Skip to main content
Top
Published in: Annals of Intensive Care 1/2022

Open Access 01-12-2022 | Probiotics | Review

The role of the microbiota in the management of intensive care patients

Authors: Piotr Szychowiak, Khanh Villageois-Tran, Juliette Patrier, Jean-François Timsit, Étienne Ruppé

Published in: Annals of Intensive Care | Issue 1/2022

Login to get access

Abstract

The composition of the gut microbiota is highly dynamic and changes according to various conditions. The gut microbiota mainly includes difficult-to-cultivate anaerobic bacteria, hence knowledge about its composition has significantly arisen from culture-independent methods based on next-generation sequencing (NGS) such as 16S profiling and shotgun metagenomics. The gut microbiota of patients hospitalized in intensive care units (ICU) undergoes many alterations because of critical illness, antibiotics, and other ICU-specific medications. It is then characterized by lower richness and diversity, and dominated by opportunistic pathogens such as Clostridioides difficile and multidrug-resistant bacteria. These alterations are associated with an increased risk of infectious complications or death. Specifically, at the time of writing, it appears possible to identify distinct microbiota patterns associated with severity or infectivity in COVID-19 patients, paving the way for the potential use of dysbiosis markers to predict patient outcomes. Correcting the microbiota disturbances to avoid their consequences is now possible. Fecal microbiota transplantation is recommended in recurrent C. difficile infections and microbiota-protecting treatments such as antibiotic inactivators are currently being developed. The growing interest in the microbiota and microbiota-associated therapies suggests that the control of the dysbiosis could be a key factor in the management of critically ill patients. The present narrative review aims to provide a synthetic overview of microbiota, from healthy individuals to critically ill patients. After an introduction to the different techniques used for studying the microbiota, we review the determinants involved in the alteration of the microbiota in ICU patients and the latter’s consequences. Last, we assess the means to prevent or correct microbiota alteration.
Literature
2.
go back to reference Lankelma JM, van Vught LA, Belzer C, Schultz MJ, van der Poll T, de Vos WM, et al. Critically ill patients demonstrate large interpersonal variation in intestinal microbiota dysregulation: a pilot study. Intensive Care Med. 2017;43:59–68.PubMedCrossRef Lankelma JM, van Vught LA, Belzer C, Schultz MJ, van der Poll T, de Vos WM, et al. Critically ill patients demonstrate large interpersonal variation in intestinal microbiota dysregulation: a pilot study. Intensive Care Med. 2017;43:59–68.PubMedCrossRef
5.
go back to reference Lagier J-C, Khelaifia S, Alou MT, Ndongo S, Dione N, Hugon P, et al. Culture of previously uncultured members of the human gut microbiota by culturomics. Nat Microbiol. 2016;1:16203.PubMedCrossRef Lagier J-C, Khelaifia S, Alou MT, Ndongo S, Dione N, Hugon P, et al. Culture of previously uncultured members of the human gut microbiota by culturomics. Nat Microbiol. 2016;1:16203.PubMedCrossRef
6.
go back to reference Scholz M, Ward DV, Pasolli E, Tolio T, Zolfo M, Asnicar F, et al. Strain-level microbial epidemiology and population genomics from shotgun metagenomics. Nat Methods. 2016;13:435–8.PubMedCrossRef Scholz M, Ward DV, Pasolli E, Tolio T, Zolfo M, Asnicar F, et al. Strain-level microbial epidemiology and population genomics from shotgun metagenomics. Nat Methods. 2016;13:435–8.PubMedCrossRef
7.
go back to reference Munk P, Knudsen BE, Lukjancenko O, Duarte ASR, Van Gompel L, Luiken REC, et al. Abundance and diversity of the faecal resistome in slaughter pigs and broilers in nine European countries. Nat Microbiol. 2018;3:898–908.PubMedCrossRef Munk P, Knudsen BE, Lukjancenko O, Duarte ASR, Van Gompel L, Luiken REC, et al. Abundance and diversity of the faecal resistome in slaughter pigs and broilers in nine European countries. Nat Microbiol. 2018;3:898–908.PubMedCrossRef
8.
go back to reference Lagier J-C, Armougom F, Million M, Hugon P, Pagnier I, Robert C, et al. Microbial culturomics: paradigm shift in the human gut microbiome study. Clin Microbiol Infect Off Publ Eur Soc Clin Microbiol Infect Dis. 2012;18:1185–93. Lagier J-C, Armougom F, Million M, Hugon P, Pagnier I, Robert C, et al. Microbial culturomics: paradigm shift in the human gut microbiome study. Clin Microbiol Infect Off Publ Eur Soc Clin Microbiol Infect Dis. 2012;18:1185–93.
10.
go back to reference Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, Chinwalla AT, et al. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–14.CrossRef Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, Chinwalla AT, et al. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–14.CrossRef
11.
go back to reference Lloyd-Price J, Mahurkar A, Rahnavard G, Crabtree J, Orvis J, Hall AB, et al. Strains, functions and dynamics in the expanded Human Microbiome Project. Nature. 2017;550:61–6.PubMedPubMedCentralCrossRef Lloyd-Price J, Mahurkar A, Rahnavard G, Crabtree J, Orvis J, Hall AB, et al. Strains, functions and dynamics in the expanded Human Microbiome Project. Nature. 2017;550:61–6.PubMedPubMedCentralCrossRef
12.
go back to reference The Integrative HMP. (iHMP) Research Network Consortium The Integrative Human Microbiome Project. Nature. 2019;569:641–8.CrossRef The Integrative HMP. (iHMP) Research Network Consortium The Integrative Human Microbiome Project. Nature. 2019;569:641–8.CrossRef
13.
go back to reference Almeida A, Nayfach S, Boland M, Strozzi F, Beracochea M, Shi ZJ, et al. A unified catalog of 204,938 reference genomes from the human gut microbiome. Nat Biotechnol. 2021;39:105–14.PubMedCrossRef Almeida A, Nayfach S, Boland M, Strozzi F, Beracochea M, Shi ZJ, et al. A unified catalog of 204,938 reference genomes from the human gut microbiome. Nat Biotechnol. 2021;39:105–14.PubMedCrossRef
14.
15.
go back to reference Belizário JE, Napolitano M. Human microbiomes and their roles in dysbiosis, common diseases, and novel therapeutic approaches. Front Microbiol. 2015;6:1050.PubMedPubMedCentralCrossRef Belizário JE, Napolitano M. Human microbiomes and their roles in dysbiosis, common diseases, and novel therapeutic approaches. Front Microbiol. 2015;6:1050.PubMedPubMedCentralCrossRef
16.
go back to reference Li J, Jia H, Cai X, Zhong H, Feng Q, Sunagawa S, et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol. 2014;32:834–41.PubMedCrossRef Li J, Jia H, Cai X, Zhong H, Feng Q, Sunagawa S, et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol. 2014;32:834–41.PubMedCrossRef
18.
go back to reference Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016;7:189–200.PubMedPubMedCentralCrossRef Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016;7:189–200.PubMedPubMedCentralCrossRef
19.
go back to reference Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci USA. 2011;108:16050–5.PubMedPubMedCentralCrossRef Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci USA. 2011;108:16050–5.PubMedPubMedCentralCrossRef
20.
go back to reference Vincent J-L. International Study of the Prevalence and Outcomes of Infection in Intensive Care Units. JAMA. 2009;302:2323.PubMedCrossRef Vincent J-L. International Study of the Prevalence and Outcomes of Infection in Intensive Care Units. JAMA. 2009;302:2323.PubMedCrossRef
22.
go back to reference Arulkumaran N, Routledge M, Schlebusch S, Lipman J, Conway MA. Antimicrobial-associated harm in critical care: a narrative review. Intensive Care Med. 2020;46(2):225–35.PubMedPubMedCentralCrossRef Arulkumaran N, Routledge M, Schlebusch S, Lipman J, Conway MA. Antimicrobial-associated harm in critical care: a narrative review. Intensive Care Med. 2020;46(2):225–35.PubMedPubMedCentralCrossRef
23.
go back to reference Ravi A, Halstead FD, Bamford A, Casey A, Thomson NM, van Schaik W, et al. Loss of microbial diversity and pathogen domination of the gut microbiota in critically ill patients. Microb Genomics. 2019;5(9):e000293.CrossRef Ravi A, Halstead FD, Bamford A, Casey A, Thomson NM, van Schaik W, et al. Loss of microbial diversity and pathogen domination of the gut microbiota in critically ill patients. Microb Genomics. 2019;5(9):e000293.CrossRef
24.
go back to reference Jernberg C, Lofmark S, Edlund C, Jansson JK. Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology. 2010;156:3216–23.PubMedCrossRef Jernberg C, Lofmark S, Edlund C, Jansson JK. Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology. 2010;156:3216–23.PubMedCrossRef
25.
go back to reference Jernberg C, Löfmark S, Edlund C, Jansson JK. Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. ISME J. 2007;1:56–66.PubMedCrossRef Jernberg C, Löfmark S, Edlund C, Jansson JK. Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. ISME J. 2007;1:56–66.PubMedCrossRef
26.
go back to reference Rashid M-U, Zaura E, Buijs MJ, Keijser BJF, Crielaard W, Nord CE, et al. Determining the long-term effect of antibiotic administration on the human normal intestinal microbiota using culture and pyrosequencing methods. Clin Infect Dis. 2015;60:S77-84.PubMedCrossRef Rashid M-U, Zaura E, Buijs MJ, Keijser BJF, Crielaard W, Nord CE, et al. Determining the long-term effect of antibiotic administration on the human normal intestinal microbiota using culture and pyrosequencing methods. Clin Infect Dis. 2015;60:S77-84.PubMedCrossRef
27.
go back to reference Slimings C, Riley TV. Antibiotics and hospital-acquired Clostridium difficile infection: update of systematic review and meta-analysis. J Antimicrob Chemother. 2014;69:881–91.PubMedCrossRef Slimings C, Riley TV. Antibiotics and hospital-acquired Clostridium difficile infection: update of systematic review and meta-analysis. J Antimicrob Chemother. 2014;69:881–91.PubMedCrossRef
28.
go back to reference Korpela K, Salonen A, Virta LJ, Kekkonen RA, Forslund K, Bork P, et al. Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children. Nat Commun. 2016;7:10410.PubMedPubMedCentralCrossRef Korpela K, Salonen A, Virta LJ, Kekkonen RA, Forslund K, Bork P, et al. Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children. Nat Commun. 2016;7:10410.PubMedPubMedCentralCrossRef
29.
go back to reference Vrieze A, Out C, Fuentes S, Jonker L, Reuling I, Kootte RS, et al. Impact of oral vancomycin on gut microbiota, bile acid metabolism, and insulin sensitivity. J Hepatol. 2014;60:824–31.PubMedCrossRef Vrieze A, Out C, Fuentes S, Jonker L, Reuling I, Kootte RS, et al. Impact of oral vancomycin on gut microbiota, bile acid metabolism, and insulin sensitivity. J Hepatol. 2014;60:824–31.PubMedCrossRef
30.
go back to reference Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci. 2011;108:4554–61.PubMedCrossRef Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci. 2011;108:4554–61.PubMedCrossRef
31.
go back to reference Soldi S, Vasileiadis S, Uggeri F, Campanale MC, Morelli L, Calanni F, et al. Modulation of the gut microbiota composition by rifaximin in non-constipated irritable bowel syndrome patients: a molecular approach. Clin Exp Gastroenterol. 2015;309:1. Soldi S, Vasileiadis S, Uggeri F, Campanale MC, Morelli L, Calanni F, et al. Modulation of the gut microbiota composition by rifaximin in non-constipated irritable bowel syndrome patients: a molecular approach. Clin Exp Gastroenterol. 2015;309:1.
32.
go back to reference Ponziani FR, Scaldaferri F, Petito V, Paroni Sterbini F, Pecere S, Lopetuso LR, et al. The role of antibiotics in gut microbiota modulation: the eubiotic effects of rifaximin. Dig Dis. 2016;34:269–78.PubMedCrossRef Ponziani FR, Scaldaferri F, Petito V, Paroni Sterbini F, Pecere S, Lopetuso LR, et al. The role of antibiotics in gut microbiota modulation: the eubiotic effects of rifaximin. Dig Dis. 2016;34:269–78.PubMedCrossRef
33.
go back to reference Stewardson AJ, Gaïa N, François P, Malhotra-Kumar S, Delémont C, de Tejada B, et al. Collateral damage from oral ciprofloxacin versus nitrofurantoin in outpatients with urinary tract infections: a culture-free analysis of gut microbiota. Clin Microbiol Infect. 2015;21(344):e1-344. Stewardson AJ, Gaïa N, François P, Malhotra-Kumar S, Delémont C, de Tejada B, et al. Collateral damage from oral ciprofloxacin versus nitrofurantoin in outpatients with urinary tract infections: a culture-free analysis of gut microbiota. Clin Microbiol Infect. 2015;21(344):e1-344.
34.
go back to reference Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG, Gratadoux J-J, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci. 2008;105:16731–6.PubMedPubMedCentralCrossRef Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG, Gratadoux J-J, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci. 2008;105:16731–6.PubMedPubMedCentralCrossRef
35.
go back to reference Weiss E, Zahar J-R, Lesprit P, Ruppe E, Leone M, Chastre J, et al. Elaboration of a consensual definition of de-escalation allowing a ranking of β-lactams. Clin Microbiol Infect. 2015;21:649.e1-649.e10.CrossRef Weiss E, Zahar J-R, Lesprit P, Ruppe E, Leone M, Chastre J, et al. Elaboration of a consensual definition of de-escalation allowing a ranking of β-lactams. Clin Microbiol Infect. 2015;21:649.e1-649.e10.CrossRef
36.
go back to reference Woerther P-L, Micol J-B, Angebault C, Pasquier F, Pilorge S, Bourhis J-H, et al. Monitoring antibiotic-resistant enterobacteria faecal levels is helpful in predicting antibiotic susceptibility of bacteraemia isolates in patients with haematological malignancies. J Med Microbiol. 2015;64:676–81.PubMedCrossRef Woerther P-L, Micol J-B, Angebault C, Pasquier F, Pilorge S, Bourhis J-H, et al. Monitoring antibiotic-resistant enterobacteria faecal levels is helpful in predicting antibiotic susceptibility of bacteraemia isolates in patients with haematological malignancies. J Med Microbiol. 2015;64:676–81.PubMedCrossRef
37.
go back to reference Ruppé E, Burdet C, Grall N, de Lastours V, Lescure F-X, Andremont A, et al. Impact of antibiotics on the intestinal microbiota needs to be re-defined to optimize antibiotic usage. Clin Microbiol Infect. 2018;24:3–5.PubMedCrossRef Ruppé E, Burdet C, Grall N, de Lastours V, Lescure F-X, Andremont A, et al. Impact of antibiotics on the intestinal microbiota needs to be re-defined to optimize antibiotic usage. Clin Microbiol Infect. 2018;24:3–5.PubMedCrossRef
38.
39.
go back to reference Woerther P-L, Barbier F, Lepeule R, Fihman V, Ruppé É. Assessing the ecological benefit of antibiotic de-escalation strategies to elaborate evidence-based recommendations. Clin Infect Dis. 2020;71:1128–9.PubMedCrossRef Woerther P-L, Barbier F, Lepeule R, Fihman V, Ruppé É. Assessing the ecological benefit of antibiotic de-escalation strategies to elaborate evidence-based recommendations. Clin Infect Dis. 2020;71:1128–9.PubMedCrossRef
40.
go back to reference Benus RF, Harmsen HJ, Welling GW, Spanjersberg R, Zijlstra JG, Degener JE, et al. Impact of digestive and oropharyngeal decontamination on the intestinal microbiota in ICU patients. Intensive Care Med. 2010;36:1394–402.PubMedPubMedCentralCrossRef Benus RF, Harmsen HJ, Welling GW, Spanjersberg R, Zijlstra JG, Degener JE, et al. Impact of digestive and oropharyngeal decontamination on the intestinal microbiota in ICU patients. Intensive Care Med. 2010;36:1394–402.PubMedPubMedCentralCrossRef
41.
go back to reference Ruppé E, Ghozlane A, Tap J, Pons N, Alvarez A-S, Maziers N, et al. Prediction of the intestinal resistome by a three-dimensional structure-based method. Nat Microbiol. 2019;4:112–23.PubMedCrossRef Ruppé E, Ghozlane A, Tap J, Pons N, Alvarez A-S, Maziers N, et al. Prediction of the intestinal resistome by a three-dimensional structure-based method. Nat Microbiol. 2019;4:112–23.PubMedCrossRef
42.
go back to reference Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science. 2016;352:565–9.PubMedPubMedCentralCrossRef Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science. 2016;352:565–9.PubMedPubMedCentralCrossRef
43.
go back to reference Imhann F, Bonder MJ, Vich Vila A, Fu J, Mujagic Z, Vork L, et al. Proton pump inhibitors affect the gut microbiome. Gut. 2016;65:740–8.PubMedCrossRef Imhann F, Bonder MJ, Vich Vila A, Fu J, Mujagic Z, Vork L, et al. Proton pump inhibitors affect the gut microbiome. Gut. 2016;65:740–8.PubMedCrossRef
44.
go back to reference Jackson MA, Goodrich JK, Maxan M-E, Freedberg DE, Abrams JA, Poole AC, et al. Proton pump inhibitors alter the composition of the gut microbiota. Gut. 2016;65:749–56.PubMedCrossRef Jackson MA, Goodrich JK, Maxan M-E, Freedberg DE, Abrams JA, Poole AC, et al. Proton pump inhibitors alter the composition of the gut microbiota. Gut. 2016;65:749–56.PubMedCrossRef
45.
go back to reference de la Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V, Velásquez-Mejía EP, Carmona JA, Abad JM, et al. Metformin Is associated with higher relative abundance of mucin-degrading Akkermansia muciniphila and several short-chain fatty acid-producing microbiota in the Gut. Diabetes Care. 2017;40:54–62.PubMedCrossRef de la Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V, Velásquez-Mejía EP, Carmona JA, Abad JM, et al. Metformin Is associated with higher relative abundance of mucin-degrading Akkermansia muciniphila and several short-chain fatty acid-producing microbiota in the Gut. Diabetes Care. 2017;40:54–62.PubMedCrossRef
46.
go back to reference Rogers MAM, Aronoff DM. The influence of non-steroidal anti-inflammatory drugs on the gut microbiome. Clin Microbiol Infect. 2016;22:178.e1-178.e9.CrossRef Rogers MAM, Aronoff DM. The influence of non-steroidal anti-inflammatory drugs on the gut microbiome. Clin Microbiol Infect. 2016;22:178.e1-178.e9.CrossRef
47.
go back to reference Schneider SM, Le Gall P, Girard-Pipau F, Piche T, Pompei A, Nano J-L, et al. Total artificial nutrition is associated with major changes in the fecal flora. Eur J Nutr. 2000;39:248–55.PubMedCrossRef Schneider SM, Le Gall P, Girard-Pipau F, Piche T, Pompei A, Nano J-L, et al. Total artificial nutrition is associated with major changes in the fecal flora. Eur J Nutr. 2000;39:248–55.PubMedCrossRef
48.
go back to reference Lombardo L, Foti M, Ruggia O, Chiecchio A. Increased incidence of small intestinal bacterial overgrowth during proton pump inhibitor therapy. Clin Gastroenterol Hepatol. 2010;8:504–8.PubMedCrossRef Lombardo L, Foti M, Ruggia O, Chiecchio A. Increased incidence of small intestinal bacterial overgrowth during proton pump inhibitor therapy. Clin Gastroenterol Hepatol. 2010;8:504–8.PubMedCrossRef
49.
go back to reference Meng J, Banerjee S, Li D, Sindberg GM, Wang F, Ma J, et al. Opioid exacerbation of gram-positive sepsis, induced by gut microbial modulation, is rescued by IL-17A neutralization. Sci Rep. 2015;5:10918.PubMedPubMedCentralCrossRef Meng J, Banerjee S, Li D, Sindberg GM, Wang F, Ma J, et al. Opioid exacerbation of gram-positive sepsis, induced by gut microbial modulation, is rescued by IL-17A neutralization. Sci Rep. 2015;5:10918.PubMedPubMedCentralCrossRef
50.
go back to reference Bassetti M, Poulakou G, Ruppe E, Bouza E, Van Hal SJ, Brink A. Antimicrobial resistance in the next 30 years, humankind, bugs and drugs: a visionary approach. Intensive Care Med. 2017;43:1464–75.PubMedCrossRef Bassetti M, Poulakou G, Ruppe E, Bouza E, Van Hal SJ, Brink A. Antimicrobial resistance in the next 30 years, humankind, bugs and drugs: a visionary approach. Intensive Care Med. 2017;43:1464–75.PubMedCrossRef
51.
go back to reference Timsit J-F, Bassetti M, Cremer O, Daikos G, de Waele J, Kallil A, et al. Rationalizing antimicrobial therapy in the ICU: a narrative review. Intensive Care Med. 2019;45:172–89.PubMedCrossRef Timsit J-F, Bassetti M, Cremer O, Daikos G, de Waele J, Kallil A, et al. Rationalizing antimicrobial therapy in the ICU: a narrative review. Intensive Care Med. 2019;45:172–89.PubMedCrossRef
53.
go back to reference Donskey CJ, Hanrahan JA, Whalen CC. Effect of antibiotic therapy on the density of vancomycin-resistant enterococci in the stool of colonized patients. N Engl J Med. 2000;343(26):1925–32.PubMedPubMedCentralCrossRef Donskey CJ, Hanrahan JA, Whalen CC. Effect of antibiotic therapy on the density of vancomycin-resistant enterococci in the stool of colonized patients. N Engl J Med. 2000;343(26):1925–32.PubMedPubMedCentralCrossRef
54.
go back to reference Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature. 2015;517:205–8.PubMedCrossRef Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature. 2015;517:205–8.PubMedCrossRef
55.
go back to reference Caballero S, Kim S, Carter RA, Leiner IM, Sušac B, Miller L, et al. Cooperating commensals restore colonization resistance to vancomycin-resistant Enterococcus faecium. Cell Host Microbe. 2017;21:592-602.e4.PubMedPubMedCentralCrossRef Caballero S, Kim S, Carter RA, Leiner IM, Sušac B, Miller L, et al. Cooperating commensals restore colonization resistance to vancomycin-resistant Enterococcus faecium. Cell Host Microbe. 2017;21:592-602.e4.PubMedPubMedCentralCrossRef
56.
go back to reference Becattini S, Littmann ER, Carter RA, Kim SG, Morjaria SM, Ling L, et al. Commensal microbes provide first line defense against Listeria monocytogenes infection. J Exp Med. 2017;214:1973–89.PubMedPubMedCentralCrossRef Becattini S, Littmann ER, Carter RA, Kim SG, Morjaria SM, Ling L, et al. Commensal microbes provide first line defense against Listeria monocytogenes infection. J Exp Med. 2017;214:1973–89.PubMedPubMedCentralCrossRef
57.
go back to reference Taur Y, Xavier JB, Lipuma L, Ubeda C, Goldberg J, Gobourne A, et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis. 2012;55:905–14.PubMedPubMedCentralCrossRef Taur Y, Xavier JB, Lipuma L, Ubeda C, Goldberg J, Gobourne A, et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis. 2012;55:905–14.PubMedPubMedCentralCrossRef
58.
go back to reference Bernard J, Armand-Lefèvre L, Luce E, El Mniai A, Chau F, Casalino E, et al. Impact of a short exposure to levofloxacin on faecal densities and relative abundance of total and quinolone-resistant Enterobacteriaceae. Clin Microbiol Infect. 2016;22:646.e1-646.e4.CrossRef Bernard J, Armand-Lefèvre L, Luce E, El Mniai A, Chau F, Casalino E, et al. Impact of a short exposure to levofloxacin on faecal densities and relative abundance of total and quinolone-resistant Enterobacteriaceae. Clin Microbiol Infect. 2016;22:646.e1-646.e4.CrossRef
59.
go back to reference Bhalla A, Pultz NJ, Ray AJ, Hoyen CK, Eckstein EC, Donskey CJ. Antianaerobic antibiotic therapy promotes overgrowth of antibiotic-resistant, gram-negative bacilli and vancomycin-resistant enterococci in the stool of colonized patients. Infect Control Hosp Epidemiol. 2003;24:644–9.PubMedCrossRef Bhalla A, Pultz NJ, Ray AJ, Hoyen CK, Eckstein EC, Donskey CJ. Antianaerobic antibiotic therapy promotes overgrowth of antibiotic-resistant, gram-negative bacilli and vancomycin-resistant enterococci in the stool of colonized patients. Infect Control Hosp Epidemiol. 2003;24:644–9.PubMedCrossRef
60.
go back to reference Ruppé E, Lixandru B, Cojocaru R, Büke Ç, Paramythiotou E, Angebault C, et al. Relative fecal abundance of extended-spectrum-β-lactamase-producing Escherichia coli strains and their occurrence in urinary tract infections in women. Antimicrob Agents Chemother. 2013;57:4512–7.PubMedPubMedCentralCrossRef Ruppé E, Lixandru B, Cojocaru R, Büke Ç, Paramythiotou E, Angebault C, et al. Relative fecal abundance of extended-spectrum-β-lactamase-producing Escherichia coli strains and their occurrence in urinary tract infections in women. Antimicrob Agents Chemother. 2013;57:4512–7.PubMedPubMedCentralCrossRef
61.
go back to reference Stiefel U, Nerandzic MM, Pultz MJ, Donskey CJ. Gastrointestinal colonization with a cephalosporinase-producing bacteroides species preserves colonization resistance against vancomycin-resistant Enterococcus and Clostridium difficile in cephalosporin-treated mice. Antimicrob Agents Chemother. 2014;58:4535–42.PubMedPubMedCentralCrossRef Stiefel U, Nerandzic MM, Pultz MJ, Donskey CJ. Gastrointestinal colonization with a cephalosporinase-producing bacteroides species preserves colonization resistance against vancomycin-resistant Enterococcus and Clostridium difficile in cephalosporin-treated mice. Antimicrob Agents Chemother. 2014;58:4535–42.PubMedPubMedCentralCrossRef
62.
go back to reference Stiefel U, Tima MA, Nerandzic MM. Metallo-β-lactamase-producing bacteroides species can shield other members of the gut microbiota from antibiotics. Antimicrob Agents Chemother. 2015;59:650–3.PubMedCrossRef Stiefel U, Tima MA, Nerandzic MM. Metallo-β-lactamase-producing bacteroides species can shield other members of the gut microbiota from antibiotics. Antimicrob Agents Chemother. 2015;59:650–3.PubMedCrossRef
63.
go back to reference Harmoinen J. Enzymic degradation of a beta-lactam antibiotic, ampicillin, in the gut: a novel treatment modality. J Antimicrob Chemother. 2003;51:361–5.PubMedCrossRef Harmoinen J. Enzymic degradation of a beta-lactam antibiotic, ampicillin, in the gut: a novel treatment modality. J Antimicrob Chemother. 2003;51:361–5.PubMedCrossRef
64.
go back to reference Kokai-Kun JF, Bristol JA, Setser J, Schlosser M. Nonclinical Safety Assessment of SYN-004: an oral β-lactamase for the protection of the gut microbiome from disruption by biliary-excreted Intravenously Administered Antibiotics. Int J Toxicol. 2016;35:309–16.PubMedCrossRef Kokai-Kun JF, Bristol JA, Setser J, Schlosser M. Nonclinical Safety Assessment of SYN-004: an oral β-lactamase for the protection of the gut microbiome from disruption by biliary-excreted Intravenously Administered Antibiotics. Int J Toxicol. 2016;35:309–16.PubMedCrossRef
65.
go back to reference Wittekamp BHJ, Oostdijk EAN, Cuthbertson BH, Brun-Buisson C, Bonten MJM. Selective decontamination of the digestive tract (SDD) in critically ill patients: a narrative review. Intensive Care Med. 2020;46:343–9.PubMedCrossRef Wittekamp BHJ, Oostdijk EAN, Cuthbertson BH, Brun-Buisson C, Bonten MJM. Selective decontamination of the digestive tract (SDD) in critically ill patients: a narrative review. Intensive Care Med. 2020;46:343–9.PubMedCrossRef
66.
go back to reference Oostdijk EAN, Kesecioglu J, Schultz MJ, Visser CE, de Jonge E, van Essen EHR, et al. Effects of decontamination of the oropharynx and intestinal tract on antibiotic resistance in ICUs: a randomized clinical trial. JAMA. 2014;312:1429.PubMedCrossRef Oostdijk EAN, Kesecioglu J, Schultz MJ, Visser CE, de Jonge E, van Essen EHR, et al. Effects of decontamination of the oropharynx and intestinal tract on antibiotic resistance in ICUs: a randomized clinical trial. JAMA. 2014;312:1429.PubMedCrossRef
67.
go back to reference Oostdijk EAN, Kesecioglu J, Schultz MJ, Visser CE, de Jonge E, van Essen EHR, et al. Notice of Retraction and Replacement: Oostdijk et al. Effects of Decontamination of the Oropharynx and Intestinal Tract on Antibiotic Resistance in ICUs: A Randomized Clinical Trial. JAMA. 2014;312(14):1429–1437. JAMA. 2017;317:1583. Oostdijk EAN, Kesecioglu J, Schultz MJ, Visser CE, de Jonge E, van Essen EHR, et al. Notice of Retraction and Replacement: Oostdijk et al. Effects of Decontamination of the Oropharynx and Intestinal Tract on Antibiotic Resistance in ICUs: A Randomized Clinical Trial. JAMA. 2014;312(14):1429–1437. JAMA. 2017;317:1583.
68.
go back to reference Halaby T, Al Naiemi N, Kluytmans J, van der Palen J, Vandenbroucke-Grauls CMJE. Emergence of colistin resistance in enterobacteriaceae after the introduction of selective digestive tract decontamination in an intensive care unit. Antimicrob Agents Chemother. 2013;57:3224–9.PubMedPubMedCentralCrossRef Halaby T, Al Naiemi N, Kluytmans J, van der Palen J, Vandenbroucke-Grauls CMJE. Emergence of colistin resistance in enterobacteriaceae after the introduction of selective digestive tract decontamination in an intensive care unit. Antimicrob Agents Chemother. 2013;57:3224–9.PubMedPubMedCentralCrossRef
69.
go back to reference Freedberg DE, Zhou MJ, Cohen ME, Annavajhala MK, Khan S, Moscoso DI, et al. Pathogen colonization of the gastrointestinal microbiome at intensive care unit admission and risk for subsequent death or infection. Intensive Care Med. 2018;44:1203–11.PubMedPubMedCentralCrossRef Freedberg DE, Zhou MJ, Cohen ME, Annavajhala MK, Khan S, Moscoso DI, et al. Pathogen colonization of the gastrointestinal microbiome at intensive care unit admission and risk for subsequent death or infection. Intensive Care Med. 2018;44:1203–11.PubMedPubMedCentralCrossRef
70.
go back to reference Ruppé E, Armand-Lefèvre L, Estellat C, Consigny P-H, El Mniai A, Boussadia Y, et al. High rate of acquisition but short duration of carriage of multidrug-resistant enterobacteriaceae after travel to the tropics. Clin Infect Dis. 2015;61:593–600.PubMedCrossRef Ruppé E, Armand-Lefèvre L, Estellat C, Consigny P-H, El Mniai A, Boussadia Y, et al. High rate of acquisition but short duration of carriage of multidrug-resistant enterobacteriaceae after travel to the tropics. Clin Infect Dis. 2015;61:593–600.PubMedCrossRef
71.
go back to reference Lerner A, Adler A, Abu-Hanna J, Cohen Percia S, Kazma Matalon M, Carmeli Y. Spread of KPC-producing carbapenem-resistant Enterobacteriaceae: the importance of super-spreaders and rectal KPC concentration. Clin Microbiol Infect Off Publ Eur Soc Clin Microbiol Infect Dis. 2015;21(470):e1-7. Lerner A, Adler A, Abu-Hanna J, Cohen Percia S, Kazma Matalon M, Carmeli Y. Spread of KPC-producing carbapenem-resistant Enterobacteriaceae: the importance of super-spreaders and rectal KPC concentration. Clin Microbiol Infect Off Publ Eur Soc Clin Microbiol Infect Dis. 2015;21(470):e1-7.
72.
go back to reference El Kafsi H, Gorochov G, Larsen M. Host genetics affect microbial ecosystems via host immunity. Curr Opin Allergy Clin Immunol. 2016;16:413–20.PubMedCrossRef El Kafsi H, Gorochov G, Larsen M. Host genetics affect microbial ecosystems via host immunity. Curr Opin Allergy Clin Immunol. 2016;16:413–20.PubMedCrossRef
73.
go back to reference Goodrich JK, Davenport ER, Waters JL, Clark AG, Ley RE. Cross-species comparisons of host genetic associations with the microbiome. Science. 2016;352:532–5.PubMedPubMedCentralCrossRef Goodrich JK, Davenport ER, Waters JL, Clark AG, Ley RE. Cross-species comparisons of host genetic associations with the microbiome. Science. 2016;352:532–5.PubMedPubMedCentralCrossRef
74.
go back to reference Bunker JJ, Flynn TM, Koval JC, Shaw DG, Meisel M, McDonald BD, et al. Innate and adaptive humoral responses coat distinct commensal bacteria with immunoglobulin A. Immunity. 2015;43:541–53.PubMedPubMedCentralCrossRef Bunker JJ, Flynn TM, Koval JC, Shaw DG, Meisel M, McDonald BD, et al. Innate and adaptive humoral responses coat distinct commensal bacteria with immunoglobulin A. Immunity. 2015;43:541–53.PubMedPubMedCentralCrossRef
75.
go back to reference Roche AM, Richard AL, Rahkola JT, Janoff EN, Weiser JN. Antibody blocks acquisition of bacterial colonization through agglutination. Mucosal Immunol. 2015;8:176–85.PubMedCrossRef Roche AM, Richard AL, Rahkola JT, Janoff EN, Weiser JN. Antibody blocks acquisition of bacterial colonization through agglutination. Mucosal Immunol. 2015;8:176–85.PubMedCrossRef
76.
go back to reference Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature. 2016;535:75–84.PubMedCrossRef Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature. 2016;535:75–84.PubMedCrossRef
77.
go back to reference Mirpuri J, Raetz M, Sturge CR, Wilhelm CL, Benson A, Savani RC, et al. Proteobacteria-specific IgA regulates maturation of the intestinal microbiota. Gut Microbes. 2014;5:28–39.PubMedCrossRef Mirpuri J, Raetz M, Sturge CR, Wilhelm CL, Benson A, Savani RC, et al. Proteobacteria-specific IgA regulates maturation of the intestinal microbiota. Gut Microbes. 2014;5:28–39.PubMedCrossRef
78.
go back to reference Peterson DA, McNulty NP, Guruge JL, Gordon JI. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe. 2007;2:328–39.PubMedCrossRef Peterson DA, McNulty NP, Guruge JL, Gordon JI. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe. 2007;2:328–39.PubMedCrossRef
79.
go back to reference Ishigame H, Kakuta S, Nagai T, Kadoki M, Nambu A, Komiyama Y, et al. Differential roles of interleukin-17A and -17F in host defense against mucoepithelial bacterial infection and allergic responses. Immunity. 2009;30:108–19.PubMedCrossRef Ishigame H, Kakuta S, Nagai T, Kadoki M, Nambu A, Komiyama Y, et al. Differential roles of interleukin-17A and -17F in host defense against mucoepithelial bacterial infection and allergic responses. Immunity. 2009;30:108–19.PubMedCrossRef
80.
go back to reference Weaver CT, Elson CO, Fouser LA, Kolls JK. The Th17 pathway and inflammatory diseases of the intestines, lungs, and skin. Annu Rev Pathol Mech Dis. 2013;8:477–512.CrossRef Weaver CT, Elson CO, Fouser LA, Kolls JK. The Th17 pathway and inflammatory diseases of the intestines, lungs, and skin. Annu Rev Pathol Mech Dis. 2013;8:477–512.CrossRef
81.
go back to reference Russell SL, Gold MJ, Hartmann M, Willing BP, Thorson L, Wlodarska M, et al. Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Rep. 2012;13:440–7.PubMedPubMedCentralCrossRef Russell SL, Gold MJ, Hartmann M, Willing BP, Thorson L, Wlodarska M, et al. Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Rep. 2012;13:440–7.PubMedPubMedCentralCrossRef
83.
go back to reference Chen H-M, Yu Y-N, Wang J-L, Lin Y-W, Kong X, Yang C-Q, et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr. 2013;97:1044–52.PubMedCrossRef Chen H-M, Yu Y-N, Wang J-L, Lin Y-W, Kong X, Yang C-Q, et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr. 2013;97:1044–52.PubMedCrossRef
84.
go back to reference De Preter V, Machiels K, Joossens M, Arijs I, Matthys C, Vermeire S, et al. Faecal metabolite profiling identifies medium-chain fatty acids as discriminating compounds in IBD. Gut. 2015;64:447–58.PubMedCrossRef De Preter V, Machiels K, Joossens M, Arijs I, Matthys C, Vermeire S, et al. Faecal metabolite profiling identifies medium-chain fatty acids as discriminating compounds in IBD. Gut. 2015;64:447–58.PubMedCrossRef
85.
go back to reference Donohoe DR, Garge N, Zhang X, Sun W, O’Connell TM, Bunger MK, et al. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011;13:517–26.PubMedPubMedCentralCrossRef Donohoe DR, Garge N, Zhang X, Sun W, O’Connell TM, Bunger MK, et al. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011;13:517–26.PubMedPubMedCentralCrossRef
86.
go back to reference Shimizu K, Ogura H, Hamasaki T, Goto M, Tasaki O, Asahara T, et al. Altered gut flora are associated with septic complications and death in critically ill patients with systemic inflammatory response syndrome. Dig Dis Sci. 2011;56:1171–7.PubMedCrossRef Shimizu K, Ogura H, Hamasaki T, Goto M, Tasaki O, Asahara T, et al. Altered gut flora are associated with septic complications and death in critically ill patients with systemic inflammatory response syndrome. Dig Dis Sci. 2011;56:1171–7.PubMedCrossRef
87.
go back to reference Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504:446–50.CrossRefPubMed Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504:446–50.CrossRefPubMed
88.
go back to reference Sokol H, Seksik P, Rigottier-Gois L, Lay C, Lepage P, Podglajen I, et al. Specificities of the fecal microbiota in inflammatory bowel disease. Inflamm Bowel Dis. 2006;12:106–11.PubMedCrossRef Sokol H, Seksik P, Rigottier-Gois L, Lay C, Lepage P, Podglajen I, et al. Specificities of the fecal microbiota in inflammatory bowel disease. Inflamm Bowel Dis. 2006;12:106–11.PubMedCrossRef
89.
go back to reference Chen W, Liu F, Ling Z, Tong X, Xiang C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS ONE. 2012;7:e39743.PubMedPubMedCentralCrossRef Chen W, Liu F, Ling Z, Tong X, Xiang C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS ONE. 2012;7:e39743.PubMedPubMedCentralCrossRef
90.
go back to reference Eppinga H, Sperna Weiland CJ, Thio HB, van der Woude CJ, Nijsten TEC, Peppelenbosch MP, et al. Similar Depletion of Protective Faecalibacterium prausnitzii in Psoriasis and Inflammatory Bowel Disease, but not in Hidradenitis Suppurativa. J Crohns Colitis. 2016;10:1067–75.PubMedCrossRef Eppinga H, Sperna Weiland CJ, Thio HB, van der Woude CJ, Nijsten TEC, Peppelenbosch MP, et al. Similar Depletion of Protective Faecalibacterium prausnitzii in Psoriasis and Inflammatory Bowel Disease, but not in Hidradenitis Suppurativa. J Crohns Colitis. 2016;10:1067–75.PubMedCrossRef
91.
go back to reference Zuo T, Zhang F, Lui GCY, Yeoh YK, Li AYL, Zhan H, et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology. 2020;159:944-955.e8.PubMedCrossRef Zuo T, Zhang F, Lui GCY, Yeoh YK, Li AYL, Zhan H, et al. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology. 2020;159:944-955.e8.PubMedCrossRef
92.
go back to reference Tilg H, Zmora N, Adolph TE, Elinav E. The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol. 2020;20:40–54.PubMedCrossRef Tilg H, Zmora N, Adolph TE, Elinav E. The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol. 2020;20:40–54.PubMedCrossRef
93.
go back to reference Wolf AJ, Underhill DM. Peptidoglycan recognition by the innate immune system. Nat Rev Immunol. 2018;18:243–54.PubMedCrossRef Wolf AJ, Underhill DM. Peptidoglycan recognition by the innate immune system. Nat Rev Immunol. 2018;18:243–54.PubMedCrossRef
96.
go back to reference Koh A, Molinaro A, Ståhlman M, Khan MT, Schmidt C, Mannerås-Holm L, et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell. 2018;175:947-961.e17.PubMedCrossRef Koh A, Molinaro A, Ståhlman M, Khan MT, Schmidt C, Mannerås-Holm L, et al. Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell. 2018;175:947-961.e17.PubMedCrossRef
97.
go back to reference Suzuki Y, Ruiz-Ortega M, Egido J. Angiotensin II: a double-edged sword in inflammation. J Nephrol. 2000;13(Suppl 3):S101-110.PubMed Suzuki Y, Ruiz-Ortega M, Egido J. Angiotensin II: a double-edged sword in inflammation. J Nephrol. 2000;13(Suppl 3):S101-110.PubMed
99.
go back to reference Senthil M, Watkins A, Barlos D, Xu D-Z, Lu Q, Abungu B, et al. Intravenous injection of trauma-hemorrhagic shock mesenteric lymph causes lung injury that is dependent upon activation of the inducible nitric oxide synthase pathway. Ann Surg. 2007;246(5):822–30.PubMedCrossRef Senthil M, Watkins A, Barlos D, Xu D-Z, Lu Q, Abungu B, et al. Intravenous injection of trauma-hemorrhagic shock mesenteric lymph causes lung injury that is dependent upon activation of the inducible nitric oxide synthase pathway. Ann Surg. 2007;246(5):822–30.PubMedCrossRef
100.
go back to reference Gou W, Fu Y, Yue L, Chen G-D, Cai X, Shuai M, et al. Gut microbiota, inflammation, and molecular signatures of host response to infection. J Genet Genomics Yi Chuan Xue Bao. 2021;48(9):792–802.PubMedCrossRef Gou W, Fu Y, Yue L, Chen G-D, Cai X, Shuai M, et al. Gut microbiota, inflammation, and molecular signatures of host response to infection. J Genet Genomics Yi Chuan Xue Bao. 2021;48(9):792–802.PubMedCrossRef
101.
go back to reference Yeoh YK, Zuo T, Lui GCY, Zhang F, Liu Q, Li AY, et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut. 2021;70(4):698–706.PubMedCrossRef Yeoh YK, Zuo T, Lui GCY, Zhang F, Liu Q, Li AY, et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut. 2021;70(4):698–706.PubMedCrossRef
102.
go back to reference Zuo T, Liu Q, Zhang F, Lui GCY, Tso EY, Yeoh YK, et al. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut. 2020;70(2):276–84.PubMed Zuo T, Liu Q, Zhang F, Lui GCY, Tso EY, Yeoh YK, et al. Depicting SARS-CoV-2 faecal viral activity in association with gut microbiota composition in patients with COVID-19. Gut. 2020;70(2):276–84.PubMed
103.
go back to reference Zaborin A, Smith D, Garfield K, Quensen J, Shakhsheer B, Kade M, et al. Membership and behavior of ultra-low-diversity pathogen communities present in the gut of humans during prolonged critical illness. MBio. 2014;5(5):e01361-e1414.PubMedPubMedCentralCrossRef Zaborin A, Smith D, Garfield K, Quensen J, Shakhsheer B, Kade M, et al. Membership and behavior of ultra-low-diversity pathogen communities present in the gut of humans during prolonged critical illness. MBio. 2014;5(5):e01361-e1414.PubMedPubMedCentralCrossRef
104.
go back to reference Rajilić-Stojanović M, de Vos WM. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev. 2014;38(5):996–1047.PubMedCrossRef Rajilić-Stojanović M, de Vos WM. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev. 2014;38(5):996–1047.PubMedCrossRef
105.
go back to reference Ojima M, Motooka D, Shimizu K, Gotoh K, Shintani A, Yoshiya K, et al. Metagenomic analysis reveals dynamic changes of whole gut microbiota in the acute phase of intensive care unit patients. Dig Dis Sci. 2016;61(6):1628–34.PubMedCrossRef Ojima M, Motooka D, Shimizu K, Gotoh K, Shintani A, Yoshiya K, et al. Metagenomic analysis reveals dynamic changes of whole gut microbiota in the acute phase of intensive care unit patients. Dig Dis Sci. 2016;61(6):1628–34.PubMedCrossRef
106.
go back to reference Yeh A, Rogers MB, Firek B, Neal MD, Zuckerbraun BS, Morowitz MJ. Dysbiosis across multiple body sites in critically ill adult surgical patients. Shock. 2016;46(6):649–54.PubMedCrossRef Yeh A, Rogers MB, Firek B, Neal MD, Zuckerbraun BS, Morowitz MJ. Dysbiosis across multiple body sites in critically ill adult surgical patients. Shock. 2016;46(6):649–54.PubMedCrossRef
107.
go back to reference Chastre J, Fagon J-Y. Ventilator-associated Pneumonia. Am J Respir Crit Care Med. 2002;165(7):867–903.PubMedCrossRef Chastre J, Fagon J-Y. Ventilator-associated Pneumonia. Am J Respir Crit Care Med. 2002;165(7):867–903.PubMedCrossRef
108.
go back to reference Ziakas PD, Thapa R, Rice LB, Mylonakis E. Trends and significance of vre colonization in the ICU: a meta-analysis of published studies. PLoS ONE. 2013;8(9):e75658.PubMedPubMedCentralCrossRef Ziakas PD, Thapa R, Rice LB, Mylonakis E. Trends and significance of vre colonization in the ICU: a meta-analysis of published studies. PLoS ONE. 2013;8(9):e75658.PubMedPubMedCentralCrossRef
109.
go back to reference Jung E, Byun S, Lee H, Moon SY, Lee H. Vancomycin-resistant Enterococcus colonization in the intensive care unit: Clinical outcomes and attributable costs of hospitalization. Am J Infect Control. 2014;42(10):1062–6.PubMedCrossRef Jung E, Byun S, Lee H, Moon SY, Lee H. Vancomycin-resistant Enterococcus colonization in the intensive care unit: Clinical outcomes and attributable costs of hospitalization. Am J Infect Control. 2014;42(10):1062–6.PubMedCrossRef
110.
go back to reference Andremont O, Armand-Lefevre L, Dupuis C, de Montmollin E, Ruckly S, Lucet J-C, et al. Semi-quantitative cultures of throat and rectal swabs are efficient tests to predict ESBL-Enterobacterales ventilator-associated pneumonia in mechanically ventilated ESBL carriers. Intensive Care Med. 2020;46(6):1232–42.PubMedCrossRefPubMedCentral Andremont O, Armand-Lefevre L, Dupuis C, de Montmollin E, Ruckly S, Lucet J-C, et al. Semi-quantitative cultures of throat and rectal swabs are efficient tests to predict ESBL-Enterobacterales ventilator-associated pneumonia in mechanically ventilated ESBL carriers. Intensive Care Med. 2020;46(6):1232–42.PubMedCrossRefPubMedCentral
111.
go back to reference Ewan VC, Sails AD, Walls AWG, Rushton S, Newton JL. Dental and microbiological risk factors for hospital-acquired pneumonia in non-ventilated older patients. PLoS ONE. 2015;10(4):e0123622.PubMedPubMedCentralCrossRef Ewan VC, Sails AD, Walls AWG, Rushton S, Newton JL. Dental and microbiological risk factors for hospital-acquired pneumonia in non-ventilated older patients. PLoS ONE. 2015;10(4):e0123622.PubMedPubMedCentralCrossRef
112.
go back to reference Taur Y, Jenq RR, Perales M-A, Littmann ER, Morjaria S, Ling L, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124:1174–82.PubMedPubMedCentralCrossRef Taur Y, Jenq RR, Perales M-A, Littmann ER, Morjaria S, Ling L, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124:1174–82.PubMedPubMedCentralCrossRef
113.
go back to reference Schuijt TJ, Lankelma JM, Scicluna BP, de Melo F, Roelofs JJTH, de Boer JD, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut. 2016;65:575–83.PubMedCrossRef Schuijt TJ, Lankelma JM, Scicluna BP, de Melo F, Roelofs JJTH, de Boer JD, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut. 2016;65:575–83.PubMedCrossRef
114.
go back to reference Yin Y, Hountras P, Wunderink RG. The microbiome in mechanically ventilated patients. Curr Opin Infect Dis. 2017;30:208–13.PubMedCrossRef Yin Y, Hountras P, Wunderink RG. The microbiome in mechanically ventilated patients. Curr Opin Infect Dis. 2017;30:208–13.PubMedCrossRef
115.
go back to reference Dickson RP, Singer BH, Newstead MW, Falkowski NR, Erb-Downward JR, Standiford TJ, et al. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat Microbiol. 2016;1:16113.PubMedPubMedCentralCrossRef Dickson RP, Singer BH, Newstead MW, Falkowski NR, Erb-Downward JR, Standiford TJ, et al. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat Microbiol. 2016;1:16113.PubMedPubMedCentralCrossRef
116.
go back to reference Emonet S, Lazarevic V, Leemann Refondini C, Gaïa N, Leo S, Girard M, et al. Identification of respiratory microbiota markers in ventilator-associated pneumonia. Intensive Care Med. 2019;45:1082–92.PubMedPubMedCentralCrossRef Emonet S, Lazarevic V, Leemann Refondini C, Gaïa N, Leo S, Girard M, et al. Identification of respiratory microbiota markers in ventilator-associated pneumonia. Intensive Care Med. 2019;45:1082–92.PubMedPubMedCentralCrossRef
117.
go back to reference Hernández-Chirlaque C, Aranda CJ, Ocón B, Capitán-Cañadas F, Ortega-González M, Carrero JJ, et al. Germ-free and antibiotic-treated mice are highly susceptible to epithelial injury in DSS colitis. J Crohns Colitis. 2016;10:1324–35.PubMedCrossRef Hernández-Chirlaque C, Aranda CJ, Ocón B, Capitán-Cañadas F, Ortega-González M, Carrero JJ, et al. Germ-free and antibiotic-treated mice are highly susceptible to epithelial injury in DSS colitis. J Crohns Colitis. 2016;10:1324–35.PubMedCrossRef
118.
go back to reference de Jonge E, Schultz MJ, Spanjaard L, Bossuyt PM, Vroom MB, Dankert J, et al. Effects of selective decontamination of digestive tract on mortality and acquisition of resistant bacteria in intensive care: a randomised controlled trial. Lancet. 2003;362:1011–6.PubMedCrossRef de Jonge E, Schultz MJ, Spanjaard L, Bossuyt PM, Vroom MB, Dankert J, et al. Effects of selective decontamination of digestive tract on mortality and acquisition of resistant bacteria in intensive care: a randomised controlled trial. Lancet. 2003;362:1011–6.PubMedCrossRef
119.
go back to reference de Smet AMGA, Kluytmans JAJW, Cooper BS, Mascini EM, Benus RFJ, van der Werf TS, et al. Decontamination of the digestive tract and oropharynx in ICU patients. N Engl J Med. 2009;360:20–31.PubMedCrossRef de Smet AMGA, Kluytmans JAJW, Cooper BS, Mascini EM, Benus RFJ, van der Werf TS, et al. Decontamination of the digestive tract and oropharynx in ICU patients. N Engl J Med. 2009;360:20–31.PubMedCrossRef
120.
go back to reference Wittekamp BH, Plantinga NL, Cooper BS, Lopez-Contreras J, Coll P, Mancebo J, et al. Decontamination strategies and bloodstream infections with antibiotic-resistant microorganisms in ventilated patients: a randomized clinical trial. JAMA. 2018;320:2087.PubMedPubMedCentralCrossRef Wittekamp BH, Plantinga NL, Cooper BS, Lopez-Contreras J, Coll P, Mancebo J, et al. Decontamination strategies and bloodstream infections with antibiotic-resistant microorganisms in ventilated patients: a randomized clinical trial. JAMA. 2018;320:2087.PubMedPubMedCentralCrossRef
121.
go back to reference Fontaine C, Armand-Lefèvre L, Magnan M, Nazimoudine A, Timsit J-F, Ruppé E. Relationship between the composition of the intestinal microbiota and the tracheal and intestinal colonization by opportunistic pathogens in intensive care patients. PLoS ONE. 2020;15:e0237260.PubMedPubMedCentralCrossRef Fontaine C, Armand-Lefèvre L, Magnan M, Nazimoudine A, Timsit J-F, Ruppé E. Relationship between the composition of the intestinal microbiota and the tracheal and intestinal colonization by opportunistic pathogens in intensive care patients. PLoS ONE. 2020;15:e0237260.PubMedPubMedCentralCrossRef
122.
go back to reference Stein-Thoeringer CK, Nichols KB, Lazrak A, Docampo MD, Slingerland AE, Slingerland JB, et al. Lactose drives Enterococcus expansion to promote graft-versus-host disease. Science. 2019;366:1143–9.PubMedPubMedCentralCrossRef Stein-Thoeringer CK, Nichols KB, Lazrak A, Docampo MD, Slingerland AE, Slingerland JB, et al. Lactose drives Enterococcus expansion to promote graft-versus-host disease. Science. 2019;366:1143–9.PubMedPubMedCentralCrossRef
123.
go back to reference Kaleko M, Bristol JA, Hubert S, Parsley T, Widmer G, Tzipori S, et al. Development of SYN-004, an oral beta-lactamase treatment to protect the gut microbiome from antibiotic-mediated damage and prevent Clostridium difficile infection. Anaerobe. 2016;41:58–67.PubMedCrossRef Kaleko M, Bristol JA, Hubert S, Parsley T, Widmer G, Tzipori S, et al. Development of SYN-004, an oral beta-lactamase treatment to protect the gut microbiome from antibiotic-mediated damage and prevent Clostridium difficile infection. Anaerobe. 2016;41:58–67.PubMedCrossRef
124.
go back to reference Roberts T, Kokai-Kun JF, Coughlin O, Lopez BV, Whalen H, Bristol JA, et al. Tolerability and pharmacokinetics of SYN-004, an orally administered β-lactamase for the prevention of clostridium difficile-associated disease and antibiotic-associated diarrhea, in two phase 1 studies. Clin Drug Investig. 2016;36:725–34.PubMedCrossRef Roberts T, Kokai-Kun JF, Coughlin O, Lopez BV, Whalen H, Bristol JA, et al. Tolerability and pharmacokinetics of SYN-004, an orally administered β-lactamase for the prevention of clostridium difficile-associated disease and antibiotic-associated diarrhea, in two phase 1 studies. Clin Drug Investig. 2016;36:725–34.PubMedCrossRef
125.
go back to reference Kokai-Kun JF, Roberts T, Coughlin O, Le C, Whalen H, Stevenson R, et al. Use of ribaxamase (SYN-004), a β-lactamase, to prevent Clostridium difficile infection in β-lactam-treated patients: a double-blind, phase 2b, randomised placebo-controlled trial. Lancet Infect Dis. 2019;19:487–96.PubMedCrossRef Kokai-Kun JF, Roberts T, Coughlin O, Le C, Whalen H, Stevenson R, et al. Use of ribaxamase (SYN-004), a β-lactamase, to prevent Clostridium difficile infection in β-lactam-treated patients: a double-blind, phase 2b, randomised placebo-controlled trial. Lancet Infect Dis. 2019;19:487–96.PubMedCrossRef
126.
go back to reference de Gunzburg J, Ducher A, Modess C, Wegner D, Oswald S, Dressman J, et al. Targeted adsorption of molecules in the colon with the novel adsorbent-based Medicinal Product, DAV132: A proof of concept study in healthy subjects. J Clin Pharmacol. 2015;55:10–6.PubMedCrossRef de Gunzburg J, Ducher A, Modess C, Wegner D, Oswald S, Dressman J, et al. Targeted adsorption of molecules in the colon with the novel adsorbent-based Medicinal Product, DAV132: A proof of concept study in healthy subjects. J Clin Pharmacol. 2015;55:10–6.PubMedCrossRef
127.
go back to reference de Gunzburg J, Ghozlane A, Ducher A, Le Chatelier E, Duval X, Ruppé E, et al. Protection of the human gut microbiome from antibiotics. J Infect Dis. 2018;217:628–36.PubMedCrossRef de Gunzburg J, Ghozlane A, Ducher A, Le Chatelier E, Duval X, Ruppé E, et al. Protection of the human gut microbiome from antibiotics. J Infect Dis. 2018;217:628–36.PubMedCrossRef
128.
go back to reference Burdet C, Sayah-Jeanne S, Nguyen TT, Miossec C, Saint-Lu N, Pulse M, et al. Protection of hamsters from mortality by reducing fecal moxifloxacin concentration with DAV131A in a Model of moxifloxacin-induced Clostridium difficile colitis. Antimicrob Agents Chemother. 2017;61(10):e00543-e617.PubMedPubMedCentralCrossRef Burdet C, Sayah-Jeanne S, Nguyen TT, Miossec C, Saint-Lu N, Pulse M, et al. Protection of hamsters from mortality by reducing fecal moxifloxacin concentration with DAV131A in a Model of moxifloxacin-induced Clostridium difficile colitis. Antimicrob Agents Chemother. 2017;61(10):e00543-e617.PubMedPubMedCentralCrossRef
129.
go back to reference Armand-Lefèvre L, Angebault C, Barbier F, Hamelet E, Defrance G, Ruppé E, et al. Emergence of imipenem-resistant gram-negative bacilli in intestinal flora of intensive care patients. Antimicrob Agents Chemother. 2013;57:1488–95.PubMedPubMedCentralCrossRef Armand-Lefèvre L, Angebault C, Barbier F, Hamelet E, Defrance G, Ruppé E, et al. Emergence of imipenem-resistant gram-negative bacilli in intestinal flora of intensive care patients. Antimicrob Agents Chemother. 2013;57:1488–95.PubMedPubMedCentralCrossRef
130.
go back to reference Tabah A, Bassetti M, Kollef MH, Zahar J-R, Paiva J-A, Timsit J-F, et al. Antimicrobial de-escalation in critically ill patients: a position statement from a task force of the European Society of Intensive Care Medicine (ESICM) and European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Critically Ill Patients Study Group (ESGCIP). Intensive Care Med. 2020;46:245–65.PubMedCrossRef Tabah A, Bassetti M, Kollef MH, Zahar J-R, Paiva J-A, Timsit J-F, et al. Antimicrobial de-escalation in critically ill patients: a position statement from a task force of the European Society of Intensive Care Medicine (ESICM) and European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Critically Ill Patients Study Group (ESGCIP). Intensive Care Med. 2020;46:245–65.PubMedCrossRef
131.
go back to reference Woerther P-L, Lepeule R, Burdet C, Decousser J-W, Ruppé É, Barbier F. Carbapenems and alternative β-lactams for the treatment of infections due to extended-spectrum β-lactamase-producing Enterobacteriaceae: what impact on intestinal colonisation resistance? Int J Antimicrob Agents. 2018;52:762–70.PubMedCrossRef Woerther P-L, Lepeule R, Burdet C, Decousser J-W, Ruppé É, Barbier F. Carbapenems and alternative β-lactams for the treatment of infections due to extended-spectrum β-lactamase-producing Enterobacteriaceae: what impact on intestinal colonisation resistance? Int J Antimicrob Agents. 2018;52:762–70.PubMedCrossRef
132.
go back to reference Grall N, Lazarevic V, Gaïa N, Couffignal C, Laouénan C, Ilic-Habensus E, et al. Unexpected persistence of extended-spectrum β-lactamase-producing Enterobacteriaceae in the faecal microbiota of hospitalised patients treated with imipenem. Int J Antimicrob Agents. 2017;50:81–7.PubMedCrossRef Grall N, Lazarevic V, Gaïa N, Couffignal C, Laouénan C, Ilic-Habensus E, et al. Unexpected persistence of extended-spectrum β-lactamase-producing Enterobacteriaceae in the faecal microbiota of hospitalised patients treated with imipenem. Int J Antimicrob Agents. 2017;50:81–7.PubMedCrossRef
133.
go back to reference Khailova L, Petrie B, Baird CH, Dominguez Rieg JA, Wischmeyer PE. Lactobacillus rhamnosus GG and bifidobacterium longum attenuate lung injury and inflammatory response in experimental sepsis. PLoS ONE. 2014;9:e97861.PubMedPubMedCentralCrossRef Khailova L, Petrie B, Baird CH, Dominguez Rieg JA, Wischmeyer PE. Lactobacillus rhamnosus GG and bifidobacterium longum attenuate lung injury and inflammatory response in experimental sepsis. PLoS ONE. 2014;9:e97861.PubMedPubMedCentralCrossRef
134.
135.
go back to reference Manzanares W, Lemieux M, Langlois PL, Wischmeyer PE. Probiotic and synbiotic therapy in critical illness: a systematic review and meta-analysis. Crit Care. 2016;20:262.PubMedCentralCrossRef Manzanares W, Lemieux M, Langlois PL, Wischmeyer PE. Probiotic and synbiotic therapy in critical illness: a systematic review and meta-analysis. Crit Care. 2016;20:262.PubMedCentralCrossRef
136.
go back to reference Petrof EO, Dhaliwal R, Manzanares W, Johnstone J, Cook D, Heyland DK. Probiotics in the critically ill: a systematic review of the randomized trial evidence. Crit Care Med. 2012;40:3290–302.PubMedCrossRef Petrof EO, Dhaliwal R, Manzanares W, Johnstone J, Cook D, Heyland DK. Probiotics in the critically ill: a systematic review of the randomized trial evidence. Crit Care Med. 2012;40:3290–302.PubMedCrossRef
137.
go back to reference Siempos II, Ntaidou TK, Falagas ME. Impact of the administration of probiotics on the incidence of ventilator-associated pneumonia: a meta-analysis of randomized controlled trials. Crit Care Med. 2010;38:954–62.PubMedCrossRef Siempos II, Ntaidou TK, Falagas ME. Impact of the administration of probiotics on the incidence of ventilator-associated pneumonia: a meta-analysis of randomized controlled trials. Crit Care Med. 2010;38:954–62.PubMedCrossRef
138.
go back to reference Johnstone J, Meade M, Lauzier F, Marshall J, Duan E, Dionne J, et al. Effect of probiotics on incident ventilator-associated pneumonia in critically ill patients: a randomized clinical trial. JAMA. 2021;326:1024.PubMedCrossRefPubMedCentral Johnstone J, Meade M, Lauzier F, Marshall J, Duan E, Dionne J, et al. Effect of probiotics on incident ventilator-associated pneumonia in critically ill patients: a randomized clinical trial. JAMA. 2021;326:1024.PubMedCrossRefPubMedCentral
140.
go back to reference Chamberlain R, Lau C. Probiotics are effective at preventing Clostridium difficile-associated diarrhea: a systematic review and meta-analysis. Int J Gen Med. 2016;9:27–37.PubMedPubMedCentralCrossRef Chamberlain R, Lau C. Probiotics are effective at preventing Clostridium difficile-associated diarrhea: a systematic review and meta-analysis. Int J Gen Med. 2016;9:27–37.PubMedPubMedCentralCrossRef
141.
go back to reference Goldenberg JZ, Yap C, Lytvyn L, et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev. 2017;12(12): CD006095. Goldenberg JZ, Yap C, Lytvyn L, et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev. 2017;12(12): CD006095.
142.
go back to reference van Prehn J, Reigadas E, Vogelzang EH, Bouza E, Hristea A, Guery B, et al. European Society of Clinical Microbiology and Infectious Diseases: 2021 update on the treatment guidance document for Clostridioides difficile infection in adults. Clin Microbiol Infect Off Publ Eur Soc Clin Microbiol Infect Dis. 2021;S1198–743X(21)00568–1. van Prehn J, Reigadas E, Vogelzang EH, Bouza E, Hristea A, Guery B, et al. European Society of Clinical Microbiology and Infectious Diseases: 2021 update on the treatment guidance document for Clostridioides difficile infection in adults. Clin Microbiol Infect Off Publ Eur Soc Clin Microbiol Infect Dis. 2021;S1198–743X(21)00568–1.
143.
go back to reference Besselink MGH, van Santvoort HC, Buskens E, Boermeester MA, van Goor H, Timmerman HM, et al. Probiotic prophylaxis in predicted severe acute pancreatitis: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;371(9613):651–9.PubMedCrossRef Besselink MGH, van Santvoort HC, Buskens E, Boermeester MA, van Goor H, Timmerman HM, et al. Probiotic prophylaxis in predicted severe acute pancreatitis: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;371(9613):651–9.PubMedCrossRef
144.
go back to reference Li C, Liu L, Gao Z, Zhang J, Chen H, Ma S, et al. Synbiotic therapy prevents nosocomial infection in critically ill adult patients: a systematic review and network meta-analysis of randomized controlled trials based on a bayesian framework. Front Med. 2021;8:693188.CrossRef Li C, Liu L, Gao Z, Zhang J, Chen H, Ma S, et al. Synbiotic therapy prevents nosocomial infection in critically ill adult patients: a systematic review and network meta-analysis of randomized controlled trials based on a bayesian framework. Front Med. 2021;8:693188.CrossRef
145.
go back to reference Knight DJW, Gardiner D, Banks A, Snape SE, Weston VC, Bengmark S, et al. Effect of synbiotic therapy on the incidence of ventilator associated pneumonia in critically ill patients: a randomised, double-blind, placebo-controlled trial. Intensive Care Med. 2009;35(5):854–61.PubMedCrossRef Knight DJW, Gardiner D, Banks A, Snape SE, Weston VC, Bengmark S, et al. Effect of synbiotic therapy on the incidence of ventilator associated pneumonia in critically ill patients: a randomised, double-blind, placebo-controlled trial. Intensive Care Med. 2009;35(5):854–61.PubMedCrossRef
146.
go back to reference Shimizu K, Yamada T, Ogura H, Mohri T, Kiguchi T, Fujimi S, et al. Synbiotics modulate gut microbiota and reduce enteritis and ventilator-associated pneumonia in patients with sepsis: a randomized controlled trial. Crit Care. 2018;22:239.PubMedPubMedCentralCrossRef Shimizu K, Yamada T, Ogura H, Mohri T, Kiguchi T, Fujimi S, et al. Synbiotics modulate gut microbiota and reduce enteritis and ventilator-associated pneumonia in patients with sepsis: a randomized controlled trial. Crit Care. 2018;22:239.PubMedPubMedCentralCrossRef
147.
go back to reference Khanna S, Pardi DS, Kelly CR, Kraft CS, Dhere T, Henn MR, et al. A novel microbiome therapeutic increases gut microbial diversity and prevents recurrent Clostridium difficile infection. J Infect Dis. 2016;214:173–81.PubMedCrossRef Khanna S, Pardi DS, Kelly CR, Kraft CS, Dhere T, Henn MR, et al. A novel microbiome therapeutic increases gut microbial diversity and prevents recurrent Clostridium difficile infection. J Infect Dis. 2016;214:173–81.PubMedCrossRef
148.
go back to reference McGovern BH, Ford CB, Henn MR, Pardi DS, Khanna S, Hohmann EL, et al. SER-109, an investigational microbiome drug to reduce recurrence after Clostridioides difficile infection: lessons learned from a phase 2 trial. Clin Infect Dis Off Publ Infect Dis Soc Am. 2021;72:2132–40.CrossRef McGovern BH, Ford CB, Henn MR, Pardi DS, Khanna S, Hohmann EL, et al. SER-109, an investigational microbiome drug to reduce recurrence after Clostridioides difficile infection: lessons learned from a phase 2 trial. Clin Infect Dis Off Publ Infect Dis Soc Am. 2021;72:2132–40.CrossRef
149.
go back to reference Sundararaman A, Ray M, Ravindra PV, Halami PM. Role of probiotics to combat viral infections with emphasis on COVID-19. Appl Microbiol Biotechnol. 2020;104:8089–104.PubMedCrossRefPubMedCentral Sundararaman A, Ray M, Ravindra PV, Halami PM. Role of probiotics to combat viral infections with emphasis on COVID-19. Appl Microbiol Biotechnol. 2020;104:8089–104.PubMedCrossRefPubMedCentral
150.
go back to reference Din AU, Mazhar M, Waseem M, Ahmad W, Bibi A, Hassan A, et al. SARS-CoV-2 microbiome dysbiosis linked disorders and possible probiotics role. Biomed Pharmacother. 2021;133:110947.PubMedCrossRef Din AU, Mazhar M, Waseem M, Ahmad W, Bibi A, Hassan A, et al. SARS-CoV-2 microbiome dysbiosis linked disorders and possible probiotics role. Biomed Pharmacother. 2021;133:110947.PubMedCrossRef
151.
go back to reference Muthukrishnan PT, Faillace R. Compassionate use of others’ immunity—understanding gut microbiome in Covid-19. Crit Care. 2020;24:1–2. Muthukrishnan PT, Faillace R. Compassionate use of others’ immunity—understanding gut microbiome in Covid-19. Crit Care. 2020;24:1–2.
152.
go back to reference Alagna L, Haak BW, Gori A. Fecal microbiota transplantation in the ICU: perspectives on future implementations. Intensive Care Med. 2019;45:998–1001.PubMedCrossRef Alagna L, Haak BW, Gori A. Fecal microbiota transplantation in the ICU: perspectives on future implementations. Intensive Care Med. 2019;45:998–1001.PubMedCrossRef
153.
go back to reference van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, et al. Duodenal Infusion of Donor Feces for Recurrent Clostridium difficile. N Engl J Med. 2013;368:407–15.PubMedCrossRef van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, et al. Duodenal Infusion of Donor Feces for Recurrent Clostridium difficile. N Engl J Med. 2013;368:407–15.PubMedCrossRef
154.
go back to reference Cammarota G, Masucci L, Ianiro G, Bibbò S, Dinoi G, Costamagna G, et al. Randomised clinical trial: faecal microbiota transplantation by colonoscopy vs. vancomycin for the treatment of recurrent Clostridium difficile infection. Aliment Pharmacol Ther. 2015;41:835–43.PubMedCrossRef Cammarota G, Masucci L, Ianiro G, Bibbò S, Dinoi G, Costamagna G, et al. Randomised clinical trial: faecal microbiota transplantation by colonoscopy vs. vancomycin for the treatment of recurrent Clostridium difficile infection. Aliment Pharmacol Ther. 2015;41:835–43.PubMedCrossRef
155.
go back to reference McDonald LC, Gerding DN, Johnson S, Bakken JS, Carroll KC, Coffin SE, et al. Clinical Practice Guidelines for Clostridium difficile Infection in Adults and Children: 2017 Update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin Infect Dis. 2018;66:e1-48.PubMedPubMedCentralCrossRef McDonald LC, Gerding DN, Johnson S, Bakken JS, Carroll KC, Coffin SE, et al. Clinical Practice Guidelines for Clostridium difficile Infection in Adults and Children: 2017 Update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin Infect Dis. 2018;66:e1-48.PubMedPubMedCentralCrossRef
156.
go back to reference Quraishi MN, Widlak M, Bhala N, Moore D, Price M, Sharma N, et al. Systematic review with meta-analysis: the efficacy of faecal microbiota transplantation for the treatment of recurrent and refractory Clostridium difficile infection. Aliment Pharmacol Ther. 2017;46:479–93.PubMedCrossRef Quraishi MN, Widlak M, Bhala N, Moore D, Price M, Sharma N, et al. Systematic review with meta-analysis: the efficacy of faecal microbiota transplantation for the treatment of recurrent and refractory Clostridium difficile infection. Aliment Pharmacol Ther. 2017;46:479–93.PubMedCrossRef
157.
go back to reference Cammarota G, Ianiro G, Tilg H, Rajilić-Stojanović M, Kump P, Satokari R, et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut. 2017;66:569–80.PubMedCrossRef Cammarota G, Ianiro G, Tilg H, Rajilić-Stojanović M, Kump P, Satokari R, et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut. 2017;66:569–80.PubMedCrossRef
158.
go back to reference Limketkai BN, Hendler S, Ting P, Parian AM. Fecal microbiota transplantation for the critically ill patient. Nutr Clin Pract. 2019;34:73–9.PubMedCrossRef Limketkai BN, Hendler S, Ting P, Parian AM. Fecal microbiota transplantation for the critically ill patient. Nutr Clin Pract. 2019;34:73–9.PubMedCrossRef
159.
go back to reference Ooijevaar RE, Terveer EM, Verspaget HW, Kuijper EJ, Keller JJ. Clinical application and potential of fecal microbiota transplantation. Annu Rev Med. 2019;70:335–51.PubMedCrossRef Ooijevaar RE, Terveer EM, Verspaget HW, Kuijper EJ, Keller JJ. Clinical application and potential of fecal microbiota transplantation. Annu Rev Med. 2019;70:335–51.PubMedCrossRef
160.
go back to reference DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, et al. Drug-resistant e coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381:2043–50.PubMedCrossRef DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, et al. Drug-resistant e coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381:2043–50.PubMedCrossRef
161.
go back to reference Lee CH, Steiner T, Petrof EO, Smieja M, Roscoe D, Nematallah A, et al. Frozen vs fresh fecal microbiota transplantation and clinical resolution of diarrhea in patients with recurrent clostridium difficile infection: a randomized clinical trial. JAMA. 2016;315:142.PubMedCrossRef Lee CH, Steiner T, Petrof EO, Smieja M, Roscoe D, Nematallah A, et al. Frozen vs fresh fecal microbiota transplantation and clinical resolution of diarrhea in patients with recurrent clostridium difficile infection: a randomized clinical trial. JAMA. 2016;315:142.PubMedCrossRef
162.
go back to reference Millan B, Park H, Hotte N, Mathieu O, Burguiere P, Tompkins TA, et al. Fecal microbial transplants reduce antibiotic-resistant genes in patients with recurrent Clostridium difficile infection. Clin Infect Dis. 2016;62:1479–86.PubMedPubMedCentralCrossRef Millan B, Park H, Hotte N, Mathieu O, Burguiere P, Tompkins TA, et al. Fecal microbial transplants reduce antibiotic-resistant genes in patients with recurrent Clostridium difficile infection. Clin Infect Dis. 2016;62:1479–86.PubMedPubMedCentralCrossRef
163.
go back to reference Bilinski J, Grzesiowski P, Sorensen N, Madry K, Muszynski J, Robak K, 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. 2017;65:364–70.PubMedCrossRef Bilinski J, Grzesiowski P, Sorensen N, Madry K, Muszynski J, Robak K, 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. 2017;65:364–70.PubMedCrossRef
164.
go back to reference Tavoukjian V. Faecal microbiota transplantation for the decolonization of antibiotic-resistant bacteria in the gut: a systematic review and meta-analysis. J Hosp Infect. 2019;102:174–88.PubMedCrossRef Tavoukjian V. Faecal microbiota transplantation for the decolonization of antibiotic-resistant bacteria in the gut: a systematic review and meta-analysis. J Hosp Infect. 2019;102:174–88.PubMedCrossRef
165.
go back to reference Huttner BD, de Lastours V, Wassenberg M, Maharshak N, Mauris A, Galperine T, et al. A 5-day course of oral antibiotics followed by faecal transplantation to eradicate carriage of multidrug-resistant Enterobacteriaceae: a randomized clinical trial. Clin Microbiol Infect. 2019;25:830–8.PubMedCrossRef Huttner BD, de Lastours V, Wassenberg M, Maharshak N, Mauris A, Galperine T, et al. A 5-day course of oral antibiotics followed by faecal transplantation to eradicate carriage of multidrug-resistant Enterobacteriaceae: a randomized clinical trial. Clin Microbiol Infect. 2019;25:830–8.PubMedCrossRef
166.
go back to reference Wei Y, Yang J, Wang J, Yang Y, Huang J, Gong H, et al. Successful treatment with fecal microbiota transplantation in patients with multiple organ dysfunction syndrome and diarrhea following severe sepsis. Crit Care. 2016;20:332.PubMedPubMedCentralCrossRef Wei Y, Yang J, Wang J, Yang Y, Huang J, Gong H, et al. Successful treatment with fecal microbiota transplantation in patients with multiple organ dysfunction syndrome and diarrhea following severe sepsis. Crit Care. 2016;20:332.PubMedPubMedCentralCrossRef
167.
go back to reference Li Q, Wang C, Tang C, He Q, Zhao X, Li N, et al. Therapeutic modulation and reestablishment of the intestinal microbiota with fecal microbiota transplantation resolves sepsis and diarrhea in a patient. Am J Gastroenterol. 2014;109:1832–4.PubMedCrossRef Li Q, Wang C, Tang C, He Q, Zhao X, Li N, et al. Therapeutic modulation and reestablishment of the intestinal microbiota with fecal microbiota transplantation resolves sepsis and diarrhea in a patient. Am J Gastroenterol. 2014;109:1832–4.PubMedCrossRef
168.
go back to reference Li Q, Wang C, Tang C, He Q, Zhao X, Li N, et al. Successful treatment of severe sepsis and diarrhea after vagotomy utilizing fecal microbiota transplantation: a case report. Crit Care. 2015;19:37.PubMedPubMedCentralCrossRef Li Q, Wang C, Tang C, He Q, Zhao X, Li N, et al. Successful treatment of severe sepsis and diarrhea after vagotomy utilizing fecal microbiota transplantation: a case report. Crit Care. 2015;19:37.PubMedPubMedCentralCrossRef
169.
go back to reference Wurm P, Spindelboeck W, Krause R, Plank J, Fuchs G, Bashir M, et al. Antibiotic-associated apoptotic enterocolitis in the absence of a defined pathogen: the role of intestinal microbiota depletion. Crit Care Med. 2017;45:e600–6.PubMedPubMedCentralCrossRef Wurm P, Spindelboeck W, Krause R, Plank J, Fuchs G, Bashir M, et al. Antibiotic-associated apoptotic enterocolitis in the absence of a defined pathogen: the role of intestinal microbiota depletion. Crit Care Med. 2017;45:e600–6.PubMedPubMedCentralCrossRef
170.
go back to reference Dai M, Liu Y, Chen W, Buch H, Shan Y, Chang L, et al. Rescue fecal microbiota transplantation for antibiotic-associated diarrhea in critically ill patients. Crit Care. 2019;23:324.PubMedPubMedCentralCrossRef Dai M, Liu Y, Chen W, Buch H, Shan Y, Chang L, et al. Rescue fecal microbiota transplantation for antibiotic-associated diarrhea in critically ill patients. Crit Care. 2019;23:324.PubMedPubMedCentralCrossRef
171.
go back to reference Ruppé E, Martin-Loeches I, Rouzé A, Levast B, Ferry T, Timsit J-F. What’s new in restoring the gut microbiota in ICU patients? Potential role of faecal microbiota transplantation. Clin Microbiol Infect. 2018;24:803–5.PubMedCrossRef Ruppé E, Martin-Loeches I, Rouzé A, Levast B, Ferry T, Timsit J-F. What’s new in restoring the gut microbiota in ICU patients? Potential role of faecal microbiota transplantation. Clin Microbiol Infect. 2018;24:803–5.PubMedCrossRef
172.
go back to reference Antonelli M, Martin-Loeches I, Dimopoulos G, Gasbarrini A, Vallecoccia MS. Clostridioides difficile (formerly Clostridium difficile) infection in the critically ill: an expert statement. Intensive Care Med. 2020;46(2):215–24.PubMedCrossRef Antonelli M, Martin-Loeches I, Dimopoulos G, Gasbarrini A, Vallecoccia MS. Clostridioides difficile (formerly Clostridium difficile) infection in the critically ill: an expert statement. Intensive Care Med. 2020;46(2):215–24.PubMedCrossRef
173.
go back to reference Jiang ZD, Ajami NJ, Petrosino JF, Jun G, Hanis CL, Shah M, et al. Randomised clinical trial: faecal microbiota transplantation for recurrent Clostridium difficile infection - fresh, or frozen, or lyophilised microbiota from a small pool of healthy donors delivered by colonoscopy. Aliment Pharmacol Ther. 2017;45:899–908.PubMedCrossRef Jiang ZD, Ajami NJ, Petrosino JF, Jun G, Hanis CL, Shah M, et al. Randomised clinical trial: faecal microbiota transplantation for recurrent Clostridium difficile infection - fresh, or frozen, or lyophilised microbiota from a small pool of healthy donors delivered by colonoscopy. Aliment Pharmacol Ther. 2017;45:899–908.PubMedCrossRef
174.
go back to reference Allegretti JR, Kassam Z, Mullish BH, Chiang A, Carrellas M, Hurtado J, et al. Effects of fecal microbiota transplantation with oral capsules in obese patients. Clin Gastroenterol Hepatol. 2020;18:855-863.e2.PubMedCrossRef Allegretti JR, Kassam Z, Mullish BH, Chiang A, Carrellas M, Hurtado J, et al. Effects of fecal microbiota transplantation with oral capsules in obese patients. Clin Gastroenterol Hepatol. 2020;18:855-863.e2.PubMedCrossRef
175.
go back to reference Fischer M, Sipe B, Cheng Y-W, Phelps E, Rogers N, Sagi S, et al. Fecal microbiota transplant in severe and severe-complicated Clostridium difficile : a promising treatment approach. Gut Microbes. 2017;8:289–302.PubMedCrossRef Fischer M, Sipe B, Cheng Y-W, Phelps E, Rogers N, Sagi S, et al. Fecal microbiota transplant in severe and severe-complicated Clostridium difficile : a promising treatment approach. Gut Microbes. 2017;8:289–302.PubMedCrossRef
176.
go back to reference Baxter M, Ahmad T, Colville A, Sheridan R. Fatal aspiration pneumonia as a complication of fecal microbiota transplant. Clin Infect Dis. 2015;61:136–7.PubMedCrossRef Baxter M, Ahmad T, Colville A, Sheridan R. Fatal aspiration pneumonia as a complication of fecal microbiota transplant. Clin Infect Dis. 2015;61:136–7.PubMedCrossRef
177.
go back to reference Allegretti JR, Kao D, Sitko J, Fischer M, Kassam Z. Early antibiotic use after fecal microbiota transplantation increases risk of treatment failure. Clin Infect Dis. 2018;66:134–5.PubMedCrossRef Allegretti JR, Kao D, Sitko J, Fischer M, Kassam Z. Early antibiotic use after fecal microbiota transplantation increases risk of treatment failure. Clin Infect Dis. 2018;66:134–5.PubMedCrossRef
178.
go back to reference Ianiro G, Valerio L, Masucci L, Pecere S, Bibbò S, Quaranta G, et al. Predictors of failure after single faecal microbiota transplantation in patients with recurrent Clostridium difficile infection: results from a 3-year, single-centre cohort study. Clin Microbiol Infect. 2017;23:337.e1-337.e3.CrossRef Ianiro G, Valerio L, Masucci L, Pecere S, Bibbò S, Quaranta G, et al. Predictors of failure after single faecal microbiota transplantation in patients with recurrent Clostridium difficile infection: results from a 3-year, single-centre cohort study. Clin Microbiol Infect. 2017;23:337.e1-337.e3.CrossRef
Metadata
Title
The role of the microbiota in the management of intensive care patients
Authors
Piotr Szychowiak
Khanh Villageois-Tran
Juliette Patrier
Jean-François Timsit
Étienne Ruppé
Publication date
01-12-2022
Publisher
Springer International Publishing
Published in
Annals of Intensive Care / Issue 1/2022
Electronic ISSN: 2110-5820
DOI
https://doi.org/10.1186/s13613-021-00976-5

Other articles of this Issue 1/2022

Annals of Intensive Care 1/2022 Go to the issue