Preview

Онкогематология

Расширенный поиск

Роль микробиома в патогенезе инфекционного и иммунологического повреждения желудочно-кишечного тракта у детей с онкогематологическими заболеваниями

https://doi.org/10.17650/1818-8346-2021-16-2-86-93

Аннотация

Инфекционные осложнения остаются одной из наиболее значимых проблем, ассоциированных с противоопухолевой терапией у онкологических пациентов. В результате проводимой цитотоксической, лучевой, а также антибактериальной терапии возникают дисбиоз и повреждение слизистого барьера желудочно-кишечного тракта. Эти изменения приводят к развитию мукозита, тем самым повышая риск эндогенной транслокации микрофлоры с последующим вероятным развитием тяжелых инфекционно-воспалительных  заболеваний. Кроме этого, существующие на сегодняшний день данные свидетельствуют о наличии взаимосвязи между нарушениями в кишечном микробиоме и развитием посттрансплантационной реакции «трансплантат против хозяина». В статье приведены существующие парадигмы в отношении определения роли функционального состояния желудочно-кишечного тракта у онкологических пациентов в целях оптимизации подходов к профилактике и противомикробному лечению.

Об авторах

Д. А. Моргачева
ФГБУ «Национальный медицинский исследовательский центр им. В.А. Алмазова» Минздрава России
Россия

Дарья Андреевна Моргачева

197341 Санкт-Петербург, ул. Аккуратова, 2

 



Ю. В. Диникина
ФГБУ «Национальный медицинский исследовательский центр им. В.А. Алмазова» Минздрава России
Россия

197341 Санкт-Петербург, ул. Аккуратова, 2



Ю. К. Тошина
ФГБУ «Национальный медицинский исследовательский центр им. В.А. Алмазова» Минздрава России
Россия

197341 Санкт-Петербург, ул. Аккуратова, 2



М. Б. Белогурова
ФГБУ «Национальный медицинский исследовательский центр им. В.А. Алмазова» Минздрава России
Россия

197341 Санкт-Петербург, ул. Аккуратова, 2



Список литературы

1. Van Vliet M.J., Tissing W.J., Dun C.A. et al. Chemotherapy treatment in pediatric patients with acute myeloid leukemia receiving antimicrobial prophylaxis leads to a relative increase of colonization with potentially pathogenic bacteria in the gut. Clin Infect Dis 2009;49(2):262–70. DOI: 10.1086/599346.

2. Kelly D.L., Lyon D.E., Yoon S.L., Horgas A.L. The Microbiome and cancer: implications for oncology nursing science. Cancer Nurs 2016;39(3):E56–62. DOI: 10.1097/NCC.0000000000000286.

3. Malard F., Gasc C., Plantamura E. et al. High gastrointestinal microbial diversity and clinical outcome in graft-versus- host disease patients. Bone Marrow Transplant 2018;53(12):1493–7. DOI: 10.1038/s41409-018-0254-x.

4. Tanaka M., Nakayama J. Development of the gut microbiota in infancy and its impact on health in later life. Allergol Int 2017;66(4):515–22. DOI: 10.1016/j.alit.2017.07.010.

5. Okada H., Kuhn C., Feillet H., Bach J.F. The “hygiene hypothesis” for autoimmune and allergic diseases: an update. Clin Exp Immunol 2010;160(1):1–9. DOI: 10.1111/j.1365–2249.2010.04139.x.

6. Alexander S., Pole J.D., Gibson P. et al. Classification of treatment-related mortality in children with cancer: a systematic assessment. Lancet Oncol 2015;16(16):e604–10. DOI: 10.1016/S1470–2045(15)00197–7.

7. Gibson P., Pole J.D., Lazor T. et al. Treatment-related mortality in newly diagnosed pediatric cancer: a population-based analysis. Cancer Med 2018;7(3):707–15. DOI: 10.1002/cam4.1362.

8. Van der Velden W.J., Herbers A.H., Netea M.G., Blijlevens N.M. Mucosal barrier injury, fever and infection in neutropenic patients with cancer: introducing the paradigm febrile mucositis. Br J Haematol 2014;167(4):441–52. DOI: 10.1111/bjh.13113.

9. Clarke R.T., Jenyon T., van Hamel Parsons V., King A.J. Neutropenic sepsis: management and complications. Clin Med 2013;13(2):185–7. DOI: 10.7861/clinmedicine.13-2-185.

10. Wright J.D., Neugut A.I., Ananth C.V. et al. Deviations from guideline-based therapy for febrile neutropenia in cancer patients and their effect on outcomes. JAMA Intern Med 2013;173(7):559–68. DOI: 10.1001/jamainternmed.2013.2921.

11. Taur Y., Xavier J.B., Lipuma L. et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis 2012;55(7):905–14. DOI: 10.1093/cid/cis580.

12. Biagi E., Zama D., Nastasi C. et al. Gut microbiota trajectory in pediatric patients undergoing hematopoietic SCT. Bone Marrow Transplant 2015;50(7): 992–8. DOI: 10.1038/bmt.2015.16.

13. Taur Y., Pamer E.G. Microbiome mediation of infections in the cancer setting. Genome Med 2016;8(1):40. DOI: 10.1186/s13073-016-0306-z.

14. Petersen C., Round J.L. Defining dysbiosis and its influence on host immunity and disease. Cell Microbiol 2014;16(7):1024–33. DOI: 10.1111/cmi.12308.

15. Miller M.M., Donald D.V., Hagemann T.M. Prevention and treatment of oral mucositis in children with cancer. J Pediatr Pharmacol Ther 2012;17(4): 340–50. DOI: 10.5863/1551-6776-17.4.340.

16. Sonis S.T., Oster G., Fuchs H. et al. Oral mucositis and the clinical and economic outcomes of hematopoietic stem-cell transplantation. J Clin Oncol 2001;19(8):2201–5. DOI: 10.1200/JCO.2001.19.8.2201.

17. Sonis S.T. The pathobiology of mucositis. Nat Rev Cancer 2004;4(4):277–84. DOI: 10.1038/nrc1318.

18. Tsuji E., Hiki N., Nomura S. et al. Simultaneous onset of acute inflammatory response, sepsis-like symptoms and intestinal mucosal injury after cancer chemotherapy. Int J Cancer 2003;107(2):303–8. DOI: 10.1002/ijc.11196.

19. Blijlevens N.M., van’t Land B., Donnelly J.P. et al. Measuring mucosal damage induced by cytotoxic therapy. Support Care Cancer 2004;12(4):227–33. DOI: 10.1007/s00520-003-0572-3.

20. Herbers A.H., de Haan A.F., van der Velden W.J. et al. Mucositis not neutropenia determines bacteremia among hematopoietic stem cell transplant recipients. Transpl Infect Dis 2014;16(2):279–85. DOI: 10.1111/tid.12195.

21. Van der Velden W.J., Herbers A.H., Feuth T. et al. Intestinal damage determines the inflammatory response and early complications in patients receiving conditioning for a stem cell transplantation. PLoS One 2010;5(12): e15156. DOI: 10.1371/journal.pone.0015156.

22. Nagalingam N.A., Lynch S.V. Role of the microbiota in inflammatory bowel diseases. Inflamm Bowel Dis 2012;18(5):968–84. DOI: 10.1002/ibd.21866.

23. Chai L.Y., de Boer M.G., van der Velden W.J. et al. The Y238X stop codon polymorphism in the human β-glucan receptor dectin‑1 and susceptibility to invasive aspergillosis. J Infect Dis 2011;203(5):736–43. DOI: 10.1093/infdis/jiq102.

24. Blijlevens N.M.A., Logan R.M., Netea M.G. Mucositis: from febrile neutropenia to febrile mucositis. J Antimicrob Chemother 2009;63(suppl_1):i36–40. DOI: 10.1093/jac/dkp081.

25. Алямкина Е.А., Долгова Е.В., Проскурина А.С. и др. Внутриклеточные системы обнаружения экзогенных нуклеиновых кислот и механизмы запуска иммунных реакций в ответ на интернализацию экзогенной ДНК. Медицинская иммунология 2013;15(5):413–30.

26. Palmblad J., Papadaki H.A. Chronic idiopathic neutropenias and severe congenital neutropenia. Curr Opin Hematol 2008;15(1):8–14. DOI: 10.1097/MOH.0b013e3282f172d3.

27. Vinolo M.A., Rodrigues H.G., Nachbar R.T., Curi R. Regulation of inflammation by short chain fatty acids. Nutrients 2011;3(10):858–76. DOI: 10.3390/nu3100858.

28. Mathewson N.D., Jenq R., Mathew A.V. et al. Gut microbiome-derived metabolites modulate intestinal epithelial cell damage and mitigate graft-versus-host disease. Nat Immunol 2016;17(5):505–13. DOI: 10.1038/ni.3400.

29. Choi S.W., Gatza E., Hou G. et al. Histone deacetylase inhibition regulates inflammation and enhances Tregs after allogeneic hematopoietic cell transplantation in humans. Blood 2015;125(5):815–9. DOI: 10.1182/ blood-2014-10-605238.

30. Taur Y., Jenq R.R., Perales M.A. et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood 2014;124(7):1174–82. DOI: 10.1182/blood‑2014-02-554725.

31. Jenq R.R., Taur Y., Devlin S.M. et al. Intestinal blautia is associated with reduced death from graft-versus- host disease. Biol Blood Marrow Transplant 2015;21(8):1373–83. DOI: 10.1016/j.bbmt.2015.04.016.

32. Sax H.C., Illig K.A., Ryan C.K., Hardy D. J. Low-dose enteral feeding is beneficial during total parenteral nutrition. Am J Surg 1996;171(6):587–90. DOI: 10.1016/s0002-9610(96)00039-6.

33. Chen Y., Zhao Y., Cheng Q. et al. The role of intestinal microbiota in acute graft-versus- host disease. J Immunol Res 2015;2015:145859. DOI: 10.1155/2015/145859.

34. Evans J.C., Hirani S.P., Needle J.J. Nutritional and post-transplantation outcomes of enteral versus parenteral nutrition in pediatric hematopoietic stem cell transplantation: a systematic review of randomized and nonrandomized studies. Biol Blood Marrow Transplant 2019;25(8):e252–9. DOI: 10.1016/j.bbmt.2019.02.023.

35. Heubi J.E. Whenever possible, use the gut! J Pediatr Hematol Oncol 1999;21(2): 88–90. DOI: 10.1097/00043426-199903000-00003.

36. Miedema K., te Poele E., Tissing W. et al. Association of polymorphisms in the TLR4 gene with the risk of developing neutropenia in children with leukemia. Leukemia 2011;25(6): 995–1000. DOI: 10.1038/leu.2011.27

37. Holle E., Rogler G., Herfarth H. et al. Both donor and recipient NOD2/ CARD15 mutations associate with transplant-related mortality and GvHD following allogeneic stem cell transplantation. Blood 2004;104(3): 889–94. DOI: 10.1182/blood-2003-10-3543.

38. Cunha C., Di Ianni M., Bozza S. et al. Dectin‑1 Y238X polymorphism associates with susceptibility to invasive aspergillosis in hematopoietic transplantation through impairment of both recipient- and donor-dependent mechanisms of antifungal immunity. Blood 2010;116(24):5394–402. DOI: 10.1182/blood‑2010-04-279307.

39. Мазурок В.А., Головкин А.С., Горелов И.И. и др. Интестинальная оксигенотерапия критических состояний. Общая реаниматология 2017;13(6):74–91.

40. Herbers A., van der Velden W., de Haan A. et al. Impact of palifermin on intestinal mucositis of HSCT recipients after BEAM. Bone Marrow Transplant 2014;49(1):8–10. DOI: 10.1038/bmt.2013.118.

41. Blijlevens N.M., Donnelly J.P., DePauw B.E. Inflammatory response to mucosal barrier injury after myeloablative therapy in allogeneic stem cell transplant recipients. Bone Marrow Transplant 2005;36(8):703–7. DOI: 10.1038/sj.bmt.1705118.

42. Noor F., Kaysen A., Wilmes P., Schneider J.G. The gut microbiota and hematopoietic stem cell transplantation: challenges and potentials. J Innate Immun 2019;11(5):405–15. DOI: 10.1159/000492943.

43. Panek M., Čipčić Paljetak H., Barešić A. et al. Methodology challenges in studying human gut microbiota – effects of collection, storage, DNA extraction and next generation sequencing technologies. Sci Rep 2018;8:5143. DOI: 10.1038/s41598-018-23296-4.

44. Gibson R.J., Keefe D.M., Lalla R.V. et al. Systematic review of agents for the management of gastrointestinal mucositis in cancer patients. Support Care Cancer 2013;21(1):313–26. DOI: 10.1007/s00520-012-1644-z.

45. Kau A.L., Ahern P.P., Griffin N.W. et al. Human nutrition, the gut microbiome and the immune system. Nature 2011;474(7351):327–36. DOI: 10.1038/nature10213.

46. Bhanja P., Saha S., Kabarriti R. et al. Protective role of R-spondin1, an intestinal stem cell growth factor, against radiation-induced gastrointestinal syndrome in mice. PLoS One 2009;4(11): e8014. DOI: 10.1371/journal.pone.0008014.

47. Fort M.M., Mozaffarian A., Stöver A.G. et al. A synthetic TLR4 antagonist has anti-inflammatory effects in two murine models of inflammatory bowel disease. J Immunol 2005;174(10):6416–23. DOI: 10.4049/jimmunol.174.10.6416.

48. Mookherjee N., Hancock R.E. Cationic host defence peptides: innate immune regulatory peptides as a novel approach for treating infections. Cell Mol Life Sci 2007;64(7–8):922–33. DOI: 10.1007/s00018-007-6475-6.

49. Ellis M., Zwaan F., Hedström U. et al. Recombinant human interleukin 11 and bacterial infection in patients with [correction of] haematological malignant disease undergoing chemotherapy: a double-blind placebo-controlled randomised trial. Lancet 2003;361(9354):275–80. DOI: 10.1016/s0140-6736(03)12322-7.

50. Bachanova V., Brunstein C.G., Burns L.J. et al. Fewer infections and lower infection-related mortality following non-myeloablative versus myeloablative conditioning for allotransplantation of patients with lymphoma. Bone Marrow Transplant 2009;43(3):237–44. DOI: 10.1038/bmt.2008.313.

51. Ren Z.G., Liu H., Jiang J.W. et al. Protective effect of probiotics on intestinal barrier function in malnourished rats after liver transplantation. Hepatobiliary Pancreat Dis Int 2011;10(5):489–96. DOI: 10.1016/s1499-3872(11)60083-0.

52. Yan F., Cao H., Cover T.L. et al. Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology 2007;132(2):562–75. DOI: 10.1053/j.gastro.2006.11.022.

53. Jones S.E., Versalovic J. Probiotic Lactobacillus reuteri biofilms produce antimicrobial and anti-inflammatory factors. BMC Microbiol 2009;9:35. DOI: 10.1186/1471-2180-9-35.

54. Mehta A., Rangarajan S., Borate U. A cautionary tale for probiotic use in hematopoietic SCT patients-Lactobacillus acidophilus sepsis in a patient with mantle cell lymphoma undergoing hematopoietic SCT. Bone Marrow Transplant 2013;48(3):461–2. DOI: 10.1038/bmt.2012.153.

55. Reyna-Figueroa J., Barrón-Calvillo E., García-Parra C. et al. Probiotic supplementation decreases chemotherapy-induced gastrointestinal side effects in patients with acute leukemia. J Pediatr Hematol Oncol 2019;41(6):468–72. DOI: 10.1097/MPH.0000000000001497.

56. Ladas E.J., Bhatia M., Chen L. et al. The safety and feasibility of probiotics in children and adolescents undergoing hematopoietic cell transplantation. Bone Marrow Transplant 2016;51(2): 262–6. DOI: 10.1038/bmt.2015.275.

57. Song Y., Garg S., Girotra M. et al. Microbiota dynamics in patients treated with fecal microbiota transplantation for recurrent Clostridium difficile infection. PLoS One 2013;8(11):e81330. DOI: 10.1371/journal.pone.0081330.

58. Kakihana K., Fujioka Y., Suda W. et al. Fecal microbiota transplantation for patients with steroid- resistant acute graft-versus-host disease of the gut. Blood 2016;128(16):2083–8. DOI: 10.1182/blood‑2016-05-717652.

59. Bilinski J., Grzesiowski P., Sorensen N. et al. Fecal microbiota transplantation in patients with blood disorders inhibits gut colonization with antibiotic-resistant bacteria: results of a prospective, single-center study. Clin Infect Dis 017;65(3): 364–70. DOI: 10.1093/cid/cix252.

60. Taur Y., Coyte K., Schluter J. et al. Reconstitution of the gut microbiota of antibiotic-treated patients by autologous fecal microbiota transplant. Sci Transl Med 2018;10(460):eaap9489. DOI: 10.1126/scitranslmed.aap9489.


Рецензия

Для цитирования:


Моргачева Д.А., Диникина Ю.В., Тошина Ю.К., Белогурова М.Б. Роль микробиома в патогенезе инфекционного и иммунологического повреждения желудочно-кишечного тракта у детей с онкогематологическими заболеваниями. Онкогематология. 2021;16(2):86-93. https://doi.org/10.17650/1818-8346-2021-16-2-86-93

For citation:


Morgacheva D.A., Dinikina Yu.V., Toshina Yu.K., Belogurova M.B. The microbiome role in pathogenesis of inflammatory and immune alterations of gastrointestinal tract in pediatric patients with cancer. Oncohematology. 2021;16(2):86-93. (In Russ.) https://doi.org/10.17650/1818-8346-2021-16-2-86-93

Просмотров: 9221


Creative Commons License
Контент доступен под лицензией Creative Commons Attribution 4.0 License.


ISSN 1818-8346 (Print)
ISSN 2413-4023 (Online)