that used in gut Crohn’s disease. Initial aims are to induce remission followed by maintenance therapy and can be largely divided into general, topical, systemic, and surgical therapy. Also relevant is whether patients have concurrent Crohn’s disease as therapy aimed at active gut disease may result in improvement of oral disease. In those with minor symptoms only or disease that remits spontaneously, it is possible that no therapy will be required. A multidisciplinary approach to management is ideal with a team consisting primarily of an oral/dental surgeon, gastroenterologist, and dietitian. General Non-specific lesions such as glossitis, aphthous-like ulcers, and oral candidiasis may often indicate concurrent hematinic and/or nutritional deficiencies [35]. Particular attention should be paid to correcting vitamin B12, folate, and iron deficiencies. Patients with malnutrition may benefit from calorie supplementation. Topical antibiotics, such as Fucidin® (Leo Pharma, Princes Risborough, Buckinghamshire, UK), should be used in cases of infected lip fissures and/or angular cheilitis. Moreover, oral hygiene and regular dental inspection should be encouraged as poor dentition and periodontitis may be present at the time of diagnosis, or be a consequence of treatment such as corticosteroids. It is also important to exclude and treat any underlying infective foci such as a dental abscess prior to considering immunosuppressive therapy, in particular biologics, as infective/ septic complications have been described [36]. Dietary The role of dietary manipulation in the form of exclusion diets has shown some promise in the last few years and supports the view that dietary antigens play a substantial role in the pathogenesis of OFG. At the authors’ institution, a cinnamon- and benzoate-free diet is used as first-line INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 21 OFG AND ORAL CROHN’S DISEASE treatment in patients with confirmed OFG. Moreover, using an oral activity score, the authors have demonstrated a 68% (39/57 patients) response rate in both lip and intra-oral disease following a cinnamon- and benzoate-free diet for a period of 12 weeks [17]. The use of enteral nutrition (either polymeric or elemental) for a period of 6 weeks as secondline therapy may also be effective, particularly in those with intra-oral compared with lip disease [37]. As with gut Crohn’s disease, compliance may be a particular problem in younger patients with OFG. However, overall, a dietary approach to the management of OFG is an effective treatment strategy and may avoid the use of corticosteroids and immunosuppression in many cases. Topical The use of topical preparations is perhaps of particular benefit in those with mild disease and without evidence of gut involvement. Remission of oral symptoms has been reported to be as high as 58% in a small cohort of patients using topical steroids [15]. Intra-lesional injections using delayed-release triamcinolone may be of benefit for lip swelling, and disease-free periods of up to 19 months have been reported [38]. Response is likely to be less optimal in those who already have histological evidence of fibrotic lip disease. Overall, the long-term use of steroids should be discouraged as the improvement is, at best, short lived and not without the risk of unacceptable side effects. Topical mesalazine (Topasa™, Tillotts Pharma AG, Basel, Switzerland) and tacrolimus have also been used with some success for both lip swelling and intra-oral disease [39–41]. Topical steroid mouthwashes, such as betamethasone (Betnesol®, GSK, Middlesex, UK), may be useful for intraoral disease but the response is usually short-lived. If pain is a particular feature, adjunctive antiseptic/analgesic mouthwashes can be helpful. limited use due to adverse effects intra-orally. Some institutions regularly use thalidomide, at doses ranging from 50–300 mg, with anecdotal success, but toxicity and restrictions limit its use [42,43]. Infliximab has demonstrated some success in severe cases where conventional immunosuppressive agents have failed [44,45]. The authors’ own experience suggests that response is greater for intraoral disease, supporting the concept of this pattern being more reflective of gut Crohn’s disease. Surgery Lip reduction procedures should be considered in severe cases with chronically enlarged, deformed fibrotic lip swelling [46]. These patients may frequently suffer unrecognized psychosocial morbidity and social isolation. Moreover, it may interfere with the ability to eat and speak. Any residual active inflammation should be excluded prior to recommending surgery and the best outcome is likely to result from a carefully planned first surgical procedure. Conclusion OFG is a rare, chronic inflammatory disease with links to Crohn’s disease, but which mainly exists as a separate entity. The presence of an associated intestinal granulomatosis should be interpreted with caution as the majority of cases do not appear to develop gut Crohn’s disease. Dietary exclusion using a cinnamon- and benzoate-free diet is effective in the majority of cases, but failing this, an immunosuppressant regimen mirroring that for the treatment of Crohn’s disease is used. Experience outside specialist centers is limited, and the best outcomes are likely to be achieved using a multi-disciplinary approach. Disclosures The authors have no relevant financial interests to disclose. Systemic References The systemic therapeutic regimen employed in OFG closely resembles that used in Crohn’s disease and includes corticosteroids, thiopurines, methotrexate, and biological therapy with infliximab. In general, systemic therapy should be considered in patients who have concomitant gut involvement and/or those who have failed dietary or topical therapy. Systemic corticosteroids are effective for more severe disease and in acute exacerbations. A dose-tapering regimen, similar to that in Crohn’s disease, is used. Steroid dependence or refractory disease should prompt the use of steroid-sparing agents, with azathioprine and 6-mercaptopurine the first choice, as in Crohn’s disease. Those intolerant to or failing on thiopurine therapy should receive methotrexate, but experience suggests a more 1. Wiesenfeld D, Ferguson MM, Mitchell DN et al. Oro-facial granulomatosis – a clinical and pathological analysis. Q J Med 1985;54:101–13. 2. James DG. Mimics of sarcoidosis. Oro-facial granulomatosis (Melkersson-Rosenthal syndrome). Sarcoidosis 1991;8:84. 3. Apaydin R, Bilen N, Bayramgurler D et al. Detection of Mycobacterium tuberculosis DNA in a patient with Melkersson-Rosenthal syndrome using polymerase chain reaction. Br J Dermatol 2000;142:1251–2. 4. van der Waal RI, Schulten EA, van der Meij EH et al. Cheilitis granulomatosa: overview of 13 patients with long-term follow-up – results of management. Int J Dermatol 2002;41:225–9. 5. Challacombe SJ. Oro-facial granulomatosis and oral Crohns disease: are they specific diseases and do they predict systemic Crohns disease? Oral Dis 1997;3:127–9. 6. Dupuy A, Cosnes J, Revuz J et al. Oral Crohn disease: clinical characteristics and long-term follow-up of 9 cases. Arch Dermatol 1999;135:439–42. 7. Basu MK, Asquith P. Oral manifestations of inflammatory bowel disease. Clin Gastroenterol 1980;9:307–21. 8. Bogenrieder T, Rogler G, Vogt T et al. Orofacial granulomatosis as the initial presentation of Crohn’s disease in an adolescent. Dermatology 2003;206:273–8. 9. Kano Y, Shiohara T, Yagita A et al. Association between cheilitis granulomatosa and Crohn’s disease. J Am Acad Dermatol 1993;28:801. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 21 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 22 CARLO NUNES ET AL. 10. Talbot T, Jewell L, Schloss E et al. Cheilitis antedating Crohn’s disease: case report and literature update of oral lesions. J Clin Gastroenterol 1984;6:349–54. 11. Girlich C, Bogenrieder T, Palitzsch KD et al. Orofacial granulomatosis as initial manifestation of Crohn’s disease: a report of two cases. Eur J Gastroenterol Hepatol 2002;14:873–6. 30. Freysdottir J, Zhang S, Tilakaratne WM et al. Oral biopsies from patients with orofacial granulomatosis with histology resembling Crohn’s disease have a prominent Th1 environment. Inflamm Bowel Dis 2007;13:439–45. 31. Nunes C, Spencer J, Escudier M et al. B-cell infiltrates in orofacial granulomatosis. Gut 2007;56:A117. 12. Sawczenko A, Sandhu BK. Presenting features of inflammatory bowel disease in Great Britain and Ireland. Arch Dis Child 2003;88:995–1000. 32. Nunes C, Escudier M, Shirlaw P et al. Orofacial granulomatosis: a tertiary referral centre experience. Gut 2006;55:A16. 13. Worsaae N, Christensen KC, Schiodt M et al. Melkersson-Rosenthal syndrome and cheilitis granulomatosa. A clinicopathological study of thirty-three patients with special reference to their oral lesions. Oral Surg Oral Med Oral Pathol 1982;54:404–13. 33. Sanderson J, Nunes C, Escudier M et al. Oro-facial granulomatosis: Crohn’s disease or a new inflammatory bowel disease? Inflamm Bowel Dis 2005;11:840–6. 14. Hornstein OP. Melkersson-Rosenthal syndrome. A neuro-muco-cutaneous disease of complex origin. Curr Probl Dermatol 1973;5:117–56. 34. Nunes C, Shirlaw P, Mistry M et al. Serum and salivary IgA responses to a 200kd Saccharomyces cerevisiae antigen in orofacial granulomatosis and Crohn’s disease. Gut 2007;56:A117. 15. Plauth M, Jenss H, Meyle J. Oral manifestations of Crohn’s disease. An analysis of 79 cases. J Clin Gastroenterol 1991;13:29–37. 16. Mignogna MD, Fedele S, Lo RL et al. Orofacial granulomatosis with gingival onset. J Clin Periodontol 2001;28:692–6. 17. White A, Nunes C, Escudier M et al. Improvement in orofacial granulomatosis on a cinnamon- and benzoate-free diet. Inflamm Bowel Dis 2006;12:508–14. 18. Greene RM, Rogers RS 3rd. Melkersson-Rosenthal syndrome: a review of 36 patients. J Am Acad Dermatol 1989;21:1263–70. 35. Halme L, Meurman JH, Laine P et al. Oral findings in patients with active or inactive Crohn’s disease. Oral Surg Oral Med Oral Pathol 1993;76:175–81. 36. Gaya DR, Aitken S, Fennell J et al. Anti-TNF-{alpha} therapy for orofacial granulomatosis: proceed with caution. Gut 2006;55:1524–5. 37. Nunes C, Lomer MC, Escudier M et al. The dietary management of orofacial granulomatosis. Gut 2007;56:A117. 19. Rogers RS 3rd. Melkersson-Rosenthal syndrome and orofacial granulomatosis. Dermatol Clin 1996;14:371–9. 38. Mignogna MD, Fedele S, Lo RL et al. Effectiveness of small-volume, intralesional, delayedrelease triamcinolone injections in orofacial granulomatosis: a pilot study. J Am Acad Dermatol 2004;51:265–8. 20. James J, Ferguson MM. Orofacial granulomatosis presenting clinically as tuberculosis of cervical lymph nodes. Br Dent J 1986;161:17–9. 39. Casson DH, Eltumi M, Tomlin S et al. Topical tacrolimus may be effective in the treatment of oral and perineal Crohn’s disease. Gut 2000;47:436–40. 21. James J, Patton DW, Lewis CJ et al. Oro-facial granulomatosis and clinical atopy. J Oral Med 1986;41:29–30. 40. Hodgson T, Hegarty A, Porter S. Topical tacrolimus and Crohn disease. J Pediatr Gastroenterol Nutr 2001;33:633. 22. Patton DW, Ferguson MM, Forsyth A et al. Oro-facial granulomatosis: a possible allergic basis. Br J Oral Maxillofac Surg 1985;23:235–42. 41. Otake K, Uchida K, Inoue M et al. Successful treatment with topical 5-aminosalicylic acid ointment and spray of refractory oral and pharyngeal ulcerations in a child with Crohn disease. J Pediatr Gastroenterol Nutr 2007;44:378–81. 23. Armstrong DK, Burrows D. Orofacial granulomatosis. Int J Dermatol 1995;34:830–3. 24. Haworth RJ, MacFadyen EE, Ferguson MM. Food intolerance in patients with oro-facial granulomatosis. Hum Nutr Appl Nutr 1986;40:447–56. 25. Armstrong DK, Biagioni P, Lamey PJ et al. Contact hypersensitivity in patients with orofacial granulomatosis. Am J Contact Dermat 1997;8:35–8. 26. Wray D, Rees SR, Gibson J et al. The role of allergy in oral mucosal diseases. QJM 2000;93:507–11. 27. Field EA, Tyldesley WR. Oral Crohn’s disease revisited – a 10-year-review. Br J Oral Maxillofac Surg 1989;27:114–23. 28. Lim SH, Stephens P, Cao QX et al. Molecular analysis of T cell receptor beta variability in a patient with orofacial granulomatosis. Gut 1997;40:683–6. 29. Gibson J, Wray D. Human leucocyte antigen typing in orofacial granulomatosis. Br J Dermatol 2000;143:1119–21. 22 42. Hegarty A, Hodgson T, Porter S. Thalidomide for the treatment of recalcitrant oral Crohn’s disease and orofacial granulomatosis. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2003;95:576–85. 43. Thomas P, Walchner M, Ghoreschi K et al. Successful treatment of granulomatous cheilitis with thalidomide. Arch Dermatol 2003;139:136–8. 44. Peitsch WK, Kemmler N, Goerdt S et al. Infliximab: a novel treatment option for refractory orofacial granulomatosis. Acta Derm Venereol 2007;87:265–6. 45. Mahadevan U, Sandborn WJ. Infliximab for the treatment of orofacial Crohn’s disease. Inflamm Bowel Dis 2001;7:38–42. 46. Kruse-Losler B, Presser D, Metze D et al. Surgical treatment of persistent macrocheilia in patients with Melkersson-Rosenthal syndrome and cheilitis granulomatosa. Arch Dermatol 2005;141:1085–91. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 23 CLINICAL REVIEWS Commentary and Analysis on Recent Key Papers Clinical reviews were prepared by Ian Arnott, MD, Federico Balzola, MD, and Charles Bernstein, MD CLINICAL OBSERVATIONS Systematic review: the potential influence of mesalazine formulation on maintenance of remission in Crohn’s disease Steinhart AH, Forbes A, Mills EC et al. Aliment Pharmacol Ther 2007;25:1389–99. The current authors performed a systematic analysis of studies of pH 6- and pH 7-dependent, and controlledrelease mesalazine formulations in the maintenance of surgically or medically induced remission of Crohn’s disease. The risk of disease relapse was found to be significantly reduced in those treated with pH 7-dependent mesalazine compared with placebo, whereas the pH 6-dependent and controlled-release formulations showed no significant reduction in the risk of relapse. Large-scale, randomized controlled trials of pH 7-dependent mesalazine in maintenance of remission are warranted. Several studies and meta-analyses have suggested that mesalazine is effective for the maintenance of surgically induced remission in Crohn’s disease, while the evidence is less clear regarding its use in maintaining medically induced remission. Thus, current guidelines recommend mesalazine, as a class, for the maintenance of disease remission postsurgery. However, there are a number of formulations of mesalazine that allow timed delivery of the active drug to specific regions of the gut. The current authors undertook a Medline/Embase literature review from January 1986 to January 2006 to identify studies of mesalazine in maintenance of medically or surgically induced remission of Crohn’s disease, with the aim of comparing effectiveness of formulations that release the drug at >pH 7 (Asacol®; Procter & Gamble Pharmaceuticals UK Ltd, Egham, UK), >pH 6 (Salofalk®; Dr Falk Pharma UK Ltd, Bourne End, UK; Mesasal®; GlaxoSmithKline, Brentford, UK), and a controlled-release form (Pentasa®; Ferring Pharmaceuticals Ltd, Langley, UK). A total of 13 randomized controlled trials were included in the analysis. Outcome measures included the pooled odds ratio (OR) of maintenance of remission, the number needed to treat (NNT) to prevent one relapse, and the percentage overall therapeutic benefit. Analysis of maintenance of surgically induced remission showed that the risk of relapse was significantly reduced in patients who were treated with pH 7-dependent mesalamine compared with placebo (OR 0.28, 95% confidence interval [CI] 0.12–0.65; p=0.0032), while there was no significant improvement in those treated with pH 6dependent formulation (OR 0.65, 95% CI 0.34–1.24; p=0.1954), or those who received controlled-release mesalazine (OR 0.72, 95% CI 0.47–1.08; p=0.1109). Similarly, treatment with pH 7-dependent mesalamine significantly reduced the risk of relapse in patients with medically induced remission (OR 0.38, 95% CI 0.17–0.85; p=0.0113), while controlled-release and pH 6-dependent formulations again demonstrated no improvement over placebo (p=0.0802 and p=0.3176, respectively). The NNT to prevent relapse of surgically or medically induced remission of Crohn’s disease were 4 and 5, respectively, for pH 7-dependent mesalamine; 11 and 23, respectively, for pH 6-dependent mesalamine; and 15 and 16, respectively, for the controlled-release formulation. Again, this demonstrated greater therapeutic advantage of the pH 7-dependent formulation, although the authors caution that the placebo relapse rates across the studies were heterogeneous. The calculated therapeutic benefit (absolute risk reduction) was also highest for pH 7dependent mesalazine (surgical 30.6%, medical 22.8%), compared with pH 6-dependent mesalazine (9.8% and 4.4%, respectively) and controlled-release mesalazine (6.9% and 6.4%, respectively). The authors discuss limitations of this type of metaanalysis, such as publication or selection bias, and also the interpretation of data when outcomes are based on single studies, as is the case with the maintenance of remission with pH 7-dependent mesalazine. However, they highlight that data are consistent for both medically and surgically induced remission in their analysis. The authors also describe INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 23 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 24 CLINICAL REVIEWS the differences between their analysis and the meta-analysis of Akobeng and Gardener [1], which included studies of azo-bonded mesalazine. Importantly, they discuss the potential suitability of pH- and azo-bonded formulations for subgroups of patients with specific disease location. Lastly, they encourage further large-scale studies of the efficacy of mesalazine formulations in CD, in order to confirm their current findings. 1. Akobeng AK, Gardener E. Oral 5-aminosalicylic acid for maintenance of medically-induced remission in Crohn’s Disease. Cochrane Database Syst Rev 2005;(1):CD003715. Address for reprints: SPL Travis, Gastroenterology Unit, John Radcliffe Hospital, Oxford, OX3 9DU, UK. Email: [email protected] Abnormal hepatic biochemistries in patients with inflammatory bowel disease Mendes FD, Levy C, Enders FB et al. Am J Gastroenterol 2007;102:344–50. The prevalence of abnormal hepatic biochemistries and chronic liver disease in patients with IBD were examined in this study. The authors found that abnormal hepatic biochemistries were present in nearly 30% of patients, but that these were unrelated to IBD activity. While primary sclerosing cholangitis (PSC) is the most widely discussed and investigated hepatobiliary abnormality in patients with IBD, it is likely not to be the most common. Although it may be the most serious chronic liver disease in IBD patients, autopsy series have also reported high rates of fatty liver in IBD patients. Patients presenting at the Mayo Clinic (Rochester, MN, USA) with IBD in 2000 were included in this study. Hepatic biochemistry analyses included blood testing for aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase. Of 621 IBD patients, 544 had hepatic biochemistry assessed; of those, 29% had abnormal test results. Chronic liver disease was diagnosed in 5.8% of patients, including 4.6% with PSC. There was no significant difference in the likelihood of having abnormal hepatic biochemistry test outcomes, whether patients had active disease or disease in remission (27% vs. 36%, respectively; p=0.06). Patients with Crohn’s disease were as likely to have abnormal hepatic biochemistries as patients with ulcerative colitis; however, they were less likely to have a diagnosis of liver disease (2% vs. 9%; p=0.0002). Patients with abnormal hepatic biochemistries were less likely to be using 5-aminosalicylate drugs (35% vs. 51%; p<0.001) and were also more likely to be dead at follow-up (proportion alive at last follow-up: 90.4% vs. 98.5%; p<0.0001, risk ratio 4.8). Of IBD patients with chronic liver disease, fewer had active disease (61% vs. 80%; p=0.006), and excessive 24 alcohol use was more common (6% vs. 1.3%; p=0.04), in comparison with IBD patients who did not have chronic liver disease. One drawback of this study was that only 75% of subjects had at least one follow-up, some of which may simply have been by telephone with further laboratory investigations not being performed. Only 226 (42%) patients had subsequent hepatic biochemistry testing. Furthermore, there was a paucity of imaging studies reported and this might have contributed to the minimal rates of fatty liver reported in this study. The most important take-home message was that hepatic biochemistry abnormalities occurred at some time in nearly 30% of patients and were unrelated to disease activity. Address for reprints: KD Lindor, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA. Chromoscopy-guided endomicroscopy increases the diagnostic yield of intraepithelial neoplasia in ulcerative colitis Kiesslich R, Goetz M, Lammersdorf K et al. Gastroenterology 2007;132:874–82. Patients with ulcerative colitis (UC) have an increased risk of developing colorectal cancer in the long term. These authors evaluated the use of combined chromoendoscopy and endomicroscopy in neoplasia surveillance in patients with UC. They concluded that chromoendoscopy-guided endomicroscopy may lead to significant improvements in the management of neoplasia surveillance in UC. The authors of this study evaluated the utility of combined chromoendoscopy and endomicroscopy in dysplasia surveillance in ulcerative colitis (UC). To facilitate this, a miniature confocal microscope was integrated into the distal tip of a conventional colonoscope and fluorescein dye was injected intravenously. This enabled histological evaluation of the mucosal layer during colonoscopy. Owing to the large surface area of the colon it would not be practical to use endomicroscopy in the entire colon of a patient with UC as this would be excessively time-consuming. For this reason, the investigators combined the endomicroscopy with chromoendoscopy such that the endomicroscopy would only be applied to lesions of interest that were identified with dye spraying. All lesions identified by endomicroscopy were digitally stored and evaluated at a later time by an investigator who was blinded to the recordings of the endoscopist. Images were graded as being good, average, or poor. For conventional colonoscopy, four biopsies were taken every 10 cm; biopsies of targeted lesions were also taken. A total of 161 subjects with INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 25 CLINICAL OBSERVATIONS UC in clinical remission were randomized in a 1:1 ratio to undergo either conventional colonoscopy with multiple surveillance biopsies, or chromoendoscopy with endomicroscopy. Eight patients were excluded due to insufficient bowel preparation that precluded satisfactory examination. Notably, 62 (39%) subjects had only histological proctitis, a condition not thought to be predispose to neoplasia and hence not included in dysplasia surveillance programs. In the conventional colonoscopy group (n=73), four were found to have dysplasia. In the endomicroscopy group (n=80), 19 dysplastic lesions were identified in 11 subjects. Fourteen of these 19 were not identified by conventional endoscopy. In the endomicroscopy group, 16 flat dysplasias were identified compared with only two in the conventional endoscopy group (p=0.002). In this latter group, 4.75-fold more neoplasia was found, despite utilizing 50% fewer biopsy specimens (there were an average of 21.2 biopsies per case in the endomicroscopy group compared with 42.2 biopsies per case in the conventional colonoscopy group). A total of 5580 confocal endomicroscopic images from 134 circumscribed lesions were compared with the histology results. Endomicroscopy was found to predict neoplasia with a sensitivity of 94.7%,specificity of 98.3%, and accuracy of 97.4%. Endomicroscopically normal tissue revealed, with 99% accuracy, non-neoplastic tissue by histological analysis. This could also reduce the need for biopsies to simply stage the extent of histological inflammation. In summary, the authors report that chromoendoscopy with endomicroscopy could increase diagnostic yield while reducing biopsy burden and enhancing neoplasia detection in the setting of UC. Address for reprints: R Kiesslich, I. Medical Clinic, University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany. Email: [email protected] Clinical usefulness of EUS for active ulcerative colitis Yoshizawa S, Kobayashi K, Katsumata T et al. Gastrointest Endosc 2007;65:253–60. The authors of this study assessed the utility of endoscopic ultrasound for predicting the response to medical treatment and the need for surgery in ulcerative colitis patients. This study assessed whether endoscopic ultrasound (EUS) could identify the depth of intestinal inflammation, predicting the response to medical treatment, and determining the necessity for surgery in active ulcerative colitis (UC). Initially, EUS was tested in vitro in 13 surgically resected specimens. In vivo, the severest lesions on colonoscopic examination were evaluated by EUS in 42 patients with active UC with the aim of potentially identifying ultrasound characteristics that predict the need for surgery. In vitro, the spread of UC identified by EUS was consistent with histological findings in 45/50 (90%) cases. The concordance for transmural inflammation between EUS and histology extending to the submucosa was 95%, to the muscularis propria was 83%, and to the subserosa was 100%. In the in vivo studies, inflammation was evaluated to extend at least as deep as the muscularis propria on EUS in 10 of the 15 cases who failed medical therapy and who ultimately underwent surgery. In five of the 27 subjects who achieved medical remission it extended to at least muscularis propria (p=0.002 for predicting surgery, based on the depth of inflammation). The authors argue that EUS in active UC can be used to help predict those likely to require surgical therapy and, therefore, might help reduce the hospitalization time, perhaps by encouraging either earlier surgery or more aggressive medical therapy. Address for reprints: S Yoshizawa, Department of Gastroenterology, Kitasato University East Hospital, 2-1-1 Asamizodai, Sagamihara-city, Kanagawa 228-8520, Japan. Incidence of minor complications and time lost from normal activities after screening or surveillance colonoscopy Ko CW, Riffle S, Shapiro JA et al. Gastrointest Endosc 2007;65:648–56. The authors of this article investigated the incidence of minor complications in patients undergoing surveillance colonoscopy during screening for colorectal cancer. Approximately one-third of patients reported minor adverse events during the first week post-procedure, and 5.9% reported complications between day 7 and day 30 after colonoscopy. The duration of the procedure was the only factor identified to be correlated with the risk of minor adverse events. Although the risk of bowel perforation and hemorrhage during colonoscopy represent the major complications of the procedure, which lead to surgery or death, the reasons for post-procedural minor complications (such as fever, light gastrointestinal bleeding, self-limited abdominal pain, shoulder pain, bloating, nausea, rash, diarrhea, or constipation) remain poorly understood and have not yet been quantified. The aims of this prospective study was to determine whether there were short-term (within 1 week) or long-term (within 1 month) procedure-related minor complications among asymptomatic patients undergoing complete colonoscopy for colorectal cancer screening or surveillance. From a total of 502 patients, 470 completed 7- and 30-day telephone interviews. The colonoscopic examination INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 25 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 26 CLINICAL REVIEWS was concluded to the cecum in 99% of subjects. In 36% of patients, a gastroenterology trainee participated in the procedure. Minor complications were reported in 33.8% during the first week (25.0% bloating, 10.5% abdominal pain, 6.3% diarrhea, 4.0% nausea, 3.8% light gastrointestinal bleeding, 1.9% shoulder pain, 1.9% constipation, and 0.8% rash), and in 5.9% during the first month (1.6% nausea, 1.6% light gastrointestinal bleeding, 1.4% constipation, 1.2% diarrhea, 1.2% abdominal pain, 0.8% fever, 0.6% bloating, 0.2% shoulder pain, and 0.2% rash). Surprisingly, the presence of minor complications was higher in women than in men, but was not influenced by age, procedure indications, or colonoscopy findings. The duration of the endoscopic procedure was the only factor directly correlated with the development of mild adverse events. This could be related to the large quantity of air insufflations or increased external abdominal pressure required to perform a difficult examination, for example as a result of a complex colonic anatomy, poor colon preparation, or presence of polyps or other abnormalities requiring biopsies or polypectomy. In conclusion, the results of this simple study on the perception of an invasive examination from the patient point of view show that screening or surveillance colonoscopy is safe and well tolerated, although one-third of patients report minor side effects. Moreover, a prior, extensive knowledge of the possible presence of immediate or delayed minor events should lead to a better acceptance and satisfaction by patients who may undergo a recurring procedure as routine surveillance screening. The colon preparation (and not the colonoscopy itself) remains the greatest concern, in terms of minor adverse events. Address for reprints: CW Ko, Division of Gastroenterology, Box 356424, University of Washington, Seattle, WA 98195, USA. Respiratory symptoms in patients with inflammatory bowel disease and the impact of dietary salicylates Sivagnanam P, Koutsoumpas A, Forbes A et al. Dig Liver Dis 2007;39:232–9. Respiratory complaints are a common extraintestinal manifestation of IBD. In this study of a cohort of 69 ulcerative colitis (UC) and 73 Crohn’s disease patients, respiratory symptoms were reported in >60% of subjects. Asthma or possible undiagnosed asthma was the most prevalent respiratory diagnosis in both UC and Crohn’s disease (43%). Thus, it is suggested that clinicians closely monitor respiratory symptoms in IBD patients. 26 Respiratory symptoms are a frequent extraintestinal manifestation of IBD. They have been described in >50% of IBD patients, although they are frequently underestimated by clinicians. This may be because their onset is reported before the manifestation of the intestinal disease (and thus may not be correlated with the presymptomatic underlying bowel disease), or because their presence can be subclinical and frequently not described by patients. The increased prevalence of both respiratory conditions and IBD reported in the past half century shows a fascinatingly similar epidemiological trend. The respiratory involvement in IBD (which includes pleuritis, airway disease, interstitial lung disease, necrobiotic nodules, pulmonary eosinophilia, thromboembolic disease, vasculitis, or granulomatous lung disease) has been postulated to be related to environmental trigger factors such as smoking and pollution, or a leaky gut in subjects with a genetically predisposed background. This altered permeability, together with an altered gut flora, can increase the passage of food antigens or bacteria into the venous blood circulation, causing direct damage to the lungs, which act as the primary filter. In addition, 5-aminosalicylates, a group of drugs used in the treatment of IBD, have been implicated in interstitial lung disease and eosinophilic pneumonia [1,2]. Salicylates are also present in certain natural food (fruits and vegetables). A salicylateexclusion diet has been suggested to be therapeutic in IBD patients with concurrent respiratory symptoms. For these reasons, the authors of this study aimed to quantify the prevalence of respiratory symptoms, pulmonary disease, and salicylate intake in a cohort of 69 ulcerative colitis (UC) and 73 Crohn’s disease patients referred to their hospital. Respiratory symptoms were reported in the 62% and 65% of Crohn’s disease and UC patients, respectively. Asthma or possible undiagnosed asthma was the prevalent respiratory diagnosis in both UC and Crohn’s disease (43%), whereas chronic bronchitis, emphysema, bronchiectasis, asbestosis, or nasal symptoms were equally distributed in 12 of the overall 142 patients. Eleven of 14 Crohn’s disease patients (79%) with extraintestinal manifestations reported defined respiratory conditions, as did three of five UC patients (60%). This study underlines the linkage between respiratory phenomena and intestinal/extraintestinal features of IBD. The weekly dietary salicylate intake was modest, ranging from 1 mg to 120 mg in both Crohn’s disease and UC patients, and did not correlate with the respiratory status. Interestingly, a significant inverse correlation between dietary salicylate intake and disease activity was found in UC but not in Crohn’s disease, where the low number of patients indicated only a slight numerical trend. In conclusion, this study confirmed the frequent association between respiratory symptoms and extraintestinal INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 27 CLINICAL OBSERVATIONS manifestations of IBD (the lung and the gastrointestinal system originate from the primitive gut); therefore, it is suggested that clinicians improve on the investigation of the respiratory status of such patients. No concomitant respiratory symptom or disease activity was reduced with a low intake of 5-ASAcontaining food, thus the salicylate-exclusion diet should not be considered as a therapeutic tool. However, an improved knowledge of the content of 5-ASA in food, together with its active absorbable action is considered important, not only to protect from potential allergies in these or other frequently atopic patients, but also to introduce specific diets with the aim of preventing IBD, colorectal cancer, or cardiovascular disease in the normal or at-risk population. 1. Moseley RH, Barwick KW, Dobuler Ket al. Sulfasalazine induced pulmonary disease. Dig Dis Sci 1985;30:901–4. 2. Tanigawa K, Sugiyama K, Matsuyama H et al. Mesalazine-induced eosinophilic pneumonia. Respiration 1999;66:69–72. Address for reprints: A Forbes, University College Hospital, 235 Euston Road, London NW1 2BU, UK. Email: [email protected] Effect of allopurinol on clinical outcomes in inflammatory bowel disease nonresponders to azathioprine or 6-mercaptopurine Sparrow MP, Hande SA, Friedman S et al. Clin Gastroenterol Hepatol 2007;5:209–14. The 6-thioguanine (6-TGN) metabolite of the prodrugs azathioprine (AZA) and 6-mercaptopurine (6-MP) appears to be the active metabolite responsible for efficacy in maintaining disease remission in IBD. Levels of another metabolite, 6-methylmercaptopurine (6-MMP), correlate with those of hepatotoxic enzymes. Allopurinol has previously been shown to optimize 6-TGN production and minimize that of 6-MMP. In this study, IBD patients classed as AZA/6-MP nonresponders who received allopurinol were demonstrated to have a significant increase in mean 6-TGN levels, a decrease in 6-MMP levels, and significant reductions of disease activity indices scores. Azathioprine (AZA) and 6-mercaptopurine (6-MP) are both inactive prodrugs that are metabolized via three main enzymatic pathways to produce the nucleotide metabolites: 6-thioguanine (6-TGN), 6-methylmercaptopurine (6-MMP), and 6-thiouracil (6-TU). 6-TGN appears to be the active metabolite responsible for therapeutic efficacy in IBD. In Crohn’s disease and in ulcerative colitis (UC), there is evidence to support the use of AZA or 6-MP for maintaining remission and as steroid-sparing agents; moreover, in Crohn’s disease patients, these drugs have been demonstrated to be effective for treatment of perianal fistulas. It has previously been assumed that the immunosuppressive actions of thiopurines are achieved by incorporation of the metabolite 6-TGN into lymphocytic DNA, thereby inhibiting cellular proliferation [1]. However, an alternative mechanism of action has recently been proposed [2]. This suggests that the 6-TGN triphosphate (6-TGTP) – a metabolite of 6-TGN – actively stimulates apoptosis of lamina propria T lymphocytes. The 6-MMP metabolite of the 6-MP does not demonstrate therapeutic efficacy, but its levels correlate with the risk of hepatotoxicity in the form of elevation of hepatic transaminase enzyme (aspartate transaminase and alanine transaminase) levels. For these reasons, a desirable metabolic profile of thiopurine drug metabolism would be one in which 6-TGN levels were maximized, and 6-MMP production was minimal. It has been shown that 6-TGN concentrations are inversely correlated with functional activity of the thiopurine methyltransferase (TPMT) enzyme, whereas elevations in 6-MMP are associated with high functional TPMT activity. The authors of this article recently demonstrated that in IBD patients displaying a high TPMT activity metabolite profile (6-MMP>6-TGN) the addition of low doses of allopurinol, a xanthine oxidase inhibitor, can safely and effectively optimize 6-TGN production and minimize that of 6-MMP [3]. In the present study, the clinical efficacy of the addition of allopurinol in AZA/6-MP nonresponders (in whom the metabolism of these drugs is preferentially directed towards 6-MMP) was investigated. Twenty AZA/6MP nonresponders with high 6-MMP metabolite levels were started on allopurinol 100 mg/day and the dose of AZA/6MP reduced to 25–50% of the original dose. A significant increase in mean 6-TGN levels, a decrease in mean 6-MMP levels, and statistically significant reductions of Harvey Bradshaw Index scores in Crohn’s disease patients and of Mayo index scores in UC patients, were observed. Furthermore, addition of allopurinol enabled a reduction in mean daily dosage of prednisone, from 18±4 mg to 2±1 mg, and led to normalization of transaminase levels in all cases. As 6-TGN levels are responsible for myelotoxicity, the addition of allopurinol, enhancing 6-TGN levels, could cause the development of leukopenia, which could be resolved with thiopurine dose reduction. This side effect could also be easily prevented by a close monitoring of white blood cell counts. 1. Lepage GA. Basic biochemical effects and mechanism of action of 6-thioguanine. Cancer Res 1963;23:1202–6. 2. Tiede I, Fritz G, Strand S et al. CD28-dependent Rac1 activation is the molecular target of azathioprine in primary human CD4+ T lymphocytes. J Clin Invest 2003;111:1133–45. 3. Sparrow MP, Hande SA, Friedman S et al. Allopurinol safely and effectively optimizes tioguanine metabolites in inflammatory bowel disease patients not responding to azathioprine and mercaptopurine. Aliment Pharmacol Ther 2005;22:441–6. Address for reprints: M Sparrow, Section of Gastroenterology and Nutrition, University of Chicago Medical Center, Chicago, IL, USA. Email: [email protected] INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 27 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 28 CLINICAL REVIEWS Narrow-band imaging compared with conventional colonoscopy for the detection of dysplasia in patients with longstanding ulcerative colitis Dekker E, van den Broek FJ, Reitsma JB et al. Endoscopy 2007;39:216–21. These investigators compared the use of standard colonoscopy with a narrow-band imaging (NBI) technique, which enables better visualization of the intestinal mucosa, for detection of dysplasia in patients with longstanding ulcerative colitis. No difference in sensitivity was observed between these techniques. Furthermore, targeted biopsies using both NBI and standard colonoscopy failed to detect one-third of patients with cancer, as identified during random biopsies using standard colonoscopy. Longstanding ulcerative colitis (UC) has been correlated with a higher risk of colorectal cancer, compared with that observed in the general population, with a cumulative incidence estimated by Rutter et al. of 2.5% at 20 years, 8% at 30 years, and 11% at 40 years [1]. Colonoscopic surveillance is recommended but is difficult for several reasons, including issues of patient compliance, the risk of missing lesions, and the presence of macroscopically invisible lesions. Narrow-band imaging (NBI) is a novel endoscopic procedure that facilitates an improved investigation of mucosal characteristics. The application of an optical filter to the light used for illumination enhances the appearance of the surface of the intestinal mucosa; specifically, the blue excitation light view has a minimal penetration depth in the mucosa, which better highlights the intestinal surface and vascular pattern. Moreover, NBI has been shown to be more practical than chromoendoscopy, a labor-intensive technique that involves staining the whole colon with dye spraying, which inexorably prolongs endoscopic examination, thus reducing its use in clinical practice. The authors prospectively investigated 42 patients with longstanding UC, with both standard white-light colonoscopy and NBI randomly performed by different endoscopists (who were blinded to the results of the first procedure) at an interval of 3 weeks. Targeted biopsies on suspicious lesions were collected using both procedures, whereas random biopsies to exclude dysplasia were taken with standard colonoscopy only. Targeted biopsies using NBI identified 52 suspicious lesions in 17 patients, while standard colonoscopy detected 28 suspicious lesions in 13 of the same patients. Histological evaluation of the biopsies identified a cancer in 11 patients; four of these cancers were detected using both NBI and standard colonoscopy, four with NBI only, and three with standard colonoscopy alone. As a comparison with the targeted biopsies, 1522 random biopsies using standard colonoscopy revealed neoplasia in six patients. In one additional patient, the 28 random biopsy identified severe dysplasia in a suspicious area, which has not been detected by targeted biopsy. In conclusion, this first, prospective, randomized study comparing NBI with standard colonoscopy did not identify a difference in sensitivity between the two techniques. Notwithstanding that NBI detected more false-positive lesions, probably because of the use of a first-generation prototype (the new NBI system is likely to have improved brightness and resolution), targeted biopsies with both procedures failed to detect approximately one-third of patients with cancer. For this reason, the importance of random biopsies, even in presence of more sensitive, novel techniques, was confirmed. 1. Rutter MD, Saunders BP, Wilkinson KH et al. Thirty-year analysis of a colonoscopic surveillance program for neoplasia in ulcerative colitis. Gastroenterology 2006;130:1030–8. Address for reprints: E Dekker, Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands. Email: [email protected] Fecal calprotectin in first-degree relatives of patients with ulcerative colitis Montalto M, Curigliano V, Santoro L et al. Am J Gastroenterol 2007;102:132–6. Fecal calprotectin (FC) has been proposed as a non-invasive marker of disease activity in IBD, with a close correlation between FC levels and disease activity reported in ulcerative colitis (UC). The current authors determined that FC levels were higher in first-degree relatives of UC patients than in healthy control subjects or patients’ spouses. They conclude that some first-degree relatives of patients with UC may have subclinical mucosal inflammation, which suggests a possible genetic component in UC. Calprotectin is a neutrophil-derived protein that is an objective, but non-specific, marker of intestinal inflammation. It is stable in feces and fecal levels of the protein correlate with the histological degree of inflammation as well as with the fecal excretion of 111-indium-labeled leukocytes in IBD. Fecal calprotectin (FC) has been proposed as a non-invasive marker of disease activity in IBD. However, it should be noted that there is closer correlation between FC levels and clinical disease activity indices in ulcerative colitis (UC) than in Crohn’s disease. FC is elevated in first-degree relatives of patients with Crohn’s disease. This elevation is greater that that seen in spouses, giving further support to the genetic predisposition of the disease. In this investigation, Montalto et al. assessed whether FC is elevated in first-degree relatives of patients with UC – a condition in which the genetic contribution is thought to be less than that found in Crohn’s disease. They examined 55 patients with UC, 167 first-degree relatives, 38 spouses, and 150 healthy control subjects. FC was assessed by enzyme- INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 29 GENETICS linked immunosorbent assay and, in patients with UC, disease activity was assessed according to the disease activity index. FC levels were found to be higher in UC patients than in all other groups. However, FC was also significantly higher in first-degree relatives of patients than in healthy controls or spouses (p=0.01), although there was no correction for multiple comparisons. In addition, spouses had higher concentrations than control subjects (p=0.01). These results suggest that some first-degree relatives of patients with UC may have subclinical mucosal inflammation; however, the nature of this inflammation was not defined in this study. These data suggest that genetic, environmental, and gene–environment interactions are all important in the pathogenesis of UC as well as Crohn’s disease; results that raise more questions than answers. Address for reprints: M Montalto, Istituto di Medicina Interna, Università Cattolica del Sacro Cuore, Largo Gemelli, 8-00168 Rome, Italy. Cytomegalovirus is frequently reactivated and disappears without antiviral agents in ulcerative colitis patients Matsuoka K, Iwao Y, Mori T et al. Am J Gastroenterol 2007;102:331–7. In order to determine the incidence of cytomegalovirus (CMV) reactivation in patients with active ulcerative colitis (UC), CMV-antigen and CMV-real-time quantitative polymerase chain reaction (PCR) tests were undertaken by the current investigators. CMV was found to be reactivated in 25 of 48 CMV-immunoglobulin G-positive patients during the study period. However, antigen levels and PCR test values were low and none of the patients showed any evidence of CMV infection on biopsy specimens. In immunocompetent individuals, primary cytomegalovirus (CMV) infections are generally mild; however, they are frequently more serious in the immunocompromised patient. CMV inclusion bodies are often identified, in varying numbers, in patients admitted to hospital with flares of ulcerative colitis (UC). This has led to considerable conjecture and debate regarding the role of the virus in perpetuating the inflammatory activity. It has been proposed that in patients with acute, severe UC, CMV may contribute to worsening of disease, steroid resistance, and even toxic megacolon. The natural history of CMV infection in patients with active UC is not known. Previously, assessing this issue has been problematic due to methodological difficulties detecting the virus in biopsy, stool, and blood specimens. The CMV antigen test and CMV quantitative real-time polymerase chain reaction (PCR) test in blood have emerged as sensitive assays of virus presence and activity. The authors of this report used these two tests to assess CMV at sequential timepoints in patients admitted with UC. A total of 69 patients who were admitted to hospital with moderately or severely active UC were enrolled in the study. Those who were CMV-immunoglobulin G (IgG)- or CMVIgM-positive had CMV antigen and PCR test levels measured every 2 weeks for 8 weeks. CMV-IgG was detected in 48 of the 69 (69.6%) patients. CMV was reactivated in 25 of the 48 (52.1%) seropositive patients during the study period. The CMV antigen and PCR values were low and none of the patients displayed any evidence of CMV infection on biopsy specimens. Gancyclovir was used in two patients but the authors felt that rates of remission and colectomy did not differ between CMV-positive and -negative patients. It is of note that CMV disappeared without therapy in most of the patients who had evidence of reactivation. CMV reactivation was more frequent in patients treated with cyclosporine but not significantly with other treatments. The authors concluded that CMV is frequently reactivated in patients with active UC. In many, it disappears without antiviral therapy; thus, such therapies should not be necessary for most UC patients with CMV reactivation provided that CMV antigen values are low. Although these data offer reassurance to the treating physician for most patients, there are individuals in whom antiviral therapy does lead to clinical improvement and careful consideration should be given to this therapeutic option if viral antigen or PCR test values are high, or if multiple inclusion bodies are seen in biopsy specimens. Address for reprints: T Hibi, Division of Gastroenterology, Department of Internal Medicine, School of Medicine, Keio University, 35 Shinamomachi, Shinjuku, Tokyo 160-8585, Japan. GENETICS Genetic variation in myosin IXB is associated with ulcerative colitis van Bodegraven AA, Curley CR, Hunt KA et al. Gastroenterology 2006;131:1768–74. Genetic variation in the 3' region of the myosin IXB (MYO9B) gene has been reported to be associated with celiac disease. The current authors examined variations of the MYO9B gene in three IBD patient cohorts. An association was observed between five of eight tested MYO9B single nucleotide polymorphisms and IBD (and celiac disease) in all three cohorts. Thus, MYO9B genetic variants appear to predispose to IBD. Many of the recent advances in the genetics of IBD have related to Crohn’s disease, and twin studies would indicate INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 29 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 30 CLINICAL REVIEWS that the genetic contribution to this condition is stronger than that seen with ulcerative colitis (UC). The authors of this study report a genetic variant that is strongly associated with UC. Recently, genetic variation in the 3' region of the myosin IXB (MYO9B) gene was reported to be associated with celiac disease [1]. In a Dutch population with celiac disease, this gene was identified by screening an area of linkage on chromosome 19p13 with 291 single nucleotide polymorphisms (SNPs). The area in 19p13 has also been associated with IBD in two genome wide scans of 2000 and 2003, and has been termed IBD6 [2,3]. Overexpression of rat myosin IXB leads to actin-related morphological changes in epithelial cells. MYO9B is most strongly expressed in human leukocytes but is also detected in intestinal epithelial cells. The mechanism by which this may predispose to celiac disease (or perhaps to IBD) is unknown, but variations in intestinal permeability have been hypothesized. The authors examined variations of the MYO9B gene in a large cohort of IBD patients from three distinct geographical areas. Eight SNPs were typed across the region of interest in 1197 patients with Crohn’s disease, 1520 with UC, and 4440 controls from the UK, Canada, and Italy. These SNPs were selected from those associated with celiac disease and from a tagging approach, to effectively cover the region. Genotyping was performed independently by each institution using a combination of Taqman (Applied Biosystems, Foster City, CA, USA) and Sequenom (Sequenom, San Diego, CA, USA) sequencing techniques. Association was observed between five of the tested MYO9B SNPs and IBD in all three cohorts. From a meta-analysis of the data, the most strongly associated SNP was found to be rs1545620, with an odds ratio of 1.2 (p=1.9×10–6). The alleles demonstrating association with IBD also showed association with celiac disease. The authors concluded that MYO9B genetic variants predispose to IBD. They note that the associated SNP, rs1545620, is a non-synonymous variant leading to an amino acid change (Ala1011Ser) in the third calmodulin-binding IQ domain of MYO9B. It is also of note that this association is considerably stronger with UC, although a weaker association with Crohn’s disease is also observed. These data imply shared causal mechanisms underlying intestinal inflammatory diseases. 1. Monsuur AJ, de Bakker PI, Alizadeh BZ et al. Myosin IXB variant increases the risk of celiac disease and points toward a primary intestinal barrier defect. Nat Genet 2005;37:1341–4. 2. Rioux JD, Silverberg MS, Daly MJ et al. Genomewide search in Canadian families with inflammatory bowel disease reveals two novel susceptibility loci. Am J Hum Genet 2000;66:1863–70. 3. van Heel DA, Dechairo BM, Dawson G et al. The IBD6 Crohn’s disease locus demonstrates complex interactions with CARD15 and IBD5 disease-associated variants. Hum Mol Genet 2003;12:2569–75. Address for reprints: DA van Heel, Centre for Gastroenterology, Institute of Cell and Molecular Science, Barts & The London, Queen Mary’s School of Medicine & Dentistry, Turner Street, London, E1 2AD, UK. Email: [email protected] 30 TUCAN (CARD8) genetic variants and inflammatory bowel disease McGovern DP, Butler H, Ahmad T et al. Gastroenterology 2006;131:1190–6. The current authors examined the role of tumor-upregulated CARD-containing antagonist of caspase nine (TUCAN; CARD8) gene variants in IBD pathogenesis. Analysis of 10 single nucleotide polymorphisms (SNPs) across the gene in control subjects, Crohn’s disease patients, and ulcerative colitis (UC) patients identified one SNP associated with Crohn’s disease. Further analyses are needed to confirm this finding. The discovery of NOD2/CARD15, a landmark in complex disease genetics, has catalyzed a surge in the efforts to find new genetic determinants in IBD. McGovern and colleagues examined the role of tumor-up-regulated CARD-containing antagonist of caspase nine (TUCAN) in the Oxford IBD cohort (Oxford, UK). TUCAN (also know as CARD8/CARDINAL) is expressed in gut epithelium, may be a negative regulator of nuclear factor-κB, and has a regulatory role in apoptosis. The gene encoding TUCAN is located beneath a peak of linkage identified on a genomewide scan performed in 2003, which showed peak logarithm of odds scores in NOD2-negative, IBD5-positive patients [1]. This suggests that TUCAN is an attractive positional and functional candidate gene for IBD. The current authors examined 10 single nucleotide polymorphisms (SNPs) across the area of linkage in 365 control subjects, 372 patients with Crohn’s disease, and 373 patients with ulcerative colitis. They subsequently constructed a predictive model using smoking status and TUCAN, NOD2, IBD5, NOD1, and TNFSF15 genotypes. A significant association between a single SNP in TUCAN and Crohn’s disease was identified. This SNP changes a cysteine residue to a stop codon at codon 10 (rs2043211). The odds ratio (OR) for the association was 1.35 (p=0.0083) with no correction for multiple comparisons. The association was stronger in patients with non-colonic disease (OR 1.52) and in Crohn’s disease patients who were negative for NOD2 variants (OR 1.50). The prognostic model showed an association between Crohn’s disease and an increasing number of positive variables. Depending on the number of variables selected, the model could provide either good sensitivity or specificity, but not both together. The provided data would make it difficult to extrapolate into clinical practice. The authors concluded that there was an association between TUCAN and Crohn’s disease. It is of note that more recent, larger studies have questioned this association [2,3]. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 31 EPIDEMIOLOGY 1. van Heel DA, Dechairo BM, Dawson G et al. The IBD6 Crohn’s disease locus demonstrates complex interactions with CARD15 and IBD5 disease-associated variants. Hum Mol Genet 2003;12:2569–75. 2. Franke A, Rosenstiel P, Balschun T et al.; IBSEN Study Group, Solberg C. No association between the TUCAN (CARD8) Cys10Stop mutation and inflammatory bowel disease in a large retrospective German and a clinically well-characterized Norwegian sample. Gastroenterology 2007;132:2080–1. 3. Fisher SA, Mirza MM, Onnie CM et al. Combined evidence from three large British Association studies rejects TUCAN/CARD8 as an IBD susceptibility gene. Gastroenterology 2007;132:2078–80. Address for reprints: DPB McGovern, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Headington, Oxford, OX3 7BN, UK. Email: [email protected] EPIDEMIOLOGY Meta-analysis: cancer risk of low-grade dysplasia in chronic ulcerative colitis Thomas T, Abrams KA, Robinson RJ et al. Aliment Pharmacol Ther 2007;25:657–68. The aim of the current meta-analysis was to determine the incidence of cancer and the relative risk of developing cancer in patients with ulcerative colitis who had lowgrade dysplasia identified on surveillance. A significant, nine-fold risk of developing cancer after a diagnosis of low-grade dysplasia was derived from this analysis. The authors of this report performed a meta-analysis of 20 studies of dysplasia surveillance in patients with ulcerative colitis (UC). This analysis included 508 lesions identified as having low-grade dysplasia in flat mucosa (n=477) or in a dysplasia-associated lesion or mass (DALM; n=31). Most of the subjects underwent continued surveillance with a mean of 4.3 colonoscopies per patient and an average 18 biopsies per colonoscopy. Some studies did not include the total number of UC patients involved and some did not include the average number of colonoscopies performed on follow-up. There were 73 (14%) advanced lesions (cancer or high-grade dysplasia) detected pre-operatively, which translates into 30 per 1000 patient-years duration and a nine-fold increased risk of developing cancer (odds ratio 9.0, 95% confidence interval 4.0–20.5) and a 12-fold increase in the risk of developing any advanced lesion if low-grade dysplasia is detected during surveillance. Of the 31 cancers detected, 45% were Dukes stages C or D. In an additional 98 cases, colectomy was performed within 6 months of the initial diagnosis of lowgrade dysplasia. In 25 (26%) cases, cancer was found at surgery, suggesting that this is the rate of having coexisting cancer when low-grade dysplasia is found. For flat, low-grade dysplasia only (excluding DALMs), the rate of concurrent cancer was 22% and the progression to cancer was not identified, whereas progression to an advanced lesion was found in 15%. The number needed to colonoscope in order to detect one cancer was eight over an average of 5.2 years. Using a meta-analysis regression, only the number of biopsies taken per colonoscopy had a significant influence on the incidence rates of advanced lesions (p=0.002) whereas the duration of colitis prior to low-grade dysplasia diagnosis, the number of pathologists reviewing histology, and the number of surveillance colonoscopies performed per patient did not influence the cancer incidence obtained. One problem with meta-analyses of this type is the inclusion of studies where modern reviews of past diagnoses of low-grade dysplasia are not undertaken; hence, it is possible that these data include case series in which many so-called low-grade dysplasias may not have been low-grade dysplasia at all. In other words, the nine-fold risk of developing cancer after a diagnosis of low-grade dysplasia derived here may even be an underestimate. In fact, there was a significant trend towards an increased incidence of cancer and advanced lesions from 1987 to the present date (p=0.05), with cancer incidence increasing by 13% per year of study. Address for reprints: R Robinson, Digestive Diseases Centre, Leicester General Hospital, Gwendolen Road, Leicester, LE4 5PW, UK. Email: [email protected] Meta-analysis: mortality in Crohn’s disease Canavan C, Abrams KR, Mayberry JF. Aliment Pharmacol Ther 2007;25:861–70. The aim of the authors of this meta-analysis was to identify the standardized mortality ratio (SMR) for Crohn’s disease. A total of 13 articles identified on the Medline database were included. From these, the estimate for standardized mortality ratio (SMR) was 1.52 (95% confidence interval 1.32–1.74). Thus, patients with Crohn’s disease have an increased mortality risk, compared with the general population. Thirteen studies addressing mortality rates in Crohn’s disease were combined in a meta-analysis. Eight of these showed an increased mortality rate for Crohn’s disease compared with the general population. The pooled estimate for standardized mortality ratio (SMR) was 1.52 (95% confidence interval [CI] 1.32–1.74). Meta-regression analysis showed that the SMR has decreased slightly over the past 30 years (p=0.08). Subanalysis by country showed that a consistently higher SMR for Crohn’s disease was reported from the UK and Scandinavia, compared with other countries. Seven studies were community-based (some of which were population-based), three were hospitalbased, two were from referral centers, and one was from the population-based UK General Practice Research Database. Not surprisingly, hospital-based and referral center-based studies that would likely bias the analysis toward an increased SMR for INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 31 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 32 CLINICAL REVIEWS Crohn’s disease did in fact show a higher SMR (1.73, 95% CI 1.45–2.47). Nonetheless, community-based studies also had a significantly increased SMR (1.48, 95% CI 1.28–1.7). Overall, there was a 2.3% decrease in SMR per median year of diagnosis (p=0.002). There was also a significant difference in SMR for those diagnosed prior to 1970 compared with those diagnosed after 1970, but both eras were associated with a significantly increased SMR over that of the general population. Furthermore, there was no significant difference in terms of year of diagnosis when assessing just the studies where diagnoses were made after 1970. A sensitivity analysis showed that no single study changed the SMR to such an extent that omitting it would change the combined SMR significantly. While no conclusions on life expectancy can be drawn from this study, the data are fairly consistent that patients with Crohn’s disease have an increased mortality risk over the general population. Whether this relates to smoking, disease-specific factors, or treatment-related factors is unknown. Address for reprints: C Canavan, Digestive Diseases Centre, Leicester General Hospital, Gwendolen Road, Leicester, LE5 4PW, UK. Email: [email protected] PATHOGENESIS Crohn’s disease, fatigue, and infliximab: is there a role for cytokines in the pathogenesis of fatigue? Minderhoud IM, Samsom M, Oldenburg B. World J Gastroenterol 2007;13:2089–93. The aim of this pilot study was to determine the role of cytokines in fatigue in patients with Crohn’s disease. The effects of infliximab infusion on fatigue and cytokine levels were assessed. No correlation between the level of fatigue and level of measured cytokines was found; however, both the infliximab and placebo infusions reduced fatigue. The cause of fatigue in Crohn’s disease may be multifactorial, but there is a paucity of studies that explore this issue. Fourteen patients with Crohn’s disease underwent infusion of placebo at baseline and received infliximab at 2 weeks; subjects were reassessed at 4 weeks from baseline. The Multidimensional Fatigue Inventory was used to assess fatigue. Scores of this Inventory range from 4–20, with higher scores reflecting increased fatigue. With a score >13 reflecting fatigue, 86% of subjects were classed as fatigued at baseline. After placebo infusion, fatigue scores decreased by 3.5±1.1 points, which was predominantly accounted for by the 33% of subjects who responded to placebo. Placebo responders with reduced fatigue had fatigue scores that returned to baseline 2 weeks later. After infliximab infusion, fatigue scores decreased by 3.8±1.4 points, 32 and remained reduced for 14 days after the infusion. Fatigue scores correlated with depression scores. There was no correlation between the level of fatigue and level of measured cytokines, including interleukin-6 (IL-6), IL-10, IL-18, and tumor necrosis factor-α (TNF-α). IL-18 levels did decrease after infliximab infusion, but there was no change in the circulating levels of the other cytokines. The reduction of fatigue after infliximab infusion is subject to a placebo effect; however, infliximab’s effect is more sustained than that achieved by placebo. It is not known why placebo can acutely reduce fatigue, or what aspect of infliximab therapy reduces fatigue. Address for reprints: B Oldenburg, Department of Gastroenterology, F02.618, PO Box 85500, 3508 GA Utrecht, The Netherlands. Email: [email protected] Increase in substance P precursor mRNA in noninflamed small-bowel sections in patients with Crohn’s disease Michalski CW, Autschbach F, Selvaggi F et al. Am J Surg 2007;193:476–81. There is significant evidence to demonstrate the involvement of the enteric nervous system in intestinal inflammation; therefore, the authors of this study investigated the role of the substance P (SP) pathway in IBD. Analysis of transmural intestinal tissue samples demonstrated upregulation of protein levels of the receptor for SP (the neurokinin-1 receptor) in IBD samples – primarily in ileal Crohn’s disease. Expression of preprotachykinin-A, which encodes for SP, was significantly upregulated in non-inflamed Crohn’s disease samples. This may indicate an influence of inflamed tissue on healthy parts of the intestine. There is increasing evidence that the nervous system plays an active role in the modulation of intestinal inflammation in IBD. Tachykinins, which include the neuropeptide substance P (SP), mediate their effects on target cells by activation of three distinct G-protein-coupled receptors: the neurokinin-1, -2, and -3 receptors (NK-1R, NK-2R, and NK-3R). Of these, the NK1R has been demonstrated to be crucial in the transmission of the biological effects of SP. The activity of SP is limited by its degradation and inactivation by neutral endopeptidase (NEP) – an enzyme that is widely distributed in mammalian tissues. There are conflicting data in the literature regarding the neuro-inflammatory role of SP and its receptors in IBD, with some reports demonstrating an increase, and others showing a decrease in the SP immunoreactivity index in samples from Crohn’s disease patients. Both a marked upregulation of NK1R mRNA levels and decreased SP receptor expression have also been reported in ulcerative colitis (UC). INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 33 PATHOGENESIS The present authors analyzed 112 IBD and control transmural intestinal tissue samples for concomitant expression of NEP, SP, and NK-1R isoforms (NK-1R-L and NK-1R-S), in addition to the expression of the preprotachykinin-A (PPT-A) gene, which encodes for SP. Analysis of different intestinal segments from Crohn’s disease and UC samples (inflamed and non-inflamed small and large bowel) revealed no significant differences in NK-1R-L and NK-1R-S mRNA expression, although there was a tendency towards overall lower NK-1R-S mRNA copy numbers. Immunoblot analysis showed that NK-1R protein levels were upregulated in the IBD samples, with a more pronounced enhancement in cases of Crohn’s disease compared with UC. NEP protein levels were similar in normal, Crohn’s disease, and UC intestinal tissue. Interestingly, a significant upregulation of PPT-A mRNA expression and a tendency towards slightly increased NK-1R-L isoform expression was found in non-inflamed Crohn’s disease tissue samples. In conclusion, the authors suggest a contribution of SP and its NK-1R isoforms, in the local inflammatory reaction in IBD, predominantly in ileal Crohn’s disease. They also speculate that the significant upregulation of PPT-A mRNA in the non-inflamed ileum suggests that inflamed intestine has an influence on healthy tissue. Address for reprints: H Friess, Department of General Surgery, University of Heidelberg, Im Neuenheimer Feld 110, D-69120 Heidelberg, Germany. Email: [email protected] Regulation of oxidant-induced intestinal permeability by metalloprotease-dependent epidermal growth factor receptor signaling Forsyth CB, Banan A, Farhadi A et al. J Pharmacol Exp Ther 2007;321:84–97. The aim of the present authors was to address the role of oxidant stress in hyperpermeability of intestinal epithelium, as observed in IBD. Specifically, they aimed to characterize in vitro the putative role of the epidermal growth factor receptor (EGFR) in mediating oxidant-induced gut epithelium hyperpermeability. Using specific inhibitors, oxidant-induced epithelial barrier hyperpermeability was demonstrated to be partly dependent on tumor necrosis factor-converting enzyme-mediated transactivation of EGFR signaling. The authors conclude that their study highlights potential novel therapeutic targets in IBD. Two key determinants of IBD pathogenesis are the hyperpermeability of the gut epithelial barrier, which allows penetration of luminal bacterial products into the mucosa, and an abnormal immune response to these products. The major permeability pathway in IBD is considered to be the paracellular route, which is regulated by tight junctions between the intestinal epithelial cells. Oxidant-induced stress and pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interferon-γ are considered to be critical mediators of intestinal hyperpermeability in IBD. The goal of this well-designed study was to identify the mechanisms of oxidant-induced intestinal hyperpermeability, and to characterize the specific role of oxidant stressmediated transactivation of the epidermal growth factor receptor (EGFR). The authors used in vitro models of intestinal epithelium (Caco-2 human colonic epithelial cells) for their analysis. Cells were grown to confluence on tissue culture plate inserts for permeability and signaling studies and on glass coverslips for microscopy studies. Permeability was measured as apical-to-basolateral flux of a fluorescent marker; Western blotting and reverse transcriptasepolymerase chain reaction techniques were employed for analyses of receptor signaling. Using specific pharmacological inhibitors and blocking antibodies, the authors found oxidant-induced epithelial barrier hyperpermeability to be partly dependent on TNFconverting enzyme (TACE)-mediated transactivation of EGFR signaling, which is convincing evidence to support an association between oxidants and TNF-α in mediating intestinal barrier disruption. The authors propose a novel intestinal hyperpermeability model: the process starts with the presence of oxidant stress, which has previously been demonstrated (and confirmed here) to activate the metalloprotease TACE. Activation of TACE results in its translocation to cell–cell contact zones where it cleaves the precursor form of transforming growth factor-α (TGF-α) to a soluble form. This soluble form of TGF-α is a ligand for the EGFR. Subsequent to ligand binding, the receptor becomes phosphorylated and activates downstream mitogenactivated protein kinases (specifically, extracellular signalregulated kinases 1/2 [ERK1/2]). Activated ERK1/2 then phosphorylates cellular targets, increasing intestinal permeability through pathways that have yet to be characterized. These data may have a significant impact on the understanding of IBD pathogenesis, and could help to identify potential targets for the development of new therapies. Address for reprints: CB Forsyth, Rush University Medical Center, Department of Internal Medicine, Section of Gastroenterology, 1725 West Harrison Street, Suite 206, Chicago, IL 60612, USA. Email: [email protected] INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 33 MEETING REPORT RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 34 40th Annual Meeting of the European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) Barcelona, Spain, 9–12 May, 2007 Johan Van Limbergen and Richard K Russell Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children, Edinburgh, UK The European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) meeting is the annual meeting of European pediatric gastroenterologists but continues to attract a global audience. This year’s meeting was held in Barcelona, Spain, which was hosting the Spanish Grand prix concurrently with ESPGHAN. IBD was, as always, well represented throughout the meeting. IBD registries Data demonstrating increasing IBD incidence rates have led many groups to establish databases/registries in their own populations [1]. Jakobsen and colleagues (Hvidovre Hospital, Copenhagen, Denmark) presented data from a population of 98 children diagnosed with IBD at age <15 years during 1998–2004 in the east of Denmark. This area represents approximately 60% of the total Danish population. The median age at diagnosis was 9.8 years. The investigators noted that the incidence of childhood Crohn’s disease increased throughout the study. The incidence of ulcerative colitis (UC), which has stabilized in most populations, also continued to rise. The incidence of IBD during the last 2 years of the study was 6.2 cases per 100 000 population. Katarzyna Karolewska-Bochenek (University of Warsaw, Warsaw, Poland) presented data on pediatric IBD patients from Poland, which were collected from 2002–2004. Notably, this was a national, prospective study. The median age at diagnosis was 11.7 years. In contrast to most other pediatric IBD study populations, a much lower incidence of IBD – 2.7 cases per 100 000 – was found; furthermore, there was a predominance of UC with an incidence rate double that of Crohn’s disease. Data from an analysis of the CEDATA registry (a combined IBD population in Austria and Germany) were presented. This study collected data on 2289 pediatric patients, 648 of whom represented new diagnoses of IBD during the study period of 2004–2006. Patients with UC had a shorter time to diagnosis 34 (approximately 4 months compared with 8 months in Crohn’s disease patients), and had significantly lower C-reactive protein levels than patients with Crohn’s disease. The most remarkable fact about this study was the extremely low number of Crohn’s disease patients in whom enteral feeding was used, at just 4%. Dominique Turck (Lille University Children’s Hospital, Lille, France) presented data from the well-established EPIMAD registry, collected from the North of France (1988–2002) on the natural history of UC diagnosed in childhood. A total of 151 cases of childhood-onset UC were identified, and follow-up data were available on 113 subjects. The study demonstrated the usual high rates of pancolitis in childhood UC (60% at follow-up). Interesting clinical data were presented demonstrating that 68% of patients required steroids within the first year after diagnosis, 26% needed immunosuppression during the first 2 years, and cumulative colectomy rates were 9%, 15%, and 20% at 1, 3, and 5 years post-diagnosis, respectively. In a sister presentation from the same registry, Gwenola Vernier-Massouille (Hôpital Huriez, Lille, France) demonstrated in 477 pediatric Crohn’s disease patients that more than one-third would require surgery at 5 years post-diagnosis. This risk was reduced in patients treated with 5-aminosalicylates, azathioprine, or infliximab. Data from the current authors’ own registry, based on the comparative analysis of 1234 adults and 353 children with IBD, demonstrated that children with Crohn’s disease were more likely to be male, have less isolated ileal and colonic disease, have more upper tract disease, and less complicated disease at diagnosis using the Montreal classification [2]. Children with UC were more likely to have extensive disease at diagnosis. Genetics The genome-wide association study reported by Duerr et al. in 2006, which implicated variants of the IL23R gene in INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 35 ESPGHAN 2007 conferring protection against Crohn’s disease, has led to several pediatric replication studies [3]. At this meeting, the present authors presented data from the Scottish pediatric population that analyzed the contribution of the Arg381Gln allele in 358 patients with IBD who were diagnosed at <17 years of age. The results demonstrated that the prevalence of this mutation in children with IBD differed significantly from healthy controls (5.9% vs. 11.1%; p=0.01). Arie Levine (Tel Aviv University, Tel Aviv, Israel) presented data on the same allele in children and adults from Israel (a total of 381 patients, of whom, 143 were pediatric). These data replicated the Scottish data, with a lower carriage rate in Crohn’s disease patients compared with healthy controls (4.3% vs. 13.6%; p=0.003). However, no differences were noted in carriage rates when comparing the adult and pediatric Crohn’s disease populations. In a second presentation, Dr Levine presented data on 721 pediatric Crohn’s disease patients from three different populations (USA, Israel, and Italy) demonstrating that the isolated, colonic form of Crohn’s disease is found in patients who carry normal (wild-type) copies of the NOD2/CARD15 Crohn’s disease susceptibility gene. This suggests that the “colonic” phenotype, which has been so well described in young children [4], may be genetically determined. However, this study did not identify the gene or genes that may be involved. Treatment Enteral nutrition A 10-year, single-center experience with enteral nutrition to induce remission in moderate-to-severe Crohn’s disease was described by Kjaergaard Nielsen and colleagues (Hvidovre Hospital). By 30 days of treatment, 63% of Crohn’s disease patients (22/35) achieved clinical remission. Following completion of 4 weeks of enteral nutrition, 50% remained in remission at 3 months. Rita Shergill-Bonner and colleagues (Royal Free Hospital, London, UK) presented their experience with food reintroduction after exclusive enteral nutrition in children presenting with newly diagnosed Crohn’s disease. Although 10 of 102 children had a documented reaction to a specific food item, only two children remained intolerant to these items after food reintroduction. The investigators concluded that gradual reintroduction over 4 weeks in order to identify specific food intolerances is not necessary, and that specific foods are only rarely responsible for symptoms in children with Crohn’s disease. Thus, they suggested that a shorter period of food-group reintroduction over 1–2 weeks after exclusive enteral nutrition may be adequate. Victor Manuel Navas (Materno-Infantil Hospital, Malaga, Spain) presented pilot data on fecal calprotectin levels after successfully inducing disease remission in children with Crohn’s disease using enteral nutrition (Modulen®, Nestle Clinical Nutrition, Glendale, CA, USA). Although 12 of 14 children had a significant reduction in Pediatric Crohn’s disease Activity Index (PCDAI) score by week 4 and week 8 (PCDAI ≤15), fecal calprotectin remained elevated (>125 μg/g) in all children. Infliximab Frank Ruemmele (Necker Enfants Malades, Paris, France) presented data on sustained remission after stopping infliximab in a cohort of 42 children with Crohn’s disease, using a combined retrospective/prospective analysis. After three infliximab infusions, 35 of 42 children (83%) were in full remission. Twenty-three of 35 responders (66%) developed infliximab dependency, as defined by the need to continue or repeat (n=13), or increase infliximab doses (n=10). Mucosal healing or marked improvement was seen in 10 of 13 children at follow-up endoscopy. Nevertheless, relapses occurred within 6 months after stopping infliximab in most of these patients. Lissy De Ridder (VU Medical Center, Amsterdam, The Netherlands) reported a similar experience with infliximab dependency in The Netherlands, in a retrospective study describing the clinical experience with infliximab in nine Dutch pediatric centers from 1992–2006. A clinical response to infliximab was observed in 75% of patients that received the agent (n=62). However, 60% of this cohort was dependent on repeat infliximab infusions. There was no difference in response between early (within 1–2 years of diagnosis) and late starting of infliximab therapy, in contrast with earlier reports from Kugathasan et al. [5]. In the Question & Answer sessions of both these studies, concerns regarding the long-term safety of infliximab maintenance therapy were raised. Over the last year, the reports of hepatosplenic γδ-T cell lymphoma in children treated with infliximab and 6-mercaptopurine/azathioprine have led to much debate at international meetings regarding the concomitant use of immunomodulators and infliximab [6]. During this ESPGHAN meeting, a symposium entitled “Young patients, Big considerations: REACHing new heights in pediatric Crohn’s disease” was held in order to discuss the latest evidence on this issue. In spite of its early morning slot, this session was one of the best attended of the whole meeting. Firstly, Gigi Veereman-Wauters (Queen Paola Children’s Hospital, Antwerp, Belgium) discussed the findings of the REACH (A Randomized, Multicenter, Open-label Study to Evaluate the Safety and Efficacy of Anti-TNF Monoclonal INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 35 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 36 JOHAN VAN LIMBERGEN AND RICHARD K RUSSELL Antibody Infliximab in Pediatric Subjects with Moderate to Severe Crohn’s Disease) study, which was published recently in the Gastroenterology journal [7]. This trial evaluated the efficacy of infliximab in inducing remission of moderate-tosevere pediatric Crohn’s disease, and maintaining remission in infliximab responders. Concomitant therapy with an immunomodulator was required for inclusion in the trial. The main conclusions of REACH were that pediatric patients responding to an induction regimen of infliximab were more likely to achieve a clinical response and be in remission at week 54 without dose adjustment when their maintenance therapy was given every 8 weeks, rather than every 12 weeks. Allowing for dose intensification in the case of relapse, remission rates but not response rates at week 54 were superior with every 8-week dosing compared with every 12week dosing. Following this overview, Anne Griffiths (Sick Kids Hospital, Toronto, ON, Canada) provided a comprehensive overview of the different strategies to optimize infliximab treatment, taking into account the latest safety data and recent reports from our colleagues that work with adult patients on infliximab efficacy relative to concomitant use of an immunomodulator. Salvatore Cucchiara (University of Rome, Rome, Italy) then summarized the available evidence and hypotheses on how early infliximab treatment could change the course of disease. During the panel discussion, both the management of loss of response to infliximab and long-term safety concerns were addressed. Differences in practice between Europe and North America regarding the withdrawal of immunomodulator therapy while continuing infliximab were debated. 36 The complexity of the question of prolonged immunosuppressive therapy was further illustrated by a case report presented at the meeting. Klaus-Michael Keller (Deutsche Klinik für Diagnostik, Wiesbaden, Germany) described the first case of a 15-year-old boy, diagnosed at 26 months with indeterminate colitis (later changed to UC), who died from a disseminated T cell lymphoma following 9 years of azathioprine therapy. The development of this lymphoma has been described in post-transplant patients receiving azathioprine, but this is the first reported case in a pediatric IBD patient. Acknowledgements Johan Van Limbergen is funded by a Research Training Fellowship from Action Medical Research, The Gay-Ramsay-Steel-Maitland or Stafford Trust, and the Hazel M Wood Charitable Trust. Disclosures The authors have no relevant financial interests to disclose. References 1. Loftus EV Jr. Clinical epidemiology of inflammatory bowel disease: incidence, prevalence, and environmental influences. Gastroenterology 2004;126:1504–17. 2. Silverberg MS, Satsangi J, Ahmad T et al. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Can J Gastroenterol 2005;19:5–36A. 3. Duerr RH, Taylor KD, Brant SR et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 2006;314:1461–3. 4. Heyman MB, Kirschner BS, Gold BD et al. Children with early-onset inflammatory bowel disease (IBD): analysis of a pediatric IBD consortium registry. J Pediatr 2005;146:35–40. 5. Kugathasan S, Werlin SL, Martinez A et al. Prolonged duration of response to infliximab in early but not late pediatric Crohn’s disease. Am J Gastroenterol 2000;95:3189–94. 6. Mackey AC, Green L, Liang LC et al. Hepatosplenic T cell lymphoma associated with infliximab use in young patients treated for inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2007;44:265–7. 7. Hyams J, Crandall W, Kugathasan S et al. Induction and maintenance infliximab therapy for the treatment of moderate-to-severe Crohn’s disease in children. Gastroenterology 2007;132:863–73. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 37 Washington, DC, USA, 20–23 May 2007 Fernando Velayos, MD University of California San Francisco, San Francisco, CA, USA The annual Digestive Diseases Week (DDW) meeting for 2007 was held in Washington, DC, USA, from May 20–23. A record number were present this year, with close to 17 000 attendees. Over 1500 abstracts related to IBD were presented. Any attempt to condense within a brief summary the wide variety of research presented over the 4-day meeting would be challenging. Therefore, this report seeks to provide a representative overview of some of the most important IBD-related clinical research presented this year in the areas of IBD therapeutics, novel therapies, and finally genetic, serological, and clinical predictors of disease. IBD therapeutics Biologics Biological agents are antibodies engineered to attack specific components of the inflammatory cascade. Pivotal randomized controlled trials presented at previous DDW meetings have demonstrated efficacy for four biologics: infliximab, adalimumab, certolizumab, and natalizumab. The first three compounds are monoclonal antibodies that inhibit the potent inflammatory cytokine, tumor necrosis factor-α (TNF-α), while natalizumab is a monoclonal antibody that targets the leukocyte adhesion molecule, α4 integrin. Infliximab and adalimumab have been approved by the US Food and Drug Administration (FDA) for the treatment of mild-to-moderate Crohn’s disease, while certolizumab and natalizumab are undergoing regulatory review. Infliximab has also been approved for the treatment of ulcerative colitis (UC). At this year’s meeting, several important post hoc analyses of these pivotal trials, as well as investigator-initiated clinical trials, sought to address as yet unanswered questions, such as whether introduction of biologics earlier in the disease course of IBD improves outcomes and whether concomitant immunomodulators are necessary for maintaining remission in patients receiving biologics. With regard to the first question, Stefan Schreiber (Christian Albrechts University, Kiel, Germany) and colleagues presented several post hoc analyses of pivotal randomized trials showing that in all but one, treatment with biologics earlier in the disease course of Crohn’s disease improved response. In the CHARM (Crohn’s Trial of the Fully Human Antibody Adalimumab for Remission Maintenance) study, 52% of patients who had disease of <2 years were in remission at week 56, compared with 35% who had 2–5 years of disease, and 33% who had >5 years of disease. In the PRECISE 2 (Pegylated Antibody Fragment Evaluation in Crohn’s Disease: Safety and Efficacy) study, 61.5% of patients treated with certolizumab who had <2 years of disease were in remission at week 26, compared with 47.9% in all treated patients. In the ENCORE (Efficacy of Natalizumab in Crohn’s Disease Response and Remission) study, 52% of patients treated with natalizumab who had <3 years of disease were in remission at week 12 compared with 38% in the entire treated population. In contrast to these positive studies, 57% of patients who had <3 years of disease and received natalizumab in the ENACT-2 (Evaluation of Natalizumab As Continuous Therapy-2) study were in remission at 12 months compared with 55% in the overall treated population. Taken together, these data are consistent with the hypothesis that earlier introduction of biologics may improve patient outcomes, although clearly this hypothesis needs to be more rigorously and formally tested in dedicated clinical trials. With regard to the second question, several studies presented this year examined whether concomitant immunomodulators improve clinical outcomes in patients receiving biological agents. This question has become particularly pertinent in light of recently reported rare but fatal cases of hepatosplenic T cell lymphoma in patients receiving both medications. Gary Lichtenstein (University of Pennsylvania, Philadelphia, PA, USA) and colleagues analyzed four clinical trials for infliximab, ACCENT I (A Crohn’s Disease Clinical Trial Evaluating Infliximab in a New Long-term Treatment Regimen I), ACCENT II, ACT I (Active Ulcerative Colitis Trial I) and ACT II, and found concomitant immunomodulators use did not appreciably influence clinical outcomes. As examples, 34% of patients receiving concomitant immunomodulators in ACT I were in remission INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 37 MEETING REPORT Digestive Diseases Week 2007 (DDW 2007) RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 38 FERNANDO VELAYOS at week 54 compared with 36% of patients who were not on immunomodulators. In ACCENT I, 37% of patients receiving concomitant immunomodulators were in remission compared with 32% without. These post hoc results are similar to those found in a prospective trial, the IMID (Infliximab Maintenance Immunosuppressive Discontinuation) trial, presented by Gert Van Assche (University of Leuven Hospitals, Leuven, Belgium) and colleagues. In this trial, 80 Crohn’s patients who were in clinical remission on both infliximab and an immunomodulator were randomized to either continue or interrupt their immunomodulator after 6 months of infliximab therapy. In the 41 patients undergoing ileocolonoscopy at 2 years, 61% of patients who continued immunomodulator therapy were in remission compared with 67% of control subjects. While both studies support the notion that concomitant immunosuppression does not impact the 1–2 year outcome of maintenance infliximab in Crohn’s disease, the longer term outcome remains unknown. Immunomodulators Patients with Crohn’s disease who undergo ileocolic resection are known to be at a significantly elevated risk of disease recurrence. This year, several presentations addressed whether immunomodulators, particularly azathioprine, could prevent endoscopic recurrence and a second surgery. In a retrospective study, Pavol Papay and colleagues (Medical University of Vienna, Vienna, Austria) identified 377 patients with Crohn’s disease who underwent a first operation; 40% of these patients (n=123) underwent a second surgery within 10 years. In a multivariate model, use of azathioprine or 6-mercaptopurine therapy for ≥3 months after the initial surgery reduced the odds of a second surgery by one-third, suggesting that azathioprine is effective for preventing a second surgery. In a prospective clinical trial, Geert D’Haens (University of Leuven) and colleagues treated all patients with Crohn’s disease who were at high risk of developing postoperative recurrence (age <40 years, more than one intestinal surgery, use of steroids at surgery) with metronidazole (750 mg/day) for 3 months immediately after surgery and randomized patients to receive azathioprine or placebo. They found that at 12 months, severe endoscopic recurrence was lower in the azathioprine-treated group than in the placebo group (44% vs. 69%; p<0.05), again suggesting that azathioprine is beneficial in maintaining surgically induced remission. 5-Aminosalicylates 5-Aminosalicylates (5-ASAs) are the cornerstone of therapy for mild-to-moderate UC. Several studies presented at DDW evaluated whether less frequent dosing was as effective as more frequent dosing for the induction and maintenance of remission in UC. Using Salofalk® (Axcan Pharma, Mont- 38 Saint-Hilaire, QC, Canada), an oral mesalamine formulation coated with a Eudagrit-L polymer, Wolfgang Kruis (University of Cologne, Cologne, Germany) and colleagues randomized 381 patients with active UC to receive either once daily 3 g mesalamine (Salofalk) or 1 g mesalamine granules three times daily for 8 weeks. After 8 weeks, both groups were equivalent with regard to the primary endpoint of clinical remission (79.1% daily mesalamine vs. 75.7% three times per day mesalamine). Using a different formulation, MMX® (Multi-Matrix System)-mesalamine (Shire Pharmaceuticals, Wayne, PA, USA), which contains a gastro-resistant, pH-dependent coating with hydrophilic and lipophilic matrices, Michael Kamm (St Mark’s Hospital, London, UK) and colleagues randomized 362 UC patients in remission to receive 12 months of MMX-mesalamine at either 2.4 g daily or 1.2 g twice daily. In this open-label study, both groups were equivalent with regard to rates of endoscopic and clinical remission (67.8% once-daily vs. 72.3% twice daily). Both of these studies highlight the efficacy of 5-ASA as a cornerstone therapy in UC and reflect current attempts to ensure efficacy while improving patient adherence through less frequent dosing. Investigational therapies This year, several promising investigational and novel therapies for Crohn’s disease and UC were presented at DDW that expand not only on the molecular targets being tested, but also the mode of delivery. Crohn’s disease CNTO 1275 CNTO 1275 is a fully human monoclonal antibody targeting the common p40 subunit of interleukin-12 (IL-12) and IL-23. In a 54-week, randomized Phase IIa trial, William Sandborn (Mayo Clinic, Rochester, MN, USA) and colleagues assessed the safety and efficacy of a single intravenous infusion of CNTO 1275 compared with four subcutaneous injections. At week 8, 49% of patients receiving CNTO 1275 were in clinical response compared with 39.6% of those who received placebo (p=0.34). Among patients with prior infliximab experience, 59.1% receiving CNTO 1275 were in response compared with 25.9% receiving placebo (p=0.02). The authors concluded that short-term treatment with CNTO 1275 was well tolerated and showed a beneficial effect in the infliximabexperienced patients. CCX282-B Unlike other investigational therapies for Crohn’s disease, which are administered intravenously or subcutaneously, INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 39 DDW 2007 CCX282-B is an orally administered antagonist of the chemokine receptor, CCR9, which is expressed by mucosahoming intestinal leukocytes. Satish Keshav (Royal Free and University College London Medical School, London, UK) and colleagues tested the safety and efficacy of CCX282-B in 71 patients with mild-to-moderate Crohn’s disease. Patients were randomized to receive a daily capsule of CCX282-B for 28 days or placebo. A total of 49% of patients treated with CCX282-B had a 70-point reduction in their Crohn’s disease activity index, which was similar to that seen in patients who received placebo (45%; p=not significant). In a subgroup analysis consisting of patients with more active disease, there was a trend toward a greater improvement (58% vs. 31% in patients receiving placebo). Although a trend for benefit was observed only on subgroup analysis, this study provides encouraging data to suggest that there may be benefit to this oral biologic for the treatment of Crohn’s disease. Visilizumab Visilizumab, a humanized anti-CD3 monoclonal antibody, has been investigated in Phase II/III clinical trials for severe UC that is resistant to intravenous steroids. This year, Daan Hommes (University of Leiden, Leiden, The Netherlands) and colleagues presented open-label, Phase I data describing its use in patients with moderate-to-severe nonfistulizing Crohn’s disease. Patients were administered an intravenous bolus of visilizumab (10 μg/kg) on two consecutive days. At day 59, a response was observed in 72% of patients, and remission in 33%. As expected, most patients experienced mild-to-moderate cytokine release. This study adds to a growing list of important inflammatory targets currently under evaluation for Crohn’s disease. It should be noted, however, that since this year’s DDW, visilizumab has been withdrawn from a trial in steriodresistant, severe UC, due to complications. Extracorporeal photoimmune therapy Data for another unique approach to Crohn’s disease therapy, extracorporeal photoimmune therapy (ECP), was presented at DDW. ECP involves treatment of peripheral blood leukocytes with 8-methoxypsoralen and ultraviolet A light to induce apoptosis. After this process, these autologous apoptotic leukocytes are re-infused back into the patient. Maria Abreu (Mount Sinai School of Medicine, New York, NY, USA) and colleagues evaluated the safety and efficacy of ECP in a multicenter, open-label study of 28 patients with moderate Crohn’s disease. Patients received twice weekly ECP treatments for 4 weeks, followed by twice weekly treatment every other week for the subsequent 6 weeks. At week 6, 50% of patients responded to ECP and 25% were in remission. Half of the patients who were refractory or intolerant to anti-TNF agents responded to this therapy. Two adverse events – health deterioration and anemia – were observed. Again, these preliminary data appear promising, leading the authors to recommend a randomized trial of ECP. Ulcerative colitis Rosiglitazone Thiazolidinedione ligands for the gamma subtype of peroxisome proliferator-activated receptors (PPARγ), which are widely used to treat type 2 diabetes mellitus, have been proposed to have anti-inflammatory properties in the colon. However, their efficacy in humans with active UC has not previously been tested. To this end, James Lewis (University of Pennsylvania, Philadelphia, PA, USA) and colleagues randomized 105 patients with mild-to-moderate UC refractory or intolerant to 5-ASAs to receive 12 weeks of either 4 mg twice daily rosigliatzone or placebo. At 12 weeks, 44% of patients treated with rosiglitazone and 23% who received placebo had achieved a clinical response (p=0.03). The main adverse effect of rosiglitazone was lower extremity swelling, which is a known side effect of this class of medications. Unfortunately, concurrent with the presentation of these data at DDW, a meta-analysis was published in the New England Journal of Medicine that linked rosiglitazone with a 43% increased risk of myocardial infarction in diabetes patients [1], prompting a review of this drug by the US FDA. Should short- and long-term safety concerns be adequately addressed, this study is important as rosiglitazone could provide a novel second line therapy for patients refractory to or intolerant of 5-ASAs. MMX budesonide Ileal-release budesonide is currently approved for treatment of mild, ileocolic Crohn’s disease, as it has preferential release in the right-sided colon. Using MMX technology, Geert D’Haens (University Hospitals Leuven) and colleagues reported the efficacy and safety of MMX-budesonide (Cosmo Technologies Ltd., Ireland) in patients with UC limited to the left colon. A total of 36 patients with moderately active, left-sided UC were treated with either 9 mg of MMX-budesonide for 8 weeks, or placebo for 4 weeks followed by MMX-budesonide for 4 weeks. Though not significant, there were trends toward clinical improvement in the MMX-budesonide group compared with the “placebo” group (47% vs. 33%). Based on these encouraging findings, the authors recommended a larger, controlled trial for this novel formulation of budesonide. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 39 RT292_1_REM_IBD_8.1_07.qxd 2/10/07 15:40 Page 40 FERNANDO VELAYOS Genetic, serological, and clinical predictors of disease severity The diverse clinical phenotypes of IBD appear to be influenced by a variety of genetic, immune, and clinical factors. There is interest in having the ability to predict which patients are at risk for developing complicated IBD, as these patients are most likely to benefit from early and aggressive medical therapy. Several studies presented this year reflect the genetic, serological, and clinical approaches for determining those who are at high risk for developing complicated IBD phenotype. A few of the important studies are presented here. Liesbet Henckaerts (University Hospital Gasthuisberg, Leuven, Belgium) and colleagues genotyped a cohort of 505 patients with Crohn’s disease for NOD2/CARD15 variants, which act as an intracellular receptor for bacterial components in monocytes, and, in turn, lead to activation of the inflammatory response. Using the Kaplan–Meier method, the authors observed a stepwise shortening of the surgery-free interval with increasing number of CARD15 variants. Based on these results, the authors suggested that genotyping for CARD15 at the time of diagnosis might be useful for identifying those patients who might benefit from early introduction of aggressive treatment. Marla Dubinsky (Cedars-Sinai Medical Center, Los Angeles, CA, USA) and colleagues tested the sera of 797 prospectively followed pediatric Crohn’s disease patients with the aim of examining the associations between the serological immune responses to microbial antigens (anti-Cbir1 [flagellin], antiouter membrane protein C [OmpC], and anti-Saccharomyces cerevisiae antibodies [ASCA]) and clinical phenotypes. The authors found that increasing immune reactivity predicted aggressive complicating disease in children, as the highest odds of developing internal penetrating or fibrostenosing disease occurred in patients positive for all three immune responses. They also tested three CARD15 variants, which were found to be associated with small bowel disease, but not with disease behavior. Based on the these results, the authors recommended that baseline serological assessment may identify children at highest risk for a complicated phenotype and might be useful in identifying those patients who may benefit from early introduction of aggressive treatment. Besides genetic and serological factors, clinical factors have also been shown to predict disabling Crohn’s disease within the first 5 years of diagnosis [2]. Recently, three clinical factors (diagnosis age <40 years, early steroids, and early perianal disease) that predicted disabling Crohn’s disease 40 within the first 5 years of diagnosis were recently determined in a large cohort of patients from France. These factors had not previously been validated in other populations. To this end, two studies attempted to validate these criteria. In a Belgian population, Edouard Louis and colleagues (University of Liège, Liège, Belgium) applied these criteria to 361 Crohn’s diease patients, distinguishing between very severe and moderately severe Crohn’s disease. They found that two to three risk factors had a positive predictive value of 67.8% for moderately disabling Crohn’s disease and 78.3% for severe disabling Crohn’s disease. In a North American population, Philippe Seksik (Hôpital Saint-Antoine, Paris, France) and colleagues reported that the positive predictive value of disabling Crohn’s disease with two and three predictors was 62% and 75%, respectively. Due to variations in definitions for disabling Crohn’s disease, the authors differed in their estimate of the percentage of patients that may benefit from a top-down, more aggressive therapeutic approach (20% in the Belgian population study, and 74% in the study of North American patients). Routine use of genetic, serological, and clinical factors to identify patients at risk of complicated disease is not currently standard care. Nevertheless, identification of such factors and refinement of relevant criteria should be encouraged in order to help create a risk profile that can aid the clinician in identifying which patients are likely to have the more aggressive clinical course, and therefore could benefit from a more aggressive medical approach. Summary Presentations at DDW 2007 examined ways of improving current therapy and tested novel targets of delivery systems that will expand the clinical options available to treat IBD. Refining which genetic, serological, and clinical predictors are associated with particular phenotypes will help to match current and emerging therapies with those patients most likely to benefit from a more aggressive medical approach. Disclosures Dr Velayos has served on speaker’s bureau for Centocor, Procter and Gamble, Prometheus, and Shire, and has received grant support from Procter and Gamble. References 1. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 2007;356:2457–71. 2. Beaugerie L, Seksik P, Nion-Larmurier I et al. Predictors of Crohn’s disease. Gastroenterology 2006;130:650–6. INFLAMMATORY BOWEL DISEASE MONITOR VOL 8 NO 1 2007 AUTHORS INFLAMMATORY BOWEL DISEASE MONITOR Reader Survey – Let Us Know What You Think! Please take a few moments to complete this survey. We value your opinion. Please photocopy this page, complete the survey below, and fax it back to Remedica on 312 372 0217 (USA, Canada) or +44 (0)20 7759 2951 (rest of the world). Or you can visit the INFLAMMATORY BOWEL DISEASE MONITOR website to provide feedback: www.ibdmonitor.com 1. We aim to provide practical information for practicing physicians, surgeons, and other healthcare professionals. How would you rate the information presented in this issue? Strongly agree Strongly disagree a) The technical quality of information included in INFLAMMATORY BOWEL DISEASE MONITOR was acceptable: 1 2 3 4 5 b) The information was relevant to my practice: 1 2 3 4 5 c) The information was presented clearly: 1 2 3 4 5 d) The Leading Articles provided new information regarding the understanding and treatment of inflammatory bowel disease: 1 2 3 4 5 e) The Clinical Review section was helpful, and I would like to see analyses in future issues: 1 2 3 4 5 2. Did you learn anything from the CME activity INFLAMMATORY BOWEL DISEASE MONITOR that will change the way you practice medicine? ■ Yes ■ No If so, what? ................................................................................................................................................................................ 3. Is there anything you learned from the CME activity INFLAMMATORY BOWEL DISEASE MONITOR that prompts you to seek further information that may influence the way you practice medicine in the future? ■ Yes ■ No If so, what? ................................................................................................................................................................................ 4. Would you like to recommend INFLAMMATORY BOWEL DISEASE MONITOR to a colleague? ■ Yes ■ No My colleague’s email is: ............................................................................................................................................................. 5. What specific topics do you think should be covered in future issues? ................................................................................................................................................................................................... Name......................................................................................... Job title ................................................................................... Institution .................................................................................................................................................................................. Address ...................................................................................................................................................................................... Country ..................................................................................... Post/zip code .......................................................................... Email ..........................................................................................................................................................................................