Review Article

Saccharomyces boulardii CNCM I-745 for Prevention of Antibiotic-Associated Diarrhea and Clostridioides difficile in China: Systematic Review and Meta-Analysis

by Lynne V. McFarland1,2*, Tong Li 3

1Public Health Reserve Corps, Seattle, Washington 98195, USA

2McFarland Consulting, Seattle, Washington 98115 USA

3Department of Gastroenterology, the First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People’s Republic of China

*Corresponding author: Lynne V. McFarland, Public Health Reserve Corps, Seattle, Washington, USA.

Received Date: 1 May 2024

Accepted Date: 10May 2024

Published Date: 14 May 2024

Citation: McFarland LV, Li T (2024) Saccharomyces boulardii CNCM I-745 for Prevention of Antibiotic-Associated Diarrhea and Clostridioides difficile in China: Systematic Review and Meta-Analysis. J Dig Dis Hepatol 9: 208. https://doi.org/10.29011/2574-3511.100208

Abstract

Probiotics are commonly used for patients receiving antibiotics to prevent antibiotic-associated diarrhea (AAD). Randomized, controlled trials (RCTs) conducted in China are often not included in probiotic reviews and meta-analyses due to the difficulties in accessing Chinese journals and translating the Chinese language. Our aim is to evaluate the efficacy and safety of S. boulardii CNCM I-745 in AAD prevention trials done in China. A literature search was conducted using PubMed, Google Scholar and Chinese databases (CNKI, CMB) (from inception to June 2023) for RCTs assessing S. boulardii CNCM I-745 for the prevention of AAD and Clostridioides difficile infections (CDI). Inclusion criteria: RCT done in China, new prescription of antibiotic, randomized to S. boulardii and control groups. Random-effect or fixed-effects models were used depending upon the degree of heterogeneity and the risk of bias for each study was determined. This review was registered with Prospero (PROSPERO CRD 429831). The literature search found 361 articles, which were screened and 46 (8,201 participants) were included in the analysis. S. boulardii CNCM I-745 significantly reduced the incidence of AAD (RR=0.43, 95% C.I. 0.40,0.48, P < 0.0001) and CDI (RR=0.30, 95% C.I. 0.10, 0.87, P = 0.03).  S. boulardii CNCM I-745 had similar degrees of efficacy for both the prevention of AAD in trials done in China and for trials done in other countries. Saccharomyces boulardii CNCM I-745 was well tolerated and effective in trials in China for both the prevention of AAD and CDI.

Keywords: Antibiotics; Diarrhea; Saccharomyces boulardii; Probiotics; Clostridioides difficile

Introduction

Antibiotic-associated diarrhea (AAD) is a common complication of antibiotic use resulting from the disruption of the normally protective intestinal microbiome. While some factors that disrupt the intestinal microbiome are well-known (such as antibiotic exposure, advanced age, diet, and health status) [1], other factors relating to geography and ethnicity are less well documented, but may have important implications for microbiome-based clinical therapies [2]. The World Health Organization (WHO) defines acute diarrhea as ≥3 loose or liquid stools per day for ≥3 days and lasting <14 days [3]. Antibiotic-associated diarrhea is defined as diarrhea occurring while on antibiotics or within 8 weeks of antibiotics [1]. The prevalence of AAD in patients receiving antibiotics can be quite high, ranging from 5-37% in adults and may be higher in children (11-40%) [1]. The onset of AAD typically occurs during antibiotic administration, but delayed onset AAD may occur in 1020% for up to 8 weeks after antibiotics have been discontinued [1]. While the severity of AAD is typically mild-moderate in severity, 16% of cases are more severe, requiring further antibiotic treatments and 10-35% may be due to Clostridioides difficile infection (CDI), which can cause healthcare-associated outbreaks. Consequences of AAD include dehydration (especially in young children), higher mortality and morbidity, increased lengths-ofstays for inpatients (4-24 days), higher risk of colectomy (1-9%), higher healthcare costs and 25-28% of those with CDI develop recurrent form of the infection [1,4].

China is the second largest consumer of antibiotics, most commonly azithromycin, clindamycin and erythromycin [5,6,7]. However, only 25-39% of antibiotics prescribed in China are given appropriately [5]. The prevalence of AAD in China ranges from 14-35%, depending upon age, hospitalization status and co-morbidities [4,8]. Efforts prevent AAD include antibiotic stewardship programs and the adjunctive use of probiotics during antibiotic use [9,10]. A survey of 138 pediatric practitioners in China found 31% gave probiotics along with antibiotics, mainly due to concerns about antibiotic resistance or development of AAD [6].

Specific types of probiotics have been recommended in clinical guidelines for the prevention of AAD, but not all probiotic strains have shown efficacy [10,11,12]. Guidelines for probiotic use to prevent AAD in children focused on Asia-Pacific populations had strong recommendations for S. boulardii CNCM I-745 and L. rhamnosus GG [13]. These two stains were also recommended for the prevention of AAD in adults in the Asia-Pacific region [14]. Indeed, the efficacy for probiotics is strain-specific and each strain or multi-strained blend must be assessed separately to determine efficacy for AAD prevention [15]. Prior meta-analyses have found Saccharomyces boulardii CNCM I-745 and Lactobacillus rhamnosus GG to have the strongest evidence for the prevention of AAD, but often have not included RCTs published in non-

English languages, due to translation problems and limited access to Chinese journals [16,17,18,19,20]. 

Saccharomyces boulardii CNCM I-745 is a strain of probiotic that has a long history of use and has demonstrated significant efficacy and safety for a variety of diseases ranging from prevention of AAD and CDI, treatment of pediatric acute gastroenteritis, to reduction of side-effects of H. pylori eradication therapies [17,20,21,22,23]. S. boulardii CNCM I-745 reaches high levels in the intestine within 2-3 days, is cleared from the colon within 5 days and can be given concurrently with antibiotics, as the yeast is not susceptible to antibiotics [24]. The ability of S. boulardii CNCM I-745 to be therapeutic is due to multiple mechanisms-of-action: direct inhibition of pathogen growth or destruction of pathogenic toxins, interference with pathogen/ toxin attachment on enteric cell surfaces, improving intestinal cell health, reduction of water secretion, immune system regulation and restoring the normally protective microbiome barrier layer [14,21,25]. S. boulardii CNCM I-745 has a remarkable safety profile in that this strain has been given to a wide variety of patient types (children, adults and the elderly, hospitalized inpatients and outpatients, patients with acute and chronic conditions) and data gathered since its introduction in Europe in the 1950s has shown few adverse reactions, most were thirst and constipation [22]. Genetic transfer of antibiotic-resistant genes has not been observed with this strain [26]. In rare cases (1/5.6 million users), immunocompromised patients or inpatients with central catheters have developed fungemia [23].

Our aim in this study is to determine its efficacy and safety of Saccharomyces boulardii CNCM I-745 for the prevention of AAD and CDI in trials done in China using a comprehensive literature search to uncover trials not previously found in nonChinese databases.

Materials and Methods

Literature search

The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed [27]. The PRISMA checklist is provided (Supplementary Table S1). Recent recommendations for reporting probiotic meta-analyses were also followed [28]. The project and protocol were prospectively registered with PROSPERO database of systematic reviews [www.

crd.york.ac.uk/PROSPERO/] on May 25, 2023 (CRD 429831).

 The following databases were searched [PubMed, Google Scholar, and Chinese databases (Chinese Biomedical Literature (CBM) and Chinese National Knowledge Infrastructure (CNKI)] from database inception to June 2023 to identify randomized controlled trials (RCTs) of probiotic use (limited to Saccharomyces boulardii CNCM I-745) to prevent antibiotic-associated diarrhea (AAD), including Clostridioides difficile infections (CDI) using randomized controlled trials done in China. The search terms were used: (antibiotic-associated diarrhea OR C. difficile AND Saccharomyces boulardii AND China). No language restrictions were imposed and publications in Chinese were translated into English.

Secondary searches of grey literature included reference lists, authors, reviews, meeting abstracts websites and clinicaltrials.gov for unpublished trials.  A recursive search was also performed, using the bibliographies of all obtained articles. 

Study selection

Inclusion criteria included: randomized, controlled clinical trials (RCTs) in children or adults receiving new prescription of antibiotics, comparison of S. boulardii versus control interventions and published in peer-reviewed journals. Only formulations of S. boulardii CNCM I-745 fulfilling the standard definition (must be living microbe, of adequate dose and conferring a health effect on the host) were included [29]. This definition excludes dead or heat-killed microbes and prebiotics. As bacterial and fungal taxonomies shift over time, the most current strain designations are presented in this review and strain identification was confirmed with the original authors or the manufacturer whenever possible. Trials with the primary outcome of prevention of AAD or CDI were included. Studies were included only if performed in China.

Exclusion criteria included: study not done in China, nonhuman studies, early phase 1 or 2 safety or mechanism of action studies, no control group, probiotic or AAD not well-described, clinical trials for the treatment of existing diarrhea, primary outcome was for the eradication of H. pylori, other probiotic strain(s) contained in the probiotic formulation, non-China setting, reviews and duplicate reports. Cross-over trials were excluded due to the potential for effect carry-over after short wash-out periods used in these trials and direct probiotic-probiotic studies were excluded if no non-probiotic control group was included.

Data extraction

The literature was searched independently by two coauthors (LM, LT). Data from all RCTs were extracted using a standardized data extraction form (Supplementary data, Form S1) initially completed by one co-author and then each study was independently scored by the other co-author following the standard methods for systematic reviews and meta-analysis [27,30]. Any disagreements were discussed until consensus was reached. The data extracted included PICOS data: (1) population (neonate, pediatric or adults, age range and country), (2) intervention (type of probiotic or controls used, daily doses, formulation, duration and follow-up times), (3) comparisons (type of control group either placebo or open, unblinded), (4) Primary outcomes (incidence of AAD or CDI), (5) secondary outcomes (stool frequency, length of hospitalization, cost data,  safety data and clinical characterization of AAD). In addition, data on potential confounding factors were collected: type of antibiotic given, type of control, study quality, setting (inpatient or outpatient), dose and duration of probiotic. For missing data not reported in the published article, we attempted to contact the author or co-authors to obtain the missing data.

Intervention

The intervention was the yeast probiotic strain Saccharomyces boulardii CNCM I-745 (Yihuo ™ or Bioflor™, Biocodex, France). If the strain designation was not reported in the published study, the strain was verified with the manufacturer or website. The intervention may be given in oral administration as capsules or sachets. The duration of the intervention was typically given for the duration of the antibiotic(s), but may have been continued after the antibiotics are discontinued.

Outcomes

The primary outcomes include the efficacy for the reduction of AAD incidence and the reduction of CDI incidence. The outcome measures included: the number of patients developing AAD (diarrhea defined as at least two loose/watery stools in young children or ≥3 loose/watery stools in older children and adults for 48 hours, not caused by rotavirus, with an onset while on antibiotics or within 8 weeks of antibiotic cessation or CDI (AAD with positive C. difficile toxin result). Secondary outcomes included: daily stool frequency at the end of the intervention, length of hospitalization, clinical presentation of AAD (measured by duration of AAD, severity of AAD symptoms and treatment effectiveness rating), costs of medical care and frequency of adverse events. Severe AAD was defined differently in the RCTs, but typically included “diarrhea with electrolyte disorders, severe dehydration, acidosis or other symptoms of toxicity”. Outcomes may be documented by patients/parents using daily diaries or by healthcare providers if hospitalized.

Risk of bias

Each included RCT was reviewed for quality and risk of bias and scored independently by both co-authors using standard methods [31]. The risk of bias (RoB) assessed with the RoB 2.0 tool and was graded (high,  some concerns, or low) for each of five domains of bias (randomization process, deviations from intended interventions, missing outcome data, measurement of outcome, selection of reported result) and a summary table and figure of bias was generated [32].

Statistical Analysis

As recommended by experts, inclusion of studies in our meta-analysis also required at least two RCTs using a common outcome measure [28]. Statistical analysis and generation of forest plots of pooled summary estimates was performed using Stata software version 16 (Stata Corporation, College Station, Texas) with meta-analysis modules [33]. Dichotomous outcomes were assessed using relative risks (RR) and 95% confidence intervals (C.I.) and continuous outcomes were assessed using standardized mean difference (SMD) and 95% C.I. using standard methods [31]. The significance level was set at P-value < 0.05. Heterogeneity across trials was evaluated using the I2 statistic, 0% indicating none and ≥50% indicating a high degree of heterogeneity across the trials [33]. Random-effects models were used for the meta-analysis if significant heterogeneity was found (I2>50%), otherwise fixed-effects models were used.  Publication bias was assessed using funnel plots and the Egger’s test [33]. Subgroup analysis was used to explore sources of heterogeneity and was assessed with the Cochrane Q test [31]. A priori subgroup analyses based on factors that might influence the magnitude of efficacy estimates were planned for the following: (a) age,  (b) daily dose S. boulardii, (c) risk of bias, (d) indication for antibiotic use (upper respiratory disease, gastrointestinal, etc.), (e) type of patient (inpatient/outpatients), (f) time of intervention initiation from onset of diarrhea, (g) rural versus urban setting, (h) route antibiotic given (IV or oral) and (i) extent of blinding. Trials were pooled in the meta-analysis if a common outcome was used and there were at least two trials within each sub-group. Sequential sensitivity analysis was done to explore the extent outcomes were dependent upon a particular trial.

Results

The literature search resulted in 361articles that were initially screened and 233 were excluded during the initial screening (Figure 1): duplicate publications (n=88), reviews (n=73), cross-over studies (n=23), pre-clinical studies (n=18) or other miscellaneous reasons (n=31). Full articles (n=128) were reviewed and 82 were excluded: treatment of existing AAD (n=18), H. pylori eradication trials (n=17), no non-probiotic controls (n=16), unclear AAD definition (n=14), direct probiotic to probiotic studies (n=12), probiotic tested was other than S. boulardii (n=2), treatment of rotaviral diarrhea (n=2) and unclear outcome data (n=1). A total of 46 RCTs published 2011-2022 were included (8,201 participants)

[34-79].  Only four RCTs were found in commonly used nonChinese databases [54,59,60,64] and the other 42 trials were found using CNKI/CMI databases. A funnel plot (Supplementary Figure S1) showed significant publication bias for the included studies Egger’s test: t= -3.15, P = 0.003) due to a lack of RCTs showing no significant efficacy of S. boulardii CNCM I-745.

 

Figure 1: PRISMA study inclusion flow-chart.

Study participant characteristics 

There was a mean of 178 + 125 enrolled subjects/trial (range 46-552), as shown in (Table 1). Most trials (n=44, 96%) were in pediatric subjects (aged neonates to 16 years old) and two trials were in elderly (>65 years old) patients [44,72]. All patients were hospitalized.

The types of antibiotics used varied, with most RCTs (n=23, 50%) using different types of antibiotics while four RCTs enrolled patients given only a single type of antibiotic (9%), but the types of antibiotics were not reported in 19 (41%) trials (Table 1). Most of the antibiotics were given for upper respiratory infections (38, 83%) or mixed types of infections (5, 11%) or not reported (n=3, 6%). The duration of antibiotics ranged from 5 to 21 days and most (26, 56.5%) were given only intravenously while a few RCTs (n=3, 6.5%) enrolled patients given either IV or oral antibiotics. The antibiotic route was not reported in 17 (37%) of the RCTs.

Ref.

Year

No.

Age range of inclusion

Type of antibiotic(s) given (route)

Antibiotic indication

Type of control

Dose (mg/day)1 for S. boulardii

Duration given (days)2

Follow up (days)

Attrition (%)

Bai SX et al. [34]

2012

120

6 mon-3 y

C,M,Ot (nr)

RTI

Open

500

On abx

5

5

Cao TR [35]

2013

70

6 mon- 2 y

nr (IV)

RTI

Open

500

On abx

14

5.7

Chen JW et al. [36]

2013

236

6 mon- 14 y

nr (nr)

RTI

Open

250

On abx

0

0

Chen JB [37]

2013

120

6 mon- 3 y

C,M,P (IV)

RTI

Open

500

On abx

0

0

Chen Juan [38]

2013

64

2 mon- 3.5 y

C,M,P (IV)

RTI

Open

250/500

On abx

0

0

Chen W et al. [39]

2012

100

1 y- 14 y

nr (IV)

RTI

Open

500

On abx

0

0

Chen YY [40]

2020

94

5 mon- 3 y

Ot (IV)

RTI

Open

500

5

0

0

Dong G [41]

2015

80

5 mon-12 y

C,P (IV)

RTI

Open

500

7

0

0

Dong X et al. [42]

2014

62

2 mon- 3 y

C, M (IV)

RTI

Open

500

On abx

0

0

Feng X et al. [43]

2015

58

7 mon- 3 y

C, M, P (nr)

RTI

Open

250/500

LOS

0

0

Guo T et al. [44]

2018

127

>65 yrs

C, E, L (IV)

RTI

Placebo

500

On abx +7

28

0

Huang MH [45]

2018

84

2 mon- 3 y

C,M (IV)

RTI

Open

250

7

0

0

Huang S et al. [46]

2017

66

1 y- 3 y

C,M,Ot (Or/IV)

RTI

Open

300

On abx

0

0

Li B et al. [47]

2011

46

5 mon- 7 y

nr (IV)

RTI

Open

500

On abx

0

0

Li JM et al. [48]

2014

2103

1 mon- 3 y

C, M, P (IV)

RTI

Open

250/500

5

0

0

Li YY et al. [49]

2021

298

<15 d

nr (IV)

Mixed

Open

250

14

14

0

Li YF [50]

2013

237

<6 mon- 1 y

nr (IV)

RTI

Open

500

On abx

0

0

Lin X et al. [51]

2018

168

1 mon- 3 yrs

C (nr)

RTI

Open

250/500/1000

7-10

0

0

Lou QY et al. [52]

2020

499

1 mon- 6 y

C, M, P (nr)

RTI

Open

250/500

7-8

14

25.8

Peng XH [53]

2018

276

2 mon- 3 y

nr (nr)

RTI

Open

250

5 + 5

0

0

Shan LS et al. [54]

2013

333

6 mon- 14 y

A, C, Ot (IV)

RTI

Open

500

14

14

15

Shan LS et al. [55]

2014

468

6 mon- 14 y

C, Ot (IV)

RTI

Open

500

8

6

0

Song YH et al. [56]

2012

60

6 mon- 14 y

Nr (nr)

RTI

Open

500

On abx

0

0

Tan G et al. [57]

2022

150

25 mon-3 y

nr (IV)

RTI

Open

250

7

0

0

Tang M [58]

2014

146

< 14 d

C,P (nr)

RTI

Open

250

On abx

0

0

Tao Z et al. [59]

2016

120

<16 y

C (IV)

RTI

Open

250/500

On abx

0

0

Wan CM et al. [60]

2017

408

1 mon- 3 y

Mixed (Or/IV)

RTI/ mixed

Open

250

On abx

14

0

Wang LH[61]

2014

100

9 mon- 7 y

nr (nr)

RTI

Open

500

On abx

0

0

Wang XY[62]

2013

126

1 y- 3 y

nr (IV)

RTI

Open

250

On abx

0

0

Wang ZZ [63]

2013

129

< 2 y

nr (IV)

RTI

Placebo

500

On abx +3

53

17

Wu XY et al. [64]

2015

180

6 mon- 8 y

C, M, P (IV)

RTI

Open

125/250/500

7

0

0

Wu YX [65]

2012

68

6 mon – 3 y

C,Ot (IV)

RTI

Open

250

On abx

0

0

Xiao J [66]

2018

92

6 mon-5 y

C (nr)

RTI

Open

250/500

On abx

7

0

Xu H [67]

2013

51

2 mon- 3 y

nr (IV)

RTI

Open

250/500

On abx

0

0

Xu LF et al. [68]

2015

552

3 mon– 14 y

C, M, P, Ot (Or/IV)

RTI or UTI

Open

500

7

0

13

Yang X [69]

2015

200

1 mon- 13 y

nr (nr)

nr

Open

250/500

On abx

14

0

Yao TX et al. [70]

2014

240

1 mon- 3 y

nr (nr)

Mixed

Open

500

On abx

0

0

Yu M [71]

2014

2714

6 mon- 3 y

nr(IV)

RTI

Open

250/500

On abx

0

0

Zhang DM et al. [72]

2017

163

>65 y

C, Ot (nr)

nr

Open

1000

21

63

0

Zhang J et al. [73]

2013

312

3 mon-3 y

nr (nr)

Mixed

Open

250/500

On abx

0

0

Zhang WX [74]

2013

220

1 mon- 8 y

Mixed (nr)

nr

Open

250/500

On abx

0

0

Zhang Y et al. [75]

2018

326

6 mon– 12 y

A, C, Ot (IV)

RTI

Open

250/500

5

0

0

Zhao G [76]

2013

96

5 mon-3 y

 nr (IV)

RTI

Open

250/500

On abx

0

0

Zhao GD[77]

2018

89

1 y- 10 y

nr (IV)

RTI

Open

250/500

On abx

0

0

Zheng SQ [78]

2013

186

10 – 16 mon

Broad-spect (nr)

RTI

Open

250

On abx

0

0

Zhu HY [79]

2017

100

1.5 y- 2 y

nr (nr)

RTI

Open

250

On abx

0

0

Table 1: Study population characteristics of 46 included trials for the prevention of AAD. 1Dose/day may vary by age of patient; 2 On abx + days extended post-antibiotics; 3Li J (treatment AAD arm excluded); 4Yu M (two study arms excluded, on for treatment of AAD by S. boulardii and another with probiotic blend). A, ampicillin; C, cephalosporin; d, days; E, ertapenum; IV, intravenous route; L, levofloxacin; LOS, during length of stay in hospital; M, macrolide; mon, months; nr, not reported in paper; No., number enrolled; OM, otitis media; On abx, given while on antibiotics; Or, oral route; Ot, other types of antibiotics; P, penicillin; Ref, reference; RTI, upper or lower respiratory tract infection; UTI, urinary tract infection; wk, weeks; y, years old.

Intervention/control characteristics

Most trials (n=44, 96%) used open controls, while two (4%) RCTs used a blinded placebo control, as shown in Table 1 [44,63]. The most common formulation for S. boulardii was as a powder (n=41 trials, 89%) or in capsules (n=1, 2%) [72], but was not reported in four trials. Many RCTs used age-dependent daily doses for S. boulardii (n=16, 35%, ranging from 150 mg to 1 g/d), while others gave a fixed daily dose, regardless of age: 250 mg/d (n=11, 24%), 300 mg/d (n=1, 2%), 500 mg/d (n=17, 37%) or 1000 mg/d (n=1, 2%). Most trials continued the intervention for the duration of the antibiotic (n= 42, 91%), three (6%) continued the intervention after antibiotics were discontinued [44,53,63] and one RCT continued for the length of the hospitalization [43]. Only 12/46 (26%) of the RCTs followed subjects post-intervention for delayed-onset AAD (follow-up times ranged from 5-63 days), while most RCTs (n=34, 74%) did not follow subjects after antibiotics were discontinued. No loss to follow-up (attrition) was reported for 40 (87%) of the trials, while six (13%) trials reported 5-26% attrition [34,35,52,54,63,68]. In 31 (67.4%) of the trials, the interventions were started within 24 hours of the antibiotic, but the initiation time was not reported in 15 (32.6%) trials.

Primary outcome: prevention AAD

Efficacy to prevent AAD was reported all included trials (n=46), as shown in (Table 2). However, the reported definitions for AAD varied (Supplementary Table S2) with 5 (11%) not reporting how AAD was defined.  Most RCTs (n=41, 89%) did report their definition of AAD in their papers, typically representing a change in stool consistency, increased frequency of watery/loose stools for longer than one day, which was associated with antibiotic use or shortly afterwards. The incidence of AAD ranged from 3/100 to 38/100 (mean 15.0 ± 7.2/100) in S. boulardii groups compared to 9/100 to 56/100 (mean 35.9 ± 10.7/100) in controls. The pooled RR for AAD was significantly lower for S. boulardii compared to controls (RR= 0.43, 95% C.I. 0.40, 0.48, P<0.0001, I2=5.5%), as shown in (Figure 2).

Ref.

Year

AAD in S. boulardii, number (%)

AAD in

controls, number (%)

P-value

CDI in S. boulardii, number (%)

CDI in control, number (%)

% AE in S. boulardii

% AE in controls

Bai SX et al [34]

2012

20/60

(33.3)

32/60 (53.3)

<0.01

nr

nr

0, state

0, state

Cao TR [35]

2013

4/34 (11.8)

12/32 (37.5)

<0.05

nr

nr

0, state

0, state

Chen JW et al [36]

2013

23/121

(19.0)

43/115

(37.4)

<0.05

nr

nr

0, state

0, state

Chen JB [37]

2013

4/60 (6.7)

14/60 (23.2)

<0.05

nr

nr

nr

nr

Chen Juan [38]

2013

6/32 (18.8)

18/32 (56.2)

<0.05

nr

nr

0, state

0, state

Chen W et al [39]

2012

7/50 (14.0)

18/50 (36.0)

<0.05

nr

nr

nr

nr

Chen YY [40]

2020

5/47 (10.6)

14/47 (29.8)

0.001

nr

nr

nr

nr

Dong G [41]

2015

4/40 (10)

13/40 (32.5)

<0.05

nr

nr

nr

nr

Dong X et al [42]

2014

5/31 (16.1)

11/31 (35.5)

<0.01

nr

nr

0, state

0, state

Feng X et al [43]

2015

8/29 (27.6)

15/29 (51.7)

<0.05

nr

nr

nr

nr

Guo T et al [44]

2018

7/63 (11.1)

17/64 (26.6)

0.04

nr

nr

19

11

Huang MH [45]

2018

7/ 42 (16.7)

15/42 (35.7)

<0.05

nr

nr

0, state

0, state

Huang S et al [46]

2017

3/33 (9.1)

9/33 (33.0)

<0.05

nr

nr

nr

nr

Li B et al [47]

2011

3/23 (13.0)

9/23 (34.6)

< 0.05

nr

nr

nr

nr

Li JM et al [48]

2014

26/150

(17.3)

25/60 (41.7)

<0.001

nr

nr

nr

nr

Li YY et al [49]

2021

17/148

(11.4)

44/150

(29.3)

<0.001

nr

nr

0, state

0, state

Li YF [50]

2013

21/120

(17.5)

43/117

(36.7)

<0.05

nr

nr

0, state

0, state

Lin X et al [51]

2018

17/90

(18.9)

34/78 (43.6)

<0.01

nr

nr

nr

nr

Lou QY et al [52]

2020

40/182

(22.0)

55/164

(33.5)

0.02

nr

nr

0, state

0, state

Peng XH [53]

2018

21/140

(15.0)

64/136

(47.1)

<0.05

nr

nr

0, state

0, state

Shan LS et al [54]

2013

6/139 (4.3)

28/144

(19.4)

<0.001

1/139 (0.7)

8/144 (5.6)

0, state

0, state

Shan LS et al [55]

2014

19/233

(8.2)

63/235

(26.8)

<0.05

2/233 (0.8)

11/235

(4.7)

0, state

0, state

Song YH et al [56]

2012

3/30 (10.0)

10/30 (33.3)

0.03

nr

nr

nr

nr

Tan G et al [ 57]

2022

9/75 (12)

26/75 (34.7)

<0.001

nr

nr

nr

nr

Tang M [58]

2014

15/73

(20.6)

34/73 (46.6)

<0.05

nr

nr

3

4

Tao Z et al [59]

2016

13/60

(21.7)

23/60 (38.3)

<0.05

nr

nr

0, state

0, state

Wan CM et al [60]

2017

27/213

(12.7)

89/195

(45.6)

<0.001

nr

nr

0, state

0, state

Wang LH [ 61]

2014

19/50

(38.0)

28/50 (56.0)

<0.05

nr

nr

nr

nr

Wang XY [62]

2013

9/60 (15.0)

20/66 (30.3)

<0.05

nr

nr

nr

nr

Wang ZZ [63]

2013

9/56 (16.1)

20/51 (39.2)

<0.05

nr

nr

nr

nr

Wu XY et al [64]

2015

21/90

(23.3)

30/90 (33.3)

<0.05

nr

nr

0, state

0, state

Wu YX [65]

2012

6/34 (17.6)

16/34 (47.0)

<0.05

nr

nr

nr

nr

Xiao J [66]

2018

3/46 (6.5)

12/46 (26.1)

< 0.05

nr

nr

0, state

0, state

Xu H [67]

2013

4/21 (19.1)

14/30 (46.7)

0.04

nr

nr

0, state

0, state

Xu LF et al [68]

2015

8/160 (5.0)

49/240

(20.4)

0.007

nr

nr

0, state

0, state

Yang X [69]

2015

17/100

(17.0)

51/100

(51.0)

<0.05

nr

nr

0, state

0, state

Yao TX et al [70]

2014

4/120 (3.3)

11/120 (9.2)

<0.05

nr

nr

nr

nr

Yu M [71]

2014

12/150

(8.0)

23/121

(19.0)

<0.05

nr

nr

0, state

0, state

Zhang DM et al [72]

2017

12/81

(14.8)

23/82 (28.0)

<0.05

3/81 (3.7)

4/82 (4.9)

nr

nr

Zhang J et al [73]

2013

11/150

(7.3)

38/162

(23.5)

<0.05

nr

nr

nr

nr

Zhang WX [74]

2013

8/110 (7.3)

29/110

(26.4)

<0.01

nr

nr

nr

nr

Zhang Y et al [75]

2018

38/171

(22.2)

53/155

(34.2)

<0.05

nr

nr

nr

nr

Zhao G [76]

2013

6/50 (12.0)

17/46 (37.0)

<0.05

nr

nr

nr

nr

Zhao GD [77]

2018

9/45 (20.0)

23/44 (52.3)

<0.05

nr

nr

0, state

0, state

Zheng SQ [78]

2013

21/100

(21.0)

38/86 (44.2)

<0.01

nr

nr

nr

nr

Zhu HY [79]

2017

4/50 (8.0)

13/50 (26.0)

<0.05

nr

nr

nr

nr

Table 2: Prevention of antibiotic-associated diarrhea (AAD), C. difficile infection or adverse events in 46 included trials. AAD, antibiotic-associated diarrhea, excluding rotaviral diarrhea; AE, adverse event; CDI, Clostridioides difficile infection;  nr, not reported; Ref, reference;  S., Saccharomyces; state, only statement of ‘No adverse events noted’.

Primary outcome: Efficacy to prevent CDI

Only three trials reported CDI as an outcome (Table 2) in their trials [54,55,72]. As shown in (Figure 3), the risk for CDI was significantly reduced for S. boulardii CNCM I-745 compared to controls (RR= 0.30, 95% C.I. 0.10, 0.87, P = 0.03, I2=22.9%).

Figure 3: Forest plot of efficacy for prevention of CDI:  S. boulardii compared to controls.DL= Der Simonian-Laird.

Secondary outcomes

Several secondary outcomes had sufficient data for analysis:

daily stool frequency at study end, clinical characterization of AAD (duration, severe AAD, effectiveness rating) and safety. Other a priori secondary outcomes were infrequently reported (rural versus urban and cost-effectiveness) and could not be analyzed.

Stool frequency

Only 16/46 (35%) reported the daily stool frequency during the intervention/control phase and 30 (65%) did not report this outcome measure (Table 3). Those given S. boulardii had significantly fewer mean stools/day compared to the controls (Figure 4), SMD: --1.25 stools/day, 95% C.I. -1.35, -1.16, P< 0.0001, I2=96%).

Figure 2: Forest plot of efficacy for prevention of AAD: S. boulardii compared to controls.DL, Der Simonian-Laird.

No. Sb

mean stools/d in Sb

Std dev in Sb

No. control

mean stools/d in control

Std dev in controls

# Sb

mean days AAD Sb

Std dev Sb

# controls

mean AAD days control

Std Dev controls

Bai SX et al [34]

2012

60

1.8

0.5

60

3.2

0.9

19

3.2

1.0

27

5.1

1.8

Cao TR [35]

2013

nr

nr

nr

nr

nr

nr

4

2.3

0.13

12

4.6

0.4

Chen JW et al [36]

2013

nr

nr

nr

nr

nr

nr

23

3.1

0.7

43

4.6

0.94

Chen JB [37]

2013

60

3.1

0.8

60

3.9

1.1

4

2.0

1.5

14

4.0

2.5

Chen Juan [38]

2013

nr

nr

nr

nr

nr

nr

6

2.4

1.3

18

4.3

1.8

Chen W et al [39]

2012

50

3.1

0.7

50

3.8

1.1

7

2.8

1.2

18

4.0

2.1

Chen YY [40]

2020

47

4.2

0.3

47

5.1

0.56

5

3.1

0.52

14

4.2

0.74

Dong G [41]

2015

nr

nr

nr

nr

nr

nr

4

0.3

1.2

13

5.4

3.1

Dong X et al [42]

2014

nr

nr

nr

nr

nr

nr

5

3.1

0.43

11

4.23

1.26

Feng X et al [43]

2015

29

1.7

0.4

29

3.4

0.8

8

3.1

0.9

15

5.2

1.7

Guo T et al [44]

2018

63

2.69

1.2

64

4.3

1.6

nr

nr

nr

nr

nr

nr

Huang MH [45]

2018

nr

nr

nr

nr

nr

nr

7

3.11

0.42

15

4.22

1.25

Huang S et al [46]

2017

nr

nr

nr

nr

nr

nr

3

1.6

0.7

9

3.9

0.7

Li B et al [47]

2011

23

3.0

0.7

23

3.8

1.1

3

2.7

1.3

9

4.1

2.1

Li JM et al [48]

2014

nr

nr

nr

nr

nr

nr

26

3.35

0.9

25

4.64

1.08

Li YY et al [49]

2021

148

2.11

0.95

150

3.15

1.27

17

3.15

1.28

44

4.75

2.16

Li YF [50]

2013

nr

nr

nr

nr

nr

nr

21

3.1

1.2

43

4.3

1.5

Lin X et al [51]

2018

90

3.68

1.1

78

5.38

1.5

17

3.73

1.33

34

4.84

1.32

Lou QY et al [52]

2020

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

Peng XH [53]

2018

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

Shan LS et al [54]

2013

nr

nr

nr

nr

nr

nr

6

2.3

0.9

28

8.97

1.1

Shan LS et al [55]

2014

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

Song YH et al [56]

2012

nr

nr

nr

nr

nr

nr

3

2.7

0.3

10

4.4

 0.93

Tan G et al [57]

2022

75

2.83

0.47

75

5.41

0.36

9

2.4

1.2

26

5.1

1.7

Tang M [58]

2014

73

5.3

1.5

73

8.2

1.4

15

3.4

1.3

34

5.0

 1.7

Tao Z et al [59]

2016

60

1.13

0.39

60

2.32

0.43

13

3.78

1.34

23

4.67

1.47

Wan CM et al [ 60]

2017

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

Wang LH [ 61]

2014

nr

nr

nr

nr

nr

nr

19

2.45

0.9

28

4.2

0.9

Wang XY [62]

2013

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

Wang ZZ [63]

2013

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

Wu XY et al [64]

2015

nr

nr

nr

nr

nr

nr

21

3.19

0.68

30

5.24

0.79

Wu YX [65]

2012

nr

nr

nr

nr

nr

nr

6

2.7

1.3

16

4.1

1.8

Xiao J [66]

2018

46

6.4

1.3

46

4.3

1.24

3

3.0

0.7

12

4.56

1.19

Xu H [67]

2013

nr

nr

nr

nr

nr

nr

4

1.5

0.4

14

4.68

0.87

Xu LF et al

[68]

2015

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

Yang X [69]

2015

nr

nr

nr

nr

nr

nr

17

4.47

1.01

51

5.1

1.19

Yao TX et al [70]

2014

120

3.01

0.7

120

3.8

1.3

4

2.7

1.3

11

4.1

2.1

Yu M [71]

2014

nr

nr

nr

nr

nr

nr

12

3.4

1.3

23

7.0

1.8

Zhang DM et al [72]

2017

81

4.3

1.7

82

6.9

2.0

12

3.0

1.1

23

5.7

1.8

Zhang J et al [73]

2013

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

Zhang WX [74]

2013

nr

nr

nr

nr

nr

nr

8

1.2

1.1

29

3.5

2.8

Zhang Y et al [75]

2018

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

nr

Zhao G [76]

2013

nr

nr

nr

nr

nr

nr

6

2.8

1.1

17

4.3

0.9

Zhao GD [77]

2018

nr

nr

nr

nr

nr

nr

9

2.85

1.3

23

4.64

1.52

Zheng SQ [78]

2013

nr

nr

nr

nr

nr

nr

21

3.26

1.05

38

5.13

1.86

Zhu HY [79]

2017

50

2.11

0.6

50

3.94

1.02

4

2.13

0.47

13

5.2

1.32

Table 3: Efficacy of S. boulardii on prevention of secondary AAD: stool frequency and duration of AAD, and stool frequency/day, AAD, antibiotic-associated diarrhea; na, not applicable; nr, not reported; Ref, reference; Sb, Saccharomyces boulardii CNCM I-745, Std dev, standard deviation.

 

Figure 4: Forest plot of efficacy for stool frequency/day: S. boulardii compared to controls. SMD, standardized mean difference; IV, Inverse variance.

Clinical characteristics of AAD

Three measures were used to determine if S. boulardii influenced the severity of AAD (duration of AAD, frequency of severe AAD and effectiveness rating).  Data was collected from 232 cases of AAD in S. boulardii treated patients and 454 cases of AAD in the controls.

Clinical characterization of AAD: duration of AAD.The duration of AAD (Table 3) was reported in 36 (78.3%)  of the 46 prevention trials, while 10 (22%) of the RCTs did not report this outcome.  Mean duration of AAD ranged from 0.3 to 5 days in the S. boulardii subjects and ranged 4 to 9 days in the controls. The reduction of the days of diarrhea was significantly reduced for those given S. boulardii compared to the controls (SMD: -1.29 days, 95% C.I. -1.43, -1.15 P<0.001, I2=68.8%), as shown in (Figure 5).

 

Figure 5: Forest plot of efficacy for reduction of AAD duration in days: S. boulardii compared to controls. SMD, standardized mean difference; IV, inverse variance.

Clinical characterization of AAD: Incidence of severe AAD. Only 17 (37%) of the RCTs reported incidence of severe AAD (Table 4) [36,38, 40, 42, 45, 46, 48, 50, 59, 61, 63, 65, 69,71,76,77,79], while 29 (63%) did not assess the severity of AAD. Of the 17 RCTs that analyzed the severity of AAD, most (70%) reported a definition of ‘severe’ AAD (Supplementary Table S2). Treatment with S. boulardii did not significantly result in less severe cases of AAD, as shown in (Supplementary Figure S2) (RR=1.00, 95% C.I. 0.57, 1.76, P = 1.0).

Ref.

Year

Number with severe AAD n/N (%)

Rated effective for AAD n/N (%)

in S. boulardii

in controls

in S. boulardii

in controls

Bai SX et al [34]

2012

nr

nr

19/20 (95)

23/32 (71.9)

Cao TR [35]

2013

nr

nr

nr

nr

Chen JW et al [36]

2013

3/23 (13.0)

12/43 (27.9)

22/23 (95.6)

28/43 (65.1)

Chen JB [37]

2013

nr

nr

nr

nr

Chen Juan [38]

2013

1/6 (16.7)

3/18 (16.7)

nr

nr

Chen W et al [39]

2012

nr

nr

nr

nr

Chen YY [40]

2020

0/5 (0)

1/14 (7)

nr

nr

Dong G [41]

2015

nr

nr

nr

nr

Dong X et al [42]

2014

1/5 (20)

4/11 (36.4)

nr

nr

Feng X et al [43]

2015

nr

nr

nr

nr

Guo T et al [44]

2018

nr

nr

nr

nr

Huang MH [45]

2018

1/7 (14.3)

5/15 (33.3)

7/7 (100)

11/15 (73.3)

Huang S et al [46]

2017

0/3 (0)

2/9 (22.2)

3/3 (100)

8/9 (88.8)

Li B et al [47]

2011

nr

nr

nr

nr

Li JM et al [48]

2014

4/26 (15.4)

8/25 (32.0)

25/26 (96.1)

17/25 (68.0)

Li YY et al [49]

2021

nr

nr

nr

nr

Li YF [50]

2013

4/21 (19)

13/43 (30.2)

20/21 (95)

30/43 (69.8)

Lin X et al [51]

2018

nr

nr

nr

nr

Lou QY et al [52]

2020

nr

nr

nr

nr

Peng XH [53]

2018

nr

nr

nr

nr

Shan LS et al [54]

2013

nr

nr

nr

nr

Shan LS et al [55]

2014

nr

nr

nr

nr

Song YH et al [56]

2012

nr

nr

nr

nr

Tan G et al [57]

2022

nr

nr

nr

nr

Tang M [58]

2014

nr

nr

nr

nr

Tao Z et al [59]

2016

3/13 (23)

7/23 (30.4)

12/13 (92)

14/23 (60.9)

Wan CM et al [60]

2017

nr

nr

nr

nr

Wang LH [61]

2014

2/19 (10.5)

10/28 (35.7)

17/19 (89.5)

16/28 (57.1)

Wang XY [62]

2013

nr

nr

nr

nr

Wang ZZ [63]

2013

2/9 (22)

9/20 (45)

nr

nr

Wu XY et al [64]

2015

nr

nr

20/21 (95.2)

22/30 (73.3)

Wu YX [65]

2012

1/6 (16.7)

6/16 (37.5)

nr

nr

Xiao J [66]

2018

nr

nr

nr

nr

Xu H [67]

2013

nr

nr

nr

nr

Xu LF et al [68]

2015

nr

nr

nr

nr

Yang X [69]

2015

1/17 (5.9)

15/51 (29.4)

nr

nr

Yao TX et al [70]

2014

nr

nr

nr

nr

Yu M [71]

2014

1/12 (8.0)

2/23 (11.5)

nr

nr

Zhang DM et al [72]

2017

nr

nr

nr

nr

Zhang J et al [73]

2013

nr

nr

nr

nr

Zhang WX [74]

2013

nr

nr

nr

nr

Zhang Y et al [75]

2018

nr

nr

nr

nr

Zhao G [76]

2013

0/6 (0)

2/17 (11.7)

nr

nr

Zhao GD [77]

2018

0/9 (0)

0/23 (0)

8/9 (88.9)

15/23 (65)

Zheng SQ [78]

2013

nr

nr

nr

nr

Zhu HY [79]

2017

0/4 (0)

2/13 (15.4)

nr

nr

Table 4: Clinical characteristics of AAD in 232 cases of AAD in S. boulardii and 454 cases of AAD in controls; %, percentage;  AAD, antibiotic-associated diarrhea; n, number in numerator; N, total number in denominator, nr, not reported.

Clinical characterization of AAD: effectiveness rating. Only 10 (22%) of the RCTs used an effectiveness rating for AAD, while most (n=36, 78%) did not report this outcome (Table 4). The range of AAD rated “effectively prevented” was higher in S. boulardii (89%-100%) compared with the controls (57%-89%) but was not significantly different when analyzed (RR=1.00, 95% C.I. 0.93, 1.08, I2=82.9%, P=1.00, as shown in (Supplementary Figure S3).

Length of hospitalization

Four RCTs found shorter length-of-stays when S. boulardii was given compared to controls (SMD: -0.75 days, 95% C.I. -0.89, -0.61, P <0.0001) [42,49,52,53], as shown in (Supplementary Figure S4). 

Safety

Many (23/46, 50%) of the RCTs provided data on adverse reactions or tolerance (Table 2), while no data on safety was provided in 23 (50%) of the RCTs. Twenty-one RCTs only provided a statement that the “probiotic was well-tolerated” or “no adverse events were noted”, while two RCTs provided more detailed data on mild adverse reactions [44,58]. Guo et al reported abdominal pain (19% in S. boulardii group and 11% in controls, P = 0.22), abdominal distension (16% and 11%, respectively, P = 0.42) and nausea (13% and 8%, respectively, P = 0.40). Tang et al. noted only mild allergic reactions in two (3%) of those given S. boulardii and in 3 (4%) of controls (P>0.05) [58]. No serious adverse events were reported in any of the trials.

Sub-groups

Age

As the enrollment age ranges were not mutually exclusive, only three age sub-groups could be analyzed: neonates (under 14 days old), pediatric (1 month to 16 years old) and elderly (>65 years old). As shown in (Supplementary Figure S5), S. boulardii significantly prevented AAD in all three age groups, with no significant differences by age groups (X2=0.8, P = 0.67).

Daily dose

The effect of S. boulardii dose was explored, which reduced the degree of heterogeneity and three dose groups had significant efficacy as shown in (Supplementary Figure S6). AAD risk was significantly reduced for 250 mg/d dose group (RR=0.36, 95% C.I.

0.33, 0.45, P < 0.001, I2=0%) and in the 500 mg/d group (RR= 0.39, 95% C.I. 0.33, 0.47, P < 0.001, I2 =8.9%) and when doses were given according to age groups in children (RR= 0.46, 95% C.I. 0.41, 0.53, P < 0.001, I2 =20.9%). Two other doses could not be analyzed as there was only one RCT for each dose: 300 mg/d [46] and 1000 mg/d [72]. There was no significant difference by dose sub-group (X2=2.52, p=0.64).

Study Quality

All the RCTs were assessed for risk of bias (Supplementary Table S3). Of the 46 RCTs, two (4.3%) were considered an overall low risk of bias (Supplementary Figure S7), 84.8% had ‘some concerns’ about bias and five (10.9%) were considered high risk bias (when more than one bias domain was rated as high risk). The only individual domain in the 46 RCTs commonly rated high-risk was for the lack of double-blinded design (96%). Other individual domains were rated as low risk. When five high risk trials for prevention of AAD were excluded [42,43,49,52,57], the reduction of AAD risk was similar (RR=0.42, 95% CI 0.38, 0.46, P<0.001, I2 =0.5%), as shown in (Supplementary Figure S8).

Other sub-groups

Some sub-groups could not be analyzed as the groups had similar characteristics or were infrequently reported. For example, respiratory tract infections were the most common indication for antibiotics (94% trials), most trials were in children (96%), 96% had open controls and all patients were hospitalized. Some data was not reported in sufficient numbers of trials (for example, 33% of the trials did not report intervention initiation time and only four trials reported the length of hospitalization (Supplementary Table S4). None of the trials reported if the hospital patients were from urban or rural healthcare facilities and none reported cost or cost-saving data.

Discussion

Our meta-analysis found S. boulardii CNCM I-745 was significantly effective for the prevention of AAD and the prevention of CDI in trials done in China and was well-tolerated. S. boulardii CNCM I-745 effectively reduced AAD in hospitalized children and adults receiving the types of antibiotics typically prescribed for respiratory tract infections in China (cephalosporins, penicillins, macrolides, etc.), which are well known risk factors for AAD and CDI [80]. S. boulardii CNCM I-745 was shown to not only significantly reduce the incidence of AAD and CDI, but also reduce the duration of AAD and the daily stool frequency. S. boulardii was well tolerated in the included trials and safety has been thoroughly documented in patients receiving antibiotics in comprehensive meta-analyses and reviews [17,21,22]. Antibiotic resistance is another safety concern associated with antibiotic use and one study reported a reduction in the antibiotic resistance gene load when H. pylori eradication therapy was combined with S. boulardii CNCM I-745 [81]. Healthcare providers and policy makers should be aware of the high rate of AAD in their patients treated with antibiotics in China and strive to implement strategies to reduce AAD.

As the intestinal microbiome is influenced by ethnicity and geography, the question often arises if probiotic efficacy is also affected by these factors [2]. The effects of differences in diet, genetic make-up, nutritional status, healthcare access and living conditions are not commonly explored in probiotic reviews. One method of controlling for these confounding factors is to limit the meta-analysis to one country or to assess the efficacy using subgroups of different geographic regions [17,82].  We showed S. boulardii significantly reduced the risk of AAD (RR=0.43) in China, which was similar to studies done in other countries and in different populations. In a meta-analysis, Szajewska et al. [17] reported S. boulardii significantly reduced the risk of AAD regardless of where the trial was conducted: trials done in Europe (RR=0.46) or in USA (RR=0.46), as shown in (Supplementary Figure S9). Guo et al. [19] reviewed different types of probiotics for the prevention of AAD in children and pooled data from nine RCTs using S. boulardii. These nine trials were done in different countries (China, France, Turkey, India and Poland) but were not assessed by country sub-groups but nevertheless found a significant reduction of AAD (RR=0.36, 95% C.I. 0.24, 0.54, P<0.0001) for the nine trials. In a network meta-analysis with 10 different probiotic types, Cai et al. [18] reported a significant reduction in AAD from 11 RCTs using S. boulardii (RR=0.41, 95% C.I. 0.29, 0.57, P<0.05) and a significant reduction in CDI from three RCTs (RR= 0.35, 95% C.I. 0.15, 0.85, P<0.05), but did not report in which countries the trials were conducted.

Study Strengths

Use of Chinese databases (CNKI and CBM) for the literature search revealed 42 trials that would have been undetected if only the most common literature databases (PubMed, Google Scholar, etc.) had been used. We also followed recently published guidelines for reporting meta-analyses involving probiotics [28]. We used rigorous inclusion criteria to include trials that adequately described the probiotic intervention and outcome measures. Subgroup analysis was done to examine factors that might influence the estimates of efficacy. As geographic region factors might influence the efficacy of a probiotic intervention, one strength was that we limited trials done in one country (China), although we acknowledge vast differences in regions within China exist. Another strength was that we limited our meta-analysis to one specific type of probiotic (S. boulardii CNCM I-745), as the efficacy of probiotics has been shown to be both strain-specific and disease-specific [15].

Study limitations

Geographic factors in China that might affect probiotic efficacy were not documented in the included trials (such as differences in diet, health status, urban/rural, etc.). Future trials may benefit in exploring these factors. Significant heterogeneity was observed in some outcomes and sub-group analyses failed to explain the possible sources of the heterogeneity. Another limitation is that some trials failed to report specific confounder data, so that not all proposed sub-group analyses could be performed. Several meta-analyses found the efficacy of a probiotic is better when the probiotic is started within 48 hours of the antibiotic [83,84], and while many RCTs in China reported the intervention was started at the same time as the antibiotics, the specific times were rarely stated. We excluded 17 trials with a primary outcome of H. pylori eradication, but S. boulardii has been shown to significantly reduce AAD in patients treated with antibiotics for H. pylori infections [21]. In a meta-analysis by McFarland et al. [85], S. boulardii CNCM I-745 significantly reduced the risk of AAD associated with H. pylori eradication therapies in eight RCTs (RR=047, 95% C.I 0.37, 0.60). Even though the 46 trials in this meta-analysis were limited to those done in China, the robustness of the efficacy estimates from trials done in other countries may show the efficacy of S. boulardii CNCM I-745 may be generalized to other populations.

Future studies

To improve the quality of future RCTs, it is recommended to provide a complete description of the method of randomization, consider using a placebo control and to follow subjects postantibiotics to detect late-onset cases of AAD. In addition, RCTs need to report safety data as this is an important clinical consideration when using probiotics. Most studies did not compare their study results with other trials using S. boulardii CNCM I-745 and this should be included in the discussion of future papers. This study shows the value of utilizing resource rich literature databases, especially in non-English language studies.

Conclusion

In summary, the results of this meta-analysis of 46 randomized, controlled trials in China demonstrated that S. boulardii CNCM I-745 was effective in both the prevention of AAD and CDI and was well tolerated. The use of Chinese databases for literature searches revealed an untapped resource to find important clinical trials and offers clinicians and policy-makers guidance for an effective strategy to prevent complications of antibiotic use.

Disclosures

Author contributions

Conceptualization: Lynne McFarland and Tong Li; Methodology: Lynne McFarland and Tong Li; Software: Lynne McFarland; Validation: Lynne McFarland and Tong Li; Formal analysis: Lynne McFarland; Resources: Lynne McFarland and Tong Li; Data curation: Lynne McFarland; Writing-original draft preparation: Lynne McFarland; Writing-review and editing: Lynne McFarland and Tong Li; Supervision: Lynne McFarland.

Funding

Funding for this study was provided by Biocodex, France.

Data Availability statement

Data is available in the Supplementary Materials.

Conflicts of Interest

Lynne McFarland is a member of the Biocodex Microbiota Foundation’s advisory board) and a paid consultant for Biocodex. Tong Li declares no conflicts of interest.

References

  1. McFarland LV, Ozen M, Dinleyici EC, Goh S (2016) Comparison of pediatric and adult antibiotic-associated diarrhea and Clostridium difficile infections. World J Gastroenterol 22: 3078-3104.
  2. Gaulke CA, Sharpton TJ (2018) The influence of ethnicity and geography on human gut microbiome composition. Nat Med 24: 14951496.
  3. World Health Organization (2023) WHO diarrhoeal disease fact sheet, 2017.
  4. Wu Y, Wang YY, Bai LL, Zhang WZ, Li GW (2022) A narrative review of Clostridioides difficile infection in China. Anaerobe 74: 102540.
  5. Wang J, Wang P, Wang X, Zheng Y, Xiao Y (2014) Use and prescription of antibiotics in primary health care settings in China. JAMA Intern Med 174: 1914-1920.
  6. Zhang J, Cameron D, Quak SH, Kadim M, Mohan N, et al. (2020) Rates and determinants of antibiotics and probiotics prescription to children in Asia-Pacific countries. Benef Microbes 11: 329-338.
  7. Liu W, Hassan Gillani A, Xu S, Chen C, Chang J, et al. (2021) Antibiotics (Macrolides and Lincosamides) consumption trends and patterns in China’s healthcare institutes. Based on a 3 Year Procurement Records, 2015–2017. Int J Environ Res Pub Healt 18:113.
  8. Chen Y, Xiang Q, Liu L (2021) Comparison of antibiotic-associated diarrhea caused by cefoperazone/sulbactam or piperacillin/tazobactam in neurosurgery patients. J Int Med Res 49: 03000605211019661.
  9. Tang Y, Liu C, Zhang Z, Zhang X (2018) Effects of prescription restrictive interventions o3n antibiotic procurement in primary care settings: a controlled interrupted time series study in China. Cost Eff Resour Alloc 16:1-11.
  10. Agamennone V, Krul CAM, Rijkers G, Kort R (2018) A practical guide for probiotics applied to the case of antibiotic-associated diarrhea in The Netherlands. BMC Gastroenterol 18: 103.
  11. Preidis GA, Weizman AV, Kashyap PC, Morgan RL (2020) AGA Technical review on the role of probiotics in the management of gastrointestinal disorders. Gastroenterol 159: 708-738.
  12. Szajewska H, Canani RB, Domellöf M, Guarino A, Hojsak I, et al. (2023) Probiotics for the management of pediatric gastrointestinal disorders: position paper of the ESPGHAN Special Interest Group on Gut Microbiota and Modifications. J Ped Gastroenterol Nut 76: 232247.
  13. Cameron D, Hock QS, Kadim M, Mohan N, Ryoo E, et al. (2017) Probiotics for gastrointestinal disorders: proposed recommendations for children of the Asia-Pacific region. World J Gastroenterol 23: 7952.
  14. Ghoshal UC, Gwee KA, Holtmann G, Li Y, Park SJ, et al. (2018) The role of the microbiome and the use of probiotics in gastrointestinal disorders in adults in the Asia‐Pacific region‐background and recommendations of a regional consensus meeting. J Gastroenterol Hepatol 33: 57-69.
  15. McFarland LV, Evans CT, Goldstein EJ (2018) Strain-specificity and disease-specificity of probiotic efficacy: a systematic review and metaanalysis. Front Med 5: 124.
  16. McFarland LV, Goh S (2013) Preventing pediatric antibiotic-associated diarrhea and Clostridium difficile infections with probiotics: a metaanalysis. World J Meta-Analysis 1: 102-120.
  17. Szajewska H, Kołodziej M (2015) Systematic review with metaanalysis: Saccharomyces boulardii in the prevention of antibioticassociated diarrhoea. Ali Pharmacol Ther 42: 793-801.
  18. Cai J, Zhao C, Du Y, Zhang Y, Zhao M, et al. (2018) Comparative efficacy and tolerability of probiotics for antibiotic‐associated diarrhea: systematic review with network meta‐analysis. UEG Journal 6: 169180.
  19. Guo Q, Goldenberg JZ, Humphrey C, El Dib R, Johnston BC (2019) Probiotics for the prevention of pediatric antibiotic‐associated diarrhea. Cochrane Database Syst Rev 4: CD004827.
  20. Szajewska H, Kołodziej M, Zalewski BM (2020) Systematic review with meta-analysis: Saccharomyces boulardii for treating acute gastroenteritis in children-a 2020 update.  Aliment Pharmacol Ther 51: 678-688.
  21. Cárdenas PA, Garcés D, Prado-Vivar B, Flores N, Fornasini M, et al. (2020) Effect of Saccharomyces boulardii CNCM I-745 as complementary treatment of Helicobacter pylori infection on gut microbiome. Euro J Clin Microbiol Infect Dis 39: 1365-1372.
  22. McFarland LV (2010) Systematic review and meta-analysis of Saccharomyces boulardii in adult patients. World J Gastroenterol 16: 2202-2222.
  23. Stier H, Bischoff SC (2016) Influence of Saccharomyces boulardii CNCM I-745 on the gut-associated immune system. Clinical Experi Gastroenterol 9: 269-279.
  24. Neut C, Mahieux S, Dubreuil LJ (2017) Antibiotic susceptibility of probiotic strains: Is it reasonable to combine probiotics with antibiotics? Medecine Maladies Infect47: 477-483.
  25. Terciolo C, Dapoigny M, Andre F (2019) Beneficial effects of Saccharomyces boulardii CNCM I-745 on clinical disorders associated with intestinal barrier disruption. Clin Experiment Gastroenterol 12: 6782.
  26. Kothari D, Patel S, Kim SK (2019) Probiotic supplements might not be universally-effective and safe: A review. Biomed Pharmacother 111: 537-547.
  27. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, et al. (2021) The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Br Med J 372: n71.
  28. McFarland LV, Hecht G, Sanders ME, Goff DA, Goldstein EJC, et al. (2023) Recommendations to improve quality of probiotic systematic reviews with meta-analyses. JAMA Network Open 6: e2346872.
  29. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, et al. (2014) Expert consensus document: The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol 11: 506-514.
  30. Moher D, Shamseer L, Clarke M, Ghersi D, Liberati A, et al. (2015) Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Rev 4: 1-9.
  31. Higgins JP, Altman DG, Gøtzsche PC, Jüni P, Moher D, et al. (2011) The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. Br Med J 343: d5928.
  32. McGuinness LA (2020) Robvis: An R package and web application for visualising risk-of-bias assessments.
  33. Palmer TM, Sterne JAC (2016) Meta-analysis in Stata: an updated collection from the Stata Journal. Second Edition. 2016. Published by Stata Press, College Station, Texas.
  34. Bai SX, Fu Y, Ding X, Zhang M, Li Z, et al. (2012) Saccharomyces boulardii for the prevention and treatment of antibiotic-associated diarrhea in infants and young children. Herald of Med 31: 605-607.
  35. Cao TR (2013) Clinical observation of Yihuo for the prevention of antibiotic-associated diarrhea in children. J Mudanjiang Med Univ 34: 57-59.
  36. Chen JW, Li S, Cai Q (2013) Observation on the efficacy of Saccharomyces boulardii in preventing antibiotic-associated diarrhea in children. Strait Pharmaceut J25: 132-133.
  37. Chen JB (2013) Clinical observation of Saccharomyces boulardii powder in the prevention of antibiotic associated diarrhea in children. Chin Foreign Med Res11:15-16.
  38. Chen J (2013) Clinical observation of Saccharomyces boulardii in prevention and treatment of 64 cases of antibiotic associated diarrhea caused by pneumonia in infants and young children. Shanxi Med J 42: 1156-1157.
  39. Chen W, Tang J (2012) Efficacy of Saccharomyces boulardiiin the prevention of antibiotic-associated diarrhea. Chin Med Innovation 9: 27.
  40. Chen YY (2020) Study on the preventive effect of Saccharomyces boulardii on antibiotic-related diarrhea in hospitalized children with pneumonia. J North Pharm 17: 83-4.
  41. Dong G (2015) Prevention of antibiotic-associated diarrhea by Saccharomyces boulardii. World Latest Med Inform 15:103-104.
  42. Dong X, Zhang J, Qi GL (2014) Observation of efficacy of a Saccharomyces boulardii preparation in prevention of antibioticassociated diarrhea in infants and young children. Chin J Gen Pract 12: 1329-1332.
  43. Feng X, Xue J, Xie K, Feng X (2015) Saccharomyces boulardii for prevention and treatment of antibiotic-associated diarrhea in infants]. Zhongguo Weishengtaxixue Zazhi [Chin J Microecol] 27: 1436-1438.
  44. Guo T, Ning X, Ni N, Bina MY, Meng SC, et al. (2018) A study of a Saccharomyces boulardii preparation for prevention of antibioticassociated diarrhea in elderly patients with lower respiratory infection. Chin J Mult Organ Dis Elderly 17: 367-371.
  45. Huang MH (2018) Prevention and treatment effect of Saccharomyces boulardii on antibiotic-associated diarrhea in infants with pneumonia. Chin J Clin Rational Drug Use 11: 71-72.
  46. Huang S, Xiang RJ, Zou, Fang ZY, Yuan ZYN, et al. (2017) Clinical analysis of Saccharomyces boulardii on prevention of antibioticassociated diarrhea in children.Contemp Med Symposium 15: 126128.
  47. Li B (2011) A clinical study of Saccharomyces boulardii and antibioticassociated diarrhea in children. Chin Gen Pract 14: 1255-1256.
  48. Li JM, Zhang HF, Xie H, Shen T (2014) A clinical study of a Saccharomyces boulardii preparation for prevention of antibioticassociated diarrhea secondary to pneumonia in infants and young children. J Pharmaceut Pract 32: 222-226. 
  49. Li YY, Feng Z, Hong X, Jing H, Jingqiu Z, et al. (2021) Saccharomyces boulardii in the prevention of antibiotics-associated diarrhea in neonates. J Ped Pharm 27: 22-25.
  50. Li YF, Huang K (2013) Clinical analysis of Saccharomyces boulardii for the prevention of antibiotic-associated diarrhea in infants. J Wenzhou Med College43: 254-256.
  51. Lin X, Cai Z, Lin GY (2018) Observation on the clinical effects of Saccharomyces boulardii sachets in preventing antibiotic-associated diarrhea in children. Chin J Microecol 30: 327-331.
  52. Lou QY, Jin L, Tang WH, Yu YP, Yuan XH, et al. (2020) A randomized controlled multicenter clinical study of a Saccharomyces boulardii preparation for prevention of diarrhea secondary to pediatric pneumonia. Chin J Pract Pediat 35: 866-874.
  53. Peng XH  (2018) Clinical observation of Saccharomyces boulardii in the prevention and treatment of antibiotic associated diarrhea in children. Tianjin Pharm 30: 28-29.
  54. Shan LS, Hou P, Wang ZJ, Liu FR, Chen N, et al. (2013) Prevention and treatment of diarrhoea with Saccharomyces boulardii in children with acute lower respiratory tract infections. Bene Microb 4: 329-334.
  55. Shan LS, Hou P, Wang Z, Chen N, Cai X, et al. (2014) Efficacy of Saccharomyces boulardii powder in the prevention and treatment of antibiotic-associated diarrhea in children with acute bronchopneumonia. J China Med Univer 3: 55-58.
  56. Song YH, Peng J, Zhang J, Yang S, Wang S (2012) A clinical study of Saccharomyces bouardii for the prevention of antibiotic-associated diarrhea in children. J Pract Med 28: 2785-2787.
  57. Tan G, Liu L, Li W (2022) Analysis on the effect of Saccharomyces boulardii in prevention and treatment of antibiotic-associated diarrhea in children. Clin Med Engineer 6: 789-780.
  58. Tang M (2014) Observation on the preventive effect of Saccharomyces boulardii on diarrhea secondary to neonatal infectious pneumonia. J Ped Pharm20: 15-17. 
  59. Tao Z, Wang Y, Zhang L, Chen N (2016) A clinical study of a Saccharomyces boulardii preparation for prevention of pediatric antibiotic-associated diarrhea. Med J West China 28: 652-655.
  60. Wan CM, Yu H, Liu G, Xu HM, Mao ZQ, et al. (2017) A multicenter randomized controlled study of Saccharomyces boulardii in the prevention of antibiotic-associated diarrhea in infants and young children. Zhonghua er ke za zhi 55: 349-354.
  61. Wang LH (2014) Clinical study of Saccharomyces boulardii in preventing antibiotic-associated diarrhea in infants. Chin J Clin Rational Drug Use 7: 73-74.
  62. Wang XY (2013) Preventive effect of Saccharomyces boulardii on antibiotic-associated diarrhea in children. Med J West China 25: 18451846.
  63. Wang ZZ (2013) Prevention of antibiotic-associated diarrhea in children with bronchial pneumonia. Chin Commun Doctors 15: 139140.
  64. Wu XY, Lu HX, Yang HX, Wu H (2015) A clinical study of Saccharomcyes boulardii powder for prevention of antibiotic-associated diarrhea. Chin J Woman Child Health Res 26: 3-9.
  65. Wu YX (2012) Saccharomyces boulardii powder for the prevention of antibiotic association observation of the clinical efficacy of diarrhea. Clin Pract 26: 46-48.
  66. Xiao J (2018) Observation on the efficacy of Saccharomyces boulardii in preventing antibiotic-associated diarrhea. Science Technolo Vision 8: 192-193.
  67. Xu H (2013) A clinical study of Saccharomyces boulardii for the prevention of antibiotic-associated diarrhea in infants and young children. Nurs Pract Res 10: 12-13.
  68. Xu LF, Guan Z, Wang Y, Wang L, Guo X, et al (2015) Randomised controlled clinical trial of probiotics for the prevention of antibioticassociated diarrhea in children: Comparison between combined Clostridium butyricum and Bifidobacterium living powders and Saccharomyces boulardii. China Ped Emer Med 22: 257-261.
  69. Yang X (2015) Clinical study of Saccharomyces boulardii in preventing antibiotic-associated diarrhea in children. Med Inform 28: 254-255.
  70. Yao TX, Luo R, Gu F, Yu S (2014) Clinical study of Saccharomyces boulardii in preventing antibiotic-associated diarrhea in children. Chin J Pub Heal Engineer13: 518-519.
  71. Yu M, Yuan X, Yang D. (2014) Efficacy of Saccharomyces boulardii in the prevention and treatment of antibiotic-associated diarrhea in infants and young children. Med Inform 27: 136.
  72. Zhang DM, Xu BB, Yu L, Zheng LF, Chen LP, et al. (2017) A prospective control study of Saccharomyces boulardii in prevention of antibioticassociated diarrhea in elderly inpatients. Zhonghua nei ke za zhi 56: 398-401.
  73. Zhang J, Niu Y, Wang Y, Li X, Yu J (2013) Observations of Saccharomyces boulardii prophylaxis for antibiotic associated diarrhea in 150 children. J Henat n Med Colleg 25:171-172.
  74. Zhang WX (2013) Saccharomyces boulardii for the prevention of antibiotic-associated diarrhea in children: 110 cases. China Pharma 22: 66-67.
  75. Zhang Y, Chen SY (2018) Preventive effect of Saccharomyces boulardii on antibiotic-associated diarrhea in hospitalized children with acute bronchial pneumonia at various age-stages. Drug Evalua Res 4: 639-644.
  76. Zhao G (2013) Clinical application of Saccharomyces boulardii in antibiotic-associated diarrhea in children. Mod Diag Treat 5: 967-969.
  77. Zhao GD (2018) Clinical observation of Saccharomyces boulardii sachets in the prevention and treatment of antibiotic associated diarrhea in children with bronchial pneumonia. J Medl Theory Pract 31:1662-1663.
  78. Zheng SQ (2013) Application of Saccharomyces boulardii in antibioticassociated diarrhea in children. Shanghai J Prevent Med25: 486-487.
  79. Zhu HY (2017) Analysis of the effect of Saccharomyces boulardii on the prevention of antibiotic associated diarrhea in infants and young children. World Latest Med Inform 17: 59.
  80. Zhou H, Xu Q, Liu Y, Guo LT (2020) Risk factors, incidence, and morbidity associated with antibiotic-associated diarrhea in intensive care unit patients receiving antibiotic monotherapy. World J Clin Cases 8: 1908-1915.
  81. Cifuentes SG, Prado MB, Fornasini M, Cohen H, Baldeón ME, et al. (2022) Saccharomyces boulardii CNCM I‐745 supplementation modifies the fecal resistome during Helicobacter pylori eradication therapy. Helicobacter 27: e12870.
  82. McFarland LV, Srinivasan R, Setty RP, Ganapathy S, Bavdekar A, et al. (2021) Specific probiotics for the treatment of pediatric acute gastroenteritis in India: a systematic review and meta-analysis. JPGN Rep 2: e079.
  83. Zhang L, Zeng X, Guo D, Zou Y, Gan H, et al. (2022) Early use of probiotics might prevent antibiotic-associated diarrhea in elderly (>65 years): a systematic review and meta-analysis. BMC Geriatr 22: 562.
  84. Liao W, Chen C, Wen T, Zhao Q. (2021) Probiotics for the prevention of antibiotic-associated diarrhea in adults: A meta-analysis of randomized placebo-controlled trials. J Clin Gastroenterol 55: 469-480.
  85. McFarland LV, Malfertheiner P, Huang Y, Wang L (2015) Meta-analysis of single strain probiotics for the eradication of Helicobacter pylori and prevention of adverse events. World J Meta-analysis 3: 97-117.

Supplementary File

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Journal of Digestive Diseases and Hepatology

rumus slot mahjongrtp slot gacorfitur slot mahjong winsrekomendasi slot pragmartp live slotpola gates of gatotkacaapk cheat slotzeus godwrath maxwinmitra slot dana resmihabanero anti gagalserver kamboja gacordaftar link togelslot pg mahjongtrik pola zeus x500slot gacor mudah menangslot mahjong pragmaticpola trik slot mahjongrtp fortune dragonrtp slot speed winnerslot kamboja mahjong waystrik mantap slot olympusnaga hitam mahjongslot tergacor mahjongtrik jitu cuan mahjongpola slot mahjong winsrtp tinggi pragmaticslot mahjong onlineslot gacor hari inislot bonanza gacorfreebet mahjong winsserver jp rtp tinggigame resmi pragmatic terbaiktaktik efektif mahjongpola mahjong rekomendasi googleaztec gems boskututorial mahjong ways2starlight princess hari inipola starlight princessrtp fortune tigerrtp pg softrtp starlight princesstrik mahjong waysperkalian x5000 banditoslot mahjong waysslot terbaik olympusslot gates of olympusdaftar slot dana maxwinbocoran pola olympusmaxwin slot bonanzabocoran rtp tinggislot samurai codemetode slot starlightslot zeusrtp slot gacor pragmaticrtp slot pg softcara menang slot onlinescatter slot mahjongslot gacor server luar rekomendasi link olympusgerbang gatot kacartp kakek olympusslot gacor andalanrtp slot pgslot mahjongslot mahjong server jepangperkalian besar starlightrtp ways of qilinslot terbaik mahjongmahjong bulan mudastrategi permainan pragmaticcheat engine gacorjackpot auto cuanmahjong mekanik tinggitrik slot mahjongtips main slotslot server thailandpola mahjong unguslot gacor menangpg soft scatterslot olympusbocoran togel terpercayaamantotorm1131aman toto