Submission of the manuscript is online via e-mail
ecgarticle@gmail.com or
cholerez@mail.ru

Tel: +7 903 250 5288

Editorial Correspondence e-mail
gastrossr@gmail.com


Publishing, Subscriptions, Sales and Advertising, Correspondence e-mail
journal@cniig.ru

Tel: +7 917 561 9505

SCImago Journal & Country Rank

№ 04 (164) 2019

  • Дорогие коллеги, читатели журнала «Экспериментальная и клиническая гастроэнтерология»!
    Технократический тип развития современного общества и появление новых медицинских технологий позволяет повысить диагностическую и лечебную значимость применяемых методов, целью которых является прежде всего, здоровье и качество жизни наших пациентов. Известны слова классик Русской литературы Н.Г Чернышевский: «Медицина слагается из науки и искусства, и над ними простирается чудесный покров героизма». Четвертый номер, по традиции, является инструментальным, в котором находит отражение прикладное клиническое использование новых медицинских технологий в области гастроэнтерологии. Открывает номер крайне интересная статья группы авторов из Первого Московского государственного медицинского университета имени И. М. Сеченова, касающаяся возможностей хирургического лечения метастазов почечно-клеточного рака в головку поджелудочной железы, редкой и крайне опасной патологии. Однако точно поставленный диагноз позволяет провести хирургическое лечение и повысить уровень безрецидивной выживаемости пациентов. Рост колоректального рака в мире заставляет медиков уделять все большее внимание заболевания толстой кишки. Возможности ранней диагностики и дифференциальной диагностики воспалительных и неопластических заболеваний толстой кишки продолжают оставаться важнейшей задачей современной гастроэнтерологии и онкологии. Генетические маркеры, эндовидеокапсульные технологии, усовершенствованые эндоскопические методы исследования толстой кишки с возможностью кругового обзора – все это далеко неполный перечень направлений исследования. В связи с этим ряд статей номера посвящены заболеваниям толстой кишки. Среди них особое место занимает исследование авторов, под руководством Луговкиной А. А. из «Волгоградского государственного медицинского университета», которая посвящена диагностике и лечению неспецифического язвенного колит. Широкое применение НПВП в ежедневной клинической практике и безконтрольный прием этой группы препаратов в быту увеличивает количество связанных с их приемом нежелательных побочных эффектов, в том числе со стороны толстой кишки. Понимание специфики и дифференциальной диагностики воспалительных заболеваний толстой кишки на фоне приема препаратов с хроническими, подчас системными, заболеваниями даёт возможности правильного понимания их генеза и назначения адекватной корригирующей терапии. Этому посвящена работа Г. В. Беловой и Е. А. Соловьева. Особенно интересен взгляд эндоскописта на данную проблему. Ранней зндоскопической флюорисцентной диагностике и дифференциальной диагностике доброкачественных и злокачественных эпителиальных новообразований толстой кишки посвящена работа М. В. Князева и В. А. Дуванского, которая в связи с вышеизложенным является крайне своевременной. Непосредственно онкологическим проблемам в области колопроктологии посвящены работы А. С. Вололеева с соавт., В. А. Дуванского с соавт., С. Н. Волкова с соавт, Д. А. Черных с соавт. Первый опыт проведения полноспектральной (панорамной) колоноскопии у детей отражен в работе Т. Н. Будкиной и М. В. Лохматова. Вызывают постоянный интерес работы из ведущих клиник Российской Федерации, посвященные эндоскопическим проблемам в гастроэнтерологии. Среди них: «Эндоскопические и морфологические факторы прогноза течения раннего рака желудка» Л. М. Михалёва, под руководством проф. Е. Д. Федорова, «Потенциал малигнизации кистозных неоплазий поджелудочной железы» Е. Г. Солоницына с соавт, под руководством проф. В. А. Кащенко из «Санкт-Петербургского государственногой университета» и «НМИЦ им. В. А. Алмазова» и «Эндоскопическая баллонная дилатация при ахалазии кардии в эпоху пероральной эндоскопической миотомии (POEM). Тридцатилетний опыт» проф. Э. А. Годжелло и проф. М. В. Хрусталевой с соавт. Из «Российскогг научного центра хирургии имени академика Б. В. Петровского». Заслуживает особого внимания, опубликованная в номере работа Т. И. Власовой и соавт. из Национального исследовательского Мордовского государственного университета им. Н. П. Огарёва, посвященная связи хронического пародонтита тяжелой степени и гастропатии, которая позволяет обратить внимание на связь заболеваний ротовой полости с заболеваниями желудочно-кишечного тракта. Новым направлением является использование синхронной КТ денситометрия в оценке влияния оперативного лечения верхних отделов желудочно-кишечного тракта на минеральную плотность кости. Работа является результатом совместного исследования, проведенным «Научно-практическим клинический центр диагностики и телемедицинских технологий Департамента здравоохранения города Москвы», Первым МГМУ им. И. М. Сеченова и Московский государственный университетом им. М. В. Ломоносова. Опубликованный материал позволяет задуматься о послеоперационном ведении пациентов в плане минерального обмена, что несомненно влияет на их качество жизни. Дискуссионные вопросы гастроэзофагеальной рефлюксной болезни на основе клинико-эндоскопических исследований отражены в работе Дронова О. Б. и соавт., включающей в себя, как всегда, вопросы клиники, патанатомии и патофизиологии. Хирургические осложнения абдоминального туберкулеза у пациентов с различным иммунным статусом отражены в совместной работе Решетникова М. Н. и соавт. Из «Московского городского научно-практического центра борьбы с туберкулезом» и «Российского национального исследовательского медицинского университета им. Н. И. Пирогова» Всегда приоритетными в области инноваций в эндоскопии являются работы Габриэля С. А. с соавт. из Краснодара. В этом номере их работы посвящены применению перорального холедохоскопа при рубцовых стриктурах общего желчного протока и собственному опыту лечения высокой обтурационной кишечной непроходимости, обусловленной билиарными конкрементами. Коллегиальность в работе гастроэнтерологов и врачей лабораторного дела общеизвестна. В связи с этим актуальным является опубликованный обзор методов лабораторной диагностики, применяемых при неалкогольной жировой болезни печени (НАЖБП) и алкогольной болезни печени (АБП) Л. В. Тарасовой и Ю. В. Цыгановой из «Чувашского государственного университета им. И. Н. Ульянова», Чебоксары и «Сургутского государственного университета и статья, посвященная Хемомикробиомному анализу Лактитола О. А. Громовой и И. Ю. Трошина. Желаю приятного чтения!
    Ответственные за выпуск редакторы.

    Заместитель главного врача по амбулаторно-поликлинической работе

    Многопрофильного медицинского центра Банка России-

    заведующая поликлиникой д. м. н.

    Белова Галина Вячеславовна  
       
    1. The State Education Institution of Higher Professional Training The First Sechenov Moscow State Medical University under Ministry of Health of the Russian Federation (Sechenov University), 119991, Moscow, Russia
    2. Moscow State University of Medicine and Dentistry named after A. I. Evdokimov, 127473, Moscow, Russia
    3. Federal State Budget Institution “A. V. Vishnevsky National Medical Research Center of Surgery” of the Ministry of Health of the Russian Federation, 117997, Moscow, Russia

    Keywords: Renal cell carcinoma, pancreatic head metastasis, surgical treatment

    Abstract: This review reflects current data on the possibilities of surgical treatment of patients with metastases of renal cell carcinoma in the pancreas in the aspect of complex therapy. The data obtained are ambiguous due to small patient samples, the extreme rarity of metastatic lesions of this localization and the ambiguity of approaches to surgical treatment. The prerequisites for performing duodenum-preserving pancreatic head resection as an operation that has a high potential of a favorable quality of life of patients in the long-term postoperative period.

      1. Chatzizacharias NA, Rosich-Medina A, Dajani K, Harper S, Huguet E, Liau SS, Praseedom RK, Jah A. Surgical management of hepato-pancreatic metastasis from renal cell carcinoma. World J Gastrointest Oncol. 2017;9(2):70-7. https://doi.org/10.4251/wjgo.v9.i2.70.
      2. Polushin OG, Fedosenko KV, Ponomareva EV, Topuzov RE, Kislitsyna ON. Late pancreatic metastases from renal cell carcinoma: A case report. Arkhiv patologii. 2016;78(5):37-39. https://doi.org/10.17116/patol201678537-39
      3. Thadani A, Pais S, Savino J. Metastasis of renal cell carcinoma to the pancreas 13 years postnenhrectomv. Gastroenterol Hepatol (N Y). 2011; 7(10):697-699.
      4. Sperti C, Moletta L, Patanè G. Metastatic tumors to the pancreas: The role of surgery. World J Gastrointest Oncol. 2014;6(10):381-392. https://doi.org/10.4251/wjgo.v6.i10.381.
      5. Shatveryan GA, Chardarov NK, Bagmet NN, Ratnikova NP, Bedzhanyan AL, Petrenko KN, Polishchuk LO, Karagyozyan GA. Isolated pancreatic metastases of renal cell carcinoma. Khirurgiia (Mosk). 2017;(12):36-40. doi: 10.17116/hirurgia20171236-40.
      6. Lee SR, Gemenetzis G, Cooper M, Javed AA, Cameron JL, Wolfgang CL, Eckhauser FE, He J, Weiss MJ. Long-Term Outcomes of 98 Surgically Resected Metastatic Tumors in the Pancreas. Ann Surg Oncol. 2017;24(3):801-807. https://doi.org/ 10.1245/s10434-016-5619-z.
      7. Strobel O, Hackert T, Hartwig W, Bergmann F, Hinz U, Wente MN, Fritz S, Schneider L, Büchler MW, Werner J. Survival data justifies resection for pancreatic metastases. Ann Surg Oncol. 2009;16(12):3340-9. https://doi.org/ 10.1245/s10434-009-0682-3.
      8. Yagi T, Hashimoto D, Taki K, Yamamura K, Chikamoto A, Ohmuraya M, Beppu T, Baba H. Surgery for metastatic tumors of the pancreas. Surg Case Rep. 2017;3(1):31. https://doi.org/ 10.1186/s40792-017-0308-0.
      9. Kriger AG, Paklina OV, Kochatkov AV, Vetsheva NN, Filippova EM, Makeeva-Malinovskaia NIu, Berelavichus SV, Svitina KA. The metastatic invasion of pancreas by renal cancer. Khirurgiia (Mosk). 2012;(9):26-31.
      10. Cheng SK, Chuah KL. Metastatic Renal Cell Carcinoma to the Pancreas: A Review. Arch Pathol Lab Med. 2016;140(6):598-602. https://doi.org/10.5858/arpa.2015-0135-RS.
      11. David AW, Samuel R, Eapen A, Vyas F, Joseph P, Sitaram V. Pancreatic metastasis from renal cell carcinoma 16 years after nephrectomy: a case report and review of the literature. Trop Gastroenterol. 2006;27(4):175-6.
      12. Sellner F, Tykalsky N, De Santis M, Pont J, Klimpfinger M. Solitary and multiple isolated metastases of clear cell renal carcinoma to the pancreas: an indication for pancreatic surgery. Ann Surg Oncol. 2006;13:75-85. https://doi.org/ 10.1245/ASO.2006.03.064
      13. Tanis PJ, van der Gaag NA, Busch OR, van Gulik TM, Gouma DJ. Systematic review of pancreatic surgery for metastatic renal cell carcinoma. Br J Surg. 2009;96:579-592. https://doi.org/ 10.1002/bjs.6606.
      14. Karmazanovskij GG, Kriger АG, Vetsheva NN, Kochatkov AV, Berelavichus SV. Metastases to the Pancreas from Renal Carcinoma (Literature Review and Case Report). Meditsinskaya vizualizatsiya. 2012; (4):49-54. (In Russ.)
      15. Pannala R, Hallberg-Wallace KM, Smith AL, Nassar A, Zhang J, Zarka M, Reynolds JP, Chen L. Endoscopic ultrasound-guided fine needle aspiration cytology of metastatic renal cell carcinoma to the pancreas: A multi-center experience. Cytojournal. 2016;13:24. https://doi.org/ 10.4103/1742-6413.192191
      16. Grassi P, Doucet L, Giglione P, Grünwald V, Melichar B, Galli L, De Giorgi U, Sabbatini R, Ortega C, Santoni M, Bamias A, Verzoni E, Derosa L, Studentova H, Pacifici M, Coppa J, Mazzaferro V, de Braud F, Porta C, Escudier B, Procopio G. Clinical Impact of Pancreatic Metastases from Renal Cell Carcinoma: A Multicenter Retrospective Analysis. PLoS One. 2016;11(4): e0151662. https://doi.org/ 10.1371/journal.pone.0151662.
      17. Grassi P, Verzoni E, Mariani L, De Braud F, Coppa J, Mazzaferro V, Procopio G. Prognostic role of pancreatic metastases from renal cell carcinoma: results from an Italian center. Clin Genitourin Cancer. 2013;11(4):484-8. https://doi.org/ 10.1016/j.clgc.2013.04.022.
      18. Heng DY, Xie W, Regan MM, Warren MA, Golshayan AR, Sahi C, Eigl BJ, Ruether JD, Cheng T, North S, Venner P, Knox JJ, Chi KN, Kollmannsberger C, McDermott DF, Oh WK, Atkins MB, Bukowski RM, Rini BI, Choueiri TK. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol. 2009;27(34):5794-9. https://doi.org/ 10.1200/JCO.2008.21.4809.
      19. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A, Zarbá JJ, Chen M, McCann L, Pandite L, Roychowdhury DF, Hawkins RE. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010;28(6):1061-8. https://doi.org/ 10.1200/JCO.2009.23.9764.
      20. Jakubowski CD, Vertosick EA, Untch BR, Sjoberg D, Wei E, Palmer FL, Patel SG, Downey RJ, Strong VE, Russo P. Complete metastasectomy for renal cell carcinoma: Comparison of five solid organ sites. J Surg Oncol. 2016;114(3):375-9. https://doi.org/ 10.1002/jso.24327.
      21. Nakagohri T, Konishi M, Inoue K, Nakamura T, Kinoshita T. Partial pancreatic head resection for pancreatic metastasis from renal cell carcinoma. Hepatogastroenterology. 2003;50(54):2236-8.
      22. Moletta L, Milanetto AC, Vincenzi V, Alaggio R, Pedrazzoli S, Pasquali C. Pancreatic secondary lesions from renal cell carcinoma. World J Surg. 2014;38(11):3002-6. https://doi.org/ 10.1007/s00268-014-2672-2.
      23. Motzer RJ, Mazumdar M, Bacik J, Berg W, Amsterdam A, Ferrara J. Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma. J Clin Oncol. 1999;17(8):2530-40. https://doi.org/10.1200/JCO.1999.17.8.2530.
      24. Nihei K, Sakamoto K, Suzuki S, Mishina T, Otaki M. A Case of Pancreatic Metastasis of Renal Cell Carcinoma. Gan To Kagaku Ryoho. 2016;43(12):2274-2276.
      25. Ko S, Yun S, Kim S, Kim TN, Seo HI. Pancreatic resection for renal cell carcinoma metastasis: a case review. Ann Hepatobiliary Pancreat Surg. 2017;21(3):176-179. https://doi.org/ 10.14701/ahbps.2017.21.3.176.
      26. Santoni M, Conti A, Partelli S, Porta C, Sternberg CN, Procopio G, Bracarda S, Basso U, De Giorgi U, Derosa L. Surgical resection does not improve survival in patients with renal metastases to the pancreas in the era of tyrosine kinase inhibitors. Ann Surg Oncol. 2015;22:2094-2100. https://doi.org/ 10.1245 / s10434-014-4256-7.
      27. Kalra S, Atkinson BJ, Matrana MR, Matin SF, Wood CG, Karam JA, Tamboli P, Sircar K, Rao P, Corn PG, Tannir NM, Jonasch E. Prognosis of patients with metastatic renal cell carcinoma and pancreatic metastases. BJU Int. 2016;117(5):761-5. https://doi.org/ 10.1111/bju.13185.
      28. Maeda H, Okabayashi T, Nishimori I, Kobayashi M, Sugimoto T, Kohsaki T, Onishi S, Hanazaki K. Duodenum-preserving pancreatic head resection for pancreatic metastasis from renal cell carcinoma: a case report. Langenbecks Arch Surg. 2007;392(5):649-52. https://doi.org/ 10.1007/s00423-007-0204-3
      29. Ballarin R, Spaggiari M, Cautero N, De Ruvo N, Montalti R, Longo C, Pecchi A, Giacobazzi P, De Marco G, D’Amico G. Pancreatic metastases from renal cell carcinoma: the state of the art. World J Gastroenterol. 2011;17:4747-4756. https://doi.org/ 10.3748/wjg.v17.i43.4747.
      30. Beger HG, Krautzberger W, Bittner R, Büchler M, Limmer J. Duodenum-preserving resection of the head of the pancreas in patients with severe chronic pancreatitis. Surgery. 1985;97(4):467-73.
      31. Ahn YJ, Kim SW, Park YC, Jang JY, Yoon YS, Park YH. Duodenal-preserving resection of the head of the pancreas and pancreatic head resection with second-portion duodenectomy for benign lesions, low-grade malignancies, and early carcinoma involving the periampullary region. Arch Surg. 2003;138(2):162-8; discussion 168.
      32. Beger HG, Nakao A, Mayer B, Poch B. Duodenum-preserving total and partial pancreatic head resection for benign tumors-systematic review and meta-analysis. Pancreatology. 2015;15(2):167-78. https://doi.org/ 10.1016/j.pan.2015.01.009.
      33. Hatori T, Kimijima A, Fujita I, Furukawa T, Yamamoto M. Duodenum-preserving total pancreatectomy for pancreatic neoplasms. J Hepatobiliary Pancreat Sci. 2010;17(6):824-30. https://doi.org/ 10.1007/s00534-009-0225-0.
      34. Bassi C, Butturini G, Falconi M, Sargenti M, Mantovani W, Pederzoli P. High recurrence rate after atypical resection for pancreatic metastases from renal cell carcinoma. Br J Surg. 2003;90:555-559. https://doi.org/ 10.1002/bjs.4072
      35. Kimura W, Nagai H. Study of surgical anatomy for duodenum-preserving resection of the head of the pancreas. Ann Surg. 1995;221(4):359-63.
      36. Kimura W, Morikane K, Futakawa N, Shinkai H, Han I, Inoue T, Muto T, Nagai H. A new method of duodenum-preserving subtotal resection of the head of the pancreas based on the surgical anatomy. Hepatogastroenterology. 1996;43(8):463-72.
      37. Nakao A. Pancreatic head resection with segmental duodenectomy and preservation of the gastroduodenal artery. Hepatogastroenterology. 1998;45(20):533-5.
      38. Isaji S, Kawarada Y. Pancreatic head resection with second-portion duodenectomy for benign lesions, low-grade malignancies, and early stage carcinomas involving the pancreatic head region. Am J Surg. 2001;181(2):172-6.
      39. Zerbi A, Ortolano E, Balzano G, Borri A, Beneduce AA, Di Carlo V. Pancreatic metastasis from renal cell carcinoma: which patients benefit from surgical resection? Ann Surg Oncol. 2008;15:1161-1168. https://doi.org/ 10.1245/s10434-007-9782-0.
     


    Full text is published :
    Zharikov Yu. O., Tupikin K. A., Baidarova M. D., Pozharskaya A. A. Metastatic renal cell carcinoma in the head of pancreas: the possibility of surgical treatment. Experimental and Clinical Gastroenterology. 2019;164(4): 4–9. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-4-9
    Read & Download full text

    1. Federal state budgetary educational institution of higher education “Volgograd state medical University” of the Ministry of health of the Russian Federation, 400131, Russian Federation, Volgograd, Pavshikh Bortsov Sq., 1

    Keywords: nonspecifi c ulcer colitis, diagnostics, treatment

    Abstract: Nonspecific ulcer colitis remains one and most complex problems in gastroenterology today. The chronic nonspecific diffusion inflammatory canker of slime and submucosal layer of a direct and large intestine is the cornerstone of this pathology that causes the clinical picture which is shown in the form of abdominal pains, tenezm, obstipation, intestinal bleedings, a frequent liquid chair with impurity of blood and pus. Also nonspecific ulcer colitis can be shown by various abenteric manifestations that causes certain difficulties in early diagnosis and treatments of this disease. Article narrates about the main strong points for the general practitioners and gastroenterologists facing this disease.

      1. osnes J., Gower-Rousseau C., Seksik P., Cortot A. Epidemiology and natural history of inflammatory bowel diseases. Gastroenterology. 2011;140:1785-94.
      2. Silverberg M.S. 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.
      3. Grigor’ev P. Ya., Yakovenko E. P. Spravochnoe rukovodstvo po gastroehnterologii [The reference guide on gastroenterology]. Moscow, Med. inform. agentstvo Publ., 1997. 480 p. (In Russ.)
      4. Baumgart DC. The Diagnosis and Treatment of Crohn’s Disease and Ulcerative Colitis. Deutsches Ärzteblatt International Dtsch Arztebl Int. 2009;106(8):123-33.
      5. D’Haens G. et al. A review of activity indices and efficacy end points for clinical trials of medical therapy in adults with ulcerative colitis. Gastroenterology 2007;132:763-86.
      6. Grigor’eva G.А., Meshalkina N. Yu. O probleme sistemnykh proyavlenij vospalitel’nykh zabolevanij kishechnika [About a problem of system manifestations of inflammatory diseases of intestines]. Farmateka, 2011, no. 15, pp. 44-49. (In Russ.)
      7. Travis S.P., Dinesen L. Remission in trials of ulcerative colitis: what does it mean? Pract. Gastroenterol. 2010;30:17-20.
      8. Masevich C. G., Sitkin S. I. Sovremennaya farmakoterapiya khronicheskikh vospalitel’nykh zabolevanij kishechnika [Modern pharmacotherapy of chronic inflammatory diseases of intestines]. Aqua Vitae, 2001, no. 1, pp. 37-41. (In Russ.)
      9. Blagodarnogo L. А, Shelygina Yu. А. Spravochnik po koloproktologii [Reference book on coloproctology]. Litterra, 2012, pp. 460-522. (In Russ.)
      10. Khalif I. L., Loranskaya I. D. Vospalitel’nye zabolevaniya kishechnika (nespetsificheskij yazvennyj kolit i bolezn’ Krona): klinika, diagnostika i lechenie [Inflammatory diseases of intestines (nonspecific ulcer colitis and disease Krone): clinic, diagnostics and treatment]. Moscow, Miklosh Publ., 2004. 88 p. (In Russ.)
      11. Shifrin O. S. Sovremennye podkhody k lecheniyu bol’nykh nespetsificheskim yazvennym kolitom [Modern approaches to treatment of patients with nonspecific ulcer colitis]. Consilium-medicum, 2002, vol. 4, no. 6, pp. 24-29. (In Russ.)
      12. Khalif I. L. Printsipy lecheniya yazvennogo kolita (rekomendatsii rossijskoj gruppy po izucheniyu vospalitel’nykh zabolevanij kishechnika) [Principles of treatment of ulcer colitis (recommendation of the Russian group on studying of inflammatory diseases of intestines)]. Koloproktologiya, 2006, no. 2, pp. 31-33. (In Russ.)
      13. Khalif I. L. Lechebnaya taktika pri yazvennom kolite [Medical tactics at ulcer colitis]. Rossijskij zhurnal gastroehnterologii, gepatologii, koloproktologii, 2006, vol. 16, no. 3, pp.58-62. (In Russ.)
      14. Belousova E. A. Iazvennyi kolit i bolezn’ Krona [Ulcer colitis and Crohn’s disease]. Moscow, Triada Publ., 2002. 128 p. (In Russ.)
      15. Belousova E. A., Nikitina N. V., Tsodikov O. M. Lechenie yazvennogo kolita legkogo i srednetyazhelogo techeniya [Treatment of ulcer colitis of an easy and medium-weight current]. Farmateka, 2013, no. 2, pp. 42-46. (In Russ.)
     


    Full text is published :
    Lugovkina A. A., Rudakova L. O., Kryukova N. A., Bessonov A. A., Skvortsov V. V. Features of diagnostics and treatment of nonspecifi c ulcer colitis. Experimental and Clinical Gastroenterology. 2019;164(4): 10–16. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-10-16
    Read & Download full text

    1. Multidisciplinary medical center of the Bank of Russia, Moscow 117593, Russia

    Keywords: NSAID-colopathy, ulcerative colitis, colonoscopy, inflammation

    Abstract: The aim: to focus on a relatively new and little-studied pathology of the colon-NSAID-colopathy, to show the similarities and differences in the clinical and endoscopic picture with ulcerative colitis.
    Materials and methods: comparison, based on the data available in the literature and own observations, using methods of endoscopic diagnosis, pathology of the colon with a similar clinical picture.
    Results: on the basis of the obtained and systematized data, key differences were formed, formulated in the form of a table for a more visual and rapid perception of information and recommendations for the differential diagnosis of NSAID-colopathy and ulcerative colitis were given.
    Conclusion: understanding the specifics and differential diagnosis of inflammatory diseases of the colon improves the quality of therapy of these pathologies.

      1. Sostres C., Gargallo C. J., Lanas A. Nonsteroidal anti-inflamma-tory drugs and upper and lower gastrointestinal mucosal damage. Arthritis Res. Ther. 2013, Vol. 15, suppl. 3, pp. S3
      2. Laine L, Curtis SP, Langman M, et al. Lower gastrointestinal events in a double-blind trial of the cyclo-oxygenase-2 selective inhibitor etoricoxib and the traditional nonsteroidal anti-inflammatory drug diclofenac. Gastroenterology. 2008 Nov;135(5):1517-25. doi: 10.1053/j.gastro.2008.07.067. Epub 2008 Aug 3.
      3. Wallace J. L. Mechanisms, prevention and clinical implications of nonsteroidal anti-inflammatory drug-enteropathy. World J. Gastroenterol. 2013, Vol. 19 (12), pp. 1861-1876
      4. Boelsterli U. A., Redinbo M. R., Saitta K. S. Multiple NSAIDinduced hits injure the small intestine: underlying mechanisms and novel strategies. Toxicol. Sci. 2013, Vol. 131 (2), pp. 654-667
      5. Pardi D. S., Kelly C. P. Gastroenterology. 2011;140:1155-. 1165.
      6. Debenham GP. Ulcer of the cecum during oxyphenbutazone (tandearil) therapy. Can Med Assoc J. 1966; 28: 1182-1184.
      7. Matsui H, Shimokawa O, Kaneko T, Nagano Y, Rai K, Hyodo I. The pathophysiology of non-steroidal antiinflammatory drug (NSAID)-induced mucosal injuries in stomach and small intestine. J Clin Biochem Nutr. 2011; 48: 107-111.
      8. Geramizadeh B, Taghavi A, Banan B. Clinical, endoscopic and pathologic spectrum of non-steroidal antiinflammatory drug-induced colitis. Indian J Gastroenterol. 2009; 28: 150-153.
      9. Ballinger A. Adverse effects of nonsteroidal anti-inflammatory drugs on the colon. Curr Gastroenterol Rep. 2008; 10: 485-489
      10. Lang J, Price AB, Levi AJ, Burke M, Gumpel JM, Bjarnason I. Diaphragm disease: pathology of disease of the small intestine induced by non-steroidal antiinflammatory drugs. J Clin Pathol. 1988; 41: 516-526
      11. Gleeson MH, Davis AJ. Non-steroidal antiinflammatory drugs, aspirin and newly diagnosed colitis: A case-control study. Aliment Pharmacol Ther. 2003; 17: 817-825.
      12. Balabantseva AP, Karateev DE. NSAID-induced colopathy: incidence, clinical and endoscopic manifestations, possibilities of drug therapy. Nauchno-Prakticheskaya Revmatologiya = Rheumatology Science and Practice. 2018;56(5):564-568.
      13. Aftab AR, Donnellan F, Zeb F, et al. NSAID-induced colopathy. A case series. J Gastrointestin Liver Dis. 2010;19:89-91.
      14. Aloysius MM, Kaye PV, Lobo DN. Non-steroidal anti-inflamatory drug (NSAID)-induced colonic strictures and perforation: a case report. Dig Liver Dis. 2006;38:276-8. doi: 10.1016/j.dld.2005.09.003.
      15. Byrne MF, McGuinness J, Smyth CM, et al. Nonsteroidal antiinflammatory drug-induced diaphragms and ulceration in the colon. Eur J Gastroenterol Hepatol. 2002;14:1265-9. doi: 10.1097/00042737-200211000-00017.
      16. El Hajj I, Hawchar M, Sharara A. NSAID-induced colopathy: case report andreview of the literature. J Med Liban. 2009;57:274-6.
      17. Mokhtare M., Valizadeh S. M., Emadian O. Lower gastrointestinal bleeding due to non-steroid anti-inflammatory druginduced colopathy case report and literature review. Middle East J. Dig. Dis. 2013, Vol. 5 (2), pp. 107-111.
      18. Geramizadeh B, Taghavi AR, Banan B. Clinical, endoscopic and pathologic spectrum of non-steroidal anti-inflammatory drug-induced colitis. Indian J Gastroenterol. 2009;28:150-3.
      19. Smith JA, Pineau BC. Endoscopic therapy of NSAID-induced colonic diaphragm disease: two cases and a review of published reports. Gastrointest Endosc. 2000;52:120-5.
      20. Halter F, Weber B, Huber T, Eigenmann F, Frey MP, Ruchti C. Diaphragm disease of the ascending colon. Association with sustained-release diclofenac. J Clin Gastroenterol. 1993;16:74-80.
      21. Adler G. Crohn’s Disease and ulcerative colitis. M, “GEOTAR-Med”, 2001, 220 P.
      22. Belousova E. A. Ulcerative colitis and Crohn’s disease. Tver: LLC “Publishing house “Triada”, 2002, 128 p.
      23. Levitan M. H., Kolosov I. A. Strekalovsky V. P. et al. Clinic and differential diagnosis of ulcerative colitis and Crohn’s disease of the colon. Moscow, 1978, 22p.
      24. Chutkan RK, Waye JD. Endoscopy in inflammatory bowel disease. In: Kirsner JB, Shorter RG, eds. Inflammatory bowel disease, 5th ed. Baltimore: Williams & Wilkins, 2000:453-477.
      25. Farrell RJ, Peppercorn M. Endoscopy in inflammatory bowel disease. In: Sartor RB, Sandborn WJ, eds. Kirsner’s Inflammatory Bowel Diseases, 6th ed. Saunders, 2004: 380-398.
     


    Full text is published :
    Belova G. V., Solovev E. A. Diff erential diagnosis of colopathy induced by nonsteroidal antiinfl ammatory drugs (NSAID-colopapthy) and ulcerative colitis. Experimental and Clinical Gastroenterology. 2019;164(4): 17–20. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-17-20
    Read & Download full text

    1. RUDN University, Moscow, Russia
    2. Skobelkin state scientifi c center of laser medicine, Moscow, Russia
    3. Branch № 2 «Medical Rehabilitation Centre of the Ministry of Economic Development of Russia», Moscow, Russia

    Keywords: endoscopy, colon, screening colonoscopy, autofluorescence, polyps, dysplasia, early cancer

    Abstract: Objective: to determine the dependence of autofluorescent staining of epithelial colon lesions, depending on the histomorphological structure.
    Material and Methods: Video colonoscopy using autofluorescence was performed in 1674 patients. The patients’ age was 58±13.9, men were 761 (45.5%), women 913 (54.5%). Epithelial lesions were detected in 269 patients (16%). Endoscopic examinations were carried out according to a standard procedure using Olympus video colonoscopes, GF-260. Results: It was established that the presence of AF staining in the purple color of the epithelial colon lesions, as a marker of dysplasia, can serve as an additional criterion in the complex indications for their endoscopic removal.
    Conclusion: The developed method of using autofluorescent endoscopy is simple to perform and allows obtaining data that have a differential value on the presence of dysplastic changes in the epithelial colon lesions.

      1. van Rijn JC, Reitsma JB, Stoker J, et al. Polyp miss rate determined by tandem colonoscopy: a systematic review. Am J Gastro. 2006;101(2):343-350
      2. Snover DC, Ahnen DJ, Burt RW, Odze RD Serrated polyps of the colon and rectum and serrated polyposis. In: Bosman FT, Carneiro F, Hruban RH, Theise ND (eds) WHO classification of tumours of the digestive system, 4th edn. IARC, Lyon, (2010), pp. 160-165.
      3. Jass J, Smith M. Sialic acid and epithelial differentiation in colorectal polyps and cancer - a morphological, mucin and lectin histochemical study. Pathol. 1992;24(4):233-42
      4. Ageĭkina NV, Duvanskiĭ VA, Kniazev MV, Mal’kov PG, Danilova NV, Kharlova OA. The alternative way of colorectal cancer developing. The histogenetic and molecular features of serrated lesions (review, continued). Eksp Klin Gastroenterol. 2014;(7):4-12.
      5. Rabeneck L, Paszat LF, Hilsden RJ, et al. Bleeding and perforation after outpatient colonoscopy and their risk factors in usual clinical practice. Gastro. 2008;135(6):1899-1906. 1906. e1;
      6. Shimoda T, Ikegami M, Fujisaki J, et al. Early colorectal carcinoma with special reference to its development de novo. Cancer 1989;64(5):1138-1146
      7. Ageĭkina NV, Duvanskiĭ VA, Kniazev MV. An alternative pathway of colorectal cancer development. Endoscopic and morphological features of serrated lesions. Review. Eksp Klin Gastroenterol. 2013;(8):3-10.
      8. Mäkinen MJ, George SM, Jernvall P, et al. Colorectal carcinoma associated with serrated adenoma - prevalence, histological features, and prognosis. J. Pathol. 2001; 193;286-294
      9. Hawkins N, et al. CpG island methylation in sporadic colorectal cancer and its relationship to microsatellite instability. Gastroenterology. 2002; 122:1376-87
      10. Mäkinen MJ. Colorectal serrated adenocarcinoma. Histopathology. 2007; 50:131-150
      11. Spring K J, Zhao Z Z, Karamatic R, et al. High prevalence of sessile serrated adenomas with BRAF mutations: a prospective study of patients undergoing colonoscopy. Gastro 2006; 131:1400-1407
      12. Weston AP, Campbell DR. Diminutive colonic polyps: histopathology, spatial distribution, concomitant significant lesions, and treatment complications. Am. J. Gastro. 1995; 90; 24-28
      13. Higuchi T, Sugihara K, Jass JR. Demographic and pathological characteristics of serrated polyps of colorectum. Histo. 2005; 47; 32-40
      14. Haws AL. Pathology of Serrated Colon Adenomas. Ed. Mamoun Younes, 2012 http://emedicine.medscape.com/article/1731536-overview
      15. Nikishaev V. I., Patiy A. R., Tumak I. N. et al. Endoscopic diagnosis of early colorectal cancer. Ukrainian Journal of Minimally Invasive and Endoscopic Surgery. 2012, vol.16, no.1, 35-55.
      16. Knyazev MV, Duvanskiy VA. Endoscopic mucosal resection with submucosal dissection esd and the first long-term results of applying this method. Eksp Klin Gastroenterol. 2015;(4):53-8.
      17. Nikishaev V. I., Patiy A. R., Tumak I. N. et al. Endoscopic diagnosis of early colorectal cancer. Ukrainian Journal of Minimally Invasive and Endoscopic Surgery. 2012, vol. 16, no.1, 35-55.
      18. Knyazev M. V., Duvanskii V. A., Ageikina N. V. Trimodal'naya endoskopiya v diagnostike zabolevanii zheludochno-kishechnogo trakta. Klin. endoskop. 2012. vol. 4. p. 2.
      19. Douvansky V. A. Kniazev M. V. Autofluorescent endoscopic diagnostics of epithelial neoplasms in the colon. Journal of Gastro and Hep. 2015; 30 (Suppl. 4): 211.2
     


    Full text is published :
    Knyazev М. V., Duvanskiy V. A., Belkov A. V. Possible autofl uorescent diagnostics of epithelial structures in the colon. Experimental and Clinical Gastroenterology. 2019;164(4): 21–26. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-21-26
    Read & Download full text

    1. Federal State Budgetary Educational Institution of Higher Education “National Research Ogarev Mordovia State University”, Saransk 430005, Republic of Mordovia, Russia

    Keywords: Remaxol, chronic periodontitis, gastropathy, lipid metabolism

    Abstract: Two groups of patients with chronic severe periodontitis and gastropathy: І (comparison group, n = 26) - under standard therapy; ІІ group (studied group, n = 22) - Remaxol is included in the standard treatment regimen. On admission and after 10 days of treatment, we studied the structural and functional state of periodontal tissues with the calculation of clinical indices, performed fibrogastroduodenoscopy with a biopsy of the gastric mucosa, followed by determination of lipid composition in its tissues and red blood cells, determined the content of H. Pylori, pepsinogen I and II, immunoglobulin A and E, malonic dialdehyde and diene conjugates, the activity of phosphalipase A2 and catalase. Chronic periodontitis was established to be accompanied by significant structural and functional lesions of the stomach (gastritis, erosion, ulcers). The role of membrane-destabilizing phenomena in epithelial cells of the gastric mucosa has been proved in the pathogenesis of gastropathy. Oxidative stress and phospholipase activity are involved in the development of these phenomena. The use of Remaxol in the treatment leads to a decrease in inflammatory necrotic processes in the mouth and stomach. The effectiveness of the drug is largely due to its ability to reduce membrane-destabilizing processes by decreasing the activity of lipoperoxidation and phospholipase A2, as well as reducing the acidity of gastric juice.

      1. Bulkina N. V., Lepilin A. V., Osadchuk M. A. The results of the treatment of chronic generalized periodontitis under chronic gastritis. Fundamental and applied research in medicine: Materials of scientific. conf. Greece, 2004. - № 9. - p. 112.
      2. Lepilin A. V., Bulkina N. V., Osadchuk M. A. Combined diseases of the oral cavity and digestive organs. Saratov: NSMU, 2005. - 125 p.
      3. Lepilin AV, Osadchuk MA, Karabushina Ya. G. Pathogenetic features of inflammatory periodontal diseases in irritable bowel syndrome. Successes of modern science. 2003, no.8, pp. 99. URL: http://natural-sciences.ru/ru/article/view?id=14852.
      4. Lukina G. I., Bazikyan E. A. Non-carious lesions of dental hard tissues in gastroenterological patients. Medical alphabet. 2010, no.4, pp.31-32.
      5. Lukina G. I., Bazikyan E. A. Possible microbial associations in the oral cavity in patients with diseases of the esophagogastroduodenal zone. DentalForum. 2010, no.4, pp. 33-35.
      6. Iordanishvili A. K., Belskikh O. A., Tishkov D. S., Karev F. A., Muzykin M. I., Libih D. A. Features of the functioning of the oral mucosa and tongue in chronic diseases of the kidneys, intestines and endocrine pathology. Kursk Scientific and Practical Journal “Man and his health”. 2015, no.4, pp. 31-40.
      7. Tytyuk S. Yu., Pykhur O. L., Tishkov D. S., Iordanishvili A. K. Morphological and functional features of the oral mucosa in persons suffering from chronic inflammatory bowel diseases. Kursk Scientific and Practical Journal “Man and his health”. 2016, no.3, pp. 49-55.
      8. Ganganna K., Shetty P., Shroff S. E. Collagen in histologic stages of oral submucous fibrosis: Apolarizing microscopic study. J. Oral Maxillofac. Pathol. 2012, Vol. 16, N2, P. 162-166.
      9. Logan R. M. Links between oral and gastrointestinal health. Curr. Opin. Support Palliat. Care. - 2010. - Vol. 4, N1 - P. 31-35.
      10. Orosz M., Sonkodi I. Oral manifestations in Crohn’s disease and dental management. Fogorv. Sz. -2004 - Vol. 97, N3 - P. 113-117.
     


    Full text is published :
    Vlasova T. I., Kondyurova E. V., Davydkin V. I., Vlasov A. P., Sheyranov N. S., Vasilyev V. V., Mitina E. A., Sinyavina K. M. Pathogenetic base for a new approach in the treatment of diseases of the digestive system upper departments. Experimental and Clinical Gastroenterology. 2019;164(4): 27–32. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-27-32
    Read & Download full text

    1. Practical and Clinical Research Center of Diagnostics and Telemedicine Technologies, Department of Healthcare of Moscow, 109029, 28–1, Srednyaya Kalitnikovskaya str., Moscow, Russia
    2. Federal State Autonomous Educational Institution of Higher Education “I. M. Sechenov First Moscow State Medical University” of the Ministry of Health of the Russian Federation (Sechenov University), 119991, 8–2, Trubetskaya str., Moscow, Russia
    3. Lomonosov Moscow State University, Faculty of Fundamental Medicine, 119192, 27/1, Lomonosovsky Ave., Moscow, Russia

    Keywords: osteoporosis, QCT, gastric cancer, pancreatic cancer, gastrectomy, pancreatoduodenal resection, malabsorption

    Abstract: Objective. Analyze the relationship between the surgical treatment of the upper gastrointestinal tract (GIT) and the decrease in bone mineral density (BMD) by comparison with reference age data and the control group. Methods. The medical data of 20 patients were analyzed with various types of surgical interventions on the upper GI tract (main group): 8 patients underwent various types of pancreatoduodenal resections, 11 - stomach resections, including two according to Billroth II, one patient underwent antral stenting. The patients’ age ranged from 53 to 84 years old, the median one being 70 years old, 13 men and 7 women. Measurements of lumbar spine BMD by the method of asynchronous CT densitometry were carried out using native images of the abdominal cavity, without contrast enhancement. The control group consisted of 35 patients who underwent an abdominal scan, without cancer pathology and without indications of a decrease in BMD. The age in the control group ranged from 43 to 86 years old, the median one being 65 years old, 22 men and 13 women. A multifactorial regression model was built to analyze the influencing factors on BMD. Results. A statistically significant dependence of the IPC on three factors was revealed: sex (p = 0.015), age (p = 0.001), and the presence of surgery. It was shown that among the main group of patients, the value of BMD was lower on average by 12.3 mg/ml as compared with the control group (p = 0.027), this corresponds to a decrease Zscore -0,35 SD. Conclusion. A statistically significant decrease BMD in patients after surgical treatment of the upper GIT, relative to the control group. It can be assumed that the emerging malabsorption syndrome is one of the prerequisites for the development of osteoporosis or osteopenia. Asynchronous CT densitometry is an effective method for monitoring bone mineral density, which provides additional information regarding BMD in patients under examination.

      1. Embutnieks Yu.V., Drozdov V. N., Chernysheva I. V., Topcheeva O. N., Koricheva E. S., Albulova E. A. Features of bone metabolism in patients with chronic diseases of digestive system. Experimental and clinical gastroenterology. 2011;(2):111-116. (In Russ.)
      2. Slohova N. K., Totrov I. N. Pathology of bone tissue in diseases of the gastrointestinal tract. Kuban Scientific Medical Bulletin. 2015;(3):97-100. (In Russ.) https://doi.org/10.25207/1608-6228-2015-3-97-100
      3. Glushkov N. I., Pertsev A. V., Belyaev E. M. Disturbances of bone tissue metabolism in elderly and senile patients after stomach resection. Grekov’s bulletin of surgery. 2009;168(1):31-32. (In Russ.).
      4. Volkiv V. E., Volkov S. V. Pathology of metabolic disturbanus in patients after total gastrectomy. Vestnik chuvashskogo universiteta. 2011;3:297-304. (In Russ.).
      5. Seo G.H., Kang H. Y., Choe E. K. Osteoporosis and fracture after gastrectomy for stomach cancer. Medicine (Baltimore). 2018;97(17): e0532. https://doi.org/10.1097/MD.0000000000010532
      6. Noh H.-M., Yoo J.-H., Jeong J. Y., Park Y. S. Bone mineral density after treatment for gastric cancer. Medicine (Baltimore). 2018;97(1): e9582. https://doi.org/10.1097/MD.0000000000009582
      7. Modi A., Sen S., Adachi J. D., Adami S., Cortet B., Cooper A. L., Geusens P., Mellström D., Weaver J. P., van den Bergh J. P., Keown P. A., Sajjan S. Association of gastrointestinal events with quality of life and treatment satisfaction in osteoporosis patients: results from the Medication Use Patterns, Treatment Satisfaction, and Inadequate Control of Osteoporosis Study (MUSIC OS). Osteoporos. Int. 2017; 28(10):2867-2876.
      8. Kuvaev R. O., Kashin S. V., Nikonov E. L., Itoh L., Gotoda T., Gono K. Early stomach cancer: the modern methods for screening, endoscopic diagnostics, and minimally invasive treatment. Evidence-based gastroenterology. 2014;3(3):44-51. (In Russ.).
      9. Therkildsen J. Winther S., Nissen L., Jørgensen H. S., Thygesen J., Ivarsen P., Frost L., Langdahl B. L., Hauge E. M., Böttcher M. Feasibility of Opportunistic Screening for Low Thoracic Bone Mineral Density in Patients Referred for Routine Cardiac CT. J. Clin. Densitom. 2018. https://doi.org/10.1016/j.jocd.2018.12.002
      10. Ziemlewicz T.J., Binkley N., Pickhardt P. J. Opportunistic Osteoporosis Screening: Addition of Quantitative CT Bone Mineral Density Evaluation to CT Colonography. J. Am. Coll. Radiol. 2015;12(10):1036-1041. https://doi.org/10.1016/j.jacr.2015.04.018
      11. Brett A.D., Brown J. K. Quantitative computed tomography and opportunistic bone density screening by dual use of computed tomography scans. J. Orthop. Transl. 2015;3(4):178-184. https://doi.org/10.1016/j.jot.2015.08.006
      12. Pickhardt P.J., Bodeen G., Brett A., Brown J. K., Binkley N. Comparison of femoral neck BMD evaluation obtained using lunar DXA and QCT with asynchronous calibration from CT colonography. J. Clin. Densitom. 2015;18(1):5-12. https://doi.org/10.1016/j.jocd.2014.03.002
      13. Engelke K., Lang T., Khosla S., Qin L., Zysset P., Leslie W. D., Shepherd J. A., Shousboe J. T. Clinical Use of Quantitative Computed Tomography-Based Advanced Techniques in the Management of Osteoporosis in Adults: The 2015 ISCD Official Positions-Part III. J. Clin. Densitom. 2015;18(3):393-407. https://doi.org/10.1016/j.jocd.2015.06.010
      14. Brown J.K., Timm W., Bodeen G., Chason A., Perry M., Vernacchia F., DeJournett R. Asynchronously Calibrated Quantitative Bone Densitometry. J. Clin. Densitom. 2017;20(2):216-225. https://doi.org/10.1016/j.jocd.2015.11.001
      15. Block J.E., Smith R., Glueer C. C.C., Steiger P., Ettinger B., Genant H. K. Models of spinal trabecular bone loss as determined by quantitative computed tomography. J. Bone Miner. Res. 1989;4(2):249-257. https://doi.org/10.1002/jbmr.5650040218
      16. Cann C.E., Genant H. K., Kolb F. O., Ettinger B. Quantitative computed tomography for prediction of vertebral fracture risk. Bone. 1985;6(1):1-7. https://doi.org/10.1016/8756-3282(85)90399-0
      17. Ministry of Health of the Russian Federation. Clinical recommendations under the supervision of Dedov I. I., Melnichenko G. A. Osteoporosis, 2016. (In Russ.)
      18. ACR-SPR-SSR practice parameter for the performance of quantitative computed tomography (QCT) bone densitometry. URL: https://www.acr.org/~/media/DE78D218C7A64526A821A9E8645AB46D.pdf. Res. 32-2013, Amended 2014 (Res. 39).
      19. Ministry of Health of the Russian Federation. Clinical recommendations under the supervision of Abdullaev A. G., Allahverdiev A. K., Besonova N. S., 2017. (In Russ.)
     


    Full text is published :
    Petraikin A. V., Soloviev A. V., Chesnokova A. O., Petryaykin F. A., Nisovtsova L. A., Sergunova K. A., Akhmad E. S., Semenov D. S., Vladzymyrskyy A. V., Morozov S. P. Asynchronous quantitative computed tomography (QCT) in assessing the impact of the upper gastrointestinal tract on bone mineral density. Experimental and Clinical Gastroenterology. 2019;164(4): 33–39. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-33-39
    Read & Download full text

    1. Orenburg State Medical University (OrSMU), 460000 Orenburg, Russia

    Keywords: gastro-esophageal junction, gastroesophageal refl ux disease, Helicobacter pylori, preneaplastic changes, Barrett’s esophagus

    Abstract: Objective: to improve the diagnosis of gastroesophageal reflux disease and its complications.
    Materials and methods: clinical and endoscopic studies were performed on 1052 patients, 110 of them healthy, 722 - with gastric ulcer and duodenal ulcer, after Billroth II stomach resection, with a hiatus hernia, gallstone disease, etc., 210 children. Studied 100 macropreparations of the esophageal-gastric junction and 70 longitudinal histotopograms from patients who died from diseases not related to diseases of the gastrointestinal tract, histotopograms of 10 cases with esophageal adenocarcinoma and esophageal-gastric junction.
    Results: The location of the Z-line is individually variable, the range is limited to two levels: up to 40 mm above and 18 mm below the outlet of the cardia. The presence of a mucous membrane with a cylindrical epithelium in the distal esophagus is a variant of the norm. In the area of the Z-line in 70% of cases there is a duplication at a distance of 10 mm, consisting of a mucous membrane with a stratified squamous epithelium, covering the mucous membrane with the gastric epithelium. High Helicobacter infection of the mucous membrane with gastric epithelium was detected, its pre-neoplastic changes in adults (17.2%), in children (10.9%) and Barrett’s esophagus are often found in mild degrees of esophagitis, and there are also indirect signs of HP in the esophagus.
    Conclusions: The presence of a mucous membrane with a cylindrical epithelium in the esophagus up to 40 mm above the outlet of the cardia is a variant of the norm. In the area of the Z - line there is in 70% of cases duplication at a distance of 10 cm, consisting of a mucous membrane with a stratified squamous epithelium, under which there is a mucous membrane with a gastric epithelium, and a step biopsy is required to diagnose changes in it. The presence of HP in the mucous membrane of gastro-esophageal junction has indirect endoscopic signs, contributes to the development of pre-neoplastic changes of the mucous membrane, which in most cases are found in mild degrees of esophagitis, which requires antihelicobacter therapy.

      1. Kalinin A. V. Gastroesofageal’naya refluksnaya bolezn’: metodicheskie ukazaniya [Gastro-esophageal reflux disease: methodical guideline]. M.: GIUV MORPH, 2004, p 5, (40p).
      2. Starostin B. D. Neerosivnaya refluksnaya bolezn’ [Non-erosive reflux disease]. Bolezni organov pishevareniya - Diseases of the digestive system. 2004, vol. 6, no 2, pp. 79-83.
      3. Lapina T. L. Ezomeprazol - pervyj ingibitor protonnoj pompy parietal’nykh kletok zheludka, sozdannyj kak monoizomer: novye dostizheniya v terapii gastroehzofageal’noj reflyuksnoj bolezni [Esomeprazole - the first proton pump inhibitor of the parietal cells of the stomach, created as a monoisomer: new advances in the treatment of gastroesophageal reflux disease]. Rossijskij zhurnal gastroehnterologii, gepatologii, koloproktologii - Russian Journal of gastroenterology, hepatology, coloproctology. 2002, vol. 12, no. 1, pp. 23-29.
      4. Kaibysheva V. O., Kucheryavyi Yu.A., Trukhmanov A. S. et al. Rezul’taty mnogotsentrovogo nablyudatel’nogo issledovaniya po primeneniyu mezhdunarodnogo oprosnika Gerd Q dlya diagnostiki gastroehzofageal’noj reflyuksnoj bolezni [The results of a multicenter observational study on the use of the international questionnaire Gerd Q for the diagnosis of gastroesophageal reflux disease]. Rossijskij zhurnal gastroehnterologii, gepatologii, koloproktologii - Russian Journal of gastroenterology, hepatology, coloproctology. 2013, vol. 23, no. 5, pp. 1-9
      5. Korolev M. P. Pishchevod Barretta (klinika, diagnostika, lecheniye) [Barrett’s esophagus (clinic, diagnosis, treatment)]. Al’manakh endoskopii - Endoscopy Almanac. 2002; no. 1, pp. 61-72.
      6. Kubyshkin V. A., Kornyak V. S. Gastroezofageal’naya reflyuksnaya bolezn’. Diagnostika, konservativnoye i operativnoye lecheniye [Gastroesophageal reflux disease. Diagnosis, conservative and surgical treatment]. Moscow, SPROS Publ., 1999, p 14, (188p).
      7. Bu X., Ma Y., Der R., Demeester T. et al. Body mass index is associated with Barrett oesophagus and cardiac mucosal metaplasia. Dig. Dis Sci. 2006. Sep; 51(9):1589-94.
      8. Casson A.Z., Williams L., Guernsey D. L. Epidemiology and molecular biology of Barrett esophagus. Semin Thorac Cardiovasc Surg. 2005, Winter; 17 (4): 284 91.
      9. Maev I. V. Ispol’zovaniye ingibitorov protonnoy pompy v lechenii gastroezofageal’noy reflyuksnoy bolezni [The use of proton pump inhibitors in the treatment of gastroesophageal reflux disease]. Klinicheskaya meditsina - Clinical medicine. 2003, no. 9, pp. 54-58.
      10. Shumeiko N. K., Kirillov V. I., Pogosova I. E. et al. Faringolaringeal’nyy reflyuks - simptom gastroezofageal’noy reflyuksnoy bolezni ili otdel’naya nozologiya [Pharyngolaryngeal reflux is a symptom of gastroesophageal reflux disease or a separate nosology]. Rossijskij zhurnal gastroehnterologii, gepatologii, koloproktologii - Russian Journal of gastroenterology, hepatology, coloproctology. 2007, vol. 17, no. 5, p. 119.
     


    Full text is published :
    Dronova O. B., Kagan I. I., Tretyakov A. A., Mironchev A. O. Discussion issues of gastroesophageal refl ux disease based on clinical and endoscopic studies. Experimental and Clinical Gastroenterology. 2019;164(4): 40–45. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-40-45
    Read & Download full text

    1. Mоscow Research and Clinical Centre for TB Control, Moscow Healthcare Department, Moscow, Russia
    2. Pirogov Russian National Research Medical University, Moscow, Russia

    Keywords: abdominal tuberculosis, intestinal tuberculosis, HIV infection, tuberculosis peritonitis, bowel obstruction

    Abstract: Aim. The aim of the research was to perform an analysis of surgical complications of abdominal tuberculosis and to determine optimal surgical treatment methods.
    Materials and methods. A retrospective analysis of the treatment of 166 patients with surgical complications of abdominal tuberculosis was conducted: 60 HIV-negative patients of group I and 106 HIV-positive patients of group II. The patients underwent a comprehensive examination, which included diagnostic radiology, endoscopic methods, and the study of biological media for the detection of Mycobacterium tuberculosis. All patients underwent various types of urgent surgery.
    Results. Perforations of tuberculosis ulcers of the intestine were diagnosed in 71 (42.8%) patients, tuberculosis peritonitis in 50 (30.1%), acute intestinal obstruction in 26 (15.7%), intestinal bleeding in 8 (4.8%), tuberculosis spleen abscesses - in 11 (6.6%). Analyzing our experience in treating patients with abdominal tuberculosis, it can be said that surgical complications are twice as likely to occur in HIV-positive patients, but the spectrum of the incidence of surgical complications does not differ.
    Conclusions. The most frequent surgical complications of abdominal tuberculosis in both patients with HIV-negative and in patients with HIV-positive status are perforations of intestinal tuberculosis ulcers, tuberculosis peritonitis and acute intestinal obstruction, which lead to the development of severe surgical complications, causing the complexity of diagnosis and treatment and high mortality. The tuberculosis lesions of various organs and systems were detected in HIV-positive patients 2.3 times more often; tuberculosis is of a generalized nature, associated with hematogenous dissemination and is accompanied by severe surgical complications.

      1. Peng-Hui Wang. Tuberculous peritonitis should always be considered as a differential diagnosis in abdominal carcinomatosis. Taiwanese Journal of Obstetrics and Gynecology. 2004;43(3):182-184. https://doi.org/10.1016/S1028-4559(09)60083-8
      2. Pattanayak S., BehuriaIs S. Аbdominal tuberculosis a surgical problem? Ann. R. Coll. Surg. Engl. 2015 Sep 1;97(6):414-419 https://doi.org/10.1308/rcsann.2015.0010
      3. Poddubnaya L. V., Zyryanova T. V., Petrenko T. I., et al. Abdominal tuberculosis in pulmonary tuberculosis patients. Experimental and Clinical Gastroenterology. 2018;153(5):38-43. (In Russ.)
      4. Bogorodskaya E. M., Sinitsyn M. V., Belilovsky E. M., Borisov S. E., Kotova E. A. Impact of HIV Infection on the structure of new tuberculosis cases detected in the city of Moscow. Tuberculosis and Lung Diseases. 2017;95(10):17-26. (In Russ.) https://doi.org/10.21292/2075-1230-2017-95-10-17-26
      5. Kawazoe A., Nagata N. Intestinal tuberculosis in an HIV-infected patient with advanced immunosuppression. Clin Gastroenterol Hepatol. 2012;10(9): A24-26. https://doi.org/10.1016/j.cgh.2012.03.013
      6. Frolova K. S., Borisov S. E. Risk of developing active TB in IBD patients treated with atni-TNF. Koloproctologia. 2018;№ 1(63):49-56. (In Russ.)
      7. UNAUDS/Fact sheet 2017. Global HIV statistics. http://www.unaids.org/sites/default/files/media_asset/UNAIDS_FactSheet_en.pdf
      8. Vasilyeva I. A., Belilovskiy E. M., Borisov S. E., Sterlikov S. A., Sinitsyn M. V. Tuberculosis combined with HIV infection in the countries of the world and the Russian Federation. Tuberculosis and Lung Diseases. 2017;95(9):8-18 (In Russ.) https://doi.org/10.21292/2075-1230-2017-95-9-8-18
      9. Getahun H., Gunneberg C., Granich R., Nunn P. HIV infection associated tuberculosis: the epidemiology and the response. Clinical Infectious Diseases, Volume 50, Issue Supplement_3, 15 May 2010, Pages S201-S207. https://doi.org/10.1086/651492
      10. Sinitsyn M. V., Belilovsky E. M., Sokolinа I. А., et.al. Extrapulmonary tuberculosis in HIV patients. Tuberculosis and Lung Diseases. 2017;95(11):19-25. (In Russ.) https://doi.org/10.21292/2075-1230-2017-95-11-19-25
      11. Debi U., Ravisankar V., Prasad K. K., Sinha S. K., Sharma A. K. Abdominal tuberculosis of the gastrointestinal tract: Revisited. World J Gastroenterol. 2014;20(40):14831-14840. http://dx.doi.org/10.3748/wjg.v20.i40.14831
      12. Plotkin D. V., Sinitsyn M. V., Reshetnikov M. N., Kharitonov S. V., Skopin M. S., Sokolina I. A. Tuberculous peritonitis. «Forgotten» disease. Khirurgiya. Zhurnal im. N. I. Pirogova. 2018;12:38-44. (In Russ.) https://doi.org/10.17116/hirurgia20181213
      13. Savonenkova L. N., Arjamkina O. L. Clinical, pathogenetic aspects of tuberculosis of digestive organs. Tuberculosis and Lung Diseases. 2005;82(6):42-45. (In Russ.)
      14. Reshetnikov M. N., Skopin M. S., Sinitsyn M. V., Plotkin D. V., Zubаn O. N. The choice of surgical tactics in perforated tuberculous intestinal ulcers in HIV patients. Tuberculosis and lung diseases. 2017;95(9):19-24. (In Russ.) https://doi.org/10.21292/2075-1230-2017-95-9-19-24
      15. Reshetnikov M.N, Plotkin D. V., Skopin M. S., Sinitsyn M. V., Rodoman G. V. Acute intestinal obstruction in tuberculosis multiple localizations. Khirurg. 2017;9-12:13-22. (In Russ.)
      16. Reshetnikov M. N., Plotkin D. V., Sinitsyn M. V., Аbu Аrqoub T. I. Abdominal tuberculosis: emergency surgery in acute intestinal obstruction. Tuberculosis and Lung Diseases. 2019;97(1):64-65. (In Russ.) https://doi.org/10.21292/2075-1230-2019-97-1-64-65
      17. Lensky E. V. Abdominal tuberculosis: difficulties in diagnosis. Siberian medical journal. 2006;59(1):5-10. (In Russ.)
      18. Lado F.L., Barrio Gomez E., Carballo Arceo E., Cabarcos Ortizide Barron A. Clinical presentation of tuberculosis and the degree of immunodeficiency in patients with HIV infection. Scandinavian Journal of Infectious Diseases. 1999;31:387-391.
      19. Savonenkova L. N., Anisimova S. V., Sidorova Yu.D., Sidorov I. A., Chunina A. F. Mortality of tuberculosis patients during HIV-infection epidemic. Ulyanovsk medico-biological journal. 2018;3:99-106. (In Russ.) Doi: https://doi.org/10.23648/UMBJ.2018.31.17220.
     


    Full text is published :
    Reshetnikov M. N., Plotkin D. V., Sinitsyn M. V., Gafarov U. O., Belentseva O. V. Surgical complications of abdominal tuberculosis in patients with diff erent immune status. Experimental and Clinical Gastroenterology. 2019;164(4): 46–53. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-46-53
    Read & Download full text

    1. State Budgetary Institution of Health Care “Region Clinic Hospital Nr 2” Health Ministry of Krasnodar Region, Krasnodar, Russia
    2. Federal State Budgetary Educational Institution of Higher Education “Kuban State Medical University” Health Ministry of Russian Federation, Krasnodar, Russia

    Keywords: cicatricial strictures, common bile duct, bougienage, choledochoscopy

    Abstract: The aim was to optimize the method of endoscopic bougienage in common bile duct strictures using the oral choledochoscope.
    Materials: the analysis of 177 patients with bile duct strictures of the main group and 38 patients of the comparison group. The nature and history of bile duct strictures, as well as the possibilities and effectiveness of application the oral choledochoscope in the narrowing of strictures in difficult clinical situations have been studied.
    Results: the method proposed by us was applied in 5 patients, in complex clinical cases when, under X-ray control, it was not possible to hold the string conductor outside the stricture zone. Comparing the proportion of effective and ineffective minimally invasive interventions, a higher rate was observed in the main group by 12.1% (91.0%) than in the comparison group (78.9%) with a statistically significant difference in the proportions (p = 0.0329, t. e. p <0.05).
    Conclusion: the introduction of the bougienage method with the choledochoscope allowed a statistically significant reduction in the failure rate of minimally invasive treatment of common bile duct strictures in the main group of observables.

      1. Pautkin Yu. F., Klimov A. E. Mechanical obstruction of the biliary tract. A guide for doctors. Moscow. Profile Pub., 2010, 224 p.
      2. Dyuzheva N. A. Endoscopic balloon dilatation of bile and pancreatic ducts. Moscow, 2014, 28 p.
      3. Vetshev P. S. Percutaneous minimally invasive technologies: history, realities and prospects. Med.vestn. South of Russia. 2014, No. 4, pp. 12-15.
      4. Costamagna, G. Longterm results of endoscopic management of postoperative bile duct strictures with increasing numbers of stents. G. Costamagna, M. Pandolfi, M. Mutignani, et al. Gastrointestinal endoscopy. 2001. Vol. 54, № 2, pp. 162-168.
      5. Lillemoe, K. D. Postoperative bile duct strictures: management and outcome in the 1990s / K. D. Lillemoe, G. B. Melton, J. I. Cameron, et al. Annals of surgery. - 2000. Vol. 232, no. 3, pp. 345-349.
      6. Halperin, E. I. Treatment of damage to extrahepatic bile ducts obtained with laparoscopic cholecystectomy. Surgery. 2001, no.1, p. 51.
      7. Balalykin A. S., Onopriev A. V., Mutsurov H. S., et al. Endoskopicheskaya diagnostika i lecheniye opukholey bol'shogo duodenal'nogo osochka [Endoscopic diagnosis and treatment of tumors of the large duodenal sediment]. [Collected materials 4 All-Russian Conf. "Endoscopy in the diagnosis of diseases of the pancreatic-biliary zone and intestines."]. SPb., 2013. pp. 22-24.
      8. Shapovalyants S. G., Ardasenov T. B., Pankov A. G., et al. Complex choledocholithiasis is the result of delayed surgical treatment of gallstone disease. RZHGGK. 2013, vol. 23, no. 4, pp. 15-21.
     


    Full text is published :
    Gabriel S. A., Durleshter V. M., Guchetl A. Ya., Dynko V. Yu., Bespechnyj M. V. The use of an oral choledochoscope for cicatricial strictures of the common bile duct. Experimental and Clinical Gastroenterology. 2019;164(4): 54–58. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-54-58
    Read & Download full text

    1. RUDN University, Moscow, Russia
    2. Skobelkin state scientifi c center of laser medicine, Moscow, Russia
    3. Herzen research oncological Institute, Moscow, Russia
    4. Eramishantsev Clinical Hospital, Moscow, Russia

    Keywords: malignant colon obstruction, colorectal stenting, bridge to surgery, IPMN, MCN, SCN, EUS

    Abstract: Malignant colonic obstruction is actual problem of emergency surgery. In various clinical situations it is possible to perform different interventions. Colorectal stenting is the favorite method of palliative treatment. However, the role of stenting in patients with curable or potentially curable tumors remains unclear. This literature review presents data from meta-analysis, randomized and cohort studies, as well as recommendations to determine the role of colorectal stenting as bridge to surgery

      1. Donlon N E, Kelly M E, Narouz F et al. Colonic stenting as a bridge to surgery in malignant large bowel obstruction: oncological outcomes. Int J Colorectal Dis. 2019 Jan 16. doi: 10.1007/s00384-019-03239-9.
      2. Jeong D S, Kim Y H, Kim K J et al. Surgical outcomes and risk factors in patients who underwent emergency colorectal surgery. Ann Coloproctol 33(6):239-244.
      3. Tejero E, Mainar A, Fernández L et al. New procedure for the treatment of colorectal neoplastic obstructions. Dis Colon Rectum 37(11):1158-1159.
      4. Hill J, Kay C, Morton D et al. CREST: randomised phase III study of stenting as a bridge to surgery in obstructing colorectal cancer - results of the UK colorectal endoscopic stenting trial (CREST). J Clin Oncol 2016, 34(15_suppl): 3507-3507
      5. Van Hooft J E, Bemelman W A, Oldenburg B et al. Colonic stenting versus emergency surgery for acute left-sided malignant colonic obstruction: a multicentre randomised trial. Lancet Oncol 2011, 12(4):344-352
      6. Zhang Y, Shi J, Shi B et al. Self-expanding metallic stent as a bridge to surgery versus emergency surgery for obstructive colorectal cancer: a meta-analysis. Surg Endosc. 2012 Jan;26(1):110-9. doi: 10.1007/s00464-011-1835-6
      7. Tan C J, Dasari B V, Gardiner K. Systematic review and meta-analysis of randomized clinical trials of self-expanding metallic stents as a bridge to surgery versus emergency surgery for malignant left-sided large bowel obstruction. Br J Surg. 2012 Apr;99(4):469-76. doi: 10.1002/bjs.8689.
      8. Pirlet I A, Slim K, Kwiatkowski F et al. Emergency preoperative stenting versus surgery for acute left-sided malignant colonic obstruction: a multicenter randomized controlled trial. Surg Endosc 2011; 25: 1814-1821.
      9. Cheung H Y, Chung C C, Tsang W W, et al. Endolaparoscopic approach versus conventional open surgery in the treatment of obstructing left-sided colon cancer: a randomized controlled trial. Arch Surg. 2009; 144: 1127-1132.
      10. Ye G Y, Cui Z, Chen L, Zhong M. Colonic stenting vs emergent surgery for acute left-sided malignant colonic obstruction: a systematic review and meta-analysis. World J Gastroenterol. 2012 Oct 21;18 (39):5608-15. doi: 10.3748/wjg.v18.i39.5608.
      11. Cirocchi R, Farinella E, Trastulli S et al. Safety and efficacy of endoscopic colonic stenting as a bridge to surgery in the management of intestinal obstruction due to left colon and rectal cancer: a systematic review and meta-analysis. Surg Oncol. 2013 Mar;22(1):14-21. doi: 10.1016/j.suronc.2012.10.003.
      12. De Ceglie A, Filiberti R, Baron T H et al. A meta-analysis of endoscopic stenting as bridge to surgery versus emergency surgery for left-sided colorectal cancer obstruction. Critical Reviews in Oncology/Hematology. 2013; 88(2): 387-403.
      13. Zhao R S, Wang H, Wang L et al. Meta-analysis of safety and efficacy of self-expending metallic stents as bridge to surgery versus emergency surgery for left-sided malignant colorectal obstruction. Zhonghua Wei Chang Wai Ke Za Zhi. 2012 Jul;15(7):697-701.
      14. Allievi N, Ceresoli M, Fugazzola P et al. Endoscopic Stenting as Bridge to Surgery versus emergency resection for left-sided malignant colorectal obstruction: an updated meta-analysis. International Journal of Surgical Oncology Volume 2017, Article ID2863272, 11 pages https://doi.org/10.1155/2017/2863272
      15. Matsuda A, Miyashita M, Matsumoto S et al. Comparison of Long-Term Outcomes of Colonic Stent as ‘‘Bridge to Surgery’’ and Emergency Surgery for Malignant Large-Bowel Obstruction: A Meta-Analysis Ann Surg Oncol. 2015;22:497-504 DOI 10.1245/s10434-014-3997-7
      16. Wang X, He J, Chen X, Yang Q. Stenting as a bridge to resection versus emergency surgery for left-sided colorectal cancer with malignant obstruction: A meta-analysis. International Journal of Surgery. 2017. doi: 10.1016/j.ijsu.2017.10.004.
      17. Huang X, Lv B, Zhang S, Meng L. Preoperative colonic stents versus emergency surgery for acute left-sided malignant colonic obstruction: a meta-analysis. J Gastrointest Surg. 2014 Mar;18(3):584-91. doi: 10.1007/s11605-013-2344-9.
      18. Foo СС, Poon S H, Chiu R H. Is bridge to surgery stenting a safe alternative to emergency surgery in malignant colonic obstruction: a meta-analysis of randomized control trials. Surg Endosc. 2019 Jan;33(1):293-302. doi: 10.1007/s00464-018-6487-3
      19. Yang P, Lin X F, Lin K, Li W. The Role of Stents as Bridge to Surgery for Acute Left-Sided Obstructive Colorectal Cancer: Meta-Analysis of Randomized Controlled Trials. Rev Invest Clin. 2018;70(6):269-278. doi: 10.24875/RIC.18002516.
      20. Malakorn S, Stein S L, Lee J H, You Y N. Urgent management of obstructing colorectal cancer: divert, stent, or resect? J Gastrointest Surg. 2018 Oct 3. doi: 10.1007/s11605-018-3990-8
      21. Hill J, Gray R, Morton D et al. Systematic review and meta-analysis of randomized clinical trials of self-expanding metallic stents as a bridge to surgery versus emergency surgery for malignant left-sided large bowel obstruction Br J Surg. 2012 Oct;99(10):1462; author reply 1462-3. doi: 10.1002/bjs.8925.
      22. Bridoux V, Schwarz L, Kianifard B et al. Systematic review and meta-analysis of randomized clinical trials of self-expanding metallic stents as a bridge to surgery versus emergency surgery for malignant left-sided large bowel obstruction Br J Surg. 2012 Oct;99(10):1464; author reply 1464-5. doi: 10.1002/bjs.8929.
      23. van Hooft J E, van Halsema E E, Vanbiervlie G et al. Self-expandable metal stents for obstructing colonic and extracolonic cancer: European Society of Gastrointestinal Endoscopy (ESGE) Clinical Guideline Endoscopy. 2014; 46: 990-1002
      24. Tung KL, Cheung HY, Ng LW et al. Endo-laparoscopic approach versus conventional open surgery in the treatment of obstructing left-sided colon cancer: long-term follow-up of a randomized trial. Asian J Endosc Surg. 2013; 6: 78-81
      25. Alcantara M, Serra-Aracil X, Falco J et al. Prospective, controlled, randomized study of intraoperative colonic lavage versus stent placement in obstructive left-sided colonic cancer. World J Surg. 2011; 35: 1904-1910
      26. Sloothaak D, van den Berg M, Dijkgraaf M et al. Recurrences after endoscopic stenting as treatment for acute malignant colonic obstruction in the Dutch Stent-In 2 trial. Conference: 21st UEG Week, October 12-16 Berlin: 2013
      27. Gorissen KJ, Tuynman JB, Fryer E et al. Local recurrence after stenting for obstructing left-sided colonic cancer. Br J Surg 2013; 100: 1805-1809
      28. Sabbagh C, Browet F, Diouf M et al. Is stenting as “a bridge to surgery” an oncologically safe strategy for the management of acute, left-sided, malignant, colonic obstruction? A comparative study with a propensity score analysis Ann Surg. 2013; 258: 107-115
      29. Kim S J, Kim H W, Park S B et al. Colonic perforation either during or after stent insertion as a bridge to surgery for malignant colorectal obstruction increases the risk of peritoneal seeding. Surg Endosc.2015 Dec; 29(12):3499-506. doi:10.1007/s00464-015-4100-6
      30. Cao Y, Deng S, Wu K et al. Oncological consequence of emergent resection of perforated colon cancer with complete obstruction after stent insertion as a bridge to surgery. Int J Colorectal Dis. 2018 Aug 8. doi: 10.1007/s00384-018-3137-0
      31. Maruthachalam K., Lash G. E., Shenton B. K., Horgan A. F. Tumour cell dissemination following endoscopic stent insertion. Br J Surg. 2007 Sep;94(9):1151-4.
      32. Thorlacius H. Tumour cell dissemination following endoscopic stent insertion. Comment on. British Journal of Surgery. 2008; 95: 125-129
      33. Takahashi G, Yamada T, Iwai T et al. Oncological Assessment of Stent Placement for Obstructive Colorectal Cancer from Circulating Cell-Free DNA and Circulating Tumor DNA Dynamics Ann Surg Oncol. 2018 Mar;25(3):737-744. doi: 10.1245/s10434-017-6300-x.
      34. Malgras, B., Brullé, L., Lo Dico, R et al. (2015). Insertion of a stent in obstructive colon cancer can induce a metastatic process in an experimental murine model. Annals of Surgical Oncology, 22(S3), 1475-1480.
      35. Broholm M, Kobborg M, Frostberg E et al. Delay of surgery after stent placement for resectable malignant colorectal obstruction is associated with higher risk of recurrence. Int J Colorectal Dis. 2017 Apr; 32(4):513-516. doi: 10.1007/s00384-016-2705-4.
      36. Kim H J, Choi G S, Park J S et al. Higher rate of perineural invasion in stent-laparoscopic approach in comparison to emergent open resection for obstructing left-sided colon cancer. Int J Colorectal Dis. 2013 Mar;28(3):407-14. doi: 10.1007/s00384-012-1556-x
      37. Matsuda A, Miyashita M, Matsumoto S. et al. Colonic stent-induced mechanical compression may suppress cancer cell proliferation in malignant large bowel obstruction Surg Endosc. 2018 Aug 31. doi: 10.1007/s00464-018-6411-x.
      38. Arezzo A, Balague C, Targarona E et al. Colonic stenting as a bridge to surgery versus emergency surgery for malignant colonic obstruction: results of a multicentre randomized controlled trial (ESCO trial). Surg Endosc. 2017 Aug;31(8):3297-3305. doi: 10.1007/s00464-016-5362-3.
      39. Kang S I, Oh H K, Yoo J S et al. Oncologic outcomes of preoperative stent insertion first versus immediate surgery for obstructing left-sided colorectal cancer. Surg Oncol. 2018 Jun; 27(2):216-224. doi: 10.1016/j.suronc.2018.04.002
      40. Yan F H, Lou Z, Liu X S. Long-term oncological outcomes of endoscopic stenting as a bridge to surgery versus emergency surgery for malignant colorectal obstruction: a comparativestudy. J Laparoendosc Adv Surg Tech A. 2017 Jun;27(6):611-617. doi: 10.1089/lap.2016.0529.
      41. Lim T-Z, Chan D K H, Tan K-K. Endoscopic stenting does not worsen long term outcomes amongst patients presenting with obstruction from colorectal cancers Ann Surg Oncol. 2017 Jun;24(6):1618-1625. doi: 10.1245/s10434-016-5724-z.
      42. Kwak M S, Kim W S, Lee J M. Does stenting as a bridge to surgery in left-sided colorectal cancer obstruction really worsen oncological outcomes? Dis Colon Rectum. 2016 Aug;59(8):725-32. doi: 0.1097/DCR.0000000000000631.
      43. Pisano M, Zorcolo L, Merli C. 2017 WSES guidelines on colon and rectal cancer emergencies: obstruction and perforation. World J Emerg Surg. 2018 Aug 13;13:36. doi: 10.1186/s13017-018-0192-3.
     


    Full text is published :
    Vodoleev A. S., Duvanskiy V. A., Yarotskov I. I. Colorectal stenting as a bridge to surgery for malignant colonic obstruction: evidence-based medicine. Experimental and Clinical Gastroenterology. 2019;164(4): 59–65. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-59-65
    Read & Download full text

    1. City clinical hospital № 24, Moscow, Russia
    2. Moscow Regional Research and Clinical Institute (“MONIKI”), Moscow, Russia
    3. Pirogov Russian National Research Medical University, Moscow, Russia

    Keywords: low rectal cancer, magnetic resonance imaging, metastases, lymph nodes, extramural vascular invasion

    Abstract: Rectal cancer occupies one of the leading places in the structure of mortality in both sexes, with the only radical method of treatment is surgery. For the planning of surgical treatment, it is important to assess the metastatic lesion of the locoregional lymph nodes, the presence of extramural lymphovascular invasion, which is an important predictor of recurrence of the disease, as well as an indicator of the early appearance of distant metastases. The effectiveness and reliability of preoperative staging of the low rectal cancer has increased significantly after the introduction into clinical practice of magnetic resonance imaging, as well as a comprehensive preoperative radiation examination of patients. The article is devoted to the problems of preoperative staging and determination of statistically significant relationships between the presence of extramural vascular invasion in cancer of the low rectal cancer and the presence of distant metastases, the identification of the correlation between T and N - stages of the tumor, their morphological types with diagnosed extramural vascular invasion.

      1. Kaprin A. D., Starinsky V. V., Petrova G. V. The status of cancer care for the population of Russia in 2015. Moscow, 2016.
      2. Piippo U., Pääkkö E., Mäkinen M., Mäkelä J. Local staging of rectal cancer using the black lumen magnetic resonance imaging technique. Scand J Surg. 2008;97:237-42. [PubMed]
      3. Zhang X.M., Zhang H. L., Yu D., Dai Y., Bi D., Prince M. R., et al. 3-T MRI of rectal carcinoma: preoperative diagnosis, staging, and planning of sphincter-sparing surgery. AJR Am J Roentgenol. 2008;190:1271-8. doi: 10.2214/AJR.07.2505. [PubMed]
      4. Slater A., Halligan S., Taylor S. A., Marshall M. Distance between the rectal wall and mesorectal fascia measured by MRI: Effect of rectal distention and implications for preoperative prediction of a tumour-free circumferential resection margin. ClinRadiol. 2006:65-70. [PubMed]
      5. Stollfuss J.C., Becker K., Sendler A., Seidl S., Settles M., Auer F., et al. Rectal carcinoma: high-spatial-resolution MRI imaging and T2 quantification in rectal cancer specimens. Radiology. 2006;241:132-41. doi: 10.1148/radiol.2411050942. [PubMed]
      6. Taylor F.G., et al. Preoperative magnetic resonance imaging assessment of circumferential resection margin predicts disease-free survival and local recurrence: 5-year follow-up results of the MERCURY study. J ClinOncol. 2014;32(1):34-43. doi:10.1200/JCO.2012.45.3258. [PubMed]
      7. Taylor F.G., Quirke P., Heald R. J., Moran B., Blomqvist L., Swift I., et al. One millimetre is the safe cut-off for magnetic resonance imaging prediction of surgical margin status in rectal cancer. Br J Surg. 2011;98:872-9. doi: 10.1002/bjs.7458. [PubMed]
      8. Taylor F.G., Swift R. I., Blomqvist L., Brown G. A systematic approach to the interpretation of preoperative staging MRI for rectal cancer. AJR Am J Roentgenol. 2008;191:1827-1835. [PubMed]
      9. Brown G, Radcliffe AG, Newcombe RG, Dallimore NS, Bourne MW, Williams GT (2003) Preoperative assessment of prognostic factors in rectal cancer using high-resolution magnetic resonance imaging. Br J Surg 90: 355-364. [PubMed]
      10. Chand M, Swift RI, Chau I, Heald RJ, Tekkis PP, Brown G (2014, а) Adjuvant therapy decisions based on magnetic resonance imaging of extramural venous invasion and other prognostic factors in colorectal cancer. Ann R Coll Surg Engl 96: 543-546. [PMC free article] [PubMed]
      11. Chand M, Swift RI, Tekkis PP, Chau I, Brown G (2014. b) Extramural venous invasion is a potential imaging predictive biomarker of neoadjuvant treatment in rectal cancer. Br J Cancer 110: 19-25. [PMC free article] [PubMed]
      12. Nougaret S, Reinhold C, Mikhael HW, Rouanet P, Bibeau F, Brown G (2013) The use of MR imaging in treatment planning for patients with rectal carcinoma: have you checked the ‘DISTANCE. Radiology 268: 330-344. [PubMed]
      13. Taylor FG, Quirke P, Heald RJ, Moran BJ, Blomqvist L, Swift IR, Sebag-Montefiore D, Tekkis P, Brown G Magnetic Resonance Imaging in Rectal Cancer European Equivalence Study, G (2014. a) Preoperative magnetic resonance imaging assessment of circumferential resection margin predicts disease-free survival and local recurrence: 5-year follow-up results of the MERCURY study. J Clin Oncol 32: 34-43. [PubMed]
      14. Taylor FGM, Quirke P, Heald RJ, Moran BJ, Blomqvist L, Swift IR, Sebag-Montefiore D, Tekkis P, Brown G (2014. b) Preoperative magnetic resonance imaging assessment of circumferential resection margin predicts disease-free survival and local recurrence: 5-year follow-up results of the MERCURY Study. J Clin Oncol 32: 34-U114. [PubMed]
      15. Chand M, Evans J, Swift RI, Tekkis PP, West NP, Stamp G, Heald RJ, Brown G (2015) The prognostic significance of postchemoradiotherapy high-resolution MRI and histopathology detected extramural venous invasion in rectal cancer. Ann Surg 261: 473-479. [PubMed]
      16. MERCURY Study Group. Extramural depth of tumor invasion at thin-section MR in patients with rectal cancer: results of the MERCURY Study. Radiology 2007; 243:132-139 [Crossref], [Medline].
      17. Harrison JC, Dean PJ, el-Zeky F, Vander Zwaaq R. From Dukes through Jass: pathological prognostic indicators in rectal cancer. Hum Patol 1994; 25: 498-505 [Crossref], [Medline].
      18. Smith NJ, Barbachano Y, Norman AR, Swift RI, Abulafi AM, Brown G. Prognostic significance of magnetic resonance imaging-detected extramural vascular invasion in rectal cancer. Surgery 2008; 95:229-236. [Crossref], [Medline].
     


    Full text is published :
    Volkova S. N., Stashuk G. A., Chermensky G. V., Naumov E. K. The role of MRI in identifying extramural vascular invasion as an indicator of the presence of regional and distant metastases of low rectal cancer. Experimental and Clinical Gastroenterology. 2019;164(4): 66–71. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-66-71
    Read & Download full text

    1. Federal state budget educational institution of higher education «Chuvash State University n. a. I. N. Ulyanov», department of Faculty and Hospital Therapy, 428015 Cheboksary, Russia
    2. EI of HE «The Surgut State University», Surgut, Russia

    Keywords: fatty liver diseases, nonalcoholic fatty liver disease, alcoholic liver disease, diagnosis of steatosis, diagnosis of steatohepatitis

    Abstract: The problem of high prevalence of fatty liver diseases is known today. The question of the diagnostic differentiation of non-alcoholic and alcoholic fatty liver diseases (NAFLD and ALD) has been studied in many studies, possible solutions have been proposed, but, to date, none of them are used in wide practice due to the presence of significant deficiencies in each method. At the same time, timely diagnosed diagnosis directly affects the quality and, often, the life expectancy of the patient. The review presents all modern methods of laboratory diagnostics that can identify differences between NAFLD and ALD, with an indication of the main known characteristics of each method.

      1. Babenko A. Yu., Laevskaya M. Yu. Non-alcoholic fatty liver disease - interrelations with metabolic syndrome. Russian Medical Journal. 2018, no. 1(I), pp. 34-40.
      2. Nikiforov I. A., Chernobrovkina T. V., Zhovnerchuk E. V., Isaev R. N. et al. Liver damage in alcoholism (literature review). Preventive medicine. 2014; 17(3): 55-61.
      3. Ress C., Kaser S. Mechanisms of intrhepatic triglyceride accumulation. World J. Gastroenterol. 2016, Vol. 22, № 4, pp. 1664-1673. DOI: 10.3748/wjg.v22.i4.1664.
      4. Balukova E. V., Uspensky Yu.P., Fominykh Yu. A. Liver affections of various genesis (toxic, medicinal, dismetabolic): from etiological heterogeneity to a single unified patient therapy. Russian Medical Journal. Medical Review. 2018, no. 1(I), pp. 35-40.
      5. Diomidova V. N., Tarasova L. V., Trukhan D. I., Tsyganova Yu.V., Vinogradova V. S. Informativeness of elastography with shear wave with elastometry in case of non-alcoholic fatty liver disease. Practical medicine. 2018; 1(112): 81-85.
      6. Toshikuni N., Tsutsumi M., Arisawa T. Clinical differences between alcoholic liver disease and nonalcoholic fatty liver disease. World J. Gastroenterol. 2014, Vol. 20(26), pp. 8393-8406. doi: 10.3748/wjg.v20.i26.8393.
      7. Ivashkin V. T., Mayevskaya M. V., Pavlov Ch.S. et al. Clinical recommendations of the Russian Society for the Study of the Liver on the management of adult patients with alcoholic liver disease. Russian Journal of Gastroenterology, Hepatology, Coloproctology. 2017; 27(6): 20-40. DOI: 10.22416 / 1382-4376-2017-27-6-20-40.
      8. Cara Torruellas, Samuel W. French, Valentina Medici. Diagnosis of alcoholic liver disease. World J. Gastroenterol. 2014, Vol. 20(33), pp. 11684-11699. doi: 10.3748/wjg.v20.i33.11684.
      9. Kruchinina M. V., Kurilovich S. A., Gromov A. A. et al. To the question of the differential diagnosis of alcoholic and non-alcoholic fatty liver disease. International Journal of Applied and Basic Research. 2016, no. 7-1, pp. 36-45. URL: http://applied-research.ru/ru/article/view?id=9753 (access date: 01/15/2019).
      10. Singal A.K., Bataller R., Ahn J., Kamath P. S., Shah V. H. ACG Clinical Guideline: Alcoholic Liver Disease. The American Journal of Gastroenterology. Vol. 113(2), pp. 175-194. doi:10.1038/ajg.2017.469.
      11. Bi W.R., Yang C. Q., Shi Q., Xu Y., Cao C. P., Ling J., Wang X. Y. Largescale analysis of factors influencing nonalcoholic fatty liver disease and its relationship with liver enzymes. Genet. Mol. Res. 2014, Vol. 13, pp. 5880-5891. doi: 10.4238/2014.August.7.3.
      12. van Beek J.H, de Moor M. H., Geels L. M. et al. The association of alcohol intake with gamma-glutamyl transferase (GGT) levels: evidence for correlated genetic effects. Drug Alcohol Depend. 2014, Vol. 134, pp. 99-105.
      13. Shigeo Sueyoshi, Setsu Sawai, Mamoru Satoh et al. Fractionation of gamma-glutamyltransferase in patients with nonalcoholic fatty liver disease and alcoholic liver disease. World J. Hepatol. 2016, Vol. 8(36), pp. 1610-1616. doi: 10.4254/wjh.v8.i36.1610.
      14. EASL Clinical Practical Guidelines: Management of Alcoholic Liver Disease. J. Hepatol. 2018, Vol. 69, pp. 154-181.
      15. Tarasova L. V. Alcoholic liver disease - the most urgent problem of modern hepatology. Remedium Volga. 2016, no. 9, pp. 15-20.
      16. Zvenigorodskaya L. A., Shinkin M. V. Alcoholic liver disease and non-alcoholic fatty liver disease. Similarities and differences. Features of treatment. Consilium Medicum. 2017; 19(8): 97-102.
      17. Polyzos S.A., Kountouras J., Anastasilakis A. D. et al. Irisin in patients with nonalcoholic fatty liver disease. Metabolism. 2014, Vol. 63(2), pp. 207-217. doi: 10.1016/j.metabol.2013.09.013.
      18. Dietrich P., Hellerbrand C. Non-alcoholic fatty liver disease, obesity and the metabolic syndrome. Best Pract. Res. Clin. Gastroenterol. 2014, Vol. 28(4), pp. 637-653.
      19. Sowa J.-P., Atmaca Ö., Kahraman A. et al. Non-Invasive Separation of Alcoholic and Non-Alcoholic Liver Disease with Predictive Modeling. PLoS ONE. Vol. 9(7). e101444. doi:10.1371/journal.pone.0101444.
      20. Elke Albrecht, Frode Norheim, Bernd Thiede et al. Irisin - a myth rather than an exercise-inducible myokine. Scientific Reports. 2015, Vol. 5, Article number: 8889.
      21. Tatsuo Inamine, Bernd Schnabl. Immunoglobulin A and liver diseases. J. Gastroenterol. 2018, Vol. 53(6). Р. 691-700. doi: 10.1007/s00535-017-1400-8.
      22. Novikov D. G., Dementi L. I., Indutny A. V., Trofimovich N. A., Nikolaev N. A. Laboratory and medical aspects of the integral assessment of the level of alcoholism in the youth environment. Modern problems of science and education. 2015, no.5, pp.352.
      23. Sanja Stojsavljević, Marija Gomerčić Palčić, Lucija Virović Jukić, Lea Smirčić Duvnjak, Marko Duvnjak. Adipokines and proinflammatory cytokines, the key mediators in the pathogenesis of nonalcoholic fatty liver disease. World J. Gastroenterol. 2014, Vol. 20(48), pp. 18070-18091. doi: 10.3748/wjg.v20.i48.18070.
      24. Samala N., Tersey S. A., Chalasani N., Anderson R. M., Mirmira R. G. Molecular mechanisms of nonalcoholic fatty liver disease: Potential role for 12-lipoxygenase. J Diabetes Complications. 2017, Vol. 11, pp. 1630-1637. doi: 10.1016/j.jdiacomp.2017.07.014.
      25. Yang M., Liu Y., Zhou G., Guo X., Zou S., Liu S., Jiang L., Liu Y., Zhu L., Guo C., Zhao J. Value of serum osteoprotegerin in noninvasive diagnosis of nonalcoholic steatohepatitis. Zhonghua Gan. Zang Bing Za Zhi. 2016, Vol. 24(2), pp. 96-101. doi: 10.3760/cma.j.issn.1007-3418.2016.02.005.
      26. Petukhov A. E., Nadezhdin A. V., Bogstrand S. T. et al. Phosphatidylethanol as a biomarker of alcohol abuse. Narcology. 2017; 2(182): 42-47.
      27. Waszkiewicz N., Chojnowska S., Zalewska A. et al. Salivary exoglycosidases as markers of alcohol dependence. Alcohol Alcohol. 2014, Vol. 49(4), pp. 409-416. doi: 10.1093/alcalc/agu005.
     


    Full text is published :
    Tarasova L. V., Tsyganova Yu. V., Opalinskaya I. V., Ivanova A. L. Overview of laboratory diagnostic methods used in nonalcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD) at the modern stage. Experimental and Clinical Gastroenterology. 2019;164(4): 72–77. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-72-77
    Read & Download full text

    1. Research Institute of Human Morphology, 117418, Moscow, Russia
    2. City Clinical Hospital № 31 of Moscow Health Department», 119415, Moscow, Russia
    3. FSBEI HPE RNSMU. N. I. Pirogov, 117997, Moscow, Russia

    Keywords: early gastric cancer, early gastric cancer, prognosis, treatment, oncology, endoscopy, pathological anatomy

    Abstract: Modern data on endoscopic and morphological factors for the prognosis of early gastric cancer (macroscopic type of tumor, its size and the presence or absence of ulceration, histological type and degree of differentiation of the tumor, immunophenotype, depth of invasion, the presence of cancer emboli in the lumen of lymphatic and blood vessels) are given. The indications for performance and criteria for the effectiveness of endoscopic surgical methods for treating early gastric cancer, recommendations for further patient management tactics are given.

      1. Fred T. Bosman, Fatima Carneiro, Ralph H. Hruban, Neil D. Theise WHO Classification of Tumors of the Digestive Sydtem. 4th Edition. Lyon, 2010.
      2. Mihaleva L. M., Birukov A.E, Plahov R. V. The algorithm of clinical and morphological diagnosis of early gastric cancer. Clinical and Experimental Morphology. 2015; 1(13):19-23.
      3. Mihaleva L. M., Birjukov A.E, Plahov R. V. Modern clinical and morphological approach to early gastric cancer. I. P. Pavlov Russian Medical Biological Herald. 2014; 4:35-41.
      4. Bing Hu, Nassim El Hajj, Scott Sittler, Nancy Lammert, et al. Gastric cancer: Classification, histology and application of molecular pathology. J Gastrointest Oncol. 2012 Sep; 3(3): 251-261.
      5. Hoon Jai Chun. Diagnosis of gastric epithelial neoplasia: Dilemma for Korean pathologists. World J Gastroenterol 2011 June 7; 17(21): 2602-2610.
      6. Min Jung Jo, Ji Yeon Park, Joon Seon Song, Myeong-Cherl Kook et al. Biopathologic features and clinical significance of micrometatasis in the lymph node of early gastric cancer. World J Gastroenterol. 2015; 21(2): 667-674.
      7. Everett S. M., Axon A. T. Early gastric cancer in Europe Gut. 1997 Aug; 41(2):142-50.
      8. Levickij A. V. Osobennosti rasprostraneniya, limfogennogo metastazirovaniya i rezul`taty` khirurgicheskogo lecheniya bol`ny`kh rannim rakom zheludka. Kand, Diss. [Features of distribution, lymphatic metastasis and the results of surgical treatment of patients with early gastric cancer. Kand, Diss.]. Moscow, 2009. 173 p.
      9. Kaprin A. D., Starinskij V. V., Petrova G. P. Zlokachestvenny`e novoobrazovaniya v Rossii v 2013 godu (zabolevaemost` i smertnost`) [Malignant tumors in Russia in 2013 (morbidity and mortality)]. Moscow, Federal State Budgetary Institution National Medical Research Radiological Center of the Ministry of Health of the Russian Federation Publ., 2015. 250 p.
      10. Kaprin A. D., Starinskij V. V., Petrova G. P. Zlokachestvenny`e novoobrazovaniya v Rossii v 2013 godu (zabolevaemost` i smertnost`) [Malignant tumors in Russia in 2017 (morbidity and mortality)]. Moscow, Federal State Budgetary Institution National Medical Research Radiological Center of the Ministry of Health of the Russian Federation Publ., 2018. 250 p.
      11. Lambert R. Participants in the Paris Workshop. The Paris endoscopic classification of superficial neoplastic lesions: esophagus, stomach, and colon: November 30 to December 1, 2002. Gastrointest. Endosc. 2003; 58 (6 Suppl): S3-43.
      12. Ohara Y., Toshikuni N., Matsueda K., Mouri H., et al. The superficial elevated and depressed lesion type is an independent factor associated with non-curative endoscopic submucosal dissection for early gastric cancer. Surg Endosc. 2016; 30 (11): 4880-88.
      13. Abe S., Oda I., Shimazu T., Kinjo T., et al. Depth-predicting score for differentiated early gastric cancer. Gastric Cancer. 2011; 14 (1): 35-40.
      14. Nakamura T., Yao T., Kakeji Y., Anai H., et al. Depressed type of intramucosal differentiated-type gastric cancer has high cell proliferation and reduced apoptosis compared with the elevated type. Gastric Cancer. 2013 Jan; 16 (1): 94-9.
      15. Neil A. Shepherd, Bryan F. Warren, Geraint T. Williams, et al. Morson and Dawson`s Gastrointestinal Pathology. 5th Edition. 2013.
      16. Bing Hu, Nassim El Hajj, Scott Sittler, Nancy Lammert, et al. Gastric cancer: Classification, histology and application of molecular pathology. J Gastrointest Oncol. 2012 Sep; 3(3):251-261.
      17. Fang C., Shi J., Sun Q., Gold J. S., et al. Risk factors of lymph node metastasis in early gastric carcinomas diagnosed by WHO criteria in 379 Chinese patients. J Dig Dis. 2016; 17 (8): 526-537.
      18. Lee J. H., Choi I. J., Kook M. C., Nam B. H., et al. Risk factors for lymph node metastasis in patients with early gastric cancer and signet ring cell histology. Br J Surg. 2010 May; 97 (5): 732-6.
      19. Fang W. L., Wu C. W., Lo S. S., Chen J. H., et al. Mucin-producing gastric cancer: clinicopathological difference between signet ring cell carcinoma and mucinous carcinoma. Hepatogastroenterology. 2009 Jul-Aug; 56 (93): 1227-31.
      20. Adachi Y., Yasuda K., Inomata M., Shiraishi N., et al. Clinicopathologic study of early-stage mucinous gastric carcinoma. Cancer. 2001 Feb 15; 91 (4): 698-703.
      21. Caruso R. A. The histogenesis of mucinous adenocarcinoma of the stomach from observations in early gastric cancer. Ann Diagn Pathol. 1999 Jun; 3 (3): 160-4.
      22. Kim Y. H., Park J. H., Park C. K., Kim J. H., et al. Histologic purity of signet ring cell carcinoma is a favorable risk factor for lymph node metastasis in poorly cohesive, submucosa-invasive early gastric carcinoma. Gastric Cancer. 2017 Jul; 20 (4): 583-590. https://doi.org/10.1007/s10120-016-0645-x
      23. Pyo J. H., Lee H., Min B. H., Lee J. H., et al. Early gastric cancer with a mixed-type Lauren classification is more aggressive and exhibits greater lymph node metastasis. J Gastroenterol. 2017; 52 (5): 594-601.
      24. Schlemper RJ, Itabashi M, Kato Y, et al. Differences in diagnostic criteria for gastric carcinoma between Japanese and western pathologists. Lancet 1997;349:1725-1729.
      25. Tsutomu Namikawa, Kazuhiro Hanazaki. Mucin phenotype of gastric cancer and clinicopathology of gastric-type differentiated adenocarcinoma. World J Gastroenterol. 2010 Oct 7; 16(37): 4634-4639.
      26. Dae Hwan Kim, Nari Shin, Gwang Ha Kim, Geum Am Song, et al. Mucin Expression in Gastric Cancer: Reappraisal of Its Clinicopathologic and Prognostic Significance. Archives of Pathology & Laboratory Medicine: August 2013, Vol. 137, No. 8, pp. 1047-1053.
      27. Koseki K, Takizawa T, Koike M, Ito M, et al. Distinction of differentiated type early gastric carcinoma with gastric type mucin expression. Cancer. 2000;89:724-732.
      28. Tsuji N, Ishiguro S, Mano M, Kasugai T, et al. Time trends for small gastric cancer in Japan. Gastric Cancer. 2000;3:123-127.
      29. Kabashima A, Yao T, Sugimachi K, Tsuneyoshi M. Relationship between biologic behavior and phenotypic expression in intramucosal gastric carcinomas. Hum Pathol. 2002;33:80-86.
      30. Kabashima A, Yao T, Maehara Y, Tsuneyoshi M. Relationship between biological behavior and phenotypic expression in undifferentiated-type gastric carcinomas. Gastric Cancer. 2005;8:220-227.
      31. Xiao-Tong Wang, Fan-Biao Kong, Wei Mai, Lei Li, et al. MUC1 Immunohistochemical Expression as a Prognostic Factor in Gastric Cancer: Meta-Analysis. Volume 2016, Article ID9421571, 8 pages http://dx.doi.org/10.1155/2016/9421571
      32. Y. Tamura, M. Higashi, S. Kitamoto et al., “MUC4 and MUC1 expression in adenocarcinoma of the stomach correlates with vessel invasion and lymph node metastasis: an immunohistochemical study of early gastric cancer”. PLoS ONE, vol. 7, no. 11, Article ID e49251, 2012.
      33. J. Y. Wang, C. T. Chang, J. S. Hsieh et al., “Role of MUC1 and MUC5AC expressions as prognostic indicators in gastric carcinomas,” Journal of Surgical Oncology, vol. 83, no. 4, pp. 253-260, 2003.
      34. J. Pinto-de-Sousa, L. David, C. A. Reis et al., “Mucins MUC1, MUC2, MUC5AC and MUC6 expression in the evaluation of differentiation and clinico-biological behaviour of gastric carcinoma,” Virchows Archiv, vol. 440, no. 3, pp. 304-310, 2002.
      35. Nalan Akyürek, GülenAkyol, AyşeDursun, DenizYamaç, et al. Expression of MUC1 and MUC2 Mucins in Gastric Carcinomas: Their Relationship with Clinicopathologic Parameters and Prognosis Pathology - Research and Practice. Volume 198, Issue 10, 2002, рр. 665-674.
      36. Chuan-Tao Zhang, Ke-Cheng He, Fei Pan, Yuan Li, et al. Prognostic value of Muc5AC in gastric cancer: A meta-analysis. World J Gastroenterol. 2015 Sep 28; 21(36): 10453-10460.
      37. James D. Brierley, Mary K. Gospodarowicz, Christian Wittekind TNM: Klassifikacziya zlokachestvenny`kh opukholej [TNM: Classification of Malignant Tumors, 8th ed]. Moscow, Logosfera Publ, 2018. 344 p.
      38. Japanese Gastric Cancer Association. Japanese classification of gastric carcinoma: 3rd English edition. Gastric Cancer. 2011 Jun; 14(2):101-12.
      39. Pedro Pimentel-Nunes, Mário Dinis-Ribeiro, Thierry Ponchon, Alessandro Repici, et al. Endoscopic submucosal dissection: European Society of Gastrointestinal Endoscopy (ESGE) Guideline. Endoscopy 2015; 47: 829-854.
      40. Hatta W, Gotoda T, Oyama T, Kawata N, et al. A scoring system to stratify curability after endoscopic submucosal dissection for early gastric cancer: “eCura system”. Am J Gastroenterol. 2017;112:874-81.
      41. Hiroyuki Ono, Kenshi Yao, Mitsuhiro Fujishiro, Ichiro Oda, et al. Guidelines for endoscopic submucosal dissection and endoscopic mucosal resection for early gastric cancer. Digestive Endoscopy. 2016;28:3-15.
      42. Besova N. S., Byahov M. Yu., Gorbunova V. A. et al. Klinicheskie rekomendaczii po diagnostike i lecheniyu bol`ny`kh rakom zheludka [Clinical guidelines for the diagnosis and treatment of patients with gastric cancer]. Assocziacziya onkologov Rossii [Association of Oncologists of Russia]. Moscow, 2014.
     


    Full text is published :
    Mikhaleva L. M., Fedorov E. D., Birukov A. E. Endoscopic and morphological prognostic factors of early gastric cancer. Experimental and Clinical Gastroenterology. 2019;164(4): 78–84. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-78-84
    Read & Download full text

    1. St. Petersburg State University, 199034, University emb, 7/9, St. Petersburg, Russia
    2. FSBU “National Almazov Medical Research Centre”, Ministry of Health, St. Petersburg, Russia

    Keywords: pancreatic cysts, cystic neoplasias of the pancreas, incident cysts, incidentaloma, pancreatic cancer, IPMN, MCN, SCN, EUS

    Abstract: Cystic neoplasms of the pancreas is a group of neoplastic changes in the epithelium of the ducts or parenchyma, which are the difficult diagnostic problems of modern medicine. With the improvement of diagnostic methods, the frequency of detection of incident cysts has increased. The article discusses the differential diagnostic characters, morphological features, and the potential for malignancy of pancreatic cystic neoplasms.

      1. Megibow AJ, Baker ME, Gore RM, Taylor A. The incidental pancreatic cyst. RadiolClin North Am. 2011; 49(2):349-359.
      2. de Jong K, Nio CY, Hermans JJ et al. High prevalence of pancreatic cysts detected by screening magnetic resonance imaging examinations. Clini Gastroenterol Hepatol. 2010; 8: 806-11.
      3. Chernyak V, Flusberg M, Haramati LB, Rozenblit AM, Bellin E. Incidental Pancreatic Cystic Lesions: Is There a Relationship with the Development of Pancreatic Adenocarcinoma and All-Cause Mortality? Radiology: Volume 274: Number 1-January 2015
      4. Tanaka M, Fernandez-del Castilio C, Adsay V, Chari S, Falconi M, et al. International consensus guidelines 2012 for the management of IPMN and MCN of the pancreas. Pancreatology. 2012; 12(3): 183-197.
      5. Birkmeyer J. D., Finlayson S. R., Tosteson A. N., Sharp S. M., Warshaw A. L., Fisher E. S. “Effect of hospital volume on in-hospital mortality with pancreaticoduodenectomy,” Surgery. vol. 125, no. 3, pp. 250-256, 1999.
      6. Gomez D., Rahman S. H., Wong L. F., Verbeke C. S., Menon K. V. “Predictors of malignant potential of cystic lesions of the pancreas,” European Journal of Surgical Oncology. vol. 34, no. 8, pp. 876-882, 2008.
      7. Birkmeyer J. D., Warshaw A. L., Finlayson S. R., Grove M. R., Tosteson A. N. “Relationship between hospital volume and late survival after pancreaticoduodenectomy,” Surgery. vol. 126, no. 2, pp. 178-183, 1999.
      8. Scheiman JM, Hwang JH, Moayyedi P. American gastroenterological association technical review on the diagnosis and management of asymptomatic neoplastic pancreatic cysts. Gastroenterology. 2015; 148: 824-48
      9. Javia S, Munigala S, Guha S, Agarwal B. EUS Morphology Is Reliable in Selecting Patients with Mucinous Pancreatic Cyst(s) Most Likely to Benefit from Surgical Resection. Gastroenterology Research and Practice. 2017.
      10. Lee LS. Diagnostic approach to pancreatic cysts. Curr Opin Gastroenterol. 2014;30:511-517.
      11. Goh BK, Tan YM, Tan PH, Ooi LL. Mucinous nonneoplastic cyst of the pancreas: a truly novel pathological entity? World J Gastroenterol. 2005; 11:2045-2047
      12. Yoon WJ, Brugge WR. Pancreatic cystic neoplasms: diagnosis and management. Gastroenterol Clin North Am. 2012;41:103-18.
      13. Spinelli KS, Fromwiller TE, Daniel RA, et al. Cystic pancreatic neoplasms: observe or operate. Ann Surg. 2004;239:651-7; discussion 657-9.
      14. Brugge W.R. “Diagnosis and management of cystic lesions of the pancreas” J Gastrointest Oncol. 2015;6(4):375-388
      15. Niki A. et al. «Pancreatic Intraepithelial Neoplasia and Pancreatic Tumorigenesis Of Mice and Men». Arch Pathol Lab Med. Vol. 133, March 2009
      16. Scheiman JM, Hwang JH, Moayyedi P. American gastroenterological association technical review on the diagnosis and management of asymptomatic neoplastic pancreatic cysts. Gastroenterology. 2015; 148: 824-48.
      17. Munigala S, Gelrud A, Agarwal B. Risk of pancreatic cancer in patients with pancreatic cyst. Gastrointestinal Endoscopy. 2016 No. 1: Volume 84, pp 81-86.
      18. Lekkerkerkera SJ, Besselinkb MG, Buschb OR. Long-term follow-up of neoplastic pancreatic cysts without high-risk stigmata: how often do we change treatment strategy because of malignant transformation?. SCANDINAVIAN JOURNAL OF GASTROENTEROLOGY. 2016 VOL. 51, NO. 9, 1138-1143
      19. Adsay, N.V.; Longnecker, D.S.; Klimstra, D. S. Pancreatic tumors with cystic dilatation of the ducts: Intraductal papillary mucinous neoplasms and intraductal oncocytic papillary neoplasms. Semin. Diagn. Pathol. 2000, 17, 16-30.
      20. Distler, M.; Kersting, S.; Niedergethmann, M.; Aust, D.E.; Franz, M.; Ruckert, F.; Ehehalt, F.; Pilarsky, C.; Post, S.; Saeger, H.D.; et al. Pathohistological subtype predicts survival in patients with intraductal papillary mucinous neoplasm (IPMN) of the pancreas. Ann. Surg. 2013, 258, 324-330
      21. Elta GH, Enestvedt BK, Sauer BG, Lennon AM. ACG Clinical Guideline: Diagnosis and Management of Pancreatic Cysts. Am J Gastroenterol advance online publication, 27 February 2018; doi: 10.1038/ajg.2018.14
      22. Morales-Oyarvide V, Fong ZV, Fernández-Del Castillo C, Warshaw AL. Intraductal Papillary Mucinous Neoplasms of the Pancreas: Strategic Considerations. Visc Med. 2017; 33(6): 466-476.
      23. Farrell JJ, Brugge WR. Intraductal papillary mucinous tumor of the pancreas. Gastrointest Endosc. 2002;55:701-14.
      24. Bauer F. Pancreatic Cystic Lesions: Diagnostic, Management and Indications for Operation. Part II Chirurgia. 2018;113: 318-334 No. 3, May - June
      25. Liu K, Joshi V, Camp L et al. Prevalence and outcomes of pancreatic cystic neoplasms in liver transplant recipients. World J Gastroenterol. 2017 December 28; 23(48): 8526-8532
      26. Choi SH, Park SH, Kim KW et al. Progression of unresected intraductal papillary mucinous neoplasms of the pancreas to cancer: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2017; 15: 1509-20.
      27. WHO Classification of Tumours of the Digestive System. Ed.by Bosman FT et.al, 2010, Lyon
      28. Basturk O, Hong SM, Wood LD, et al. Baltimore Consensus Meeting: A revised classification system and recommendations from the Baltimore consensus meeting for neoplastic precursor lesions in the pancreas. Am J Surg Pathol. 2015; 39: 1730-1741.
      29. Furukawa T, Hatori T, Fujita I, et al. Prognostic relevance of morphological types of intraductal papillary mucinous neoplasms of the pancreas. Gut. 2011;60:509-16.
      30. Morales-Oyarvide V, Fong ZV, Castillo CF, Warshaw AL. Intraductal Papillary Mucinous Neoplasms of the Pancreas: Strategic Considerations Visc Med 2017;33:466-476 467
      31. Pusateri AJ, Krishna SG. Pancreatic Cystic Lesions: Pathogenesis and Malignant Potential Diseases. 2018, 6, 50; doi:10.3390
      32. Pittman ME, Rao R, Hruban RH. Classification, Morphology, Molecular Pathogenesis, and Outcome of Premalignant Lesions of the Pancreas Arch Pathol Lab Med. Vol. 141, December 2017
      33. Grutzmann, R.; Niedergethmann, M.; Pilarsky, C.; Kloppel, G.; Saeger, H. D. Intraductal papillary mucinous tumors of the pancreas: Biology, diagnosis, and treatment. Oncologist. 2010, 15, 1294-1309.
      34. Koh, Y.X.; Zheng, H.L.; Chok, A.Y.; Tan, C.S.; Wyone, W.; Lim, T.K.; Tan, D.M.; Goh, B. K. Systematic review and meta-analysis of the spectrum and outcomes of different histologic subtypes of noninvasive and invasive intraductal papillary mucinous neoplasms. Surgery. 2015, 157, 496-509.
      35. Kechagias D, Laspas F. (2015) Imaging of Pancreatic Cystic Neoplasms. In: Sakorafas G, Smyrniotis V, Sarr M, (Eds). Pancreatic Cystic Neoplasms: From Imaging to Differential Diagnosis and Management. Springer, Milano, Italia, pp. 37-52.
      36. Baker, M.L.; Seeley, E.S.; Pai, R.; Suriawinata, A.A.; Mino-Kenudson, M.; Zamboni, G.; Kloppel, G.; Longnecker, D. S. Invasive mucinous cystic neoplasms of the pancreas. Exp. Mol. Pathol. 2012, 93, 345-349.
      37. Bai, X.; Ye, L.; Zhang, Q.; Prasoon, P.; Wang, J.; Liang, T. Surgical resection and outcome of pancreatic cystic neoplasms in China: Analysis of a 16-year experience from a single high-volume academic institution. World J. Surg. Oncol. 2014, 12, 228
      38. Spinelli KS, Fromwiller TE, Daniel RA, Kiely JM, Nakeeb A, et al. (2004) Cystic pancreatic neoplasms: observe or operate. Ann Surg. 239(5):651-657.
      39. Jang KT, Park SM, Basturk O, et al. Clinicopathologic characteristics of 29 invasive carcinomas arising in 178 pancreatic mucinous cystic neoplasms with ovarian-type stroma: implications for management and prognosis. Am J Surg Pathol. 2015; 39(2):179-187
      40. Di Paola V, Manfredi R, Mehrabi S, et al. Pancreatic mucinous cystoadenomas and cystoadenocarcinomas: differential diagnosis by means of MRI. Br J Radiol. 2016;89(1057):20150536. doi:10.1259/bjr.20150536.
      41. Jang, K.T.; Park, S.M.; Basturk, O.; Bagci, P.; Bandyopadhyay, S.; Stelow, E.B.; Walters, D.M.; Choi, D.W.; Choi, S.H.; Heo, J.S.; et al. Clinicopathologic characteristics of 29 invasive carcinomas arising in 178 pancreatic mucinous cystic neoplasms with ovarian-type stroma: Implications for management and prognosis. Am. J. Surg. Pathol. 2015, 39, 179-187.
      42. Crippa, S.; Salvia, R.; Warshaw, A.L.; Dominguez, I.; Bassi, C.; Falconi, M.; Thayer, S.P.; Zamboni, G.; Lauwers, G.Y.; Mino-Kenudson, M.; et al. Mucinous cystic neoplasm of the pancreas is not an aggressive entity: Lessons from 163 resected patients. Ann. Surg. 2008, 247, 571-579.
      43. Dijck WP, Groot VP, Brosens LA, Hagendoorn J, Rinkes IB, et al. IQ Rare Case of an Epithelial Cyst in an Intrapancreatic Accessory Spleen Treated by Robot-Assisted Spleen Preserving Distal Pancreatectomy. Case Reports in Gastrointestinal Medicine. 2016; Volume 2016.
      44. The European Study Group on Cystic Tumours of the Pancreas. Gut. 2018;67:789-804. doi:10.1136/gutjnl-2018-316027
      45. Postlewait LM, Ethun CG, Mclnnis MR Association of Preoperative Risk Factors With Malignancy in Pancreatic Mucinous Cystic Neoplasms: A Multicenter Study. JAMA Surg. 2017 January 01; 152(1): 19-25.
      46. Bauer F. Pancreatic Cystic Lesions: Diagnostic, Management and Indications for Operation. Part I. Chirurgia (Bucur). 2017; 112(2):97-109.
      47. Graham R, Smyrk T. Pathology of Pancreatic Cystic Neoplasms. In Sakorafas G, Smyrniotis V, Sarr M, editors. Pancreatic Cystic Neoplasms: From Imaging to Differential Diagnosis and Management. Ed. Springer; 2015. p. 11-24.
      48. Strobel O, Z’graggen K, Schmitz-Winnenthal FH, et al. Risk of malignancy in serous cystic neoplasms of the pancreas. Digestion. 2003; 68: 24-33.
      49. Tseng JF, Warshaw AL, Sahani DV, et al. Serous cystadenoma of the pancreas: tumor growth rate and recommendations for treatment. Ann Surg. 2005; 242: 413-421.
      50. George DH, Murphy F, Michalski R, et al. Serous cystadenocarcinoma of the pancreas: a new entity? Am J Surg Pathol. 1989; 13: 61-66.
      51. Kamei K, Funabiki T, Ochiai M, Amano H, Kasahara M, et al. Multifocal pancreatic serous cystadenoma with atypical cells and focal perineural invasion. Int J Pancreatol. 1991; 10(2): 161-172.
      52. Yoshimi N, Sugie S, Tanaka T, Aijin W, Bunai Y, et al. A rare case of serous cystadenocarcinoma of the pancreas. Cancer. 1992; 69(10): 2449-2453.
      53. Ohta T, Nagakawa T, Itoh H, Fonseca L, Miyazaki I, et al. A case of serous cystadenoma of the pancreas with focal malignant changes. Int J Pancreatol. 1993; 14(3): 283-289.
      54. Widmaier U, Mattfekdt T, Siech M, Beger HG. Serous cystadenocarcinoma of the pancreas. Int J Pancreatol. 1996; 20(2): 135-139.
      55. Eriguchi N, Aoyagi S, Nakayama T, Hara M, Miyazaki T, et al. Serous cystadenocarcinoma of the pancreas with liver metastases. J Hepatobiliary PancreatSurg. 1998; 5(4): 467-470.
      56. Abe H, Kubota K, Mori M, Miki K, Minagawa M, et al. Serous cystadenoma of the pancreas with invasive growth: benign or malignant?. Am J Gastroenterol. 1998; 93(10): 1963-1966.
      57. Shintaku M, Arimoto A, Sakita N. Serous cystadenocarcinoma of the pancreas. PatholInt. 2005; 55(7): 573-580.
      58. Matsumoto T, Hirano S, Yada K, Shibata K, Sasaki A, et al. Malignant serous cystic neoplasm of the pancreas: report of a case and review of the literature. J ClinGastroenterol. 2005; 39(3): 253-256.
      59. Friebe V, Keck T, Mattern D, Schmitt-Graeff A, Werner M, et al. Serous cystadenocarcinoma of the pancreas: management of a rare entity. Pancreas. 2005; 31(2): 182-187.
      60. Hruban RH, Klimstra DS, Pitman MB. Atlas of Tumor Pathology. Tumors of the Pancreas. 4th series. Washington, American Institute of Pancreatology, 2006, pp 45-47.
      61. Reid MD, Choi HJ, Memis B, et al. Serous neoplasms of the pancreas: a clinicopathologic analysis of 193 cases and literature review with new insights on macrocystic and solid variants and critical reappraisal of so-called ‘‘serous cystadenocarcinoma.’’ Am J Surg Pathol. 2015;39(12):1597-1610.
      62. Fukasawa M, Maguchi H, Takahashi K, Katanuma A, Osanai M, et al. (2010) Clinical Features and Natural History of Serous Cystic Neoplasm of the Pancreas. Pancreatology. 10(6):695-701.
      63. Strobel O, Z’graggen K, Schmitz-Winnenthal FH, et al. Risk of malignancy in serous cystic neoplasms of the pancreas. Digestion. 2003; 68: 24-33.
      64. Galanis C, Zamani A, Cameron JL, et al. Resected serous cystic neoplasms of the pancreas: a review of 158 patients with recommendations for treatment. J Gastrointest Surg. 2007;11:820-6.
      65. Khashab MA, Shin EJ, Amateau S, et al. Tumor size and location correlate with behavior of pancreatic serous cystic neoplasms. Am J Gastroenterol. 2011;106:1521-6.
      66. Abe H, Kubota K, Mori M, et al. Serous cystadenoma of the pancreas with invasive growth: benign or malignant? Am J Gastroenterol. 1998; 93: 1963-1966.
      67. Hwang HK, Chung YE, Kim HK et al. Serous Cystic Neoplasm: Do We Have to Wait Till It Causes Trouble? Korean Journal of HBP Surgery. Vol. 15, No. 2, May 2011.
      68. Tipton SG, Smyrk TC, Sarr MG, et al. Malignant potential of solid pseudopapillary neoplasm of the pancreas. Br J Surg. 2006;93:733-7.
      69. Chiang AL, Lee LS. Clinical approach to incidental pancreatic cysts World J Gastroenterol. 2016 January 21; 22(3): 1236-1245.
      70. S. Reddy, J. L. Cameron, J. Scudiere et al. “Surgical management of solid-pseudopapillary neoplasms of the pancreas (Franz or Hamoudi Tumors): a large single-institutional series,” Journal of the American College of Surgeons, vol. 208, no. 5, pp. 950-957, 2009.
      71. Jani N, Hani MB, Schulick RD, Hruban R, Cunningham SC. Diagnosis and Management of Cystic Lesions of the Pancreas Diagnostic and Therapeutic Endoscopy. Volume 2011, Article ID478913, 10 pages
      72. Martin RCG, Klimstra DS, Brennan MF, Conlon KC. “Solid-pseudopapillary tumor of the pancreas: a surgical enigma?” Annals of Surgical Oncology, vol. 9, no. 1, pp. 35-40, 2002.
      73. Chang MK, Kyung SK, Jin SC, Kim H, Woo JL, Byong RK. “Solid pseudopapillary tumor of the pancreas suggesting malignant potential,” Pancreas, vol. 32, no. 3, pp. 276-280, 2006.
      74. Song H, Dong M, Zhou J, Sheng W, Zhong B, Gao W. Solid Pseudopapillary Neoplasm of the Pancreas: Clinicopathologic Feature, Risk Factors of Malignancy, and Survival Analysis of 53 Cases from a Single Center. Hindawi BioMed Research International. Volume 2017, Article ID5465261, 7 pages
      75. Bauer F. Pancreatic Cystic Lesions: Diagnostic, Management and Indications for Operation. Part II Chirurgia. 2018; 113: 318-334 No. 3, May - June
      76. Koh YX, Chok AY, Zheng HL, et al. A systematic review and metaanalysis of the clinicopathologic characteristics of cystic versus solid pancreatic neuroendocrine neoplasms. Surgery. 2014; 156(1): 83-96.e2, doi: 10.1016/j.surg.2014.03.026, indexed in Pubmed: 24878455.
      77. Hurtado-Pardo L, A Cienfuegos J, Ruiz-Canela M, et al. Cystic pancreatic neuroendocrine tumors (cPNETs): a systematic review and meta-analysis of case series. Rev Esp Enferm Dig. 2017; 109(11): 778-787, doi: 10.17235/ reed.2017.5044/2017, indexed in Pubmed: 29072081.
      78. Caglià P, Cannizzaro MT, Tracia A, et al. Cystic pancreatic neuroendocrine tumors: To date a diagnostic challenge. Int J Surg. 2015; 21 Suppl 1: S44-S49, doi: 10.1016/j.ijsu.2015.04.087, indexed in Pubmed: 26118611.
      79. Bordeianou L, Vagefi PA, Sahani D, et al. Cystic pancreatic endocrine neoplasms: a distinct tumor type? J Am Coll Surg. 2008; 206(6): 1154-1158, doi: 10.1016/j.jamcollsurg.2007.12.040, indexed in Pubmed: 18501813.
      80. Dąbkowski K, Kos-Kudł B, Andrysiak-Mamos E, Syrenicz A, Pilch-Kowalczyk J, Starzyńska T. “Cystic pancreatic neuroendocrine tumours - a gastroenterologist’s point of view” Endokrynologia Polska. Vol. 69; no 3/2018
      81. Kashchenko VA, Solonitsyn EG, Vasyukova EL, Berko OM, Bakirov II. Differential Diagnosis of Pancreatic Cyst Tumors. Gastroenterology Medicine & Research. 2018; ISSN2637-7632 Volume 2 Issue 3
     


    Full text is published :
    Kashchenko V. A., Solonitsyn E. G., Danilov I. N., Vanyan A. V., Glistenkova D. D., Neklyudova P. I., Kamalova V. F. The potential of the malignancy of cystic neoplasms of the pancreas. Experimental and Clinical Gastroenterology. 2019;164(4): 85–92. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-85-92
    Read & Download full text

    1. Skobelkin state scientifi c center of laser medicine, Moscow, Russia
    2. RUDN University, Moscow, Russia

    Keywords: colorectal cancer, colonoscopy, adenoma detection rate (ADR), chromoendoscopy, virtual chromoendoscopy, cap, Endocuff , Endorings, FUSE

    Abstract: Colonoscopy has been established as the gold standard for the detection of colorectal neoplasms. Adenoma detection rate (ADR) is one of the most important quality indicator of colonoscopy, and, as it has been recommended by a European Society of Gastrointestinal Endoscopy (ESGE), the benchmark for ADR is 20%. However, ADR widely varies, and almost one fourth of polyps and adenomas are missed. Currently several techniques have been developed with the aim to improve the quality of colonoscopy. Among them there are image-enhanced endoscopy (IEE) and techniques or devices that improve mucosal observation. IEE includes chromoendoscopy and virtual chromoendoscopy (NBI, FICE, AFI, i-scan); among techniques which enable better observation of the mucosa there are add-on devices, such as transparent cap, Endocuff, Endocuff Vision, Endorings; and wide-angle colonoscopes (full spectrum endocopy - FUSE). In the literature review, these mentioned techniques and their effect on ADR are described.

      1. International Agency for Research on Cancer. World cancer observatory. GLOBOCAN2018. Lyon: International Agency for Research on Cancer; 2018. http://gco.iarc.fr/ Дата обращения 22.01.2019.
      2. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018 Nov;68(6):394-424.
      3. Kaprin A. D., Starinsky V. V., Petrova G. V. Malignant neoplasms in Russia in 2017 (morbidity and mortality). Moscow: MNII them. P. A. Herzen - branch of the Federal State Budgetary Institution Scientific Research Center for Radiology of the Ministry of Health of Russia; 2018, 250 p.
      4. Ananyev V. S., Artamonova E. V., Achkasov S. I. et al. Clinical recommendations for the diagnosis and treatment of patients with colon cancer. Moscow. 2014, 13 p.
      5. Winawer S., Classen M., Lambert R., et al. World Gastroenterological Society (GGO) and International Digestive Cancer Prevention Practical Guide: Screening for Colorectal Cancer. World Gastroenterology Organisation; 2008. 17 P.
      6. Ageĭkina NV, Duvanskiĭ VA, Kniazev MV. An alternative pathway of colorectal cancer development. Endoscopic and morphological features of serrated lesions. Review. Eksp Klin Gastroenterol. 2013;(8):3-10.
      7. Kaminski MF, Thomas-Gibson S, Bugajski M, et al. Performance measures for lower gastrointestinal endoscopy: a European Society of Gastrointestinal Endoscopy (ESGE) quality improvement initiative. United European Gastroenterol J. 2017 Apr;5(3):309-334.
      8. Kashin S. V., Nekhaykova N. V. Quality indicators in screening colonoscopy for colorectal polyps and cancer. Russian journal of Evidence-based gastroenterology = Dokazatel'naya gastroenterologiya. 2016;5(2):13-21. https://doi.org/10.17116/dokgastro20165213-21
      9. Heresbach D, Barrioz T, Lapalus MG, et al. Miss rate for colorectal neoplastic polyps: a prospective multicenter study of back-to-back video colonoscopies.Endoscopy. 2008 Apr;40(4):284-290.
      10. Leufkens AM, van Oijen MG, Vleggaar FP, Siersema PD. Factors influencing the miss rate of polyps in a back-to-back colonoscopy study. Endoscopy. 2012; 44:470-475.
      11. Xiang L, Zhan Q, Zhao XH, et al. Risk factors associated with missed colorectal flat adenoma: a multicenter retrospective tandem colonoscopy study. World J Gastroenterol. 2014; 20:10927-10937.
      12. Barclay RL1, Vicari JJ, Doughty AS, et al. Colonoscopic withdrawal times and adenoma detection during screening colonoscopy. N Engl J Med. 2006 Dec 14;355(24):2533-2541.
      13. Bartel MJ, Picco MF, Wallace MB. Chromocolonoscopy. Gastrointest Endosc Clin N Am. 2015 Apr;25(2):243-260.
      14. Pohl J1, Schneider A, Vogell H, et al. Pancolonic chromoendoscopy with indigo carmine versus standard colonoscopy for detection of neoplastic lesions: a randomised two-centre trial. Gut. 2011 Apr;60(4):485-490.
      15. Brooker JC, Saunders BP, Shah SG, et al. Total colonic dye-spray increases the detection of diminutive adenomas during routine colonoscopy: a randomized controlled trial. Gastrointest Endosc. 2002 Sep;56(3):333-338.
      16. Hurlstone DP, Cross SS, Slater R et al. Detecting diminutive colorectal lesions at colonoscopy: a randomised controlled trial of pan-colonic versus targeted chromoscopy. Gut. 2004 Mar;53(3):376-380.
      17. Brown SR, Baraza W, Din S, Riley S. Chromoscopy versus conventional endoscopy for the detection of polyps in the colon and rectum. Cochrane Database Syst Rev. 2016 Apr 7;4: CD006439.
      18. Saitoh Y, Waxman I, West AB, et al. Prevalence and distinctive biologic features of flat colorectal adenomas in a North American population. Gastroenterology. 2001 Jun;120(7):1657-1665.
      19. Paggi S, Radaelli F, Amato A, et al. The impact of narrow band imaging in screening colonoscopy: a randomized controlled trial. Clin Gastroenterol Hepatol. 2009 Oct;7(10):1049-1054.
      20. Kaltenbach T, Friedland S, Soetikno R. A randomised tandem colonoscopy trial of narrow band imaging versus white light examination to compare neoplasia miss rates. Gut. 2008 Oct;57(10):1406-1412.
      21. Rex DK, Helbig CC. High yields of small and flat adenomas with high-definition colonoscopes using either white light or narrow band imaging. Gastroenterology. 2007 Jul;133(1):42-47.
      22. Adler A, Pohl H, Papanikolaou IS, et al. A prospective randomised study on narrow-band imaging versus conventional colonoscopy for adenoma detection: does narrow-band imaging induce a learning effect? Gut. 2008 Jan;57(1):59-64.
      23. Inoue T, Murano M, Murano N, et al. Comparative study of conventional colonoscopy and pan-colonic narrow-band imaging system in the detection of neoplastic colonic polyps: a randomized, controlled trial. J Gastroenterol. 2008;43(1):45-50.
      24. Uraoka T, Saito Y, Matsuda T, et al. Detectability of colorectal neoplastic lesions using a narrow-band imaging system: a pilot study. J Gastroenterol Hepatol. 2008 Dec;23(12):1810-1815.
      25. Nagorni A, Bjelakovic G, Petrovic B. Narrow band imaging versus conventional white light colonoscopy for the detection of colorectal polyps. Cochrane Database Syst Rev. 2012 Jan 18;1: CD008361.
      26. Leung WK, Lo OS, Liu KS, et al. Detection of colorectal adenoma by narrow band imaging (HQ190) vs. high-definition white light colonoscopy: a randomized controlled trial. Am J Gastroenterol. 2014 Jun;109(6):855-863.
      27. Belkov A. V., Duvanskiy V. A. Spectral color separation technology with endoscopic diagnosis of colon tumors. Experimental and Clinical Gastroenterology. 2017;144(08):23-25.
      28. Aminalai A, Rösch T, Aschenbeck J, et al. Live image processing does not increase adenoma detection rate during colonoscopy: a randomized comparison between FICE and conventional imaging (Berlin Colonoscopy Project 5, BECOP-5). Am J Gastroenterol. 2010 Nov;105(11):2383-2388.
      29. Chung SJ, Kim D, Song JH, et al. Efficacy of computed virtual chromoendoscopy on colorectal cancer screening: a prospective, randomized, back-to-back trial of Fuji Intelligent Color Enhancement versus conventional colonoscopy to compare adenoma miss rates. Gastrointest Endosc. 2010 Jul;72(1):136-142.
      30. Pohl J, Lotterer E, Balzer C, et al. Computed virtual chromoendoscopy versus standard colonoscopy with targeted indigocarmine chromoscopy: a randomised multicentre trial. Gut. 2009 Jan;58(1):73-78.
      31. Knyazev M. V., Duvanskii V. A., Ageikina N. V. Trimodal'naya endoskopiya v diagnostike zabolevanii zheludochno-kishechnogo trakta. Klin. endoskop. 2012. vol. 4. p. 2.
      32. Douvansky V. A. Kniazev M. V. Autofluorescent endoscopic diagnostics of epithelial neoplasms in the colon. Journal of Gastroenterology and Hepatology. 2015;30 (Suppl. 4): 211.
      33. Aihara H, Tajiri H, Suzuki T. Application of Autofluorescence Endoscopy for Colorectal Cancer Screening: Rationale and an Update. Gastroenterol Res Pract. 2012; 2012: 971383.
      34. Matsuda T, Saito Y, Fu KI, Uraoka T, et al. Does autofluorescence imaging videoendoscopy system improve the colonoscopic polyp detection rate? - a pilot study. Am J Gastroenterol. 2008 Aug;103(8):1926-1932.
      35. Ramsoekh D, Haringsma J, Poley JW, et al. A back-to-back comparison of white light video endoscopy with autofluorescence endoscopy for adenoma detection in high-risk subjects. Gut. 2010 Jun;59(6):785-793.
      36. Takeuchi Y, Inoue T, Hanaoka N, et al. Autofluorescence imaging with a transparent hood for detection of colorectal neoplasms: a prospective, randomized trial. Gastrointest Endosc. 2010 Nov;72(5):1006-1013.
      37. Hoffman A, Sar F, Goetz M, et al. High definition colonoscopy combined with i-Scan is superior in the detection of colorectal neoplasias compared with standard video colonoscopy: a prospective randomized controlled trial. Endoscopy. 2010 Oct;42(10):827-833.
      38. Hong SN, Choe WH, Lee JH, et al. Prospective, randomized, back-to-back trial evaluating the usefulness of i-SCAN in screening colonoscopy. Gastrointest Endosc. 2012 May;75(5):1011-1021.
      39. Rex DK, Repici A, Gross SA, et al. High-definition colonoscopy versus Endocuff versus EndoRings versus full-spectrum endoscopy for adenoma detection at colonoscopy: a multicenter randomized trial. Gastrointest Endosc. 2018 Aug;88(2):335-344.
      40. Kondo S, Yamaji Y, Watabe H, et al. A randomized controlled trial evaluating the usefulness of a transparent hood attached to the tip of the colonoscope. Am J Gastroenterol. 2007;102:75-81.
      41. Harada Y, Hirasawa D, Fujita N, et al. Impact of a transparent hood on the performance of total colonoscopy: a randomized controlled trial. Gastrointest Endosc. 2009;69:637-644.
      42. Tang Z, Zhang DS, Thrift AP, Patel KK. Impact of cap-assisted colonoscopy on the learning curve and quality in colonoscopy: a randomized controlled trial. Gastrointest Endosc. 2018;87:723-732.
      43. Park SM, Lee SH, Shin KY, et al. The cap-assisted technique enhances colonoscopy training: prospective randomized study of six trainees. Surg Endosc. 2012;26:2939-2943.
      44. Floer M, Biecker E, Fitzlaff R, et al. Higher adenoma detection rates with endocuff-assisted colonoscopy-a randomized controlled multicenter trial. PLoS One. 2014;9: e114267.
      45. Biecker E, Floer M, Heinecke A, et al. Novel endocuff-assisted colonoscopy significantly increases the polyp detection rate: a randomized controlled trial. J Clin Gastroenterol. 2015;49:413-418.
      46. Triantafyllou K, Polymeros D, Apostolopoulos P, et al. Endocuff-assisted colonoscopy is associated with a lower adenoma miss rate: a multicenter randomized tandem study. Endoscopy. 2017;49:1051-1060
      47. De Palma GD, Giglio MC, Bruzzese D, et al. Cap cuff-assisted colonoscopy versus standard colonoscopy for adenoma detection: a randomized back-to-back study. Gastrointest Endosc. 2018;87:232-240.
      48. Ngu WS1, Bevan R2, Tsiamoulos ZP et al. Improved adenoma detection with Endocuff Vision: the ADENOMA randomised controlled trial. Gut. 2019 Feb;68(2):280-288.
      49. Dik VK, Gralnek IM, Segol O, et al. Multicenter, randomized, tandem evaluation of EndoRings colonoscopy - results of the CLEVER study. Endoscopy. 2015;47:1151-1158.
      50. Deenadayalu VP, Chadalawada V, Rex DK. 170 degrees wide-angle colonoscope: effect on efficiency and miss rates. Am J Gastroenterol. 2004 Nov;99(11):2138-2142.
      51. Fatima H, Rex DK, Rahmani E, et al. Wide-Angle (WA) (170° Angle of View) Versus Standard (ST) (140°Angle of View) Colonoscopy. Gastrointest Endosc. 2006;63: AB204.
      52. Gralnek IM, Siersema PD2, Halpern Z, et al. Standard forward-viewing colonoscopy versus full-spectrum endoscopy: an international, multicentre, randomised, tandem colonoscopy trial. Lancet Oncol. 2014 Mar;15(3):353-360.
     


    Full text is published :
    Duvanskiy V. A., Chesalina Ya. O. Advanced endoscopic technologies for detection of colorectal neoplasms. Experimental and Clinical Gastroenterology.2019;164(4): 93–99. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-93-99
    Read & Download full text

    1. Skobelkin state scientifi c center of laser medicine, Moscow, Russia
    2. RUDN University, Moscow, Russia
    3. Leningrad Regional Clinical Hospital, St. Petersburg, Russia

    Keywords: lateral spreading tumor, endoscopic resection, submucosal injection, carbon dioxide, hemostasis

    Abstract: Objective. The purpose of the literature review is to present the latest literature data on endoscopic resection of a laterally spreading colon tumor. Conclusions. Most non-malignant colon neoplasias can be removed by the endoscopic method. When removing LST, it is necessary to focus on high-quality endoscopic imaging, magnification, chromoscopy and narrow-spectrum endoscopy. LST resection should be carried out according to the accepted method. Before making a decision on endoscopic resection of neoplasia, all its characteristics should be carefully evaluated. The analysis of cases of incomplete removal and recurrence of formation is associated with a size of more than 40 mm, localization in the ileocecal colon, the characteristics of the patching pattern and the probability of invasion.

      1. Jung JS, Hong JY, Oh HH, Kweon SS, Lee J, Kim SW, Seo GS, Kim HS, Joo YE. Clinical outcomes of endoscopic resection for colorectal laterally spreading tumors with advanced histology. Surg Endosc. 2018 Oct 22. doi: 10.1007/s00464-018-6550-0.
      2. Knyazev MV, Duvanskiy VA. Endoscopic mucosal resection with submucosal dissection esd and the first long-term results of applying this method. Eksp Klin Gastroenterol. 2015;(4):53-8.
      3. Ageĭkina NV, Duvanskiĭ VA, Kniazev MV. An alternative pathway of colorectal cancer development. Endoscopic and morphological features of serrated lesions. Review. Eksp Klin Gastroenterol. 2013;(8):3-10.
      4. Uraoka T., Saito Y., Matsuda T., Ikehara H., Gotoda T., Saito D. et al. Endoscopic indications for endoscopic mucosal resection of laterally spreading tumours in the colorectum. Gut. 2006; 55(11): 1592-7.
      5. Duvansky V. A., Knyazev M. V. Endoscopic treatment of gastrointestinal neoplasia - evolution of the method. Grekov's Bulletin of Surgery. 2015;174(2):130-134. (In Russ.)
      6. Moss A, Bourke MJ, Williams SJ. et al. Endoscopic mucosal resection outcomes and prediction of submucosal cancer from advanced colonic mucosal neoplasia. Gastroenterology 2011; 140: 1909-1918 DOI: 10.1053/j.gastro.2011.02.062
      7. Buchner AM, Guarner-Argente C, Ginsberg GG. Outcomes of EMR of defiant colorectal lesions directed to an endoscopy referral center. Gastrointest Endosc 2012; 76: 255-263 DOI: 10.1016/j.gie.2012.02.060
      8. Oba S, Tanaka S, Oka S. et al. Characterization of colorectal tumors using narrow-band imaging magnification: combined diagnosis with both pit pattern and microvessel features. Scand J Gastroenterol 2010; 45: 1084-1092 DOI: 10.3109/00365521003734166
      9. Shimura T, Ebi M, Yamada T. et al. Magnifying chromoendoscopy and endoscopic ultrasonography measure invasion depth of early stage colorectal cancer with equal accuracy on the basis of a prospective trial. Clin Gastroenterol Hepatol 2014; 12: 662-668 e661-662 DOI: 10.1016/j.cgh.2013.06.022
      10. Jang HW, Park SJ, Cheon JH. et al. Does magnifying narrow-band imaging or magnifying chromoendoscopy help experienced endoscopists assess invasion depth of large sessile and flat polyps?. Dig Dis Sci 2014; 59: 1520-1528 doi: 10.3748/wjg.v16.i14.1727
      11. Beaton C, Twine CP, Williams GL. et al. Systematic review and meta-analysis of histopathological factors influencing the risk of lymph node metastasis in early colorectal cancer. Colorectal Dis 2013; 15: 788-797 DOI: 10.1111/codi.12129
      12. Kethu SR, Banerjee S, Desilets D. et al. Endoscopic tattooing. Gastrointest Endosc 2010; 72: 681-685 DOI: 10.1016/j.gie.2010.06.020
      13. Askin MP, Waye JD, Fiedler L. et al. Tattoo of colonic neoplasms in 113 patients with a new sterile carbon compound. Gastrointest Endosc 2002; 56: 339-342
      14. Voiosu TA, Margarit C, Rimbas M. et al. Polypectomy practices in a real life setting. Do we do enough for our patients? A review of 1061 colonoscopies. Rom J Intern Med 2011; 49: 257-265
      15. Pohl H, Srivastava A, Bensen SP. et al. Incomplete polyp resection during colonoscopy-results of the complete adenoma resection (CARE) study. Gastroenterology 2013; 144: 74-80 e71. DOI: 10.1053/j.gastro.2012.09.043
      16. Muniraj T, Sahakian A, Ciarleglio MM. et al. Cold snare polypectomy for large sessile colonic polyps: a single-center experience. Gastroenterol Res Pract 2015; 2015: 175959 DOI: 10.1155/2015/175959
      17. Watabe H, Yamaji Y, Okamoto M. et al. Risk assessment for delayed hemorrhagic complication of colonic polypectomy: polyp-related factors and patient-related factors. Gastrointest Endosc 2006; 64: 73-78
      18. Kim HS, Kim TI, Kim WH. et al. Risk factors for immediate postpolypectomy bleeding of the colon: a multicenter study. Am J Gastroenterol 2006; 101: 1333-1341
      19. Longcroft-Wheaton G, Duku M, Mead R. et al. Risk stratification system for evaluation of complex polyps can predict outcomes of endoscopic mucosal resection. Dis Colon Rectum 2013; 56: 960-966 DOI: 10.1097/DCR.0b013e31829193e0
      20. Augusto Barros R, Monteverde MJ, Federico Barros R. et al. Safety and efficacy of cold snare resection of non-polypoid colorectal lesions (0-IIa and 0-IIb). Acta Gastroenterol Latinoam 2014; 44: 27-32
      21. Dobrowolski S, Dobosz M, Babicki A. et al. Blood supply of colorectal polyps correlates with risk of bleeding after colonoscopic polypectomy. Gastrointest Endosc 2006; 63: 1004-1009
      22. Nanda KS, Tutticci N, Burgess NG. et al. Endoscopic mucosal resection of laterally spreading lesions involving the ileocecal valve: technique, risk factors for failure, and outcomes. Endoscopy 2015; 47: 710-718 DOI: 10.1055/s-0034-1391732
      23. Swan MP, Bourke MJ, Alexander S. et al. Large refractory colonic polyps: is it time to change our practice? A prospective study of the clinical and economic impact of a tertiary referral colonic mucosal resection and polypectomy service (with videos). Gastrointest Endosc 2009; 70: 1128-1136 DOI: 10.1016/j.gie.2009.05.039
      24. Brooker JC, Saunders BP, Shah SG. et al. Endoscopic resection of large sessile colonic polyps by specialist and non-specialist endoscopists. Br J Surg 2002; 89: 1020-1024 DOI: 10.1046/j.1365-2168.2002.02157.x
      25. Hong YM, Kim HW, Park SB. et al. Endoscopic mucosal resection with circumferential incision for the treatment of large sessile polyps and laterally spreading tumors of the colorectum. Clin Endosc 2015; 48: 52-58 doi: 10.5946/ce.2015.48.1.52
      26. Masci E, Viale E, Notaristefano C. et al. Endoscopic mucosal resection in high- and low-volume centers: a prospective multicentric study. Surg Endosc 2013; 27: 3799-3805 doi: 10.1007/s00464-013-2977-5
      27. Bahin FF, Pellise M, Williams SJ. et al. Extended endoscopic mucosal resection does not reduce recurrence compared with standard endoscopic mucosal resection of large laterally spreading colorectal lesions. Gastrointest Endosc 2016; DOI: 10.1016/j.gie.2016.05.015.
      28. Desomer L, Tutticci N, Tate DJ. et al. A standardized imaging protocol is accurate in detecting recurrence after endoscopic mucosal resection. Gastrointest Endosc 2016; Jun 22. pii: S0016-5107(16)30277-2. DOI: 10.1016/j.gie.2016.06.031.
      29. Nanda KS, Tutticci N, Burgess NG. et al. Endoscopic mucosal resection of laterally spreading lesions involving the ileocecal valve: technique, risk factors for failure, and outcomes. Endoscopy 2015; 47: 710-718 DOI: 10.1055/s-0034-1391732.
      30. Hassan C, Repici A, Sharma P. et al. Efficacy and safety of endoscopic resection of large colorectal polyps: a systematic review and meta-analysis. Gut 2016; 65: 806-820 DOI: 10.1136/gutjnl-2014-308481.
      31. Fujiya M, Tanaka K, Dokoshi T. et al. Efficacy and adverse events of EMR and endoscopic submucosal dissection for the treatment of colon neoplasms: a meta-analysis of studies comparing EMR and endoscopic submucosal dissection. Gastrointest Endosc 2015; 81: 583-595 DOI: 10.1016/j.gie.2014.07.034
      32. Holt BA, Bourke MJ. Wide field endoscopic resection for advanced colonic mucosal neoplasia: current status and future directions. Clin Gastroenterol Hepatol 2012; 10: 969-979 DOI: 10.1016/j.cgh.2012.05.020
      33. Kishihara T, Chino A, Uragami N. et al. Usefulness of sodium hyaluronate solution in colorectal endoscopic mucosal resection. Dig Endosc 2012; 24: 348-352 DOI: 10.1111/j.1443-1661.2012.01244.x
      34. Katsinelos P, Kountouras J, Paroutoglou G. et al. A comparative study of 50% dextrose and normal saline solution on their ability to create submucosal fluid cushions for endoscopic resection of sessile rectosigmoid polyps. Gastrointest Endosc 2008; 68: 692-698 doi: 10.2147/CEG.S29704
      35. Hurlstone DP, Fu KI, Brown SR. et al. EMR using dextrose solution versus sodium hyaluronate for colorectal Paris type I and 0-II lesions: a randomized endoscopist-blinded study. Endoscopy 2008; 40: 110-114 DOI: 10.1055/s-2007-966987.
      36. Puli SR, Kakugawa Y, Gotoda T. et al. Meta-analysis and systematic review of colorectal endoscopic mucosal resection. World J Gastroenterol 2009; 15: 4273-4277 doi: 10.3748/wjg.15.4273.
      37. Miller K, Waye JD. Polyp retrieval after colonoscopic polypectomy: use of the Roth Retrieval Net. Gastrointest Endosc 2001; 54: 505-507.
      38. Singh N, Harrison M, Rex DK. A survey of colonoscopic polypectomy practices among clinical gastroenterologists. Gastrointest Endosc 2004; 60: 414-418.
      39. Carter D, Beer-Gabel M, Zbar A. et al. A survey of colonoscopic polypectomy practice amongst Israeli gastroenterologists. Ann Gastroenterol 2013; 26: 135-140.
      40. Parra-Blanco A, Kaminaga N, Kojima T. et al. Colonoscopic polypectomy with cutting current: is it safe?. Gastrointest Endosc 2000; 51: 676-681.
      41. Wu J, Hu B. The role of carbon dioxide insufflation in colonoscopy: a systematic review and meta-analysis. Endoscopy 2012; 44: 128-136 DOI: 10.1055/s-0031-1291487.
      42. Hsu WF, Hu WH, Chen YN. et al. Carbon dioxide insufflation can significantly reduce toilet use after colonoscopy: a double-blind randomized controlled trial. Endoscopy 2014; 46: 190-195 DOI: 10.1055/s-0034-1365016.
      43. Bahin FF, Naidoo M, Williams SJ. et al. Prophylactic endoscopic coagulation to prevent bleeding after wide-field endoscopic mucosal resection of large sessile colon polyps. Clin Gastroenterol Hepatol 2015; 13: 724-730 DOI: 10.1016/j.cgh.2014.07.063.
      44. Shioji K, Suzuki Y, Kobayashi M. et al. Prophylactic clip application does not decrease delayed bleeding after colonoscopic polypectomy. Gastrointest Endosc 2003; 57: 691-694 DOI: 10.1067/mge.2003.193.
      45. Liaquat H, Rohn E, Rex DK. Prophylactic clip closure reduced the risk of delayed postpolypectomy hemorrhage: experience in 277 clipped large sessile or flat colorectal lesions and 247 control lesions. Gastrointest Endosc 2013; 77: 401-407 DOI: 10.1016/j.gie.2012.10.024.
      46. Burgess NG, Bassan MS, McLeod D. et al. Deep mural injury and perforation after colonic endoscopic mucosal resection: a new classification and analysis of risk factors. Gut 2016; Jul 27. pii: gutjnl-2015-309848. doi: DOI: 10.1136/gutjnl-2015-309848.
     


    Full text is published :
    Chernykh D. A., Duvanskiy V. A. Endoscopic resection of a laterally spreading colon tumors. Experimental and Clinical Gastroenterology.2019;164(4): 100–104. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-100-104
    Read & Download full text

    1. The Federal State Budgetary Scientifi c Institution “Petrovsky National Research Center of Surgery”, 119991, Moscow, Russia

    Keywords: cardiospasm, achalasia cardia, endoscopic balloon dilation of the cardia, cardia pneumatic dilation, cardia hydrodilatation, POEM

    Abstract: Aim: the unresolved problem with the large amount of treatment options for the achalasia cardia indicates the absence of a perfect treatment method. The large interest in peroral endoscopic myotomy, as in any innovation, leads to disregard of other treatment methods and the absence of an alternative. Therefore, attention to a simple, low-cost and effective way of dilating the cardia should again be drawn. Materials and methods. This article summarizes the results of 30 years of experience in treating functional obstruction of the cardia with endoscopic balloon dilation in 353 patients. The technical features of the intervention with the specially designed balloons fixed to the endoscope, as well as large-diameter wire-guided balloons are described in the article. Results. Criteria for assessing the end of endoscopic treatment of achalasia and cardiospasm are given. It is shown that the correct technique of the intervention gives good and satisfactory immediate results up to 95%, long-term - up to 80% of cases with 1 to 26 years remission. Conclusion: endoscopic balloon dilation is an equal, well-developed minimally invasive method for treating achalasia cardia and can technically be performed at any stage of the disease.

      1. Chernousov A. F., Bogopolsky P. M., Kurbanov F. S. Esophageal surgery. A guide for doctors. Moscow. Medicine Publ., 2000, 350 p.
      2. Traube M., Hongo M., Magyar L., McCallum R. W. Effects of nifedipine in achalasia and in patients with high-amplitude peristaltic esophageal contractions. JAMA 1984; 252: 1733-6.
      3. Bassotti G., Annese V. Review article: pharmacological options in achalasia. Aliment. Pharmacol. Ther. 1999; 13: 1391-6.
      4. Chernousov A. F., Gadzhiev A. N., Shestakov A. L. Treatment cardiospasm and achalasia of cardia by pneumocardial dilatation. Annals of Surgery. 2000, No. 3, pp. 50-53.
      5. Ivashkin V. T., Trukhmanov A. S., Godzhello E. A. et al. Diagnostics and treatment of cardiac achalasia and cardiospasm: guidelines of the Russian gastroenterological association. Russian Journal of Gastroenterology, Hepatology, Coloproctology. 2016;26(4):36-54. (In Russ.) https://doi.org/10.22416/1382-4376-2016-26-4-36-54
      6. Vaezi M.F., Richter J. E. Diagnosis and management of achalasia. American College of Gastroenterology Practice Parameter Committee. Am J Gastroenterol. 1999; 94: 3406-3412.
      7. Gallinger Yu. I., Godzhello E. A. Operative endoscopy esophagus. Moscow. RSCH RAMS, 1999, 273 p.
      8. Chernousov A. F., Khorobrykh T. V., Vetshev F. P. et al. Esophageal achalasia and cardiospasm - contemporary principles of treatment. Annals of Surgery. 2012, No. 3, pp. 5-10.
      9. Inoue H., Minami H., Kobayashi Y. et al. Peroral endoscopic myotomy (POEM) for esophageal achalasia. Endoscopy 2010; 42: 265-71.
      10. Inoue H., Santi E. G., Onimaru M., Kudo S. E. Submucosal endoscopy: from ESD to POEM and beyond.Gastrointest Endosc Clin N Am. 2014; 24: 257-264.
      11. Allahverdyan A. S., Mazurin V. S., Titov A. G., Frolov A. V. Esophagocardiomyotomy with partial anteroposterior fundoplication in the treatment of cardia achalasia: a Training manual. Moscow, 2011, 24 p.
      12. Shestakov AL, Chernousov FA, Pastukhov DV. Surgical treatment of the benign diseases of the esophagus. Pirogov Russian Journal of Surgery = Khirurgiya. Zhurnal imeni N. I. Pirogova. 2013;(5):36-39.
      13. Sweet M.P, Nipomnick I., Gasper W.J et al. The outcome of laparoscopic Heller myotomy for achalasia is not influenced by the degree of esophageal dilatation. J. Gastrointest. Surg. 2008; 12:159-65.
      14. Duranceau A., Liberman M., Martin J., Ferraro P. End-stage achalasia. Dis. Esophagus 2012; 25: 319-30.
      15. Gadzhiev A. N. Clinic, diagnosis, treatment of cardiospasm, cardiac achalasia and diffuse esophagospasm. Diss. Doct. Moscow, 2001.
      16. Kahrilas P.J., Bredenoord A. J., Fox M. et al. The Chicago classification of esophageal motility disorders, v3.0. Neurogastroenterol. Motil. 2015; 27: 160-74.
     


    Full text is published :
    Godzhello E. A., Khrustaleva M. V., Bulganina N. A., Shatveryan D. G., Dekhtyar M. A. Endoscopic Balloon Dilation for Achalasia in the Era of Peroral Endoscopic Myotomy (POEM). Thirty years’ experience. Experimental and Clinical Gastroenterology. 2019;164(4): 105–110. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-105-110
    Read & Download full text

    1. FRC CSC RAS, Institute of Pharmacoinformatics, 119333, Moscow, Vavilova, 42, Russia
    2. Big Data Storage and Analysis Center (CCHD), Moscow State University, Moscow, Russia
    3. Federal State Budgetary Educational Institution of Further Professional Education “Russian Medical Academy of Continuous Professional Education” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia

    Keywords: prebiotics, lactitol, microbiome, human microbiota, area under the growth curve, chemoinformatics, data mining, system biology

    Abstract: Recently, more attention has been paid to studying the relationship between the state of the intestinal microbiome and the state of both the intestine itself and other human organs. In this connection, interest arises in studying the effect on the microbiome of both well-known molecules with a prebiotic effect and the lactose derivative, lactitol, which is relatively recently used in clinical practice. Prebiotic molecules differ significantly in their effects on the microbiome of the human intestine. A systematic study of this issue has not been conducted previously. This work presents the results of a comparative chemomicrobiome analysis of lactitol, lactulose, sorbitol, xylitol and fructose, carried out using modern technologies for the analysis of "big data". For each of the molecules studied, estimates of the area under the growth curve were obtained for a representative sample of human microbiota, including 36 commensal bacteria, including bifidobacteria and lactobacilli. It is shown that lactitol stimulates the growth of the population of bifidobacteria and beneficial eubacteria, as well as microorganisms producing butyric acid, to a greater extent than comparison molecules, and thus, to a greater extent than comparison molecules, it improves the condition of the colon.

      1. Gibson, G.R.; Probert, H.M.; van Loo, J.; Rastall, R.A.; Roberfroid, M. B. Dietary modulation of the human colonic microbiota: Updating the concept of prebiotics. Nutr. Res. Rev. 2004, 17, 259-275
      2. Vasquez KS, Shiver AL, Huang KC. Cutting the Gordian Knot of the Microbiota. Mol Cell. 2018 Jun 7;70(5):765-767. doi: 10.1016/j.molcel.2018.05.034. Epub 2018 Jun 7. PubMed ID:29883604
      3. Shawcross DL, Jalan R. Treatment of hepatic encephalopathy: it’s not lactulose. BMJ. 2004 Jul 10;329(7457):112; author reply 112. doi: 10.1136/bmj.329.7457.112. PubMed ID:15242927
      4. Maguire A, Rugg-Gunn AJ. Xylitol and caries prevention - is it a magic bullet? Br Dent J. 2003 Apr 26;194(8):429-36. doi: 10.1038/sj.bdj.4810022. PubMed ID:12778091
      5. Grimble GK, Patil DH, Silk DB. Assimilation of lactitol, an ‘unabsorbed’ disaccharide in the normal human colon. Gut. 1988 Dec;29(12):1666-71. PubMed ID:3220306
      6. Andersen JM, Barrangou R, Hachem MA, Lahtinen SJ, Goh YJ, Svensson B, Klaenhammer TR. Transcriptional analysis of prebiotic uptake and catabolism by Lactobacillus acidophilus NCFM. PLoS One. 2012;7(9): e44409. doi: 10.1371/journal.pone.0044409. Epub 2012 Sep 19. PubMed ID:23028535
      7. Torshin I. Y., Rudakov K. V. On the application of the combinatorial theory of solvability to the analysis of chemographs: Part 2. local completeness of invariants of chemographs in view of the combinatorial theory of solvability. Pattern Recognition and Image Analysis. 2014, Vol. 24, no. 2, pp. 196-208.
      8. Torshin I. Yu, Rudakov K. V. On metric spaces arising during formalization of problems of recognition and classification. Part 2: Density properties. Pattern Recognit. Image Anal. (2016) Vol. 26. № 3. p. 483-496.
      9. Torshin I. Y., Rudakov K. V. On metric spaces arising during formalization of recognition and classification problems. Part 1: Properties of compactness. Pattern Recognition and Image Analysis, 2016, Vol. 26, № 2, p. 274-284.
      10. Torshin I. Y. On solvability, regularity, and locality of the problem of genome annotation. Pattern Recognition and Image Analysis (Advances in Mathematical Theory and Applications). 2010. Т. 20. № 3. p. 386-395.
      11. Torshin I. Yu, Rudakov K. V. On the theoretical basis of metric analysis of poorly formalized problems of recognition and classification. Pattern Recognition and Image Analysis (2015). Vol. 25. № 4. p. 577-587.
      12. Torshin I. Yu, Rudakov K. V. On the procedures of generation of numerical features over the splits of a set of objects and the problem of prediction of numeric target variables. Pattern Recognition and Image Analysis (2019). Vol. 29. № 3. p. 65-75.
      13. Barclay T, Ginic-Markovic M, Johnston MR, Cooper P, Petrovsky N. Observation of the keto tautomer of D-fructose in D(2) O using (1) H NMR spectroscopy. Carbohydr Res. 2012 Jan 10;347(1):136-41. doi: 10.1016/j.carres.2011.11.003. Epub 2011 Nov 12. PubMed ID:22129837
      14. Maier L, Pruteanu M, Kuhn M, Zeller G, Telzerow A, Anderson EE, Brochado AR, Fernandez KC, Dose H, Mori H, Patil KR, Bork P, Typas A. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018 Mar 29; 555(7698):623-628. doi: 10.1038/nature25979. PMID: 29555994
      15. A framework for human microbiome research. Nature. 2012 Jun 13;486(7402):215-21. doi: 10.1038/nature11209. PubMed ID:22699610
      16. The Integrative Human Microbiome Project: dynamic analysis of microbiome-host omics profiles during periods of human health and disease. Cell Host Microbe. 2014 Sep 10;16(3):276-89. doi: 10.1016/j.chom.2014.08.014. PubMed ID:25211071
      17. Kim S, Chen J, Cheng T, Gindulyte A, He J, He S, Li Q, Shoemaker BA, Thiessen PA, Yu B, Zaslavsky L, Zhang J, Bolton EE. PubChem 2019 update: improved access to chemical data. Nucleic Acids Res. 2019 Jan 8;47(D1): D1102-D1109. doi: 10.1093/nar/gky1033. PubMed ID:30371825
      18. Barkham TM. Laboratory safety aspects of SARS at Biosafety Level 2.Ann Acad Med Singapore. 2004 Mar;33(2):252-6. PubMed ID:15098644
      19. Aleshkin V. A., Afanasyev S. S., Karaulov A. V. et al. MICROBIOCENOSIS AND HUMAN HEALTH. Moscow, 2015.
      20. Baron EJ. Bilophila wadsworthia: a unique Gram-negative anaerobic rod. Anaerobe. 1997 Apr-Jun;3(2-3):83-6. doi: 10.1006/anae.1997.0075. PubMed ID:16887567
      21. Donohoe DR, Garge N, Zhang X, Sun W, O’Connell TM, Bunger MK, Bultman SJ. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011 May 4;13(5):517-26. doi: 10.1016/j.cmet.2011.02.018. PubMed ID:21531334
      22. Kassinen A, Krogius-Kurikka L, Makivuokko H, Rinttila T, Paulin L, Corander J, Malinen E, Apajalahti J, Palva A. The fecal microbiota of irritable bowel syndrome patients differs significantly from that of healthy subjects. Gastroenterology. 2007 Jul;133(1):24-33. doi: 10.1053/j.gastro.2007.04.005. Epub 2007 Apr 14. PubMed ID:17631127
      23. Ulsemer P, Toutounian K, Schmidt J, Karsten U, Goletz S. Preliminary safety evaluation of a new Bacteroides xylanisolvens isolate.Appl Environ Microbiol. 2012 Jan;78(2):528-35. doi: 10.1128/AEM.06641-11. Epub 2011 Nov 18. PubMed ID:22101046
      24. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human intestinal microbial flora.Science. 2005 Jun 10;308(5728):1635-8. doi: 10.1126/science.1110591. Epub 2005 Apr 14. PubMed ID:15831718
      25. Zimmerman MA, Singh N, Martin PM, Thangaraju M, Ganapathy V, Waller JL, Shi H, Robertson KD, Munn DH, Liu K. Butyrate suppresses colonic inflammation through HDAC1-dependent Fas upregulation and Fas-mediated apoptosis of T cells. Am J Physiol Gastrointest Liver Physiol. 2012 Jun 15;302(12): G1405-15. doi: 10.1152/ajpgi.00543.2011. Epub 2012 Apr 19. PubMed ID:22517765
      26. Zeng H, Lazarova DL, Bordonaro M. Mechanisms linking dietary fiber, gut microbiota and colon cancer prevention.World J Gastrointest Oncol. 2014 Feb 15;6(2):41-51. doi: 10.4251/wjgo.v6.i2.41. PubMed ID:24567795
      27. Hsiao A, Ahmed AM, Subramanian S, Griffin NW, Drewry LL, Petri WA Jr, Haque R, Ahmed T, Gordon JI. Members of the human gut microbiota involved in recovery from Vibrio cholerae infection. Nature. 2014 Nov 20;515(7527):423-6. doi: 10.1038/nature13738. Epub 2014 Sep 17. PubMed ID:25231861
      28. Dabard J, Bridonneau C, Phillipe C, Anglade P, Molle D, Nardi M, Ladire M, Girardin H, Marcille F, Gomez A, Fons M. Ruminococcin A, a new lantibiotic produced by a Ruminococcus gnavus strain isolated from human feces.Appl Environ Microbiol. 2001 Sep;67(9):4111-8. PubMed ID:11526013
      29. Patterson AM, Mulder IE, Travis AJ, Lan A, Cerf-Bensussan N, Gaboriau-Routhiau V, Garden K, Logan E, Delday MI, Coutts AGP, Monnais E, Ferraria VC, Inoue R, Grant G, Aminov RI. Human Gut Symbiont Roseburia hominis Promotes and Regulates Innate Immunity.Front Immunol. 2017 Sep 26;8:1166. doi: 10.3389/fimmu.2017.01166. eCollection 2017. PubMed ID:29018440
      30. Burton JP, Drummond BK, Chilcott CN, Tagg JR, Thomson WM, Hale JD, Wescombe PA. Influence of the probiotic Streptococcus salivarius strain M18 on indices of dental health in children: a randomized double-blind, placebo-controlled trial.J Med Microbiol. 2013 Jun;62(Pt 6):875-84. doi: 10.1099/jmm.0.056663-0. Epub 2013 Feb 28. PubMed ID:23449874
      31. Burton JP, Chilcott CN, Moore CJ, Speiser G, Tagg JR. A preliminary study of the effect of probiotic Streptococcus salivarius K12 on oral malodour parameters.J Appl Microbiol. 2006 Apr;100(4):754-64. doi: 10.1111/j.1365-2672.2006.02837.x. PubMed ID:16553730
      32. Mohandas, Rajesh; Poduval, Rajiv D.; Unnikrishnan, Dilip; Corpuz, Marilou (2001). “Clostridium ramosum Bacteremia and Osteomyelitis in a Patient with Infected Pressure Sores”. Infectious Diseases in Clinical Practice. 10 (2): 123-24. doi:10.1097/00019048-200102000-00010.
      33. Moser SA, Savage DC. Bile salt hydrolase activity and resistance to toxicity of conjugated bile salts are unrelated properties in lactobacilli. Appl Environ Microbiol. 2001 Aug;67(8):3476-80. PMID: 11472922
      34. Ardatskaya M. D., Butorova L. I., Grigorieva Y. V., Loginov V. A., Loshchinina Y. N., Cheremushkin S. V. Clinical-metabolic efficiency of lactitol in the therapy of chronic constipation (results of the non-interventional prospective observation program “OSMOAID”). Experimental and Clinical Gastroenterology. 2018;(2):149-160. (In Russ.)
     


    Full text is published :
    Torshin I. Yu., Gromova O. A., Zakcharova I. N., Maximov V. A. Hemomikrobiomny lactitol analysis. Experimental and Clinical Gastroenterology. 2019;164(4): 111–121. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-111-121
    Read & Download full text

    1. Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation (Scientifi c Center for Children’s Health), Russia, 119991, Moscow
    2. Federal State Autonomous Educational Institution of Higher Education I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Russia, Moscow

    Keywords: full-spectrum colonoscopy, panoramic colonoscopy, FUSE, children, colorectal polyps, polypectomy, inflammatory bowel diseases

    Abstract: The aim was to assess the potential of the usage and efficacy of full-spectrum colonoscopy in children. Materials and methods: 22 children (1-17 years) were assessed with a full-spectrum colonoscope (FUSE system). Colorectal polyps were detected and removed in five of them. A new FUSE colonoscope has three cameras and allows a panoramic to 330 ° field of view. Results. With this full-spectrum colonoscope, endoscopists are able to view difficult- or impossible- to- view areas within the colon (“blind spots”), that allows to improve diagnostics of colorectal lesions. FUSE system helps in navigation within the colon. FUSE colonoscopy appears to be safe and efficient diagnostic and therapeutic method in children.

      1. Tringali A. et al. Pediatric gastrointestinal endoscopy: European Society of Gastrointestinal Endoscopy (ESGE) and European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) Guideline. Executive summary. Endoscopy 2017; 49: 83-91. DOI http://dx.doi.org/10.1055/s-0042-111002.
      2. Park J. H. Role of colonoscopy in the diagnosis and treatment of pediatric lower gastrointestinal disorders. Korean J Pediatr 2010; 53(9): 824-829. DOI: 10.3345/kjp.2010.53.9.824.
      3. Lokhmatov M. M., Budkina T. N., Oldakovsky V. I., Tupylenko A. V., Ibragimov S. I. Intraluminal endoscopy in children - past, present, future. Rossiiskiy Pediatricheskiy Zhurnal. 2018; 21(4): 230-236. DOI: 10.18821/1560-9561-2018-21-4-230-236.
      4. Gralnek IM. Evaluation of the EndoChoice full spectrum endoscopy (Fuse) platform for upper endoscopy and colonoscopy. Expert Rev Med Devices. 2016;13(4):313-9. DOI: 10.1586/17434440.2016.
      5. Roepstorff S., Hadi S. A., Rasmussen M. Scand J Gastroenterol. Full spectrum endoscopy (FUSE) versus standard forward-viewing endoscope (SFV) in a high-risk population. 2017 Nov; 52(11): 1298-1303. DOI: 10.1080/00365521.2017.1363278.
      6. Song JY, Cho YH, Kim MA, Kim JA, Lee CT, Lee MS. Feasibility of full-spectrum endoscopy: Korea’s first full-spectrum endoscopy colonoscopic trial. World J Gastroenterol. 2016; 22(8): 2621-9. doi:10.3748/wjg.v22.i8.2621.
      7. Gralnek IM, Siersema PD, Halpern Z, et al. Standard forward-viewing colonoscopy versus full-spectrum endoscopy: an international, multicentre, randomised, tandem colonoscopy trial. Lancet Oncol. 2014; 15(3): 353-360. doi: 10.1016/S1470-2045(14)70020-8.
      8. Kudo, T., Saito, Y., Ikematsu, H., et al. New-generation full-spectrum endoscopy versus standard forward-viewing colonoscopy: a multicenter, randomized, tandem colonoscopy trial (J-FUSE Study). Gastrointestinal Endoscopy. 2018 Nov; 88(5): 854-864. doi: 10.1016/j.gie.2018.06.011.
      9. Hassan C, Senore C, Radaelli F, et al. Full-spectrum (FUSE) versus standard forward-viewing colonoscopy in an organised colorectal cancer screening programme. Gut. 2017 Nov; 66(11): 1949-1955. doi: 10.1136/gutjnl-2016-311906.
      10. Facciorusso A., Del Prete V., Buccino V., et al. Full-spectrum versus standard colonoscopy for improving polyp detection rate: A systematic review and meta-analysis. J Gastroenterol Hepatol. 2018 Feb; 33(2): 340-346. doi: 10.1111/jgh.13859.
      11. Leong R.W., Ooi M., Corte C., et al. Full-Spectrum Endoscopy Improves Surveillance for Dysplasia in Patients With Inflammatory Bowel Diseases. Gastroenterology. 2017 May; 152(6): 1337-1344.e3. doi: 10.1053/j.gastro.2017.01.008.
      12. Veselov V. V., Nechipai A. M., Poltoryhina E. A., Vasilchenko A. V. First experience in full-spectrum colonoscopy. Coloproctology. 2017; 2 (60): 36-46.
      13. Thakkar K, Fishman DS, Gilger MA. Colorectal polyps in childhood. Curr Opin Pediatr. 2012 Oct; 24(5): 632-7. doi: 10.1097/MOP.0b013e328357419f.
     


    Full text is published :
    Budkina T. N., Lokhmatov M. M., Dyakonova E. Yu., Ibragimov S. I., Oldakovsky V. I., Tupylenko A. V. First experience of using full-spectrum colonoscopy in children. Experimental and Clinical Gastroenterology. 2019;164(4): 122–126. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-122-126.
    Read & Download full text

    1. State Budgetary Healthcare Establishment “Regional Clinical Hospital № 2” of the Ministry of Healthcare of the Krasnodar Region (GBUZ “KKB№ 2”) Krasnykh Partizan ul. 6, building 2 Krasnodar, 350012, Russia

    Keywords: Cholecysto-duodenal fi stula, choledocholithiasis, small bowel obstruction

    Abstract: The interest of the above clinical case is that cholecysto-duodenal fistula, which arose against the background of cholelithiasis and caused high obstructive small intestinal obstruction by biliary calculi migrated from it, is a very rare complication of gallstones and was diagnosed at the preoperative stage. Patient B, 75 years old, was admitted to GBUZ “KKB No. 2” in Krasnodar on July 21, 2016 with a diagnosis of Cholelithiasis. Chronic calculous cholecystitis. Choledocholithiasis. High small bowel obstruction? The first step was performed diagnostic esophagogastroduodenoscopy. When endoscopy at the output level of the LDPC, along the back wall, a wall defect in the form of a cholecysto-duodenal fistula with visualization of the gallbladder cavity is determined. At the level of the lower horizontal branch of the duodenum, there are mobile multiple biliary concrements that completely overlap the lumen of the duodenum. It was decided to perform endoscopic mechanical litoextraction of these stones. With the Dormia basket probe, movable duodenal calculi were captured and removed. After deleting the mobile calculus data, we found a large calculus that completely covered the duodenal lumen. An attempt was made to electrohydraulic lithotripsy of this large calculus of duodenum. The calculus was fragmented. But when trying to hold an endoscope for calculus fragments, in order to capture them with a basket of Dormia, the ischemic wall of the duodenum perforated. The patient was transferred to the operating room, where laparotomy, duodenotomy, removal of the wedged calculus of the lower horizontal branch of the duodenum, suturing of the duodenum defect were performed. On day 12 after surgery, the patient was discharged from the hospital in a satisfactory condition.

      1. Beltran M. A., Csendes A., Cruces K. S. The relationship of Mirizzi syndrome and cholecystoenteric fistula: validation of a modified classification. World J. Surg. 2008; 32 (10): 2237-2243. PMID: 18587614.
      2. Mithani R., Schwesinger W. H., Bingener J., Sirinek R., Gross G. W. The Mirizzi syndrome: multidisciplinary management promotes optimal outcomes. J. Gastrointest. Surg. 2008; 12 (6): 1022-1028. PMID: 17874273.
      3. Bedin V. V., Zarutskaya N. V., Pel’ts V. A. Ten-year experience of surgical treatment of patients with biliodigestive and cholecystoc holedocheal fistulas. Annaly khirurgicheskoy gepatologii. 2006; 11 (3): 72. (In Russian).
      4. Timerbulatov V. M., Garipov R. M., Khunafin S. N., Nurmukhametov A. A. Vnutrennie zhelchnie svischi. Sovremennie tehnologii v diagnostike i lechenii [Internal biliary fistulas. Modern technologies in diagnostics and treatment]. Moscow: Triada-X, 2003. 160 p. (In Russian).
      5. Topchiashvili Z. A., Kaprov I. B. Spontannie vnutrennie zhelchnie svischi. [Spontaneous internal biliary fistulas]. Tbilisi: Ganatleba, 1988. 132 p. (In Russian).
      6. Velsky AV, Ermolaev VV, Zakharova NG, Myshkin KI. Internal biliary fistulas in cholelithiasis. Surgery. 1978, no. 5, pp. 30-32.
      7. Matviychuk B. O., Bilyak S. S. Bouveret II Syndrome. Clinical Surgery. 1993, no. 6, p. 69.
      8. Tsybyrne K. A., Popov S. D., Chalganov A, I. Biliary fistula. Chisinau, Shtiintsa. 1983. pp. 120-142.
      9. Bilo M., Dzupa K. Akutny uzaver pyloru zicovym kamenom // Rozhl. CHIR.-1989.-v.68, N4., s.258-260.
      10. De Santis G., Pancotii G. Sindroime di Boweret. Presentazione dl un caso // MIN, GHIK. 1988, v. 435, N17, P. 1403-1405.
      11. Vantsyana E. N. External and internal fistulas. Moscow. Medicine, 1990, pp. 76-132.
      12. Sonak R., Tusek D., Rusche H. H., Mackrodi H. G. Das Bouveret-Sendrome - einesel-tene Form der Magenausgangsstenose. Zen-tralblatt fur Chirurgie. 1995, vol. 120(1), P. 75-78.
      13. Vidal 0., Seco J.L„ Alvarez A., Trinanes J. P., Serrano L. P., Serrano S. R. Sindrome de Bouveret: cinco casos. Revista Espanjla de Enfermedades Digestivas. l994, vol.86(5), P. 839-844.
      14. Baudet-Bourgarel A., Boruchowicz А„ Gambiez L., Paris L. С. Syndrome de Bouvere revele par une hematemese. Gastroenterologie Cliniqie et Biologique. 1996, vol.20 (1), P. 112-113.
     


    Full text is published :
    Gabriel S. A., Dynko V. Yu., Guchetl A. Ya., Tlekhuray R. M., Bespechnyj M. V., Mamishev A. K. Attempt to endoscopic treatment of high obstructive intestinal obstruction, due to biliary calculus. Experimental and Clinical Gastroenterology. 2019;164(4): 127–130. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-127-130
    Read & Download full text

    1. FSBEI of HE “The Chuvash State University n. a. I. N. Ulyanov”, 15 Moscow avenue, 428015 Cheboksary, Russia
    2. SAI SVE “The Institute of Postgraduate Medical Education” of the Chuvash Republic Ministry Healthcare, 27 Sespel street, 428018 Cheboksary, Russia
    3. BI of HE “The Surgut State University”, 1 Lenina street, 628412 Khanty-Mansi Autonomous Region — Ugra, Surgut, Russia
    4. BI "City clinical hospital № 1" of the Ministry of Нealthcare of the Chuvash Republic, Cheboksary, Russia

    Keywords: non-alcoholic fatty liver disease, steatosis, steatohepatitis, ursodeoxycholic acid

    Abstract:

      1. Vovk E. I. Nealkogolynaya ghirovaya bolezn' pecheni kak proaterogennoe zabolevanie: diagnostika I lechenie v obschey praktike. RMZh. Меditsinskoe obozrenie. 2017, no.2, pp. 68-79.
      2. Lazebnik L. B., Radchenko V. G., Golovanova E. V. et al. Nonalcoholic fatty liver disease: clinic, diagnostics, treatment (Recommendations for therapists, 2nd edition). Experimental and Clinical Gastroenterology Journal. 2017;138(02):22-37.
      3. Ivashkin V. T., Mayevskaya M. V., Pavlov Ch.S., i soavt. Кlinicheskiye rekomendatsii ро diagnostikе i lecheniyu nealkogol’noy zhirovoy bolezni pecheni Rossiiskogo obschestva po izucheniyu pecheni I Rossiiskoi gastroenterologicheskoiassociathii. Rossiyskiy zhurnal gastroenterologii, gepatologii, koloproktologii. 2016, no.2, pp. 24-42.
      4. Erymina E. Yu., Zvereva S. I., Morozova N. T. et al. Alkogol’naya bolezn’ pecheni. Sovremennye vozmozhnosti patogeneticheskoi medikamentoznoi terapii. Меditsinsky alfavit. Prakticheskaya gastroenterologiya. 2018, vol.1, no.7, pp. 54-61.
      5. Tarasova L. V., Diomidova V. N., Tsyganova Yu. V. Secretted frizzled related protein-4 kak odin is pokazatelei differentsial’noi diagnostiki zhirovih boleznei pecheni. Vestnik SurGU. Меdicina. 2019, no.1(39), pp. 73-33.
      6. Ermolova T. V., Ermolov S. Yu., Belyaeva E. L. Nealkogol’naya zhirovaya bolezn’ pecheni: sovremennyy vzglyad na problem. Effectivnaya farmakoterapiya. Gastroenterologiya. 2015, no.3, pp. 24-34.
      7. Mekhtiyev S. N. Sovremennyy vzglyad na perspektivy terapii nealkogol’noy bolezni pecheni. Effektivnaya farmakoterapiya. 2011, no.2, pp. 50.
      8. Ivashkin V. T., Drapkina O. M., Mayev I. V. i soavt. Rasprostranennost’ nealkogol’noy zhirovoy bolezni pecheni u patsiyentov ambulatorno-poliklinicheskoy praktiki v Rossiyskoy Federatsii: rezul’taty issledovaniya DIREG 2. Rossiyskiy zhurnal gastroenterologii, gepatologii, koloproktologii. 2015, no.6, pp.31-41.
      9. Zheludochno-kishechnyi trakt i ozhirenie u detei. Pod red. V.P Novikovoi, M. M. Gurovoi. SPb, SpetsLit, 2016, 302 p.
      10. Fedulaev Yu. N., Akhmatova F. D. Nealkogol’naya zhirovaya bolezn’ pecheni v obshei vrachebnoi praktike. Меditsinsky alfavit. Sovremennaya poliklinika. 2018, vol. 1, no.12 (349), pp. 19-25.
      11. Minushkin O. N. Preperaty ursodesoksiholevoi kisloty v klinicheskoi praktike (metodicheskiye posobie). Мoscow, 2016, 48 p.
      12. Tarasova L. V., Вusalaeva E. I., Illarionova C. V. Nealkogol’naya zhirovaya bolezn’ pecheni u molodoi patsientki (klinicheskyi sluchai) [Electronnii resurs с]. Acta medica Eurasica. 2017, no.2, pp. 30-34. - URL: http://acta-medica-eurasica.ru/single/2017/2/6.
      13. Banerjee R, Pavlides M, Tunnicliffe EM, et al. Multiparametricmagnetic resonance for the non-invasive diagnosis of liver disease. J. Hepatol. 2014, no.60, pp. 69-77.
     


    Full text is published :
    Tarasova L. V., Busalaeva E. I., Tsyganova J. V., Diomidova V. N. Clinical case of non-alcoholic fatty liver disease successful treatment in a young patient. Experimental and Clinical Gastroenterology. 2019;164(4): 131–135. (In Russ.) DOI: 10.31146/1682-8658-ecg-164-4-131-135
    Read & Download full text