Таргетная терапия антибиотик-ассоциированной диареи у детей
Б.М. Блохин
А.Д. Прохорова
Т.Х. Мирзоев

Ключевые слова

дисбиоз кишечника
антибиотик-ассоциированная диарея
пробиотики в детской практике
intestinal dysbiosis
antibiotic-associated diarrhea
probiotics in pediatric practice

Как цитировать

Блохин Б., Прохорова А., Мирзоев Т. Таргетная терапия антибиотик-ассоциированной диареи у детей // Кремлевская медицина. Клинический вестник. 2016. Т. № 3. С. 98-102.

Аннотация

В условиях постоянного развития возможностей антибактериальной терапии, увеличения спектраантибактериальных препаратов, расширения возможностей их применения проблема дисбиоза кишечника скаждым годом становится все более актуальной. Изменение состава микрофлоры кишечника становится причинойаутоинтоксикации, нарушения процессов всасывания, регенерации, способствует хронизации заболеванийжелудочно-кишечного тракта. В России примерно 90% населения страдают от тех или иных проявлений дисбиоза,зачастую связанных с последствиями применения антибиотиков. Антибиотик-ассоциированная диарея (ААД) является одним из нежелательных побочных эффектовантибактериальной терапии. Это состояние развивается либо во время приема антибиотика, или в течение восьминедель после того, как прием антибиотика был прекращен. Дети раннего возраста, получающие антибиотикиширокого спектра действия (пенициллины, цефалоспорины и др.) относятся к группе риска развития антибиотикассоциированнойдиареи. С целью коррекции нежелательных последствий антибактериальной терапии успешно могут применятьсяпробиотические препараты. Доказана эффективность терапии пробиотиками при таких патологических состояниях, какантибиотик-ассоциированная и С. difficile-аccoциированная диарея. Эти препараты являются не только эффективными,но и обладают высоким профилем безопасности, что немаловажно в детской практике.At the utero stage, the fetal gastrointestinal tract is sterile. During the labours a new born child is colonizing its digestivetract through its mouth when passing by the mother's birth tract. E.coli and streptococci can be found in the digestive tract ina few hours after birth, and they spread from the mouth to the anus. Different strains ofbifidobacteria and bacteroides appearin the digestive tract 10 days after birth. Intestinal dysbiosis (ID) is a bacteriological concept characterizing the misbalancebetween the qualitative content of bacteria and their quantitative ratio. ID is always secondary and is mediated by theprimary disease. Hence, it explains the absence of such diagnosis as "dysbiosis" or "intestinal dysbiosis" in the InternationalClassification of Human Diseases (ICD-10) which has been adopted in our country as well as throughout the world. One of themajor causes of dysbiosis is antibioticotherapy. The antibiotic-associated diarrhea (AAD) is defined as diarrhea caused by withantibiotic effects either during its administration or within eight weeks after this therapy. AAD risk factors in children are early age ( 1- 2 years) and antibiotics of broad-spectrum (penicillins, cephalosporins,and others.). Thus, prescription of probiotics for the treatment and prophylactics of antibiotic-associated and C. difficile diarrhea is a logical step if to bear in mind the pathogenesis of this condition.

Литература

1. Hall M.A., Cole С.В., Smith S.L., Fuller R., Rolles CJ.
Factors influencing the presence of faecal lactobacilli in early
infancy. Arch. Dis. Child. 1990: 65: 185-188.
2. Braun O.H. Effect of consumption of human milk and other
formulas on intesti-nal bacterial flora in infants. In: Lebenthal B.
ed. Gasfroenterology and nutrition In infancy. New York: Raven
Press, 1981: 247-251.
3. Senna V., Sawada К., Mitsuoka Т. The intestinal
microtlora of infants: composi-tion of fecal flora in breast-led and
bottle-fed infants. Microbiol. Immunol. 1984; 28: 975-986.
4. Plummer S., Weawer M., Dee P., Hunter J. Clostridium
difficile pilot study: effects of probiotic supplementation on the
incidence of Cdifficile diarrhea. Int. Microbiol. 2004; 7(1): 5962.
5. Международный классификатор заболеваний человека
(МКБ-10). М., 1997.
6. Mukherjee P.K., Sendid B., Hoarau G. et al. Mycobiota in
gastrointestinal diseases. Nat. Rev. Gastroenterol. Hepatol. 2015;
12(2): 77-87.13.
7. Beyer G., Heimer-Bau M. et al. Impact of Moxifloxacin
versus Claritromycin on normal oropharyngeal microflora. Eur. J.
Clin. Microbiol. Inf. Dis. 2000; 7: 548–550.
8. World Health Organization. WHO definition of diarrhea.
Accessed on November 13. 2015.
9. McFarland L.V. Epidemiology, risk factors and treatments
for antibiotic-associated diarrhea. Dig. Dis. 1998 Sep-Oct; 16(5):
292-307.
10. McFarland L.V. Epidemiology, risk factors and treatments
for antibiotic-associated diarrhea. Dig. Dis. 1998 Sep-Oct; 16(5):
292-307.
11. Turck D., Bernet J.P., Marx J. et al. Incidence and risk
factors of oral antibiotic-associated diarrhea in an outpatient
pediatric population. J. Pediatr. Gastroenterol. Nutr. 2003 Jul;
37(1): 22-26.
12. Szajewska H. et al. Probiotics in the prevention of
antibiotic-associated diarrhea in children: a meta-analysis of
randomized controlled trials. J. Pediatr. 2006; 149: 367–372.
13. Alam S., Mushtaq M. Antibiotic associated diarrhea in
children. Indian Pediatr. 2009 Jun; 46(6): 491-496.
14. Jaimes E.C. Lincocinamides and the incidence of
antibiotic-associated colitis. Clin Ther 1991; 13: 270.
15. Lynne Vernice McFarland, Metehan Ozen, Ener Cagri
Dinleyici, Shan Goh Comparison of pediatric and adult antibioticassociated
diarrhea and Clostridium difficile infections. World
J.

Gastroenterol.
2016 Mar 21; 22(11): 3078–3104.
16. Mylonaki M., Langmead L., Pantes A. et al. Enteric
infection in relapse of inflammatory bowel disease: importance
of microbiological examination of stool. Eur. J. Gastroenterol.
Hepatol. 2004 Aug; 16(8): 775-778.
17. Cohen S.H., Gerding D.N., Johnson S. et al. Society for
Healthcare Epidemiology of America, Infectious Diseases Society
of America. Clinical practice guidelines for Clostridium difficile
infection in adults: 2010 update by the society for healthcare
epidemiology of America (SHEA) and the infectious diseases
society of America (IDSA). Infect. Control. Hosp. Epidemiol.
2010 May; 31(5): 431-455.
18. Rupnik M., Wilcox M.H., Gerding D.N. Clostridium
difficile infection: new developments in epidemiology and
pathogenesis.Nat. Rev. Microbiol. 2009 Jul; 7(7): 526-536
19. Silva Júnior M. Recent changes in Clostridium difficile
infection. Einstein (Sao Paulo). 2012; 10: 105–109.
20. Oake N., Taljaard M., van Walraven C. et al. The effect
of hospital-acquired Clostridium difficile infection on in-hospital
mortality. Arch. Intern. Med. 2010 Nov 8; 170(20): 1804-1810.
21. McFarland L.V., Brandmarker S.A., Guandalini S.
Pediatric Clostridium difficile: a phantom menace or clinical
reality? J. Pediatr. Gastroenterol. Nutr. 2000 Sep; 31(3): 220231.
22. Jangi S., Lamont J.T. Asymptomatic colonization by
Clostridium difficile in infants: implications for disease in later
life. J. Pediatr. Gastroenterol. Nutr. 2010 Jul; 51(1): 2-7.
23. Sathyendran V., McAuliffe G.N., Swager T. et al.
Clostridium difficile as a cause of healthcare-associated
diarrhoea among children in Auckland, New Zealand: clinical
and molecular epidemiology. Eur. J. Clin. Microbiol. Infect. Dis.
2014 Oct; 33(10): 1741-1747.
24. Sammons J.S., Localio R., Xiao R. et al. Clostridium
difficile infection is associated with increased risk of death and
prolonged hospitalization in children. Clin. Infect. Dis. 2013 Jul;
57(1): 1-8.
25. Banerjee P., Merkel G.J., Bhunia A.K. Lactobacillus
delbrueckii ssp. bulgaricus B-30892 can inhibit cytotoxic effects
and adhesion of pathogenic Clostridium difficile to Caco-2 cells.
Gut. Pathog. 2009 Apr 27; 1(1): 8.
26. Kondepudi K.K., Ambalam P., Karagin P.H. et al. A novel
multi-strain probiotic and synbiotic supplement for prevention
of Clostridium difficile infection in a murine model. Microbiol.
Immunol. 2014 Oct; 58(10): 552-558.
27. Klaenhammer T.R.. Bacteriocins of lactic acid bacteria.
Biochimie. 1988 Mar; 70(3): 337-349.
28. Nemcová R. Criteria for selection of lactobacilli for
probiotic use. Vet. Med. (Praha). 1997 Jan; 42(1): 19-27.
29. Venturi A., Gionchetti P., Rizzello F. et al. Impact on the
composition of the faecal flora by a new probiotic preparation:
preliminary data on maintenance treatment of patients with
ulcerative colitis. Aliment. Pharmacol. Ther. 1999 Aug; 13(8):
1103-1108.
30. Grosse C., Scherer J., Koch D. et al. A new ferrous ironuptake
transporter,
EfeU (YcdN),
from Escherichia coli. Mol

Microbiol.
2006 Oct; 62(1): 120-131.
31. Zhang Z., Hinrichs D.J., Lu H. et al. After interleukin12p40,
are interleukin-23 and interleukin-17 the next therapeutic

targets
for inflammatory bowel disease? Int. Immunopharmacol.

2007
Apr; 7(4): 409-416.
32. Trejo F.M., De Antoni G.L., Pérez P.F. Protective effect
of bifidobacteria in an experimental model of Clostridium difficile
associated colitis. J. Dairy. Res. 2013 Aug; 80(3): 263-269.
33. Advice for travelers.Treat Guidel Med Lett. 2004 May;
2(21): 33-40.
34. Захарова И.Н., Мумладзе Э.Б., Свинцицкая В.И.,
Кучерова В.В., Дмитриева Ю.А. Пробиотики в профилактике
антибиотик-ассоциированных диарей у детей. Журнал
практическая медицина. 2009; 7 (39).
35. Szajewska, Hania; Canani, Roberto Berni; Guarino,
Alfredo et al. on Behalf of the ESPGHAN Working Group
for ProbioticsPrebiotics Probiotics for the Prevention of
Antibiotic-Associated Diarrhea in Children Journal of Pediatric
Gastroenterology & Nutrition: March 2016 - Volume 62 - Issue
3: 495–506.
36. McFarland L.V. Antibiotic-associated diarrhea:
epidemiology, trends and treatment. Future Microbiol. 2008 Oct;
3(5): 563-578.