Abstract
Antibiotic resistance (AR) has been identified by the World Health Organization as an “increasingly serious threat to global public health.” Many mechanisms of AR have become widespread due to global selective pressures as widespread antibiotic use and/or chemotherapy. The intestine has become the primary reservoir for many resulting multidrug resistant organisms (MDROs), leading to the intestinal microbiome being dubbed the “resistome.” Intestinal decolonization has been explored as a strategy to eradicate MDROs with selective digestive tract decontamination and probiotics being notable examples with mixed results. This review focuses on fecal microbiota transplantation (FMT) and the early evidence suggesting its efficacy in decolonizing MDROs and potential mechanisms of action to reduce AR genes. Current evidence suggests FMT may have promise in restoring healthy microbial diversity and reducing AR, and clinical trials are underway to better characterize its safety and efficacy.
Keywords: fecal microbiota transplantation, Intestinal microbiota, multidrug resistant organisms, sequencing
Introduction
Bacterial Antimicrobial Resistance
The World Health Organization describes antimicrobial resistance (AR) as an “increasingly serious threat to global public health.”1 Multidrug-resistant organisms (MDROs) have been growing in prevalence2 and global distribution, accompanied by a related rise in costs, disability, and mortality.3–5 These MDROs have emerged from a variety of phyla with new species continually being identified.
Methicillin-resistant Staphylococcus aureus (MRSA) was one of the first recognized MDROs, identified in the United Kingdom in 1961.6 Since that time, MRSA has been associated with increased inpatient mortality, higher hospital costs, worse quality of life, and greater financial costs post-discharge.7,8 The impact of MRSA extends beyond the hospital setting with community-acquired MRSA (CA-MRSA) emerging as common pathogen in the United States in the early-2000s.9–11
Similarly, extended-spectrum β-lactamase (ESBL)-producing Enterobacteriaceae have become a significant source of infection in both nosocomial and community settings, especially with recognition of community-acquired ESBL urinary tract infections (UTIs)12 that can be complicated by bacteremia requiring inpatient management.13–15 Further, according to surveillance by the Centers for Disease Control and Prevention (CDC), a rising number of carbapenem-resistant Enterobacteriaceae have been identified since 2001,16 most prominently in southern Europe and Asia.17 Infections with carbapenem-resistant Enterobacteriaceae (CRE), including Klebsiella pneumoniae carbapenemase-(KPC) producing organisms, have a mortality rate of up to 71.9%, in solid organ transplant recipients.18,19
Another notable MDRO to emerge is vancomycin-resistant enterococcus (VRE), which includes Enterococcus faecium and faecalis. In one meta-analysis, patients with VRE bacteremia were approximately 2.5 times more likely to die than those with Enterococcus bacteremia susceptible to vancomycin.20 VRE bacteremia has also been associated with increased hospital costs21,22 and length of stay.23,24 There are several patient groups at risk for VRE, including the critically ill requiring intensive care unit (ICU) stay, those living in a long-term care facility, and the immunosuppressed.25 In one study of adult patients undergoing allogeneic hematopoietic cell transplantation, 10% developed VRE bacteremia with an associated mortality of 88% within 1.1 months.26
The Resistome
There are numerous mechanisms by which bacteria such as the aforementioned MDROs may become resistant to antibiotics. These include variations in antimicrobial targets of activity through changes in gene expression levels or target modification, physical barriers against antibiotics, phenotypic or behavioral changes such as expression or activation of efflux pumps, and inactivation of antimicrobial molecules. Determinants of these mechanisms are often coded by genes that reside on chromosomes or plasmids, the latter being more amenable to horizontal transmission.27 Novel mechanisms can develop as a result of spontaneous mutations and lateral gene transfer, resulting in the creation and spread of AR genes,28 which has been noted in varied niches from the soil27 to the human intestine. The human intestinal microbiota is largely comprised of bacteria, which accounts for its potential for accumulation and transmission of AR genes. Strains of AR bacteria passing through the intestine after ingestion can introduce additional AR genes29–31 with the gut serving as a reservoir for these drug-resistant opportunistic pathogens. This composite sum of AR genes in a microbial community has been dubbed the “resistome.”32,33
For the MDROs in the resistome to become pathogenic, changes in microbiome composition may account for the transition from asymptomatic colonization to development of infectious disease.33,34 Heathy intestinal microbiota is primarily comprised of a variety of anaerobic species including Lactobacillus, Faecalibacterium, and Bacteroides that demonstrate a mutualistic relationship with the body. Pathogenic species such as Escherichia spp. and Enterococcus spp. often become more abundant when there is reduced intestinal microbiome density and diversity, particularly with reduced presence of the commensal anaerobes.35,36
A well-described cause of intestinal dysbiosis is antibiotic use. Extensive research using mouse models has demonstrated effects of antibiotics on the microbiome with studies dating back to the 1970s.37,38 In a more recent study, antibiotic treatment was demonstrated to increase resistant bacteria that were then noted to translocate from the intestine to the rest of the body across the epithelial barrier.39 Ubeda, et al. showed that antibiotic administration promoted Enterococcus, including VRE, Enterobacteriaceae, and Clostridium spp. colonization of the small and large intestines.40 In another murine study, antibiotics that suppress anaerobes such as piperacillin-tazobactam and clindamycin promoted KPC colonization.41 Caballero et al. demonstrated that the intestinal colonization of one MDRO, namely VRE, did not impede that of another, in this case KPC. The researchers suggested that while these MDROs may be present in the same intestinal lumen, they grow independently through distinct mechanisms within their niches in the microbiome.42 In humans, the resistome and AR potential have been shown to vary across countries and have been suspected to be related to historically different antibiotic usage patterns in each place.32 Furthermore, patients who received anti-anaerobic antibiotic therapy have been found to have significantly more MDRO colonization, such as with VRE, than those who had received minimal anti-anaerobic treatment.43
There are additional causes of dysbiosis that increase the risk of MDRO infection in certain patient populations. In patients preparing for hematopoietic stem cell transplantation (HSCT), chemotherapy was associated with significantly increased concentration of Proteobacteria, Firmicutes, and Actinobacteria.44 The HSCT itself, in combination with antibiotics that may have been started for empiric or prophylactic therapy, reduces microbiome diversity, leading to dominance of certain bacteria. Enterococcus found to be the most common genus at 40.4% of patients in one study; of these patients, 92% had vanA gene expression consistent with VRE.45 A mouse model suggests that burn injury may also lead to dysbiosis as it was found to decrease aerobic and anaerobic species in a manner similar to antibiotic therapy.46
Decolonization as Treatment
Given the importance of the intestinal microbiome as a reservoir for AR, decolonization of the intestine has been studied as a treatment option to eradicate MDROs. Studies of the natural history of MDRO colonization suggest that spontaneous clearance of these organisms is hard to predict and frequently incomplete. In two studies of VRE colonization in oncology patients, at least 60% of patients had either intermittent or persistent presence of VRE on surveillance after at least two weeks.47,48 Similarly, in a study of renal and liver transplant recipients, only 35% of the patients were deemed to have VRE clearance, defined as negative rectal cultures on three consecutive occasions at least one week apart, with a subsequent 11% relapse rate, which prompted the authors to conclude that VRE colonization often persists for months to years in this population.49 In an ICU population, approximately 50% of patients were decolonized after two months with a median time for clearance of all MDROs of 4.8 months.50 In patients at a long-term acute care hospital, KPC colonization was lost in only 17% of patients within four weeks with more than half of the patients still colonized on readmission over nine months later.51 In another study of patients with history of positive CRE culture, mean time to CRE negativity was calculated to be 387 days with 39% of patients still with positive cultures at one year.52 A systematic review calculated via pooled analysis the median clearance time of MRSA to be 88 weeks and that of VRE to be 26 weeks.53
Selective digestive tract decontamination
Proactive MDRO decolonization has also been pursued with selective digestive tract decontamination (SDD), which involves prophylactic parenteral and systemic antibiotic therapy in the ICU setting. In one randomized control trial (RCT), SDD consisting of intravenous cefotaxime and topical application of tobramycin, colistin, and amphotericin B in the oropharynx and stomach reduced the ICU mortality rate by ~3.5% at day 28.54 Another RCT similarly showed a significantly lower ICU mortality of 15% in the SDD group versus 23% in the control as well as a significantly lower hospital mortality of 24% compared to 31% in the control.55 Oostdijk, et al. demonstrated that SDD led to decolonization in 60–80% of cases, depending on the MDRO.56 However, in a more specific look at the resistome, SDD was shown to have a high patient-specific variance on the eradication of aminoglycoside resistance genes with SDD in some patients actually increasing the copy number of resistance genes, as determined by quantitative polymerase chain reaction.57 In another study from Oostdijk, et al., gram-negative bacteria resistance to either ceftazidime, tobramycin, or ciprofloxacin increased from 5, 7, and 7%, respectively, to 15, 13, and 13% after SDD. There was a brief period during SDD where resistance levels decreased, but ceftazidime resistance in particular significantly increased during SDD itself with all resistance levels peaking months after the intervention, which was in part theorized to be a rebound effect.58 Similarly, another study revealed significantly increased colistin and tobramycin resistance as well as more frequent bacteremia among patients with ESBL-Klebsiella pneumoniae isolates treated with SDD.59 Such concerns have raised doubt over SDD and whether it may be counterproductive in the long-term.60
Probiotics
Probiotics have become a more mainstream method of altering the gut microbiome. The principle with probiotics is that the introduction of commensal bacteria, often including Lactobacillus and Bifidobacterium, would promote health61 through various mechanisms including enhanced immunity62 and infection prevention.63 In terms of MDRO decolonization, research regarding the role of probiotics has produced mixed results.
In vitro and murine studies have demonstrated Lactobacillus species being effective in limiting growth of MRSA isolates64 and decolonization of MDR E. coli.65 Early isolated case reports demonstrated a lactic acid bacteria preparation removing MRSA from a decubitus ulcer66 as well as a treatment regimen involving oral vancomycin and oral administration of Saccharomyces boulardii, a yeast species, contributing to resolution of antibiotic-associated diarrhea from MRSA.67 In another report, Roos, et. al presented a case series involving seven patients among whom five had resolution of throat and nasal carriage of MRSA with a Lactobacillus probiotic.68
There have since been several clinical trials further exploring the efficacy of probiotics for MDRO decolonization. In an open prospective trial, a probiotic drink containing two species of Lactobacillus, Bifidobacterium, and Streptococcus thermophilus reduced the nasal carriage of several organisms, including S. aureus, Streptococcus pneumoniae, β-hemolytic streptococci, and Haemophilus influenzae, by 19% though they were not necessarily drug-resistant strains.69 Conversely, in a randomized controlled trial (RCT) involving ICU patients, there was no significant difference in acquisition or loss of colonization of MDROs, including ESBL/CRE, VRE, and Pseudomonas aeruginosa, between patients who received a Lactobacillus rhamnosus probiotic versus standard of care.70 Similarly, upon evaluating a probiotic strain of E. coli, Tannock et. al concluded that the probiotic did not effectively compete against MDR E. coli for gut colonization in a group of patients from a long-term care facility.71 Likewise, a RCT in Brazil found that a Lactobacillus probiotic was not effective in decolonizing hospitalized patients with MDR gram-negative bacilli.72
In another study of patients receiving either a L. rhamnosus probiotic versus placebo for four weeks, gastrointestinal (GI) carriage of MRSA decreased by only 3% compared to 12% in the placebo group. However, three patients with VRE colonization tested negative after the four weeks of probiotics.73 In a more recent clinical trial, a L. rhamnosus probiotic reduced the odds of methicillin-sensitive S. aureus (MSSA) carriage in the gastrointestinal tract by 73% and all S. aureus presence by 83% compared to placebo; however, there was no data regarding efficacy with MRSA decolonization.74
In addition to the mixed data on the efficacy of probiotics, concerns have been raised regarding its potential dangers. This includes various gastrointestinal side effects ranging from minor ones such as abdominal cramping and nausea75 to bowel ischemia.76 More notably, Doron and Snydman found numerous reports of systemic infection; they cited 33 cases of fungemia as well as at least eight cases of Lactobacillus bacteremia and nine cases of sepsis. The Food and Drug Administration (FDA) has highlighted the particular risk in the immunosuppressed population, many of whom may be risk of intestinal dysbiosis due to chemotherapy exposure.75 Thus, despite extensive research into several microbiome restoration techniques, including SDD and probiotics, definitive solutions have yet to be found.
Fecal Microbiota Transplantation
Mechanism
Considering the limitations and risks of the aforementioned modalities, fecal microbiota transplantation (FMT) has begun to emerge as a promising approach to eradicating MDROs from the intestine. The safety and efficacy of FMT has been evaluated extensively, primarily through studies involving its role in treating recurrent Clostridium difficile infection, and appears to be related to a variety of factors. Firstly, it has been shown to increase microbial diversity. After successful FMT, the microbiome composition of a recipient becomes similar to that of the donor with a demonstrated decrease in pathogenic Enterobacteriaceae and increase in commensal bacteria, including Bacteroides and Firmicutes.77,78 Also observed after FMT via deep gene sequencing are increases in Streptococcaceae and Enterococcaceae79 as well as an overall increase in phylogenetic diversity.80 While the microbiome may diverge from that of the donor in the long-term, it appears to do so in a manner similar to the natural dynamic change seen in healthy patients77 rather than through a rebound of opportunistic pathogens as described with SDD.
Genetic studies have also revealed another mechanism by which FMT may be effective – decreasing AR genes. Millan, et al. demonstrated FMT decreasing the number of AR genes from a mean of 34.5 (± 6.7) to 12.2 (± 7.0) and decreasing the diversity of genes. This was posited to be clinically significant as the patients whose CDI did not initially respond to FMT also did not have a significant decrease in the number of AR genes; when they underwent a second round of FMT that was successful, a significant decrease in AR genes was noted as well.81 In response to this study, Jouhten et al. presented similar findings with a reduction in the number and diversity of AR genes in 20 patients post-FMT.82 Another study showed a 2.4-fold decrease in the abundance of AR genes in a patient post-FMT that persisted after one year; the resistome was also noted to have a profile similar to that of the donor.83 Metagenomic sequencing data in a more recent study demonstrated the depletion of 95 AR genes, including clinically relevant loss of quinolone, β-lactamase, ESBL, and vancomycin resistance genes in the FMT recipients. There was also acquisition of 37 AR genes from the donors though only a few of these were clinically relevant; this was thought to be expected since there are some AR genes endogenous to the commensal bacteria.84
Murine models also suggest a beneficial immune response associated with FMT. In one study, dysbiosis was established through administration of ceftriaxone, resulting in intestinal membrane compromise and increased expression of inflammatory cytokines. After FMT, the intestinal barrier was noted to recover with cytokine levels returning to normal levels after three weeks.85 Similarly, in another study, mice were subjected to eight weeks of broad-spectrum antibiotics. This resulted in a decreased presence of B and T lymphocytes as well as less active dendritic cells, particularly in the intestinal lamina propria. FMT was associated with a reversal of these findings with restoration of the lymphocytes and dendritic cell activity to that seen in mice that were not administered antibiotics.86
Specific human cases of MDRO elimination with FMT
Given FMT’s more established role in the treatment of CDI, there have been cases incidentally hinting at FMT’s possible effect on other infections in patients who were being treated for CDI. For example, in a study of patients treated with FMT for recurrent CDI, the frequency of recurrent urinary tract infections (UTIs) was found to decrease from a median of four episodes per year to one episode in the year after FMT. Furthermore, the UTIs that did occur post-FMT rarely involved bacterial isolates with any significant resistance.87 Another patient with recurrent CDI was also found to have intestinal colonization with KPC; FMT not only successfully treated the CDI but also resulted in follow-up stool cultures being negative for KPC up to 100 days post-FMT.88 In another case involving KPC colonization, an elderly patient with recurrent CDI was found to have eradication of VIM-1 producing Klebsiella oxytoca.89
Similarly, in an organ transplant recipient treated with FMT for recurrent CDI who had also had multiple episodes of VRE infections, FMT decreased Enterococcus abundance from 84% to 24% after three months and 0.2% after seven months with increased microbiota diversity.90 In another case report, a patient with quadriplegia resulting from a spinal epidural abscess who had CDI treated with FMT had a marked reduction in colonization of several MDROs, including CRE, MRSA, and MDR Acinetobacter baumannii. In total, the MDROs found on cultures decreased from 12 to 4 by 15 weeks after FMT, and while he had recurrent infections and episodes of sepsis prior to FMT, he only had one UTI in the post-FMT period from a newly acquired pathogen not previously isolated in the patient and required a 5-day course with a single antibiotic. Furthermore, despite ongoing care in the ICU post-FMT for his other conditions, the number of MDROs remained low, and he did not re-colonize with CRE, CR Pseudomonas species, or VRE. Over the following two years, he only became recolonized with MRSA in the urine and had one episode of sepsis secondary to Streptococcus pyogenes bacteremia that was treated with cefazolin.91
Mouse models have more directly explored the effect of FMT on MDROs. A study found that reintroducing a diverse intestinal microbiome through FMT to mice densely colonized with VRE resulted in clearance of VRE. They particularly noted a negative correlation between recolonization of Barnesiella, a member of the Bacteroidetes phylum, and VRE colonization.92 In another study, mice were infected with Citrobacter rodentium, which was used as a model for human enteropathogenic and enterohaemorrhagic E. coli. FMT resulted in improved innate defense, particularly with increased IL-22 gene expression, and delayed C. rodentium recolonization as well as reduced mortality with these effects persisting at least four weeks post-FMT.93 Mahieu, et al. inoculated mice with VRE and a strain of Escherichia coli producing a New Delhi metallo-β-lactamase-1 (NDM-1) and then performed FMT. After FMT, the MDRO colonization significantly decreased, particularly with VRE though in this case, the bacteria were not definitively eradicated.94 Murine studies have even looked into the role of FMT in treating tuberculosis (TB). FMT reconstituted the intestinal microbiota and reduced Mycobacterium tuberculosis (MTB) load in the lungs and spleen. Mice treated with FMT also had fewer and smaller granulomas with lesser infiltration of lymphocytes and were thus noted to have less severe cases of TB.95
Most notably, there are a growing number of cases of FMT being used solely to manage MDROs in humans. In response to Mahieu’s murine study, Leung et al. described VRE clearance in seven out of eight patients three months after FMT.96 In a more extensive prospective study, 20 patients with hematologic disorders whose intestines were colonized with various MDROs, including NDM-1+ Klebsiella pneumoniae, carbapenem-resistant Acinetobacter baumannii, and Pseudomonas aeruginosa, underwent 25 FMTs. Partial MDRO decolonization was achieved in 20/25 (80%) of cases at one month and 13/14 (93%) of cases at six months. When accounting for some of the patients who required multiple rounds of FMT, 15/20 (75%) of the participants had complete decolonization. In particular, the gene encoding carbapenemase was eradicated in 9/17 (53%) of the patients at one month and 8/9 (89%) of those at six months. After one month, 6/10 (60%) cases of NDM-1+ K. pneumoniae were eradicated while 100% of ESBL (11 patients) and oxacillinase-48 (OXA-48) (1 patient) E. coli were eradicated. Also of note in this high-risk population, there were no significant adverse events noted.97
Another report of FMT eradicating OXA-48 has been documented in an 82 year old patient with KPC whose stool cultures were negative 7 and 14 days post-FMT.98 In another case, here involving an end-stage renal disease (ESRD) patient with recurrent episodes of ESBL E. coli induced pyelonephritis, FMT resulted in the eradication of the MDRO at 2-week follow-up that persisted 12 weeks post-FMT. During this period, the patient had no recurrence of infection and was able to be actively listed for renal transplantation.99 In a 14 year old patient with a hematologic disorder who had recurrent KPC infections, FMT resulted in documented fecal clearance of MDRO 8 months later with no subsequent infections noted for 1.5 years.100 FMT has also been used in active infection; one such report found five patients with MRSA enteritis that clinically resolved along with eradication of MRSA in the feces using FMT.101 Thus, there is a growing literature highlighting the potential of FMT and suggesting its long term role in clinical practice to address MDRO colonization and infection.
Limitations and Future Considerations
Despite the cases described above, there are some limitations and concerns raised regarding FMT. There have been some documented reports of MDRO colonization persisting despite FMT or returning shortly after the procedure, both in mouse models102 and patients.103,104 One particular report presented a patient with recurrent UTIs from MDR Pseudomonas aeruginosa with both Pseudomonas and ESBL-Escherichia coli colonization; FMT resulted in Pseudomonas clearance with no more infections in the next 18 months of follow-up, but the ESBL-E. coli persisted.105 In studies of FMT in CDI, adverse effects have been described,106 including bacteremia107–109 and fatal aspiration.109,110 These appear to be rare with the more common complications being relatively mild in morbidity, such as abdominal cramping, distention, and diarrhea.106 Nevertheless, there is a limited long term outcomes data so there may be unintended consequences of FMT that are presently unknown.
As described above, the literature involving FMT in MDRO colonization and infection has consisted primarily of isolated case reports, small series of cases and small uncontrolled clinical trials. There is a lack of standardization of products used in FMT, lack of standardization of delivery, lack of characterization of the microbiome in the FMT produces, all confounding the comparability of studies. Just as with FMT to treat CDI,111 structured, randomized, controlled trials are needed to better characterize the safety and efficacy of FMT for MDRO elimination, many of which are currently underway (www.clinicaltrials.gov). Nevertheless, given the limitations of the current antibiotic development pipeline and infection prevention methods, these early signals of FMT support its investigation and potential application as a therapeutic option for MDRO colonization and infection in the future.
Acknowledgments
Source of funding: C.S.K. has funding through an Emory University Research Committee grant, as well as by the Center for AIDS Research (P30 A1050409). MHW and CSK are supported by supported by the National Center for Advancing Translational Sciences of the National Institutes of Health under Award number UL1TR000454. MHW is supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under Award Number UM1AI104681.
Footnotes
Conflict of Interest: All authors report no conflicts of interest relevant to this article.
References
- 1.World Health Organization. Antimicrobial resistance [Internet]. 2017;Available from: http://www.who.int/mediacentre/factsheets/fs194/en/
- 2.Manges AR, Steiner TS, Wright AJ. Fecal microbiota transplantation for the intestinal decolonization of extensively antimicrobial-resistant opportunistic pathogens: A review. Infect. Dis. (Auckl) 2016;48(8):587–92. [DOI] [PubMed] [Google Scholar]
- 3.Martens E, Demain AL. The antibiotic resistance crisis, with a focus on the United States. J. Antibiot. (Tokyo) 2017;70(5):520–6. [DOI] [PubMed] [Google Scholar]
- 4.Zhang R, Eggleston K, Rotimi V, et al. Antibiotic resistance as a global threat: Evidence from China, Kuwait and the United States. Global Health 2006;2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Goossens H, Ferech M, Vander Stichele R, et al. Outpatient antibiotic use in Europe and association with resistance: A cross-national database study. Lancet 2005;365(9459):579–87. [DOI] [PubMed] [Google Scholar]
- 6.Barber M Methicillin-resistant staphylococci. J Clin Pathol [Internet] 1961;14(February):385–93. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2483095&tool=pmcentrez&rendertype=abstract [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Pada SK, Ding Y, Ling ML, et al. Economic and clinical impact of nosocomial meticillin-resistant Staphylococcus aureus infections in Singapore: A matched case-control study. J Hosp Infect 2011;78(1):36–40. [DOI] [PubMed] [Google Scholar]
- 8.Cosgrove SE, Qi Y, Kaye KS, et al. The impact of methicillin resistance in Staphylococcus aureus bacteremia on patient outcomes: mortality, length of stay, and hospital charges. Infect Control Hosp Epidemiol [Internet] 2005;26(2):166–74. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15756888 [DOI] [PubMed] [Google Scholar]
- 9.Fridkin SK, Hageman JC, Morrison M, et al. Methicillin-resistant Staphylococcus aureus disease in three communities. NEnglJMed [Internet] 2005;352(1533–4406):1436–44. Available from: c:%5CKarsten%5CPDFs%5CStaphylokokken-PDFs%5CStaph-2003%5CFridkin et al.-Methicillin-resistant Staphylococcus aureus disease in three communities.pdf [DOI] [PubMed] [Google Scholar]
- 10.Moran GJ, Krishnadasan A, Gorwitz RJ, et al. Methicillin-resistant S. aureus infections among patients in the emergency department. N Engl J Med [Internet] 2006;355(7):666–74. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16914702 [DOI] [PubMed] [Google Scholar]
- 11.Stefani S, Chung DR, Lindsay JA, et al. Meticillin-resistant Staphylococcus aureus (MRSA): global epidemiology and harmonisation of typing methods. Int J Antimicrob Agents [Internet] 2012. [cited 2018 Apr 14];39(4):273–82. Available from: http://linkinghub.elsevier.com/retrieve/pii/S0924857911004687 [DOI] [PubMed] [Google Scholar]
- 12.Pitout JDD, Nordmann P, Laupland KB, et al. Emergence of Enterobacteriaceae producing extended-spectrum β-lactamases (ESBLs) in the community. J Antimicrob Chemother 2005;56(1):52–9. [DOI] [PubMed] [Google Scholar]
- 13.Pitout JD, Laupland KB. Extended-spectrum β-lactamase-producing Enterobacteriaceae: an emerging public-health concern. Lancet Infect. Dis 2008;8(3):159–66. [DOI] [PubMed] [Google Scholar]
- 14.Ben-Ami R, Schwaber MJ, Navon-Venezia S, et al. Influx of extended-spectrum beta-lactamase-producing enterobacteriaceae into the hospital. Clin Infect Dis [Internet] 2006;42(7):925–34. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16511754 [DOI] [PubMed] [Google Scholar]
- 15.Rodríguez-Baño J, Navarro MD, Romero L, et al. Bacteremia due to extended-spectrum beta -lactamase-producing Escherichia coli in the CTX-M era: a new clinical challenge. Clin Infect Dis 2006;43:1407–14. [DOI] [PubMed] [Google Scholar]
- 16.Centers for Disease Control and Prevention (CDC). Vital signs: carbapenem-resistant Enterobacteriaceae. MMWR Morb Mortal Wkly Rep [Internet] 2013. [cited 2018 Apr 14];62(9):165–70. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23466435 [PMC free article] [PubMed] [Google Scholar]
- 17.Nordmann P, Naas T, Poirel L. Global spread of Carbapenemase-producing Enterobacteriaceae. Emerg Infect Dis [Internet] 2011;17(10):1791–8. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3310682&tool=pmcentrez&rendertype=abstract [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Borer A, Saidel-Odes L, Riesenberg K, et al. Attributable mortality rate for carbapenem-resistant Klebsiella pneumoniae bacteremia. Infect Control Hosp Epidemiol [Internet] 2009;30(10):972–6. Available from: http://www.researchgate.net/publication/26771968_Attributable_Mortality_Rate_for_Carbapenem-Resistant_Klebsiella_pneumoniae_Bacteremia [DOI] [PubMed] [Google Scholar]
- 19.Perez F, Van Duin D. Carbapenem-resistant enterobacteriaceae: A menace to our most vulnerable patients. Cleve Clin J Med 2013;80(4):225–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.DiazGranados CA, Zimmer SM, Mitchel K, Jernigan JA. Comparison of Mortality Associated with Vancomycin-Resistant and Vancomycin-Susceptible Enterococcal Bloodstream Infections: A Meta-analysis. Clin Infect Dis [Internet] 2005;41(3):327–33. Available from: https://academic.oup.com/cid/article-lookup/doi/10.1086/430909 [DOI] [PubMed] [Google Scholar]
- 21.Stosor V, Peterson LR, Postelnick M, et al. Enterococcus faecium bacteremia: Does vancomycin resistance make a difference? Arch Intern Med [Internet] 1998;158(5):522–7. Available from: https://www.scopus.com/inward/record.uri?eid=2-s2.0-0032498719&partnerID=40&md5=05f022bccf2d951a6ed3c0dc3290dd79 [DOI] [PubMed] [Google Scholar]
- 22.Jung E, Byun S, Lee H, et al. 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. [DOI] [PubMed] [Google Scholar]
- 23.Lucas GM, Lechtzin N, Puryear DW, et al. Vancomycin-resistant and vancomycin-susceptible enterococcal bacteremia: comparison of clinical features and outcomes. Clin Infect Dis 1998;26(5):1127–33. [DOI] [PubMed] [Google Scholar]
- 24.Lloyd-Smith P, Younger J, Lloyd-Smith E, et al. Economic analysis of vancomycin-resistant enterococci at a Canadian hospital: Assessing attributable cost and length of stay. J Hosp Infect 2013;85(1):54–9. [DOI] [PubMed] [Google Scholar]
- 25.Olivier CN, Blake RK, Steed LL, et al. Risk of Vancomycin-Resistant Enterococcus (VRE) Bloodstream Infection Among Patients Colonized With VRE. Infect Control Hosp Epidemiol [Internet] 2008;29(5):404–9. Available from: https://www.cambridge.org/core/product/identifier/S0195941700026801/type/journal_article [DOI] [PubMed] [Google Scholar]
- 26.Tavadze M, Rybicki L, Mossad S, et al. Risk factors for vancomycin-resistant enterococcus bacteremia and its influence on survival after allogeneic hematopoietic cell transplantation. Bone Marrow Transplant 2014;49(10):1310–6. [DOI] [PubMed] [Google Scholar]
- 27.Allen HK, Donato J, Wang HH, et al. Call of the wild: Antibiotic resistance genes in natural environments. Nat. Rev. Microbiol 2010;8(4):251–9. [DOI] [PubMed] [Google Scholar]
- 28.Ochman H, Lawrence JG, Grolsman EA. Lateral gene transfer and the nature of bacterial innovation. Nature. 2000;405(6784):299–304. [DOI] [PubMed] [Google Scholar]
- 29.Salyers AA, Gupta A, Wang Y. Human intestinal bacteria as reservoirs for antibiotic resistance genes. Trends Microbiol. 2004;12(9):412–6. [DOI] [PubMed] [Google Scholar]
- 30.Simonsen GS, Haaheim H, Dahl KH, et al. Transmission of VanA-type vancomycin-resistant enterococci and vanA resistance elements between chicken and humans at avoparcin-exposed farms. Microb Drug Resist [Internet] 1998;4(4):313–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9988050 [DOI] [PubMed] [Google Scholar]
- 31.Van Braak N Den, Van Belkum A, Van Keulen M, et al. Molecular characterization of vancomycin-resistant enterococci from hospitalized patients and poultry products in the Netherlands. J Clin Microbiol 1998;36(7):1927–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Forslund K, Sunagawa S, Kultima JR, et al. Country-specific antibiotic use practices impact the human gut resistome. Genome Res 2013;23(7):1163–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.van Schaik W The human gut resistome. Philos Trans R Soc B Biol Sci [Internet] 2015;370(1670):20140087–20140087. Available from: http://rstb.royalsocietypublishing.org/cgi/doi/10.1098/rstb.2014.0087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Stecher B, Maier L, Hardt WD. “Blooming” in the gut: How dysbiosis might contribute to pathogen evolution. Nat. Rev. Microbiol. 2013;11(4):277–84. [DOI] [PubMed] [Google Scholar]
- 35.Araos R, Tai AK, Snyder GM, et al. Predominance of Lactobacillus spp. among Patients Who Do Not Acquire Multidrug-Resistant Organisms. Clin Infect Dis 2016;63(7):937–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Buffie CG, Pamer EG. Microbiota-mediated colonization resistance against intestinal pathogens. Nat. Rev. Immunol 2013;13(11):790–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Van Der Waaij D, Berghuis-de Vries JM, Lekkerkerk-Van Der Wees JEC. Colonization resistance of the digestive tract in conventional and antibiotic-treated mice. J Hyg (Lond) 1971;69(3):405–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Thijm HA, van der Waaij D. The effect of three frequently applied antibiotics on the colonization resistance of the digestive tract of mice. J Hyg (Lond) 1979;82(3):397–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yu LC-H, Shih Y-A, Wu L-L, et al. Enteric dysbiosis promotes antibiotic-resistant bacterial infection: systemic dissemination of resistant and commensal bacteria through epithelial transcytosis. AJP Gastrointest Liver Physiol [Internet] 2014;307(8):G824–35. Available from: http://ajpgi.physiology.org/cgi/doi/10.1152/ajpgi.00070.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ubeda C, Taur Y, Jenq RR, et al. Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans. J Clin Invest 2010;120(12):4332–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Perez F, Pultz MJ, Endimiani A, et al. Effect of antibiotic treatment on establishment and elimination of intestinal colonization by KPC-producing Klebsiella pneumoniae in mice. Antimicrob Agents Chemother 2011;55(6):2585–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Caballero S, Carter R, Ke X, et al. Distinct but Spatially Overlapping Intestinal Niches for Vancomycin-Resistant Enterococcus faecium and Carbapenem-Resistant Klebsiella pneumoniae. PLoS Pathog 2015;11(9):1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Donskey CJ, Chowdhry TK, Hecker MT, et al. Effect of Antibiotic Therapy on the Density of Vancomycin-Resistant Enterococci in the Stool of Colonized Patients. N Engl J Med [Internet] 2000;343(26):1925–32. Available from: http://www.nejm.org/doi/abs/10.1056/NEJM200012283432604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Montassier E, Gastinne T, Vangay P, et al. Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment Pharmacol Ther 2015;42(5):515–28. [DOI] [PubMed] [Google Scholar]
- 45.Taur Y, Xavier JB, 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] [PMC free article] [PubMed] [Google Scholar]
- 46.Kuethe JW, Armocida SM, Midura EF, et al. Fecal microbiota transplant restores mucosal integrity in a murine model of burn injury. Shock 2016;45(6):647–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Montecalvc MA, Chung M, Carraher M, et al. Natural History of Colonization with Vancomycin-Resistant Enterococcus Faecium. Infect Control Hosp Epidemiol [Internet] 1995;16(12):680–5. Available from: https://www.scopus.com/inward/record.uri?eid=2-s2.0-0029440719&partnerID=40&md5=83521b59669b90485fb2f23003722ee5 [DOI] [PubMed] [Google Scholar]
- 48.Roghmann MC, Qaiyumi S, Schwalbe R, et al. Natural history of colonization with vancomycin-resistant Enterococcus faecium. Infect Control Hosp Epidemiol [Internet] 1997. [cited 2018 Apr 15];18(10):679–80. Available from: http://www.ncbi.nlm.nih.gov/pubmed/9350457 [DOI] [PubMed] [Google Scholar]
- 49.Patel R, Allen SL, Manahan JM, et al. Natural history of vancomycin-resistant enterococcal colonization in liver and kidney transplant recipients. Liver Transplant 2001;7(1):27–31. [DOI] [PubMed] [Google Scholar]
- 50.Haverkate MR, Derde LPG, Brun-Buisson C, et al. Duration of colonization with antimicrobial-resistant bacteria after ICU discharge. Intensive Care Med 2014;40(4):564–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Haverkate MR, Weiner S, Lolans K, et al. Duration of colonization with Klebsiella pneumoniae carbapenemase-producing bacteria at long-term acute care hospitals in Chicago, Illinois. Open Forum Infect Dis 2016;3(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Zimmerman FS, Assous M V., Bdolah-Abram T, et al. Duration of carriage of carbapenem-resistant Enterobacteriaceae following hospital discharge. Am J Infect Control 2013;41(3):190–4. [DOI] [PubMed] [Google Scholar]
- 53.Shenoy ES, Paras ML, Noubary F, et al. Natural history of colonization with methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus (VRE): A systematic review. BMC Infect Dis 2014;14(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.de Smet AMGA, Kluytmans JAJW, Cooper BS, et al. Decontamination of the digestive tract and oropharynx in ICU patients. N Engl J Med [Internet] 2009;360(1):20–31. Available from: http://content.nejm.org/cgi/reprint/360/1/20.pdf%5Cnhttp://ovidsp.ovid.com/ovidweb.cgi?T=JS&PAGE=reference&D=emed9&NEWS=N&AN=2009099920%5Cnhttp://www.ncbi.nlm.nih.gov/pubmed/19118302 [DOI] [PubMed] [Google Scholar]
- 55.de Jonge E, Schultz MJ, Spanjaard L, et al. Effects of selective decontamination of digestive tract on mortality and acquisition of resistant bacteria in intensive care: a randomised controlled trial. Lancet [Internet] 2003;362(9389):1011–6. Available from: http://www.ncbi.nlm.nih.gov/pubmed/14522530 [DOI] [PubMed] [Google Scholar]
- 56.Oostdijk EAN, De smet AMGA, Kesecioglu J, et al. Decontamination of cephalosporin-resistant enterobacteriaceae during selective digestive tract decontamination in intensive care units. J Antimicrob Chemother 2012;67(9):2250–3. [DOI] [PubMed] [Google Scholar]
- 57.Buelow E, Gonzalez TB, Versluis D, et al. Effects of selective digestive decontamination (SDD) on the gut resistome. J Antimicrob Chemother 2014;69(8):2215–23. [DOI] [PubMed] [Google Scholar]
- 58.Oostdijk EA, de Smet AM, Blok HE, et al. Ecological effects of selective decontamination on resistant gram-negative bacterial colonization. Am J Respir Crit Care Med [Internet] 2010;181(5):452–7. Available from: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=19965807 [DOI] [PubMed] [Google Scholar]
- 59.Halaby T, Naiemi N Al, Kluytmans J, et al. Emergence of colistin resistance in Enterobacteriaceae after the introduction of selective digestive tract decontamination in an intensive care unit. Antimicrob Agents Chemother 2013;57(7):3224–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.van der Meer JWM, Vandenbroucke-Grauls CMJE. Resistance to selective decontamination: The jury is still out. Lancet Infect. Dis. 2013;13(4):282–3. [DOI] [PubMed] [Google Scholar]
- 61.Pamer EG. Resurrecting the intestinal microbiota to combat antibiotic-resistant pathogens. Science (80-. ). 2016;352(6285):535–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science (80- ) 2015;350(6264):1084–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Fukuda S, Toh H, Hase K, et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 2011;469(7331):543–9. [DOI] [PubMed] [Google Scholar]
- 64.Sikorska H, Smoragiewicz W. Role of probiotics in the prevention and treatment of meticillin-resistant Staphylococcus aureus infections. Int. J. Antimicrob. Agents 2013;42(6):475–81. [DOI] [PubMed] [Google Scholar]
- 65.Kumar M, Dhaka P, Vijay D, et al. Antimicrobial effects of Lactobacillus plantarum and Lactobacillus acidophilus against multidrug-resistant enteroaggregative Escherichia coli. Int J Antimicrob Agents 2016;48(3):265–70. [DOI] [PubMed] [Google Scholar]
- 66.Kimura S, Takeuchi Y, Tago H, et al. Multiple antibiotic-resistant lactic acid bacteria preparation eliminated MRSA from the decubitus of a bed-ridden elderly patient. Chin Med J (Engl) 1997;110(2):157–9. [PubMed] [Google Scholar]
- 67.Sizemore EN, Rivas KM, Valdes J, et al. Enteral vancomycin and probiotic use for methicillin-resistant Staphylococcus aureus antibiotic-associated diarrhoea. BMJ Case Rep 2012; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Roos K, Simark-Mattsson C, Grahn Håkansson E, et al. Can probiotic lactobacilli eradicate persistent carriage of meticillin-resistant Staphylococcus aureus? J. Hosp. Infect 2011;78(1):77–8. [DOI] [PubMed] [Google Scholar]
- 69.Glück U, Gebbers JO. Ingested probiotics reduce nasal colonization with pathogenic bacteria (Staphylococcus aureus, Streptococcus pneumoniae, and β-hemolytic streptococci). Am J Clin Nutr 2003;77(2):517–20. [DOI] [PubMed] [Google Scholar]
- 70.Kwon JH, Bommarito KM, Reske KA, et al. Randomized controlled trial to determine the impact of probiotic administration on colonization with multidrug-resistant organisms in critically ill patients. Infect Control Hosp Epidemiol [Internet] 2015;36(12):1451–4. Available from: 10.1017/ice.2015.195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Tannock GW, Tiong IS, Priest P, et al. Testing probiotic strain Escherichia coli Nissle 1917 (Mutaflor) for its ability to reduce carriage of multidrug-resistant E. coli by elderly residents in long-term care facilities. J Med Microbiol 2011;60(3):366–70. [DOI] [PubMed] [Google Scholar]
- 72.Salomão MCC, Heluany-Filho MA, Menegueti MG, et al. A randomized clinical trial on the effectiveness of a symbiotic product to decolonize patients harboring multidrug-resistant Gram-negative bacilli. Rev Soc Bras Med Trop 2016;49(5):559–66. [DOI] [PubMed] [Google Scholar]
- 73.Warrack S, Panjikar P, Duster M, et al. Tolerability of a probiotic in subjects with a history of methicillin-resistant Staphylococcus aureus colonisation. Benef Microbes 2014;5(4):389–95. [DOI] [PubMed] [Google Scholar]
- 74.Eggers S, Barker AK, Valentine S, et al. Effect of Lactobacillus rhamnosus HN001 on carriage of Staphylococcus aureus: results of the impact of probiotics for reducing infections in veterans (IMPROVE) study. BMC Infect Dis [Internet] 2018. [cited 2018 Apr 17];18(1):129 Available from: http://www.ncbi.nlm.nih.gov/pubmed/29540160 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Doron S, Snydman DR. Risk and Safety of Probiotics. Clin Infect Dis [Internet] 2015. [cited 2018 Jul 22];60(suppl_2):S129–34. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25922398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Besselink MG, van Santvoort HC, Buskens E, et al. Probiotic prophylaxis in predicted severe acute pancreatitis: a randomised, double-blind, placebo-controlled trial. Lancet [Internet] 2008. [cited 2018 Jul 22];371(9613):651–9. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18279948 [DOI] [PubMed] [Google Scholar]
- 77.Weingarden A, González A, Vázquez-Baeza Y, et al. Dynamic changes in short- and long-term bacterial composition following fecal microbiota transplantation for recurrent Clostridium difficile infection. Microbiome 2015;3(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Seekatz AM, Aas J, Gessert CE, et al. Recovery of the gut microbiome following fecal microbiota transplantation. MBio 2014;5(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.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). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Shahinas D, Silverman M, Sittler T, et al. Toward an Understanding of Changes in Diversity Associated with Fecal Microbiome Transplantation Based on 16S rRNA Gene Deep Sequencing. MBio [Internet] 2012. [cited 2018 Apr 17];3(5):e00338–12–e00338–12. Available from: http://www.ncbi.nlm.nih.gov/pubmed/23093385 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Millan B, Park H, Hotte N, et al. Fecal Microbial Transplants Reduce Antibiotic-resistant Genes in Patients with Recurrent Clostridium difficile Infection. Clin Infect Dis 2016;62(12):1479–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Jouhten H, Mattila E, Arkkila P, et al. Reduction of Antibiotic Resistance Genes in Intestinal Microbiota of Patients With Recurrent Clostridium difficile Infection After Fecal Microbiota Transplantation. Clin. Infect. Dis 2016;63(5):710–1. [DOI] [PubMed] [Google Scholar]
- 83.Moss EL, Falconer SB, Tkachenko E, et al. Long-term taxonomic and functional divergence from donor bacterial strains following fecal microbiota transplantation in immunocompromised patients. PLoS One 2017;12(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Leung V, Vincent C, Edens TJ, et al. Antimicrobial Resistance Gene Acquisition and Depletion Following Fecal Microbiota Transplantation for Recurrent Clostridium difficile Infection. Clin Infect Dis 2018;66(3):456–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Li M, Liang P, Li Z, et al. Fecal microbiota transplantation and bacterial consortium transplantation have comparable effects on the re-establishment of mucosal barrier function in mice with intestinal dysbiosis. Front Microbiol 2015;6(JUL). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ekmekciu I, von Klitzing E, Fiebiger U, et al. Immune responses to broad-spectrum antibiotic treatment and fecal microbiota transplantation in mice. Front Immunol 2017;8(APR). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Tariq R, Pardi DS, Tosh PK, et al. Fecal Microbiota Transplantation for Recurrent Clostridium difficile Infection Reduces Recurrent Urinary Tract Infection Frequency. Clin Infect Dis 2017;65(10):1745–7. [DOI] [PubMed] [Google Scholar]
- 88.Ponte A, Pinho R, Mota M. Fecal microbiota transplantation: is there a role in the eradication of carbapenem-resistant Klebsiella pneumoniae intestinal carriage? Rev Esp Enferm Dig [Internet] 2017;109(5):392 Available from: http://www.ncbi.nlm.nih.gov/pubmed/28196423 [DOI] [PubMed] [Google Scholar]
- 89.García-Fernández S, Morosini M-I, Cobo M, et al. Gut eradication of VIM-1 producing ST9 Klebsiella oxytoca after fecal microbiota transplantation for diarrhea caused by a Clostridium difficile hypervirulent R027 strain. Diagn Microbiol Infect Dis [Internet] 2016. [cited 2018 Apr 17];86(4):470–1. Available from: http://linkinghub.elsevier.com/retrieve/pii/S0732889316302917 [DOI] [PubMed] [Google Scholar]
- 90.Stripling J, Kumar R, Baddley JW, et al. Loss of vancomycin-resistant enterococcus fecal dominance in an organ transplant patient with Clostridium difficile colitis after fecal microbiota transplant. Open Forum Infect Dis 2015;2(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Crum-Cianflone NF, Sullivan E, Ballon-Landa G. Fecal microbiota transplantation and successful resolution of multidrug-resistant-organism colonization. J Clin Microbiol 2015;53(6):1986–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ubeda C, Bucci V, Caballero S, et al. Intestinal microbiota containing Barnesiella species cures vancomycin-resistant Enterococcus faecium colonization. Infect Immun 2013;81(3):965–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Willing BP, Vacharaksa A, Croxen M, et al. Altering host resistance to infections through microbial transplantation. PLoS One 2011;6(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Mahieu R, Cassisa V, Hilliquin D, et al. Impact of faecal microbiota transplantation on mouse digestive colonization with two extensively resistant bacteria. J. Infect 2017;75(1):75–7. [DOI] [PubMed] [Google Scholar]
- 95.Khan N, Vidyarthi A, Nadeem S, et al. Alteration in the gut microbiota provokes susceptibility to tuberculosis. Front Immunol 2016;7(NOV). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Davido B, Batista R, Fessi H, et al. Impact of faecal microbiota transplantation to eradicate vancomycin-resistant enterococci (VRE) colonization in humans. J. Infect 2017;75(4):376–7. [DOI] [PubMed] [Google Scholar]
- 97.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 [Internet] 2017;65(3):364–70. Available from: https://academic.oup.com/cid/article-lookup/doi/10.1093/cid/cix252 [DOI] [PubMed] [Google Scholar]
- 98.Lagier JC, Million M, Fournier PE, et al. Faecal microbiota transplantation for stool decolonization of OXA-48 carbapenemase-producing Klebsiella pneumoniae. J. Hosp. Infect 2015;90(2):173–4. [DOI] [PubMed] [Google Scholar]
- 99.Singh R, Nood E, Nieuwdorp M, et al. Donor feces infusion for eradication of Extended Spectrum beta‐Lactamase producing Escherichia coli in a patient with end stage renal disease. Clin Microbiol Infec 2014;20(11):O977–8. [DOI] [PubMed] [Google Scholar]
- 100.Freedman A, Eppes S. Use of Stool Transplant to Clear Fecal Colonization with Carbapenem-Resistant Enterobacteraciae (CRE): Proof of Concept. Open Forum Infect Dis 2014;1(suppl_1):S65–S65. [Google Scholar]
- 101.Wei Y, Gong J, Zhu W, et al. Fecal microbiota transplantation restores dysbiosis in patients with methicillin resistant Staphylococcus aureus enterocolitis. BMC Infect Dis 2015;15(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Myers-Morales T, Bussell KM, D’Orazio SE. Fecal transplantation does not transfer either susceptibility or resistance to food borne listeriosis in C57BL/6 and BALB/c/By mice. F1000Research [Internet] 2013;Available from: http://f1000research.com/articles/2-177/v1 [DOI] [PMC free article] [PubMed]
- 103.Jang MO, An JH, Jung SI, et al. Refractory Clostridium difficile Infection Cured With Fecal Microbiota Transplantation in Vancomycin-Resistant Enterococcus Colonized Patient. Intest Res [Internet] 2015;13(1):80–4. Available from: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4316227/pdf/ir-13-80.pdf [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Sohn KM, Cheon S, Kim YS. Can Fecal Microbiota Transplantation (FMT) Eradicate Fecal Colonization with Vancomycin-Resistant Enterococci (VRE)? Infect. Control Hosp. Epidemiol 2016;37(12):1519–21. [DOI] [PubMed] [Google Scholar]
- 105.Stalenhoef JE, Terveer EM, Knetsch CW, et al. Fecal microbiota transfer for multidrug-resistant gram-negatives: A clinical success combined with microbiological failure. Open Forum Infect Dis 2017;4(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Baxter M, Colville A. Adverse events in faecal microbiota transplant: a review of the literature. J Hosp Infect [Internet] 2016. [cited 2018 Apr 17];92(2):117–27. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26803556 [DOI] [PubMed] [Google Scholar]
- 107.Solari PR, Fairchild PG, Noa LJ, et al. Tempered enthusiasm for fecal transplant. Clin Infect Dis [Internet] 2014. [cited 2018 Apr 17];59(2):319 Available from: https://academic.oup.com/cid/article-lookup/doi/10.1093/cid/ciu278 [DOI] [PubMed] [Google Scholar]
- 108.Quera R, Espinoza R, Estay C, et al. Bacteremia as an adverse event of fecal microbiota transplantation in a patient with Crohn’s disease and recurrent Clostridium difficile infection. J Crohns Colitis [Internet] 2014;8(3):252–3. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24184170 [DOI] [PubMed] [Google Scholar]
- 109.Baxter M, Ahmad T, Colville A, et al. Fatal aspiration pneumonia as a complication of fecal microbiota transplant. Clin. Infect. Dis 2015;61(1):136–7. [DOI] [PubMed] [Google Scholar]
- 110.Kelly CR, Ihunnah C, Fischer M, et al. Fecal Microbiota Transplant for Treatment of Clostridium difficile Infection in Immunocompromised Patients. Am J Gastroenterol [Internet] 2014;109(7):1065–71. Available from: http://www.nature.com/doifinder/10.1038/ajg.2014.133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.van Nood E, Vrieze A, Nieuwdorp M, et al. Duodenal Infusion of Donor Feces for Recurrent Clostridium difficile. N Engl J Med [Internet] 2013;368(5):407–15. Available from: http://www.nejm.org/doi/10.1056/NEJMoa1205037 [DOI] [PubMed] [Google Scholar]