Sei sulla pagina 1di 8

ORIGINAL ARTICLE: GASTROENTEROLOGY

Effect of Probiotics on Diarrhea in Children With Severe


Acute Malnutrition: A Randomized Controlled
Study in Uganda

Benedikte Grenov, yHanifa Namusoke, yBetty Lanyero, yNicolette Nabukeera-Barungi,

Christian Ritz, Christian Mlgaard, Henrik Friis, and Kim F. Michaelsen

ABSTRACT

Objectives: The aim of the study was to assess the effect of probiotics on What Is Known
diarrhea during in- and outpatient treatment of children with severe acute
malnutrition (SAM).  Severe acute malnutrition is responsible for 0.5 to 1
Methods: A randomized, double-blind, placebo-controlled study was
million child deaths annually and diarrhea is a com-
conducted involving 400 children admitted with SAM. Patients received 1
mon morbidity associated with mortality.
daily dose of a blend of Bifidobacterium animalis subsp lactis and  Probiotics reduce the duration of acute diarrhea by 1
Lactobacillus rhamnosus (10 billion colony-forming units, 50:50) or
day in well-nourished or mildly malnourished chil-
placebo during hospitalization followed by an 8- to 12-week outpatient
dren and reduce the risk of antibiotic-associated
treatment period, depending on patients recovery rate. All outcomes were
diarrhea.
reported for in- and outpatient treatment separately. The primary outcome was
number of days with diarrhea during hospitalization. Secondary outcomes
included other diarrhea outcomes, pneumonia, weight gain, and recovery. What Is New
Results: There was no difference in number of days with diarrhea between
 The probiotics Lactobacillus rhamnosus and Bifido-
the probiotic (n 200) and placebo (n 200) groups during inpatient
treatment (adjusted difference 0.2 days, 95% confidence interval 0.8 bacterium animalis subsp lactis had no effect on
to 1.2, P 0.69); however, during outpatient treatment, probiotics reduced diarrhea during inpatient treatment of children with
days with diarrhea (adjusted difference 2.2 days 95% confidence interval severe acute malnutrition, but reduced days with
3.5 to 0.3, P 0.025). There were no effects of probiotics on diarrhea diarrhea during outpatient treatment by 26%.
 The study results do not support using probiotics for
incidence and severity or pneumonia, weight gain or recovery during in- or
outpatient treatment. Twenty-six patients died in the probiotic versus 20 in inpatient treatment of children with severe acute
the placebo group (P 0.38). malnutrition, but may have a role in outpatient
Conclusions: Bifidobacterium animalis subsp lactis and Lactobacillus treatment.
rhamnosus had no effect on diarrhea in children with SAM during
hospitalization, but reduced the number of days with diarrhea in
outpatient treatment by 26%. Probiotics may have a role in follow-up of
hospitalized children with SAM or in community-based treatment
of malnourished children, but further studies are needed to confirm this. susceptible and case fatality rates in many sub-Saharan hospitals are
Key Words: diarrhea, low-income country, probiotic, severe acute often above 20% (3). Diarrhea is a major complication to SAM
malnutrition, young children associated with increased morbidity, longer hospitalization, and
death (4,5).
(JPGN 2017;64: 396403) Meta-analyses have shown that probiotics reduce the
duration of acute infectious diarrhea by approximately 1 day and
reduce the risk of acute diarrhea lasting 4 days or more (6). The

S evere acute malnutrition (SAM) is a major challenge in low-


income countries and results in 0.5 to 1 million child deaths
annually (1,2). Hospitalized children with SAM are particularly
dosage and the strains used were, however, different. Investigation
of strain-specific effects have led to recommendation of 2 strains,
Lactobacillus rhamnosus (LGG) and Saccharomyces boulardii, in

Received August 29, 2016; accepted December 27, 2016. www.isrctn.com registration number: ISRCTN16454889.
From the Department of Nutrition, Exercise, and Sports, University of Chr. Hansen A/S, University of Copenhagen and Innovation Fund Denmark
Copenhagen, Denmark, and the yMwanamugimu Nutrition Unit, Depart- funded the study. Chr. Hansen A/S suggested the intervention
ment of Pediatrics and Child Health, Mulago National Referral Hospital (combination of probiotic strains and dosage) but did not have any
Kampala, Uganda. role in study design, data collection, data analysis, data interpretation, or
Address correspondence and reprint requests to Prof Kim F. Michaelsen, Dr writing of the report. The corresponding author had full access to all the
Med Sci, Department of Nutrition, Exercise, and Sports, Faculty of data in the study and had final responsibility for the decision to submit
Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg for publication. B.G. was employed by Chr. Hansen A/S as an industrial
C, Denmark (e-mail: KFM@nexs.ku.dk). PhD student with parallel enrolment at University of Copenhagen. Half
Supplemental digital content is available for this article. Direct URL citations of her salary was sponsored by Innovation Fund Denmark. K.F.M., H.F.,
appear in the printed text, and links to the digital files are provided in the and C.M. received a grant from Chr. Hansen and Innovation Fund
HTML text of this article on the journals Web site (www.jpgn.org). Denmark for the current study, and K.F.M. and C.M. received a grant

396 JPGN  Volume 64, Number 3, March 2017

Copyright ESPGHAN and NASPGHAN. All rights reserved.


JPGN  Volume 64, Number 3, March 2017 Effect of Probiotics on Diarrhea in Children With SAM

the treatment of acute gastroenteritis in children (7). Both strains weight-for-Length z score (WHZ/WLZ) <3 or bipedal pitting
seem to reduce duration of acute diarrhea by 1 day and the risk of edema) were eligible. Patients with shock or severe
having diarrhea on day 3 or 4 by approximately 50% (7). Probiotics respiratory difficulty at admission, weight below 4.0 kg, obvious
may also reduce the risk of antibiotic-associated diarrhea and the disability or significant congenital or malignant disease
duration of persistent diarrhea (810). and patients admitted with SAM the previous 6 months
Pneumonia is the most frequent infectious cause of mortality were excluded.
in children below 5 years and the risk of dying if severely mal-
nourished is several folds higher (11). Studies on the effects of Randomization, Allocation Concealment,
probiotics on pneumonia are scarce; however, a number of studies
have indicated that probiotics may reduce the risk of upper respir-
and Blinding
atory infections (12). Furthermore, a few probiotic studies have The study products were labeled with a 4-digit number. There
shown a small improvement in growth (13). were 4 different 4-digit numbers, 2 for placebo and 2 for probiotics.
Some of the studies mentioned above included mildly to Only the study supply coordinator at Chr. Hansen A/S had access to
moderately malnourished children from low-income countries, but the blinding code. The randomization list was generated by a person
most studies were conducted in well-nourished children from high- not involved in the study using the web site Randomization.com
income countries. The large ProNUT study (an intervention study (http://www.randomization.com, accessed February 6, 2014).
testing probiotics and prebiotics supplied in PlumpyNut for chil- Randomization to probiotic or placebo treatments was performed
dren with SAM) investigated the effects of a mixture of pro- and in a 1:1:1:1 ratio in blocks of 4 and 8 in random order. The
prebiotics in Malawian children with SAM (14). There was no randomization list was kept by the head of MNU and only made
effect on nutritional cure, or on death, weight gain, or time to cure, available to staff responsible for pre-packaging of study products.
but a trend toward lower mortality (relative risk [RR] 0.65, These staff members were not involved in patient enrolment or
P 0.06) among patients receiving pro- and prebiotics in the treatment. Subject allocation was performed by assigning eligible
outpatient period was observed. subjects to the first available randomization number in consecutive
Bifidobacterium animalis subsp lactis (BB-12) and LGG are order. All participants, caregivers, investigators, and staff involved
among the most widely consumed and studied probiotic strains. in the study were blinded until the database was locked. The
Earlier studies have indicated that these strains reduce diarrhea and appearance, taste, and smell of the products were identical, except
upper respiratory infections in well-nourished children (1518) and for the 4-digit number.
that LGG may reduce diarrhea in mildly to moderately under-
nourished children (1921). We aimed at assessing the effect of a
combination of BB-12 and LGG on diarrhea, pneumonia, and Intervention and Procedures
growth in children admitted with SAM.
Patients received 1 sachet of study product daily in addition
METHODS to standard treatment of SAM. The study product was administered
from hospital admission to discharge and throughout an outpatient
Design and Ethics treatment period of minimum 8 weeks and maximum 12 weeks,
The ProbiSAM study was a randomized, double-blind, placebo- depending on the nutritional recovery of each child. Each sachet
controlled, 2-arm parallel-group study in children with SAM. contained 1 g of white powder: maltodextrin with or without a
The study was conducted at Mwanamugimu Nutrition Unit combination of the 2 probiotic strains BB-12 and LGG (dosage 10
(MNU), Department of Pediatrics and Child Health, Mulago billion colony-forming units [CFU], 50:50). Study products were
National Referral Hospital, Kampala, Uganda. The mortality rate manufactured by Chr. Hansen A/S, Hrsholm, Denmark.
at the unit is approximately 20%. During hospitalization, study personnel administered, regis-
The study was performed in accordance with the principles in tered, and supervised consumption of study products, which were
the Declaration of Helsinki. Ethical approval was obtained from the provided together with the morning food. Standard treatment of
Makerere University School of Medicine Research Ethics Committee SAM was given in compliance with World Health Organization
in Uganda and a consultative approval was given by The National (WHO) recommendations (22) and the Ugandan national protocol
Committee of Health Research Ethics in Denmark. Oral and written (23). Standard treatment involved a stabilization phase where
information about the study was provided and written; informed children received F-75 formula (Nutriset, Malaunay, France) fol-
consent was obtained from all caregivers before enrolment in the lowed by a rehabilitation phase with step-wise transitioning to
study. An independent data safety monitoring board (DSMB) was ready-to-use-therapeutic-food (RUTF; PlumpyNut, Nutriset) or
established to monitor patient safety throughout the study. F-100 formula (Nutriset) if RUTF was not tolerated well. A
commercial soy-based, lactose-free infant formula (Isomil, Abbott,
Participants Chicago, IL) was used if patients were suspected to have lactose
intolerance based on acidic stool pH and profuse diarrhea with >10
Children of age 6 to 59 months admitted with SAM stools per day. Breast-feeding was encouraged throughout the study.
(mid-upper-arm-circumference <11.5 cm or weight-for-height/ Study staff measured weight daily and assessed children daily for

from Chr. Hansen for another probiotic study with Danish infants. The article distributed under the terms of the Creative Commons Attribution-
remaining authors report no conflicts of interest. Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it
Copyright # 2017 The Author(s). Published by Wolters Kluwer Health, Inc. is permissible to download and share the work provided it is properly
on behalf of the European Society for Pediatric Gastroenterology, cited. The work cannot be changed in any way or used commercially
Hepatology, and Nutrition and the North American Society for Pediatric without permission from the journal.
Gastroenterology, Hepatology, and Nutrition. This is an open access DOI: 10.1097/MPG.0000000000001515

www.jpgn.org 397

Copyright ESPGHAN and NASPGHAN. All rights reserved.


Grenov et al JPGN  Volume 64, Number 3, March 2017

vital signs, signs of pneumonia (respiratory rate, chest in-drawing, score that includes grading of stool frequency, diarrhea duration,
respiratory auscultation, oxygen saturation), degree of edema and vomit frequency and duration, temperature, dehydration, and need
dehydration, and other signs of illness or adverse events. Triple of hospitalization. Dehydration or temperature during outpatient
measurements of weight and length/height were performed weekly treatment was not assessed. To be conservative, children were
and a thorough physical examination and anthropometry was considered not to be dehydrated and if the caregiver ticked
performed at admission and discharge. Antibiotics were adminis- fever in the stool diary, it was considered to be the lowest
tered as part of standard treatment for minimum 5 days. Ampicillin fever score on the Vesikari scale. The Vesikari scale categorizes
and gentamycin were first-line antibiotics, and second- and third- diarrhea episodes according to the following ranges: 7 mild, 7 to
line antibiotics included chloramphenicol, ceftriaxone, cloxacillin, 10 moderate, and 11 severe. Second, pneumonia incidence,
and ciprofloxacin. duration and severity for inpatients, and pneumonia incidence
Diarrhea data were collected using a stool diary in which for outpatients. Pneumonia was diagnosed according to clinical
caregivers ticked every time their child passed stool. Each stool was assessment by a pediatrician. Severity of pneumonia was cate-
categorized as watery, abnormally loose, loose, or normal according gorized as mild-moderate or severe. Duration and severity
to a photo scale. Caregivers were thoroughly trained in how to use could not be assessed during outpatient treatment as children
the diary and nutritionists supported and evaluated caregivers were only observed at follow-up visits every second week. Third,
ability to score stool consistency and fill out the stool diary weight gain (g/kg bodyweight per day) for in- and outpatients,
correctly. Vomiting, fever, and consumption of the study product respectively, and recovery defined as WHZ/WLZ >2 at study
were also recorded in the diary. Development and validation termination. Fourth, other outcomes included days with fever or
of the stool diary is described elsewhere (24). Stool frequency vomiting during in- and outpatient treatment and duration
and consistency scoring had a high validity, good reliability, and of hospitalization.
high sensitivity. Safety outcomes included mortality and other adverse events.
During outpatient treatment, children received RUTF at Due to the high background morbidity and mortality in the study
200 kcal/kg bodyweight per day. Follow-up visits were scheduled population, only medical conditions that were judged by a study
every second week to assess the children with respect to anthro- pediatrician to be uncommon in this population were recorded as
pometry, medical history, physical examination, follow-up on stool other adverse events.
diary data, and to deliver new supplies of RUTF, study products, and
stool diaries. The study staff did not attempt to fill the diary together
with the caregiver if data were missing. Caregivers were contacted Statistical Analysis
by phone once a week to ask about the status of their child and to Sample Size Calculation
remind them about study procedures and visit dates. If caregivers To have 80% power at 5% significance level to detect a 0.3
missed a scheduled follow-up visit, they were again contacted by SD reduction in number of days with diarrhea, 178 children were
phone or a home visit was done to assess reasons for failure to return needed per study arm. To account for loss to follow-up, 200 children
for a follow-up visit. Study product compliance during outpatient were recruited per arm. Assuming that the SD of days with diarrhea
care was estimated based on ticks in stool diaries, returned empty or at MNU was 3 days, it would be possible to detect a 1-day reduction
unused sachets, and differences between visit dates and number of in days with diarrhea, which is similar to what is found in meta-
sachets supplied. analyses (6).
Outcomes
Statistical Analyses
The primary outcome was duration of diarrhea during hos-
pitalization. Duration was defined as number of days with diar- Data were double-entered in EpiData v.3.1 (EpiData,
rhea of each patient. Diarrhea was defined as 3 loose or watery Odense, Denmark) and analyzed using statistical software R version
stools per 24 hours (25) based on stool diary data. A diarrhea 3.1.1 (2014-07-10) (27).
episode started when the diarrhea definition was fulfilled and was The primary outcome and the secondary outcomes of diar-
considered to have ceased when the child passed <3 loose or watery rhea severity, weight gain, hospitalization, fever, and vomiting were
stools per day. If diarrhea re-appeared after <48 hours, it was analyzed using linear-mixed models with subject-specific random
considered part of the same diarrhea episode, but only days with effects. Remaining secondary outcomes were analyzed either using
3 loose or watery stools were counted as diarrhea days. The logistic regression (mixed-effects) models (incidence of diarrhea
protocol was amended during the clinical study and approved by the and pneumonia, fever, recovery) or log-linear Poisson models with
ethical committee. In the original protocol, the primary outcome adjustment for overdispersion (diarrhea days in outpatients, pneu-
was phrased as duration of diarrhea episodes without any monia duration, and severity inpatients). All models were adjusted
specification of in- or outpatient treatment. We considered the total for age, sex, human immunodeficiency virus (HIV) status, baseline
number of days with diarrhea of each child to be clinically more edema, and WHZ/WLZ. Mortality was analyzed using a Cox-
important than the duration of each episode. In addition, we decided regression adjusted for sex and age only as HIV data were missing
to split analyses in in- and outpatient treatment as the populations for a number of patients that died. In addition, analyses of inpatient
and data collection were different in the 2 periods with more data included adjustment for duration of hospitalization and base-
critically ill patients and closer monitoring of patients during line diarrhea or pneumonia. All analyses of diarrhea inpatient data
inpatient treatment. were repeated with additional adjustment for Isomil treatment and
Secondary outcomes were, first, number of days with reduction in days with diarrhea during outpatient treatment was
diarrhea during outpatient treatment and incidence and severity repeated with adjusted for duration of outpatient treatment. Effect
of diarrhea during inpatient and outpatient treatment. Diarrhea modification was also investigated for age, sex, HIV status, duration
incidence was defined as the proportion of children with mini- of hospitalization and baseline edema, WHZ/WLZ, and diarrhea.
mum 1 day of diarrhea. Severity was defined as the Vesikari score Model checking was based on residuals and predicted random
for inpatients and a modified Vesikari score for outpatients (26). effects, which were evaluated visually using (cumulated) residual
The Vesikari score is a multidomain diarrhea episode severity plots and normal probability plots.

398 www.jpgn.org

Copyright ESPGHAN and NASPGHAN. All rights reserved.


JPGN  Volume 64, Number 3, March 2017 Effect of Probiotics on Diarrhea in Children With SAM

Intention-to-treat analysis was performed on all patients with boys, 66% had edematous malnutrition, 14% were HIV seroposi-
any available data related to the specific outcome, assuming that tive, and 34% had an HIV-positive mother. The total loss to follow-
drop-outs occurred at random. Intermittent missing values in inpa- up, including deceased patients, was 18% (n 73/400) and 12%
tient diarrhea data were imputed to obtain complete episodes, which (n 38/327) during in- and outpatient treatment, respectively. The
were needed for the primary outcome and for calculation of the numbers were equally distributed between the 2 study groups. No
Vesikari score. Specifically, deterministic hot-deck imputation was patients were discharged to outpatient treatment with edema, but
used: gaps in stool diaries were imputed using data from multiple 44% still had SAM with no medical complications (see Supple-
matched patients with complete data for the same days of hospital- mental Digital Content 1, Table, http://links.lww.com/MPG/A882).
ization as the gap occurred and an identical diarrhea pattern before/ The mean duration of hospitalization was 181  9.2 days and most
after the gap as the patient having the gap (28). For the primary patients (>80%) were discharged from outpatient treatment after 8
outcome, a per-protocol analysis was also carried out and a subgroup weeks. The number of patients included in intention-to-treat diar-
analysis was performed to investigate if the subgroup of children who rhea analyses during in- and outpatient treatment was n 369
were discharged and analyzed in the outpatient phase showed a (probiotics n 187, placebo n 182) and n 289 (probiotics
different result in the primary outcome than the total population. n 147, placebo n 145), respectively. The main reasons for
At predefined, regular intervals, the DSMB monitored patients not being included in the statistical models were lack of
safety with specific focus on mortality by evaluating unblinded data on HIV status or lack of stool diaries in the outpatient period.
individual case fatality reports, serious adverse events, and the Compliance to study product consumption was 98% for both
reasons of patients lost to follow-up. The study was registered at groups during inpatient treatment based on study staff registrations.
www.isrctn.com as ISRCTN16454889. During the outpatient phase, compliance was estimated at 93% and
96% for probiotics and placebo, respectively, based on comparison
of the number of study products delivered and the number of days
RESULTS between follow-up visits.
Of 757 children screened, 400 children were randomized to There was no difference in the primary outcome between the
receive probiotics (n 200) or placebo (n 200) (Fig. 1). Patients probiotic and placebo groups with an adjusted difference of mean
were recruited between March 10, 2014 and July 8, 2015 and number of days with diarrhea of 0.2 days (95% confidence
followed until October 2015. interval [CI] 0.8 to 1.2, P 0.69) (Table 2). During outpatient
Baseline characteristics were comparable between the 2 treatment, number of days with diarrhea was lower in the probiotic
study groups (Table 1). Mean age was 17.0 months, 58% were compared with the placebo group with an adjusted difference of

757 screened
357 excluded
307 ineligible
17 declined
33 other
400 randomized

200 probiotics 200 placebo

37 lost to follow-up 36 lost to follow-up


23 deceased 16 deceased
13 self-discharged 16 self-discharged
1 other 4 other

187 included in 182 included in


inpatient ITT-analysis inpatient ITT-analysis

163 discharged 164 discharged

20 lost to follow-up 17 lost to follow-up


3 deceased 4 deceased
17 self-discharged 13 self-discharged

147 included in 145 included in


outpatient ITT-analysis outpatient ITT-analysis

143 completed 145 completed

FIGURE 1. Trial profile. Dotted line: patients that were lost to follow-up were included in the intention-to-treat analysis on an available case basis.
The main reasons for patients not being included in the analyses were lack of data on HIV status or no stool diaries available (mainly during
outpatient treatment). ITT intention-to-treat.

www.jpgn.org 399

Copyright ESPGHAN and NASPGHAN. All rights reserved.


Grenov et al JPGN  Volume 64, Number 3, March 2017

TABLE 1. Baseline characteristics subpopulation of inpatients that were included in the outpatient
analysis showed an adjusted difference of 0.1 days (95% CI 1.1
Probiotics Placebo to 1.2 days, P 0.91). There was no effect on modification of the
(n 200) (n 200) primary outcome by any of the covariates (data not shown). Days
with diarrhea during outpatient treatment resulted in similar results
Age (mo  SD) 17.5  85 16.5  84 after adjustment for duration of outpatient treatment (data not
Sex (male, %) 115 (58%) 115 (58%) shown).
Weight (kg  SD) 7.0  1.7 6.9  1.7
Height or length or height 71.1  6.6 71.0  6.1
(cm  SD) DISCUSSION
MUAC (cm  SD) 11.6  1.4 11.5  1.5 Probiotics did not reduce number of days with diarrhea
Height/length-for-age z score 3.2  1.4 3.0  1.4 during hospitalization, whereas days with diarrhea were reduced
Weigth-for-age z score 3.5  1.3 3.5  1.3 by 2.2 days corresponding to 26% of the mean number of days with
Weight-for-height/length 2.6  1.5 2.7  1.6 diarrhea in the outpatient phase. The different results may be
z score explained by more severe illness and a compromised gut barrier
Edema (n, %) 132 (66%) 132 (66%) in children during hospitalization. Children admitted to MNU were
HIV-positive children 28 (15%) 24 (13%) often ill with multiple life-threatening conditions and it may have
(serology) (n, %) affected their ability to respond to probiotic treatment. More
HIV-positive mothers (n, %) 56 (34%) 53 (34%) specifically, their gut function may have been impaired resulting
Presenting symptoms at admission n (%) in poor adhesion of the probiotics to the mucosa.
Fever 101 (50%) 110 (55%) Antibiotic use was also different during in- and outpatient
Cough 131 (66%) 131 (66%) treatment. Broad-spectrum antibiotics were administered intrave-
Diarrhea 118 (59%) 126 (63%) nously as part of standard treatment during hospitalization whereas
Vomiting 84 (42%) 87 (44%) oral antibiotics were used only when children developed respiratory
Medical treatment 182 (91%) 184 (93%) or other infections during outpatient treatment. Both oral and
Household n (%) intravenously administered antibiotics are known to cause diarrhea
Child lives with mother 160 (80%) 157 (79%) in some patients probably by disturbing the gut microbiota and
Mother education primary 94 (47%) 117 (59%) reducing the colonization resistance to pathogens. The 2 probiotic
school or lower strains were sensitive to most of the used antibiotics and the
antibiotics may therefore have influenced the effectiveness of the
HIV human immunodeficiency virus; MUAC mid-upper-arm-
circumference; SD standard deviation.
probiotics. On the contrary, probiotic studies including LGG have
been shown to reduce the risk of antibiotic-associated diarrhea after
administration of broad-spectrum antibiotics (29).
2.2 days (95% CI 3.5 to 0.3, P 0.025). Supplemental Finally, diarrhea was more severe with higher stool frequency,
Digital Content 2, Figure, http://links.lww.com/MPG/A883, shows more dehydration, fever, and vomiting during inpatient compared to
the distribution of diarrhea days in the probiotic versus the placebo outpatient treatment. Meta-analyses of probiotics in general (6) or
group during outpatient care. As seen, the proportion of patients LGG alone (30) have indicated that the effect on acute diarrhea is
with diarrhea for 20 or more days was reduced in the probiotic higher in community-based than inpatient studies. The results, how-
group. ever, vary and a study with LGG in hospitalized children with acute
The incidence of diarrhea was 89% versus 85% in the diarrhea showed effect in patients with profuse diarrhea (19).
probiotic versus placebo group during hospitalization, odds ratio The ProNUT study investigated a combination of pro- and
(OR) 1.6 (95% CI 0.8 to 3.3, P 0.17) and 70% versus 76% in the prebiotics in children with SAM and also observed differences
outpatient period, OR 0.7 (95% CI 0.4 to 1.2, P 0.17). The during in- and outpatient periods (14). They found more vomiting,
severity of diarrhea episodes measured by the Vesikari score severe diarrhea (6 stools per day) and cough, and a small,
was comparable between the study groups during in- and outpatient nonsignificant increase in mortality in inpatients, whereas they
treatment. Episodes observed during inpatient treatment were more reported a trend toward reduced mortality and fewer cases of severe
severe (592 episodes, mean score 10.0 (520)) compared with diarrhea in outpatients receiving pro- and prebiotics. The increase in
outpatient episodes (752 episodes, mean score 4.3 (313)). severe diarrhea among inpatients was suggested to be related to the
Pneumonia incidence, duration, and severity were not differ- intake of prebiotics.
ent between the study groups during inpatient treatment. The Based on our results, it is not possible to conclude if the effect
incidence during outpatient treatment was 5% (n 8) in the pro- in outpatients is depending on probiotic administration initiated
biotic group and 10% (n 16) in the placebo group, but the before discharge. However, studies on treatment or prevention of
difference was insignificant (OR 0.5, 95% CI 0.2 to 1.3, diarrhea either started probiotic treatment together with or maxi-
P 0.17). Nutritional recovery, total weight gain (g/kg bodyweight mum 2 days before exposure to the cause of diarrhea (15,16,31,32).
per day), fever, vomiting, and duration of hospitalization did not Meta-analyses of probiotic studies with LGG on treatment of
differ significantly between the groups. acute diarrhea have shown a reduction of diarrhea duration by 1 day
Forty-six patients died during the course of the study; 39 (23 (30) and a study on persistent diarrhea reduced duration by 4 days
probiotics, 16 placebo) during hospitalization and 7 (3 probiotics, 4 (9). The meta-analyses on acute diarrhea found that LGG seems to
placebo) during outpatient treatment. There was no difference have slightly higher effect in studies with a dose of 10 billion CFU/
between the probiotic and placebo groups during the entire study day compared with studies with lower doses and there was a
(hazard ratio [HR] 1.3, 95% CI 0.7 to 2.3, P 0.38). Other tendency toward lower effect in non-European countries compared
adverse events were not reported. with European countries (30). Some older studies with BB-12 found
The per-protocol analysis (probiotics n 176, placebo an effect on diarrhea (33,34), but 2 new larger studies in hospital-
n 169) resulted in an adjusted difference of the primary outcome ized and community-based children showed no effect on diarrhea
of 0.2 days (95% CI 0.8 to 1.2, P 0.68) and analysis of the (35,36).

400 www.jpgn.org

Copyright ESPGHAN and NASPGHAN. All rights reserved.


JPGN  Volume 64, Number 3, March 2017 Effect of Probiotics on Diarrhea in Children With SAM

TABLE 2. Results of probiotic treatment on diarrhea, pneumonia and nutritional recovery during in- and outpatient treatment

Outcome Probiotics (N 200) Placebo (N 200) Adjusted effect size (95% CI) probiotics vs placebo

Diarrhea

Duration, days
Inpatient 6.9  6.0 6.5  6.4 0.2 (0.8 to 1.2), P 0.69
Outpatient 6.0  8.2 8.5  10.9 2.2 (3.5 to 0.3), P 0.025
Incidencey n (%)
Inpatient 177 (89%) 169 (85%) OR 1.6 (0.8 to 3.3), P 0.17
Outpatient 107 (70%) 116 (76%) OR 0.7 (0.4 to 1.2), P 0.17
Severity (Vesikari score 020)z
Inpatient episodes 299 (51%) 293 (49%)
Inpatient scores 10.2  3.8 9.9  3.7 0.1 (0.6 to 0.8), P 0.74
Outpatient episodes 354 (47%) 398 (53%)
Outpatient scores 4.3  1.7 4.4  1.7 0.2 (0.5 to 0.1), P 0.16
Pneumonia
Duration, days
Inpatient 1.9  3.6 2.0  3.7 0.2 (0.7 to 0.4), P 0.51
Outpatient NA NA NA
Incidence
Inpatient 77 (39%) 80 (41%) OR 0.9 (0.6 to 1.5), P 0.80
Outpatient 8 (5%) 15 (10%) OR 0.5 (0.2 to 1.3), P 0.17
Severity (score)
Inpatient 2.6  5.5 2.7  5.3 0.3 (1.0 to 0.7), P 0.50
Outpatient NA NA NA
Nutritional recovery
Weight gain (g/kg bodyweight per day)jj
Inpatient 6.5  4.7 6.1  4.2 0.3 (0.6 to 1.3), P 0.49
Outpatient 3.0  2.1 3.2  2.3 0.1 (0.6 to 0.4), P 0.80
Total 4.1  2.2 4.2  2.2 0.02 (0.5 to 0.5), P 0.94
Recovered n (%) 133 (66%) 128 (64%) OR 1.2 (0.6 to 2.4), P 0.68
Other
Fever, days
Inpatient 7.0  5.1 6.7  4.4 0.3 (0.3 to 1.0), P 0.29
Outpatient 1.6  2.9 2.0  3.9 0.5 (1.3 to 0.2), P 0.16
Vomit, days
Inpatient 2.1  3.3 2.0  4.4 0.002 (0.7 to 0.7), P > 099
Outpatient 1.2  2.3 1.6  6.0 0.3 (1.4 to 0.7), P 0.53
Hospitalization, days
Inpatient 18.3  9.1 18.0  9.3 0.1 (1.7 to 1.9), P 0.93

Data are reported as mean  SD or n (%) unless otherwise indicated.


CI confidence interval; OR odds ratio.

Total number of days with diarrhea. Number of subjects analyzed: Inpatients: probiotics n 187, placebo n 182. Outpatients: probiotic n 147, placebo
n 145.
y
Incidence of diarrhea: Number of patients with minimum 1 day of diarrhea.
z
Vesikari score according to Lewis (23). For outpatients a modified Vesikari score was calculated, see methods.

Pneumonia was diagnosed based on clinical symptoms. Number of subjects analyzed: Inpatients: Probiotics, n 189, placebo n 183. Outpatients:
probiotics, n 148, placebo, n 149. Incidence: number of children with minimum 1 day with pneumonia. Severity: sum of daily pneumonia scores (0 no
pneumonia, 1 pneumonia, 2 severe pneumonia) during hospitalization. Pneumonia duration and severity were not possible to measure in outpatients as they
were only seen by a medical doctor every second week.
jj
Weight gain during inpatient treatment was calculated as the difference between discharge weight and the minimum weight during hospitalization and days
were total number of hospitalization days. Outpatient weight gain was calculated from discharge to week 8. Total weight gain was the difference between
minimum weight during hospitalization and week 8. Number of subjects analyzed: Inpatients probiotics n 159, placebo n 155, outpatients and total weight
gain: probiotics n 138, placebo n 142.

Recovery was defined as weight-for-length >2.

Diarrhea in the outpatient phase was usually mild according in the number of children with long diarrhea periods could improve
to the Vesikari scale, but a number of patients had prolonged the long-term nutritional status and reduce the risk of hospital
periods of diarrhea, especially in the placebo group (Supplemental admissions in community-based children with SAM.
Digital Content 2, Fig. http://links.lww.com/MPG/A883). This Regarding respiratory tract infections probiotics are mainly
could be associated with presence of environmental enteric dys- reported to prevent upper respiratory tract infections (12). Both BB-
function (EED), which involves increased gut permeability, 12andLGGhavebeenreportedtoreduceupperrespiratorytractinfection
reduced absorptive capacity and inflammation (37). A reduction (1517); however, sometimes with conflicting results (18,35,36).

www.jpgn.org 401

Copyright ESPGHAN and NASPGHAN. All rights reserved.


Grenov et al JPGN  Volume 64, Number 3, March 2017

We did not find any difference in weight gain between the 2. World Health Organization, World Food Programme, United Nations
probiotic and placebo groups, neither during hospitalization nor System Standing Committee on Nutrition, The United Nations Chil-
during outpatient treatment. The overall evidence regarding effect drens fund. Community-based management of severe acute malnutri-
of probiotics on growth is scarce (38). Onubi performed a systema- tion. A Joint Statement by the World Health Organization, the World
tic review of the effect of probiotics on growth in children and Food Programme, the United Nations System Standing Committee on
evaluated 12 studies (13). Five studies from low-income countries, Nutrition and the United Nations Childrens Fund, 2007. Report No.:
ISBN: 978-92-806-4147-9. http://www.who.int/nutrition/topics/State-
including 4 studies with undernourished children, showed a positive ment_community_based_man_sev_acute_mal_eng.pdf. Accessed Au-
effect on weight gain whereas 7 studies from high-income countries gust 10, 2016.
did not. Both studies on BB-12 or other strains belonging to the 3. Brewster DR. Inpatient management of severe malnutrition: time for a
same subspecies BB-12 and LGG were included in the review. change in protocol and practice. Ann Trop Paediatr 2011;31:97107.
Safety of probiotics in immune compromised and critically 4. Irena AH, Mwambazi M, Mulenga V. Diarrhea is a major killer of
ill patients has been discussed mainly due to a concern about risk of children with severe acute malnutrition admitted to inpatient set-up in
probiotic-related sepsis (39). The mortality rate was therefore Lusaka, Zambia. Nutr J 2011;10:110.
carefully followed throughout the study period by investigators 5. Talbert A, Thuo N, Karisa J, et al. Diarrhoea complicating severe acute
and the DSMB. In the current study, there was a small, nonsigni- malnutrition in Kenyan children: a prospective descriptive study of risk
factors and outcome. PloS One 2012;7:e38321.
ficantly higher number of patients that died in the probiotic group 6. Allen SJ, Martinez EG, Gregorio GV, et al. Probiotics for treating acute
(26 patients) compared with the placebo group (20 patients) infectious diarrhoea. Cochrane Database Syst Rev (11)2010CD003048.
(P 0.38). The mortality reports showed multiple severe medical 7. Szajewska H, Guarino A, Hojsak I, et al., European Society for Pediatric
complications in most of the children, complicating assessment of Gastroenterology, Hepatology, and Nutrition. Use of probiotics for
the exact cause of death. The most common causes of death, management of acute gastroenteritis: a position paper by the ESPGHAN
according to the mortality reports, were respiratory failure/severe Working Group for Probiotics and Prebiotics. J Pediatr Gastroenterol
pneumonia and shock/dehydration related to severe diarrhea. Sep- Nutr 2014;58:5319.
ticemia was considered to be a direct cause of death in 4 patients in 8. Goldenberg JZ, Lytvyn L, Steurich J, et al. Probiotics for the prevention
of pediatric antibiotic-associated diarrhea. Cochrane Database Syst Rev
each group and contributed to the underlying cause of death in 6
(12)2015CD004827.
patients in each group. There were no signs of consistent differences 9. Szajewska H, Canani RB, Guarino A, et al. Probiotics for the prevention
between the groups. of antibiotic-associated diarrhea in children. J Pediatr Gastroenterol
Strengths of the study include a randomized, double-blind Nutr 2016;62:495506.
controlled design, use of a validated stool diary and thorough 10. Bernaola Aponte G, Bada Mancilla CA, Carreazo NY, et al. Probiotics
training and monitoring of caregivers when they recorded childrens for treating persistent diarrhea in children. Cochrane Database Syst Rev
stool pattern. Lack of data on diarrhea etiology is a limitation of the (8)2013CD007401.
study. In children with SAM, diarrhea can be caused by both 11. Chisti MJ, Tebruegge M, La Vincente S, et al. Pneumonia in severely
infectious and noninfectious agents. This includes infections with malnourished children in developing countriesmortality risk, aetiol-
bacteria, viruses or parasites and diarrhea due to malabsorption, for ogy and validity of WHO clinical signs: a systematic review. Trop Med
Int Health 2009;14:117389.
example, secondary lactose intolerance and enteropathy. Probiotics 12. Hao Q, Dong BR, Wu T. Probiotics for preventing acute upper respira-
are likely to have different effects on these diarrhea etiologies, but tory tract infections. Cochrane Database Syst Rev (2)2015CD006895.
this was not assessed. The loss to follow-up may have resulted in 13. Onubi OJ, Poobalan AS, Dineen B, et al. Effects of probiotics on child
lower power, imprecise estimates of effects, and attrition bias. The growth: a systematic review. J Health Popul Nutr 2015;34:8.
drop-outs were, however, equally distributed in the probiotic and 14. Kerac M, Bunn J, Seal A, et al. Probiotics and prebiotics for severe acute
placebo groups. Finally, the results may not be generalizable to all malnutrition (PRONUT study): a double-blind efficacy randomised
children with SAM as children below 6 months, children with an controlled trial in Malawi. Lancet 2009;374:13644.
admission weight below 4 kg and children in shock or with severe 15. Hojsak I, Abdovic S, Szajewska H, et al. Lactobacillus GG in the
prevention of nosocomial gastrointestinal and respiratory tract infec-
respiratory distress were excluded from the study. Children with
tions. Pediatrics 2010;125:e1171.
any of these criteria belong to the most vulnerable children 16. Hojsak I, Snovak N, Abdovic S, et al. Lactobacillus GG in the preven-
with SAM. tion of gastrointestinal and respiratory tract infections in children who
The current results do not support using probiotics for the attend day care centers: a randomized, double-blind, placebo-controlled
treatment of hospitalized children with SAM and severe medical trial. Clin Nutr 2010;29:3126.
complications. The reduction of days with diarrhea in the outpatient 17. Taipale T, Pienihakkinen K, Isolauri E, et al. Bifidobacterium animalis
phase, especially among children with long diarrhea duration, may, subsp. lactis BB-12 in reducing the risk of infections in infancy. Br J
however, be important in future community-based treatment of Nutr 2011;105:40916.
children with SAM and may reduce admissions and mortality. But 18. Weizman Z, Asli G, Alsheikh A. Effect of a probiotic infant formula on
further studies are needed to clarify this potential effect. infections in child care centers: comparison of two probiotic agents.
Pediatrics 2005;115:59.
19. Basu S, Paul DK, Ganguly S, et al. Efficacy of high-dose Lactobacillus
Acknowledgments: The authors thank children and caregivers rhamnosus GG in controlling acute watery diarrhea in Indian children: a
for their contribution to the study, especially for filling out stool randomized controlled trial. J Clin Gastroenterol 2009;43:20813.
diaries and attending follow-up visits. The authors also thank the 20. Basu S, Chatterjee M, Ganguly S, et al. Effect of Lactobacillus
study team for their persistent commitment to the study and patient rhamnosus GG in persistent J Clin Gastroenterol 2007;41:756-60.
care. Funding was received from Chr. Hansen A/S, University of 21. Oberhelman RA, Gilman RH, Sheen P, et al. A placebo-controlled trial
Copenhagen and Innovation Fund Denmark with gratitude. of Lactobacillus GG to prevent diarrhea in undernourished Peruvian
children. J Pediatr 1999;134:1520.
22. World Health Organization. WHO. Guideline: Updates on the manage-
REFERENCES ment of severe acute malnutrition in infants and children. Geneva:
1. Black RE, Victora CG, Walker SP, et al. Maternal and child under- World Health Organization; 2013. Report No.: ISBN 978 92 4 150632 8.
nutrition and overweight in low-income and middle-income countries. http://www.who.int/nutrition/publications/guidelines/updates_manage-
Lancet 2013;382:42751. ment_SAM_infantandchildren/en/. Accessed August 10, 2016.

402 www.jpgn.org

Copyright ESPGHAN and NASPGHAN. All rights reserved.


JPGN  Volume 64, Number 3, March 2017 Effect of Probiotics on Diarrhea in Children With SAM

23. Ministry of Health, Uganda. Integrated Management of Acute Malnu- 32. Hilton E, Kolakowski P, Singer C, et al. Efficacy of Lactobacillus GG as
trition Guidelines; 2010. www.unicef.org/uganda/IMAM_Guidelines_ a diarrheal preventive in travelers. J Travel Med 1997;4:413.
final_version.pdf. Accessed August 10, 2016. 33. Saavedra JM, Bauman NA, Oung I, et al. Feeding of Bifidobacterium
24. Grenov B, Namusoke H, Nabukeera-Barungi N, et al. Validation of a bifidum and Streptococcus thermophilus to infants in hospital for
simple stool diary used by caregivers to document diarrhea among prevention of diarrhoea and shedding of rotavirus. Lancet
young children in a low-income country. J Pediatr Gastroenterol Nutr 1994;344:10469.
2016 [Epub ahead of print]. 34. Chouraqui J-P, Van Egroo L-D, Fichot M-C. Acidified milk formula
25. World Health Organization. Diarrhoea definition. http://who.int/topics/ supplemented with Bifidobacterium lactis: impact on infant diarrhea
diarrhoea/en/. Accessed August 10, 2016 in residential care settings. J Pediatr Gastroenterol Nutr 2004;38:
26. Lewis K, PATH. Vesikari Clinical Severity Scoring System Manual; 28892.
2011 May. http://www.path.org/publications/detail.php?i=2228. Ac- 35. Hojsak I, Tokic Pivac V, Mocic Pavic A, et al. Bifidobacterium animalis
cessed August 10, 2016. subsp. lactis fails to prevent common infections in hospitalized children:
27. R Core Team. R: A language and environment for statistical computing. a randomized, double-blind, placebo-controlled study. Am J Clin Nutr
R Foundation for Statistical Computing, Vienna, Austria: R Core Team; 2015;101:6804.
2013. http://www.R-project.org/. Accessed August 10, 2016. 36. Hojsak I, Mocic Pavic A, Kos T, et al. Bifidobacterium animalis subsp.
28. Little RJ, Yosef M, Cain KC, et al. A hot-deck multiple imputation lactis in prevention of common infections in healthy children attending
procedure for gaps in longitudinal data on recurrent events. Stat Med day care centers: randomized, double blind, placebo-controlled study.
2008;27:10320. Clin Nutr Edinb Scotl 2016;35:58791.
29. Szajewska H, Koodziej M. Systematic review with meta-analysis: Lacto- 37. Crane RJ, Jones KD, Berkley JA. Environmental enteric dysfunction: an
bacillus rhamnosus GG in the prevention of antibiotic-associated diarrhoea overview. Food Nutr Bull 2015;36(suppl 1):76S87S.
in children and adults. Aliment Pharmacol Ther 2015;42:114957. 38. Braegger C, Chmielewska A, Decsi T, et al. Supplementation of infant
30. Szajewska H, Skorka A, Ruszczynski M, et al. Meta-analysis: Lactobacillus formula with probiotics and/or prebiotics: a systematic review and
GG for treating acute gastroenteritis in childrenupdated analysis of comment by the ESPGHAN committee on nutrition. J Pediatr Gastro-
randomised controlled trials. Aliment Pharmacol Ther 2013;38:46776. enterol Nutr 2011;52:23850.
31. Vanderhoof JA, Whitney DB, Antonson DL, et al. Lactobacillus GG in 39. Urben LM, Wiedmar J, Boettcher E, et al. Bugs or drugs: are
the prevention of antibiotic-associated diarrhea in children. J Pediatr probiotics safe for use in the critically ill? Curr Gastroenterol Rep
1999;135:5648. 2014;16:388.

www.jpgn.org 403

Copyright ESPGHAN and NASPGHAN. All rights reserved.

Potrebbero piacerti anche