Z Gastroenterol 2022; 60(03): 332-418
DOI: 10.1055/a-1713-3941
Leitlinie

Aktualisierte S3-Leitlinie „Diagnostik und Therapie des Morbus Crohn“ der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS) – August 2021 – AWMF-Registernummer: 021-004

Andreas Sturm
1   Klinik für Innere Medizin mit Schwerpunkt Gastroenterologie, DRK Kliniken Berlin Westend, Berlin, Deutschland
,
Raja Atreya
2   Medizinische Klinik 1, Universitätsklinikum Erlangen, Deutschland
,
Dominik Bettenworth
3   Praxis für Innere Medizin, Münster, Deutschland
,
Bernd Bokemeyer
4   Gastroenterologische Gemeinschaftspraxis Minden, Deutschland
,
Axel Dignaß
5   Medizinische Klinik I, Agaplesion Markus Krankenhaus, Frankfurt am Main, Deutschland
,
Robert Ehehalt
6   Praxis für Gastroenterologie, Heidelberg, Deutschland
,
Christoph Germer
7   Chirurgische Klinik I, Universitätsklinikum Würzburg, Deutschland
,
Philip C. Grunert
8   Klinik für Innere Medizin IV (Gastroenterologie, Hepatologie und Infektiologie), Universitätsklinikum Jena, Deutschland
,
Ulf Helwig
9   Internistische Praxengemeinschaft, Oldenburg, Deutschland
,
Klaus Herrlinger
10   Innere Medizin I, Asklepios Klinik Nord, Hamburg, Deutschland
,
Peter Kienle
11   Allgemein- und Viszeralchirurgie, Theresienkrankenhaus und Sankt Hedwig-Klinik GmbH, Mannheim, Deutschland
,
Martin E. Kreis
12   Klinik für Allgemein-, Viszeral- und Gefäßchirurgie, Charité Campus Benjamin Franklin – Universitätsmedizin Berlin, Deutschland
,
Torsten Kucharzik
13   Klinik für Allgemeine Innere Medizin und Gastroenterologie, Klinikum Lüneburg, Deutschland
,
Jost Langhorst
14   Klinik für Integrative Medizin und Naturheilkunde, Klinikum am Bruderwald, Bamberg, Deutschland
,
Christian Maaser
15   Gastroenterologie, Ambulanzzentrum Lüneburg, Deutschland
,
Johann Ockenga
16   Medizinische Klinik II, Klinikum Bremen Mitte – Gesundheit Nord, Bremen, Deutschland
,
Claudia Ott
17   Gastroenterologie Facharztzentrum, Regensburg, Deutschland
,
Britta Siegmund
18   Medizinische Klinik I, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Deutschland
,
Sebastian Zeißig
19   Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Deutschland
,
Andreas Stallmach
8   Klinik für Innere Medizin IV (Gastroenterologie, Hepatologie und Infektiologie), Universitätsklinikum Jena, Deutschland
,
Collaborators:
› Author Affiliations


Publication History

Received: 15 October 2021

Accepted: 03 December 2021

Article published online:
09 March 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • Literatur

  • 1 Bundes, G.d. Diagnosedaten der Krankenhäuser ab 2000 (Eckdaten der vollstationären Patienten und Patientinnen). Gliederungsmerkmale: Jahre, Behandlungs-/Wohnort, ICD10. 2021 Available from: https://www.gbe-bund.de/gbe/pkg_isgbe5.prc_menu_olap?p_uid=gast&p_aid=81229970&p_sprache=D&p_help=3&p_indnr=550&p_indsp=&p_ityp=H&p_fid=
  • 2 Lammert F, Jansen PL, Lerch MM. Weissbuch Gastroenterologie 2020/2021. ed. Frank L, Petra Lynen J, Markus ML. De Gruyter; 2019
  • 3 Lennard-Jones JE, Shivananda S. Clinical uniformity of inflammatory bowel disease a presentation and during the first year of disease in the north and south of Europe. EC-IBD Study Group. Eur J Gastroenterol Hepatol 1997; 9: 353-359
  • 4 Forrest K, Symmons D, Foster P. Systematic review: is ingestion of paracetamol or non-steroidal anti-inflammatory drugs associated with exacerbations of inflammatory bowel disease?. Aliment Pharmacol Ther 2004; 20: 1035-1043
  • 5 Takeuchi K. et al. Prevalence and mechanism of nonsteroidal anti-inflammatory drug-induced clinical relapse in patients with inflammatory bowel disease. Clin Gastroenterol Hepatol 2006; 4: 196-202
  • 6 Korzenik JR, Podolsky DK. Selective use of selective nonsteroidal anti-inflammatory drugs in inflammatory bowel disease. Clin Gastroenterol Hepatol 2006; 4: 157-159
  • 7 Long MD. et al. Role of Nonsteroidal Anti-Inflammatory Drugs in Exacerbations of Inflammatory Bowel Disease. J Clin Gastroenterol 2016; 50: 152-156
  • 8 Beaugerie L, Rahier JF, Kirchgesner J. Predicting, Preventing, and Managing Treatment-Related Complications in Patients With Inflammatory Bowel Diseases. Clin Gastroenterol Hepatol 2020; 18: 1324-1335 e2
  • 9 Singh S. et al. Comparative Risk of Serious Infections With Biologic and/or Immunosuppressive Therapy in Patients With Inflammatory Bowel Diseases: A Systematic Review and Meta-Analysis. Clin Gastroenterol Hepatol 2020; 18: 69-81 e3
  • 10 Schumacher G, Kollberg B, Sandstedt B. A prospective study of first attacks of inflammatory bowel disease and infectious colitis. Histologic course during the 1st year after presentation. Scand J Gastroenterol 1994; 29: 318-332
  • 11 Garcia Rodriguez LA, Ruigomez A, Panes J. Acute gastroenteritis is followed by an increased risk of inflammatory bowel disease. Gastroenterology 2006; 130: 1588-1594
  • 12 Ott C. et al. Smoking increases the risk of extraintestinal manifestations in Crohn’s disease. World J Gastroenterol 2014; 20: 12269-12276
  • 13 Scharrer S. et al. Passive Smoking Increases the Risk for Intestinal Surgeries in Patients With Crohn’s Disease. Inflamm Bowel Dis 2021; 27: 379-385
  • 14 Orholm M. et al. Familial occurrence of inflammatory bowel disease. N Engl J Med 1991; 324: 84-88
  • 15 Schreiber S. et al. Genetics of Crohn disease, an archetypal inflammatory barrier disease. Nat Rev Genet 2005; 6: 376-388
  • 16 Fagan EA. et al. Serum levels of C-reactive protein in Crohn’s disease and ulcerative colitis. Eur J Clin Invest 1982; 12: 351-359
  • 17 Vermeire S, Van Assche G, Rutgeerts P. C-reactive protein as a marker for inflammatory bowel disease. Inflamm Bowel Dis 2004; 10: 661-665
  • 18 Sands BE. Biomarkers of Inflammation in Inflammatory Bowel Disease. Gastroenterology 2015; 149: 1275-1285 e2
  • 19 Henriksen M. et al. C-reactive protein: a predictive factor and marker of inflammation in inflammatory bowel disease. Results from a prospective population-based study. Gut 2008; 57: 1518-1523
  • 20 Solem CA. et al. Correlation of C-reactive protein with clinical, endoscopic, histologic, and radiographic activity in inflammatory bowel disease. Inflamm Bowel Dis 2005; 11: 707-712
  • 21 Schoepfer AM. et al. Fecal calprotectin correlates more closely with the Simple Endoscopic Score for Crohn’s disease (SES-CD) than CRP, blood leukocytes, and the CDAI. Am J Gastroenterol 2010; 105: 162-169
  • 22 Bou-Fakhredin R. et al. Insights into the diagnosis and management of iron deficiency in inflammatory bowel disease. Expert Rev Hematol 2017; 10: 801-808
  • 23 Cappellini MD. et al. Iron deficiency across chronic inflammatory conditions: International expert opinion on definition, diagnosis, and management. Am J Hematol 2017; 92: 1068-1078
  • 24 Stein J, Hartmann F, Dignass AU. Diagnosis and management of iron deficiency anemia in patients with IBD. Nat Rev Gastroenterol Hepatol 2010; 7: 599-610
  • 25 Langhorst J. et al. Noninvasive markers in the assessment of intestinal inflammation in inflammatory bowel diseases: performance of fecal lactoferrin, calprotectin, and PMN-elastase, CRP, and clinical indices. Am J Gastroenterol 2008; 103: 162-169
  • 26 Geerling BJ. et al. Comprehensive nutritional status in recently diagnosed patients with inflammatory bowel disease compared with population controls. Eur J Clin Nutr 2000; 54: 514-521
  • 27 Filippi J. et al. Nutritional deficiencies in patients with Crohn’s disease in remission. Inflamm Bowel Dis 2006; 12: 185-191
  • 28 Ibd Working Group of the European Society for Paediatric Gastroenterology, H. and Nutrition. Inflammatory bowel disease in children and adolescents: recommendations for diagnosis--the Porto criteria. J Pediatr Gastroenterol Nutr 2005; 41: 1-7
  • 29 Ogura Y. et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature 2001; 411: 603-606
  • 30 Hugot JP. et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature 2001; 411: 599-603
  • 31 Lesage S. et al. CARD15 / NOD2 mutational analysis and genotype-phenotype correlation in 612 patients with inflammatory bowel disease. Am J Hum Genet 2002; 70: 845-857
  • 32 Radlmayr M. et al. The c-insertion mutation of the NOD2 gene is associated with fistulizing and fibrostenotic phenotypes in Crohn’s disease. Gastroenterology 2002; 122: 2091-2092
  • 33 Peeters M. et al. Diagnostic value of anti-Saccharomyces cerevisiae and antineutrophil cytoplasmic autoantibodies in inflammatory bowel disease. Am J Gastroenterol 2001; 96: 730-734
  • 34 Zholudev A. et al. Serologic testing with ANCA, ASCA, and anti-OmpC in children and young adults with Crohn’s disease and ulcerative colitis: diagnostic value and correlation with disease phenotype. Am J Gastroenterol 2004; 99: 2235-2241
  • 35 Reese GE. et al. Diagnostic precision of anti-Saccharomyces cerevisiae antibodies and perinuclear antineutrophil cytoplasmic antibodies in inflammatory bowel disease. Am J Gastroenterol 2006; 101: 2410-2422
  • 36 Smids C. et al. The value of serum antibodies in differentiating inflammatory bowel disease, predicting disease activity and disease course in the newly diagnosed patient. Scand J Gastroenterol 2017; 52: 1104-1112
  • 37 Plevy S. Do serological markers and cytokines determine the indeterminate?. J Clin Gastroenterol 2004; 38 (Suppl. 01) S51-S56
  • 38 Aadland E, Fagerhol MK. Faecal calprotectin: a marker of inflammation throughout the intestinal tract. Eur J Gastroenterol Hepatol 2002; 14: 823-825
  • 39 Sipponen T. et al. Crohn’s disease activity assessed by fecal calprotectin and lactoferrin: correlation with Crohn’s disease activity index and endoscopic findings. Inflamm Bowel Dis 2008; 14: 40-46
  • 40 van Rheenen PF, Van de Vijver E, Fidler V. Faecal calprotectin for screening of patients with suspected inflammatory bowel disease: diagnostic meta-analysis. BMJ 2010; 341: c3369
  • 41 Mylonaki M. et al. Enteric infection in relapse of inflammatory bowel disease: importance of microbiological examination of stool. Eur J Gastroenterol Hepatol 2004; 16: 775-778
  • 42 Ananthakrishnan AN, McGinley EL, Binion DG. Excess hospitalisation burden associated with Clostridium difficile in patients with inflammatory bowel disease. Gut 2008; 57: 205-210
  • 43 Coremans G. et al. The value of ileoscopy with biopsy in the diagnosis of intestinal Crohn’s disease. Gastrointest Endosc 1984; 30: 167-172
  • 44 Samuel S. et al. Endoscopic skipping of the distal terminal ileum in Crohn’s disease can lead to negative results from ileocolonoscopy. Clin Gastroenterol Hepatol 2012; 10: 1253-1259
  • 45 Dong J. et al. Ultrasound as a diagnostic tool in detecting active Crohn’s disease: a meta-analysis of prospective studies. Eur Radiol 2014; 24: 26-33
  • 46 Fiorino G. et al. Prospective comparison of computed tomography enterography and magnetic resonance enterography for assessment of disease activity and complications in ileocolonic Crohn’s disease. Inflamm Bowel Dis 2011; 17: 1073-1080
  • 47 Panes J. et al. Systematic review: the use of ultrasonography, computed tomography and magnetic resonance imaging for the diagnosis, assessment of activity and abdominal complications of Crohn’s disease. Aliment Pharmacol Ther 2011; 34: 125-145
  • 48 Puylaert CA. et al. Grading of Crohn’s disease activity using CT, MRI, US and scintigraphy: a meta-analysis. Eur Radiol 2015; 25: 3295-3313
  • 49 Maaser C. et al. ECCO-ESGAR Guideline for Diagnostic Assessment in IBD Part 1: Initial diagnosis, monitoring of known IBD, detection of complications. J Crohns Colitis 2019; 13: 144-164
  • 50 Horsthuis K, Stokkers PC, Stoker J. Detection of inflammatory bowel disease: diagnostic performance of cross-sectional imaging modalities. Abdom Imaging 2008; 33: 407-416
  • 51 Taylor SA. et al. Diagnostic accuracy of magnetic resonance enterography and small bowel ultrasound for the extent and activity of newly diagnosed and relapsed Crohn’s disease (METRIC): a multicentre trial. Lancet Gastroenterol Hepatol 2018; 3: 548-558
  • 52 Calabrese E. et al. Bowel Ultrasonography in the Management of Crohn’s Disease. A Review with Recommendations of an International Panel of Experts. Inflamm Bowel Dis 2016; 22: 1168-1183
  • 53 Calabrese E. et al. Accuracy of small-intestine contrast ultrasonography, compared with computed tomography enteroclysis, in characterizing lesions in patients with Crohn’s disease. Clin Gastroenterol Hepatol 2013; 11: 950-955
  • 54 Parente F. et al. Oral contrast enhanced bowel ultrasonography in the assessment of small intestine Crohn’s disease. A prospective comparison with conventional ultrasound, × ray studies, and ileocolonoscopy. Gut 2004; 53: 1652-1657
  • 55 Rieder F. et al. Crohn’s disease complicated by strictures: a systematic review. Gut 2013; 62: 1072-1084
  • 56 Bettenworth D. et al. Assessment of Crohn’s disease-associated small bowel strictures and fibrosis on cross-sectional imaging: a systematic review. Gut 2019; 68: 1115-1126
  • 57 Annunziata ML. et al. Upper gastrointestinal involvement of Crohn’s disease: a prospective study on the role of upper endoscopy in the diagnostic work-up. Dig Dis Sci 2012; 57: 1618-1623
  • 58 Diaz L. et al. Upper Gastrointestinal Involvement in Crohn Disease: Histopathologic and Endoscopic Findings. South Med J 2015; 108: 695-700
  • 59 Ripolles T. et al. Contrast-enhanced ultrasound in the differentiation between phlegmon and abscess in Crohn’s disease and other abdominal conditions. Eur J Radiol 2013; 82: e525-e531
  • 60 Desmond AN. et al. Crohn’s disease: factors associated with exposure to high levels of diagnostic radiation. Gut 2008; 57: 1524-1529
  • 61 Kopylov U. et al. Small bowel capsule endoscopy in the management of established Crohn’s disease: clinical impact, safety, and correlation with inflammatory biomarkers. Inflamm Bowel Dis 2015; 21: 93-100
  • 62 Kopylov U. et al. Diagnostic yield of capsule endoscopy versus magnetic resonance enterography and small bowel contrast ultrasound in the evaluation of small bowel Crohn’s disease: Systematic review and meta-analysis. Dig Liver Dis 2017; 49: 854-863
  • 63 Katsinelos P. et al. Diagnostic yield and clinical impact of wireless capsule endoscopy in patients with chronic abdominal pain with or without diarrhea: a Greek multicenter study. Eur J Intern Med 2011; 22: e63-e66
  • 64 De Bona M. et al. Capsule endoscopy findings in patients with suspected Crohn’s disease and biochemical markers of inflammation. Dig Liver Dis 2006; 38: 331-335
  • 65 Kopylov U. et al. Fecal calprotectin for the prediction of small-bowel Crohn’s disease by capsule endoscopy: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol 2016; 28: 1137-1144
  • 66 Nemeth A. et al. Use of patency capsule in patients with established Crohn’s disease. Endoscopy 2016; 48: 373-379
  • 67 Liao Z. et al. Indications and detection, completion, and retention rates of small-bowel capsule endoscopy: a systematic review. Gastrointest Endosc 2010; 71: 280-286
  • 68 Vernier-Massouille G. et al. Natural history of pediatric Crohn’s disease: a population-based cohort study. Gastroenterology 2008; 135: 1106-1113
  • 69 de Ridder L. et al. Single-balloon enteroscopy, magnetic resonance enterography, and abdominal US useful for evaluation of small-bowel disease in children with (suspected) Crohn’s disease. Gastrointest Endosc 2012; 75: 87-94
  • 70 Heine GD. et al. Double-balloon enteroscopy: indications, diagnostic yield, and complications in a series of 275 patients with suspected small-bowel disease. Endoscopy 2006; 38: 42-48
  • 71 Gay G, Delvaux M. Double balloon enteroscopy in Crohn’s disease and related disorders: our experience. Gastrointest Endosc 2007; 66: S82-S90
  • 72 Manes G. et al. Use of double-balloon enteroscopy in the management of patients with Crohn’s disease: feasibility and diagnostic yield in a high-volume centre for inflammatory bowel disease. Surg Endosc 2009; 23: 2790-2795
  • 73 Wang P. et al. Outcomes and safety of double-balloon enteroscopy in small bowel diseases: a single-center experience of 1531 procedures. Surg Endosc 2020; 35: 576-583
  • 74 Benmassaoud A. et al. The Use of Balloon-assisted Enteroscopy at a Large Volume Centre: A Retrospective Analysis. J Can Assoc Gastroenterol 2018; 1: 33-39
  • 75 Arulanandan A. et al. Systematic review: Safety of balloon assisted enteroscopy in Crohn’s disease. World J Gastroenterol 2016; 22: 8999-9011
  • 76 Di Nardo G. et al. Usefulness of single-balloon enteroscopy in pediatric Crohn’s disease. Gastrointest Endosc 2012; 75: 80-86
  • 77 Despott EJ. et al. Effective dilation of small-bowel strictures by double-balloon enteroscopy in patients with symptomatic Crohn’s disease (with video). Gastrointest Endosc 2009; 70: 1030-1036
  • 78 Pohl J. et al. Diagnostic and therapeutic yield of push-and-pull enteroscopy for symptomatic small bowel Crohn’s disease strictures. Eur J Gastroenterol Hepatol 2007; 19: 529-534
  • 79 Hirai F. et al. Efficacy of Endoscopic Balloon Dilation for Small Bowel Strictures in Patients With Crohn’s Disease: A Nationwide, Multi-centre, Open-label, Prospective Cohort Study. J Crohns Colitis 2018; 12: 394-401
  • 80 Eglinton TW. et al. Clinical and genetic risk factors for perianal Crohn’s disease in a population-based cohort. Am J Gastroenterol 2012; 107: 589-596
  • 81 Wewer MD. et al. The Incidence and Disease Course of Perianal Crohn’s Disease: A Danish Nationwide Cohort Study, 1997–2015. J Crohns Colitis 2020; 15: 5-13
  • 82 Regueiro M. The role of endoscopy in the evaluation of fistulizing Crohn’s disease. Gastrointest Endosc Clin N Am 2002; 12: 621-633
  • 83 Spradlin NM. et al. A randomized prospective trial of endoscopic ultrasound to guide combination medical and surgical treatment for Crohn’s perianal fistulas. Am J Gastroenterol 2008; 103: 2527-2535
  • 84 Savoye-Collet C. et al. Fistulizing perianal Crohn’s disease: contrast-enhanced magnetic resonance imaging assessment at 1 year on maintenance anti-TNF-alpha therapy. Inflamm Bowel Dis 2011; 17: 1751-1758
  • 85 Reginelli A. et al. MRI of perianal fistulas in Crohn’s disease. Acta Biomed 2020; 91: 27-33
  • 86 Dagli U. et al. Transrectal ultrasound in the diagnosis and management of inflammatory bowel disease. Endoscopy 1999; 31: 152-157
  • 87 Luglio G. et al. Diagnostic Accuracy of 3-Dimensional Endoanal Ultrasound in Identifying Perianal Crohn’s Fistulas. Dis Colon Rectum 2018; 61: 931-937
  • 88 Siddiqui MR. et al. A diagnostic accuracy meta-analysis of endoanal ultrasound and MRI for perianal fistula assessment. Dis Colon Rectum 2012; 55: 576-585
  • 89 Maconi G. et al. Transperineal ultrasound in the detection of perianal and rectovaginal fistulae in Crohn’s disease. Am J Gastroenterol 2007; 102: 2214-2219
  • 90 Bor R. et al. Prospective Comparison of Magnetic Resonance Imaging, Transrectal and Transperineal Sonography, and Surgical Findings in Complicated Perianal Crohn Disease. J Ultrasound Med 2016; 35: 2367-2372
  • 91 Bezzio C. et al. New horizons in the imaging of perianal Crohn’s disease: transperineal ultrasonography. Expert Rev Gastroenterol Hepatol 2017; 11: 523-530
  • 92 Maconi G, Greco MT, Asthana AK. Transperineal Ultrasound for Perianal Fistulas and Abscesses – A Systematic Review and Meta-Analysis. Ultraschall Med 2017; 38: 265-272
  • 93 Peyrin-Biroulet L. et al. Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE): Determining Therapeutic Goals for Treat-to-Target. Am J Gastroenterol 2015; 110: 1324-1338
  • 94 Mosli MH. et al C-Reactive Protein, Fecal Calprotectin, and Stool Lactoferrin for Detection of Endoscopic Activity in Symptomatic Inflammatory Bowel Disease Patients: A Systematic Review and Meta-Analysis. Am J Gastroenterol 2015; 110: 802-819 ; quiz 820
  • 95 Kiss LS. et al. Early clinical remission and normalisation of CRP are the strongest predictors of efficacy, mucosal healing and dose escalation during the first year of adalimumab therapy in Crohn’s disease. Aliment Pharmacol Ther 2011; 34: 911-922
  • 96 Vermeire S, Van Assche G, Rutgeerts P. Laboratory markers in IBD: useful, magic, or unnecessary toys?. Gut 2006; 55: 426-431
  • 97 Zubin G, Peter L. Predicting Endoscopic Crohn’s Disease Activity Before and After Induction Therapy in Children: A Comprehensive Assessment of PCDAI, CRP, and Fecal Calprotectin. Inflamm Bowel Dis 2015; 21: 1386-1391
  • 98 Sipponen T, Kolho KL. Fecal calprotectin in diagnosis and clinical assessment of inflammatory bowel disease. Scand J Gastroenterol 2015; 50: 74-80
  • 99 Colombel JF. et al. Effect of tight control management on Crohn’s disease (CALM): a multicentre, randomised, controlled phase 3 trial. Lancet 2018; 390: 2779-2789
  • 100 Bryant RV. et al. Gastrointestinal ultrasound in inflammatory bowel disease: an underused resource with potential paradigm-changing application. Gut 2018; 67: 973-985
  • 101 Kucharzik T. et al. Use of Intestinal Ultrasound to Monitor Crohn’s Disease Activity. Clin Gastroenterol Hepatol 2017; 15: 535-542 e2
  • 102 Kucharzik T. et al Intestinal ultrasound after 48 weeks of treatment with ustekinumab in Crohn’s disease: STARDUST trial substudy. UEG Week Virtual October 2020; 8 (Suppl. 01) 1265-1266
  • 103 Peyrin-Biroulet L. et al. Clinical disease activity, C-reactive protein normalisation and mucosal healing in Crohn’s disease in the SONIC trial. Gut 2014; 63: 88-95
  • 104 Baert F. et al Mucosal healing predicts sustained clinical remission in patients with early-stage Crohn’s disease. Gastroenterology 2010; 138: 463-468 ; quiz e10–e11
  • 105 Colombel JF. et al. Infliximab, azathioprine, or combination therapy for Crohn’s disease. N Engl J Med 2010; 362: 1383-1395
  • 106 Colombel JF. et al. Four-year maintenance treatment with adalimumab in patients with moderately to severely active ulcerative colitis: Data from ULTRA 1, 2, and 3. Am J Gastroenterol 2014; 109: 1771-1780
  • 107 Shah SC. et al. Systematic review with meta-analysis: mucosal healing is associated with improved long-term outcomes in Crohn’s disease. Aliment Pharmacol Ther 2016; 43: 317-333
  • 108 Colombel JF. et al. Randomised clinical trial: deep remission in biologic and immunomodulator naive patients with Crohn’s disease – a SONIC post hoc analysis. Aliment Pharmacol Ther 2015; 41: 734-746
  • 109 D’Inca R. et al. Can calprotectin predict relapse risk in inflammatory bowel disease?. Am J Gastroenterol 2008; 103: 2007-2014
  • 110 Tibble JA. et al. Surrogate markers of intestinal inflammation are predictive of relapse in patients with inflammatory bowel disease. Gastroenterology 2000; 119: 15-22
  • 111 Costa F. et al. Calprotectin is a stronger predictive marker of relapse in ulcerative colitis than in Crohn’s disease. Gut 2005; 54: 364-368
  • 112 Heida A, Park KT, van Rheenen PF. Clinical Utility of Fecal Calprotectin Monitoring in Asymptomatic Patients with Inflammatory Bowel Disease: A Systematic Review and Practical Guide. Inflamm Bowel Dis 2017; 23: 894-902
  • 113 Ben-Horin S, Chowers Y. Review article: loss of response to anti-TNF treatments in Crohn’s disease. Aliment Pharmacol Ther 2011; 33: 987-995
  • 114 Guerbau L. et al. Patients with Crohn’s Disease with High Body Mass Index Present More Frequent and Rapid Loss of Response to Infliximab. Inflamm Bowel Dis 2017; 23: 1853-1859
  • 115 Zhang QW. et al. Loss of response to scheduled infliximab therapy for Crohn’s disease in adults: A systematic review and meta-analysis. J Dig Dis 2019; 20: 65-72
  • 116 Hendy P, Hart A, Irving P. Anti-TNF drug and antidrug antibody level monitoring in IBD: a practical guide. Frontline Gastroenterol 2016; 7: 122-128
  • 117 Moore C, Corbett G, Moss AC. Systematic Review and Meta-Analysis: Serum Infliximab Levels During Maintenance Therapy and Outcomes in Inflammatory Bowel Disease. J Crohns Colitis 2016; 10: 619-625
  • 118 Karmiris K. et al. Influence of trough serum levels and immunogenicity on long-term outcome of adalimumab therapy in Crohn’s disease. Gastroenterology 2009; 137: 1628-1640
  • 119 Vande Casteele N. et al. Trough concentrations of infliximab guide dosing for patients with inflammatory bowel disease. Gastroenterology 2015; 148: 1320-1329 e3
  • 120 Ungar B. et al. Prospective Observational Evaluation of Time-Dependency of Adalimumab Immunogenicity and Drug Concentrations: The POETIC Study. Am J Gastroenterol 2018; 113: 890-898
  • 121 Battat R. et al. Association Between Ustekinumab Trough Concentrations and Clinical, Biomarker, and Endoscopic Outcomes in Patients With Crohn’s Disease. Clin Gastroenterol Hepatol 2017; 15: 1427-1434 e2
  • 122 Williet N. et al. Association Between Low Trough Levels of Vedolizumab During Induction Therapy for Inflammatory Bowel Diseases and Need for Additional Doses Within 6 Months. Clin Gastroenterol Hepatol 2017; 15: 1750-1757 e3
  • 123 Steenholdt C. et al. Individualised therapy is more cost-effective than dose intensification in patients with Crohn’s disease who lose response to anti-TNF treatment: a randomised, controlled trial. Gut 2014; 63: 919-927
  • 124 D’Haens G. et al. Increasing Infliximab Dose Based on Symptoms, Biomarkers, and Serum Drug Concentrations Does Not Increase Clinical, Endoscopic, and Corticosteroid-Free Remission in Patients With Active Luminal Crohn’s Disease. Gastroenterology 2018; 154: 1343-1351 e1
  • 125 Vande Casteele N. et al. Therapeutic Drug Monitoring of Tumor Necrosis Factor Antagonists in Crohn Disease: A Theoretical Construct to Apply Pharmacokinetics and Guidelines to Clinical Practice. Inflamm Bowel Dis 2020; 27: 1346-1355
  • 126 Srinivasan A. et al. Anti-TNF Re-induction Is as Effective, Simpler, and Cheaper Compared With Dose Interval Shortening for Secondary Loss of Response in Crohn’s Disease. J Crohns Colitis 2018; 12: 280-288
  • 127 Peeters H. et al. Efficacy of switching to infliximab in patients with Crohn’s disease with loss of response to adalimumab. Acta Gastroenterol Belg 2018; 81: 15-21
  • 128 Chaparro M. et al. Outcome after a dose “de-intensification” strategy with anti-TNF drugs in patients with Crohn’s disease. Gastroenterol Hepatol 2016; 39: 255-260
  • 129 Peyrin-Biroulet L. et al. Loss of Response to Vedolizumab and Ability of Dose Intensification to Restore Response in Patients With Crohn’s Disease or Ulcerative Colitis: A Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol 2019; 17: 838-846 e2
  • 130 Fumery M. et al. Effectiveness And Safety Of Ustekinumab Intensification At 90 Mg Every Four Weeks In Crohn’s Disease: A Multicenter Study. J Crohns Colitis 2020; 15: 222-227
  • 131 Brandes A. et al. Real-world biologic treatment and associated cost in patients with inflammatory bowel disease. Z Gastroenterol 2019; 57: 843-851
  • 132 Papamichael K. et al. Improved Long-term Outcomes of Patients With Inflammatory Bowel Disease Receiving Proactive Compared With Reactive Monitoring of Serum Concentrations of Infliximab. Clin Gastroenterol Hepatol 2017; 15: 1580-1588 e3
  • 133 Papamichael K. et al. Proactive Infliximab Monitoring Following Reactive Testing is Associated With Better Clinical Outcomes Than Reactive Testing Alone in Patients With Inflammatory Bowel Disease. J Crohns Colitis 2018; 12: 804-810
  • 134 Papamichael K. et al. Proactive Therapeutic Drug Monitoring of Adalimumab Is Associated With Better Long-term Outcomes Compared With Standard of Care in Patients With Inflammatory Bowel Disease. J Crohns Colitis 2019; 13: 976-981
  • 135 Ricciuto A. et al. Clinical Outcomes With Therapeutic Drug Monitoring in Inflammatory Bowel Disease: A Systematic Review With Meta-Analysis. J Crohns Colitis 2018; 12: 1302-1315
  • 136 Assa A. et al. Proactive Monitoring of Adalimumab Trough Concentration Associated With Increased Clinical Remission in Children With Crohn’s Disease Compared With Reactive Monitoring. Gastroenterology 2019; 157: 985-996 e2
  • 137 Roblin X. et al. Addition of azathioprine to the switch of anti-TNF in patients with IBD in clinical relapse with undetectable anti-TNF trough levels and antidrug antibodies: a prospective randomised trial. Gut 2020; 69: 1206-1212
  • 138 Issa M. et al. Impact of Clostridium difficile on inflammatory bowel disease. Clin Gastroenterol Hepatol 2007; 5: 345-351
  • 139 Rodemann JF. et al. Incidence of Clostridium difficile infection in inflammatory bowel disease. Clin Gastroenterol Hepatol 2007; 5: 339-344
  • 140 Bossuyt P. et al. Increasing incidence of Clostridium difficile-associated diarrhea in inflammatory bowel disease. J Crohns Colitis 2009; 3: 4-7
  • 141 Ricciardi R. et al. Epidemiology of Clostridium difficile colitis in hospitalized patients with inflammatory bowel diseases. Dis Colon Rectum 2009; 52: 40-45
  • 142 Nguyen GC. et al. A national survey of the prevalence and impact of Clostridium difficile infection among hospitalized inflammatory bowel disease patients. Am J Gastroenterol 2008; 103: 1443-1450
  • 143 Singh H. et al. Higher Incidence of Clostridium difficile Infection Among Individuals With Inflammatory Bowel Disease. Gastroenterology 2017; 153: 430-438 e2
  • 144 Kucharzik T. et al. Aktualisierte S3-Leitlinie Colitis ulcerosa – Living Guideline. Z Gastroenterol 2020; 58: e241-e326
  • 145 Dimitroulia E. et al. Frequent detection of cytomegalovirus in the intestine of patients with inflammatory bowel disease. Inflamm Bowel Dis 2006; 12: 879-884
  • 146 Roblin X. et al. Prevalence of cytomegalovirus infection in steroid-refractory Crohn’s disease. Inflamm Bowel Dis 2012; 18: E1396-E1397
  • 147 Cottone M. et al. Prevalence of cytomegalovirus infection in severe refractory ulcerative and Crohn’s colitis. Am J Gastroenterol 2001; 96: 773-775
  • 148 Hommes DW. et al. The pathogenicity of cytomegalovirus in inflammatory bowel disease: a systematic review and evidence-based recommendations for future research. Inflamm Bowel Dis 2004; 10: 245-250
  • 149 Kredel LI. et al. Accuracy of diagnostic tests and a new algorithm for diagnosing cytomegalovirus colitis in inflammatory bowel diseases: a diagnostic study. Int J Colorectal Dis 2019; 34: 229-237
  • 150 Halpin SJ, Ford AC. Prevalence of symptoms meeting criteria for irritable bowel syndrome in inflammatory bowel disease: systematic review and meta-analysis. Am J Gastroenterol 2012; 107: 1474-1482
  • 151 Perera LP. et al. Presence of Irritable Bowel Syndrome Symptoms in Quiescent Inflammatory Bowel Disease Is Associated with High Rate of Anxiety and Depression. Dig Dis Sci 2019; 64: 1923-1928
  • 152 Lemann M. et al. A randomized, double-blind, controlled withdrawal trial in Crohn’s disease patients in long-term remission on azathioprine. Gastroenterology 2005; 128: 1812-1818
  • 153 Wenzl HH. et al. Withdrawal of long-term maintenance treatment with azathioprine tends to increase relapse risk in patients with Crohn’s disease. Dig Dis Sci 2015; 60: 1414-1423
  • 154 Vilien M. et al. Randomized controlled azathioprine withdrawal after more than two years treatment in Crohn’s disease: increased relapse rate the following year. Aliment Pharmacol Ther 2004; 19: 1147-1152
  • 155 Gisbert JP, Marin AC, Chaparro M. The Risk of Relapse after Anti-TNF Discontinuation in Inflammatory Bowel Disease: Systematic Review and Meta-Analysis. Am J Gastroenterol 2016; 111: 632-647
  • 156 Torres J. et al. Systematic Review of Effects of Withdrawal of Immunomodulators or Biologic Agents From Patients With Inflammatory Bowel Disease. Gastroenterology 2015; 149: 1716-1730
  • 157 Kennedy NA. et al. Relapse after withdrawal from anti-TNF therapy for inflammatory bowel disease: an observational study, plus systematic review and meta-analysis. Aliment Pharmacol Ther 2016; 43: 910-923
  • 158 Louis E. et al Maintenance of remission among patients with Crohn’s disease on antimetabolite therapy after infliximab therapy is stopped. Gastroenterology 2012; 142: 63-70 e5 ; quiz e31
  • 159 Rutgeerts P. et al. Natural history of recurrent Crohn’s disease at the ileocolonic anastomosis after curative surgery. Gut 1984; 25: 665-672
  • 160 Rutgeerts P. et al. Predictability of the postoperative course of Crohn’s disease. Gastroenterology 1990; 99: 956-963
  • 161 Rutgeerts P. et al. Ornidazole for prophylaxis of postoperative Crohn’s disease recurrence: a randomized, double-blind, placebo-controlled trial. Gastroenterology 2005; 128: 856-861
  • 162 Peyrin-Biroulet L. et al. Azathioprine and 6-mercaptopurine for the prevention of postoperative recurrence in Crohn’s disease: a meta-analysis. Am J Gastroenterol 2009; 104: 2089-2096
  • 163 Regueiro M. et al Infliximab prevents Crohn’s disease recurrence after ileal resection. Gastroenterology 2009; 136: 441-450 e1 ; quiz 716
  • 164 Hukkinen M. et al. Fecal calprotectin in the prediction of postoperative recurrence of Crohn’s disease in children and adolescents. J Pediatr Surg 2016; 51: 1467-1472
  • 165 Yamamoto T. et al. Faecal calprotectin and lactoferrin as markers for monitoring disease activity and predicting clinical recurrence in patients with Crohn’s disease after ileocolonic resection: A prospective pilot study. United European Gastroenterol J 2013; 1: 368-374
  • 166 Qiu Y. et al. Fecal calprotectin for evaluating postoperative recurrence of Crohn’s disease: a meta-analysis of prospective studies. Inflamm Bowel Dis 2015; 21: 315-322
  • 167 Tham YS. et al. Fecal calprotectin for detection of postoperative endoscopic recurrence in Crohn’s disease: systematic review and meta-analysis. Therap Adv Gastroenterol 2018; 11: 1756284818785571
  • 168 Parente F. et al. Behaviour of the bowel wall during the first year after surgery is a strong predictor of symptomatic recurrence of Crohn’s disease: a prospective study. Aliment Pharmacol Ther 2004; 20: 959-968
  • 169 Castiglione F. et al. Oral contrast-enhanced sonography for the diagnosis and grading of postsurgical recurrence of Crohn’s disease. Inflamm Bowel Dis 2008; 14: 1240-1245
  • 170 Castiglione F. et al. Bowel wall thickness at abdominal ultrasound and the one-year-risk of surgery in patients with Crohn’s disease. Am J Gastroenterol 2004; 99: 1977-1983
  • 171 Maconi G. et al. Preoperative characteristics and postoperative behavior of bowel wall on risk of recurrence after conservative surgery in Crohn’s disease: a prospective study. Ann Surg 2001; 233: 345-352
  • 172 Calabrese E. et al. Severity of postoperative recurrence in Crohn’s disease: correlation between endoscopic and sonographic findings. Inflamm Bowel Dis 2009; 15: 1635-1642
  • 173 Pallotta N. et al. Ultrasonographic detection and assessment of the severity of Crohn’s disease recurrence after ileal resection. BMC Gastroenterol 2010; 10: 69
  • 174 Koilakou S. et al. Endoscopy and MR enteroclysis: equivalent tools in predicting clinical recurrence in patients with Crohn’s disease after ileocolic resection. Inflamm Bowel Dis 2010; 16: 198-203
  • 175 Soyer P. et al. Suspected anastomotic recurrence of Crohn disease after ileocolic resection: evaluation with CT enteroclysis. Radiology 2010; 254: 755-764
  • 176 Biancone L. et al. Virtual colonoscopy compared with conventional colonoscopy for stricturing postoperative recurrence in Crohn’s disease. Inflamm Bowel Dis 2003; 9: 343-350
  • 177 Cammarota T. et al. Role of bowel ultrasound as a predictor of surgical recurrence of Crohn’s disease. Scand J Gastroenterol 2013; 48: 552-555
  • 178 Sailer J. et al. Anastomotic recurrence of Crohn’s disease after ileocolic resection: comparison of MR enteroclysis with endoscopy. Eur Radiol 2008; 18: 2512-2521
  • 179 Kono T. et al. Prospective postsurgical capsule endoscopy in patients with Crohn’s disease. World J Gastrointest Endosc 2014; 6: 88-98
  • 180 Bourreille A. et al. Wireless capsule endoscopy versus ileocolonoscopy for the diagnosis of postoperative recurrence of Crohn’s disease: a prospective study. Gut 2006; 55: 978-983
  • 181 Yung DE. et al. Capsule Endoscopy, Magnetic Resonance Enterography, and Small Bowel Ultrasound for Evaluation of Postoperative Recurrence in Crohn’s Disease: Systematic Review and Meta-Analysis. Inflamm Bowel Dis 2017; 24: 93-100
  • 182 Surawicz CM. et al. Rectal biopsy in the diagnosis of Crohn’s disease: value of multiple biopsies and serial sectioning. Gastroenterology 1981; 80: 66-71
  • 183 Surawicz CM, Belic L. Rectal biopsy helps to distinguish acute self-limited colitis from idiopathic inflammatory bowel disease. Gastroenterology 1984; 86: 104-113
  • 184 Seldenrijk CA. et al. Histopathological evaluation of colonic mucosal biopsy specimens in chronic inflammatory bowel disease: diagnostic implications. Gut 1991; 32: 1514-1520
  • 185 Jenkins D. et al. Guidelines for the initial biopsy diagnosis of suspected chronic idiopathic inflammatory bowel disease. The British Society of Gastroenterology Initiative. J Clin Pathol 1997; 50: 93-105
  • 186 Bentley E. et al. How could pathologists improve the initial diagnosis of colitis? Evidence from an international workshop. J Clin Pathol 2002; 55: 955-960
  • 187 Dejaco C. et al. Diagnosing colitis: a prospective study on essential parameters for reaching a diagnosis. Endoscopy 2003; 35: 1004-1008
  • 188 Geboes K. et al. Is ileoscopy with biopsy worthwhile in patients presenting with symptoms of inflammatory bowel disease?. Am J Gastroenterol 1998; 93: 201-206
  • 189 McHugh JB, Appelman HD, McKenna BJ. The diagnostic value of endoscopic terminal ileum biopsies. Am J Gastroenterol 2007; 102: 1084-1089
  • 190 Tanaka M. et al. Morphologic criteria applicable to biopsy specimens for effective distinction of inflammatory bowel disease from other forms of colitis and of Crohn’s disease from ulcerative colitis. Scand J Gastroenterol 1999; 34: 55-67
  • 191 Theodossi A. et al. Observer variation and discriminatory value of biopsy features in inflammatory bowel disease. Gut 1994; 35: 961-968
  • 192 Mowat C. et al. Guidelines for the management of inflammatory bowel disease in adults. Gut 2011; 60: 571-607
  • 193 Petritsch W. et al. [Diagnosis in chronic inflammatory bowel diseases--report of the Austrian Chronic Inflammatory Bowel Disease Study Group]. Acta Med Austriaca 1998; 25: 37-43
  • 194 Rubio CA, Sjodahl K, Lagergren J. Lymphocytic esophagitis: a histologic subset of chronic esophagitis. Am J Clin Pathol 2006; 125: 432-437
  • 195 Ebach DR. et al. Lymphocytic esophagitis: a possible manifestation of pediatric upper gastrointestinal Crohn’s disease. Inflamm Bowel Dis 2011; 17: 45-49
  • 196 Bataille F. et al. Histopathological parameters as predictors for the course of Crohn’s disease. Virchows Arch 2003; 443: 501-507
  • 197 Bernstein CN. et al. Cancer risk in patients with inflammatory bowel disease: a population-based study. Cancer 2001; 91: 854-862
  • 198 Jess T. et al. Increased risk of intestinal cancer in Crohn’s disease: a meta-analysis of population-based cohort studies. Am J Gastroenterol 2005; 100: 2724-2729
  • 199 Canavan C, Abrams KR, Mayberry J. Meta-analysis: colorectal and small bowel cancer risk in patients with Crohn’s disease. Aliment Pharmacol Ther 2006; 23: 1097-1104
  • 200 Rubio CA. et al. Further studies on the frequency and length of the glandulo-metaplastic esophageal mucosa in baboons. In Vivo 2009; 23: 955-958
  • 201 Laukoetter MG. et al. Intestinal cancer risk in Crohn’s disease: a meta-analysis. J Gastrointest Surg 2011; 15: 576-583
  • 202 Maykel JA. et al. Crohn’s colitis: the incidence of dysplasia and adenocarcinoma in surgical patients. Dis Colon Rectum 2006; 49: 950-957
  • 203 Bergeron V. et al. Risk factors for neoplasia in inflammatory bowel disease patients with pancolitis. Am J Gastroenterol 2010; 105: 2405-2411
  • 204 Palascak-Juif V. et al. Small bowel adenocarcinoma in patients with Crohn’s disease compared with small bowel adenocarcinoma de novo. Inflamm Bowel Dis 2005; 11: 828-832
  • 205 Feldstein RC, Sood S, Katz S. Small bowel adenocarcinoma in Crohn’s disease. Inflamm Bowel Dis 2008; 14: 1154-1157
  • 206 Egan L. et al. Non-colorectal intestinal tract carcinomas in inflammatory bowel disease: results of the 3rd ECCO Pathogenesis Scientific Workshop (II). J Crohns Colitis 2014; 8: 19-30
  • 207 Odze RD. et al. Premalignant lesions of the digestive tract, in WHO Classification of Tumours of the Digestive System. Bosman FT. et al., Editors Lyon (France): IARC Press; 2010: 10-12
  • 208 Riddell RH. et al. Dysplasia in inflammatory bowel disease: standardized classification with provisional clinical applications. Hum Pathol 1983; 14: 931-968
  • 209 Laine L. et al. SCENIC international consensus statement on surveillance and management of dysplasia in inflammatory bowel disease. Gastrointest Endosc 2015; 81: 489-501 e26
  • 210 Sjoqvist U. et al. Colorectal cancer in colonic Crohn’s disease--high frequency of DNA-aneuploidy. Anticancer Res 2005; 25: 4393-4397
  • 211 Nathanson JW. et al. p53 mutations are associated with dysplasia and progression of dysplasia in patients with Crohn’s disease. Dig Dis Sci 2008; 53: 474-480
  • 212 Preiss JC. et al. [Updated German clinical practice guideline on “Diagnosis and treatment of Crohn’s disease” 2014]. Z Gastroenterol 2014; 52: 1431-1484
  • 213 Dabritz J. et al. Inflammatory Bowel Disease in Childhood and Adolescence. Dtsch Arztebl Int 2017; 114: 331-338
  • 214 Schwerd T, Koletzko S. Inflammatory bowel disease. Monatsschr Kinderheilkd 2017; 165: 257-270
  • 215 Crandall WV. et al. Improved outcomes in a quality improvement collaborative for pediatric inflammatory bowel disease. Pediatrics 2012; 129: e1030-e1041
  • 216 Samson CM. et al. Improved outcomes with quality improvement interventions in pediatric inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2012; 55: 679-688
  • 217 Levine A. et al. ESPGHAN revised porto criteria for the diagnosis of inflammatory bowel disease in children and adolescents. J Pediatr Gastroenterol Nutr 2014; 58: 795-806
  • 218 Levine A. et al. Pediatric modification of the Montreal classification for inflammatory bowel disease: the Paris classification. Inflamm Bowel Dis 2011; 17: 1314-1321
  • 219 Turner D. et al. Which PCDAI Version Best Reflects Intestinal Inflammation in Pediatric Crohn Disease?. J Pediatr Gastroenterol Nutr 2017; 64: 254-260
  • 220 van Rheenen PF. et al. The Medical Management of Paediatric Crohn’s Disease: an ECCO-ESPGHAN Guideline Update. J Crohns Colitis 2020; 15: 171-194
  • 221 Cagol L. et al. Vaccination rate and immunity of children and adolescents with inflammatory bowel disease or autoimmune hepatitis in Germany. Vaccine 2020; 38: 1810-1817
  • 222 Ruemmele FM. et al. Consensus guidelines of ECCO/ESPGHAN on the medical management of pediatric Crohn’s disease. J Crohns Colitis 2014; 8: 1179-1207
  • 223 Ricciuto A. et al. Predicting Outcomes in Pediatric Crohn’s Disease for Management Optimization: Systematic Review and Consensus Statements From the Pediatric Inflammatory Bowel Disease-Ahead Program. Gastroenterology 2020; 160: 403-436
  • 224 Thangarajah D. et al. Systematic review: Body composition in children with inflammatory bowel disease. Aliment Pharmacol Ther 2015; 42: 142-157
  • 225 Posovszky C. Barth TFE. [The gut: center of immunity: Rare inflammatory bowel diseases caused by immunodeficiencies]. Pathologe 2020; 41: 211-223
  • 226 Uhlig HH. et al. The diagnostic approach to monogenic very early onset inflammatory bowel disease. Gastroenterology 2014; 147: 990-1007 e3
  • 227 Thomson M. et al. Paediatric Gastrointestinal Endoscopy: European Society for Paediatric Gastroenterology Hepatology and Nutrition and European Society of Gastrointestinal Endoscopy Guidelines. J Pediatr Gastroenterol Nutr 2017; 64: 133-153
  • 228 Buderus S. Endoscopy and histologic evaluation in chronic inflammatory bowel diseases. Diagnostic importance in pediatrics. Monatsschr Kinderheilkd 2020; 168: 332-343
  • 229 Hauer A. Laboratory diagnostics in chronic inflammatory bowel diseases. Monatsschr Kinderheilkd 2020; 168: 314-322
  • 230 Dabritz J, Musci J, Foell D. Diagnostic utility of faecal biomarkers in patients with irritable bowel syndrome. World J Gastroenterol 2014; 20: 363-375
  • 231 E, N. and S. E. Radiological diagnostics of chronic inflammatory bowel diseases in children and adolescents. Monatsschr Kinderheilkd 2020; 168: 323-331
  • 232 Arndt H. et al. [Imaging of chronic inflammatory bowel diseases in childhood and adolescence: Repetitorium]. Radiologe 2020; 60: 1085-1096
  • 233 Naviglio S. et al. How to predict response to anti-tumour necrosis factor agents in inflammatory bowel disease. Expert Rev Gastroenterol Hepatol 2018; 12: 797-810
  • 234 Cornish JS, Wirthgen E, Dabritz J. Biomarkers Predictive of Response to Thiopurine Therapy in Inflammatory Bowel Disease. Front Med (Lausanne) 2020; 7: 8
  • 235 Barre A, Colombel JF, Ungaro R. Review article: predictors of response to vedolizumab and ustekinumab in inflammatory bowel disease. Aliment Pharmacol Ther 2018; 47: 896-905
  • 236 Musci JO, Cornish JS, Dabritz J. Utility of surrogate markers for the prediction of relapses in inflammatory bowel diseases. J Gastroenterol 2016; 51: 531-547
  • 237 Su C. et al. A meta-analysis of the placebo rates of remission and response in clinical trials of active Crohn’s disease. Gastroenterology 2004; 126: 1257-1269
  • 238 Cholapranee A. et al. Systematic review with meta-analysis: comparative efficacy of biologics for induction and maintenance of mucosal healing in Crohn’s disease and ulcerative colitis controlled trials. Aliment Pharmacol Ther 2017; 45: 1291-1302
  • 239 Froslie KF. et al. Mucosal healing in inflammatory bowel disease: results from a Norwegian population-based cohort. Gastroenterology 2007; 133: 412-422
  • 240 Neurath MF, Travis SP. Mucosal healing in inflammatory bowel diseases: a systematic review. Gut 2012; 61: 1619-1635
  • 241 Ungaro RC. et al. Deep Remission at 1 Year Prevents Progression of Early Crohn’s Disease. Gastroenterology 2020; 159: 139-147
  • 242 Turner D. et al. STRIDE-II: An Update on the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE) Initiative of the International Organization for the Study of IBD (IOIBD): Determining Therapeutic Goals for Treat-to-Target strategies in IBD. Gastroenterology 2021; 160: 1570-1583
  • 243 Best WR. et al. Development of a Crohn’s disease activity index. National Cooperative Crohn’s Disease Study. Gastroenterology 1976; 70: 439-444
  • 244 Thia KT. et al. Defining the optimal response criteria for the Crohn’s disease activity index for induction studies in patients with mildly to moderately active Crohn’s disease. Am J Gastroenterol 2008; 103: 3123-3131
  • 245 Harvey RF, Bradshaw JM. A simple index of Crohn’s-disease activity. Lancet 1980; 1: 514
  • 246 Daperno M. et al. Development and validation of a new, simplified endoscopic activity score for Crohn’s disease: the SES-CD. Gastrointest Endosc 2004; 60: 505-512
  • 247 Khanna R. et al. Reliability among central readers in the evaluation of endoscopic findings from patients with Crohn’s disease. Gut 2016; 65: 1119-1125
  • 248 Ordas I. et al. Accuracy of magnetic resonance enterography in assessing response to therapy and mucosal healing in patients with Crohn’s disease. Gastroenterology 2014; 146: 374-382 e1
  • 249 Panes J. et al. Imaging techniques for assessment of inflammatory bowel disease: joint ECCO and ESGAR evidence-based consensus guidelines. J Crohns Colitis 2013; 7: 556-585
  • 250 Siddiki HA. et al. Prospective comparison of state-of-the-art MR enterography and CT enterography in small-bowel Crohn’s disease. Am J Roentgenol 2009; 193: 113-121
  • 251 Solem CA. et al. Small-bowel imaging in Crohn’s disease: a prospective, blinded, 4-way comparison trial. Gastrointest Endosc 2008; 68: 255-266
  • 252 Hanauer SB. et al Human anti-tumor necrosis factor monoclonal antibody (adalimumab) in Crohn’s disease: the CLASSIC-I trial. Gastroenterology 2006; 130: 323-333 ; quiz 591
  • 253 Jones J. et al. Relationships between disease activity and serum and fecal biomarkers in patients with Crohn’s disease. Clin Gastroenterol Hepatol 2008; 6: 1218-1224
  • 254 Kiss LS. et al. High-sensitivity C-reactive protein for identification of disease phenotype, active disease, and clinical relapses in Crohn’s disease: a marker for patient classification?. Inflamm Bowel Dis 2012; 18: 1647-1654
  • 255 Lamireau T. et al. Efficacy and tolerance of infliximab in children and adolescents with Crohn’s disease. Inflamm Bowel Dis 2004; 10: 745-750
  • 256 Rutgeerts P. et al. Efficacy and safety of retreatment with anti-tumor necrosis factor antibody (infliximab) to maintain remission in Crohn’s disease. Gastroenterology 1999; 117: 761-769
  • 257 Stack WA. et al. Randomised controlled trial of CDP571 antibody to tumour necrosis factor-alpha in Crohn’s disease. Lancet 1997; 349: 521-524
  • 258 Targan SR. et al. A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn’s disease. Crohn’s Disease cA2 Study Group. N Engl J Med 1997; 337: 1029-1035
  • 259 Reinisch W. et al. C-reactive protein, an indicator for maintained response or remission to infliximab in patients with Crohn’s disease: a post-hoc analysis from ACCENT I. Aliment Pharmacol Ther 2012; 35: 568-576
  • 260 Sandborn WJ. et al. Certolizumab pegol for the treatment of Crohn’s disease. N Engl J Med 2007; 357: 228-238
  • 261 Schreiber S. et al. A randomized, placebo-controlled trial of certolizumab pegol (CDP870) for treatment of Crohn’s disease. Gastroenterology 2005; 129: 807-818
  • 262 Yzet C. et al. No Benefit of Concomitant Immunomodulator Therapy on Efficacy of Biologics That Are Not Tumor Necrosis Factor Antagonists in Patients With Inflammatory Bowel Diseases: A Meta-Analysis. Clin Gastroenterol Hepatol 2020; 19: 668-679
  • 263 Singh S. et al. Systematic review and network meta-analysis: first- and second-line biologic therapies for moderate-severe Crohn’s disease. Aliment Pharmacol Ther 2018; 48: 394-409
  • 264 Florin TH. et al. Clinically active Crohn’s disease in the presence of a low C-reactive protein. Scand J Gastroenterol 2006; 41: 306-311
  • 265 De Cruz P. et al. Crohn’s disease management after intestinal resection: a randomised trial. Lancet 2015; 385: 1406-1417
  • 266 Schnitzler F. et al. Mucosal healing predicts long-term outcome of maintenance therapy with infliximab in Crohn’s disease. Inflamm Bowel Dis 2009; 15: 1295-1301
  • 267 Bennett JL. et al. Optimizing perioperative Crohn’s disease management: role of coordinated medical and surgical care. World J Gastroenterol 2015; 21: 1182-1188
  • 268 Calvet X. et al. Delphi consensus statement: Quality Indicators for Inflammatory Bowel Disease Comprehensive Care Units. J Crohns Colitis 2014; 8: 240-251
  • 269 Hibi T. et al. The 5C Concept and 5S Principles in Inflammatory Bowel Disease Management. J Crohns Colitis 2017; 11: 1302-1308
  • 270 Kemp K. et al. Second N-ECCO Consensus Statements on the European Nursing Roles in Caring for Patients with Crohn’s Disease or Ulcerative Colitis. J Crohns Colitis 2018; 12: 760-776
  • 271 Louis E. et al. Optimising the Inflammatory Bowel Disease Unit to Improve Quality of Care: Expert Recommendations. J Crohns Colitis 2015; 9: 685-691
  • 272 Panes J. et al. Improving quality of care in inflammatory bowel disease: what changes can be made today?. J Crohns Colitis 2014; 8: 919-926
  • 273 Ricci C, Lanzarotto F, Lanzini A. The multidisciplinary team for management of inflammatory bowel diseases. Dig Liver Dis 2008; 40 (Suppl. 02) S285-S288
  • 274 Chan WPW, Mourad F, Leong RW. Crohn’s disease associated strictures. J Gastroenterol Hepatol 2018; 33: 998-1008
  • 275 Clancy C. et al. A Meta-analysis of Percutaneous Drainage Versus Surgery as the Initial Treatment of Crohn’s Disease-related Intra-abdominal Abscess. J Crohns Colitis 2016; 10: 202-208
  • 276 de Groof EJ. et al. Abdominal abscess in Crohn’s disease: multidisciplinary management. Dig Dis 2014; 32 (Suppl. 01) 103-109
  • 277 Goldstone RN, Steinhagen RM. Abdominal Emergencies in Inflammatory Bowel Disease. Surg Clin North Am 2019; 99: 1141-1150
  • 278 Mohan HM, Coffey JC. Surgical treatment of intestinal stricture in inflammatory bowel disease. J Dig Dis 2020; 21: 355-359
  • 279 Sahnan K. et al. Medical-surgical Combined Approach in Perianal Fistulizing Crohn’s Disease (CD): Doing it Together. Curr Drug Targets 2019; 20: 1373-1383
  • 280 Shen B. et al. Practical guidelines on endoscopic treatment for Crohn’s disease strictures: a consensus statement from the Global Interventional Inflammatory Bowel Disease Group. Lancet Gastroenterol Hepatol 2020; 5: 393-405
  • 281 Toh JW. et al. Indications and surgical options for small bowel, large bowel and perianal Crohn’s disease. World J Gastroenterol 2016; 22: 8892-8904
  • 282 Lai C. et al. Patient decision-making in severe inflammatory bowel disease: the need for improved communication of treatment options and preferences. Colorectal Dis 2019; 21: 1406-1414
  • 283 Siegel CA. et al. Novel Statistical Approach to Determine Inflammatory Bowel Disease: Patients’ Perspectives on Shared Decision Making. Patient 2016; 9: 79-89
  • 284 Veilleux S. et al. Patients’ perception of their involvement in shared treatment decision making: Key factors in the treatment of inflammatory bowel disease. Patient Educ Couns 2018; 101: 331-339
  • 285 van der Have M. et al. Non-adherence to Anti-TNF Therapy is Associated with Illness Perceptions and Clinical Outcomes in Outpatients with Inflammatory Bowel Disease: Results from a Prospective Multicentre Study. J Crohns Colitis 2016; 10: 549-555
  • 286 Wentworth BJ. et al. Nonadherence to Biologic Therapies in Inflammatory Bowel Disease. Inflamm Bowel Dis 2018; 24: 2053-2061
  • 287 Kennedy NA. et al. Predictors of anti-TNF treatment failure in anti-TNF-naive patients with active luminal Crohn’s disease: a prospective, multicentre, cohort study. Lancet Gastroenterol Hepatol 2019; 4: 341-353
  • 288 Sazonovs A. et al. HLA-DQA1*05 Carriage Associated With Development of Anti-Drug Antibodies to Infliximab and Adalimumab in Patients With Crohn’s Disease. Gastroenterology 2020; 158: 189-199
  • 289 Siegmund B. Cytomegalovirus infection associated with inflammatory bowel disease. Lancet Gastroenterol Hepatol 2017; 2: 369-376
  • 290 Burisch J. et al. The use of 5-aminosalicylate for patients with Crohn’s disease in a prospective European inception cohort with 5 years follow-up – an Epi-IBD study. United European Gastroenterol J 2020; 8: 949-960
  • 291 Torres J. et al. ECCO Guidelines on Therapeutics in Crohn’s Disease: Medical Treatment. J Crohns Colitis 2020; 14: 4-22
  • 292 Lim WC. et al. Aminosalicylates for induction of remission or response in Crohn’s disease. Cochrane Database Syst Rev 2016; 7: CD008870
  • 293 Moja L. et al. Systematic review with network meta-analysis: comparative efficacy and safety of budesonide and mesalazine (mesalamine) for Crohn’s disease. Aliment Pharmacol Ther 2015; 41: 1055-1065
  • 294 Coward S. et al. Comparative Effectiveness of Mesalamine, Sulfasalazine, Corticosteroids, and Budesonide for the Induction of Remission in Crohn’s Disease: A Bayesian Network Meta-analysis: Republished. Inflamm Bowel Dis 2017; 23: E26-E37
  • 295 Hanauer SB, Stromberg U. Oral Pentasa in the treatment of active Crohn’s disease: A meta-analysis of double-blind, placebo-controlled trials. Clin Gastroenterol Hepatol 2004; 2: 379-388
  • 296 Suzuki Y. et al. Efficacy and tolerability of oral budesonide in Japanese patients with active Crohn’s disease: a multicentre, double-blind, randomized, parallel-group Phase II study. J Crohns Colitis 2013; 7: 239-247
  • 297 Tremaine WJ. et al. Budesonide CIR capsules (once or twice daily divided-dose) in active Crohn’s disease: a randomized placebo-controlled study in the United States. Am J Gastroenterol 2002; 97: 1748-1754
  • 298 Rezaie A. et al. Budesonide for induction of remission in Crohn’s disease. Cochrane Database Syst Rev 2015; 6: CD000296
  • 299 Greenberg GR. et al. Oral budesonide for active Crohn’s disease. Canadian Inflammatory Bowel Disease Study Group. N Engl J Med 1994; 331: 836-841
  • 300 Thomsen OO. et al. A comparison of budesonide and mesalamine for active Crohn’s disease. International Budesonide-Mesalamine Study Group. N Engl J Med 1998; 339: 370-374
  • 301 Tromm A. et al Budesonide 9 mg is at least as effective as mesalamine 4.5g in patients with mildly to moderately active Crohn’s disease. Gastroenterology 2011; 140: 425-434 e1 ; quiz e13–e14
  • 302 Yokoyama T. et al. Efficacy and Safety of Oral Budesonide in Patients with Active Crohn’s Disease in Japan: A Multicenter, Double-Blind, Randomized, Parallel-Group Phase 3 Study. Inflamm Intest Dis 2018; 2: 154-162
  • 303 Benchimol EI. et al. Traditional corticosteroids for induction of remission in Crohn’s disease. Cochrane Database Syst Rev 2008; 2: CD006792
  • 304 Malchow H. et al. European Cooperative Crohn’s Disease Study (ECCDS): results of drug treatment. Gastroenterology 1984; 86: 249-266
  • 305 Summers RW. et al. National Cooperative Crohn’s Disease Study: results of drug treatment. Gastroenterology 1979; 77: 847-869
  • 306 Singleton JW. et al. National Cooperative Crohn’s Disease Study: adverse reactions to study drugs. Gastroenterology 1979; 77: 870-882
  • 307 Mottet C. et al. Treatment of gastroduodenal Crohn’s disease. Digestion 2005; 71: 37-40
  • 308 Mowat C. et al. Guidelines for the management of inflammatory bowel disease in adults. Gut 2011; 60: 571-607
  • 309 Alric H. et al. The effectiveness of either ustekinumab or vedolizumab in 239 patients with Crohn’s disease refractory to anti-tumour necrosis factor. Aliment Pharmacol Ther 2020; 51: 948-957
  • 310 Hamdeh S. et al. Early vs Late Use of Anti-TNFa Therapy in Adult Patients With Crohn Disease: A Systematic Review and Meta-Analysis. Inflamm Bowel Dis 2020; 26: 1808-1818
  • 311 Schreiber S. et al. Increased response and remission rates in short-duration Crohn’s disease with subcutaneous certolizumab pegol: an analysis of PRECiSE 2 randomized maintenance trial data. Am J Gastroenterol 2010; 105: 1574-1582
  • 312 Akobeng AK, Zachos M. Tumor necrosis factor-alpha antibody for induction of remission in Crohn’s disease. Cochrane Database Syst Rev 2004; 1: CD003574
  • 313 Behm BW, Bickston SJ. Tumor necrosis factor-alpha antibody for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2008; 1: CD006893
  • 314 Hanauer SB. et al. Maintenance infliximab for Crohn’s disease: the ACCENT I randomised trial. Lancet 2002; 359: 1541-1549
  • 315 Sandborn WJ. et al. Adalimumab induction therapy for Crohn disease previously treated with infliximab: a randomized trial. Ann Intern Med 2007; 146: 829-838
  • 316 Stidham RW. et al. Systematic review with network meta-analysis: the efficacy of anti-TNF agents for the treatment of Crohn’s disease. Aliment Pharmacol Ther 2014; 39: 1349-1362
  • 317 Schreiber S. et al. Noninferiority of novel subcutaneous infliximab (CT-P13) to intravenous infliximab (CT-P13) in patients with active Crohn’s disease and ulcerative colitis: week 30 results from a mutlicrentre, randomised controlled pivital trial. UEGWeek 2019; 7: 1412 ; LB02 (A)
  • 318 Ben-Horin S. et al. OP24 A novel subcutaneous infliximab (CT-P13): 1-year results including switching results from intravenous infliximab (CT-P13) in patients with active Crohn’s disease and ulcerative colitis. Journal of Crohn’s and Colitis 2020; 14: 021-022
  • 319 Matsumoto T. et al. Adalimumab Monotherapy and a Combination with Azathioprine for Crohn’s Disease: A Prospective, Randomized Trial. J Crohns Colitis 2016; 10: 1259-1266
  • 320 Jones JL. et al Effects of Concomitant Immunomodulator Therapy on Efficacy and Safety of Anti-Tumor Necrosis Factor Therapy for Crohn’s Disease: A Meta-analysis of Placebo-controlled Trials. Clin Gastroenterol Hepatol 2015; 13: 2233-2240 e1-2 ; quiz e177–e178
  • 321 Kopylov U. et al. Adalimumab monotherapy versus combination therapy with immunomodulators in patients with Crohn’s disease: a systematic review and meta-analysis. J Crohns Colitis 2014; 8: 1632-1641
  • 322 Feagan BG. et al. Methotrexate in combination with infliximab is no more effective than infliximab alone in patients with Crohn’s disease. Gastroenterology 2014; 146: 681-688 e1
  • 323 Kirchgesner J. et al. Risk of Serious and Opportunistic Infections Associated With Treatment of Inflammatory Bowel Diseases. Gastroenterology 2018; 155: 337-346 e10
  • 324 Marehbian J. et al. Adverse events associated with common therapy regimens for moderate-to-severe Crohn’s disease. Am J Gastroenterol 2009; 104: 2524-2533
  • 325 Feagan BG. et al. Treatment of active Crohn’s disease with MLN0002, a humanized antibody to the alpha4beta7 integrin. Clin Gastroenterol Hepatol 2008; 6: 1370-1377
  • 326 Feagan BG. et al. Ustekinumab as Induction and Maintenance Therapy for Crohn’s Disease. N Engl J Med 2016; 375: 1946-1960
  • 327 MacDonald JK. et al. Anti-IL-12/23p40 antibodies for induction of remission in Crohn’s disease. Cochrane Database Syst Rev 2016; 11: CD007572
  • 328 Sandborn WJ. et al. A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn’s disease. Gastroenterology 2008; 135: 1130-1141
  • 329 Sandborn WJ. et al. Vedolizumab as induction and maintenance therapy for Crohn’s disease. N Engl J Med 2013; 369: 711-721
  • 330 Sandborn WJ. et al. Ustekinumab induction and maintenance therapy in refractory Crohn’s disease. N Engl J Med 2012; 367: 1519-1528
  • 331 Sands BE. et al. Effects of vedolizumab induction therapy for patients with Crohn’s disease in whom tumor necrosis factor antagonist treatment failed. Gastroenterology 2014; 147: 618-627 e3
  • 332 Engel T. et al. Vedolizumab in IBD-Lessons From Real-world Experience; A Systematic Review and Pooled Analysis. J Crohns Colitis 2018; 12: 245-257
  • 333 Engel T. et al. Effectiveness and safety of Ustekinumab for Crohn’s disease; systematic review and pooled analysis of real-world evidence. Dig Liver Dis 2019; 51: 1232-1240
  • 334 Macaluso FS. et al. Effectiveness and safety of Ustekinumab for the treatment of Crohn’s disease in real-life experiences: a meta-analysis of observational studies. Expert Opin Biol Ther 2020; 20: 193-203
  • 335 Schreiber S. et al. Systematic review with meta-analysis: real-world effectiveness and safety of vedolizumab in patients with inflammatory bowel disease. J Gastroenterol 2018; 53: 1048-1064
  • 336 Kawalec P, Mocko P. An indirect comparison of ustekinumab and vedolizumab in the therapy of TNF-failure Crohn’s disease patients. J Comp Eff Res 2018; 7: 101-111
  • 337 Burisch J. et al. Natural disease course of Crohn’s disease during the first 5 years after diagnosis in a European population-based inception cohort: an Epi-IBD study. Gut 2019; 68: 423-433
  • 338 Wong DJ. et al. Surgery in the age of biologics. Gastroenterol Rep (Oxf) 2019; 7: 77-90
  • 339 An V. et al. Early surgery in Crohn’s disease a benefit in selected cases. World J Gastrointest Surg 2016; 8: 492-500
  • 340 Aratari A. et al. Early versus late surgery for ileo-caecal Crohn’s disease. Aliment Pharmacol Ther 2007; 26: 1303-1312
  • 341 de Buck van Overstraeten A. et al. Short- and medium-term outcomes following primary ileocaecal resection for Crohn’s disease in two specialist centres. Br J Surg 2017; 104: 1713-1722
  • 342 Golovics PA. et al. Is early limited surgery associated with a more benign disease course in Crohn’s disease?. World J Gastroenterol 2013; 19: 7701-7710
  • 343 Latella G. et al. Clinical course of Crohn’s disease first diagnosed at surgery for acute abdomen. Dig Liver Dis 2009; 41: 269-276
  • 344 Lee JM. et al. Postoperative course of Crohn disease according to timing of bowel resection: Results from the CONNECT Study. Medicine (Baltimore) 2018; 97: e0459
  • 345 Ryan EJ. et al. Meta-analysis of early bowel resection versus initial medical therapy in patient’s with ileocolonic Crohn’s disease. Int J Colorectal Dis 2020; 35: 501-512
  • 346 Ponsioen CY. et al. Laparoscopic ileocaecal resection versus infliximab for terminal ileitis in Crohn’s disease: a randomised controlled, open-label, multicentre trial. Lancet Gastroenterol Hepatol 2017; 2: 785-792
  • 347 Stevens TW. et al. Laparoscopic ileocaecal resection versus infliximab for terminal ileitis in Crohn’s disease: retrospective long-term follow-up of the LIR!. C trial. Lancet Gastroenterol Hepatol 2020; 5: 900-907
  • 348 Peyrin-Biroulet L. et al. Systematic review: outcomes and post-operative complications following colectomy for ulcerative colitis. Aliment Pharmacol Ther 2016; 44: 807-816
  • 349 Patel KV. et al. Patient optimization for surgery relating to Crohn’s disease. Nat Rev Gastroenterol Hepatol 2016; 13: 707-719
  • 350 Brennan GT. et al. Does preoperative enteral or parenteral nutrition reduce postoperative complications in Crohn’s disease patients: a meta-analysis. Eur J Gastroenterol Hepatol 2018; 30: 997-1002
  • 351 Bemelman WA. et al. ECCO-ESCP Consensus on Surgery for Crohn’s Disease. J Crohns Colitis 2018; 12: 1-16
  • 352 Gionchetti P. et al. 3rd European Evidence-based Consensus on the Diagnosis and Management of Crohn’s Disease 2016: Part 2: Surgical Management and Special Situations. J Crohns Colitis 2017; 11: 135-149
  • 353 Subramanian V. et al. Preoperative steroid use and risk of postoperative complications in patients with inflammatory bowel disease undergoing abdominal surgery. Am J Gastroenterol 2008; 103: 2373-2381
  • 354 Huang W. et al. Risk factors for postoperative intra-abdominal septic complications after surgery in Crohn’s disease: A meta-analysis of observational studies. J Crohns Colitis 2015; 9: 293-301
  • 355 Lightner AL, Shen B. Perioperative use of immunosuppressive medications in patients with Crohn’s disease in the new “biological era”. Gastroenterol Rep (Oxf) 2017; 5: 165-177
  • 356 Serradori T. et al. The effect of immune therapy on surgical site infection following Crohn’s Disease resection. Br J Surg 2013; 100: 1089-1093
  • 357 Aberra FN. et al. Corticosteroids and immunomodulators: postoperative infectious complication risk in inflammatory bowel disease patients. Gastroenterology 2003; 125: 320-327
  • 358 Rizzo G. et al. Anti-TNF-alpha therapies do not increase early postoperative complications in patients with inflammatory bowel disease. An Italian single-center experience. Int J Colorectal Dis 2011; 26: 1435-1444
  • 359 Xu Y. et al. Meta-Analysis: The Influence of Preoperative Infliximab Use on Postoperative Complications of Crohn’s Disease. Inflamm Bowel Dis 2019; 25: 261-269
  • 360 Brouquet A. et al. Anti-TNF Therapy Is Associated With an Increased Risk of Postoperative Morbidity After Surgery for Ileocolonic Crohn Disease: Results of a Prospective Nationwide Cohort. Ann Surg 2018; 267: 221-228
  • 361 Lin YS. et al. Systematic review with meta-analysis: risk of post-operative complications associated with pre-operative exposure to anti-tumour necrosis factor agents for Crohn’s disease. Aliment Pharmacol Ther 2019; 49: 966-977
  • 362 Cohen B. et al. 415a – Anti-Tumor Necrosis Factor Therapy is Not Associated with Post-Operative Infection: Results from Prospective Cohort of Ulcerative Colitis and Crohn’s Disease Patients Undergoing Surgery to Identify Risk Factors for Postoperative Infection I (Puccini). Gastroenterology 2019; 156: 80
  • 363 Soop M, Hancock L, Davies J. Anti-TNF therapy before intestinal surgery for Crohn’s disease and the risks of postoperative complications. J Crohns Colitis 2021; 15: 1777
  • 364 Lightner AL. et al. Postoperative Outcomes in Vedolizumab-Treated Patients Undergoing Major Abdominal Operations for Inflammatory Bowel Disease: Retrospective Multicenter Cohort Study. Inflamm Bowel Dis 2018; 24: 871-876
  • 365 Law CCY. et al. Systematic Review and Meta-Analysis: Preoperative Vedolizumab Treatment and Postoperative Complications in Patients with Inflammatory Bowel Disease. J Crohns Colitis 2018; 12: 538-545
  • 366 Novello M. et al. Case-matched Comparison of Postoperative Outcomes Following Surgery for Inflammatory Bowel Disease After Exposure to Vedolizumab vs Other Biologics. J Crohns Colitis 2020; 14: 185-191
  • 367 Shim HH. et al. Preoperative Ustekinumab Treatment Is Not Associated With Increased Postoperative Complications in Crohn’s Disease: A Canadian Multi-Centre Observational Cohort Study. J Can Assoc Gastroenterol 2018; 1: 115-123
  • 368 Lightner AL. et al. Postoperative Outcomes in Ustekinumab-Treated Patients Undergoing Abdominal Operations for Crohn’s Disease. J Crohns Colitis 2018; 12: 402-407
  • 369 Chande N. et al. Azathioprine or 6-mercaptopurine for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2015; 10: CD000067
  • 370 Chande N. et al. Azathioprine or 6-mercaptopurine for induction of remission in Crohn’s disease. Cochrane Database Syst Rev 2016; 10: CD000545
  • 371 Bastida Paz G. et al. [Optimization of immunomodulatory treatment with azathioprine or 6-mercaptopurine in inflammatory bowel disease]. Gastroenterol Hepatol 2007; 30: 511-516
  • 372 Feagan BG. et al. A comparison of methotrexate with placebo for the maintenance of remission in Crohn’s disease. North American Crohn’s Study Group Investigators. N Engl J Med 2000; 342: 1627-1632
  • 373 Feagan BG. et al. Methotrexate for the treatment of Crohn’s disease. The North American Crohn’s Study Group Investigators. N Engl J Med 1995; 332: 292-297
  • 374 McDonald JW. et al. Methotrexate for induction of remission in refractory Crohn’s disease. Cochrane Database Syst Rev 2014; 8: CD003459
  • 375 Patel V. et al. Methotrexate for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2014; 8: CD006884
  • 376 Cohen-Dolev N. et al. Differences in Outcomes Over Time With Exclusive Enteral Nutrition Compared With Steroids in Children With Mild to Moderate Crohn’s Disease: Results From the GROWTH CD Study. J Crohns Colitis 2018; 12: 306-312
  • 377 Shamir R, Phillip M, Levine A. Growth retardation in pediatric Crohn’s disease: pathogenesis and interventions. Inflamm Bowel Dis 2007; 13: 620-628
  • 378 Navarro FA, Hanauer SB, Kirschner BS. Effect of long-term low-dose prednisone on height velocity and disease activity in pediatric and adolescent patients with Crohn disease. J Pediatr Gastroenterol Nutr 2007; 45: 312-318
  • 379 Escher JC. I.B.D. European Collaborative Research Group on Budesonide in Paediatric. Budesonide versus prednisolone for the treatment of active Crohn’s disease in children: a randomized, double-blind, controlled, multicentre trial. Eur J Gastroenterol Hepatol 2004; 16: 47-54
  • 380 Levine A. et al. A comparison of budesonide and prednisone for the treatment of active pediatric Crohn disease. J Pediatr Gastroenterol Nutr 2003; 36: 248-252
  • 381 Heuschkel RB. et al. Enteral nutrition and corticosteroids in the treatment of acute Crohn’s disease in children. J Pediatr Gastroenterol Nutr 2000; 31: 8-15
  • 382 Yu Y, Chen KC, Chen J. Exclusive enteral nutrition versus corticosteroids for treatment of pediatric Crohn’s disease: a meta-analysis. World J Pediatr 2019; 15: 26-36
  • 383 Borrelli O. et al. Polymeric diet alone versus corticosteroids in the treatment of active pediatric Crohn’s disease: a randomized controlled open-label trial. Clin Gastroenterol Hepatol 2006; 4: 744-753
  • 384 Cameron FL. et al. Clinical progress in the two years following a course of exclusive enteral nutrition in 109 paediatric patients with Crohn’s disease. Aliment Pharmacol Ther 2013; 37: 622-629
  • 385 Frivolt K. et al. Repeated exclusive enteral nutrition in the treatment of paediatric Crohn’s disease: predictors of efficacy and outcome. Aliment Pharmacol Ther 2014; 39: 1398-1407
  • 386 Connors J. et al. Exclusive Enteral Nutrition Therapy in Paediatric Crohn’s Disease Results in Long-term Avoidance of Corticosteroids: Results of a Propensity-score Matched Cohort Analysis. J Crohns Colitis 2017; 11: 1063-1070
  • 387 Werkstetter KJ. et al. Influence of exclusive enteral nutrition therapy on bone density and geometry in newly diagnosed pediatric Crohn’s disease patients. Ann Nutr Metab 2013; 63: 10-16
  • 388 Day AS. et al. Exclusive enteral feeding as primary therapy for Crohn’s disease in Australian children and adolescents: a feasible and effective approach. J Gastroenterol Hepatol 2006; 21: 1609-1614
  • 389 Rubio A. et al. The efficacy of exclusive nutritional therapy in paediatric Crohn’s disease, comparing fractionated oral vs. continuous enteral feeding. Aliment Pharmacol Ther 2011; 33: 1332-1339
  • 390 Swaminath A. et al. Systematic review with meta-analysis: enteral nutrition therapy for the induction of remission in paediatric Crohn’s disease. Aliment Pharmacol Ther 2017; 46: 645-656
  • 391 Heerasing N. et al. Exclusive enteral nutrition provides an effective bridge to safer interval elective surgery for adults with Crohn’s disease. Aliment Pharmacol Ther 2017; 45: 660-669
  • 392 Sharma S. et al. Efficacy and tolerability of exclusive enteral nutrition in adult patients with complicated Crohn’s disease. Intest Res 2020; 19: 291-300
  • 393 Yang Q. et al. Efficacy of exclusive enteral nutrition in complicated Crohn’s disease. Scand J Gastroenterol 2017; 52: 995-1001
  • 394 Markowitz J. et al. A multicenter trial of 6-mercaptopurine and prednisone in children with newly diagnosed Crohn’s disease. Gastroenterology 2000; 119: 895-902
  • 395 Colman RJ. et al. Methotrexate for the Treatment of Pediatric Crohn’s Disease: A Systematic Review and Meta-analysis. Inflamm Bowel Dis 2018; 24: 2135-2141
  • 396 Hyams J. et al Induction and maintenance infliximab therapy for the treatment of moderate-to-severe Crohn’s disease in children. Gastroenterology 2007; 132: 863-873 ; quiz 1165–1166
  • 397 Pinto Pais I. et al. Optimizing Antitumor Necrosis Factor Treatment in Pediatric Inflammatory Bowel Disease With Therapeutic Drug Monitoring. J Pediatr Gastroenterol Nutr 2020; 71: 12-18
  • 398 Winter DA. et al. Pharmacokinetics, Pharmacodynamics, and Immunogenicity of Infliximab in Pediatric Inflammatory Bowel Disease: A Systematic Review and Revised Dosing Considerations. J Pediatr Gastroenterol Nutr 2020; 70: 763-776
  • 399 Joosse ME. et al. Malignancy and mortality in paediatric-onset inflammatory bowel disease: a 3-year prospective, multinational study from the paediatric IBD Porto group of ESPGHAN. Aliment Pharmacol Ther 2018; 48: 523-537
  • 400 Newby EA. et al. Interventions for growth failure in childhood Crohn’s disease. Cochrane Database Syst Rev 2005; 3: CD003873
  • 401 Singh Ranger G. et al. Surgery results in significant improvement in growth in children with Crohn’s disease refractory to medical therapy. Pediatr Surg Int 2006; 22: 347-352
  • 402 Munkholm P. et al. Disease activity courses in a regional cohort of Crohn’s disease patients. Scand J Gastroenterol 1995; 30: 699-706
  • 403 Henriksen M. et al. Clinical course in Crohn’s disease: results of a five-year population-based follow-up study (the IBSEN study). Scand J Gastroenterol 2007; 42: 602-610
  • 404 Solberg IC. et al. Clinical course in Crohn’s disease: results of a Norwegian population-based ten-year follow-up study. Clin Gastroenterol Hepatol 2007; 5: 1430-1438
  • 405 Beaugerie L. et al. Predictors of Crohn’s disease. Gastroenterology 2006; 130: 650-656
  • 406 Allez M. et al. Long term outcome of patients with active Crohn’s disease exhibiting extensive and deep ulcerations at colonoscopy. Am J Gastroenterol 2002; 97: 947-953
  • 407 Allez M, Lemann M. Role of endoscopy in predicting the disease course in inflammatory bowel disease. World J Gastroenterol 2010; 16: 2626-2632
  • 408 Wilson B, Lonnfors S, Hommes DW. et al. A European Crohn’s and ulcerative colitis patient life IMPACT survey. JCC 2012; 6 (Suppl. 01) S171
  • 409 Bokemeyer B, Katalinic A, Klugmann T. et al. Predictive factors for a mild course of Crohn’s disease. Journal of Crohn’s and Colitis 2009; 3: S82-S83
  • 410 Hauser W. et al. Anxiety and depression in patients with inflammatory bowel disease: comparisons with chronic liver disease patients and the general population. Inflamm Bowel Dis 2011; 17: 621-632
  • 411 Takagi S. et al. Effectiveness of an ‘half elemental diet’ as maintenance therapy for Crohn’s disease: A randomized-controlled trial. Aliment Pharmacol Ther 2006; 24: 1333-1340
  • 412 Wilschanski M. et al. Supplementary enteral nutrition maintains remission in paediatric Crohn’s disease. Gut 1996; 38: 543-548
  • 413 Mahid SS. et al. Active and passive smoking in childhood is related to the development of inflammatory bowel disease. Inflamm Bowel Dis 2007; 13: 431-438
  • 414 Mahid SS. et al. Smoking and inflammatory bowel disease: a meta-analysis. Mayo Clin Proc 2006; 81: 1462-1471
  • 415 Timmer A, Sutherland LR, Martin F. Oral contraceptive use and smoking are risk factors for relapse in Crohn’s disease. The Canadian Mesalamine for Remission of Crohn’s Disease Study Group. Gastroenterology 1998; 114: 1143-1150
  • 416 Kane SV, Flicker M, Katz-Nelson F. Tobacco use is associated with accelerated clinical recurrence of Crohn’s disease after surgically induced remission. J Clin Gastroenterol 2005; 39: 32-35
  • 417 Johnson GJ, Cosnes J, Mansfield JC. Review article: smoking cessation as primary therapy to modify the course of Crohn’s disease. Aliment Pharmacol Ther 2005; 21: 921-931
  • 418 Cosnes J. et al. Effects of cigarette smoking on the long-term course of Crohn’s disease. Gastroenterology 1996; 110: 424-431
  • 419 Breuer-Katschinski BD, Hollander N, Goebell H. Effect of cigarette smoking on the course of Crohn’s disease. Eur J Gastroenterol Hepatol 1996; 8: 225-228
  • 420 Steinhart AH. et al. Corticosteroids for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2003; 4: CD000301
  • 421 Smith RC. et al. Low dose steroids and clinical relapse in Crohn’s disease: a controlled trial. Gut 1978; 19: 606-610
  • 422 Steinhart H. Maintenance therapy in Crohn’s disease. Can J Gastroenterol 2000; 14: 23C-28C
  • 423 Benchimol EI. et al. Budesonide for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2009; 1: CD002913
  • 424 Prefontaine E. et al. Azathioprine or 6-mercaptopurine for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2009; 1: CD000067
  • 425 Lemann M. et al. A randomized, double-blind, controlled withdrawal trial in Crohn’s disease patients in long-term remission on azathioprine. Gastroenterology 2005; 128: 1812-1818
  • 426 O’Donoghue DP. et al. Double-blind withdrawal trial of azathioprine as maintenance treatment for Crohn’s disease. Lancet 1978; 2: 955-957
  • 427 Panes J. et al. Early azathioprine therapy is no more effective than placebo for newly diagnosed Crohn’s disease. Gastroenterology 2013; 145: 766-774 e1
  • 428 Cosnes J. et al Early administration of azathioprine vs conventional management of Crohn’s Disease: a randomized controlled trial. Gastroenterology 2013; 145: 758-765 e2 ; quiz e14–e15
  • 429 Patel V. et al. Methotrexate for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2009; 4: CD006884
  • 430 Mate-Jimenez J. et al. 6-mercaptopurine or methotrexate added to prednisone induces and maintains remission in steroid-dependent inflammatory bowel disease. Eur J Gastroenterol Hepatol 2000; 12: 1227-1233
  • 431 Oren R. et al. Methotrexate in chronic active Crohn’s disease: a double-blind, randomized, Israeli multicenter trial. Am J Gastroenterol 1997; 92: 2203-2209
  • 432 Peyrin-Biroulet L. et al. Efficacy and safety of tumor necrosis factor antagonists in Crohn’s disease: meta-analysis of placebo-controlled trials. Clin Gastroenterol Hepatol 2008; 6: 644-653
  • 433 Ford AC. et al Efficacy of biological therapies in inflammatory bowel disease: systematic review and meta-analysis. Am J Gastroenterol 2011; 106: 644-659 , quiz 660
  • 434 Sandborn WJ. et al. Efficacy and Safety of Vedolizumab Subcutaneous Formulation in a Randomized Trial of Patients With Ulcerative Colitis. Gastroenterology 2020; 158: 562-572 e12
  • 435 Hupe M. et al. Comparative efficacy and safety of vedolizumab and infliximab in ulcerative colitis after failure of a first subcutaneous anti-TNF agent: a multicentre cohort study. Aliment Pharmacol Ther 2020; 51: 852-860
  • 436 Parigi TL. et al. Evolution of infliximab biosimilar in inflammatory bowel disease: from intravenous to subcutaneous CT-P13. Expert Opin Biol Ther 2020; 21: 1-10
  • 437 Chen C. et al. DOP16 An evaluation of the exposure–efficacy relationship for subcutaneous vedolizumab maintenance treatment of Crohn’s disease: Pharmacokinetic findings from VISIBLE 2. Journal of Crohn’s and Colitis 2020; 14: 056-057
  • 438 European Medicines Agency: Guideline on similar biological medicinal products. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2014/10/WC500176768.pdf , Oct23, 2014.
  • 439 Park W. et al. A randomised, double-blind, multicentre, parallel-group, prospective study comparing the pharmacokinetics, safety, and efficacy of CT-P13 and innovator infliximab in patients with ankylosing spondylitis: the PLANETAS study. Ann Rheum Dis 2013; 72: 1605-1612
  • 440 Yoo DH. et al. A randomised, double-blind, parallel-group study to demonstrate equivalence in efficacy and safety of CT-P13 compared with innovator infliximab when coadministered with methotrexate in patients with active rheumatoid arthritis: the PLANETRA study. Ann Rheum Dis 2013; 72: 1613-1620
  • 441 Ye BD. et al. Efficacy and safety of biosimilar CT-P13 compared with originator infliximab in patients with active Crohn’s disease: an international, randomised, double-blind, phase 3 non-inferiority study. Lancet 2019; 393: 1699-1707
  • 442 Jorgensen KK. et al. Switching from originator infliximab to biosimilar CT-P13 compared with maintained treatment with originator infliximab (NOR-SWITCH): a 52-week, randomised, double-blind, non-inferiority trial. Lancet 2017; 389: 2304-2316
  • 443 Numan S, Faccin F. Non-medical Switching from Originator Tumor Necrosis Factor Inhibitors to Their Biosimilars: Systematic Review of Randomized Controlled Trials and Real-World Studies. Adv Ther 2018; 35: 1295-1332
  • 444 Luber RP. et al. Sa1882 SWITCHING INFLIXIMAB BIOSIMILAR: NO ADVERSE IMPACT ON IBD CONTROL OR DRUG LEVELS WITH FIRST OR SECOND SWITCH. Gastroenterology 2020; 158: 464
  • 445 Blauvelt A. et al. Phase III randomized study of the proposed adalimumab biosimilar GP2017 in psoriasis: impact of multiple switches. Br J Dermatol 2018; 179: 623-631
  • 446 Kruis W. et al Predictive factors for an uncomplicated long-term course of Crohn’s disease: A retrospective analysis. J Crohns Colitis 2012; PMID:23182164. 7: 1263-1270
  • 447 Bokemeyer B. et al. Clinical status, psychosocial impairments, medical treatment and health care costs for patients with inflammatory bowel disease (IBD) in Germany: An online IBD registry. J Crohns Colitis 2013; 7: 355-368
  • 448 Akobeng AK, Thomas AG. Enteral nutrition for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2007; 3: CD005984
  • 449 Steinhart AH. et al. Systematic review: the potential influence of mesalazine formulation on maintenance of remission in Crohn’s disease. Aliment Pharmacol Ther 2007; 25: 1389-1399
  • 450 Ford AC. et al. Efficacy of 5-aminosalicylates in Crohn’s disease: systematic review and meta-analysis. Am J Gastroenterol 2011; 106: 617-629
  • 451 Akobeng AK. et al. Oral 5-aminosalicylic acid for maintenance of medically-induced remission in Crohn’s disease. Cochrane Database Syst Rev 2016; 9: CD003715
  • 452 National Institute for Health and Clinical Excellence, N.C.G.C., Crohn’s disease. Management in adults and children. 2012
  • 453 Duricova D. et al. 5-aminosalicyclic acid dependency in Crohn’s disease: a Danish Colitis Database study. J Crohns Colitis 2010; 4: 575-581
  • 454 Pariente B. et al. Development of the Crohn’s disease digestive damage score, the Lemann score. Inflamm Bowel Dis 2011; 17: 1415-1422
  • 455 Kamm MA. Review article: chronic active disease and maintaining remission in Crohn’s disease. Aliment Pharmacol Ther 2004; 20 (Suppl. 04) 102-105
  • 456 Ludwig D, Stange EF. Efficacy of azathioprine in the treatment of chronic active Crohn’s disease: prospective one-year follow-up study. German Imurek Study Group. Z Gastroenterol 1999; 37: 1085-1091
  • 457 Hanauer SB. et al. Maintenance infliximab for Crohn’s disease: the ACCENT I randomised trial. Lancet 2002; 359: 1541-1549
  • 458 Colombel JF. et al. Adalimumab for maintenance of clinical response and remission in patients with Crohn’s disease: the CHARM trial. Gastroenterology 2007; 132: 52-65
  • 459 D’Haens GR. et al The London Position Statement of the World Congress of Gastroenterology on Biological Therapy for IBD with the European Crohn’s and Colitis Organization: when to start, when to stop, which drug to choose, and how to predict response?. Am J Gastroenterol 2011; 106: 199-212 ; quiz 213
  • 460 Hindorf U. et al. Mercaptopurine treatment should be considered in azathioprine intolerant patients with inflammatory bowel disease. Aliment Pharmacol Ther 2009; 29: 654-661
  • 461 Bokemeyer B. et al. Adherence to thiopurine treatment in out-patients with Crohn’s disease. Aliment Pharmacol Ther 2007; 26: 217-225
  • 462 Thomsen SB. et al. Outcome of concomitant treatment with thiopurines and allopurinol in patients with inflammatory bowel disease: A nationwide Danish cohort study. United European Gastroenterol J 2020; 8: 68-76
  • 463 Chavoushi SF. et al. Thiopurines with low-dose allopurinol (ThiLDA)-a prospective clinical one-way crossover trial. Eur J Clin Pharmacol 2019; 75: 1669-1674
  • 464 Friedman AB. et al. Randomised clinical trial: efficacy, safety and dosage of adjunctive allopurinol in azathioprine/mercaptopurine nonresponders (AAA Study). Aliment Pharmacol Ther 2018; 47: 1092-1102
  • 465 Reinshagen M. et al. 6-thioguanine nucleotide-adapted azathioprine therapy does not lead to higher remission rates than standard therapy in chronic active crohn disease: results from a randomized, controlled, open trial. Clin Chem 2007; 53: 1306-1314
  • 466 Colombel JF. et al. Infliximab, azathioprine, or combination therapy for Crohn’s disease. N Engl J Med 2010; 362: 1383-1395
  • 467 Boyapati RK. et al. Withdrawal of immunosuppressant or biologic therapy for patients with quiescent Crohn’s disease. Cochrane Database Syst Rev 2018; 5: CD012540
  • 468 Roblin X. et al. Azathioprine dose reduction in inflammatory bowel disease patients on combination therapy: an open-label, prospective and randomised clinical trial. Aliment Pharmacol Ther 2017; 46: 142-149
  • 469 Van Assche G. et al. Withdrawal of immunosuppression in Crohn’s disease treated with scheduled infliximab maintenance: a randomized trial. Gastroenterology 2008; 134: 1861-1868
  • 470 Chalhoub JM. et al. Systematic Review and Meta-analysis: Adalimumab Monotherapy Versus Combination Therapy with Immunomodulators for Induction and Maintenance of Remission and Response in Patients with Crohn’s Disease. Inflamm Bowel Dis 2017; 23: 1316-1327
  • 471 Rutgeerts P. et al Scheduled maintenance treatment with infliximab is superior to episodic treatment for the healing of mucosal ulceration associated with Crohn’s disease. Gastrointest Endosc 2006; 63: 433-442 ; quiz 464
  • 472 Beaugerie L. et al. Lymphoproliferative disorders in patients receiving thiopurines for inflammatory bowel disease: a prospective observational cohort study. Lancet 2009; 374: 1617-1625
  • 473 Taylor NS. et al. The impact of an inflammatory bowel disease nurse-led biologics service. Frontline Gastroenterol 2016; 7: 283-288
  • 474 López-Parra M. et al. A review of nursing consultancy to control patients with inflammatory bowel disease treated with thiopurines: Analysis and implementation of improvements. Journal of Crohn’s and Colitis 2014; 8: 358
  • 475 Belling R, McLaren S, Woods L. Specialist nursing interventions for inflammatory bowel disease. Cochrane Database Syst Rev 2009; 4: CD006597
  • 476 Hernandez-Sampelayo P. et al. Contribution of nurses to the quality of care in management of inflammatory bowel disease: a synthesis of the evidence. J Crohns Colitis 2010; 4: 611-622
  • 477 Price T, Lithgo K, Johnsson M. Cost savings and outpatient clinic appointments saved: A 2 year review of a nurse led telephone advice line for inflammatory bowel disease. gut 2013; 62: A34
  • 478 Mukherjee S, Sloper P, Lewin R. The meaning of parental illness to children: the case of inflammatory bowel disease. Child Care Health Dev 2002; 28: 479-485
  • 479 Andersson G, Marsden E. Audit of an inflammatory bowel disease telephone helpline set up and managed by IBD specialist nurses. Gastroinestinal Nursing 2012; 10: 24-30
  • 480 Hueppe A, Langbrandtner J, Raspe H. Inviting patients with inflammatory bowel disease to active involvement in their own care: a randomized controlled trial. Inflammatory bowel diseases 2014; 20: 1057-1069
  • 481 Bager P, Hentze R, Markussen T. IBD patients in remission strongly prefer annual telephone calls by IBD nurses compared to outpatient visits. Inflammatory bowel diseases 2011; 5: 175
  • 482 Sack C. et al. A chronic care model significantly decreases costs and healthcare utilisation in patients with inflammatory bowel disease. J Crohns Colitis 2012; 6: 302-310
  • 483 Huppe A. et al. The Effectiveness of Actively Induced Medical Rehabilitation in Chronic Inflammatory Bowel Disease. Dtsch Arztebl Int 2020; 117: 89-96
  • 484 Treton X. et al. Azathioprine withdrawal in patients with Crohn’s disease maintained on prolonged remission: a high risk of relapse. Clin Gastroenterol Hepatol 2009; 7: 80-85
  • 485 Kandiel A. et al. Increased risk of lymphoma among inflammatory bowel disease patients treated with azathioprine and 6-mercaptopurine. Gut 2005; 54: 1121-1125
  • 486 Te HS. et al. Hepatic effects of long-term methotrexate use in the treatment of inflammatory bowel disease. Am J Gastroenterol 2000; 95: 3150-3156
  • 487 Louis E. et al Maintenance of remission among patients with Crohn’s disease on antimetabolite therapy after infliximab therapy is stopped. Gastroenterology 2012; 142: 63-70 e5 ; quiz e31
  • 488 Long MD, Kappelman MD, Pipkin CA. Nonmelanoma skin cancer in inflammatory bowel disease: a review. Inflamm Bowel Dis 2011; 17: 1423-1427
  • 489 Maddox JS, Soltani K. Risk of nonmelanoma skin cancer with azathioprine use. Inflamm Bowel Dis 2008; 14: 1425-1431
  • 490 Long MD. et al. Risk of melanoma and nonmelanoma skin cancer among patients with inflammatory bowel disease. Gastroenterology 2012; 143: 390-399 e1
  • 491 Kennedy ED. et al. Do patients consider postoperative maintenance therapy for Crohn’s disease worthwhile?. Inflamm Bowel Dis 2008; 14: 224-235
  • 492 Caprilli R, Taddei G, Viscido A. In favour of prophylactic treatment for post-operative recurrence in Crohn’s disease. Ital J Gastroenterol Hepatol 1998; 30: 219-225
  • 493 Breslin NP, Sutherland LR. The case against routine post-operative therapy for prevention of recurrence in Crohn’s disease. Ital J Gastroenterol Hepatol 1998; 30: 226-230
  • 494 Froehlich F. et al. Maintenance of surgically induced remission of Crohn’s disease. Digestion 2007; 76: 130-135
  • 495 Auzolle C. et al. Male gender, active smoking and previous intestinal resection are risk factors for post-operative endoscopic recurrence in Crohn’s disease: results from a prospective cohort study. Aliment Pharmacol Ther 2018; 48: 924-932
  • 496 Swoger JM, Regueiro M. Preventive therapy in postoperative Crohn’s disease. Curr Opin Gastroenterol 2010; 26: 337-343
  • 497 Ng SC, Kamm MA. Management of postoperative Crohn’s disease. Am J Gastroenterol 2008; 103: 1029-1035
  • 498 Sutherland LR, Steinhart AH. Mesalazine as a maintenance treatment in Crohn’s disease. Gut 1998; 42: 143-144
  • 499 Akobeng AK. Review article: the evidence base for interventions used to maintain remission in Crohn’s disease. Aliment Pharmacol Ther 2008; 27: 11-18
  • 500 Ford AC. et al. 5-aminosalicylates prevent relapse of Crohn’s disease after surgically induced remission: systematic review and meta-analysis. Am J Gastroenterol 2011; 106: 413-420
  • 501 Yang Z. et al. A network meta-analysis on the efficacy of 5-aminosalicylates, immunomodulators and biologics for the prevention of postoperative recurrence in Crohn’s disease. Int J Surg 2014; 12: 516-522
  • 502 Singh S. et al Comparative efficacy of pharmacologic interventions in preventing relapse of Crohn’s disease after surgery: a systematic review and network meta-analysis. Gastroenterology 2015; 148: 64-76 e2 ; quiz e14
  • 503 Burr NE. et al. Systematic Review and Network Meta-Analysis of Medical Therapies to Prevent Recurrence of Post-Operative Crohn’s Disease. J Crohns Colitis 2019; 13: 693-701
  • 504 Feng JS. et al. Strategies for Preventing Endoscopic Recurrence of Crohn’s Disease 1 Year after Surgery: A Network Meta-Analysis. Gastroenterol Res Pract 2017; 2017: 7896160
  • 505 Gjuladin-Hellon T. et al. Oral 5-aminosalicylic acid for maintenance of surgically-induced remission in Crohn’s disease. Cochrane Database Syst Rev 2019; 6: CD008414
  • 506 Hanauer SB. et al. Postoperative maintenance of Crohn’s disease remission with 6-mercaptopurine, mesalamine, or placebo: a 2-year trial. Gastroenterology 2004; 127: 723-729
  • 507 Ardizzone S. et al. Azathioprine and mesalamine for prevention of relapse after conservative surgery for Crohn’s disease. Gastroenterology 2004; 127: 730-740
  • 508 De Cruz P. et al. Efficacy of thiopurines and adalimumab in preventing Crohn’s disease recurrence in high-risk patients – a POCER study analysis. Aliment Pharmacol Ther 2015; 42: 867-879
  • 509 Bakouny Z. et al. Comparative Efficacy of Anti-TNF Therapies For The Prevention of Postoperative Recurrence of Crohn’s Disease: A Systematic Review and Network Meta-Analysis of Prospective Trials. J Clin Gastroenterol 2019; 53: 409-417
  • 510 Kaimakliotis P. et al. A Systematic Review Assessing Medical Treatment for Rectovaginal and Enterovesical Fistulae in Crohn’s Disease. J Clin Gastroenterol 2016; 50: 714-721
  • 511 Hotouras A. et al. Gracilis muscle interposition for rectovaginal and anovaginal fistula repair: a systematic literature review. Colorectal Dis 2015; 17: 104-110
  • 512 Adamina M. et al. ECCO Guidelines on Therapeutics in Crohn’s Disease: Surgical Treatment. J Crohns Colitis 2020; 14: 155-168
  • 513 Present DH. et al. Infliximab for the treatment of fistulas in patients with Crohn’s disease. N Engl J Med 1999; 340: 1398-1405
  • 514 Sands BE. et al. Infliximab maintenance therapy for fistulizing Crohn’s disease. N Engl J Med 2004; 350: 876-885
  • 515 Yarur AJ. et al. Higher infliximab trough levels are associated with perianal fistula healing in patients with Crohn’s disease. Aliment Pharmacol Ther 2017; 45: 933-940
  • 516 Colombel JF. et al. Adalimumab for the treatment of fistulas in patients with Crohn’s disease. Gut 2009; 58: 940-948
  • 517 Lichtiger S. et al. The CHOICE trial: adalimumab demonstrates safety, fistula healing, improved quality of life and increased work productivity in patients with Crohn’s disease who failed prior infliximab therapy. Aliment Pharmacol Ther 2010; 32: 1228-1239
  • 518 Thia KT. et al. Ciprofloxacin or metronidazole for the treatment of perianal fistulas in patients with Crohn’s disease: a randomized, double-blind, placebo-controlled pilot study. Inflamm Bowel Dis 2009; 15: 17-24
  • 519 Dewint P. et al. Adalimumab combined with ciprofloxacin is superior to adalimumab monotherapy in perianal fistula closure in Crohn’s disease: a randomised, double-blind, placebo controlled trial (ADAFI). Gut 2014; 63: 292-299
  • 520 Khan KJ. et al. Antibiotic therapy in inflammatory bowel disease: a systematic review and meta-analysis. Am J Gastroenterol 2011; 106: 661-673
  • 521 Feagan BG. et al. Efficacy of Vedolizumab in Fistulising Crohn’s Disease: Exploratory Analyses of Data from GEMINI 2. J Crohns Colitis 2018; 12: 621-626
  • 522 Chapuis-Biron C. et al. Vedolizumab for perianal Crohn’s disease: a multicentre cohort study in 151 patients. Aliment Pharmacol Ther 2020; 51: 719-727
  • 523 Sands BE. et al. Fistula Healing in Pivotal Studies of Ustekinumab in Crohn’s Disease. Gastroenterology 2017; 152: S185
  • 524 Chapuis-Biron C. et al. Ustekinumab for Perianal Crohn’s Disease: The BioLAP Multicenter Study From the GETAID. Am J Gastroenterol 2020; 115: 1812-1820
  • 525 Sonnenberg A, Gavin MW. Timing of surgery for enterovesical fistula in Crohn’s disease: decision analysis using a time-dependent compartment model. Inflamm Bowel Dis 2000; 6: 280-285
  • 526 Taxonera C. et al. Outcomes of Medical and Surgical Therapy for Entero-urinary Fistulas in Crohn’s Disease. J Crohns Colitis 2016; 10: 657-662
  • 527 Zhang W. et al. The respective role of medical and surgical therapy for enterovesical fistula in Crohn’s disease. J Clin Gastroenterol 2014; 48: 708-711
  • 528 Amiot A. et al. Long-term outcome of enterocutaneous fistula in patients with Crohn’s disease treated with anti-TNF therapy: a cohort study from the GETAID. Am J Gastroenterol 2014; 109: 1443-1449
  • 529 Buhr HJ, Kroesen AJ, Herfarth C. [Surgical therapy of recurrent Crohn disease]. Chirurg 1995; 66: 764-773
  • 530 Sandborn WJ. et al. AGA technical review on perianal Crohn’s disease. Gastroenterology 2003; 125: 1508-1530
  • 531 O’Donoghue DP, Hyland JM. Perianal Crohn’s disease. Eur J Gastroenterol Hepatol 1997; 9: 235-236
  • 532 Sangwan YP. et al. Perianal Crohn’s disease. Results of local surgical treatment. Dis Colon Rectum 1996; 39: 529-535
  • 533 Lichtenstein GR. Treatment of fistulizing Crohn’s disease. Gastroenterology 2000; 119: 1132-1147
  • 534 Solomon MJ. Fistulae and abscesses in symptomatic perianal Crohn’s disease. Int J Colorectal Dis 1996; 11: 222-226
  • 535 Mardini HE, Schwartz DA. Treatment of Perianal Fistula and Abscess: Crohn’s and Non-Crohn’s. Curr Treat Options Gastroenterol 2007; 10: 211-220
  • 536 Steele SR. et al. Practice parameters for the management of perianal abscess and fistula-in-ano. Dis Colon Rectum 2011; 54: 1465-1474
  • 537 Kasparek MS. et al. Long-term quality of life in patients with Crohn’s disease and perianal fistulas: influence of fecal diversion. Dis Colon Rectum 2007; 50: 2067-2074
  • 538 Singh B, George BD, Mortensen NJ. Surgical therapy of perianal Crohn’s disease. Dig Liver Dis 2007; 39: 988-992
  • 539 Yamamoto T, Allan RN, Keighley MR. Effect of fecal diversion alone on perianal Crohn’s disease. World J Surg 2000; 24: 1258-1262 ; discussion 1262–1263
  • 540 Singh S. et al. Systematic review with meta-analysis: faecal diversion for management of perianal Crohn’s disease. Aliment Pharmacol Ther 2015; 42: 783-792
  • 541 Galandiuk S. et al Perianal Crohn disease: predictors of need for permanent diversion. Ann Surg 2005; 241: 796-801 ; discussion 801–802
  • 542 Marti-Gallostra M. et al. The role of a defunctioning stoma for colonic and perianal Crohn’s disease in the biological era. Scand J Gastroenterol 2017; 52: 251-256
  • 543 Kroesen AJ. et al. [Therapy of refractory proctocolitis and Crohn’s disease. Incisionless laparoscopic proctocolectomy with a Brooke ileostomy]. Chirurg 2009; 80: 730-733
  • 544 Shwaartz C. et al. Fistula-Associated Anorectal Cancer in the Setting of Crohn’s Disease. Dis Colon Rectum 2016; 59: 1168-1173
  • 545 Gecse KB. et al. A global consensus on the classification, diagnosis and multidisciplinary treatment of perianal fistulising Crohn’s disease. Gut 2014; 63: 1381-1392
  • 546 de Groof EJ. et al. Treatment of perianal fistula in Crohn’s disease: a systematic review and meta-analysis comparing seton drainage and anti-tumour necrosis factor treatment. Colorectal Dis 2016; 18: 667-675
  • 547 Tanaka S. et al. Clinical advantages of combined seton placement and infliximab maintenance therapy for perianal fistulizing Crohn’s disease: when and how were the seton drains removed?. Hepatogastroenterology 2010; 57: 3-7
  • 548 Seyfried S. et al. Fistulectomy with primary sphincter reconstruction. Int J Colorectal Dis 2018; 33: 911-918
  • 549 Rozalen V. et al. Advancement Flap Technique for Anal Fistula in Patients With Crohn’s Disease: A Systematic Review of the Literature. Cir Esp 2017; 95: 558-565
  • 550 Stellingwerf ME. et al. Systematic review and meta-analysis of endorectal advancement flap and ligation of the intersphincteric fistula tract for cryptoglandular and Crohn’s high perianal fistulas. BJS Open 2019; 3: 231-241
  • 551 Gottgens KWA. et al. Ligation of the Intersphincteric Fistula Tract for High Transsphincteric Fistula Yields Moderate Results at Best: Is the Tide Turning?. Dis Colon Rectum 2019; 62: 1231-1237
  • 552 Gingold DS, Murrell ZA, Fleshner PR. A prospective evaluation of the ligation of the intersphincteric tract procedure for complex anal fistula in patients with Crohn’s disease. Ann Surg 2014; 260: 1057-1061
  • 553 Senejoux A. et al. Fistula Plug in Fistulising Ano-Perineal Crohn’s Disease: a Randomised Controlled Trial. J Crohns Colitis 2016; 10: 141-148
  • 554 Fichera A. et al. Guidelines for the surgical treatment of Crohn’s perianal fistulas. Inflamm Bowel Dis 2015; 21: 753-758
  • 555 Panes J. et al. Expanded allogeneic adipose-derived mesenchymal stem cells (Cx601) for complex perianal fistulas in Crohn’s disease: a phase 3 randomised, double-blind controlled trial. Lancet 2016; 388: 1281-1290
  • 556 Molendijk I. et al. Allogeneic Bone Marrow-Derived Mesenchymal Stromal Cells Promote Healing of Refractory Perianal Fistulas in Patients With Crohn’s Disease. Gastroenterology 2015; 149: 918-927 e6
  • 557 Dige A. et al. Efficacy of Injection of Freshly Collected Autologous Adipose Tissue Into Perianal Fistulas in Patients With Crohn’s Disease. Gastroenterology 2019; 156: 2208-2216 e1
  • 558 WHO. Haemoglobin concentrations for the diagnosis of anaemia and assessment of severity. Vitamin and Mineral Nutrition Information System. Geneva, World Health Organization. 2011 (WHO/NMH/NHD/MNM/11.1) http://www.who.int/vmnis/indicators/haemoglobin
  • 559 Gasche C. et al. Guidelines on the diagnosis and management of iron deficiency and anemia in inflammatory bowel diseases. Inflamm Bowel Dis 2007; 13: 1545-1553
  • 560 Bager P. et al. High burden of iron deficiency and different types of anemia in inflammatory bowel disease outpatients in Scandinavia: a longitudinal 2-year follow-up study. Scand J Gastroenterol 2013; 48: 1286-1293
  • 561 Filmann N. et al. Prevalence of anemia in inflammatory bowel diseases in european countries: a systematic review and individual patient data meta-analysis. Inflamm Bowel Dis 2014; 20: 936-945
  • 562 Voegtlin M. et al. Prevalence of anaemia in inflammatory bowel disease in Switzerland: a cross-sectional study in patients from private practices and university hospitals. J Crohns Colitis 2010; 4: 642-648
  • 563 Patel D, Trivedi C, Khan N. Management of Anemia in Patients with Inflammatory Bowel Disease (IBD). Current Treatment Options in Gastroenterology 2018; 16: 112-128
  • 564 Stein J. et al. Management of inflammatory bowel disease-related anemia and iron deficiency with specific reference to the role of intravenous iron in current practice. Expert Opin Pharmacother 2017; 18: 1721-1737
  • 565 Dignass AU. et al. European consensus on the diagnosis and management of iron deficiency and anaemia in inflammatory bowel diseases. J Crohns Colitis 2015; 9: 211-222
  • 566 Daude S. et al. Comparative accuracy of ferritin, transferrin saturation and soluble transferrin receptor for the diagnosis of iron deficiency in inflammatory bowel disease. Alimentary Pharmacology & Therapeutics 2020; 51: 1087-1095
  • 567 Oustamanolakis P, Koutroubakis IE. Soluble transferrin receptor-ferritin index is the most efficient marker for the diagnosis of iron deficiency anemia in patients with IBD. Inflamm Bowel Dis 2011; 17: E158-E159
  • 568 Skikne BS. et al. Improved differential diagnosis of anemia of chronic disease and iron deficiency anemia: a prospective multicenter evaluation of soluble transferrin receptor and the sTfR/log ferritin index. Am J Hematol 2011; 86: 923-927
  • 569 Auerbach M, Adamson JW. How we diagnose and treat iron deficiency anemia. Am J Hematol 2016; 91: 31-38
  • 570 Evstatiev R. et al. FERGIcor, a randomized controlled trial on ferric carboxymaltose for iron deficiency anemia in inflammatory bowel disease. Gastroenterology 2011; 141: 846-853.e1-2
  • 571 Weng NP. Aging of the immune system: how much can the adaptive immune system adapt?. Immunity 2006; 24: 495-499
  • 572 Kulnigg S. et al. A novel intravenous iron formulation for treatment of anemia in inflammatory bowel disease: the ferric carboxymaltose (FERINJECT) randomized controlled trial. Am J Gastroenterol 2008; 103: 1182-1192
  • 573 Reinisch W. et al. A randomized, open-label, non-inferiority study of intravenous iron isomaltoside 1000 (Monofer) compared with oral iron for treatment of anemia in IBD (PROCEED). Am J Gastroenterol 2013; 108: 1877-1888
  • 574 Lindgren S. et al. Intravenous iron sucrose is superior to oral iron sulphate for correcting anaemia and restoring iron stores in IBD patients: A randomized, controlled, evaluator-blind, multicentre study. Scand J Gastroenterol 2009; 44: 838-845
  • 575 Erichsen K. et al. Oral ferrous fumarate or intravenous iron sucrose for patients with inflammatory bowel disease. Scand J Gastroenterol 2005; 40: 1058-1065
  • 576 Schröder O. et al. Intravenous iron sucrose versus oral iron supplementation for the treatment of iron deficiency anemia in patients with inflammatory bowel disease--a randomized, controlled, open-label, multicenter study. Am J Gastroenterol 2005; 100: 2503-2509
  • 577 Aksan A. et al. Systematic review with network meta-analysis: comparative efficacy and tolerability of different intravenous iron formulations for the treatment of iron deficiency anaemia in patients with inflammatory bowel disease. Aliment Pharmacol Ther 2017; 45: 1303-1318
  • 578 Abhyankar A, Moss AC. Iron Replacement in Patients with Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. Inflamm Bowel Dis 2015; 21: 1976-1981
  • 579 Bonovas S. et al. Intravenous Versus Oral Iron for the Treatment of Anemia in Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Medicine (Baltimore) 2016; 95: e2308
  • 580 Nielsen OH. et al. Management of Iron-Deficiency Anemia in Inflammatory Bowel Disease: A Systematic Review. Medicine (Baltimore) 2015; 94: e963
  • 581 Gasche C. et al. Ferric Maltol Is Effective in Correcting Iron Deficiency Anemia in Patients with Inflammatory Bowel Disease: Results from a Phase-3 Clinical Trial Program. Inflammatory Bowel Diseases 2014; 21: 579-588
  • 582 Ganzoni AM. [Intravenous iron-dextran: therapeutic and experimental possibilities]. Schweiz Med Wochenschr 1970; 100: 301-303
  • 583 Kulnigg S. et al. Rapid recurrence of IBD-associated anemia and iron deficiency after intravenous iron sucrose and erythropoietin treatment. Am J Gastroenterol 2009; 104: 1460-1467
  • 584 Schreiner P. et al. Nutrition in Inflammatory Bowel Disease. Digestion 2020; 101 (Suppl. 01) 120-135
  • 585 Domènech E. et al. Infliximab for the Treatment of Chronic Anemia in Crohn’s Disease. Official journal of the American College of Gastroenterology | ACG 2005; 100: 496
  • 586 Gasché C. et al. Intravenous iron and erythropoietin for anemia associated with Crohn disease. A randomized, controlled trial. Ann Intern Med 1997; 126: 782-787
  • 587 Gasche C. et al. Sequential treatment of anemia in ulcerative colitis with intravenous iron and erythropoietin. Digestion 1999; 60: 262-267
  • 588 Schreiber S. et al. Recombinant erythropoietin for the treatment of anemia in inflammatory bowel disease. N Engl J Med 1996; 334: 619-623
  • 589 Koutroubakis IE. et al. Effectiveness of darbepoetin-alfa in combination with intravenous iron sucrose in patients with inflammatory bowel disease and refractory anaemia: a pilot study. Eur J Gastroenterol Hepatol 2006; 18: 421-425
  • 590 Manganelli C, Turco S, Balestrazzi E. Ophthalmological aspects of IBD. Eur Rev Med Pharmacol Sci 2009; 13 (Suppl. 01) 11-13
  • 591 Cury DB, Moss AC. Ocular manifestations in a community-based cohort of patients with inflammatory bowel disease. Inflammatory Bowel Diseases 2009; 16: 1393-1396
  • 592 Walldorf J. et al. High frequency of secondary, but not primary ocular manifestations of inflammatory bowel disease in patients treated at a tertiary care center. European Journal of Gastroenterology & Hepatology 2018; 30: 1502-1506
  • 593 Troncoso LL. et al. Ophthalmic manifestations in patients with inflammatory bowel disease: A review. World J Gastroenterol 2017; 23: 5836-5848
  • 594 Glaubitz M. Rheumatic Manifestations in Chronic Inflammatory Bowel Disease: Clinical Features, Diagnosis and Treatment. Aktuelle Rheumatologie 2017; 42: 505-511
  • 595 Orchard TR, Wordsworth BP, Jewell DP. Peripheral arthropathies in inflammatory bowel disease: their articular distribution and natural history. Gut 1998; 42: 387-391
  • 596 Palm O. et al. The prevalence and incidence of peripheral arthritis in patients with inflammatory bowel disease, a prospective population-based study (the IBSEN study). Rheumatology (Oxford) 2001; 40: 1256-1261
  • 597 Palm O. et al. Prevalence of ankylosing spondylitis and other spondyloarthropathies among patients with inflammatory bowel disease: a population study (the IBSEN study). J Rheumatol 2002; 29: 511-515
  • 598 Harbord M. et al. The First European Evidence-based Consensus on Extra-intestinal Manifestations in Inflammatory Bowel Disease. J Crohns Colitis 2016; 10: 239-254
  • 599 Meserve J. et al. Retrospective Analysis of Safety of Vedolizumab in Patients With Inflammatory Bowel Diseases. Clin Gastroenterol Hepatol 2019; 17: 1533-1540 e2
  • 600 Herfarth H. et al. Improvement of arthritis and arthralgia after treatment with infliximab (Remicade) in a German prospective, open-label, multicenter trial in refractory Crohn’s disease. Am J Gastroenterol 2002; 97: 2688-2690
  • 601 Whittle SL. et al. Opioid therapy for treating rheumatoid arthritis pain. Cochrane Database Syst Rev 2011; 11: CD003113
  • 602 Miao XP. et al. Tolerability of selective cyclooxygenase 2 inhibitors used for the treatment of rheumatological manifestations of inflammatory bowel disease. Cochrane Database of Systematic Reviews 2014; 10: CD007744
  • 603 El Miedany Y. et al. The gastrointestinal safety and effect on disease activity of etoricoxib, a selective cox-2 inhibitor in inflammatory bowel diseases. Am J Gastroenterol 2006; 101: 311-317
  • 604 Sandborn WJ. et al. Safety of celecoxib in patients with ulcerative colitis in remission: a randomized, placebo-controlled, pilot study. Clin Gastroenterol Hepatol 2006; 4: 203-211
  • 605 Haibel H. et al. No efficacy of subcutaneous methotrexate in active ankylosing spondylitis: a 16-week open-label trial. Ann Rheum Dis 2007; 66: 419-421
  • 606 van der Heijde D. et al. 2010 Update of the international ASAS recommendations for the use of anti-TNF agents in patients with axial spondyloarthritis. Ann Rheum Dis 2011; 70: 905-908
  • 607 van der Heijde D. et al. 2016 update of the ASAS-EULAR management recommendations for axial spondyloarthritis. Ann Rheum Dis 2017; 76: 978-991
  • 608 Fragoulis GE. et al. Inflammatory bowel diseases and spondyloarthropathies: From pathogenesis to treatment. World J Gastroenterol 2019; 25: 2162-2176
  • 609 Jones G. et al. The effect of treatment on radiological progression in rheumatoid arthritis: a systematic review of randomized placebo-controlled trials. Rheumatology (Oxford) 2003; 42: 6-13
  • 610 Farhi D. et al. Significance of erythema nodosum and pyoderma gangrenosum in inflammatory bowel diseases: a cohort study of 2402 patients. Medicine (Baltimore) 2008; 87: 281-293
  • 611 Roth N. et al. Occurrence of skin manifestations in patients of the Swiss Inflammatory Bowel Disease Cohort Study. PLoS One 2019; 14: e0210436
  • 612 Ampuero J. et al. Predictive factors for erythema nodosum and pyoderma gangrenosum in inflammatory bowel disease. J Gastroenterol Hepatol 2014; 29: 291-295
  • 613 Agarwal A, Andrews JM. Systematic review: IBD-associated pyoderma gangrenosum in the biologic era, the response to therapy. Aliment Pharmacol Ther 2013; 38: 563-572
  • 614 Yadav S. et al. Hidradenitis Suppurativa in Patients With Inflammatory Bowel Disease: A Population-Based Cohort Study in Olmsted County, Minnesota. Clin Gastroenterol Hepatol 2016; 14: 65-70
  • 615 Deckers IE. et al. Inflammatory bowel disease is associated with hidradenitis suppurativa: Results from a multicenter cross-sectional study. J Am Acad Dermatol 2017; 76: 49-53
  • 616 Vavricka SR. et al. Chronological Order of Appearance of Extraintestinal Manifestations Relative to the Time of IBD Diagnosis in the Swiss Inflammatory Bowel Disease Cohort. Inflamm Bowel Dis 2015; 21: 1794-1800
  • 617 Guerra I. et al. Induction of psoriasis with anti-TNF agents in patients with inflammatory bowel disease: a report of 21 cases. J Crohns Colitis 2012; 6: 518-523
  • 618 Harrison MJ. et al. Rates of new-onset psoriasis in patients with rheumatoid arthritis receiving anti-tumour necrosis factor alpha therapy: results from the British Society for Rheumatology Biologics Register. Ann Rheum Dis 2009; 68: 209-215
  • 619 Mert A. et al. Erythema nodosum: an evaluation of 100 cases. Clin Exp Rheumatol 2007; 25: 563-570
  • 620 Baumgart DC. et al. Frequency, phenotype, outcome, and therapeutic impact of skin reactions following initiation of adalimumab therapy: experience from a consecutive cohort of inflammatory bowel disease patients. Inflamm Bowel Dis 2011; 17: 2512-2520
  • 621 Karmiris K. et al. Prevalence and Characteristics of Extra-intestinal Manifestations in a Large Cohort of Greek Patients with Inflammatory Bowel Disease. J Crohns Colitis 2016; 10: 429-436
  • 622 Vavricka SR. et al. Anti-TNF Treatment for Extraintestinal Manifestations of Inflammatory Bowel Disease in the Swiss IBD Cohort Study. Inflamm Bowel Dis 2017; 23: 1174-1181
  • 623 Peyrin-Biroulet L. et al. Systematic Review of Tumor Necrosis Factor Antagonists in Extraintestinal Manifestations in Inflammatory Bowel Disease. Clin Gastroenterol Hepatol 2017; 15: 25-36.e27
  • 624 Greuter T, Navarini A, Vavricka SR. Skin Manifestations of Inflammatory Bowel Disease. Clin Rev Allergy Immunol 2017; 53: 413-427
  • 625 Maverakis E. et al. Diagnostic Criteria of Ulcerative Pyoderma Gangrenosum: A Delphi Consensus of International Experts. JAMA Dermatol 2018; 154: 461-466
  • 626 Baranska-Rybak W. et al. A retrospective study of 12 cases of pyoderma gangrenosum: why we should avoid surgical intervention and what therapy to apply. Am Surg 2011; 77: 1644-1649
  • 627 Brooklyn TN. et al. Infliximab for the treatment of pyoderma gangrenosum: a randomised, double blind, placebo controlled trial. Gut 2006; 55: 505-509
  • 628 Saunte DML, Jemec GBE. Hidradenitis Suppurativa: Advances in Diagnosis and Treatment. Jama 2017; 318: 2019-2032
  • 629 Tzanetakou V. et al. Safety and Efficacy of Anakinra in Severe Hidradenitis Suppurativa: A Randomized Clinical Trial. JAMA Dermatol 2016; 152: 52-59
  • 630 Blok JL. et al. Ustekinumab in hidradenitis suppurativa: clinical results and a search for potential biomarkers in serum. Br J Dermatol 2016; 174: 839-846
  • 631 Boehncke W-H, Schön MP. Psoriasis. The Lancet 2015; 386: 983-994
  • 632 Nast A. et al. European S3-Guidelines on the systemic treatment of psoriasis vulgaris--Update 2015--Short version--EDF in cooperation with EADV and IPC. J Eur Acad Dermatol Venereol 2015; 29: 2277-2294
  • 633 Fousekis FS. et al. Hepatobiliary Manifestations and Complications in Inflammatory Bowel Disease: A Review. Gastroenterology Res 2018; 11: 83-94
  • 634 Lin A. et al. Prevalence of Nonalcoholic Fatty Liver Disease in Patients With Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. Inflamm Bowel Dis 2020; 27: 947-955
  • 635 Gizard E. et al. Systematic review: The epidemiology of the hepatobiliary manifestations in patients with inflammatory bowel disease. Aliment Pharmacol Ther 2014; 40: 3-15
  • 636 Dave M. et al. Primary sclerosing cholangitis: meta-analysis of diagnostic performance of MR cholangiopancreatography. Radiology 2010; 256: 387-396
  • 637 Nicoletti A, Maurice JB, Thorburn D. Guideline review: British Society of Gastroenterology/UK-PSC guidelines for the diagnosis and management of primary sclerosing cholangitis. Frontline Gastroenterology 2021; 12: 62-66
  • 638 Lindor KD. et al. High-dose ursodeoxycholic acid for the treatment of primary sclerosing cholangitis. Hepatology 2009; 50: 808-814
  • 639 Lindstrom L. et al. High dose ursodeoxycholic acid in primary sclerosing cholangitis does not prevent colorectal neoplasia. Aliment Pharmacol Ther 2012; 35: 451-457
  • 640 Pardi DS. et al. Ursodeoxycholic acid as a chemopreventive agent in patients with ulcerative colitis and primary sclerosing cholangitis. Gastroenterology 2003; 124: 889-893
  • 641 Lindor KD. Ursodiol for primary sclerosing cholangitis. Mayo Primary Sclerosing Cholangitis-Ursodeoxycholic Acid Study Group. N Engl J Med 1997; 336: 691-695
  • 642 Lindström L. et al. Association between reduced levels of alkaline phosphatase and survival times of patients with primary sclerosing cholangitis. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association 2013; 11: 841-846
  • 643 Aljiffry M. et al. Analytical review of diagnosis and treatment strategies for dominant bile duct strictures in patients with primary sclerosing cholangitis. HPB (Oxford) 2011; 13: 79-90
  • 644 Strassburg CP. S2k Leitlinie Autoimmune Lebererkrankungen. Zeitschrift für Gastroenterologie 2017; 55: 1135-1226
  • 645 Sgambato D. et al. Bone alterations in inflammatory bowel diseases. World journal of clinical cases 2019; 7: 1908-1925
  • 646 Klaus J. et al. High prevalence of osteoporotic vertebral fractures in patients with Crohn’s disease. Gut 2002; 51: 654-658
  • 647 Vazquez MA. et al. Vertebral fractures in patients with inflammatory bowel disease compared with a healthy population: a prospective case-control study. BMC Gastroenterol 2012; 12: 47
  • 648 Thomasius F. DVO Leitlinie 2017 zur Prophylaxe, Diagnostik und Therapie der Osteo- porose bei postmenopausalen Frauen und Männern. Osteologie 2018; 27: 154-160
  • 649 Bundesministerium für Gesundheit, Bekanntmachung eines Beschlusses des Gemeinsamen Bundesausschusses über eine Änderung der Richtlinie Methoden vertragsärztliche Versorgung (MVV-RL): Osteodensitometrie bei Osteoporose – vom: 21.02.2013. BAnz AT 10;05.2013: B3.
  • 650 Cravo M. et al. Risk factors for metabolic bone disease in Crohn’s disease patients. Inflamm Bowel Dis 2010; 16: 2117-2124
  • 651 Lo B. et al. Incidence, Risk Factors and Evaluation of Osteoporosis in Patients With Inflammatory Bowel Disease: A Danish Population-Based Inception Cohort With 10 Years of Follow-Up. J Crohns Colitis 2020; 14: 904-914
  • 652 Bechtold-Dalla Pozza S. Knochendichtemessungen beim wachsenden Skelett und die klinischen Konsequenzen. Z Rheumatol 2011; 70: 844-852
  • 653 Aksan A. et al. Measuring Vitamin D Status in Chronic Inflammatory Disorders: How does Chronic Inflammation Affect the Reliability of Vitamin D Metabolites in Patients with IBD?. Journal of Clinical Medicine 2020; 9: 547
  • 654 Levin AD. et al. Vitamin D deficiency in children with inflammatory bowel disease. Dig Dis Sci 2011; 56: 830-836
  • 655 Sentongo TA. et al. Vitamin D status in children, adolescents, and young adults with Crohn disease. Am J Clin Nutr 2002; 76: 1077-1081
  • 656 Siffledeen JS. et al. The frequency of vitamin D deficiency in adults with Crohn’s disease. Can J Gastroenterol 2003; 17: 473-478
  • 657 Mouli VP, Ananthakrishnan AN. Review article: vitamin D and inflammatory bowel diseases. Aliment Pharmacol Ther 2014; 39: 125-136
  • 658 Robinson RJ. et al. Effect of a low-impact exercise program on bone mineral density in Crohn’s disease: a randomized controlled trial. Gastroenterology 1998; 115: 36-41
  • 659 Manson JE. et al. Vitamin D Deficiency — Is There Really a Pandemic?. New England Journal of Medicine 2016; 375: 1817-1820
  • 660 Holick MF. et al. Evaluation, treatment, and prevention of vitamin D deficiency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2011; 96: 1911-1930
  • 661 Ross AC. et al. The 2011 report on dietary reference intakes for calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin Endocrinol Metab 2011; 96: 53-58
  • 662 Soo I. et al. Risedronate improves bone mineral density in Crohn’s disease: a two year randomized controlled clinical trial. J Crohns Colitis 2012; 6: 777-786
  • 663 Fagagnini S. et al. Risk factors for gallstones and kidney stones in a cohort of patients with inflammatory bowel diseases. PloS one 2017; 12: e0185193-e0185193
  • 664 Corica D, Romano C. Renal Involvement in Inflammatory Bowel Diseases. Journal of Crohn’s and Colitis 2015; 10: 226-235
  • 665 Worcester EM. Stones from bowel disease. Endocrinol Metab Clin North Am 2002; 31: 979-999
  • 666 Fink HA. et al. Diet, fluid, or supplements for secondary prevention of nephrolithiasis: a systematic review and meta-analysis of randomized trials. Eur Urol 2009; 56: 72-80
  • 667 Lieske JC. et al. Diet, but not oral probiotics, effectively reduces urinary oxalate excretion and calcium oxalate supersaturation. Kidney Int 2010; 78: 1178-1185
  • 668 Taylor EN, Stampfer MJ, Curhan GC. Dietary factors and the risk of incident kidney stones in men: new insights after 14 years of follow-up. J Am Soc Nephrol 2004; 15: 3225-3232
  • 669 Knoll T. et al. S2k-Leitlinie zur Diagnostik, Therapie und Metaphylaxe der Urolithiasis (AWMF 043/025). Der Urologe 2016; 55: 904-922
  • 670 Grainge MJ, West J, Card TR. Venous thromboembolism during active disease and remission in inflammatory bowel disease: a cohort study. Lancet 2010; 375: 657-663
  • 671 Kappelman MD. et al. Thromboembolic risk among Danish children and adults with inflammatory bowel diseases: a population-based nationwide study. Gut 2011; 60: 937-943
  • 672 Andrade AR. et al. Risk of thrombosis and mortality in inflammatory bowel disease. Clin Transl Gastroenterol 2018; 9: 142
  • 673 Cohen JB. et al. Inflammatory Bowel Disease and Thrombosis: A National Inpatient Sample Study. TH Open 2020; 4: e51-e58
  • 674 McCurdy JD. et al. A clinical predictive model for post-hospitalisation venous thromboembolism in patients with inflammatory bowel disease. Aliment Pharmacol Ther 2019; 49: 1493-1501
  • 675 Faye AS. et al. Minor Hematochezia Decreases Use of Venous Thromboembolism Prophylaxis in Patients with Inflammatory Bowel Disease. Inflammatory Bowel Diseases 2019; 26: 1394-1400
  • 676 Ra G. et al. Predictors and safety of venous thromboembolism prophylaxis among hospitalized inflammatory bowel disease patients. Journal of Crohn’s and Colitis 2013; 7: e479-e485
  • 677 Sultan K. et al. Increased Transfusion Requirements with Pharmacologic Thromboembolism Prophylaxis During Inflammatory Bowel Disease Exacerbation. Digestive Diseases and Sciences 2019; 64: 3256-3262
  • 678 Johannesdottir SA. et al. Use of glucocorticoids and risk of venous thromboembolism: a nationwide population-based case-control study. JAMA Intern Med 2013; 173: 743-752
  • 679 Lambin T, Faye AS, Colombel J-F. Inflammatory Bowel Disease Therapy and Venous Thromboembolism. Current Treatment Options in Gastroenterology 2020; 18: 462-475
  • 680 Hach-Wunderle V. et al. Diagnostik und Therapie der Venenthrombose und der Lungenembolie. 2010
  • 681 Lobo JL. et al. Anticoagulant therapy for venous thromboembolism in patients with inflammatory bowel disease. Eur J Gastroenterol Hepatol 2018; 30: 526-530
  • 682 Van Langenberg DR, Gibson PR. Systematic review: fatigue in inflammatory bowel disease. Alimentary Pharmacology & Therapeutics 2010; 32: 131-143
  • 683 Cohen BL. et al. Fatigue is highly associated with poor health-related quality of life, disability and depression in newly-diagnosed patients with inflammatory bowel disease, independent of disease activity. Aliment Pharmacol Ther 2014; 39: 811-822
  • 684 Czuber-Dochan W. et al. Development and psychometric testing of inflammatory bowel disease fatigue (IBD-F) patient self-assessment scale. J Crohns Colitis 2014; 8: 1398-1406
  • 685 Kreijne JE. et al. Practical Guideline for Fatigue Management in Inflammatory Bowel Disease. Journal of Crohn’s and Colitis 2015; 10: 105-111
  • 686 Kreijne JE. et al. Practical Guideline for Fatigue Management in Inflammatory Bowel Disease. J Crohns Colitis 2016; 10: 105-111
  • 687 van Erp LW. et al. Improvement of Fatigue and Quality of Life in Patients with Quiescent Inflammatory Bowel Disease Following a Personalized Exercise Program. Dig Dis Sci 2021; 66: 597-604
  • 688 Vogelaar L. et al. Solution focused therapy: A promising new tool in the management of fatigue in Crohn’s disease patients: Psychological interventions for the management of fatigue in Crohn’s disease. Journal of Crohn’s and Colitis 2011; 5: 585-591
  • 689 Bager P. et al. Randomised clinical trial: high-dose oral thiamine versus placebo for chronic fatigue in patients with quiescent inflammatory bowel disease. Aliment Pharmacol Ther 2021; 53: 79-86
  • 690 Jawaid N. et al. Paediatric to Adult Transition of Care in IBD: Understanding the Current Standard of Care Among Canadian Adult Academic Gastroenterologists. J Can Assoc Gastroenterol 2020; 3: 266-273
  • 691 Schutz L. et al. Long-term implications of structured transition of adolescents with inflammatory bowel disease into adult health care: a retrospective study. BMC Gastroenterol 2019; 19: 128
  • 692 Trivedi I. et al. Integrating Adolescents and Young Adults into Adult-Centered Care for IBD. Curr Gastroenterol Rep 2016; 18: 21
  • 693 Pearlstein H. et al. Predicting Sub-Optimal Transitions in Adolescents with Inflammatory Bowel Disease. J Pediatr Gastroenterol Nutr 2020; 72: 563-568
  • 694 Buderus S. et al. Inflammatory bowel disease in pediatric patients: Characteristics of newly diagnosed patients from the CEDATA-GPGE Registry. Dtsch Arztebl Int 2015; 112: 121-127
  • 695 Dodds CM. et al. Physicians’ Perceptions of Shared Decision Making in Chronic Disease and Its Barriers and Facilitators. J Pediatr 2016; 171: 307-309 e1-2
  • 696 Lipstein EA. et al. High Levels of Decisional Conflict and Decision Regret When Making Decisions About Biologics. J Pediatr Gastroenterol Nutr 2016; 63: e176-e181
  • 697 Siegel CA. et al. Gastroenterologists’ Views of Shared Decision Making for Patients with Inflammatory Bowel Disease. Dig Dis Sci 2015; 60: 2636-2645
  • 698 Picoraro JA, Rosh JR. Communicating the benefits and risks of inflammatory bowel disease therapy to patients and families. Curr Opin Pediatr 2017; 29: 572-577
  • 699 Afzal NA. et al. Colonic Crohn’s disease in children does not respond well to treatment with enteral nutrition if the ileum is not involved. Dig Dis Sci 2005; 50: 1471-1475
  • 700 Hansen T, Duerksen DR. Enteral Nutrition in the Management of Pediatric and Adult Crohn’s Disease. Nutrients 2018; 10: 537
  • 701 Russell CA. Role of dietitians in enteral feeding. Gut 1986; 27 (Suppl. 01) 58-60
  • 702 Baird DD, Narendranathan M, Sandler RS. Increased risk of preterm birth for women with inflammatory bowel disease. Gastroenterology 1990; 99: 987-994
  • 703 Hudson M. et al. Fertility and pregnancy in inflammatory bowel disease. Int J Gynaecol Obstet 1997; 58: 229-237
  • 704 Ording Olsen K. et al. Ulcerative colitis: female fecundity before diagnosis, during disease, and after surgery compared with a population sample. Gastroenterology 2002; 122: 15-19
  • 705 Senates E. et al. Serum anti-Mullerian hormone levels are lower in reproductive-age women with Crohn’s disease compared to healthy control women. J Crohns Colitis 2013; 7: e29-e34
  • 706 Selinger CP. et al. Inflammatory bowel disease and pregnancy: lack of knowledge is associated with negative views. J Crohns Colitis 2013; 7: e206-e213
  • 707 de Lima A. et al. Preconception Care Reduces Relapse of Inflammatory Bowel Disease During Pregnancy. Clin Gastroenterol Hepatol 2016; 14: 1285-1292 e1
  • 708 Khosla R, Willoughby CP, Jewell DP. Crohn’s disease and pregnancy. Gut 1984; 25: 52-56
  • 709 Hanan IM, Kirsner JB. Inflammatory bowel disease in the pregnant woman. Clin Perinatol 1985; 12: 669-682
  • 710 Reddy D. et al. Relapses of inflammatory bowel disease during pregnancy: in-hospital management and birth outcomes. Am J Gastroenterol 2008; 103: 1203-1209
  • 711 Abhyankar A, Ham M, Moss AC. Meta-analysis: the impact of disease activity at conception on disease activity during pregnancy in patients with inflammatory bowel disease. Aliment Pharmacol Ther 2013; 38: 460-466
  • 712 Fonager K. et al. Pregnancy outcome for women with Crohn’s disease: a follow-up study based on linkage between national registries. Am J Gastroenterol 1998; 93: 2426-2430
  • 713 Baiocco PJ, Korelitz BI. The influence of inflammatory bowel disease and its treatment on pregnancy and fetal outcome. J Clin Gastroenterol 1984; 6: 211-216
  • 714 Broms G. et al. Birth outcomes in women with inflammatory bowel disease: effects of disease activity and drug exposure. Inflamm Bowel Dis 2014; 20: 1091-1098
  • 715 Mahadevan U. et al. Pregnancy outcomes in women with inflammatory bowel disease: a large community-based study from Northern California. Gastroenterology 2007; 133: 1106-1112
  • 716 Naganuma M. et al. Conception and pregnancy outcome in women with inflammatory bowel disease: A multicentre study from Japan. J Crohns Colitis 2011; 5: 317-323
  • 717 Cornish J. et al. A meta-analysis on the influence of inflammatory bowel disease on pregnancy. Gut 2007; 56: 830-837
  • 718 Kammerlander H. et al. The Effect of Disease Activity on Birth Outcomes in a Nationwide Cohort of Women with Moderate to Severe Inflammatory Bowel Disease. Inflamm Bowel Dis 2017; 23: 1011-1018
  • 719 van der Woude CJ. et al. The second European evidenced-based consensus on reproduction and pregnancy in inflammatory bowel disease. J Crohns Colitis 2015; 9: 107-124
  • 720 Carmichael SL. et al Maternal corticosteroid use and orofacial clefts. Am J Obstet Gynecol 2007; 197: 585 e1–7 ; discussion 683–684, e1–7.
  • 721 Hviid A, Molgaard-Nielsen D. Corticosteroid use during pregnancy and risk of orofacial clefts. CMAJ 2011; 183: 796-804
  • 722 Martel MJ. et al. Use of inhaled corticosteroids during pregnancy and risk of pregnancy induced hypertension: nested case-control study. BMJ 2005; 330: 230
  • 723 Hutson JR. et al. The fetal safety of thiopurines for the treatment of inflammatory bowel disease in pregnancy. J Obstet Gynaecol 2013; 33: 1-8
  • 724 Akbari M. et al. Systematic review and meta-analysis on the effects of thiopurines on birth outcomes from female and male patients with inflammatory bowel disease. Inflamm Bowel Dis 2013; 19: 15-22
  • 725 Matro R. et al. Exposure Concentrations of Infants Breastfed by Women Receiving Biologic Therapies for Inflammatory Bowel Diseases and Effects of Breastfeeding on Infections and Development. Gastroenterology 2018; 155: 696-704
  • 726 Mahadevan U. et al. Pregnancy and Neonatal Outcomes After Fetal Exposure to Biologics and Thiopurines Among Women With Inflammatory Bowel Disease. Gastroenterology 2020; 160: 1131-1139
  • 727 Chaparro M. et al. Long-Term Safety of In Utero Exposure to Anti-TNFalpha Drugs for the Treatment of Inflammatory Bowel Disease: Results from the Multicenter European TEDDY Study. Am J Gastroenterol 2018; 113: 396-403
  • 728 Kammerlander H. et al. Anti-TNF-alpha Use During the Third Trimester of Pregnancy in Women with Moderate-severe Inflammatory Bowel Disease and the Risk of Preterm Birth and Low Birth Weight. Inflamm Bowel Dis 2017; 23: 1916-1923
  • 729 Nielsen OH. et al. Biologics for Inflammatory Bowel Disease and their Safety in Pregnancy: A Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol 2020; 20: 74-87
  • 730 Kozlowski RD. et al. Outcome of first-trimester exposure to low-dose methotrexate in eight patients with rheumatic disease. Am J Med 1990; 88: 589-592
  • 731 Smithells RW, Newman CG. Recognition of thalidomide defects. J Med Genet 1992; 29: 716-723
  • 732 O’Morain C. et al. Reversible male infertility due to sulphasalazine: studies in man and rat. Gut 1984; 25: 1078-1084
  • 733 Birnie GG, McLeod TI, Watkinson G. Incidence of sulphasalazine-induced male infertility. Gut 1981; 22: 452-455
  • 734 Kane S, Lemieux N. The role of breastfeeding in postpartum disease activity in women with inflammatory bowel disease. Am J Gastroenterol 2005; 100: 102-105
  • 735 Roy A. et al. Exposure to Biologic Therapy and Childhood Development among Offspring of Women with Inflammatory Bowel Disease: Results from the Piano Registry. Gastroenterology 2017; 152: S85-S86
  • 736 Hatch Q. et al. Crohn’s disease and pregnancy: the impact of perianal disease on delivery methods and complications. Dis Colon Rectum 2014; 57: 174-178
  • 737 Mahadevan U. et al. Inflammatory Bowel Disease in Pregnancy Clinical Care Pathway: A Report From the American Gastroenterological Association IBD Parenthood Project Working Group. Gastroenterology 2019; 156: 1508-1524
  • 738 Mahadevan U. Pregnancy and inflammatory bowel disease. Med Clin North Am 2010; 94: 53-73
  • 739 Sturm A. et al. European Crohn’s and Colitis Organisation Topical Review on IBD in the Elderly. J Crohns Colitis 2017; 11: 263-273
  • 740 Hruz P. et al. Management of the Elderly Inflammatory Bowel Disease Patient. Digestion 2020; 101 (Suppl. 01) 105-119
  • 741 Rahier JF. et al. Second European evidence-based consensus on the prevention, diagnosis and management of opportunistic infections in inflammatory bowel disease. J Crohns Colitis 2014; 8: 443-468
  • 742 Akerkar GA. et al. Corticosteroid-associated complications in elderly Crohn’s disease patients. Am J Gastroenterol 1997; 92: 461-464
  • 743 Brassard P. et al Oral corticosteroids and the risk of serious infections in patients with elderly-onset inflammatory bowel diseases. Am J Gastroenterol 2014; 109: 1795-1802 ; quiz 1803
  • 744 Lichtenstein GR. et al. Serious infection and mortality in patients with Crohn’s disease: more than 5 years of follow-up in the TREAT registry. Am J Gastroenterol 2012; 107: 1409-1422
  • 745 Kelly CP, LaMont JT. Clostridium difficile--more difficult than ever. N Engl J Med 2008; 359: 1932-1940
  • 746 Schneeweiss S. et al. Infliximab and other immunomodulating drugs in patients with inflammatory bowel disease and the risk of serious bacterial infections. Aliment Pharmacol Ther 2009; 30: 253-264
  • 747 Fidder H. et al. Long-term safety of infliximab for the treatment of inflammatory bowel disease: a single-centre cohort study. Gut 2009; 58: 501-508
  • 748 Asscher VER. et al. Anti-tumor necrosis factor therapy in patients with inflammatory bowel disease; comorbidity, not patient age, is a predictor of severe adverse events. Int J Colorectal Dis 2020; 35: 2331-2338
  • 749 Siegel CA. et al. Risk of lymphoma associated with combination anti-tumor necrosis factor and immunomodulator therapy for the treatment of Crohn’s disease: a meta-analysis. Clin Gastroenterol Hepatol 2009; 7: 874-881
  • 750 Colombel JF. et al. The safety profile of infliximab in patients with Crohn’s disease: the Mayo clinic experience in 500 patients. Gastroenterology 2004; 126: 19-31
  • 751 Desai A. et al. Older age is associated with higher rate of discontinuation of anti-TNF therapy in patients with inflammatory bowel disease. Inflamm Bowel Dis 2013; 19: 309-315
  • 752 Cottone M. et al. Advanced age is an independent risk factor for severe infections and mortality in patients given anti-tumor necrosis factor therapy for inflammatory bowel disease. Clin Gastroenterol Hepatol 2011; 9: 30-35
  • 753 Piovani D. et al. Systematic review with meta-analysis: biologics and risk of infection or cancer in elderly patients with inflammatory bowel disease. Aliment Pharmacol Ther 2020; 51: 820-830
  • 754 Hardt J. et al. [Inflammatory bowel diseases as multi-focal disorders: results from a multi-regional survey on bodily and psychosocial problems in IBD patients]. Z Gastroenterol 2010; 48: 381-391
  • 755 Lo CH. et al. Dietary Inflammatory Potenzial and Risk of Crohn’s Disease and Ulcerative Colitis. Gastroenterology 2020; 159: 873-883 e1
  • 756 Levine A. et al. Dietary Guidance From the International Organization for the Study of Inflammatory Bowel Diseases. Clin Gastroenterol Hepatol 2020; 18: 1381-1392
  • 757 Godny L. et al. Fruit Consumption is Associated with Alterations in Microbial Composition and Lower Rates of Pouchitis. J Crohns Colitis 2019; 13: 1265-1272
  • 758 Ritchie JK. et al. Controlled multicentre therapeutic trial of an unrefined carbohydrate, fibre rich diet in Crohn’s disease. Br Med J (Clin Res Ed) 1987; 295: 517-520
  • 759 Limketkai BN. et al. Dietary interventions for induction and maintenance of remission in inflammatory bowel disease. Cochrane Database Syst Rev 2019; 2: CD012839
  • 760 Albenberg L. et al. A Diet Low in Red and Processed Meat Does Not Reduce Rate of Crohn’s Disease Flares. Gastroenterology 2019; 157: 128-136 e5
  • 761 Narula N. et al. Enteral nutritional therapy for induction of remission in Crohn’s disease. Cochrane Database Syst Rev 2018; 4: CD000542
  • 762 Comeche JM. et al. Enteral Nutrition in Patients with Inflammatory Bowel Disease. Systematic Review, Meta-Analysis, and Meta-Regression. Nutrients 2019; 11: 2657
  • 763 Lee D. et al. Comparative Effectiveness of Nutritional and Biological Therapy in North American Children with Active Crohn’s Disease. Inflamm Bowel Dis 2015; 21: 1786-1793
  • 764 Pigneur B. et al. Mucosal Healing and Bacterial Composition in Response to Enteral Nutrition Vs Steroid-based Induction Therapy-A Randomised Prospective Clinical Trial in Children With Crohn’s Disease. J Crohns Colitis 2019; 13: 846-855
  • 765 Adamji M, Day AS. An overview of the role of exclusive enteral nutrition for complicated Crohn’s disease. Intest Res 2019; 17: 171-176
  • 766 Levine A. et al. Crohn’s Disease Exclusion Diet Plus Partial Enteral Nutrition Induces Sustained Remission in a Randomized Controlled Trial. Gastroenterology 2019; 157: 440-450 e8
  • 767 Sigall-Boneh R. et al. Partial enteral nutrition with a Crohn’s disease exclusion diet is effective for induction of remission in children and young adults with Crohn’s disease. Inflamm Bowel Dis 2014; 20: 1353-1360
  • 768 Sigall Boneh R. et al. Dietary Therapy With the Crohn’s Disease Exclusion Diet is a Successful Strategy for Induction of Remission in Children and Adults Failing Biological Therapy. J Crohns Colitis 2017; 11: 1205-1212
  • 769 Yanai H, Levine A, Sigall Boneh R. et al Crohn’s disease exclusion diet induces remission in adults with mild to moderate Crohn’s disease: preliminary report from a randomized controlled trial (CDED-AD trial), in United European Gastroenterology Journal. 2020 SAGE Publications. 466
  • 770 Bischoff SC. et al. S3-Leitlinie der Deutschen Gesellschaft für Ernährungsmedizin (DGEM) in Zusammenarbeit mit der Gesellschaft für klinische Ernährung der Schweiz (GESKES), der Österreichischen Arbeitsgemeinschaft für klinische Ernährung (AKE) und der Deutsche Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS). Aktuelle Ernährungsmedizin 2014; 39: e72-e98
  • 771 Valentini L. et al. Malnutrition and impaired muscle strength in patients with Crohn’s disease and ulcerative colitis in remission. Nutrition 2008; 24: 694-702
  • 772 Nguyen GC, Munsell M, Harris ML. Nationwide prevalence and prognostic significance of clinically diagnosable protein-calorie malnutrition in hospitalized inflammatory bowel disease patients. Inflamm Bowel Dis 2008; 14: 1105-1111
  • 773 Gomes F. et al. Association of Nutritional Support With Clinical Outcomes Among Medical Inpatients Who Are Malnourished or at Nutritional Risk: An Updated Systematic Review and Meta-analysis. JAMA Netw Open 2019; 2: e1915138
  • 774 Sandhu A. et al. Self-Screening for Malnutrition Risk in Outpatient Inflammatory Bowel Disease Patients Using the Malnutrition Universal Screening Tool (MUST). JPEN J Parenter Enteral Nutr 2016; 40: 507-510
  • 775 Cox SR. et al. Effects of Low FODMAP Diet on Symptoms, Fecal Microbiome, and Markers of Inflammation in Patients With Quiescent Inflammatory Bowel Disease in a Randomized Trial. Gastroenterology 2020; 158: 176-188 e7
  • 776 Burden S. et al. Pre-operative nutrition support in patients undergoing gastrointestinal surgery. Cochrane Database Syst Rev 2012; 11: CD008879
  • 777 Adamina M. et al. Perioperative Dietary Therapy in Inflammatory Bowel Disease. J Crohns Colitis 2020; 14: 431-444
  • 778 Spinelli A. et al. Review article: optimal preparation for surgery in Crohn’s disease. Aliment Pharmacol Ther 2014; 40: 1009-1022
  • 779 Helwig U. et al. A Prospective Multicenter Study on the Prevalence of Fructose Malabsorption in Patients with Chronic Inflammatory Bowel Disease. Digestion 2020; 102: 397-403
  • 780 Szilagyi A, Galiatsatos P, Xue X. Systematic review and meta-analysis of lactose digestion, its impact on intolerance and nutritional effects of dairy food restriction in inflammatory bowel diseases. Nutr J 2016; 15: 67
  • 781 Buning C. et al. The C/C(-13910) and G/G(-22018) genotypes for adult-type hypolactasia are not associated with inflammatory bowel disease. Scand J Gastroenterol 2003; 38: 538-542
  • 782 Omer B. et al. Steroid-sparing effect of wormwood (Artemisia absinthium) in Crohn’s disease: a double-blind placebo-controlled study. Phytomedicine 2007; 14: 87-95
  • 783 Krebs S, Omer TN, Omer B. Wormwood (Artemisia absinthium) suppresses tumour necrosis factor alpha and accelerates healing in patients with Crohn’s disease – A controlled clinical trial. Phytomedicine 2010; 17: 305-309
  • 784 Gerhardt H. et al. [Therapy of active Crohn disease with Boswellia serrata extract H 15]. Z Gastroenterol 2001; 39: 11-17
  • 785 Holtmeier W. et al. Randomized, placebo-controlled, double-blind trial of Boswellia serrata in maintaining remission of Crohn’s disease: good safety profile but lack of efficacy. Inflamm Bowel Dis 2011; 17: 573-582
  • 786 Torres J. et al. European Crohn’s and Colitis Organisation Topical Review on Complementary Medicine and Psychotherapy in Inflammatory Bowel Disease. J Crohns Colitis 2019; 13: 673-685e
  • 787 Nielsen AA. et al. Omega-3 fatty acids inhibit an increase of proinflammatory cytokines in patients with active Crohn’s disease compared with omega-6 fatty acids. Aliment Pharmacol Ther 2005; 22: 1121-1128
  • 788 Eivindson M. et al. Insulin-like growth factors (IGFs) and IGF binding proteins in active Crohn’s disease treated with omega-3 or omega-6 fatty acids and corticosteroids. Scand J Gastroenterol 2005; 40: 1214-1221
  • 789 Lev-Tzion R. et al. Omega 3 fatty acids (fish oil) for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2014; 2: CD006320
  • 790 Turner D. et al. Omega 3 fatty acids (fish oil) for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2009; 1: CD006320
  • 791 Turner D. et al. Maintenance of remission in inflammatory bowel disease using omega-3 fatty acids (fish oil): a systematic review and meta-analyses. Inflamm Bowel Dis 2011; 17: 336-445
  • 792 Joos S. et al. Acupuncture and moxibustion in the treatment of active Crohn’s disease: a randomized controlled study. Digestion 2004; 69: 131-139
  • 793 Bao CH. et al. Randomized controlled trial: moxibustion and acupuncture for the treatment of Crohn’s disease. World J Gastroenterol 2014; 20: 11000-11011
  • 794 Scholmerich J. Trichuris suis ova in inflammatory bowel disease. Dig Dis 2013; 31: 391-395
  • 795 Langhorst J. et al. Systematic review of complementary and alternative medicine treatments in inflammatory bowel diseases. J Crohns Colitis 2015; 9: 86-106
  • 796 Derwa Y. et al. Systematic review with meta-analysis: the efficacy of probiotics in inflammatory bowel disease. Aliment Pharmacol Ther 2017; 46: 389-400
  • 797 Malchow HA. Crohn’s disease and Escherichia coli. A new approach in therapy to maintain remission of colonic Crohn’s disease?. J Clin Gastroenterol 1997; 25: 653-658
  • 798 Schultz M. et al. Lactobacillus GG in inducing and maintaining remission of Crohn’s disease. BMC Gastroenterol 2004; 4: 5
  • 799 Willert RP. et al. T1235 Randomised, Double-Blinded, Placebo-Controlled Study of VSL#3 Versus Placebo in the Maintenance of Remission in Crohns Disease. Gastroenterology 2010; 138: S-517-S-518
  • 800 Bourreille A. et al. Saccharomyces boulardii does not prevent relapse of Crohn’s disease. Clin Gastroenterol Hepatol 2013; 11: 982-987
  • 801 Fedorak RN. et al. The probiotic VSL#3 has anti-inflammatory effects and could reduce endoscopic recurrence after surgery for Crohn’s disease. Clin Gastroenterol Hepatol 2015; 13: 928-935 e2
  • 802 Marteau P. et al. Ineffectiveness of Lactobacillus johnsonii LA1 for prophylaxis of postoperative recurrence in Crohn’s disease: a randomised, double blind, placebo controlled GETAID trial. Gut 2006; 55: 842-847
  • 803 Prantera C. et al. Ineffectiveness of probiotics in preventing recurrence after curative resection for Crohn’s disease: a randomised controlled trial with Lactobacillus GG. Gut 2002; 51: 405-409
  • 804 Van Gossum A. et al. Multicenter randomized-controlled clinical trial of probiotics (Lactobacillus johnsonii, LA1) on early endoscopic recurrence of Crohn’s disease after lleo-caecal resection. Inflamm Bowel Dis 2007; 13: 135-142
  • 805 Atzeni F. et al. Rheumatic manifestations in inflammatory bowel disease. Autoimmun Rev 2014; 13: 20-23
  • 806 Palm O. et al. Fibromyalgia and chronic widespread pain in patients with inflammatory bowel disease: a cross sectional population survey. J Rheumatol 2001; 28: 590-594
  • 807 Martinis F. et al. Clinical and sonographic discrimination between fibromyalgia and spondyloarthopathy in inflammatory bowel disease with musculoskeletal pain. Rheumatology (Oxford) 2020; 59: 2857-2863
  • 808 Chen JH. et al. Is Fibromyalgia Risk Higher Among Male and Young Inflammatory Bowel Disease Patients? Evidence from a Taiwan Cohort of One Million. Pain Physician 2018; 21: E257-E264
  • 809 Buskila D. et al. Fibromyalgia in inflammatory bowel disease. J Rheumatol 1999; 26: 1167-1171
  • 810 Petzke F. et al. [General treatment principles, coordination of care and patient education in fibromyalgia syndrome: Updated guidelines 2017 and overview of systematic review articles]. Schmerz 2017; 31: 246-254
  • 811 Coates MD. et al. Opioid Analgesics Do Not Improve Abdominal Pain or Quality of Life in Crohn’s Disease. Dig Dis Sci 2020; 65: 2379-2387
  • 812 Burr NE. et al. Increasing Prescription of Opiates and Mortality in Patients With Inflammatory Bowel Diseases in England. Clin Gastroenterol Hepatol 2018; 16: 534-541 e6
  • 813 Hauser W. et al. [Recommendations of the second update of the LONTS guidelines: Long-term opioid therapy for chronic noncancer pain]. Schmerz 2020; 34: 204-244
  • 814 Kafil TS. et al. Cannabis for the treatment of Crohn’s disease. Cochrane Database Syst Rev 2018; 11: CD012853
  • 815 Naftali T. et al. Cannabis induces a clinical response in patients with Crohn’s disease: a prospective placebo-controlled study. Clin Gastroenterol Hepatol 2013; 11: 1276-1280 e1
  • 816 Petzke F. et al. [Position paper on medical cannabis and cannabis-based medicines in pain medicine]. Schmerz 2019; 33: 449-465
  • 817 Naftali T. et al. Medical cannabis for inflammatory bowel disease: real-life experience of mode of consumption and assessment of side-effects. Eur J Gastroenterol Hepatol 2019; 31: 1376-1381
  • 818 Leitlinienprogramm Onkologie (Deutsche Krebsgesellschaft, Deutsche Krebshilfe, AWMF): Palliativmedizin für Patienten mit einer nicht-heilbaren Krebserkrankung. 2020
  • 819 Walker JR. et al. The Manitoba IBD cohort study: a population-based study of the prevalence of lifetime and 12-month anxiety and mood disorders. Am J Gastroenterol 2008; 103: 1989-1997
  • 820 Bernstein MT. et al. Assessing the Relationship between Sources of Stress and Symptom Changes among Persons with IBD over Time: A Prospective Study. Can J Gastroenterol Hepatol 2016; 2016: 1681507
  • 821 Bernstein CN. et al. A prospective population-based study of triggers of symptomatic flares in IBD. Am J Gastroenterol 2010; 105: 1994-2002
  • 822 Bitton A. et al. Predicting relapse in Crohn’s disease: a biopsychosocial model. Gut 2008; 57: 1386-1392
  • 823 Camara RJ. et al. The role of psychological stress in inflammatory bowel disease: quality assessment of methods of 18 prospective studies and suggestions for future research. Digestion 2009; 80: 129-139
  • 824 Knowles SR, Monshat K, Castle DJ. The efficacy and methodological challenges of psychotherapy for adults with inflammatory bowel disease: a review. Inflamm Bowel Dis 2013; 19: 2704-2715
  • 825 Timmer A. et al. Psychological interventions for treatment of inflammatory bowel disease. Cochrane Database Syst Rev 2011; 2: CD006913
  • 826 Targownik LE. et al The Relationship Among Perceived Stress, Symptoms, and Inflammation in Persons With Inflammatory Bowel Disease. Am J Gastroenterol 2015; 110: 1001-1012 ; quiz 1013
  • 827 Langhorst J, Häuser W. KA. Entzündliche Darmerkrankungen. in Egle, Heim, Strauß, von Känel. (Hrsg): Psychosomatik. Neurobiologisch fundiert und evidenzbasiert. Ein Lehr- und Handbuch. Kohlhammer; 2020: 860
  • 828 Dignass A. et al. Aktualisierte Leitlinie zur Diagnostik und Therapie der Colitis ulcerosa 2011 – Ergebnisse einer Evidenzbasierten Konsensuskonferenz. Z Gastroenterol 2011; 49: 1276-1341
  • 829 Kucharzik T. et al. Aktualisierte S3-Leitlinie Colitis ulcerosa. Z Gastroenterol 2019; 57: 1321-1405
  • 830 Gracie DJ. et al. Bi-directionality of Brain-Gut Interactions in Patients With Inflammatory Bowel Disease. Gastroenterology 2018; 154: 1635-1646 e3
  • 831 Langhorst J. et al. Short-term stress, but not mucosal healing nor depression was predictive for the risk of relapse in patients with ulcerative colitis: a prospective 12-month follow-up study. Inflamm Bowel Dis 2013; 19: 2380-2386
  • 832 Berrill JW. et al. Mindfulness-based therapy for inflammatory bowel disease patients with functional abdominal symptoms or high perceived stress levels. J Crohns Colitis 2014; 8: 945-955
  • 833 Wynne B. et al. Acceptance and Commitment Therapy Reduces Psychological Stress in Patients With Inflammatory Bowel Diseases. Gastroenterology 2019; 156: 935-945 e1
  • 834 Vogelaar L. et al. Fatigue management in patients with IBD: a randomised controlled trial. Gut 2014; 63: 911-918
  • 835 Corey G. Theory and practice of counseling and psychotherapy. 8th ed. Thomson/Brooks/Cole; 2009
  • 836 Margraf J, Schneider S. Lehrbuch der Verhaltenstherapie, Band 1 – Grundlagen, Diagnostik, Verfahren und Rahmenbedingungen psychologischer Therapie. 4. Auflage ed. Berlin Heidelberg: Springer- Verlag; 2018
  • 837 Gödde GB, Michael B. Band 1: Psychologie als Wissenschaft der Komplementarität. Der Besen, mit dem die Hexe fliegt Wissenschaft und Therapeutik des Unbewussten. Psychosozial-Verlag; 2012: 669
  • 838 Bennebroek Evertsz F. et al. Effectiveness of cognitive-behavioral therapy on quality of life, anxiety, and depressive symptoms among patients with inflammatory bowel disease: A multicenter randomized controlled trial. J Consult Clin Psychol 2017; 85: 918-925
  • 839 Mikocka-Walus A. et al. Cognitive-Behavioural Therapy for Inflammatory Bowel Disease: 24-Month Data from a Randomised Controlled Trial. Int J Behav Med 2017; 24: 127-135
  • 840 Mikocka-Walus A. et al. Cognitive-behavioural therapy has no effect on disease activity but improves quality of life in subgroups of patients with inflammatory bowel disease: a pilot randomised controlled trial. BMC Gastroenterol 2015; 15: 54
  • 841 Levy RL. et al. Effects of a Cognitive Behavioral Therapy Intervention Trial to Improve Disease Outcomes in Children with Inflammatory Bowel Disease. Inflamm Bowel Dis 2016; 22: 2134-2148
  • 842 Stapersma L. et al. Effectiveness of Disease-Specific Cognitive Behavioral Therapy on Anxiety, Depression, and Quality of Life in Youth With Inflammatory Bowel Disease: A Randomized Controlled Trial. J Pediatr Psychol 2018; 43: 967-980