Shan 2013

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Wageningen Academic

Beneficial Microbes, December 2013; 4(4): 329-334 P u b l i s h e r s

Prevention and treatment of diarrhoea with Saccharomyces boulardii in children with


acute lower respiratory tract infections

L.-S. Shan1, P. Hou1, Z.-J. Wang1, F.-R. Liu1, N. Chen1, L.-H. Shu1, H. Zhang1, X.-H. Han1, X.-X. Cai1, Y.-X. Shang1
and Y. Vandenplas2

1Department of Pediatric Pneumology, Shengjing Hospital of China Medical University, Shenyang 110004, China; 2UZ
Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; shangyx@sj-hospital.org

Received: 17 January 2013 / Accepted: 23 July 2013


© 2013 Wageningen Academic Publishers

RESEARCH PAPER
Abstract

The aim of this study was to determine whether Saccharomyces boulardii prevents and treats diarrhoea and antibiotic-
associated diarrhoea (AAD) in children. A total of 333 hospitalised children with acute lower respiratory tract
infection were enrolled in a 2-phase open randomised controlled trial. During the 1st phase, all children received
intravenous antibiotics (AB). They were randomly allocated to group A (S. boulardii 500 mg/day + AB, n=167) or
group B (AB alone, n=166) and followed for 2 weeks. Diarrhoea was defined as ≥3 loose/watery stools/day during at
least 2 days, occurring during treatment and/or up to 2 weeks after AB therapy had stopped. AAD was considered
when diarrhoea was caused by Clostridium difficile or when stool cultures remained negative. In the 2nd phase of the
study, group B patients who developed diarrhoea were randomly allocated to two sub-groups: group B1 (S. boulardii
+ oral rehydration solution (ORS)) and group B2 (ORS alone). Data from 283 patients were available for analysis.
Diarrhoea prevalence was lower in group A than in group B (11/139 (7.9%) vs. 42/144 (29.2%); relative risk (RR):
0.27, 95% confidence interval (CI): 0.1-0.5). S. boulardii reduced the risk of AAD (6/139 (4.3%) vs. 28/144 (19.4%);
RR: 0.22; 95% CI: 0.1-0.5). When group B patients developed diarrhoea (n=42), S. boulardii treatment during 5
days (group B1) resulted in lower stool frequency (P<0.05) and higher recovery rate (91.3% in group B1 vs. 21.1%
in B2; P<0.001). The mean duration of diarrhoea in group B1 was shorter (2.31±0.95 vs. 8.97±1.07 days; P<0.001).
No adverse effects related to S. boulardii were observed. S. boulardii appeared to be effective in the prevention and
treatment of diarrhoea and AAD in children treated with intravenous antibiotics.

Keywords: antibiotic-associated diarrhoea, Clostridium difficile, Saccharomyces boulardii, acute lower respiratory tract
infection, hospitalised children

1. Introduction (Bartlett et al., 1978; McFarland, 1998; Ruszczyński et al.,


2008; Turck et al., 2003; Wistrom et al., 2001).
Antibiotic-associated diarrhoea (AAD) is an acute
inflammation of the intestinal mucosa caused by the Probiotics are defined as microorganisms that, when
administration of antibiotics (Bartlett et al., 1978). administered in adequate amounts, confer a health benefit
Clostridium difficile is the most commonly bacterial agent to the host (Vandenplas et al., 2011). Saccharomyces
associated with AAD. Almost all antibiotics can cause boulardii is a probiotic yeast isolated from the peel of
AAD, especially broad-spectrum antibiotics (Elstner et fruits growing in Indochina, such as lychees, and belonging
al., 1983; Turck et al., 2003), such as aminopenicillins, to a species closely related to Saccharomyces cerevisiae
clavulanate, cephalosporins and clindamycin (Barbut et al., (Hennequin et al., 2001). S. boulardii has shown to be
1997; McFarland et al., 1990). The prevalence of AAD is effective in the treatment of acute infectious diarrhoea,
approximately 5-35%, depending on patients and antibiotics the prevention of AAD and as an adjunctive eradication
therapy of Helicobacter pylori (Buts, 2009; Erdeve et al.,

ISSN 1876-2833 print, ISSN 1876-2891 online, DOI 10.3920/BM2013.0008329


L.-S. Shan et al.

2004; Guandalini, 2011; Kotowska et al., 2005; Szajewska neutral envelope was opened each time the next patient
and Mrukowicz, 2005; Vandenplas et al., 2007, 2009, 2011). was included in the study. To evaluate the role of S. boulardii
in prevention, the incidence of diarrhoea and AAD was
Our study aimed to evaluate the efficacy of S. boulardii evaluated every day up to 2 weeks after the antibiotic
in the prevention and treatment of diarrhoea and AAD in therapy had stopped.
hospitalised children with an acute lower respiratory tract
infection. This is the first study evaluating the efficacy of The Bristol stool scale (BSS) classifying stool consistency
S. boulardii in hospitalised children developing diarrhoea in seven categories was used to evaluate stool consistency
during antibiotic treatment. (Lewis and Heaton, 1997). The definition of diarrhoea was
≥3 loose or watery stools (BSS type 5, 6 and 7) per day
2. Materials and methods during at least 2 days, occurring during treatment and/
or up to 2 weeks after the antibiotic therapy had stopped.
Subjects AAD was defined as diarrhoea caused by C. difficile or
diarrhoea with negative stool cultures (Bartlett et al., 1978).
The study was performed between January and June 2012 in Recovery was defined as a decrease in stool frequency and
children (age range from 6 months to 14 years) hospitalised BSS type ≤4. The number of stools, stool consistency, and
in the Department of Pediatric Pulmonology, Shengjing any other symptom were recorded daily in a diary during
Hospital of China Medical University, Shenyang, China hospitalisation and during a two week follow-up period.
because of an acute lower respiratory infection and the When diarrhoea occurred, stool samples were analysed
need of administration of intravenous antibiotics. Acute for rotavirus antigen using a colloidal gold test device
lower respiratory infections included bronchitis, bronchial (Rotavirus Group A diagnostic kit; Beijing Wantai Biological
pneumonia, extended lobar pneumonia, and asthma attacks Pharmacy Enterprise Co., Beijing, China); standard stool
together with lower respiratory tract infections. Exclusion cultures (Salmonella, Shigella, Yersinia, Campylobacter,
criteria were obvious malnutrition, severe dehydration, Escherichia coli) were performed and C. difficile toxins A
unconsciousness, gastrointestinal disease, a central line, and B were looked for using an enzyme immunoassay (C.
and use of antifungal drugs or probiotics. Participation in difficile TOX A/B Test; Tech Lab Inc., Blacksburg, VA,
this clinical study was voluntary. Informed consent was USA). Treatment compliance was evaluated by returning
obtained from the children’s guardian. The study protocol medication after the intervention period.
was approved by the University Hospital Ethical Committee.
Children who developed diarrhoea were rehydrated with
Sample size oral or intravenous fluids, but antibiotic treatment was
continued. Children from group B who developed diarrhoea
It was assumed that the incidence rate of diarrhoea were divided in two sub-groups: group B1, S. boulardii
development in the control groups would be equal to 25%. (daily 2×250 mg for 5 days) + oral rehydration solution
Based on this, we estimated that a minimum sample size (ORS), and group B2, ORS only. The enrolment method
of 129 children in each treatment group would show 15% for the second part of the study was identical to that of
reduction in the proportion of children with AAD in the the first part.
intervention groups with a significance level of 5% and
power of 90%. It was assumed that the study withdrawal Outcome measures
rate would be at least 20-25% and, therefore, the sample
size was increased to compensate for the potential influence The outcome measures were twofold. For the first part
of loss to follow-up. of the study, the development of diarrhoea between the
start of antibiotic treatment up to two weeks later was
Study design the primary outcome. The secondary outcome was AAD,
when all cultures were negative or when diarrhoea was
This study was an open, randomised, controlled clinical caused by C. difficile. For the second part of the study,
trial. All patients included were given the standard only groups B1 and B2 were considered. In those patients,
intravenous antibiotic treatment according to the hospital duration of diarrhoea was the primary endpoint. Duration
recommendations and were randomly allocated to two of diarrhoea was determined by the number of days of
groups. Group A received S. boulardii daily 2×250 mg continuous diarrhoea, from the first diarrheic stool until
(Bioflor; CMS·Shenzhen Kangzhe Pharmaceutical Co. the first normal stool.
Ltd., Shenzhen, China) for the duration of the antibiotic
treatment (n=167) and group B received no additional
therapy (n=166). Randomisation was done according to
a computer-determined allocation to group A or B. The
sequence was concealed in an envelope, and the next

330 Beneficial Microbes 4(4)


 S. boulardii in diarrhoea and antibiotic-associated diarrhoea

Statistical analysis Table 1. Baseline demographics of patients receiving


Saccharomyces boulardii during antibiotic treatment or
Continuous variables were presented as mean ± standard antibiotics alone.
deviation and analysed using the t-test, approximating a
normal distribution. Statistical analysis was performed Group A: Group B:
using ‘available case analysis’, i.e. analysis in which data S. boulardii iv AB1
are analysed for every participant for whom an outcome + iv AB1
was obtained. Categorical variables were expressed in
percentage and compared using a chi-square test. The Number of patients 167 166
relative risk (RR) and 95% confidence interval (CI) were Sex (male/female) 82/85 84/82
calculated. The difference was considered significant when Age (months, range) 49.8±36 48.7±34
the P-value was <0.05. Statistical analyses were performed (6.2-158) (6.1-161)
by SPSS version 13.0 (IBM, Armonk, NY, USA). Diagnosis
Bronchitis 36 (53.7%) 31 (46.3%)
3. Results Pneumonia 70 (48.6%) 74 (51.4%)
Asthma + LRT infection1 39 (48.1%) 42 (51.9%)
Clinical characteristics Others 22 (53.6%) 19 (46.3%)
Antibiotic
333 patients were included in the trial. The patients’ Cefepime 26 24
stool frequency and characteristics were normal prior to Cefoperazone + sulbactam 24 28
admission. 167 patients received S. boulardii (group A) Cefuroxime 37 34
shortly after the first dose of antibiotics; the other 166 Amoxicillin+ clavulanic acid 43 45
patients (group B) were not given S. boulardii (Figure 1). Erythromycin 22 24
Baseline demographics, clinical characteristics and Others 12 11
antibiotics did not differ significantly between both groups
(Table 1). In total, 50 (15%) patients were considered drop- 1 iv AB = intravenous antibiotics, LRT = lower respiratory tract.
outs (28 (16.7%) in group A and 22 (13.2%) in group B)
because of withdrawal of consent (n=12), antibiotic stop
within 48 h (n=8), non-compliance (n=10), and lost to Diarrhoea was diagnosed in 53/283 patients (18.7%)
follow-up (n=20). Data of 283/333 children were available: (Table 2): 11/139 (7.9%) in group A compared to 42/144
139/283 in group A and 144/283 in group B. (29%) in group B (RR=0.27; 95% CI = 0.13-0.55; P<0.01).

Acute lower respiratory


tract infections
(n=333)

Group A: SB + iv AB Group B: iv AB
(n=167) (n=166)

drop out drop out


n=28 (17 %) n=22 (13 %)

n=139 n=144

No diarrhoea Diarrhoea No diarrhoea Diarrhoea


n=128 n=11 n=102 n=42
(92%) (8%) (71%) (29%)

Group B1: Group B2:


SB + ORS ORS
n=23 (56%) n=19 (45%)

Figure 1. Study flow chart. Abbreviations used: iv AB = intravenous antibiotics, n = number of patients, ORS = oral rehydration
solution.

Beneficial Microbes 4(4) 331


L.-S. Shan et al.

Table 2. Primary and secondary outcomes in patients receiving Saccharomyces boulardii during antibiotic treatment or antibiotics
alone.

Group A: Group B: RR (95% CI)2


S. boulardii + iv AB1 (n=139) iv AB1 (n=144)

Diarrhoea 11/139 (7.9%) 42/144 (29.2%) 0.27 (0.13-0.55)


Cause of diarrhoea
Rotavirus 5 (3.6%) 14 (9.7%) 0.37 (0.13-1.06)
Clostridium difficile 1 (0.7%) 8 (5.6%) 0.13 (0.02-1.05)
Unexplained diarrhoea 5 (3.6%) 20 (13.9%) 0.26 (0.09-0.71)
AAD3 (C. difficile, unexplained diarrhoea) 6 (4.3%) 28 (19.4%) 0.22 (0.09-0.55)

1 iv AB = intravenous antibiotics.
2 RR = relative risk, CI = confidence interval.
3 AAD = antibiotic-associated diarrhoea.

AAD was diagnosed in 34/53 (64%) patients. The AAD recovery rate was significantly higher than in group B2 (83
risk was lower in group A (RR=0.22; 95% CI = 0.09-0.56), vs. 16%; χ2=18.62, P<0.001) (Table 4). The mean duration
especially in patients treated with cefuroxime (1/30 of diarrhoea in group B1 was significantly shorter than in
(3.3%) vs. 8/31 (25.8%); RR=0.13; 95% CI = 0.02-0.97), or group B2 (2.31±0.95 vs. 8.97±1.07 days, t=21.36, P<0.001).
amoxicillin/clavulanic acid (2/36 (5.6%) vs. 9/37 (24.3%);
RR=0.23; 95% CI = 0.05-0.99) (Table 3). The incidence Adverse reactions
of rotavirus gastroenteritis was similar in both groups
(Table 2). No adverse reaction to S. boulardii and no case of drug
discontinuation (neither antibiotics nor S. boulardii) have
Therapeutic effect of Saccharomyces boulardii on been reported.
diarrhoea
4. Discussion
For the second part of the study, the sample size calculation
was not performed. The 42 patients with diarrhoea in group This study confirms the efficacy of S. boulardii in the
B were further randomly allocated: 23 to group B1 (ORS prevention of diarrhoea and AAD developing during or
+ S. boulardii) and 19 to group B2 (ORS). After 3 days of short after antibiotic treatment. The administration of
administration of S. boulardii to patients in group B1, the S. boulardii during 5 days in the group of children that

Table 3. Features of patients with antibiotic-associated diarrhoea.

Group A: Group B: P-value RR (95% CI)2


S. boulardii + iv AB1 (n=6) iv AB1 (n=28)

Age (months±SD)3 (range) 16.5±14 (3.3-33.5) 18.8±19 (3.7-30.6) 0.69


Onset diarrhoea (days±SD) (range) 4.8±1.7 (3-8) 4.9±2.5 (2-10) 0.89 -
Antibiotics4
Cefepime 1/21 (4.5%) 3/21 (14.3%) 0.57 0.32 (0.04-2.82)
Cefoperazone + sulbactam 1/21 (4.5%) 2/23 (8.7%) 1.0 0.50 (0.05-5.14)
Cefuroxime 1/30 (3.3%) 8/31 (25.8%) 0.03 0.13 (0.02-0.97)
Amoxicillin + clavulanic acid 2/36 (5.6%) 9/37 (24.3%) 0.02 0.23 (0.05-0.98)
Erythromycin 0/18 (0%) 4/23 (17.4%) 0.12 0
Others 1/10 (10%) 2/9 (22.2%) 0.92 0.45 (0.05-4.16)

1 iv AB = intravenous antibiotics.
2 RR = relative risk, CI = confidence interval.
3 SD = standard deviation.
4 The difference in number of children with antibiotics between Tables 1 and 3 is due to the withdrawals.

332 Beneficial Microbes 4(4)


 S. boulardii in diarrhoea and antibiotic-associated diarrhoea

Table 4. Efficacy of Saccharomyces boulardii in children from the initial control group that developed diarrhoea.

Group B1 (n=23): Group B2 (n=19): t-value or χ2 P-value


S. boulardii + ORS1 ORS1

Day 0
Stool frequency (n/day) 4.03±0.61 3.98±0.52 0.28 0.779
Day 3
Stool frequency (n/day) 2.22±1.89 4.01±2.15 2.87 0.006
Recovery rate (%) 19/23 (82.6) 3/19 (15.8) χ2=18.62 <0.001
Day 5
Stool frequency (n/day) 1.01±0.85 3.98±1.89 6.766 <0.001
Recovery rate (%) 21/23 (91.3) 4/19 (21.1) χ2=21.31 <0.001

1 ORS = oral rehydration solution..

developed diarrhoea during antibiotic treatment or boulardii is not inactivated by gastric acid and antibiotics,
during the following two weeks, shortened the duration except for fungicides. These properties make S. boulardii an
of diarrhoea. After 3 days, diarrhoea had stopped in 83% attractive probiotic to test in the prevention and treatment
of the children in the S. boulardii group compared to of AAD.
only 16% in the untreated group. There were no adverse
reactions. These results suggest that S. boulardii offers an A major shortcoming of this trial is its ‘open’ design.
appropriate and safe therapeutic intervention to prevent Because there is no placebo control group, a placebo effect
and treat diarrhoea and AAD. cannot be excluded. However, a previous placebo-controlled
trial reported similar results (Johnston et al., 2011). The
Antibiotic use in children is three times higher than in age-range of the patients included is quite wide, but other
adults due to more frequent infections in that age group studies used similar inclusion criteria (McFarland, 1998).
(Szajewska et al., 2006). Besides being an effective treatment Another limitation of this study is the 2-week follow-up.
for bacterial infections, antibiotics may also induce AAD AAD may occur within 2 months after antibiotic treatment
by disrupting the balance of the intestinal flora. Antibiotics had stopped, therefore, some AAD cases might not have
may cause direct damage to the intestinal mucosa and been included. In addition, we only tested for the major
atrophy of intestinal epithelium, and reduce the activity of stool pathogens; other pathogens may have been missed.
cellular enzymes (disaccharidases), which leads to diarrhoea Therefore, AAD in this study was defined as diarrhoea
and abdominal pain (Johnston et al., 2011). Children may caused by C. difficile or as unexplained diarrhoea that
be prone to AAD because of their still-developing immune might be related to antibiotics (McFarland, 1998). Only
system. All antibiotics can cause AAD depending on type the children without acute or chronic diarrhoea and without
and duration of administration (Bergogne-Berezin, 2000) other chronic gastrointestinal diseases were enrolled in this
rather than dosage or administration route (McFarland, study to reduce interference.
1998). While AAD in children is estimated at 11-40%, it has
been reported that incidence for cephalosporin, penicillin, 5. Conclusions
and clindamycin may be higher (Kotowska et al., 2005).
Similar results were obtained in our study. The results of the present study confirm that prophylactic
use of oral S. boulardii reduces the risk to develop
The yeast S. boulardii secretes proteins that lower the (antibiotic-associated) diarrhoea during treatment or during
cAMP activity of intestinal epithelium cells and, therefore, two weeks after antibiotic administration had stopped. It is
reduces the secretory watery diarrhoea (Czerucka et al., for the first time that the efficacy of S. boulardii is suggested
1994). Moreover, S. boulardii increases production of when diarrhoea occurs during antibiotic treatment or
polyamines and activates mucosal enzymes (Buts et al., the following two weeks. Future double-blind placebo-
1999; Vandenplas et al., 2007). Furthermore, as it secretes controlled trials are needed to confirm these data.
proteases, S. boulardii can directly hydrolyse the toxins
secreted by C. difficile, resulting in a reduction of the Conflict of interest
occurrence of AAD. In addition, S. boulardii can stimulate
the gastrointestinal immune system, such as intestinal Y. Vandenplas is consultant for Biocodex and United
phagocytosis, intestinal secretary IgA, and other non- Pharmaceuticals.
specific immunity functions (Czerucka et al., 2007). S.

Beneficial Microbes 4(4) 333


L.-S. Shan et al.

References Kotowska, M., Albrecht, P. and Szajewska, H., 2005. Saccharomyces


boulardii in the prevention of antibiotic-associated diarrhoea in
Barbut, F., Meynard, J.L., Guiguet, M., Avesani, V., Bochet, M.V., children: a randomized double-blind placebo-controlled trial.
Meyohas, M.C., Delmee, M., Tilleul, P., Frottier, J. and Petit, J.C., Alimentary Pharmacology and Therapeutics 21: 583-590.
1997. Clostridium difficile-associated diarrhea in HIV-infected Lewis, S.J. and Heaton, K.W., 1997. Stool form scale as a useful guide
patients: epidemiology and risk factors. Journal of Acquired Immune to intestinal transit time. Scandinavian Journal of Gastroenterology
Deficiency Syndromes and Human Retrovirology 16: 176-181. 32: 920-924.
Bartlett, J.G., Chang, T.W., Gurwith, M., Gorbach, S.L. and Onderdonk, McFarland, L.V., 1998. Epidemiology, risk factors and treatments for
A.B., 1978. Antibiotic-associated pseudomembranous colitis due antibiotic-associated diarrhea. Digestive Diseases 16: 292-307.
to toxin-producing clostridia. New England Journal of Medicine McFarland, L.V., Surawicz, C.M. and Stamm, W.E., 1990. Risk factors
298: 531-534. for Clostridium difficile carriage and C. difficile-associated diarrhea
Bergogne-Berezin, E., 2000. Treatment and prevention of antibiotic in a cohort of hospitalized patients. Journal of Infectious Diseases
associated diarrhea. International Journal of Antimicrobial Agents 162: 678-684.
16: 521-526. Ruszczynski, M., Radzikowski, A. and Szajewska, H., 2008. Clinical
Buts, J.P., 2009. Twenty-five years of research on Saccharomyces trial: effectiveness of Lactobacillus rhamnosus (strains E/N, Oxy
boulardii trophic effects: updates and perspectives. Digestive and Pen) in the prevention of antibiotic-associated diarrhoea in
Diseases and Sciences 54: 15-18. children. Alimentary Pharmacology and Therapeutics 28: 154-161.
Buts, J.P., De Keyser, N., Marandi, S., Hermans, D., Sokal, E.M., Szajewska, H., Ruszczynski, M. and Radzikowski, A., 2006. Probiotics
Chae, Y.H., Lambotte, L., Chanteux, H. and Tulkens, P.M., 1999. in the prevention of antibiotic-associated diarrhea in children: a
Saccharomyces boulardii upgrades cellular adaptation after proximal meta-analysis of randomized controlled trials. Journal of Pediatrics
enterectomy in rats. Gut 45: 89-96. 149: 367-372.
Czerucka, D., Piche, T. and Rampal, P., 2007. Review article: yeast as Turck, D., Bernet, J.P., Marx, J., Kempf, H., Giard, P., Walbaum, O.,
probiotics – Saccharomyces boulardii. Alimentary Pharmacology Lacombe, A., Rembert, F., Toursel, F., Bernasconi, P., Gottrand, F.,
and Therapeutics 26: 767-778. McFarland, L.V. and Bloch, K., 2003. Incidence and risk factors
Czerucka, D., Roux, I. and Rampal, P., 1994. Saccharomyces of oral antibiotic-associated diarrhea in an outpatient pediatric
boulardii inhibits secretagogue-mediated adenosine 3’,5’-cyclic population. Journal of Pediatric Gastroenterology and Nutrition
monophosphate induction in intestinal cells. Gastroenterology 37: 22-26.
106: 65-72. Vandenplas, Y., Brunser, O. and Szajewska, H., 2009. Saccharomyces
Elstner, C.L., Lindsay, A.N., Book, L.S. and Matsen, J.M., 1983. Lack of boulardii in childhood. European Journal of Pediatrics 168: 253-265.
relationship of Clostridium difficile to antibiotic-associated diarrhea Vandenplas, Y., Salvatore, S., Vieira, M., Devreker, T. and Hauser,
in children. Pediatric Infectious Disease 2: 364-366. B., 2007. Probiotics in infectious diarrhoea in children: are they
Erdeve, O., Tiras, U. and Dallar, Y., 2004. The probiotic effect of indicated? European Journal of Pediatrics 166: 1211-1218.
Saccharomyces boulardii in a pediatric age group. Journal of Tropical Vandenplas, Y., Veereman-Wauters, G., De Greef, E., Peeters, S.,
Pediatrics 50: 234-236. Casteels, A., Mahler, T., Devreker, T. and Hauser, B., 2011. Probiotics
Guandalini, S., 2011. Probiotics for prevention and treatment of and prebiotics in prevention and treatment of diseases in infants
diarrhea. Journal of Clinical Gastroenterology 45 Suppl.: S149-S153. and children. Jornal de Pediatria 87: 292-300.
Hennequin, C., Thierry, A., Richard, G.F., Lecointre, G., Nguyen, H.V., Wistrom, J., Norrby, S.R., Myhre, E.B., Eriksson, S., Granstrom, G.,
Gaillardin, C. and Dujon, B., 2001. Microsatellite typing as a new Lagergren, L., Englund, G., Nord, C.E. and Svenungsson, B., 2001.
tool for identification of Saccharomyces cerevisiae strains. Journal Frequency of antibiotic-associated diarrhoea in 2462 antibiotic-
of Clinical Microbiology 39: 551-559. treated hospitalized patients: a prospective study. Journal of
Johnston, B.C., Goldenberg, J.Z., Vandvik, P.O., Sun, X. and Guyatt, Antimicrobial Chemotherapy 47: 43-50.
G.H., 2011. Probiotics for the prevention of pediatric antibiotic-
associated diarrhea. Cochrane Database of Systematic Reviews 11
Art. No. CD004827. DOI: http://dx.doi.org/10.1002/14651858.
CD004827.pub3.

334 Beneficial Microbes 4(4)

You might also like