Skip to main content
Erschienen in: Critical Care 1/2016

Open Access 01.12.2016 | Research

Probiotic and synbiotic therapy in critical illness: a systematic review and meta-analysis

verfasst von: William Manzanares, Margot Lemieux, Pascal L. Langlois, Paul E. Wischmeyer

Erschienen in: Critical Care | Ausgabe 1/2016

Abstract

Background

Critical illness is characterized by a loss of commensal flora and an overgrowth of potentially pathogenic bacteria, leading to a high susceptibility to nosocomial infections. Probiotics are living non-pathogenic microorganisms, which may protect the gut barrier, attenuate pathogen overgrowth, decrease bacterial translocation and prevent infection. The purpose of this updated systematic review is to evaluate the overall efficacy of probiotics and synbiotic mixtures on clinical outcomes in critical illness.

Methods

Computerized databases from 1980 to 2016 were searched. Randomized controlled trials (RCT) evaluating clinical outcomes associated with probiotic therapy as a single strategy or in combination with prebiotic fiber (synbiotics). Overall number of new infections was the primary outcome; secondary outcomes included mortality, ICU and hospital length of stay (LOS), and diarrhea. Subgroup analyses were performed to elucidate the role of other key factors such as probiotic type and patient mortality risk on the effect of probiotics on outcomes.

Results

Thirty trials that enrolled 2972 patients were identified for analysis. Probiotics were associated with a significant reduction in infections (risk ratio 0.80, 95 % confidence interval (CI) 0.68, 0.95, P = 0.009; heterogeneity I 2 = 36 %, P = 0.09). Further, a significant reduction in the incidence of ventilator-associated pneumonia (VAP) was found (risk ratio 0.74, 95 % CI 0.61, 0. 90, P = 0.002; I 2 = 19 %). No effect on mortality, LOS or diarrhea was observed. Subgroup analysis indicated that the greatest improvement in the outcome of infections was in critically ill patients receiving probiotics alone versus synbiotic mixtures, although limited synbiotic trial data currently exists.

Conclusion

Probiotics show promise in reducing infections, including VAP in critical illness. Currently, clinical heterogeneity and potential publication bias reduce strong clinical recommendations and indicate further high quality clinical trials are needed to conclusively prove these benefits.
Hinweise
An erratum to this article is available at http://​dx.​doi.​org/​10.​1186/​s13054-017-1622-4.
A comment to this article is available at http://​dx.​doi.​org/​10.​1186/​s13054-016-1565-1.

Background

Critical illness is characterized by a loss of commensal flora and an overgrowth of potentially pathogenic bacteria, leading to a high susceptibility to acquired nosocomial infections [1, 2]. Further, sepsis following infection is still a leading cause of death worldwide [3]. The U.S. Centers for Disease Control indicates death rates from critical illness/sepsis have increased at a rate greater than any other common cause of mortality in the last year for which data were available [4]. Thus, therapies to reduce the risk and incidence of infection and sepsis in critical illness are urgently needed.
According to the World Health Organization and the Food and Agriculture Organization, probiotics are living non-pathogenic microorganisms, which have demonstrated well-documented beneficial health effects administered in optimum amounts in the prevention and treatment of several disease states [5]. So far, several mechanisms by which probiotics may exert beneficial effects have been described, including modification of the gut flora by inducing host cell antimicrobial peptides, release of antimicrobial factors, suppression of the immune cell proliferation, stimulation of mucus and IgA production, anti-oxidative activity, inhibition of epithelial cell nuclear factor kappa B activation, and other potentially vital gut epithelial barrier protective effects [68]. As the gut is hypothesized to play a central role in the progression of critical illness, sepsis and multiple organ dysfunction syndrome [9], maintenance of the gut barrier and a healthy gut microbiome, potentially via reintroduction of commensal bacteria (probiotic therapy), may be essential to optimizing outcomes in critically ill patients.
According to current literature, the efficacy of probiotics in the prevention of infectious complications has been extensively evaluated in many animal studies and clinical trials in heterogenous intensive care unit (ICU) patient populations. These studies suggest that probiotics may reduce the incidence of infection, particularly ventilator-associated pneumonia (VAP) [10], which is a common serious complication in intubated, mechanically ventilated patients [11]. Nonetheless, the effect of probiotics on the prevention of VAP still remains controversial and inconclusive [1217]. In fact, its effect depends on the patient population and the probiotic strain studied. Despite the outcome benefits of probiotics therapy, recent guidelines have been unable to make a definitive recommendation for the routine use of probiotics in ICU patients. To date, these guidelines have suggested the use of probiotic therapy in select medical and surgical patient populations in whom trials have documented safety and clinical benefits [18, 19].
Over the last few years, several systematic reviews and meta-analyses have evaluated the effects of probiotics in critically ill patients [1217]. In 2012, after aggregating 11 trials that reported on infections [14], we demonstrated that probiotics may reduce infections, including the incidence of VAP, although the effect on VAP was not statistically significant given the available data. Moreover, probiotics were associated with a trend toward reduced ICU mortality, but did not influence hospital mortality. Since our last systematic review and meta-analyses, seven new trials of probiotic therapy have been published [2026]. Further, to date, no recent meta-analysis has examined the effect of probiotic versus synbiotic (probiotic and prebiotic fiber) therapy. Finally, a Canadian survey [27] on the use of probiotics as a prophylactic strategy for VAP showed that most Canadian ICU pharmacists have used probiotics at least once, although routine use is considered controversial and considerable practice variability exists. Thus, any increased understanding that the newly published trials can yield will be vital to clarifying clinical probiotic use in the ICU and areas in need of future research focus.
Therefore, as probiotic use in the ICU remains widespread and controversial, current guidelines are not conclusive, and with a significant number of new trials of probiotic use published recently we conducted a comprehensive systematic review and meta-analysis of probiotic and synbiotic use in critically ill patients. Our aim was to elucidate the overall efficacy of probiotics, as a single strategy or in combination with fiber therapy (synbiotics) on relevant clinical outcomes, particularly infection and VAP, in adult critically ill patients.

Methods

Search strategy and study identification

A literature search was conducted in MEDLINE, Embase, CINAHL, the Cochrane Central Register of Controlled Trials and the Cochrane Database of Systematic Reviews to identify all relevant randomized controlled trials (RCTs) published between 1980 and April 2016. The literature search used broad search terms containing “randomized,” “clinical trial,” “nutrition support,” “enteral nutrition”, “probiotics,” and “synbiotics”. No language restrictions were applied. Personal files and reference lists of relevant review articles were also reviewed.

Eligibility criteria

We included trials with the following characteristics:
1.
Type of study: randomized controlled parallel group trials
 
2.
Population: adult (≥18 years of age) critically ill patients. If the study population was unclear, we considered a mortality rate higher than 5 % in the control group to be consistent with critical illness
 
3.
Intervention: Probiotics alone or associated with prebiotics (synbiotics) compared to a placebo
 
4.
Outcomes: pre-specified clinical outcomes in ICU patients such as infectious complications, VAP, mortality, ICU and hospital length of stay (LOS), and diarrhea
 
We excluded trials that reported only nutrition, biochemical, metabolic, or immunologic outcomes. Data published in abstract form were included only if additional information about the study design was obtained from the authors. The methodological quality of the included trials was assessed in duplicate by two reviewers independently using a data abstraction form with a scoring system from 0 to 14 according to the following criteria:
1.
The extent to which randomization was concealed
 
2.
Blinding
 
3.
Analysis based on the intention-to-treat (ITT) principle
 
4.
Comparability of groups at baseline
 
5.
Extent of follow up
 
6.
Description of treatment protocol
 
7.
Co-interventions
 
8.
Definition of clinical outcomes
 
Consensus between both reviewers on the individual scores of each of the categories was obtained. We attempted to contact the authors of included studies and requested additional information not contained in published articles. We designated studies as level I if all of the following criteria were fulfilled: concealed randomization, blinded outcome adjudication and an ITT analysis, all which are the strongest methodological tools to reduce bias. A study was considered as level II if any one of the above-described characteristics were unfulfilled.

Data synthesis

All analyses, except the test for asymmetry, were conducted using RevMan 5.3 (Cochrane IMS, Oxford, UK) with a random effects model. We combined data from all trials to estimate the overall weighted mean difference (WMD) with 95 % confidence intervals for LOS data the pooled risk ratio (RR) with 95 % confidence intervals (CIs) for the incidence of infections and mortality, and diarrhea. WMDs were estimated by the inverse variance approach and pooled RRs were calculated using the Mantel-Haenszel estimator. The random effects model of DerSimonian and Laird was used to estimate variances for the Mantel-Haenszel and inverse variance estimators [28]. RRs were undefined and excluded for studies with no event in either arm. Heterogeneity was tested by a weighted Mantel-Haenszel χ2 test and quantified by the I 2 statistic as implemented in RevMan. Differences between subgroups were analyzed using the test of subgroup differences described by Deeks et al., and the results expressed using the P values. We considered P <0.05 to be statistically significant and P <0.10 as an indicator of trends. Funnel plots were used to assess the possibility of publication bias and the Egger regression test was used to measure funnel plot asymmetry [29].

Clinical outcomes

Overall infections were the primary outcome for this meta-analysis. Secondary outcomes were VAP, mortality, ICU and hospital LOS, and finally diarrhea. We used definitions of infections as defined by the authors in their original articles. From all trials, we combined hospital mortality where reported. Mortality specified at either 28 days or 90 days was not considered as ICU or hospital mortality, respectively. Nonetheless, if the mortality time frame was not specified as either ICU or hospital, it was presumed to be the later.

Subgroup analysis

We utilized predefined subgroup analyses to assess a number of possible influences on the effect of probiotic supplementation on clinical outcomes, and thus to explore the possible causes of heterogeneity. On the basis that the higher the daily dose the greater the effect, we first examined trials that administered a high dose of probiotics defined as >5 × 109 colony-forming units (CFU)/day vs. lower dose probiotics defined as <5 × 109 CFU/day. Second, we compared the results of RCTs that administered Lactobacillus plantarum as probiotic therapy vs. no L. Plantarum, and compared trials using Lactobacillus rhamnosus strain GG (LGG) vs. those administering other non-LGG strains.
Moreover, based on a larger treatment effect in those more seriously ill patients with higher risk of death, we compared studies including patients with higher mortality vs. lower mortality. Mortality was considered to be high or low based on whether it was greater or less than the median control group mortality of all the trials. Trials of higher quality, defined as those with a methodological score equal to or higher than the median quality score, may demonstrate a lower treatment effect.

Results

Study identification and selection

A total of 79 relevant citations were identified from the search of computerized bibliographic databases and a review of reference lists from related articles. Of these, we excluded 49 due to the following reasons: 21 trials did not include ICU patients (mostly surgical patients); 12 articles were systematic reviews and meta-analyses; 4 trials were published as an abstract and we were unable to obtain the data from the authors to complete our data abstraction process; 5 articles were duplicates of included trials; 3 studies did not evaluate clinical outcomes; 2 trials tested multiple interventions; 1 study was not a RCT, and finally 1 study administered probiotics as oral swabs.
Finally, 30 RCTs [10, 2026, 3051] met our inclusion criteria and were included, covering a total of 2972 patients (see Tables 1 and 2). The reviewers reached 100 % agreement on the inclusion of the trials. The mean methodological score of all trials was 9, whereas the median value was 9.5 on a maximum of 14 (range 5–13). Randomization was concealed in 9/30 trials (30 %), ITT analysis was performed in 18/30 trials (60 %), and double blinding was done in 20/30 of the studies (67 %). There were five level-I studies and 25 level-II studies. The details of the methodological quality of the individual trials are shown in Table 1.
Table 1
Randomized studies evaluating probiotics in critically ill patients
 
Study
Population
Methods score
Type of probiotic/intervention
Delivery vehicle
Intervention/dose/duration
Control
1
Tempe 1983 [30]
ICU patients
n = 40
C.Random: yes
ITT: yes
Blinding: double
Score: 10
Viability (intervention): NR
EN tube
EN (unknown) + Ultra-Levure (Saccharomyces boulardii), 1010/1 L solution for 11–21 days
EN (unknown) + placebo (sterile solution)
2
Schlotterer 1987 [31]
Patients with burns
n = 18
C.Random: no
ITT: no
Blinding: double
Score: 8
Viability (intervention): NR
NG tube
EN (Polydiet or Nutrigil) + Saccharomyces boulardi
500 mg QID for 8-28 days
EN (Polydiet or Nutrigil) + placebo
3
Heimburger 1994 [32]
Mixed ICU patients:
83 % received antibiotics
n = 62
C.Random: no
ITT: no
Blinding: double
Score: 9
Viability (intervention): NR
EN tube
EN (standard) + 1 g of Lactinex (Lactobacillus acidophilus and Lactobaccilus bulgaricus) 2 × 106TID for 5–10 days
EN (standard) + placebo (0.5 g dextrose + 0.5 g lactose)
4
Bleichner 1997 [33]
Mixed ICU patients
n = 128
C.Random: not sure
ITT: yes
Blinding: double
Score: 13
Viability (intervention): NR
EN tube
EN (unknown) + Saccharomyces boulardii
500 mg QID for 21 days or until EN stopped
EN (unknown) + placebo (powder)
5
Kecskes 2003 [34]
ICU patients on antibiotics
n = 45
C.Random: no
ITT: no
Blinding: double
Score: 8
Viability (intervention): yes
NJ tube
EN (Nutrison fiber) + fermented oatmeal formula with Lactobacillus plantarum 299 10 9 BID and fiber for 7 days
EN (Nutrison fiber) + heat-killed Lactobacillus plantarum 299 BID + fiber (non-viable)
6
Jain 2004 [36]
ICU patients
n = 90
C.Random: no
ITT: yes
Blinding: double
Score: 10
Viability (intervention): NR
Oral or NG tube
EN or PN + Trevis™ 1 capsule TID + 7.5 g Raftilose (oligofructose)
BID until hospital discharge
EN or PN + placebo (powdered sucrose capsules)
7
Lu 2004 [35]
Patients with burns
n = 40
C.Random: no
ITT: yes
Blinding: double
Score: 9
Viability (intervention): NR
NR
EN + synbiotics (4 types of probiotics + 4 types of unspecified prebiotics) for 21 days
EN + 4 types of prebiotics
8
Klarin 2005 [37]
Critically ill patients on antibiotics
n = 17
C.Random: no
ITT: no
Blinding: no
Score: 6
Viability (intervention): NR
Mixed in fermented oatmeal, given via NG tube
EN + Lactobacillus plantarum 299v, 109/day 50 ml every 6 h × 3 days then 25 ml every 6 h until ICU discharge
EN (Impact or Nutrodrip fiber). Some patients needed PN
9
McNaught 2005 [38]
ICU patients on antibiotics
n = 130
C.Random: no
ITT: yes
Blinding: no
Score: 7
Viability (intervention): NR
Oral, NJ tube
EN or PN + Proviva, (oatmeal and fruit drink) 5 × 107 CFU/ml of L. plantarum 299 v × 500 mls until hospital discharge or beyond
EN or PN alone
10
Kotzampassi 2006 [39]
Patients with multiple trauma from 5 ICUs
n = 77
C.Random: no
ITT: no
Blinding: double
Score: 8
Viability (intervention): NR
VAP determination: clinical
Endoscopic gastrostomy or NG tube
EN or PN + Synbiotic 2000 Forte 1011, 1 sachet/day for 15 days until ICU discharge
EN or PN + placebo (maltodextrin), mixed in tap water
11
Alberda 2007 [40]
ICU patients
n = 28
C.Random: no
ITT: yes;
Blinding: double
Score: 10
Viability (intervention): No for VSL # 3; Yes for bacteria sonicates
NG tube
Jevity Plus (EN) (10 g fructooligosaccharides/1000 mL and 12 g of soluble and insoluble fiber blend) +
VSL # 3, 1 package BID,
9 × 1011 /day for 7 days until ICU discharge or EN discontinuation
Jevity Plus + placebo
12
Li 2007 [41]
Patients with severe acute pancreatitis
n = 25
C.Random: no
ITT: yes
Blinding: no
Score: 7
Viability (intervention): NR
Given enterally
Jinshuangqi (bifidobacteria, lactobacillus and streptococcus) 2.0 g TID on basis of traditional treatment
Duration: NR
Traditional treatment
13
Olah 2007 [42]
Patients with severe acute pancreatitis
n = 83
C.Random: no
ITT: no
Blinding: no
Score: 9
Viability (intervention): NR
NJ tube
EN (Nutricion fiber) + Synbiotic 2000, 4 × 1010 CFU for 7 days
EN (Nutricion fiber) + 10 g plant fibers ((2.5 g each of Betaglucan, inulin, pectin and resistant starch) (prebiotics) BID for at least 2 days
14
Forestier 2008 [44]
Mixed ICU patients, 50 % on antibiotics
n = 208
C.Random: not sure
ITT: no
Blinding: double
Score: 8
Viability (intervention): NR
VAP determination: objective
NG tube or oral (after tube removal)
Lactobacillus casei rhamnosum, 109 CFU BID until ICU discharge
Placebo (growth medium never exposed to bacteria).
15
Besselink 2008 [43]
Patients with severe acute pancreatitis from 15 ICUs
n = 298
C.Random: not sure
ITT: yes
Blinding: double
Score:11
Viability (intervention): NR
VAP determination: clinical
NJ tube or oral
EN (Nutrison multifiber)
+ Ecologic 641 1010 CFU BID for 28 days
EN (Nutrison multifiber) + placebo (cornstarch + maltodextrins)
16
Klarin 2008 [45]
ICU patients from 5 ICUs, on antibiotics for C. Difficile
n = 68
C.Random: yes
ITT: no
Blinding: double
Score: 10
Viability (intervention): NR
Mixed in fermented oatmeal added to enteral feeds NG tube
299 Lactobacillus plantarum,
8 × 108 CFU/ml given as 6 × 100 ml doses every 12 h and after 50 ml given BID until ICU discharge
Same oatmeal gruel mixed with lactic acid
17
Knight 2009 [46]
General ICU patients
n = 300
C.Random: yes
ITT: no
Blinding: double
Score: 10
Viability (intervention): NR
VAP determination: clinical
NJ or OG (orogastric) tube
EN (Nutrition Energy) +
Synbiotic 2000 FORTE
4 × 1011 species/sachet
BID for 28 days or ICU discharge
EN (Nutrison Energy) + Placebo
18
Barraud 2010 [47]
Mechanically ventilated ICU patients, 80 % on antibiotics
n = 167
C.Random: yes
ITT: yes;
Blinding: double
Score: 12
Viability (intervention): NR
VAP determination: objective
NG tube
EN (Fresubin) + Ergyphilus
2 × 1010 per capsule + potato starch 5 capsules/day for 28 days
EN (fresubin) + placebo capsules (excipient of potato starch)
19
Morrow 2010 [10]
ICU patients
n = 146
C.Random: no;
ITT: yes;
Blinding: double; Score:10
Viability (intervention): yes
VAP determination: objective
Oropharynx and NG tube
EN (routine care) + Lactobacillus rhamnosus GG, 2 × 109 BID as lubricant and mixed with water until extubation
EN (routine care) + inert plant starch inulin (prebiotic) BID as lubricant and mixed with water
20
Frohmader 2010 [48]
General ICU patients
on antibiotics
n = 45
C.Random: yes
ITT: yes
Blinding: double
Score: 11
Viability (intervention): yes
NG or NJ tube
EN (Standard) + VSL #3 mixed in nutritional supplement (Sustagen), BID until hospital discharge
EN (Standard) + placebo mixed in nutritional supplement (Sustagen), BID
21
Ferrie 2011 [49]
Critically ill patients with diarrhea,
n = 36
C.Random: no
ITT: yes
Blinding: double
Score: 10
Viability (intervention): yes
NG tube
EN (Standard) + Culturelle (Lactobacillus rhamnosus GG), 1010 species/capsule
+280 mg inulin powder for 7 days
EN (Standard) + Raftiline, gelatin capsule with 280 mg inulin powder (prebiotic)
22
Sharma 2011 [50]
Patients with acute pancreatitis
n = 50
C.Random: yes
ITT: yes
Blinding: double
Score: 11
Viability (intervention): yes
Oral, NJ or NG
EN (standard) or oral
4 sachets each 2.5 × 109
Lactobacillus acidophilus, Bifidobacterium longus, Bifidobacterium bifidum & Bifidobacterium infantalis + 25 gms fructose for 7 days
EN (Standard) + placebo
23
Tan 2011 [51]
Patients with closed head injury
n = 52
C.Random: yes
ITT: yes
Blinding: single
Score: 10
Viability (intervention): yes
VAP determination: clinical
NG tube
EN (standard) total of 109 bacteria i.e.,
7 sachets each 0.5 × 108 Bifidobacterium longum, 0.5 × 1071 Lactobacillus bulgaricus and 0.5 × 107 Streptococcus thermophilus for 21 days
EN (standard)
24
Cui 2013 [20]
Patients with severe acute pancreatitis
n = 70
C.Random: no
ITT: yes
Blinding: no
Score: 9
Viability (intervention): yes
EN
EN + bifidobacterium, 4 capsules (each 210 mg, 2.604 × 109) every 12 h, given through nasal gastric tube. Total dose per day 20.832 × 109
EN
25
Tan 2013 [21]
Severe craniocerebral trauma
n = 52
C.Random: no
ITT: other
Blinding: no
Score: 11
Viability (intervention): yes
NG tube
EN + 1 × 109 bacteria of viable probiotics (Golden Bifid, 3.5 g 3 times per day) per day for 21 days.
EN (standard)
26
Wang 2013 [22]
Severe acute pancreatitis with intestinal ileus or abdominal distention.
n = 183
C.Random: no
ITT: yes
Blinding: no
Score: 6
Viability (intervention): NR
SBFT
EN (standard) + capsules 0.5 g TID containing Bacillus subtilis and Enterococcus faecium (5.0 × 107Bacillus subtilis and 4.5 × 108 Enterococcus faecium per 250 g capsule). Unclear timeframe.
EN (standard)
27
Lopez de Toro 2014 [23]
Medical and surgical ICU patients with multi-organ failure
n = 89
C.Random: yes
ITT: yes
Blinding: no
Score: 11
Viability (intervention): NR
EN
EN + symbiotic drink with streptococcus Thermophilus, lactobacillus bulgaricus, Lactobacillus casei, lactobacillus acidophilus, bifidobacterium, Escherichia coli, coliformes × 7 days (max 4.8 × 109 UFC/ml).
EN and PN
28
Sanaie 2014 [24]
Critically ill pts, SIRS, expected length of stay ≥7 days
n = 40
C.Random: yes
ITT: yes
Blinding: double
Score: 9
Viability (intervention): yes
NG tube
EN (standard) + 2 sachets VSL#3 BID × 7 days.
EN (standard) + placebo
29
Rongrungruang 2015 [25]
Critically ill patients, expected to receive mechanical ventilation at least 72 h and had no VAP at enrollment
n = 150
C.Random: no
ITT: no
Blinding: no
Score: 6
Viability (intervention): yes
EN
80 ml of 8 × 109 cfu of Lactobacillus casei (Shirota strain) (Yakult) for oral care after the standard oral care once daily
An additional 80 ml of the product was given via enteral feeding once daily for 28 days or when their endotracheal tubes were removed
EN (standard) + oral care with 2 % chlorhexidine solution 4 times per day
30
Zeng 2016 [26]
Critically ill patients, expected to receive mechanical ventilation at least 48 h
n = 235
C.Random: yes
ITT: no
Blinding: no
Score: 5
Viability (intervention): yes
NG tube
1 capsule (Medilac-S, China) 0.5 g three times daily. Each probiotic capsule contained
active Bacillus subtilis and Enterococcus faecalis at a concentration of 4.5 × 109 /0.25 g and 0.5 × 109/0.25 g, respectively
EN (standard)
CFU colony forming units, C.Random concealed randomization, EN enteral nutrition, FOS fructooligosaccharides, NG nasogastric, NJ nasojejunal, NR not reported, OG orogastric, ITT intention to treat, SIRS, systemic inflammatory response syndrome, VAP ventilator-associated pneumonia, BID twice daily. Trevis™: 1 capsule = Lactobacillus acidophilus La5, Bifidobacterium lactis Bb12, Streptococcus thermophilus, Lactobacillus bulgaricus, 4 × 10 9 /total; Synbiotic 2000 Forte: 1011 CFU each of Pediococcus pentoseceus 5-33:3, Leuconostoc mesenteroides 32-77:1, L. paracasei ssp paracasei 19, L. plantarum 2362, and 2.5 g each of inulin, oat bran, pectin and resistant starch; Ergyphilus: 1010 Lactobaccilus rhamnosus GG, Lactobacillus casei, L. acidophilus, Bifidobacterium bifidus; VSL # 3: >1010 Bifidobacterium longum, Bifidobacterium breve, >10 10/g Bifidobacterium infantis, >10 11/g L. acidophulus, L. plantarum, L. casei, L. bulgaris, and Streptococcus thermophiles; Jinshuangqi: B. longum >10 7 CFU , L. bulgaricus >10 6 CFU , and S. Thermophilus >10 6 CFU ; Ecologic 641: L. acidophilus, Lactobacillus salivarius, Lactococcus lactis, B. bifidus, and Bifidobacterium lactis; Synbiotic 2000: 10 10 CFU each of P. pentoseceus 5-33:3, Leuconostoc mesenteroides 32-77:1, L. paracasei ssp paracasei 19, L. plantarum 2362, and 2.5 g each of betaglucan, inulin, pectin and resistant starch; Golden Bifid: B. bifidum, L. bulgaricus, and S. thermophilus triple-human probiotic-supplemented oligosaccharides FOS (bifidus factor)
Table 2
Reported clinical outcomes in RCTs evaluating probiotics in critically ill patients
 
Study
Mortality
Infections
Length of stay
Diarrhea
Intervention
Control
Intervention
Control
Intervention
Control
Intervention
Control
1
Tempe 1983 [30]
3/20 (15)
3/20 (15)
NR
NR
NR
NR
Diarrhea days 34/389 (9)
Diarrhea days 63/373 (17)
2
Schlotterer 1987 [31]
NR
NR
NR
NR
NR
NR
Diarrhea days 3/150 (2)
Diarrhea days 19/143 (13)
3
Heimburger 1994 [32]
NR
NR
NR
NR
NR
NR
Diarrhea 5/16 (31)
Diarrhea 2/18 (11)
4
Bleichner 1997 [33]
NR
NR
NR
NR
NR
NR
Diarrhea 18/64 (28)
Days w/diarrhea 91/648 (14)
Diarrhea 24/64 (38)
Days w/diarrhea 134/683 (20)
5
Kecskes 2003 [34]
Hospital 1/22 (5)
Hospital 2/23 (9)
Septic compl 1/22 (5)
Septic compl 7/23 (30)
Hospital 13.7 ± 8.7
Hospital 21.4 ± 17.9
NR
NR
6
Jain 2004 [36]
Hospital 22/45 (49)
Hospital 20/45 (45)
Septic compl 33/45 (73)
Septic compl 26/45 (58)
Hospital 24.0 ± 31.5
ICU 11.9 ± 13.1
Hospital 18.7 ± 13.5
ICU 9.0 ± 8.9
NR
NR
7
Lu 2004 [35]
Hospital 2/20 (10)
Hospital 1/20 (5)
Infectious compl 8/20 (40)
Infectious compl 11/20 (55)
NR
NR
NR
NR
8
Klarin 2005 [37]
Hospital 2/8 (25)
ICU 1/8 (12)
Hospital 2/7 (29)
ICU 2/7 (29)
NR
NR
Hospital 48.3 ± 30.4
ICU 14.2 ± 10.6
Hospital 34.3 ± 15.4
ICU 16.3 ± 15.7
NR
NR
9
McNaught 2005 [38]
18/52 (35)
18/51 (35)
Septic morbidity 21/52 (40)
Septic morbidity 22/51 (43)
ICU 5 (2–9)
ICU 4 (2–7)
NR
NR
10
Kotzampassi 2006 [39]
ICU 5/35 (14)
ICU 9/30 (30)
Infections 22/35 (63)
VAP 19/35 (54)
Septic compl 17/35 (49)
Central venous line infections 13/35 (37)
Wound infections 6/35 (17)
UTI 6/35 (17)
Infections 27/30 (90)
VAP 24/30 (80)
Septic compl 23/30 (77)
Central venous line infections 20/30 (66)
Wound infections 8/30 (26)
UTI 13/30 (43)
ICU 27.7 ± 15.2
ICU 41.3 ± 20.5
Diarrhea 5/35 (14)
Diarrhea 10/30 (30)
11
Alberda 2007 [40]
ICU 1/10 (10)
ICU 1/9 (11)
NR
NR
NR
NR
Diarrhea 1/10 (14)
Diarrhea 2/9 (23)
12
Li 2007 [41]
NR
NR
Infections 8/14 (58)
Infections 10/11 (91)
Hospital 42 ± 5.0
Hospital 49 ± 6.8
NR
NR
13
Olah 2007 [42]
Hospital 2/33 (6)
Hospital 6/29 (21)
Infections 9/33 (27)
Septic compl 7/33 (12)
Pancreatic abscess 2/33 (6)
Infected pancreatic necrosis 2/33 (6)
UTI 3/33 (9)
Infections 15/29 (52)
Septic compl 17/29 (28)
Pancreatic abscess 2/29 (7)
Infected pancreatic necrosis 6/29 (21)
UTI 3/33 (9)
Hospital 14.9 ± 3.3
Hospital 19.7 ± 4.5
NR
NR
14
Forestier 2008 [44]
NR
NR
VAP 19/102 (19)
VAP 21/106 (20)
ICU 22.5 ± 20.6
ICU 19.7 ± 16.7
NR
NR
15
Besselink 2008 [43]
24/152 (16)
9/144 (6)
Infections 46/152 (30)
VAP 24/152 (16)
Bacteremia 33/152 (22)
Infected necrosis 21/152 (14)
Urosepsis 1/52 (2)
Infections 41/144 (28)
VAP 16/144 (11)
Bacteremia 22/144 (15)
Infected necrosis 14/144 (10)
Urosepsis 2/144 (1)
Hospital 28.9 ± 41.5
ICU 6.6 ± 17
Hospital 23.5 ± 25.9
ICU 3.0 ± 9.3
Diarrhea 25/152 (16)
Diarrhea 28/144 (19)
16
Klarin 2008 [45]
Hospital 3/22 (5)
ICU 2/22 (9)
Hospital 2/22 (0)
ICU 2/22 (9)
C. difficile + fecal samples 0/71
C. difficile + fecal samples 4/80
Hospital 25.8 ± 19.4
ICU 8.0 ± 5.4
Hospital 50.3 ± 75.2
ICU 11.6 ± 14
NR
NR
17
Knight 2009 [46]
Hospital 35/130 (27)
ICU 28/130 (22)
Hospital 42/129 (33)
ICU 34/129 (26)
VAP 12/130 (9)
VAP 17/129 (13)
ICU 6 (3–11)
ICU 7 (3–14)
Diarrhea 7/130 (5)
Diarrhea 9/129 (7)
18
Barraud 2010 [47]
ICU 21/87 (24)
28 days 22/87 (25)
90 days 27/87 (31)
ICU 21/80 (26)
28 days 19/80 (24)
90 days 24/80 (30)
All infections 30/87 (34)
Infection > 96 h 26/87 (30)
VAP 23/87 (26)
Catheter-related BSI 3/87 (4)
UTI 4/87 (5)
All infections 30/80 (38)
Infection > 96 h 29/80 (36)
VAP 15/80 (19)
Catheter-related BSI 11/80 (14)
UTI 4/89 (5)
Hospital 26.6 ± 22.3
ICU 18.7 ± 12.4
Hospital 28.9 ± 26.4
ICU 20.2 ± 20.8
Diarrhea 48/87 (55)
Diarrhea 42/80 (53)
19
Morrow 2010 [10]
12/68 (18)
15/70 (21)
VAP 13/73 (18)
VAP 28/73 (38)
Hospital 21.4 ± 14.9
ICU 14.8 ± 11.8
Hospital 21.7 ± 17.4
ICU 14.6 ± 11.6
Non C. difficile diarrhea 42/68 (62)
C. difficile diarrhea 4/68 (6)
Non C. difficile diarrhea 44/70 (63)
C. difficile diarrhea 13/70 (19)
20
Frohmader 2010 [48]
5/20 (25)
3/25 (12)
NR
NR
ICU 7.3 ± 5.7
ICU 8.1 ± 4
Diarrhea episodes/pt/day 0.53 ± 0.54
Diarrhea episodes/pt/day 1.05 ± 1.08
21
Ferrie 2011 [49]
Hospital 2/18 (11)
6 months 7/18 (39)
Hospital 2/18 (11)
6 months 5/18 (28)
Infections 14/18 (78)
Infections 16/18 (89)
Hospital 54.50 ± 31.26
ICU 32.04 ± 24.46
Hospital 59.04 ± 33.92
ICU 29.75 ± 18.81
Duration of diarrhea 3.83 ± 2.39
Loose stools/day 1.58 ± 0.88
Duration of diarrhea 2.56 ± 1.85
Loose stools/day 1.10 ± 0.79
22
Sharma 2011 [50]
Hospital 2/24 (8)
Hospital 2/26 (8)
NR
NR
Hospital 13.23 ± 18.19
ICU 4.94 ± 9.54
Hospital 9.69 ± 9.69
ICU 4.0 ± 5.86
NR
NR
23
Tan 2011 [51]
28 days 3/26 (12)
28 days 5/26 (19)
Infections 9/26 (35)
VAP 7/26 (27)
Infections 15/26 (58)
VAP 13/26 (50)
ICU 6.8 ± 3.8
ICU 10.7 ± 7.3
NR
NR
24
Cui 2013 [20]
Hospital 1/23 (4)
Hospital 1/25 (4)
N/A
N/A
Hospital 10.4 ± 3.9 (23)
Hospital 13.4 ± 5.2 (25)
NR
NR
25
Tan 2013 [21]
28 days 23/26 (12)
28 days 5/26 (19)
NR
NR
ICU 6.8 ± 3.8 (26)
ICU 10.7 ± 7.3 (26)
NR
NR
26
Wang 2013 [22]
Unspecified 1/62 (8.1)
Unspecified 3/61 (9.8)
Pancreatic sepsis 8/62 (13)
MODS 7/62 (11.3)
Pancreatic sepsis 13/61 (21)
MODS 15/61 (25)
NR
NR
NR
NR
27
Lopez de Toro 2014 [23]
Hospital 19/46 (41)
ICU 15/46 (33)
Hospital 18/43 (42)
ICU 14/43 (33)
Hospital acquired infections 9/46 (20)
Hospital acquired infections 13/43 (30)
Hospital 18.5 (10–36)
ICU 9 (3–19)
Hospital 24.5 (10–38)
ICU 8 (2.5–16.5)
NR
NR
28
Sanaie 2014 [24]
28 days 2/20 (10)
28 days 5/20 (25)
Bacteremia 2/20(10)
Bacteremia 5/20(25)
ICU 13.85 ± 6.96
ICU 14.16 ± 5.97
NR
NR
29
Rongrungruang 2015 [25]
28 days 18/75 (24)
90 day 25/75 (33)
28 days 17/75 (22.7)
90 day 26/75 (34.7)
VAP 18/75 (24)
VAP 22/75 (29.3)
Hospital 20 (2–106)
ICU 30.5 (4–98)
Hospital 19 (3–171)
ICU 19 (5–30)
Diarrhea 19/75 (25.3)
Diarrhea 14/75 (18.7)
30
Zeng 2016 [26]
Hospital 26/118 (22)
ICU 15/118 (12.7)
Hospital 25/117 (21.4)
ICU 9/117 (7.7)
VAP 43/118 (36.4)
VAP 59/117 (50.4)
Hospital 13.5 ± 12.4
ICU 18 (14–32)
Hospital 10.6 ± 10.2
ICU 22 (11–56)
NR
NR
BSI blood stream infection, ICU intensive care unit, NR not reported, RCT randomized control trial, UTI urinary tract infection, VAP ventilator associated pneumonia, N/A non-attributable, compl complications, MODS Multiple Organ Dysfunction Syndrome, Days w/ diarrhea - days with diarrhea

Primary outcome: infections

Overall effect on new infections

Aggregating the results of the 14 trials reporting overall infections, probiotics were associated with a significant reduction in infections (RR 0.80, 95 % CI 0.68, 0.95, P = 0.009; heterogeneity I 2 = 36 %, P = 0.09; Fig. 1).

Secondary outcomes

Ventilator associated pneumonia

Aggregating the data from 9 trials that reported VAP, there was a significant reduction in the incidence of VAP (RR 0.74, 95 % CI 0.61, 0.90, P = 0.002; I2 = 19 %, P = 0.27; Fig. 2).

Overall effect on mortality

A total of 17 studies reported on hospital mortality. When statistically aggregated, probiotic therapy did not significantly affect mortality (RR 0.98, 95 % CI 0.82, 1.18, P = 0.85; I 2 = 0 %, Fig. 3). Moreover, probiotics did not show any effect on ICU mortality (RR 0.90, 95 % CI 0.70, 1.17, P = 0.44; I 2 = 0 %).

Overall effect on ICU length of stay

Aggregating the data from the 14 RCTs reporting on ICU LOS, there were no significant differences between the groups (weighted mean difference (WMD) -3.26, 95 % CI -7.82, 1.31, P = 0.16; I 2 = 93 %, P < 0.00001).

Overall effect on hospital length of stay

Aggregating the data from the nine RCTs that reported hospital LOS, there were no significant differences between the groups (WMD -0.58, 95 % CI -3.66, 2.50, P = 0.71; I 2 = 74 %, P < 0.00001).

Diarrhea

Aggregating the data from nine trials that reported on diarrhea, probiotics had no effect (RR 0.97; 95 % CI 0.82, 1.15; P = 0.74; I 2 5 %, P = 0.39; Fig. 4).

Antibiotic days

When we aggregated the data of four trials reporting on antibiotic days, we found that probiotics were significantly associated with a reduction in the duration of antibiotic therapy (WMD -1.12, 95 % CI -1.72, -0.51, P = 0.0003; I 2 = 32 %, P = 0.22; Fig. 5).

Subgroup analysis

Probiotics daily dose

There were similar rates of infectious complications in RCTs using high-dose probiotic therapy (n = 8 trials) (0.87; 95 % CI 0.72–1.06; P =0.18; I 2 = 43 %) and in those using lower daily doses (n = 2 trials) RR 0.40; 95 % CI 0.11–1.50; P = 0.18; I 2 = 48 %. The difference between subgroups was not significant, P = 0.25).

L. plantarum vs. non L. plantarum

Subgroup analyses showed that four trials administering L. plantarum, either alone or in combination with other probiotics, were associated with a significant reduction in overall infections (RR = 0.70, 95 % CI 0.50, 0.97; P = 0.03; I 2 = 36 %). However, in the 10 trials that did not include L. plantarum, there was no significant effect on overall infectious complications (RR = 0.88, 95 % CI 0.74,1.04; P = 0.15; I 2 = 21 %). Test for subgroup differences between groups was not significant (P = 0.21).

L. rhamnosus GG vs. other probiotics

In two trials using LGG there was no significant effect on reduction in infectious complications (RR 0.86; 95 % CI 0.67–1.10; P = 0.22; I 2 = 0 %). However, in 12 trials that supplemented other probiotics there was a significant reduction in overall infections (RR 0.77; 95 % CI 0.62–0.95; P = I 2 = 45 %); P = 0.52 for the difference between groups.

Synbiotics vs. other strategies

In subgroup analyses of the four trials that administered synbiotics there was no effect on infections (RR = 0.80, 95 % CI 0.49, 1.30, P = 0.36; I 2 = 66 %, P = 0.03) (Fig. 6). However, in 10 studies that administered probiotics alone there was a significant reduction in the incidence of infections (RR 0.79, 95 % CI 0.68, 0.92, P = 0.002; I 2 = 9 %, P = 0.36). The P value for subgroup differences was not significant (P = 0.98). (Fig. 6).

Higher vs. lower mortality

The median hospital mortality rate of all the trials (or ICU mortality when hospital mortality was not reported) in the control group was 19 %. After aggregating nine studies with a higher mortality rate, probiotics significantly reduced the incidence of infections (RR 0.74; 95 % CI 0.57, 0.96; P = 0.02; I 2 = 58 %, P = 0.01) (Fig. 7). However, probiotics did not have an effect on infections in the five studies with lower mortality (RR 0.85; 95 % CI 0.66, 1.11; P = 0.24; I 2 = 23 %, P = 0.27). The test for subgroup differences was not significant (P = 0.43) (Fig. 7).

Higher vs. lower methodological score

The median methodological score was 9.5. In six trials with a higher score (≥9.5) there was no effect on infections (RR 0.93; 95 % CI 0.76, 1.15; P = 0.51; I 2 = 35 %, P = 0.17), whereas in eight trials with a lower methodological score (<9.5) there was a significant reduction in infectious complications (RR 0.69, 95 % CI 0.57, 0.83, P < 0.0001; I 2 = 0 %) (Fig. 8); the overall test for subgroup differences was significant for these subgroups (P = 0.03).

Publication bias

There was indication that potential publication bias influenced the observed aggregated results. In fact, funnel plots were created for each study outcome and the tests of asymmetry were significant for overall new infections (odds ratio (OR) -2.30, 95 % CI -3.56, -1.05, P = 0.001; Fig. 9) and hospital LOS (OR -3.32, 95 % CI -6.12, -0.52, P = 0.024). However, the test for asymmetry was not significant for any other outcome (VAP, P = 0.76; mortality, P = 0.80; ICU LOS, P = 0.47; diarrhea, P = 0.18).

Discussion

To date, our systematic review and meta-analysis is the largest and most updated evaluation of the overall effects of probiotics in the critically ill. It is also the first to include an analysis of symbiotic (probiotic/fiber combinations). Based on the analysis of 30 trials enrolling 2972 patients we demonstrated that probiotics are associated with a significant reduction in ICU-acquired infections, including VAP, which is the most common infectious complication in the critically ill. This significant effect on VAP is a new finding from our previous systematic reviews. Further, the beneficial effect of probiotics on reduction of infections is stronger with the publication of the new trials and the data no longer show a statistically significant effect of heterogeneity on this endpoint. Despite the probiotic effect of reducing infectious complications, this therapy did not influence ICU or hospital mortality, although none of the trials were powered to detect an effect on mortality. Overall, there was a tendency towards a reduction in ICU LOS and probiotic therapy did not influence other clinical endpoints such as hospital LOS, and diarrhea. Statistical and clinical heterogeneity was observed for some endpoints, although this was significant for the key endpoints of infectious complications and VAP. In addition, publication bias for overall infections and hospital LOS means that larger, well-powered, and more definitive clinical trials are urgently needed aimed to avoid these biases. Moreover, subgroup analysis showed that those trials with lower methodological quality exhibit the best treatment effect, which is another issue indicating that larger, well-designed studies are needed. Again, with the exception of four trials, most of the included studies (n = 14) that reported mortality had small sample sizes, and hence were underpowered and inadequate to detect any clinically important treatment effects of probiotic therapy on mortality. Moreover, the inferences we can make from our current findings are further weakened, as randomization was concealed in 30 % of trials, whereas double-blinding was performed in 67 % of trials.
Over recent years, several systematic reviews and meta-analyses have been conducted, although our meta-analysis is the largest and most current to date, as it contains the seven new suitable trials published since the most recent comprehensive meta-analysis publication on this topic, which focused on overall infections and other outcomes (not primarily VAP) in 2012 [14]. Further, these previous systematic reviews did not include analysis of synbiotic therapy. Overall, we have examined several relevant clinical outcomes in a heterogenous ICU patient population, and therefore our results could be applied to a broad group of critically ill patients with sepsis, trauma, severe pancreatitis, or who have undergone surgery. Specific to pancreatitis, concerns have been raised about the safety of probiotic therapy following the 2008 trial, Probiotic prophylaxis in patients with predicted severe acute pancreatitis (PROPATRIA) [43], which showed that Ecologic 641® given with fiber post-pyloric was associated with higher mortality and bowel ischemia. This post-pyloric method of administration was associated with an increase in small bowel necrosis, which was subsequently associated with death in a number of patients receiving the prebiotic fiber/probiotic mixture. It is possible that the post-pyloric administration of this fiber/multiple probiotic strain mixture in patients with pancreatitis may carry significant risk and should likely be avoided [52]. Unfortunately, there were significant ethical and statistical concerns raised about the conduct of the trial [53], limiting the utility of the data. Further, more recently, a systematic review and meta-analysis by Gou et al. [54] found that probiotics had neither beneficial nor adverse effects in patients with pancreatitis.
Despite the limitations of the PROPRIATA trial, it has contributed to concerns around the safety of probiotic administration in critical illness and limited the design of larger-scale clinical trials and/or more routine clinical administration of live probiotics. To address this, the American Health Care Research and Quality (AHRQ) agency reviewed and reported on the safety of probiotic therapy in over 600 published clinical trials and case reports [55]. It should be reassuring to future investigators that the overall conclusion of the extensive AHRQ report indicates that probiotic therapy in both adults and pediatric populations was not been found to be associated with any increased risk of infectious or other adverse events in either healthy or ill patients. Importantly, their report revealed a trend towards less adverse events in probiotic-treated critically ill patients, although isolated adverse effects of probiotic administration have been reported [56]. In any case, careful and appropriate safety monitoring in all future probiotic clinical trials should be conducted.
Recent data indicate that infection during critical illness continues to be a major challenge worldwide. A multi-national ICU study of 14,414 patients in 1265 ICUs from 75 countries, revealed that 51 % of ICU patients were considered infected on the day of survey and 71 % were receiving antibiotics [57]. Of the infections in this study, 64 % were of respiratory origin and the ICU mortality rate in infected patients was more than twice that of non-infected patients (25 % vs. 11 %, P < 0.001), as was the hospital mortality rate (33 % infected vs. 15 % non-infected, P < 0.001) [57].
Currently, VAP is the second most common nosocomial infection in the USA and the most prevalent ICU-acquired infection. Notwithstanding, its incidence is highly variable depending on diagnostic criteria used to identify this infectious complication. In fact, in 2015 Ego et al. [58] reported that the incidence of VAP ranged from 4 % to 42 % when using the six published sets of criteria and from 0 % to 44 % when using the 89 combinations of criteria for hypoxemia, inflammatory response, bronchitis, chest radiography, and microbiologic findings. In our systematic review we found that the incidence of VAP ranged from 9 % [46] to 80 % [39]. Additionally, the apparent effect of probiotics on VAP is largely driven by the studies of Kotzampassi et al. [39] study and the Zeng et al. [26]; both trials explain 45.5 % of the signal and thus, provide an unstable estimate. Moreover, current knowledge shows that VAP is associated with high cost and poor clinical outcomes [59]. In 2002, Rello et al. [60] demonstrated that VAP leads to an additional US$40,000 in hospital charges per patient, and recently it has been suggested that the use of prophylactic probiotics may be cost-effective for prevention of VAP from a hospital perspective [61].
Probiotic therapy may prevent VAP and other infections by restoring non-pathogenic flora, which competes with nosocomial pathogens inhibiting their overgrowth, modulating local and systemic immune response, and improving gut barrier function. However, in spite of these protective effects the role of probiotics as a non-pharmacological strategy in preventing VAP has previously been inconclusive. In 2010, Siempos et al. [12] aggregated five probiotic trials demonstrating a reduction in the incidence of VAP, whereas in 2012 Petrof et al. [14] and subsequently Barraud et al. [13] and Wang et al. [15] did not demonstrate any significant effect of probiotic therapy on VAP. More recently, a Cochrane review of probiotic therapy specifically for VAP [17], found with low quality of evidence that probiotic therapy is associated with a reduction in the incidence of VAP. Our current systematic review demonstrates a significant treatment effect of probiotics in reducing VAP and did not demonstrate statistical heterogeneity, strengthening the signal that this may be an effective therapy for VAP. Recently, a Canadian survey [27] on the use of probiotics as a prophylactic strategy for VAP showed that most Canadian ICU pharmacists have used probiotics at least once, although they do not routinely recommend probiotics for the prevention of VAP.
Currently, a large number of clinical trials have demonstrated that probiotics may reduce the incidence of antibiotic-associated diarrhea and Clostridium difficile infections, and systematic reviews have confirmed a significant signal of benefit on reduction of diarrhea and C. difficile-related colitis in all patients (not confined to ICU patients) [62, 63]. Our results, when focused on ICU patients do not currently demonstrate a treatment benefit of probiotics in preventing and treating diarrhea in the critically ill, including antibiotic-associated diarrhea.
An interesting finding of our meta-analysis was a reduction in antibiotic use in those patients who received probiotics. Nonetheless, only four trials [10, 21, 26, 48] comprising 13 % of included studies reported duration of antibiotic therapy as an outcome. In addition, the study of Zeng et al. contributed to 90 % of the signal, which is a very unstable estimate that weakens our finding. Therefore, probiotics may shorten the duration of antibiotic therapy, although the limited clinical trial data available for this endpoint limits the strength of these findings and further investigation of this effect is needed.
We currently have a greater understanding about the potential benefits of probiotics therapy in critical illness, although much more data are needed. Subgroup analysis found that certain strains such as L. plantarum alone or in combination was associated with a significant reduction in overall infections, although the test for subgroup differences was not significant (P = 0.21). Certain specific biological properties have been described for L. plantarum, including an ability to prevent adhesion of pathogens to the intestinal epithelium secondary to the production of adhesins, enolase, and phosphoglycerate kinase on the bacterial surface [64, 65]. These mechanisms may be crucial to reduction of bacterial translocation and modulation of local inflammatory response, and therefore the effect of this strain on systemic infectious complications. Interestingly, probiotics alone had a greater effect than synbiotics on infections, although the difference between these subgroups was not significant (P = 0.98) and more data on the specific effects of different prebiotic fibers are needed. Finally, future trials also need to focus on evaluating the changes in the microbiome following critical illness and the effect of probiotic or synbiotics on restoring a healthy microbiome in treated patients [66]. Recent advances in microbiome sequencing technology (16 s rRNA) in the last few years have resulted in an unprecedented growth in the amount of sequence data that can be collected at a previously unattainable low cost [66]. Thus, if we speculate that a specific probiotic or synbiotic therapy can be used to treat dysbiosis (a pathological change in the patient’s bacterial flora) and restore a healthy microbiome, we need to evaluate this with the new accessible microbiome analysis techniques currently available. This may help us target probiotic or probiotic mixtures in the future and increase the personalization of care.
The strength of this current systematic review includes the use of several methods to reduce bias (comprehensive literature search, duplicate data abstraction, specific criteria for searching and analysis), and the analysis of relevant clinical outcomes in the critically ill. However, several important limitations in drawing strong treatment inferences are present. These include the significant potential for publication bias for the infection and hospital LOS outcomes, and the small numbers of trials included in subgroup analyses. In addition, the variety of probiotic strains, wide range of daily doses, and length of administration of probiotic therapy among the different trials weaken any possible clinical conclusions and recommendations. We were also unable to perform subgroup analysis for all clinical outcomes due to the limited number of studies evaluating each endpoint.
Based on our current data, there is not currently sufficient evidence to make a final strong recommendation for probiotics to be utilized in the prevention of infections, including VAP, in the critically ill. However, our current guideline recommendations suggest that probiotics should be considered to improve outcome in critically ill patients [19]. Future trials continue to need to address questions about timing, daily dose, and duration of therapy, which still remain unanswered.

Conclusion

In the largest systematic review and meta-analysis of probiotics to date, we demonstrated that in 30 trials enrolling 2972 patients, probiotics significantly reduced the incidence of infectious complications, including new episodes of VAP in critically ill patients. This finding is limited by clinical heterogeneity and potential publication bias for the overall infection outcome. This precludes a more meaningful statistical conclusion of the efficacy of probiotic therapy on overall infections and potentially the prevention of VAP in critical illness. Moreover, according to our findings probiotics has been demonstrated to be more effective in those trials with higher mortality in the control group. Probiotic therapy with L. plantarum currently demonstrates the most significant effect on the reduction of infections. Overall, the variety of strains, wide range of daily doses, and length of administration of probiotics weakens the strength of our conclusion. Certainly, additional large-scale, adequately powered, well-designed clinical trials, aimed at confirming our observations, are needed and warranted.

Key messages

  • Critical illness is characterized by a loss of commensal flora and an overgrowth of potentially pathogenic bacteria, leading to a high susceptibility of nosocomial infections
  • Probiotics are living non-pathogenic microorganisms, which may protect the gut barrier, attenuate pathogen overgrowth, decrease bacterial translocation and prevent infection in ICU patients
  • Probiotic use in the ICU remains widespread and controversial, current guidelines are not conclusive, and a significant number of new trials of probiotics have been published recently, which requires a current and comprehensive systematic analysis of probiotic and synbiotic therapy in critically ill patients
  • Probiotics were associated with a significant reduction in infections and a significant reduction in the incidence of ventilator-associated pneumonia (VAP) was found in critically ill patients receiving probiotics alone versus synbiotic mixtures, demonstrating the greatest improvement in infectious outcome, limited synbiotic trial data are currently available
  • Currently, clinical heterogeneity and potential publication bias reduce strong clinical recommendations and indicate further high-quality clinical trials are needed to conclusively prove these benefits
  • Probiotics shows promise for the reduction of infections, including VAP in critical illness, and should be considered in critically ill patients

Abbreviations

CFU, colony-forming unit; CI, confidence interval; C.Random, concealed randomization; EN, enteral nutrition; ICU, intensive care unit; Ig A, immunoglobulin A; ITT, intention to treat; LGG, Lactobacillus rhamnosus strain GG; LOS, length of stay; MV, mechanical ventilation; NA, non-attributable; NR, non-reported; OR, odds ratio; RCT, randomized controlled trial; RNA, ribonucleic acid; RR, risk ratio; VAP, ventilator-associated pneumonia; WMD, weighted mean difference

Funding

No funding for the development, writing or submission of this manuscript was received.

Authors’ contributions

WM contributed to development of the concept of the manuscript, study grading, study selection, evaluation and interpretation of data, and also performed primary authoring and editing of all drafts of the manuscript. ML contributed to study grading, selection, evaluation and interpretation of data, performed much of the primary statistical analysis, meta-analysis and data analysis, and also contributed to the writing of the manuscript. PL contributed to development of study grading, study selection, evaluation and interpretation of data, and also contributed substantially to the writing of the manuscript. PW contributed to development of the concept of the manuscript, evaluation and interpretation of data, and also performed authoring and editing of all drafts of the manuscript. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Alverdy JC, Chang FB. The reemerging role of the intestinal flora in critical illness and inflammation: why the gut hypothesis of sepsis syndrome will not go away. J Leukoc Biol. 2008;83:461–6.CrossRefPubMed Alverdy JC, Chang FB. The reemerging role of the intestinal flora in critical illness and inflammation: why the gut hypothesis of sepsis syndrome will not go away. J Leukoc Biol. 2008;83:461–6.CrossRefPubMed
2.
Zurück zum Zitat Latorre M, Krishnareddy S, Freedberg DE. Microbiome as mediator: Does systemic infection start in the gut? World J Gastroenterol. 2015;21:10487–92.CrossRefPubMedPubMedCentral Latorre M, Krishnareddy S, Freedberg DE. Microbiome as mediator: Does systemic infection start in the gut? World J Gastroenterol. 2015;21:10487–92.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Watson RS, Carcillo JA, Linde-Zwirble WT, Clermont G, Lidicker J, Angus DC. The epidemiology of severe sepsis in children in the United States. Am J Respir Crit Care Med. 2003;167:695–701.CrossRefPubMed Watson RS, Carcillo JA, Linde-Zwirble WT, Clermont G, Lidicker J, Angus DC. The epidemiology of severe sepsis in children in the United States. Am J Respir Crit Care Med. 2003;167:695–701.CrossRefPubMed
4.
Zurück zum Zitat Milbrandt EB, Kersten A, Rahim MT, Dremsizov TT, Clermont G, Cooper LM, Angus DC, Linde-Zwirble WT. Growth of intensive care unit resource use and its estimated cost in Medicare. Crit Care Med. 2008;36:2504–10.CrossRefPubMed Milbrandt EB, Kersten A, Rahim MT, Dremsizov TT, Clermont G, Cooper LM, Angus DC, Linde-Zwirble WT. Growth of intensive care unit resource use and its estimated cost in Medicare. Crit Care Med. 2008;36:2504–10.CrossRefPubMed
6.
Zurück zum Zitat Luyer MD, Buurman WA, Hadfoune M, Speelmans G, Knol J, Jacobs JA, Dejong CH, Vriesema AJ, Greve JW. Strain-specific effects of probiotics on gut barrier integrity following hemorrhagic shock. Infect Immun. 2005;73:3686–92.CrossRefPubMedPubMedCentral Luyer MD, Buurman WA, Hadfoune M, Speelmans G, Knol J, Jacobs JA, Dejong CH, Vriesema AJ, Greve JW. Strain-specific effects of probiotics on gut barrier integrity following hemorrhagic shock. Infect Immun. 2005;73:3686–92.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Tok D, Ilkgul O, Bengmark S, Aydede H, Erhan Y, Taneli F, Ulman C, Vatansever S, Kose C, Ok G. Pretreatment with pro- and synbiotics reduces peritonitis-induced acute lung injury in rats. J Trauma. 2007;62:880–5.CrossRefPubMed Tok D, Ilkgul O, Bengmark S, Aydede H, Erhan Y, Taneli F, Ulman C, Vatansever S, Kose C, Ok G. Pretreatment with pro- and synbiotics reduces peritonitis-induced acute lung injury in rats. J Trauma. 2007;62:880–5.CrossRefPubMed
8.
Zurück zum Zitat Corr SC, Li Y, Riedel CU, O'Toole PW, Hill C, Gahan CG. Bacteriocin production as a mechanism for the antiinfective activity of Lactobacillus salivarius UCC118. Proc Natl Acad Sci U S A. 2007;104:7617–21.CrossRefPubMedPubMedCentral Corr SC, Li Y, Riedel CU, O'Toole PW, Hill C, Gahan CG. Bacteriocin production as a mechanism for the antiinfective activity of Lactobacillus salivarius UCC118. Proc Natl Acad Sci U S A. 2007;104:7617–21.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Mittal R, Coopersmith CM. Redefining the gut as the motor of critical illness. Trends Mol Med. 2014;20(4):214–23.CrossRefPubMed Mittal R, Coopersmith CM. Redefining the gut as the motor of critical illness. Trends Mol Med. 2014;20(4):214–23.CrossRefPubMed
10.
Zurück zum Zitat Morrow LE, Kollef MH, Casale TB. Probiotic prophylaxis of ventilator-associated pneumonia: a blinded, randomized, controlled trial. Am J Respir Crit Care Med. 2010;182:1058–64.CrossRefPubMedPubMedCentral Morrow LE, Kollef MH, Casale TB. Probiotic prophylaxis of ventilator-associated pneumonia: a blinded, randomized, controlled trial. Am J Respir Crit Care Med. 2010;182:1058–64.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Hranjec T, Sawyer RG. Management of infections in critically ill patients. Surg Infect. 2014;15:474–8.CrossRef Hranjec T, Sawyer RG. Management of infections in critically ill patients. Surg Infect. 2014;15:474–8.CrossRef
12.
Zurück zum Zitat Siempos II, Ntaidou TK, Falagas ME. Impact of the administration of probiotics on the incidence of ventilator-associated pneumonia: a meta-analysis of randomized controlled trials. Crit Care Med. 2010;38:954–62.CrossRefPubMed Siempos II, Ntaidou TK, Falagas ME. Impact of the administration of probiotics on the incidence of ventilator-associated pneumonia: a meta-analysis of randomized controlled trials. Crit Care Med. 2010;38:954–62.CrossRefPubMed
13.
Zurück zum Zitat Barraud D, Bollaert PE, Gibot S. Impact of the administration of probiotics on mortality in critically ill adult patients. Chest. 2013;143:646–55.CrossRefPubMed Barraud D, Bollaert PE, Gibot S. Impact of the administration of probiotics on mortality in critically ill adult patients. Chest. 2013;143:646–55.CrossRefPubMed
14.
Zurück zum Zitat Petrof E, Dhaliwal R, Manzanares W, Johnstone J, Cook D, Heyland DK. Probiotics in the critically ill: a systematic review of the randomized trial evidence. Crit Care Med. 2012;40:3290–302.CrossRefPubMed Petrof E, Dhaliwal R, Manzanares W, Johnstone J, Cook D, Heyland DK. Probiotics in the critically ill: a systematic review of the randomized trial evidence. Crit Care Med. 2012;40:3290–302.CrossRefPubMed
15.
Zurück zum Zitat Wang J, Liu KX, Ariani F, Tao LL, Zhang J, Qu JM. Probiotics for preventing ventilator-associated pneumonia: a systematic review and meta-analysis of high-quality randomized controlled trials. PLoS One. 2013;8, e83934.CrossRefPubMedPubMedCentral Wang J, Liu KX, Ariani F, Tao LL, Zhang J, Qu JM. Probiotics for preventing ventilator-associated pneumonia: a systematic review and meta-analysis of high-quality randomized controlled trials. PLoS One. 2013;8, e83934.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Gu WJ, Wei CY, Yin RX. Lack of efficacy of probiotics in preventing ventilator-associated pneumonia: a systematic review and meta-analysis of randomized controlled trials. Chest. 2012;142:859–68.CrossRefPubMed Gu WJ, Wei CY, Yin RX. Lack of efficacy of probiotics in preventing ventilator-associated pneumonia: a systematic review and meta-analysis of randomized controlled trials. Chest. 2012;142:859–68.CrossRefPubMed
17.
Zurück zum Zitat Bo L, Li J, Tao T, Bai Y, Ye X, Hotchkiss RS, Kollef MH, Crooks NH, Deng X. Probiotics for preventing ventilator-associated pneumonia. Cochrane Database Syst Rev. 2014;(10)CD009066. Bo L, Li J, Tao T, Bai Y, Ye X, Hotchkiss RS, Kollef MH, Crooks NH, Deng X. Probiotics for preventing ventilator-associated pneumonia. Cochrane Database Syst Rev. 2014;(10)CD009066.
18.
Zurück zum Zitat McClave SA, Taylor BE, Martindale RG, Warren MM, Johnson DR, Braunschweig C, McCarthy MS, Davanos E, Rice TW, Cresci GA, Gervasio JM, Sacks GS, Roberts PR, Compher C, Society of Critical Care Medicine; American Society for Parenteral and Enteral Nutrition. Guidelines for the Provision and Assessment of Nutrition Support Therapy in the Adult Critically Ill Patient: Society of Critical Care Medicine (SCCM) and American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.). JPEN J Parenter Enteral Nutr. 2016;40:159–211.CrossRefPubMed McClave SA, Taylor BE, Martindale RG, Warren MM, Johnson DR, Braunschweig C, McCarthy MS, Davanos E, Rice TW, Cresci GA, Gervasio JM, Sacks GS, Roberts PR, Compher C, Society of Critical Care Medicine; American Society for Parenteral and Enteral Nutrition. Guidelines for the Provision and Assessment of Nutrition Support Therapy in the Adult Critically Ill Patient: Society of Critical Care Medicine (SCCM) and American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.). JPEN J Parenter Enteral Nutr. 2016;40:159–211.CrossRefPubMed
20.
Zurück zum Zitat Cui LH, Wang XH, Peng LH, Yu L, Yang YS. The effects of early enteral nutrition with addition of probiotics on the prognosis of patients suffering from severe acute pancreatitis. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 2013;25:224–8.PubMed Cui LH, Wang XH, Peng LH, Yu L, Yang YS. The effects of early enteral nutrition with addition of probiotics on the prognosis of patients suffering from severe acute pancreatitis. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 2013;25:224–8.PubMed
21.
Zurück zum Zitat Tan M, Lu X, Duan JW, Peng H, Zhu JC. Effects of probiotics on blood glucose levels and clinical outcomes in patients with severe cranocerebral trauma. Chin Crit Care Med. 2013;25:627–30. Tan M, Lu X, Duan JW, Peng H, Zhu JC. Effects of probiotics on blood glucose levels and clinical outcomes in patients with severe cranocerebral trauma. Chin Crit Care Med. 2013;25:627–30.
22.
Zurück zum Zitat Wang G, Wen J, Xu L, Zhou S, Gong M, Wen P, Xiao X. Effect of enteral nutrition and ecoimmunonutrition on bacterial translocation and cytokine production in patients with severe acute pancreatitis. J Surg Res. 2013;183:592–7.CrossRefPubMed Wang G, Wen J, Xu L, Zhou S, Gong M, Wen P, Xiao X. Effect of enteral nutrition and ecoimmunonutrition on bacterial translocation and cytokine production in patients with severe acute pancreatitis. J Surg Res. 2013;183:592–7.CrossRefPubMed
23.
Zurück zum Zitat López de Toro Martín-Consuegra I, Sanchez-Casado M, Pérez-Pedrero Sánchez-Belmonte MJ, López-Reina Torrijos P, Sánchez-Rodriguez P, Raigal-Caño A, Heredero-Galvez E, Zubigaray SB, Arrese-Cosculluela MÁ. The influence of symbiotics in multi-organ failure: randomised trial. Med Clin (Barc). 2014;143:143–9.CrossRef López de Toro Martín-Consuegra I, Sanchez-Casado M, Pérez-Pedrero Sánchez-Belmonte MJ, López-Reina Torrijos P, Sánchez-Rodriguez P, Raigal-Caño A, Heredero-Galvez E, Zubigaray SB, Arrese-Cosculluela MÁ. The influence of symbiotics in multi-organ failure: randomised trial. Med Clin (Barc). 2014;143:143–9.CrossRef
24.
Zurück zum Zitat Sanaie S, Ebrahimi-Mameghani M, Hamishehkar H, Mojtahedzadeh M, Mahmoodpoor A. Effect of a multispecies probiotic on inflammatory markers in critically ill patients: A randomized, double-blind, placebo-controlled trial. J Res Med Sci. 2014;19:827–33.PubMedPubMedCentral Sanaie S, Ebrahimi-Mameghani M, Hamishehkar H, Mojtahedzadeh M, Mahmoodpoor A. Effect of a multispecies probiotic on inflammatory markers in critically ill patients: A randomized, double-blind, placebo-controlled trial. J Res Med Sci. 2014;19:827–33.PubMedPubMedCentral
25.
Zurück zum Zitat Rongrungruang Y, Krajangwittaya D, Pholtawornkulchai K, Tiengrim S, Thamlikitkul V. Randomized controlled study of probiotics containing Lactobacillus casei (Shirota strain) for prevention of ventilator-associated pneumonia. J Med Assoc Thai. 2015;98:253–159. Rongrungruang Y, Krajangwittaya D, Pholtawornkulchai K, Tiengrim S, Thamlikitkul V. Randomized controlled study of probiotics containing Lactobacillus casei (Shirota strain) for prevention of ventilator-associated pneumonia. J Med Assoc Thai. 2015;98:253–159.
26.
Zurück zum Zitat Zeng J, Wang CT, Zhang FS, Qi F, Wang SF, Ma S, Wu TJ, Tian H, Tian ZT, Zhang SL, Qu Y, Liu LY, Li YZ, Cui S, Zhao HL, Du QS, Ma Z, Li CH, Li Y, Si M, Chu YF, Meng M, Ren HS, Zhang JC, Jiang JJ, Ding M, Wang YP. Effect of probiotics on the incidence of ventilator-associated pneumonia in critically ill patients: a randomized controlled multicenter trial. Intensive Care Med. 2016. Epub ahead of print. Zeng J, Wang CT, Zhang FS, Qi F, Wang SF, Ma S, Wu TJ, Tian H, Tian ZT, Zhang SL, Qu Y, Liu LY, Li YZ, Cui S, Zhao HL, Du QS, Ma Z, Li CH, Li Y, Si M, Chu YF, Meng M, Ren HS, Zhang JC, Jiang JJ, Ding M, Wang YP. Effect of probiotics on the incidence of ventilator-associated pneumonia in critically ill patients: a randomized controlled multicenter trial. Intensive Care Med. 2016. Epub ahead of print.
27.
Zurück zum Zitat Wheeler KE, Cook DJ, Mehta S, Calce A, Guenette M, Perreault MM, Thiboutot Z, Duffett M, Burry L. Use of probiotics to prevent ventilator-associated pneumonia: A survey of pharmacists' attitudes. J Crit Care. 2016;31:221–6.CrossRefPubMed Wheeler KE, Cook DJ, Mehta S, Calce A, Guenette M, Perreault MM, Thiboutot Z, Duffett M, Burry L. Use of probiotics to prevent ventilator-associated pneumonia: A survey of pharmacists' attitudes. J Crit Care. 2016;31:221–6.CrossRefPubMed
28.
Zurück zum Zitat DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7:177–88.CrossRefPubMed DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7:177–88.CrossRefPubMed
30.
Zurück zum Zitat Tempe JD, Steidel AL, Blehaut H, Hasselmann M, Lutun P, Maurier F. Prevention of diarrhea administering Saccharomyces boulardii during continuous enteral feeding. Sem Hosp. 1983;59:1409–12. Tempe JD, Steidel AL, Blehaut H, Hasselmann M, Lutun P, Maurier F. Prevention of diarrhea administering Saccharomyces boulardii during continuous enteral feeding. Sem Hosp. 1983;59:1409–12.
31.
Zurück zum Zitat Schlotterer M, Bernasconi P, Lebreton F, Wasserman D. Intérêt de Saccharomyces boulardii dans la tolérance digestive de la nutrition entérale à debit continu chez le brulé. Nutr Clin Métabol. 1987;1:31–4.CrossRef Schlotterer M, Bernasconi P, Lebreton F, Wasserman D. Intérêt de Saccharomyces boulardii dans la tolérance digestive de la nutrition entérale à debit continu chez le brulé. Nutr Clin Métabol. 1987;1:31–4.CrossRef
32.
Zurück zum Zitat Heimburger DC, Sockwell DG, Geels WJ. Diarrhea with enteral feeding: prospective reappraisal of putative causes. Nutrition. 1994;10:392.PubMed Heimburger DC, Sockwell DG, Geels WJ. Diarrhea with enteral feeding: prospective reappraisal of putative causes. Nutrition. 1994;10:392.PubMed
33.
Zurück zum Zitat Bleichner G, Blehaut H, Mentec H, Moyse D. Saccharomyces boulardii prevents diarrhea in critically ill tube-fed patients. A multicenter, randomized, double-blind placebo-controlled trial. Intensive Care Med. 1997;23:517–23.CrossRefPubMed Bleichner G, Blehaut H, Mentec H, Moyse D. Saccharomyces boulardii prevents diarrhea in critically ill tube-fed patients. A multicenter, randomized, double-blind placebo-controlled trial. Intensive Care Med. 1997;23:517–23.CrossRefPubMed
34.
Zurück zum Zitat Kecskés G, Belágyi T, Oláh A. Early jejunal nutrition with combined pre- and probiotics in acute pancreatitis--prospective, randomized, double-blind investigations [Article in Hungarian]. Magy Seb. 2003;56:3–8.PubMed Kecskés G, Belágyi T, Oláh A. Early jejunal nutrition with combined pre- and probiotics in acute pancreatitis--prospective, randomized, double-blind investigations [Article in Hungarian]. Magy Seb. 2003;56:3–8.PubMed
35.
Zurück zum Zitat Lu X, Han CM, Yu JX, Fu SZ. Preliminary comparative study on the effects of early enteral supplementation of synbiotics on severely burned patients. [Article in Chinese]. Zhonghua Shao Shang Za Zhi. 2004;20:198–201.PubMed Lu X, Han CM, Yu JX, Fu SZ. Preliminary comparative study on the effects of early enteral supplementation of synbiotics on severely burned patients. [Article in Chinese]. Zhonghua Shao Shang Za Zhi. 2004;20:198–201.PubMed
36.
Zurück zum Zitat Jain PK, McNaught CE, Anderson AD, MacFie J, Mitchell CJ. Influence of synbiotic containing Lactobacillus acidophilus La5, Bifidobacterium lactis Bb 12, Streptococcus thermophilus, Lactobacillus bulgaricus and oligofructose on gut barrier function and sepsis in critically ill patients: a randomised controlled trial. Clin Nutr. 2004;23:467–75.CrossRefPubMed Jain PK, McNaught CE, Anderson AD, MacFie J, Mitchell CJ. Influence of synbiotic containing Lactobacillus acidophilus La5, Bifidobacterium lactis Bb 12, Streptococcus thermophilus, Lactobacillus bulgaricus and oligofructose on gut barrier function and sepsis in critically ill patients: a randomised controlled trial. Clin Nutr. 2004;23:467–75.CrossRefPubMed
37.
Zurück zum Zitat Klarin B, Johansson ML, Molin G, Larsson A, Jeppsson B. Adhesion of the probiotic bacterium Lactobacillus plantarum 299v onto the gut mucosa in critically ill patients: a randomised open trial. Crit Care. 2005;9:R285–293.CrossRefPubMedPubMedCentral Klarin B, Johansson ML, Molin G, Larsson A, Jeppsson B. Adhesion of the probiotic bacterium Lactobacillus plantarum 299v onto the gut mucosa in critically ill patients: a randomised open trial. Crit Care. 2005;9:R285–293.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat McNaught CE, Woodcock NP, Anderson AD, MacFie J. A prospective randomised trial of probiotics in critically ill patients. Clin Nutr. 2005;24:211–9.CrossRefPubMed McNaught CE, Woodcock NP, Anderson AD, MacFie J. A prospective randomised trial of probiotics in critically ill patients. Clin Nutr. 2005;24:211–9.CrossRefPubMed
39.
Zurück zum Zitat Kotzampassi K, Giamarellos-Bourboulis EJ, Voudouris A, Kazamias P, Eleftheriadis E. Benefits of a symbiotic formula (Synbiotic 2000Forte) in critically ill trauma patients: early results of a randomized controlled trial. World J Surg. 2006;30:1848–55.CrossRefPubMed Kotzampassi K, Giamarellos-Bourboulis EJ, Voudouris A, Kazamias P, Eleftheriadis E. Benefits of a symbiotic formula (Synbiotic 2000Forte) in critically ill trauma patients: early results of a randomized controlled trial. World J Surg. 2006;30:1848–55.CrossRefPubMed
40.
Zurück zum Zitat Alberda C, Gramlich L, Meddings J, Field C, McCargar L, Kutsogiannis D, Fedorak R, Madsen K. Effects of probiotic therapy in critically ill patients: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr. 2007;85:816–23.PubMed Alberda C, Gramlich L, Meddings J, Field C, McCargar L, Kutsogiannis D, Fedorak R, Madsen K. Effects of probiotic therapy in critically ill patients: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr. 2007;85:816–23.PubMed
41.
Zurück zum Zitat Li YM. Adjuvant therapy for probiotics in patients with severe acute pancreatitis: an analysis of 14 cases. World Chin J Digestol. 2007;15:302–4. Li YM. Adjuvant therapy for probiotics in patients with severe acute pancreatitis: an analysis of 14 cases. World Chin J Digestol. 2007;15:302–4.
42.
Zurück zum Zitat Olah A, Belagyi T, Poto L, Romics Jr L, Bengmark S. Synbiotic control of inflammation and infection in severe acute pancreatitis: a prospective, randomized, double blind study. Hepatogastroenterolgy. 2007;54:590–4. Olah A, Belagyi T, Poto L, Romics Jr L, Bengmark S. Synbiotic control of inflammation and infection in severe acute pancreatitis: a prospective, randomized, double blind study. Hepatogastroenterolgy. 2007;54:590–4.
43.
Zurück zum Zitat Besselink MG, van Santvoort HC, Buskens E, Boermeester MA, van Goor H, Timmerman HM, Nieuwenhuijs VB, Bollen TL, van Ramshorst B, Witteman BJ, Rosman C, Ploeg RJ, Brink MA, Schaapherder AF, Dejong CH, Wahab PJ, van Laarhoven CJ, van der Harst E, van Eijck CH, Cuesta MA, Akkermans LM, Gooszen HG, Dutch Acute Pancreatitis Study Group. Probiotic prophylaxis in predicted severe acute pancreatitis: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;371:651–9.CrossRefPubMed Besselink MG, van Santvoort HC, Buskens E, Boermeester MA, van Goor H, Timmerman HM, Nieuwenhuijs VB, Bollen TL, van Ramshorst B, Witteman BJ, Rosman C, Ploeg RJ, Brink MA, Schaapherder AF, Dejong CH, Wahab PJ, van Laarhoven CJ, van der Harst E, van Eijck CH, Cuesta MA, Akkermans LM, Gooszen HG, Dutch Acute Pancreatitis Study Group. Probiotic prophylaxis in predicted severe acute pancreatitis: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;371:651–9.CrossRefPubMed
44.
Zurück zum Zitat Forestier C, Guelon D, Cluytens V, Gillart T, Sirot J, De Champs C. Oral probiotic and prevention of Pseudomonas aeruginosa infections: a randomized, double-blind, placebo-controlled pilot study in intensive care unit patients. Crit Care. 2008;12:R69.CrossRefPubMedPubMedCentral Forestier C, Guelon D, Cluytens V, Gillart T, Sirot J, De Champs C. Oral probiotic and prevention of Pseudomonas aeruginosa infections: a randomized, double-blind, placebo-controlled pilot study in intensive care unit patients. Crit Care. 2008;12:R69.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Klarin B, Wullt M, Palmquist I, Molin G, Larsson A, Jeppsson B. Lactobacillus plantarum 299v reduces colonisation of Clostridium difficile in critically ill patients treated with antibiotics. Acta Anaesthesiol Scand. 2008;52:1096–102.CrossRefPubMed Klarin B, Wullt M, Palmquist I, Molin G, Larsson A, Jeppsson B. Lactobacillus plantarum 299v reduces colonisation of Clostridium difficile in critically ill patients treated with antibiotics. Acta Anaesthesiol Scand. 2008;52:1096–102.CrossRefPubMed
46.
Zurück zum Zitat Knight DJ, Gardiner D, Banks A, Snape SE, Weston VC, Bengmark S, Girling KJ. Effect of synbiotic therapy on the incidence of ventilator associated pneumonia in critically ill patients: a randomised, double-blind, placebo-controlled trial. Intensive Care Med. 2009;35:854–61.CrossRefPubMed Knight DJ, Gardiner D, Banks A, Snape SE, Weston VC, Bengmark S, Girling KJ. Effect of synbiotic therapy on the incidence of ventilator associated pneumonia in critically ill patients: a randomised, double-blind, placebo-controlled trial. Intensive Care Med. 2009;35:854–61.CrossRefPubMed
47.
Zurück zum Zitat Barraud D, Blard C, Hein F, Marçon O, Cravoisy A, Nace L, Alla F, Bollaert PE, Gibot S. Probiotics in the critically ill patient: a double blind, randomized, placebo-controlled trial. Intensive Care Med. 2010;36:1540–7.CrossRefPubMed Barraud D, Blard C, Hein F, Marçon O, Cravoisy A, Nace L, Alla F, Bollaert PE, Gibot S. Probiotics in the critically ill patient: a double blind, randomized, placebo-controlled trial. Intensive Care Med. 2010;36:1540–7.CrossRefPubMed
48.
Zurück zum Zitat Frohmader TJ, Chaboyer WP, Robertson IK, Gowardman J. Decrease in frequency of liquid stool in enterally fed critically ill patients given the multispecies probiotic VSL#3: a pilot trial. Am J Crit Care. 2010;19:e1–11.CrossRefPubMed Frohmader TJ, Chaboyer WP, Robertson IK, Gowardman J. Decrease in frequency of liquid stool in enterally fed critically ill patients given the multispecies probiotic VSL#3: a pilot trial. Am J Crit Care. 2010;19:e1–11.CrossRefPubMed
49.
Zurück zum Zitat Ferrie S, Daley M. Lactobacillus GG as treatment for diarrhea during enteral feeding in critical illness: randomized controlled trial. JPEN J Parenter Enteral Nutr. 2011;35:43–9.CrossRefPubMed Ferrie S, Daley M. Lactobacillus GG as treatment for diarrhea during enteral feeding in critical illness: randomized controlled trial. JPEN J Parenter Enteral Nutr. 2011;35:43–9.CrossRefPubMed
50.
Zurück zum Zitat Sharma B, Srivastava S, Singh N, Sachdev V, Kapur S, Saraya A. Role of probiotics on gut permeability and endotoxemia in patients with acute pancreatitis: a double-blind randomized controlled trial. J Clin Gastroenterol. 2011;45:442–8.CrossRefPubMed Sharma B, Srivastava S, Singh N, Sachdev V, Kapur S, Saraya A. Role of probiotics on gut permeability and endotoxemia in patients with acute pancreatitis: a double-blind randomized controlled trial. J Clin Gastroenterol. 2011;45:442–8.CrossRefPubMed
51.
Zurück zum Zitat Tan M, Zhu JC, Du J, Zhang LM, Yin HH. Effects of probiotics on serum levels of Th1/Th2 cytokine and clinical outcomes in severe traumatic brain-injured patients: a prospective randomized pilot study. Crit Care. 2011;15:R290.CrossRefPubMedPubMedCentral Tan M, Zhu JC, Du J, Zhang LM, Yin HH. Effects of probiotics on serum levels of Th1/Th2 cytokine and clinical outcomes in severe traumatic brain-injured patients: a prospective randomized pilot study. Crit Care. 2011;15:R290.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Morrow LE, Gogineni V, Malesker MA. Synbiotics and probiotics in the critically ill after the PROPATRIA trial. Curr Opin Clin Nutr Metab Care. 2012;15:147–50.CrossRefPubMed Morrow LE, Gogineni V, Malesker MA. Synbiotics and probiotics in the critically ill after the PROPATRIA trial. Curr Opin Clin Nutr Metab Care. 2012;15:147–50.CrossRefPubMed
53.
Zurück zum Zitat Sheldon T. Dutch probiotics study is criticised for its "design, approval, and conduct". BMJ. 2010;340:c77.CrossRefPubMed Sheldon T. Dutch probiotics study is criticised for its "design, approval, and conduct". BMJ. 2010;340:c77.CrossRefPubMed
54.
Zurück zum Zitat Gou S, Yang Z, Liu T, Wu H, Wang C. Use of probiotics in the treatment of severe acute pancreatitis: a systematic review and meta-analysis of randomized controlled trials. Crit Care. 2014;18:R57.CrossRefPubMedPubMedCentral Gou S, Yang Z, Liu T, Wu H, Wang C. Use of probiotics in the treatment of severe acute pancreatitis: a systematic review and meta-analysis of randomized controlled trials. Crit Care. 2014;18:R57.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Hempel S, Newberry S, Ruelaz A, Wang Z, Miles JNV, Suttorp MJ, Johnsen B, Shanman R, Slusser W, Fu N, Smith A, Roth E, Polak J, Motala A, Perry T, Shekelle PG. Safety of Probiotics to Reduce Risk and Prevent or Treat Disease. Evidence Report/Technology Assessment No. 200. (Prepared by the Southern California Evidence-based Practice Center under Contract No. 290-2007-10062-I.) AHRQ Publication No. 11-E007. April 2011. Available at: www.ahrq.gov/clinic/tp/probiotictp.htm. Accessed 15 June 2016. Hempel S, Newberry S, Ruelaz A, Wang Z, Miles JNV, Suttorp MJ, Johnsen B, Shanman R, Slusser W, Fu N, Smith A, Roth E, Polak J, Motala A, Perry T, Shekelle PG. Safety of Probiotics to Reduce Risk and Prevent or Treat Disease. Evidence Report/Technology Assessment No. 200. (Prepared by the Southern California Evidence-based Practice Center under Contract No. 290-2007-10062-I.) AHRQ Publication No. 11-E007. April 2011. Available at: www.​ahrq.​gov/​clinic/​tp/​probiotictp.​htm. Accessed 15 June 2016.
56.
Zurück zum Zitat De Groote MA, Frank DN, Dowell E, Glode MP, Pace NR. Lactobacillus rhamnosus GG bacteremia associated with probiotic use in a child with short gut syndrome. Pediatr Infect Dis J. 2005;24:278–80.CrossRefPubMed De Groote MA, Frank DN, Dowell E, Glode MP, Pace NR. Lactobacillus rhamnosus GG bacteremia associated with probiotic use in a child with short gut syndrome. Pediatr Infect Dis J. 2005;24:278–80.CrossRefPubMed
57.
Zurück zum Zitat Vincent JL, Rello J, Marshall J, Silva E, Anzueto A, Martin CD, Moreno R, Lipman J, Gomersall C. International study of the prevalence and outcomes of infection in intensive care units. JAMA. 2009;302:2323–9.CrossRefPubMed Vincent JL, Rello J, Marshall J, Silva E, Anzueto A, Martin CD, Moreno R, Lipman J, Gomersall C. International study of the prevalence and outcomes of infection in intensive care units. JAMA. 2009;302:2323–9.CrossRefPubMed
58.
Zurück zum Zitat Ego A, Preiser JC, Vincent JL. Impact of diagnostic criteria on the incidence of ventilator-associated pneumonia. Chest. 2015;147:347–55.CrossRefPubMed Ego A, Preiser JC, Vincent JL. Impact of diagnostic criteria on the incidence of ventilator-associated pneumonia. Chest. 2015;147:347–55.CrossRefPubMed
59.
Zurück zum Zitat Kollef MH, Shorr A, Tabak YP, Gupta V, Liu LZ, Johannes RS. Epidemiology and outcomes of health-care-associated pneumonia: results from a large US database of culture positive pneumonia. Chest. 2005;128:3854–62.CrossRefPubMed Kollef MH, Shorr A, Tabak YP, Gupta V, Liu LZ, Johannes RS. Epidemiology and outcomes of health-care-associated pneumonia: results from a large US database of culture positive pneumonia. Chest. 2005;128:3854–62.CrossRefPubMed
60.
Zurück zum Zitat Rello J, Ollendorf DA, Oster G, Vera-Llonch M, Bellm L, Redman R, Kollef MH, VAP Outcomes Scientific Advisory Group. Epidemiology and outcomes of ventilator-associated pneumonia in a large US database. Chest. 2002;122:2115–21.CrossRefPubMed Rello J, Ollendorf DA, Oster G, Vera-Llonch M, Bellm L, Redman R, Kollef MH, VAP Outcomes Scientific Advisory Group. Epidemiology and outcomes of ventilator-associated pneumonia in a large US database. Chest. 2002;122:2115–21.CrossRefPubMed
61.
Zurück zum Zitat Branch-Elliman W, Wright SB, Howell MD. Determining the ideal strategy for ventilator-associated pneumonia. Am J Respir Crit Care Med. 2015;192:57–63.CrossRefPubMed Branch-Elliman W, Wright SB, Howell MD. Determining the ideal strategy for ventilator-associated pneumonia. Am J Respir Crit Care Med. 2015;192:57–63.CrossRefPubMed
62.
Zurück zum Zitat Hempel S, Newberry SJ, Maher AR, Wang Z, Miles JN, Shanman R, Johnsen B, Shekelle PG. Probiotics for the prevention and treatment of antibiotic-associated diarrhea: a systematic review and meta-analysis. JAMA. 2012;307:1959–69.CrossRefPubMed Hempel S, Newberry SJ, Maher AR, Wang Z, Miles JN, Shanman R, Johnsen B, Shekelle PG. Probiotics for the prevention and treatment of antibiotic-associated diarrhea: a systematic review and meta-analysis. JAMA. 2012;307:1959–69.CrossRefPubMed
63.
Zurück zum Zitat Goldenberg JZ, Ma SS, Saxton JD, Martzen MR, Vandvik PO, Thorlund K, Guyatt GH, Johnston BC. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev. 2013;5, CD006095. Goldenberg JZ, Ma SS, Saxton JD, Martzen MR, Vandvik PO, Thorlund K, Guyatt GH, Johnston BC. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev. 2013;5, CD006095.
64.
Zurück zum Zitat Glenting J, Beck HC, Vrang A, Riemann H, Ravn P, Hansen AM, Antonsson M, Ahrné S, Israelsen H, Madsen S. Anchorless surface associated glycolytic enzymes from Lactobacillus plantarum299v bind to epithelial cells and extracellular matrix proteins. Microbiol Res. 2013;168:245–53.CrossRefPubMed Glenting J, Beck HC, Vrang A, Riemann H, Ravn P, Hansen AM, Antonsson M, Ahrné S, Israelsen H, Madsen S. Anchorless surface associated glycolytic enzymes from Lactobacillus plantarum299v bind to epithelial cells and extracellular matrix proteins. Microbiol Res. 2013;168:245–53.CrossRefPubMed
65.
Zurück zum Zitat Gross G, Snel J, Boekhorst J, Smits MA, Kleerebezem M. Biodiversity of mannose-specific adhesion in Lactobacillus plantarum revisited: strain-specific domain composition of the mannose-adhesin. Benef Microbes. 2010;1:61–6.CrossRefPubMed Gross G, Snel J, Boekhorst J, Smits MA, Kleerebezem M. Biodiversity of mannose-specific adhesion in Lactobacillus plantarum revisited: strain-specific domain composition of the mannose-adhesin. Benef Microbes. 2010;1:61–6.CrossRefPubMed
Metadaten
Titel
Probiotic and synbiotic therapy in critical illness: a systematic review and meta-analysis
verfasst von
William Manzanares
Margot Lemieux
Pascal L. Langlois
Paul E. Wischmeyer
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Critical Care / Ausgabe 1/2016
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-016-1434-y

Weitere Artikel der Ausgabe 1/2016

Critical Care 1/2016 Zur Ausgabe

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.