Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Human Microbiome Journal 16 (2020) 100071

Contents lists available at ScienceDirect

Human Microbiome Journal


journal homepage: www.elsevier.com/locate/humic

Fecal microbiota transplantation for antibiotic resistant bacteria T


decolonization
Sophie Amrane, Jean-Christophe Lagier

Aix Marseille Univ, IRD, AP-HM, MEPHI, IHU-Méditerranée Infection, Marseille, France

ARTICLE INFO ABSTRACT

Keywords: Patients colonized with antibiotic resistant bacteria are at risk of infections and spontaneous decolonization
Fecal microbiota transplantation delays are highly variable between patients. The management of these patients is therefore time-consuming;
Decolonization requires patient isolation, and cohort policies. Fecal microbiota transplantation (FMT) has been used with the
Antibiotic resistance aim of shortening this gut colonization. Here, we proposed a comprehensive literature review on FMT utilization
Vancomycin resistant Enterococcus faecium
for gut antibiotics resistant bacteria decolonization. After literature research through Pubmed indexed for
And carbapenem resistant Enterobacteriaceae
MEDLINE, we analyzed 23 studies with description of FMT utilization and analyze of gut decolonization. In total,
the data involved 197 patients, 153 of whom underwent FMT. Overall, 66.7% (102/153) of the patients were
decolonized after FMT. However, we noticed a lot of interpretation bias, such as variation in colonization de-
finition and high disparities in FMT administration modalities. Two disparities were of special interest: repeated
FMT seems to increase the effectiveness of decolonization, and gut decolonization with antibiotics before FMT
was proposed by some authors, but with too few studies to draw a conclusion. Overall, the use of FMT is a
promising perspective for intestinal decolonization, but it requires greater standardization.

1. Introduction: organization.
The management of patients colonized with antibiotics resistant
Intestinal colonization with highly drug-resistant enteric bacteria i.e bacteria is time-consuming, requires patient isolation, screening of
Vancomycin resistant Enterococcus faecium (VRE) and carbapenem re- contact patients and cohort policies. In a literature review in 2010,
sistant Enterobacteriaceae can be high, reaching up to 109 micro- Abad et al. recorded negative impact of isolation on patients well being
organism/g of feces in an animal model [1], which facilitated spread and safety [6]. They mentioned among others, higher scores of anxiety
between patients in hospital. Patients colonized with antibiotics re- or depression with HADS score elevation in proportion to the duration
sistant bacteria are at risk of infection. In a study conducted on im- of isolation, health care workers half as likely to enter patients' rooms in
munocompromised patients, VRE colonization was associated with an contact isolation, more errors in processes of care (incomplete re-
increased risk of VRE infection (adjusted odd ratio 3.61(2.01–6.47)) cording vital signs, more days with no physician or nursing notes in the
[2]. Malignant hematologic population has a high risk of both coloni- charts), patients were eight times more likely to experience supportive
zation with antibiotics resistant bacteria and translocation and then care failure, such as fall, ulcers or electrolytes abnormalities. For co-
infection with those bacteria during neutropenia and damage to the lonized patients, the duration of hospitalization increased and their
gastro-intestinal tract [3]. Amit et al. [4] retrospectively studied the access to reeducation centers are limited [7]. The economic cost of
occurrence of blood stream infection in the 45 days following carba- antibiotics resistant bacteria is estimated at $20 billion in excess med-
penem resistant -K. pneumoniae colonization portage detection. They ical spending each year in the United States [8]. In a German analysis of
did not found specific exposition risk for carbapenem resistant -K. antibiotic resistant bacteria colonization, additional costs were eval-
pneumoniae bloodstream infection, but mentioned C. difficile infection uated at €1480 for each hospitalization [9].
or use of broad-spectrum antibiotics as exposition risk for gram-nega- Spontaneous decolonization delays are highly variable between
tive roads blood stream infections. Antibiotic resistance consequences studies. After ICU admission in 8 European countries, decolonization
were evaluated in 2015 at 33,110 death a year in Europa [5]. However, occurred in a median of 4.8 month [10]. In a multicentric French study,
this number is probably overestimated compared to reality [5] and spontaneous decolonization of VRE and carbapenem resistant En-
antibiotic resistance consequences are first experienced in day care terobacteriaceae occurred for 48.2% of the patient in a median duration


Corresponding author at: Aix Marseille Univ, IRD, APHM, MEPHI, IHU - Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13005 Marseille, France..
E-mail address: jclagier@yahoo.fr (J.-C. Lagier).

https://doi.org/10.1016/j.humic.2020.100071

Available online 06 May 2020


2452-2317/ © 2020 Elsevier Ltd. This is an open access article under the CC BY license (http://creativecommons.org/licenses/BY/4.0/).
Table 1
(Part 1): Literature review: Case or study with utilization of FMT for antibiotic-resistant bacteria decolonization.
Study Design Nr Age Sex IS Stool or Rectal swab FMT for Bowel PPI Admini Stool Fresh Stool Antibiotics Eradication
positive with lavage stration route donor Frozen volume

Singh, 2014 [36] Case report 1 60 M Yes ESBL E. coli D Yes nd ND U Fresh nd No D14
Nederland D-1 in 500 mL
S. Amrane and J.-C. Lagier

Freedman, 2014 [23] Case report 1 14 F Yes CR K.pneumoniae D Yes Yes ND R Fresh 30 g No Yes
USA D-2, D-1 (nd) in 70 mL
Jang, 2015 [26] Korea Case report 1 33 M No VRE CDI No nd 1st E R Fresh 300 g Vancomycin No
C. difficile 2nd ND in 400 mL Metronidazole until
FMT
Lagier, 2015 [42] Case report 1 82 F nd CR K. pneumoniae D Yes nd NG U Frozen 50 g in Colimycin D7
France D-1 400 mL 2,5 MUI qid,
Gentamycin
100 mg qid D-1
Lombardo, 2015 [33] Retrospective analyze of 8 Nd Nd Nd VRE CDI Yes nd Capsules U Frozen 150 g Until FMT 8/8 D28
USA prospective data C. difficile D-1 treated
with
ethanol
Bilinski, 2016 [38] Case report 1 51 M Yes CR K. pneumoniae D Yes Yes ND U Fresh 100 g No D10
Poland ESBL E. coli D-1 D-1 bid in 100 mL
Dubberke, 2016 [27] Retrospective analyze of 11 75 4M No VRE CDI nd nd E U Frozen 50 g Vancomycin 8/11 at M6, 4/5 at D30
USA prospective data 7F C. difficile in 150 mL until FMT
Eysenbach, 2016 [32] Retrospective case control 16 Nd nd nd VRE CDI nd nd nd nd nd nd nd 9/9 in FMT group
USA study C. difficile 3/7 in control group
(autologous FMT)

(Part 2): Literature review: Case or study with utilization of FMT for antibiotic-resistant bacteria decolonization.

2
Study Design Nr Age Sex IS Stool or Rectal FMT for Bowel PPI Administration Stool Fresh Stool Antibiotics Eradication
swab positive with lavage route donor Frozen volume

Garcia-Fernande, 2016 Case report 1 84 F No CR K. oxytoca CDI nd nd C R nd 100g No D6


[28] Spain C. difficile in 500mL
Sohn, 2016 [29] Korea Case report 3 72, 73 and F No VRE D nd nd E R nd 100g in No No
79 C. difficile (2/3) 200mL
Bilinski, 2017 [40] Prospective 20 51 14M Yes CRE, D Yes Yes ND U nd 100g in No 15/20 D30
Poland monocentric (22–77) 6F CR P. aeruginosa, D-1 D-1 qd, 100mL
uncontrolled study VRE, ESBL D0 bid
Davido, 2017 [37] Prospective 8 69 6M No CRE D Yes Yes ND U Frozen 50g in No 2/8 D30, 3/8 M2
France monocentric (43–87) 2F VRE D-1 D-2 qd, 250mL
uncontrolled study D-1 qd
Innes, 2017 [41] UK Case report 1 36 M Yes CRE D Yes Yes NG U Frozen nd in Vancomycin 500 mg D7
D-1 H-1 100mL qid,
Neomycin 500 mg
qid, D-5 - > D-1
Lahtien, 2017 [51] Case report 1 31 F No ESBL D nd nd C nd nd nd nd Yes D42
Finland
(continued on next page)
Human Microbiome Journal 16 (2020) 100071
Table 1 (continued)

(Part 2): Literature review: Case or study with utilization of FMT for antibiotic-resistant bacteria decolonization.

Study Design Nr Age Sex IS Stool or Rectal FMT for Bowel PPI Administration Stool Fresh Stool Antibiotics Eradication
swab positive with lavage route donor Frozen volume

Ponte, 2017 [52] Case report 1 66 F nd CR K. pneumoniae CDI nd nd ND nd nd nd in nd D15


S. Amrane and J.-C. Lagier

Portugal C. difficile 50mL


Stalenhoef, 2017 [25] Case report 1 34 M No CR P. aeruginosa, D Yes nd ND U Fresh 75g in No Yes D7, No
Nederland ESBL D-1 300mL eradication of ESBL

(Part 3): Literature review: Case or study with utilization of FMT for antibiotic-resistant bacteria decolonization

Study Design Nr Age Sex IS Stool or Rectal FMT for Bowel PPI Administration Stool Fresh Stool Antibiotics Eradication
swab positive lavage route donor Frozen Volume
with

Dias, 2018 [31] Cases report 2 66 and 70 F No CRE CDI nd nd nd nd nd nd nd D30


Portugal C. difficile
Dinh, 2018 [35] Prospective 17 73 11 M 6F No CRE D Yes Yes NG U Frozen 70-100g No 3/8 CRE, 3/9 VRE D7; 4/8
France multicentric (43–87) VRE D-1 D-2 qd, in 250mL CRE, 7/8 VRE M3 (no
controlled study D-1 qd statistical difference)
Saidani, 2018 [7] Retrospective 30 59,9 24 M No CRE D Yes Yes NG U Frozen 50-100g Colimycin 8/10 D14,
France matched case (21–88) 6F A. baumanii D-5, D-1 D-1 qd, in 400mL 6MUI qid, 2/20 in control group
control study H-1, Aminoglycoside
D1-D2 200mg qid
qd D-6 - > D-1
Singh, Prospective 15 56 5M Yes ESBL D Yes nd ND U Fresh 50g No 3/15 D14 (20%),
2018 [34] monocentric (21–76) 10F (5/ D-1 in nd 2 FMT for 7 patients

3
Nederland uncontrolled study 15) with 3/7 success
- > 6/15 (40%) D14
Battipaglia, Retrospective 10 48 4M Yes CRE, D nd nd E (9), U (2) Frozen 8, 84g No 4/4 before allo HSCT, 3/6
2019 [24] monocentric study (16–64) 6F CR P. NG (1) R (8) Fresh 2 in 200mL after allo HSCT,
France aeruginosa, VRE 2 FMT for 3 patients
(time to decolonization
not precised)
Davido, 2019 [53] Prospective 8 65,5 5M nd VRE D Yes Yes NG U (2 Frozen 50g No 5/8 M1, 7/8 M3
France monocentric (50–80) 3F D-1 D-2 qd, donors) in 250mL
uncontrolled study D-1 qd
Huttner, 2019 [22] Randomized open 39 65 18 M No CRE, D No Yes NG U Frozen, 40g Colimycin 9/22 M1,
Switzerland label superiority (57–72) 21F ESBL D-1 qd, or in 80mL 2MUI qid, 5/17 in control group,
trial D0 qd Capsules Neomycin OR 1,7 (0,4–6,4)
350 mg qid,
D-6- > D-1

Nr: Number of patients, ID: Immunosuppressed patients, FMT: fecal microbiota transplantation, PPI: Proton pomp inhibitors, M: Male, F: Female, CRE: Carbapenem Resistant Enterobacteria, VRE: Vancomycine resistant
E. faecium, ESBL: extended spectrum β-lactamase Enterobacteriaceae, D: Decolonization, CDI: C. difficile infection, NG: Nasogastric, ND: Nasoduodenal, E: Enema, C: Colonoscopy, U: unrelated, R: related, nd: no data
available.
Human Microbiome Journal 16 (2020) 100071
S. Amrane and J.-C. Lagier Human Microbiome Journal 16 (2020) 100071

Table 2
Number of patients treated by FMT for gut colonization with antibiotic resistant bacteria and success rate classified by bacteria.
Colonization with N° of patients N° of eradication Delay of eradication References

Acinetobacter 2 2 (100%) D14 D30 [7,40]


CR Enterobacteriaceae 66 44 (66.7%) D7-M2 [7,22–24,28,30,31,35,37,38,40]–[42]
ESBL 52 28 (53.8%) D7-M2 [22,25,34,36,38,40,51]
CR P. aeruginosa 7 6 (85.7%) D7- D30 [24,25,40]
S. maltophila 1 1 (100%) D30 [40]
VRE 54 44 (81.5%) D10-M6 [24,26,27,29,32,33,35,37,40,53]

of 49 days (1–1091) [11]. In a German study, decolonization occurred reports or case series. In total, the data involved 197 patients, 153 of
for majority (65%) of the patients after 6 months of follow up [12], the whom underwent FMT. Only two case reports concerned an infant (14
duration of colonization was correlated with the length of hospital stay. and 16 years-old) [23,24]. Antibiotic resistant bacteria colonization
In an other study, without intervention colonization rates of carba- were various between studies, concerning VRE, carbapenem resistant
penem resistant Enterobacteriaceae and extended spectrum beta-lacta- Enterobacteriaceae, carbapenem resistant-P. aeruginosa, A. baumanii, or
mase (ESBL) Enterobacteriaceae dropped from 76.7% at one month to ESBL Enterobacteriacae (Table 2). In two case reports and one study
35.2% at twelve months of follow up [13]. [23–25] (or for five patients), antibiotics resistant bacteria were im-
To shorten this decolonization period, fecal microbiota transplan- plicated in systemic infections and not only colonization. Primary in-
tation (FMT) is currently experienced. Gut microbiota composition dication of FMT was the treatment of CDI for 40 patients in four case
analysis [14] led to a new approach in understanding of some diseases. reports and three studies [26–33].
Disruption of the gut microbiota composition has been fund during Regarding the efficiency, overall, 66.7% (102/153) of the patients
Clostridium difficile infection (CDI) and identified as the triggering factor were decolonized after FMT. In the twelve case reports, 11/15 (73%)
of the diseases [15]. In the aim to restore gut homeostasis, FMT method patients were decolonized within two months following FMT. For the
has been developed for CDI treatment, mainly for recurrent infections, eleven other studies, the decolonization rate was highly variable, from
but as a primary indication for serious infections [16]. Millan et al. [17] 20% [34] to 100% [32,33]. Dinh et al. [35] proposed comparison of
demonstrated by shotgun metagenomics and DNA microarray targeting FMT efficiency for gut decolonization of carbapenem resistant En-
354 resistant genes, a reduction of antibiotic resistant bacteria genes terobacteriaceae versus VRE, and reported no statistical differences.
after FMT (formerly done for CDI treatment). These data were con- Saidani et al. [7], in a retrospective match controlled study, reported a
firmed by Jouhten et al. [18]. The authors draw attention to the risk of decolonization rate of 80% (8/10) two weeks after FMT, compared to
antibiotic resistant bacteria genes transmission from donor to receiver, 10% (2/20) in the control group. Decolonization protocol included
donor screening for antibiotic resistant bacteria gene should be pro- three days of naso-pharyngeal decolonization with chlorhexidine, two
posed in selection criteria. bowel lavages, five days oral antibiotic decolonization and a patient
Here, we proposed a comprehensive literature review on the use of room change after FMT. Huttner at al. [22], in a randomized trial,
FMT for gut antibiotics resistant bacteria decolonization. obtained decolonization for 41% patients (9/22) versus 30% (5/17) in
the control group, OR 1.7 (0.4–6.4).
1.1. Method Only one serious adverse event was described: a woman followed
for liver cirrhosis with former recurrent encephalopathy was hospita-
We conducted a literature review from January 2000 to March 2020 lized two weeks after FMT for acute encephalopathy [22]. For the other
through Pubmed indexed for MEDLINE with “FMT AND colonization”; studies, no severe secondary effects were described after FMT.
“FMT AND decolonization”; “FMT and multi-drug resistant”; “FMT and
carbapenem resistant”; “FMT and vancomycin resistant” queries. The 2.2. Interpretation biases:
search was limited to articles written in English language and involving
humans. We selected by reading the title or the abstract of case reports Several biases limit the interpretation of these results. First, we
or clinical studies on this subject. Several cases were added by cross- observed a lack of standardization for colonization and decolonization
referencing using references cited in the articles. definitions. For colonization definition, most of the authors just men-
We obtained a selection of 26 studies with a description of the use of tioned a positive culture more or less PCR confirmation. Singh et al.,
FMT for intestinal decolonization. By reading the articles, we excluded Sohn et al., Davido et al. added some details: colonization was defined
three studies: the first because no specific evaluation of antibiotics re- by three or four consecutive positive rectal swabs one week apart
sistant bacteria colonization was performed, authors just mentioned a [29,36,37]. Davido et al. specified that the last positive swab should be
decreased of events (colonization or infection) after FMT performed for performed in the week prior to FMT [37]. Decolonization definition was
CDI infection [19], the second because VRE colonization was char- also often unclear: negative culture more or less negative PCR result.
acterized by metagenomic relative abundance (which declined after Some authors added details: with two or three negative rectal swab at
FMT) [20], and the third because FMT was proposed for MRSA enteritis one week interval [7,24,38,39]. Secondly, the modes of evaluation and
treatment without indication on gut colonization with enteric anti- length of follow up were strongly heterogeneous in the different studies.
biotics resistant bacteria [21]. As an example in prospective studies, Billinski et al. and Singh et al.
proposed follow up until one month after FMT [34,40], Davido at al.
2. Results: and Dinh at al. until three months [35,37,39], and Huttner et al. after
six months [22]. Thirdly, in the different studies reviewed, FMT ad-
2.1. FMT indication, efficiency and side effects ministration was very diverse with volume of stool from 30 to 300 g.
Stools were more often reconstituted with a sterile saline solution.
Overall, we analyzed 23 studies with a description of the use of FMT Product was often frozen and donors unrelated. Preferred route of ad-
and an analysis of intestinal decolonization (Table 1). Articles reviewed ministration was naso-duodenal or nasogastric. Two authors used an
were highly heterogeneous; they ranged from 2014 to 2019. They have oral administration with capsules: in Huttner et al. [22] study, two
all been conducted in rich countries: Europa, USA, Israel or Korea. We centers administrated FMT though lyophilized capsules. Lombardo
found only one randomized controlled trial [22] and twelve cases et al. [33] used a preparation called SER-109: stools from donors were

4
S. Amrane and J.-C. Lagier Human Microbiome Journal 16 (2020) 100071

treated with ethanol solution with the aim of promoting sporulation


Estimated Study Completion Date
and selected Firmicutes bacteria, which were then administrated in
capsules. Other parts of the protocol varied, such as bowel preparation,
mentioned in thirteen studies, and often performed the day prior to
FMT. Only one author proposed two bowel lavages (one five days prior
to FMT and one the day before) [7]. Fasting before the FMT was spe-
cified only in two studies [33,41]. If notified, authors used proton pump
March

2019 June

2022 June
2023 Nov
inhibitor before FMT administration. Saidani et al. administered also
Dec

2019 Dec

2020 Dec

2023 Dec
Jun

2025 Jun
2024 Jan

2019 Fev
2019 Oct
2020 Oct
Jul

150 mL bicarbonate sodium before FMT [7], Innes et al. mentioned


2020
2018
2022
2021

administration of metoclopramide one hour before FMT for its proki-


netic effect [41]. Finally, antibiotics for decolonization are used by
some teams before FMT administration [7,22,41,42], other antibiotics
were for C. difficile treatment.
Estimated/Actual Study

2.3. FMT implication in treatment of other colonization sites


2019 March

2019 March

2020 March
started Date

2017 June

Among the articles reviewed, some authors analyzed other coloni-


2022 Sept
2014 Nov

2019 Nov
Aug

2017 Aug
Dec

2019 Dec
Fev

2018 Fev

2018 Fev
Jul

zation sites: Singh et al. proposed swabbing of pelvic region and throat
2015
2016
2016
2016

[36], Stalenhoef et al. and Saidani et al. proposed swabbing of the skin,
throat and a sample of urine [7,25]. In another study, Singh et al.
Switzerland

Hong Kong

sampled urine [34]. However, the results of colonization or decoloni-


Belgium
Country

Canada

Canada
France

zation of these other locations have not been reported in the articles.
Israel
Israel

Israel
USA

USA
USA

USA

USA

USA

Gut decolonization could have an impact on urinary infections fre-


quency. Stripling et al. mentioned no VRE infection relapse (urinary
Fecal Transplant, a Hope to Eradicate Colonization of Patient Harboring eXtreme Drug Resistant

A Trial of Encapsulated Fecal Microbiota for Vancomycin Resistant Enterococcus Decolonization

tract infection) after FMT (vesicle device was not mentioned in the
Fecal Microbiota Transplantation for Eradication of Carbapenem-resistant Enterobacteriaceae

Decolonization of Gram-negative Multi-resistant Organisms (MDRO) With Donor Microbiota

article) [20]. In 2014, Sing et al. proposed the use of FMT for gut de-
Eradication of Antibiotic-resistant Bacteria Through Antibiotics and Fecal Bacteriotherapy

FMT for multidrug resistant organisms Colonization After Infection in Renal Transplant

colonization of an ESBL-E. coli [36]. One patient had recurrent urinary


Autologous Fecal Microbiota Transplantation to Prevent Antibiotic Resistant Bacteria

Effectiveness of Fecal Flora Alteration for Eradication of Carbapenemase-producing

tract infection with ESBL microorganism with severe renal graft failure.
After FMT, he presented no more urinary tract infection. In a retro-
Fecal Microbiota Transplantation for Carbapenem-resistant Enterobacteriaceae
FMT for multidrug resistant organisms Colonization in Solid Organ Transplant

spective case controlled study, Tariq et al. analyzed urinary tract in-
fection frequency before and after FMT (for recurrent CDI treatment).
Phase II Trial of Fecal Microbiota Transplant for VRE and CRE Patients

They demonstrated a reduction in the incidence of these infections with


a median of four infections in the year preceding the FMT and one in
the year following the FMT [43]. In a recent publication, Hocquart et al.
presented a case of a woman with inflammatory bowel disease and
Fecal Microbiota Transplantation for Eradication of CRE

recurrent urinary tract infection, eight months after FMT she had no
recurrence of urinary infection [44].
FMT for Multidrug Resistant Organism Reversal

Fecal Microbiota Transplantation for CRE/VRE

Fecal Transplant for MDRO Decolonization

2.4. Modality of FMT protocol that could modify success rates

Among the decolonization procedures described, some author pro-


Enterobacteriaceae Colonization

posed oral antibiotics before FMT. In a literature review, Cheng et al.


[45] reported former uses of oral antibiotics for gut VRE decolonization
(more or less probiotic administration) with success rates ranging from
17.8% to 100%. In their work, the authors were the first to propose a
bowel lavage during the decontamination procedure; they obtained
VRE decolonization for 75% of their patients. Machuca et al. [46] de-
Colonization

Colonization
Recipients

monstrated a reduction of global mortality and a reduction carbapenem


Bacteria?
Current interventional study on FMT and gut decolonization.

(FMT)

resistant -K. pneumoniae infection with oral decontamination with


Title

aminoglycosides. Among the articles reviewed, two case reports


[41,42] proposed oral antibiotics for gut carbapenem resistant En-
terobacteriaceae decolonization before FMT, procedure succeeded in
Single group open label trial

Single group open label trial

Single group open label trial

Single group open label trial


Type of interventional study

Randomized open label trial

Randomized open label trial

Randomized open label trial

Randomized open label trial

Randomized open label trial


Randomized open label trial
Non randomized open label
Randomized blinded trial

Randomized blinded trial

Randomized blinded trial

Randomized blinded trial

both cases. Saidani et al. used antibiotics (Colimycin and Aminogly-


cosides) in their protocol with success for 8/10 (80%) patients [7].
Huttner et al. also included antibiotics (Colimycin and Neomycin) in
their protocol, eradication was obtained for 9/22 (41%) of the patients
[22].
Three authors mentioned more than one FMT for their patients with
trial

increased decolonization rate. Singh et al. [34] treated 15 patients, they


obtained 3/15 (20%) decolonization after one FMT, seven patients
Clinical trial number

underwent two FMT with a success rate of 3/7 (43%), bringing the
overall success rate to 6/15 (40%). Battipaglia et al. [24] treated ten
NCT02312986
NCT02472600
NCT02816437
NCT02922816

NCT03029078

NCT03061097

NCT03063437
NCT03167398
NCT03391674

NCT03479710

NCT03643887
NCT03802461

NCT04146337
NCT04181112
NCT04188743

patients, they obtained 5/10 (50%) decolonization after one FMT, 2/3
(67%) after two FMT with an overall success rate at 7/10 (70%). Sai-
Table 3

dani et al. [7], treated ten patients, they obtained 4/10 (40%) decolo-
nization after one FMT, 4/5 (80%) after two FMT with and overall

5
S. Amrane and J.-C. Lagier Human Microbiome Journal 16 (2020) 100071

success rate at 8/10 (80%) patients. Repeating FMT could increase the strategies for ESBL/carbapenem-resistant Enterobacteriaceae carriage: Systematic
chances of decolonization by a factor of almost two. review and meta-analysis. J Antimicrob Chemother 2016;71(10).
[14] Qin J, et al. A human gut microbial gene catalogue established by metagenomic
sequencing. Nature 2010;464(7285):59–65.
3. Conclusion: [15] Seekatz AM, Rao K, Santhosh K, Young VB. Dynamics of the fecal microbiome in
patients with recurrent and nonrecurrent Clostridium difficile infection. Genome
Med 2016;8(1):47.
We reviewed 23 articles on the use of FMT for gut decolonization. [16] Hocquart M, et al. Early Faecal Microbiota Transplantation Improves Survival in
To date, only one randomized controlled trial has been published. Severe Clostridium difficile Infections. Clin Infect Dis 2017.
Fifteen other trials results are expected (Table 3), their results will [17] Millan B, et al. Fecal microbial transplants reduce antibiotic-resistant genes in pa-
tients with recurrent clostridium difficile infection. Clin Infect Dis
contribute to expend our knowledge on this problematic. Some of these 2016;62(12):1479–86.
trials propose FMT via oral capsule administration, this route of ad- [18] Jouhten H, Mattila E, Arkkila P, Satokari R. Reduction of antibiotic resistance genes
ministration has been used by Huttner et al. for 16/22 patients [22]. in intestinal microbiota of patients with recurrent clostridium difficile infection
after fecal microbiota transplantation. Clin Infect Dis 2016;63(5):710–1.
Oral capsules administration has former been evaluated for CDI treat-
[19] Crum-Cianflone NF, Sullivan E, Ballon-Landa G. Fecal microbiota transplantation
ment [47] with non-inferiority compare to FMT administration via and successful resolution of multidrug-resistant-organism colonization. J Clin
colonoscopy. Repeated FMT appears to be necessary in some cases for Microbiol 2015;53(6):1986–9.
the eradication of antibiotic-resistant bacteria in the intestine, the use [20] Stripling J, et al. Loss of vancomycin-resistant enterococcus fecal dominance in an
organ transplant patient with clostridium difficile colitis after fecal microbiota
of capsules could facilitate repeated administration. transplant. Open Forum Infect Dis 2015;2(2):ofv078.
Another area of research on the decolonization of antibiotic-re- [21] Wei Y, et al. Fecal microbiota transplantation restores dysbiosis in patients with
sistant bacteria with microbiota manipulation could be the adminis- methicillin resistant Staphylococcus aureus enterocolitis. BMC Infect Dis
2015;15(1):265.
tration of specific bacteria. Ubeda et al. explored by 16S rRNA gene [22] Huttner BD, et al. A 5-day course of oral antibiotics followed by faecal transplan-
sequencing bacteria population before and after reintroduction of a tation to eradicate carriage of multidrug-resistant Enterobacteriaceae: a rando-
diverse intestinal microbiota in a mice model. Barnesiella species were mized clinical trial. Clin Microbiol Infect 2019.
[23] Freedman A, Eppes S. Use of stool transplant to clear fecal colonization with car-
associated with VRE clearance [1]. Lombardo et al. administrated a bapenem-resistant enterobacteraciae (CRE): proof of concept. Open Forum Infect
consortium of bacteria (Firmicutes) to treat patients with recurrent C. Dis 2014.
difficile infection and observed eradication of VRE carriage for 8/8 [24] Battipaglia G, et al. Fecal microbiota transplantation before or after allogeneic
hematopoietic transplantation in patients with hematological malignancies carrying
patients [33]. Association of selected bacteria have been tested with multidrug-resistance bacteria. Haematologica 2019. p. haematol.2018.198549.
success for C. difficile infection treatment [48,49] and tentative to find [25] Stalenhoef JE, et al. Fecal microbiota transfer for multidrug-resistant gram-nega-
the good consortium of bacteria are in progress [50]. The same tenta- tives: a clinical success combined with microbiological failure. Open Forum Infect
Dis 2017;4(2):ofx047.
tive could be explored for the eradication of antibiotic-resistant bac-
[26] Jang M-O, An JH, Jung S-I, Park K-H. Refractory Clostridium difficile infection
teria. cured with fecal microbiota transplantation in vancomycin-resistant enterococcus
colonized patient. Intest Res 2015;13(1):80–4.
Competing interest [27] Dubberke ER, et al. Clearance of vancomycin-resistant enterococcus concomitant
with administration of a microbiota-based drug targeted at recurrent Clostridium
difficile infection. Open Forum Infect Dis 2016;3(3):ofw133.
None to declare. [28] García-Fernández S, et al. Gut eradication of VIM-1 producing ST9 Klebsiella oxy-
toca after fecal microbiota transplantation for diarrhea caused by a Clostridium
difficile hypervirulent R027 strain. Diagn Microbiol Infect Dis 2016;86(4):470–1.
Funding [29] Sohn KM, Cheon S, Kim Y-S. Can fecal microbiota transplantation (FMT) Eradicate
fecal colonization with vancomycin-resistant enterococci (VRE)? Infect Control
None to declare. Hosp Epidemiol 2016;37(12):1519–21.
[30] Ponte A, Pinho R, Mota M. Fecal microbiota transplantation: Is there a role in the
eradication of carbapenem-resistant Klebsiella pneumoniae intestinal carriage?
References Revista Espanola de Enfermedades Digestivas 2017;109(5).
[31] Dias C, Pipa S, Duarte-Ribeiro F, Mota M. Fecal microbiota transplantation as a
potential way to eradicate multiresistant microorganisms. IDCases 2018;13:e00432.
[1] Ubeda C, et al. Intestinal microbiota containing Barnesiella species cures vanco-
[32] Eysenbach L, et al. Clearance of vancomycin-resistant enterococcus colonization
mycin-resistant Enterococcus faecium colonization. Infect Immun
with fecal microbiota transplantation among patients with recurrent Clostridium
2013;81(3):965–73.
difficile infection. Open Forum Infect Dis 2016;3(suppl_1).
[2] Russell DL, et al. Outcomes of colonization with MRSA and VRE among liver
[33] Lombardo M-J, et al. Vancomycin-resistant enterococcal iters diminish among pa-
transplant candidates and recipients. Am J Transplant 2008;8(8):1737–43.
tients with recurrent Clostridium difficile infection after administration of SER-109,
[3] Taur Y, Pamer EG. The intestinal microbiota and susceptibility to infection in im-
a novel microbiome agent. Open Forum Infect Dis 2015;2(suppl_1).
munocompromised patients. Curr Opin Infect Dis 2013;26(4):332–7.
[34] Singh R, et al. Fecal microbiota transplantation against intestinal colonization by
[4] Amit S, Mishali H, Kotlovsky T, Schwaber MJ, Carmeli Y. Bloodstream infections
extended spectrum beta-lactamase producing Enterobacteriaceae: a proof of prin-
among carriers of carbapenem-resistant Klebsiella pneumoniae: etiology, incidence
ciple study. BMC Res Notes 2018;11(1):190.
and predictors. Clin Microbiol Infect 2015;21(1):30–4.
[35] Dinh A, et al. Clearance of carbapenem-resistant Enterobacteriaceae vs vancomycin-
[5] Raoult D, Leone M, Roussel Y, Rolain J-M. Attributable deaths caused by infections
resistant enterococci carriage after faecal microbiota transplant: a prospective
with antibiotic-resistant bacteria in France. Lancet Infect Dis 2019;19(2):128–9.
comparative study. J Hosp Infect 2018;99(4):481–6.
[6] Abad C, Fearday A, Safdar N. Adverse effects of isolation in hospitalised patients: a
[36] Singh R, et al. Donor feces infusion for eradication of extended spectrum beta-
systematic review. J Hosp Infect 2010;76(2):97–102.
lactamase producing Escherichia coli in a patient with end stage renal disease. Clin
[7] Saïdani N, et al. Faecal microbiota transplantation shortens the colonisation period
Microbiol Infect 2014;20(11):O977–8.
and allows re-entry of patients carrying carbapenamase-producing bacteria into
[37] Davido B, et al. Is faecal microbiota transplantation an option to eradicate highly
medical care facilities. Int J Antimicrob Agents 2019;53(4):355–61.
drug-resistant enteric bacteria carriage? J Hosp Infect 2017;95(4):433–7.
[8] Marston HD, Dixon DM, Knisely JM, Palmore TN, Fauci AS. Antimicrobial
[38] Biliński J, et al. Fecal Microbiota Transplantation Inhibits Multidrug-Resistant Gut
Resistance. JAMA 2016;316(11):1193–204.
Pathogens: Preliminary Report Performed in an Immunocompromised Host. Arch
[9] Engler-Hüsch S, Heister T, Mutters NT, Wolff J, Kaier K. In-hospital costs of com-
Immunol Ther Exp (Warsz) 2016;64(3):255–8.
munity-acquired colonization with multidrug-resistant organisms at a German
[39] Davido B, et al. Fecal microbiota transplantation to eradicate vancomycin-resistant
teaching hospital. BMC Health Serv Res 2018;18(1):737.
enterococci colonization in case of an outbreak. Médecine Mal Infect 2018.
[10] Haverkate MR, Derde LPG, Brun-Buisson C, Bonten MJM, Bootsma MCJ. Duration
[40] Bilinski J, et al. Fecal microbiota transplantation in patients with blood disorders
of colonization with antimicrobial-resistant bacteria after ICU discharge. Intensive
inhibits gut colonization with antibiotic-resistant bacteria: results of a prospective,
Care Med 2014;40(4):564–71.
single-center study. Clin Infect Dis 2017;65(3):364–70.
[11] Davido B, et al. Germs of thrones - spontaneous decolonization of carbapenem-
[41] Innes AJ, et al. Faecal microbiota transplant: a novel biological approach to ex-
resistant enterobacteriaceae (CRE) and vancomycin-resistant enterococci (VRE) in
tensively drug-resistant organism-related non-relapse mortality. Bone Marrow
Western Europe: is this myth or reality? Antimicrob Resist Infect Control
Transplant 2017;52(10):1452–4.
2018;7(1):100.
[42] Lagier JC, Million M, Fournier PE, Brouqui P, Raoult D. Faecal microbiota trans-
[12] Lübbert C, et al. Long-term carriage of Klebsiella pneumoniae carbapenemase–2-
plantation for stool decolonization of OXA-48 carbapenemase-producing Klebsiella
producing K pneumoniae after a large single-center outbreak in Germany. Am J
pneumoniae. J Hosp Infect 2015;90(2):173–4.
Infect Control 2014;42(4):376–80.
[43] Tariq R, Pardi DS, Tosh PK, Walker RC, Razonable RR, Khanna S. Fecal Microbiota
[13] Bar-Yoseph H, Hussein K, Braun E, Paul M. Natural history and decolonization
Transplantation for Recurrent Clostridium difficile Infection Reduces Recurrent

6
S. Amrane and J.-C. Lagier Human Microbiome Journal 16 (2020) 100071

Urinary Tract Infection Frequency. Clin Infect Dis 2017;65(10):1745–7. difficile-associated diarrhoea: results from a case series of 55 patients in Denmark
[44] Hocquart M, Pham T, Kuete E, Tomei E, Lagier JC, Raoult D. Successful fecal mi- 2000–2012. Clin Microbiol Infect 2015;21(1):48–53.
crobiota transplantation in a patient suffering from irritable bowel syndrome and [49] Petrof EO, et al. Stool substitute transplant therapy for the eradication of
recurrent urinary tract infections. Open Forum Infect Dis 2019;6(10). Clostridium difficile infection: ‘RePOOPulating’ the gut. Microbiome 2013;1(1):3.
[45] Cheng VCC, et al. Decolonization of gastrointestinal carriage of vancomycin-re- [50] Amrane S, et al. Metagenomic and culturomic analysis of gut microbiota dysbiosis
sistant Enterococcus faecium: case series and review of literature. BMC Infect Dis during Clostridium difficile infection. Sci Rep 2019;9(1):12807.
2014;14(1):514. [51] Lahtinen P, et al. Faecal microbiota transplantation in patients with Clostridium
[46] Machuca I, et al. Oral decontamination with aminoglycosides is associated with difficile and significant comorbidities as well as in patients with new indications: A
lower risk of mortality and infections in high-risk patients colonized with colistin- case series. World J Gastroenterol 2017;23(39):7174–84.
resistant, KPC-producing Klebsiella pneumoniae. J Antimicrob Chemother [52] Ponte A, Pinho R, Mota M. Fecal microbiota transplantation: Is there a role in the
2016;71(11). eradication of carbapenem-resistant Klebsiella pneumoniae intestinal carriage? Rev
[47] Kao D, et al. Effect of oral capsule- vs colonoscopy-delivered fecal microbiota Española Enfermedades Dig 2017;109(5):392.
transplantation on recurrent Clostridium difficile infection: a randomized clinical [53] Davido B, et al. Fecal microbiota transplantation to eradicate vancomycin-resistant
trial. JAMA 2017;318(20):1985–93. enterococci colonization in case of an outbreak. Médecine Mal Infect
[48] Tvede M, Tinggaard M, Helms M. Rectal bacteriotherapy for recurrent Clostridium 2019;49(3):214–8.

You might also like