Новые подходы к диагностике и лечению болезни Крона


Цитировать

Полный текст

Аннотация

Аннотация. Представлены современные взгляды на диагностику и консервативное лечение болезни Крона (БК). Рассмотрены современные принципы классификации, оценки тяжести и диагностические критерии рассматриваемого заболевания. Приведены актуальные схемы консервативной терапии БК, регламентированные проектом Российских клинических рекомендаций по диагностике и лечению БК у взрослых (2013), составленных ведущими отечественными экспертами на основании Европейского доказательного консенсуса по диагностике и лечению БК (2010). Рассмотрены перспективные методы лечения больных БК, влияющие на ключевые молекулы патогенеза данного заболевания.

Полный текст

Новые подходы к диагностике и лечению болезни Крона. - Аннотация. Представлены современные взгляды на диагностику и консервативное лечение болезни Крона (БК). Рассмотрены современные принципы классификации, оценки тяжести и диагностические критерии рассматриваемого заболевания. Приведены актуальные схемы консервативной терапии БК, регламентированные проектом Российских клинических рекомендаций по диагностике и лечению БК у взрослых (2013), составленных ведущими отечественными экспертами на основании Европейского доказательного консенсуса по диагностике и лечению БК (2010). Рассмотрены перспективные методы лечения больных БК, влияющие на ключевые молекулы патогенеза данного заболевания.
×

Об авторах

И В Маев

ГБОУ ВПО "Московский государственный медико-стоматологический университет им. А.И. Евдокимова" Минздрава России

Д Н Андреев

ГБОУ ВПО "Московский государственный медико-стоматологический университет им. А.И. Евдокимова" Минздрава России

Email: dna-mit8@mail.ru

Список литературы

  1. Парфенов А.И. Болезнь Крона: к 80-летию описания. Тер арх. 2013; 8: 35-42.
  2. Sands B.E., Siegel C.A. Crohn's disease. In: Feldman M, Friedman LS, Brandt LJ, eds. Sleisenger & Fordtran's Gastrointestinal and Liver Disease. 9th ed. Philadelphia, Pa: Saunders Elsevier; 2010: chap 111.
  3. Рекомендации Российской гастроэнтерологической ассоциации по лечению болезни Крона у взрослых (проект). Рос журн гастроэнтерол гепатол колопроктол 2012; 6: 66-82.
  4. Burisch J., Munkholm P. Inflammatory bowel disease epidemiology. Curr Opin Gastroenterol 2013; 29 (4): 357-362.
  5. Molodecky N.A., Soon I.S., Rabi D.M. et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology 2012; 142 (1): 46-54.
  6. Kappelman M.D., Rifas-Shiman S.L., Kleinman K. et al. The prevalence and geographic distribution of Crohn's disease and ulcerative colitis in the United States. Clin Gastroenterol Hepatol 2007; 5:1424-1429.
  7. Ponder A., Long M.D. A clinical review of recent findings in the epidemiology of inflammatory bowel disease. Clin Epidemiol 2013; 5:237-247.
  8. Воробьев Г.И., Халиф И.Л. Неспецифические воспалительные заболевания кишечника. М: Миклош 2008.
  9. Bernstein C.N., Wajda A., Svenson L.W. et al. The epidemiology of inflammatory bowel disease in Canada: a population-based study. Am J Gastroenterol 2006; 101:1559-1568.
  10. Barrett J.C., Hansoul S., Nicolae D.L. et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease. Nat Genet 2008; 40: 955-962.
  11. Lee Y.H., Song G.G. Pathway analysis of a genome-wide association study of ileal Crohn's disease. DNA Cell Biol 2012; 31 (10): 1549-1554.
  12. Tsianos E.V., Katsanos K.H., Tsianos V.E. Role of genetics in the diagnosis and prognosis of Crohn's disease. World J Gastroenterol 2012; 18 (2): 105-118.
  13. Маев И.В., Андреев Д.Н. Молекулярно-генетические механизмы развития болезни Крона. Мол мед 2014; 3: 21-27.
  14. Hugot J.P., Chamaillard M., Zouali H. et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature 2001; 411: 599-603.
  15. Ogura Y., Bonen D.K., Inohara N. et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature 2001; 411: 603-606.
  16. Girardin S.E., Boneca I.G., Viala J. et al. Nod2 is a general sensor of peptidoglycan through muramyl dipeptide (MDP) detection. J Biol Chem 2003; 278 (11): 8869-8872.
  17. Grimes C.L., Ariyananda Lde Z., Melnyk J.E., O'Shea E.K. The innate immune protein Nod2 binds directly to MDP, a bacterial cell wall fragment. J Am Chem Soc 2012; 134 (33):13535-13537.
  18. Maev I.V., Andreev D.N. Role of mutations in NOD2/CARD15, ATG16L1, and IRGM in the pathogenesis of Crohn's disease. Intern J Biomed 2014; 4 (1): 7-10.
  19. Maev I.V., Andreev D.N. Moderne vedute sul contributo delle mutazioni del gene NOD2/CARD15 nell'eziopatogenesi della malattia di Crohn. Italian Science Rev 2014; 3 (12): 122-126.
  20. Zhou Z., Lin X.Y., Akolkar P.N. et al. Variation at NOD2/CARD15 in familial and sporadic cases of Crohn's disease in the Ashkenazi Jewish population. Am J Gastroenterol 2002; 97: 3095-3101.
  21. Hampe J., Franke A., Rosenstiel P. et al. A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat Genet 2007; 39 (2): 207-211.
  22. Rioux J.D., Xavier R.J., Taylor K.D. et al. Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat Genet 2007; 39: 596-604.
  23. Cooney R., Baker J., Brain O. et al. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat Med 2010; 16 (1): 90-97.
  24. Travassos L.H., Carneiro L.A., Ramjeet M. et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat Immunol 2010; 11: 55-62.
  25. Das K.M., Seril D.N. Mycobacterium avium Subspecies paratuberculosis in Crohn's disease: the puzzle continues. J Clin Gastroenterol 2012; 46 (8): 627-628.
  26. Smith E.J., Thompson A.P., O'Driscoll A., Clarke D.J. Pathogenesis of adherent-invasive Escherichia coli. Future Microbiol 2013; 8 (10): 1289-1300
  27. Duerr R.H., Taylor K.D., Brant S.R. et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 2006; 314 (5804): 1461-1463.
  28. Siakavellas S.I., Bamias G. Role of the IL-23/IL-17 axis in Crohn's disease. Discov Med 2012; 14 (77): 253-262.
  29. Polgar N., Csongei V., Szabo M. et al. Investigation of JAK2, STAT3 and CCR6 polymorphisms and their gene-gene interactions in inflammatory bowel disease. Int J Immunogenet 2012; 39 (3): 247-252.
  30. Конович Е.А., Халиф И.Л., Шапина М.В. Иммунопатогенез воспалительных заболеваний кишечника. Рос журн гастроэнтерол гепатол колопроктол 2013; 4: 69-78.
  31. Marcuzzi A., Bianco A.M., Girardelli M. et al. Genetic and functional profiling of Crohn's disease: autophagy mechanism and susceptibility to infectious diseases. Biomed Res Int 2013; 2013: 297501.
  32. Jess T., Riis L., Vind I. et al. Changes in clinical characteristics, course, and prognosis of inflammatory bowel disease during the last 5 decades: A population-based study from Copenhagen, Denmark. Inflamm. Bowel Dis 2007; 13: 481-489.
  33. Annunziata M.L., Caviglia R., Papparella L.G., Cicala M. 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.
  34. Silverberg M.S., Satsangi J., Ahmad T. et al. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: Report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Can J Gastroenterol 2005; 19 (suppl. A): 5-36.
  35. Cosnes J., Cattan S., Blain A. et al. Long-term evolution of disease behavior of Crohn's disease. Inflamm Bowel Dis 2002; 8: 244-250.
  36. Best W.R., Becktel J.M., Singleton J.W., Kern F. Jr. Development of a Crohn's disease activity index. National Cooperative Crohn's Disease Study. Gastroenterology 1976; 70 (3): 439-444.
  37. Pimentel M., Chang M., Chow E.J. et al. Identification of a prodromal period in Crohn's disease but not ulcerative colitis. Am J Gastroenterol 2000; 95: 3458.
  38. Burgmann T., Clara I., Graff L. et al. The Manitoba Inflammatory Bowel Disease Cohort Study: prolonged symptoms before diagnosis - how much is irritable bowel syndrome? Clin Gastroenterol Hepatol 2006; 4: 614.
  39. Podolsky D.K. Inflammatory bowel disease. N Engl J Med 2002; 347: 417-429.
  40. Ingle S.B., Loftus E.V. Jr. The natural history of perianal Crohn's disease. Dig Liver Dis 2007; 39 (10): 963-969.
  41. Yamaguchi A., Matsui T., Sakurai T. et al. The clinical characteristics and outcome of intraabdominal abscess in Crohn's disease. J Gastroenterol 2004; 39: 441-448.
  42. Гастроэнтерология. Национальное руководство. Под ред. В.Т. Ивашкина, Т.Л. Лапиной. М: ГЭОТАР-Медиа 2008.
  43. Bernstein C.N., Blanchard J.F., Rawsthorne P., Yu N. The prevalence of extraintestinal diseases in inflammatory bowel disease: A population-based study. Am J Gastroenterol 2001; 96: 1116-1122.
  44. Jose F.A., Garnett E.A., Vittinghoff E. et al. Development of extraintestinal manifestations in pediatric patients with inflammatory bowel disease. Inflamm Bowel Dis 2009; 15: 63-68.
  45. Vavricka S.R., Brun L., Ballabeni P. et al. Frequency and risk factors for extraintestinal manifestations in the Swiss inflammatory bowel disease cohort. Am J Gastroenterol 2011; 106: 110-119.
  46. Lapidus A., Bernell O., Hellers G. et al. Clinical course of colorectal Crohn's disease: a 35-year follow-up study of 507 patients. Gastroenterology 1998; 114: 1151-1160.
  47. Baumgart D.C., Sandborn W.J. Crohn's disease. Lancet 2012; 380 (9853): 1590-1605.
  48. Lennard-Jones J.E., 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 (4): 353-359.
  49. Panés J., Bouzas R., Chaparro M. 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.
  50. Lin M.F., Narra V. Developing role of magnetic resonance imaging in Crohn's disease. Curr Opin Gastroenterol 2008; 24: 135.
  51. Dotan I. Serologic markers in inflammatory bowel disease: tools for better diagnosis and disease stratification. Expert Rev Gastroenterol Hepatol 2007; 1: 265.
  52. Mosli M., Al Beshir M., Al-Judaibi B. et al. Advances in the diagnosis and management of inflammatory bowel disease: Challenges and uncertainties. Saudi J Gastroenterol 2014; 20 (2): 81-101.
  53. Vermeire S., Van Assche G., Rutgeerts P. C-reactive protein as a marker for inflammatory bowel disease. Inflamm Bowel Dis 2004; 10: 661.
  54. Solem C.A., Loftus E.V., Tremaine W.J. Correlation of C-reactive protein (CRP) with clinical, radiographic, and endoscopic activity in inflammatory bowel disease (abstract). Gastroenterology 2004; 26 (Suppl): A477.
  55. Chamouard P., Richert Z., Meyer N. et al. Diagnostic value of C-reactive protein for predicting activity level of Crohn's disease. Clin Gastroenterol Hepatol 2006; 4: 882.
  56. Boirivant M., Leoni M., Tariciotti D. et al. The clinical significance of serum C reactive protein levels in Crohn's disease. Results of a prospective longitudinal study. J Clin Gastroenterol 1988; 10: 401.
  57. Consigny Y., Modigliani R., Colombel J.F. Biological markers of short term relapse in Crohn's disease (abstract). Gastroenterology 2001; 20:A53.
  58. Jürgens M., Mahachie John J.M., Cleynen I. et al. Levels of C-reactive protein are associated with response to infliximab therapy in patients with Crohn's disease. Clin Gastroenterol Hepatol 2011; 9: 421.
  59. Kuna A.T. Serological markers of inflammatory bowel disease. Biochem Med (Zagreb). 2013; 23 (1): 28-42.
  60. Peeters M., Joossens S., Vermeire S. et al. Diagnostic value of anti-Saccharomyces cerevisiae and antineutrophil cytoplasmic autoantibodies in inflammatory bowel disease. Am J Gastroenterol 2001; 96:730.
  61. Mow W.S., Vasiliauskas E.A., Lin Y.C. et al. Association of antibody responses to microbial antigens and complications of small bowel Crohn's disease. Gastroenterology 2004; 126: 414.
  62. Turner D., Leach S.T., Mack D. et al. Faecal calprotectin, lactoferrin, M2-pyruvate kinase and S100A12 in severe ulcerative colitis: A prospective multicentre comparison of predicting outcomes and monitoring response. Gut 2010; 59: 1207-1212.
  63. van Rheenen P.F., Van de Vijver E., Fidler V. Faecal calprotectin for screening of patients with suspected inflammatory bowel disease: diagnostic meta-analysis. BMJ 2010; 341: c3369.
  64. Carter M.J., Lobo A.J., Travis S.P. IBD Section, British Society of Gastroenterology. Guidelines for the management of inflammatory bowel disease in adults. Gut 2004; 53 (Suppl 5): V1-V16.
  65. Steinhart A.H., Ewe K., Griffiths A.M. et al. Corticosteroids for maintaining remission of Crohn's disease. Cochrane Database Syst Rev 2003 (4): CD000301.
  66. Dignass A., Van Assche G., Lindsay J.O. et al. The second European evidence-based Consensus on the diagnosis and management of Crohn's disease: Current management. J Crohns Colitis 2010; 4 (1): 28-62.
  67. Проект клинических рекомендаций по диагностике и лечению взрослых пациентов с болезнью Крона. Колопроктология 2013; 3: 22-38.
  68. Bar-Meir S., Chowers Y., Lavy A. et al. Budesonide versus prednisone in the treatment of active Crohn's disease. The Israeli Budesonide Study Group. Gastroenterology 1998; 115: 835-840.
  69. Халиф И.Л., Белоусова Е.А. Консервативная терапия воспалительных заболеваний кишечника в схемах: пособие для врачей. М: Форте принт 2014.
  70. Tromm A., Bunganic I., Tomsova E. et al. Double-blind, double-dummy, randomised, multicentre study to compare the efficacy and safety of oral budesonide (9mg) and oral mesalazine (4.5g) in moderately active Crohn's disease patients. Gastroenterology 2009; 139 (Suppl. 1): 391.
  71. Hanauer S.B., 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.
  72. Sandborn W.J., Feagan B.G., Lichtenstein G.R. Medical management of mild to moderate Crohn's disease: Evidence-based treatment algorithms for induction and maintenance of remission. Aliment Pharmacol Ther 2007; 26: 987-1003.
  73. Белоусова Е.А. Рекомендации по диагностике и лечению взрослых пациентов с болезнью Крона (2013). Фарматека 2013; 14: 34-43.
  74. Toussirot E. The IL23/Th17 pathway as a therapeutic target in chronic inflammatory diseases. Inflamm Allergy Drug Targets 2012; 11 (2): 159-168.
  75. Mannon P.J., Fuss I.J., Mayer L. et al. The Anti-IL-12 Crohn's Disease Study Group. Anti-interleukin-12 antibody for active Crohn's disease. N Engl J Med 2004; 351: 2069-2079.
  76. Sandborn W.J., Feagan B.G., Fedorak R.N. et al. The Ustekinumab Crohn's Disease Study Group. 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.
  77. Khanna R., Feagan B.G. Ustekinumab for the treatment of Crohn's disease. Immunotherapy 2013; 5 (8): 803-815.
  78. Mozaffari S., Nikfar S., Abdolghaffari A.H., Abdollahi M. New biologic therapeutics for ulcerative colitis and Crohn's disease. Expert Opin Biol Ther 2014; 14 (5): 583-600.
  79. Thomas S., Baumgart D.C. Targeting leukocyte migration and adhesion in Crohn's disease and ulcerative colitis. Inflammopharmacology 2012; 20 (1): 1-18.
  80. Bamias G., Clark D.J., Rivera-Nieves J. Leukocyte traffic blockade as a therapeutic strategy in inflammatory bowel disease. Curr Drug Targets 2013; 14 (12):1490-500.
  81. Berlin C., Berg E.L., Briskin M.J. et al. Alpha 4 beta 7 integrin mediates lymphocyte binding to the mucosal vascular addressin MAdCAM-1. Cell. 1993; 74: 185-195.
  82. Briskin M., Winsor-Hines D., Shyjan A. et al. Human mucosal addressin cell adhesion molecule-1 is preferentially expressed in intestinal tract and associated lymphoid tissue. Am J Pathol 1997; 151: 97-110.
  83. Adams D.H., Eksteen B. Aberrant homing of mucosal T cells and extra-intestinal manifestations of inflammatory bowel disease. Nat Rev Immunol 2006; 6: 244-251.
  84. Sakuraba A., Keyashian K., Correia C. et al. Natalizumab in Crohn's disease: results from a US tertiary inflammatory bowel disease center. Inflamm Bowel Dis 2013; 19 (3): 621-626.
  85. Toussirot E., Bereau M. The Risk of Progressive Multifocal Leukoencephalopathy Under Biological Agents Used in the Treatment of Chronic Inflammatory Diseases. Inflamm Allergy Drug Targets. 2014; in press.
  86. Soler D., Chapman T., Yang L.L. et al. The binding specificity and selective antagonism of vedolizumab, an anti-alpha4beta7 integrin therapeutic antibody in development for inflammatory bowel diseases. J Pharmacol Exp Ther 2009; 330: 864-875.
  87. Sandborn W.J., Feagan B.G., Rutgeerts P. et al. Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med 2013; 369 (8): 711-721.
  88. Lobatón T., Vermeire S., Van Assche G., Rutgeerts P. Review article: anti-adhesion therapies for inflammatory bowel disease. Aliment Pharmacol Ther 2014; 39 (6): 579-594.

Дополнительные файлы

Доп. файлы
Действие
1. JATS XML

© ООО "Консилиум Медикум", 2014

Creative Commons License
Эта статья доступна по лицензии Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License.
 

Адрес издателя

  • 127055, г. Москва, Алабяна ул., 13, корп.1

Адрес редакции

  • 127055, г. Москва, Алабяна ул., 13, корп.1

По вопросам публикаций

  • +7 (926) 905-41-26
  • editor@ter-arkhiv.ru

По вопросам рекламы

  • +7 (495) 098-03-59

 

 


Данный сайт использует cookie-файлы

Продолжая использовать наш сайт, вы даете согласие на обработку файлов cookie, которые обеспечивают правильную работу сайта.

О куки-файлах