Next Article in Journal
Dietary Responses of Dementia-Related Genes Encoding Metabolic Enzymes
Next Article in Special Issue
Latilactobacillus sakei Wikim0066 Protects Skin through MMP Regulation on UVB-Irradiated In Vitro and In Vivo Model
Previous Article in Journal
Saturated Fatty Acids Consumed in Smoothies Increase Glucose and Metabolic Load Index in Young Adults Compared to Polyunsaturated Omega-3-Fatty Acids
Previous Article in Special Issue
Novel Horizons in Postbiotics: Lactobacillaceae Extracellular Vesicles and Their Applications in Health and Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathogen-Specific Benefits of Probiotic and Synbiotic Use in Childhood Acute Gastroenteritis: An Updated Review of the Literature

by
Maria Oana Săsăran
1,
Cristina Oana Mărginean
2,*,
Heidrun Adumitrăchioaiei
3 and
Lorena Elena Meliț
2
1
Department of Pediatrics III Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania
2
Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania
3
Doctoral School of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania
*
Author to whom correspondence should be addressed.
Submission received: 8 January 2023 / Revised: 24 January 2023 / Accepted: 25 January 2023 / Published: 27 January 2023
(This article belongs to the Special Issue Role of Lactobacillus and Probiotics in Human Health and Diseases)

Abstract

:
Probiotics represent viable microorganisms which are found within the normal gut microbiota, that exert strain-specific benefits in the management of several gastrointestinal disorders in children, including acute gastroenteritis. This review aims to evaluate the pathogen-specific role of probiotic supplementation in childhood diarrhea. A search of scientific databases was conducted to identify studies which investigated efficacy of probiotics and synbiotics in influencing outcome of acute gastroenteritis of known etiology. We identified 32 studies, most of which analyzed impact of probiotic supplementation in rotavirus gastroenteritis, while a very limited number of these conducted a separate analysis on bacterial diarrhea. Lactobacillus rhamnosus (L. rhamnosus), L. reuteri and S. boulardii still remain the most researched strains, with a proven role in decreasing diarrhea and hospitalization duration, especially in the setting of rotavirus infection. Combined products containing at least one of the aforementioned strains also performed similarly and might also influence rotavirus fecal shedding. Rotavirus immunization status has also been proposed as a significant influencing factor of probiotic use impact. The paucity of research focusing on bacterial etiologies, as well as of clinical trials conducted within ambulatory care units leaves room for further research on the matter, which needs to include larger cohort studies.

1. Introduction

Probiotics have been defined as oral supplements which contain viable microorganisms, in the form of bacteria and yeasts similar to those found within the microbiota of the normal, healthy gut [1,2]. The first evidence available in the literature regarding their existence comes from Henry Tissier, who was the first person to remark on a poorer stool bacterial culture in children infected with diarrhea, as well as in those fed with formula, as opposed to healthy and breastfed infants [3,4]. Since the publication of these first data in 1907, many studies have emerged, but their poor design and inadequate cultivation of bacteria in substrates other than human milk initially hindered the acquisition of reliable evidence. However, later research, and the ability to isolate and characterize specific bacterial cultures, showed the numerous health benefits of probiotics, which have been proven to improve intestinal health, alleviate symptoms related to lactose intolerance and decrease the risk of developing disorders such as inflammatory bowel disease, infectious diarrhea or allergies [3,5]. Thus, their administration in adequate amounts can be beneficial for the host and their lack of side effects has encouraged their widespread use [6].
Ensuring the equilibrium and integrity of intestinal microflora facilitates a functioning intestinal mucosal defense system, which in turn will trigger an appropriate immune response of the intestinal mucosa after exposure to non-self-antigens [7]. Probiotic products can interfere with the ability of invasive species to interact with endothelial receptors, can positively impact the integrity of enterocyte tight junctions, can stimulate local mucin production (Lactobacillus species) or the production of lactic acid and hydrogen peroxide which lowers intraluminal pH [8,9]. Thus, probiotic strains not only create a hostile environment for potential pathogens, but also enhance protection of the intestinal mucosa from external aggression. Probiotic effects vary from one strain to the other. For example, Saccharomyces boulardii (S. boulardii) is one of the protease-producing probiotics which promote neutralization of toxins such as those produced by Escherichia coli (E. coli), Clostridium difficile or Vibrio cholerae [10]. Lactobacilli species, on the other hand, produce β-galactosidase, an enzyme with a proven role in preventing diarrhea and facilitating lactose digestion [11].
As ongoing research provided insights into the role of probiotics in modulating mucosal immune response and combating antigen invasion, multiple randomized controlled trials assessed the role of probiotics in decreasing severity and duration of diarrhea-related symptoms and results obtained have constituted the basis of meta-analysis publications [12,13,14]. Most of these published trials have included pediatric populations, as diarrhea represents a major health burden at young ages, constituting one of the leading causes of morbidity and mortality under the age of 5 years [15]. The strains which were most frequently analyzed were Lactobacillus rhamnosus GG (L. rhamnosus GG) and S. boulardii, which also constitute the two major probiotics universally recommended through consensus statements, alongside with L. reuteri, in the management of acute gastroenteritis in children for decreasing symptom intensity and duration [16]. European guidelines sustain the use of L. rhamnosus GG and S. boulardii for 5–7 days, as adjuvant to oral rehydration solutions in childhood acute gastroenteritis, after consistent expert agreement, whereas American guidelines sustain the use of probiotic preparations in both infectious and antimicrobial-related diarrhea in children and adults, but only based on weak and moderate evidence [16,17]. Moreover, an update on the treatment of childhood acute gastroenteritis provided in 2020 by the Working Group on Probiotics and Prebiotics of the European Society for Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) also sustained the use of a combination between L. rhamnosus and L. reuteri and recommended against the simultaneous use of L. helveticus and L. rhamnosus, as well as against the use of Bacillus clausii (B. clausii) strains [18]. Thus, in spite of multiple probiotic strains available on the market, only a few have demonstrated their superiority in decreasing symptom intensity and duration of acute gastroenteritis compared to placebo and are as a result recommended as adjuvant therapies.
The beneficial probiotic effects seem to be strain specific, not only in the setting of acute gastroenteritis, but also in the prevention of antibiotic-associated diarrhea, the management of infantile colic or the treatment of other digestive tract conditions, such as Helicobacter pylori gastritis [19,20]. However, there are few reports available regarding the pathogen-specific benefits of probiotic strains, with a particular focus given in recent years to the outcome of probiotic supplementation upon the evolution and severity of diarrheic episodes with known etiology [21,22].
This review aims to investigate the pathogen-specific role of probiotic and synbiotic supplementation in pediatric acute gastroenteritis, in light of recent literature data.

2. Methods

We searched the Pubmed, Web of Science and Google Scholar databases for randomized controlled trials and pediatric population-based studies which have evaluated efficacy of probiotics and synbiotics in influencing outcome of acute gastroenteritis. We only focused on those studies which included patients with a known infectious etiology of the diarrheic disease or which had also performed a separate analysis on subgroups in which a certain viral or bacterial causative agent was identified or on those cohorts in which rotavirus vaccination status was known. We refrained from reporting data provided by publications limited to abstract formats or by in-extenso manuscripts written in a language different from English. Search terms used included “probiotic” AND “diarrhea” AND “child”, or “probiotic” AND “gastroenteritis” AND “child” or “probiotic” AND “rotavirus” or “prebiotic” AND “diarrhea” AND “child”, or “prebiotic” AND “gastroenteritis” AND “child” or “prebiotic” AND “rotavirus” or “synbiotic” AND “diarrhea” AND “child”, or “synbiotic” AND “gastroenteritis” AND “child” or “synbiotic” AND “rotavirus”.

3. Results

We identified 32 studies which complied with our inclusion and exclusion criteria, as summarized in Table 1, Table 2, Table 3 and Table 4. Most of these studies assessed the influence of probiotics on rotaviral diarrhea, whereas a very limited number of clinical trials evaluated the impact of probiotics upon bacterial diarrhea-related symptoms and aftermath. The most intensely researched probiotics on this subject still remain L. rhamnosus, L. reuteri and S. boulardii, but a few others have also showed promising potential in combating the burden of rotaviral or bacterial diarrhea-related hospitalizations and complications. In this paper, we will provide a background on probiotics and synbiotic products and detail how these impact the outcome of childhood acute gastroenteritis with an identified etiology.

3.1. Lactobacillus rhamnosus

L. rhamnosus has been proven to be one of the probiotics that can positively influence hospital stay length, as well as diarrhea duration (Table 1) [23]. A European multicenter trial, which randomized an impressive number of children aged 1 month to 3 years of age demonstrated that a combination of oral rehydration and L. rhamnosus is superior to oral rehydration alone in terms of reducing hospitalization duration and number of watery stools after 3 days of diarrhea evolution. Recovery from watery stools was significantly more rapid in those subjects whose stool samples were positive for rotavirus infection. Bacterial etiologies were scarcely detected, not allowing for a convenient statistical analysis, but no differences were reported in terms of diarrhea duration in these cases, independently of the study group allotment [24]. Likewise, a randomized controlled trial conducted in India highlighted the benefits of L. rhamnosus in decreasing diarrhea extent and timespan of parenteral rehydration, but only in those subjects confirmed with rotavirus infection, as opposed to other etiologies [25]. Similar results were reported by Guarino et al., with a slightly more consistent reduction of diarrhea duration in children diagnosed with rotavirus, as well as of fecal viral identification after 6 days of diarrhea onset [26]. Another study, conducted in Taiwan, suggested that L. rhamnosus also aids in decreasing the extent of fecal elimination of rotavirus, in a dose-dependent manner [27]. On the other hand, increase in stool consistency and recovery from diarrheic stools took place after a shorter amount of time with L. rhamnosus administration, but independently of the presence of rotavirus fecal shredding, according to the study of Aggarwal et al. [28]. Moreover, a meta-analysis reported a better performance of L. rhamnosus in children who had not received a vaccine against rotavirus and a lack of significant difference in gastroenteritis outcome in those who were immunized [29]. In terms of bacterial infections, data are scarce; one Indian study in which a little over 60% of the stool cultures were negative, but which individually assessed the role of L. rhamnosus in relation to each particular type of pathogen identified in the stool samples, concluded that its efficacy is limited to reducing diarrhea duration in Clostridium difficile positive patients. However, this conclusion is supported by a very limited number of cases [23].
Two randomized clinical trials which analyzed the efficacy of a combined probiotic strain containing L. rhamnosus and L. helveticus reported no virus-specific benefit over placebo in terms of reducing viral load at 28 days post-enrollment, nor in decreasing intensity of clinical symptoms. Both studies included children diagnosed with gastroenteritis of viral etiologies (norovirus, rotavirus, adenovirus), as well as of bacterial etiologies [21,22]. Still, a decrease in number of diarrhea episodes caused by adenovirus infection was noted (Table 1) [22].
There are limited reports regarding the efficacy of L. rhamnosus in the absence of hospitalization. An Indian study reported a significant reduction in diarrhea episodes during a follow-up period of 4 weeks in recipients of L. rhamnosus diagnosed with rotavirus gastroenteritis, as opposed to those with Crytosporidial diarrhea, in whom administration of the same probiotic showed no benefit compared to placebo. L. rhamnosus also seemed to augment specific IgG (and not IgA) production against rotavirus, but did not influence antibody levels for Cryptosporidium species (Table 1) [30]. Another randomized controlled trial, conducted in Uganda, highlighted the benefit of both L. rhamnosus and S. boulardi in reducing diarrhea episodes only in outpatient settings in children with severe acute malnutrition, and not in those who required hospitalizations. The study suggested that both strains do not influence the outcome and evolution of severe gastroenteritis, but provided no data regarding etiology of the diarrheic manifestations [31].
Table 1. Characteristics of clinical studies which assessed overall and pathogen specific impact of L. rhamnosus supplementation in childhood diarrhea.
Table 1. Characteristics of clinical studies which assessed overall and pathogen specific impact of L. rhamnosus supplementation in childhood diarrhea.
Reference (Author, Year)Type of StudyPopulation and Study Group AssignmentType of InterventionMain OutcomePathogen Specific Benefits
Intervention/Study GroupControl GroupDuration of Treatment/Dosage
Guandalini et al., 2000 [24]Double-blind RCT287 children, with ages: 1 month–3 years
147 in intervention group
140 in placebo group
L. rhamnosus + ORS
ORSDosage: 1010 CFU/250 mL 2×/days;
up to 7 days
Reduction in duration of diarrhea and hospitalization period in the intervention group
  • Rotavirus positive patients: reduction in duration of diarrhea, in number of watery stools starting from the 3rd day of treatment and in hospitalization period in the intervention group
  • Invasive pathogens—no significant differences between intervention and control group
Szymański et al., 2006 [25]Double-blind RCT87 children with ages 2 months–6 years
49 in intervention group
44 in control group
Three L. rhamnosus strains. (573L/1, 573L/2, 573L/3) + ORS/IV rehydrationORS/IV rehydrationDosage: 1.2 × 1010 CFU 2×/days, 5 daysNo influence upon duration of diarrhea, of parenteral rehydration, nor of weight gain within the entire intervention groupRotavirus positive patients: significant reduction of diarrhea duration, especially within the first 72 h of treatment; overall reduction in duration of parenteral rehydration
Guarino et al., 1997 [26]Double-blind RCT100 children with ages: 3–36 months
52 in intervention group
48 in control group
L. rhamnosus + oral rehydration therapyOral rehydration therapyDosage: 3 × 109 CFU in 200 mL milk or formula, 2×/days, up to 5 daysSignificant reduction in diarrhea duration in the intervention group versus control group Rotavirus positive patients: slightly greater efficacy of L. rhamnosus in reducing diarrhea duration; significant reduction in viral excretion 6 days after the onset of diarrhea in the intervention group
Fang et al., 2009 [28]RCT23 patients with ages: 9–72 months
9 in the low-dose group, 8 in the high-dose group, 6 in control group
low-dose group and high-dose group—L. rhamnosus+ SimethiconeSimethicone 80 mg/dayDosage:
low-dose group—2 × 108 L. rhamnosus + Simethicone 80 mg/day
high-dose group—6 × 108 L. rhamnosus + Simethicone 80 mg/day, 3 days
  • Significant reduction in fecal rotavirus concentration in the high-dose group after 3 days of treatment
  • No significant difference in fecal rotavirus shedding in the low-dose and control group after 3 days
The study included only rotavirus positive patients
Aggarwal et al., 2014 [28]Open Label
RCT
200 children with ages: 6 months–5 years
100 in intervention group
100 control group
L. rhamnosus + Standard manage-ment (ORS/IV rehydration + zinc 20 mg/day for 14 days)Standard managementDosage: 1010 CFU once daily, 5 daysSignificant reduction in duration of diarrhea and in time to improvement in stool consistencySignificant reduction in duration of diarrhea and in time to improvement in stool consistency independently of the presence/absence of Rotavirus fecal antigen
Basu et al., 2007 [23]Double-blind RCT235 children with ages: 2–6 years
117 in intervention group
118 control group
L. rhamnosus + ORSORSDosage: 60 million cells dissolved in ORS twice daily, at least 7 days/until diarrhea stoppedSignificant reduction in duration of diarrhea and hospital stayC. difficile positive patients: Significant reduction in duration of diarrhea
Freedman et al., 2020 [21,22]Double-blind RCT816 patients with ages: 3–48 months
408 in intervention group
408 control group
L. rhamnosus RO011 + L. helveticus RO052 (95:5 ratio) + standard therapyStandard therapyDosage: 4 × 109 CFU L. rhamnosus RO011 + L. helveticus RO052(95:5 ratio) + standard therapy twice daily, 5 days No significant changes in disease severity, assessed using the modified Vesicari score [32]No significant reduction in stool viral and bacterial load
Freedman et al., 2022 [22]Two Double-blind RCT1565 patients with ages: 3–48 months
778 in intervention group
787 control group
US Group—370 with L. rhamnosus GG
Canadian Group—408 L. rhamnosus RO011 + L. helveticus RO052 (95:5 ratio) + standard therapy
Standard therapyDosage:
US Group—received only L. rhamnosus—1010 CFU, twice daily, 5 days
Canadian Group—4 × 109 CFU L. rhamnosus RO011 + L. helveticus RO052 (95:5 ratio) + standard therapy twice daily, 5 days
No significant changes in disease severity, assessed using the modified Vesicari score [32]No significant reduction in stool viral and bacterial load
Adenovirus positive patients: significant decrease in number of diarrhea episodes in those supplemented with L. rhamnosus RO011 + L. helveticus RO052
Sindhu et al., 2014 [30]Double-blind
RCT
124 children with ages: 6 months–5 years (82 with Rotavirus and 42 with Cryptosporidium
Species)
65 in intervention group
59 in control group
L. rhamnosus GG + antibiotic treatment (in case of positive stool culture)Placebo + antibiotic treatment (in case of positive stool culture)Dosage: 1010 CFU once per daily for 4 weeksNo influence of the probiotic product upon diarrhea duration, severity, fever, frequency of vomiting episodes, dehydration status or hospital stayRotavirus positive patients: significant reduction in diarrhea episodes, augmentation of specific IgG antibody production against rotavirus
Cryptosporidium positive patients: no influence upon diarrhea duration compared to placebo; no impact on specific antibody levels
Legend: CFU—colony forming units; C. difficileClostridium difficile; IV—intravenous, L. helveticus—Lactobacillus helveticus; L. rhamnosus—Lactobacillus rhamnosus; ORS—oral rehydration solution; RCT—randomized controlled trial.

3.2. Saccharomyces boulardii

S. boulardii has been regarded as the most effective agent in reducing duration of diarrhea (including the risk of an evolution of loose stools over 2 days) and is apparently superior to other strains such as B. lactis, L. rhamnosus, L. reuteri and L. paracasei, according to a meta-analysis conducted on 84 studies [33]. Moreover, its advantages over B. clausii in shortening diarrhea extent have been highlighted within a randomized trial, which also showed comparable performance of the two strains in terms of hospitalization, fever duration and number of vomiting episodes [34]. These results contradict former scientific reports such as that of Gaon et al., which claimed that S. boulardii’s efficacy is similar to the efficacy of a combination between L. casei and L. acidophilus in diminishing diarrhea and vomiting, when compared to placebo and is independent of Rotaviral etiology (Table 2) [35].
Table 2 details characteristics and results of studies which analyzed overall and pathogen-specific impact of S. boulardii in pediatric gastroenteritis. One randomized controlled trial conducted in India certified the advantages of S. boulardii over a similar product in rotavirus-associated diarrhea, without any adverse events, but only in terms of hospitalization duration. No difference from controls was seen in terms of fever duration, vomiting episodes, proportion of children requiring parenteral rehydration or with diarrhea lasting for more than one week [36]. Similarly, another study conducted by Dalgic et al. established a significant decrease in duration of hospitalization and diarrhea in children diagnosed with rotavirus infection, supplemented with both S. boulardii and zinc, as opposed to controls. Moreover, the authors also demonstrated that this adjuvant therapeutic combination can also accentuate resolution of vomiting [37]. On the other hand, a Bolivian randomized controlled trial reported that number of recurrent vomiting episodes is decreased in patients receiving an oral rehydration therapy in combination with S. boulardii, B. longum, L. rhamnosus and L. acidophilus as opposed to those which only benefited from the addition of S. boulardii in their treatment or who received placebo together with oral rehydration solutions (ORS). Still, S. boulardii alone showed a better outcome than the four-strain product and placebo in shortening the number of days with fever and diarrhea in children infected with rotavirus [38]. Apparently, a three-day treatment with S. boulardii performed even better in inhibiting release of watery stools in pre-school children who tested positive for rotavirus, unlike in those who were negative for the virus [39].
Persistent, prolonged elimination of rotavirus in feces has been reported even in vaccinated children and represents a source of community-acquired infection [40]. S. boulardi might be a potential inhibitor of rotavirus fecal shedding, with absence of fecal virus detection after 5 days of administration, as suggested by a multicenter, case-control clinical trial. This conclusion was, however, supported by a very limited number of cases [41].
Table 2. Characteristics of clinical studies which assessed overall and pathogen specific impact of S. boulardii supplementation in childhood diarrhea.
Table 2. Characteristics of clinical studies which assessed overall and pathogen specific impact of S. boulardii supplementation in childhood diarrhea.
Reference (Author, Year)Type of StudyPopulation and Study Group AssignmentType of InterventionMain OutcomePathogen Specific Benefits
Intervention/Study GroupControl GroupDuration of Treatment/Dosage
Gaón et al., 2003 [35]Double-blind
RCT
89 children with ages: 6–24 months
29 in control group (group 1).
30 in S. boulardii (group 2)
30 in Lactobacilli group (group 3)
Group 2: S. boulardii
Group 3: Pasteurized cow milk with L. casei
and L. acidophilus
Group 1: Pasteurized cow milk + ORS Dosage:
S. boulardii group 1010 CFU/g twice daily for 5 days
Group with: Pasteurized cow milk with L. casei
and L. acidophilus: 1010–1012 CFU/g twice daily for 5 days
Similar significant reduction in vomiting, diarrhea duration and number of stools in both study groupsNo specific benefits in relation to the presence/absence of rotavirus fecal antigen
Das et al., 2016 [36]Double-blind
RCT
60 children with ages: 3 months–5 years
30 in intervention group
30 in control group
S. boulardiiPlaceboDosage: 250 mg 2×/day for 5 daysSignificant reduction in diarrhea duration and hospitalization period
No influence on fever and vomiting duration nor on proportion of children who required parenteral rehydration or presented diarrhea lasting for >7 days
The study included only rotavirus positive patients
Dalgic et al., 2011 [37]Single
Blind
RCT
480 children with ages: 1–28 months, divided into 7 intervention groups and 1 control group (60 patients included in each group)Group 1: S. boulardii
Group 2: zinc (Zn)
Group 3: lactose-free formula (LF)
Group 4: S. boulardii + Zn
Group 5: S. boulardii + LF
Group 6: Zn + LF,
Group 7: Zn + LF + S. boulardii
+
ORS and/or parenteral rehydration
Group 8 (control group): −ORS and/or parenteral rehydrationDosage:
S. boulardii—250 mg once daily 5 days
Zn—10 mg 2×/day in infants < 6 months; 20 mg 2×/day in infants/children > 6 months
Statistically significant decrease in
diarrhea duration and hospitalization time in groups 2 and 4 versus controls
The study included only rotavirus positive patients
Grandy et al., 2010 [39]Double-blind
RCT
64 hospitalized children with ages: 1–23 months divided in 3 groups
Group GC (control): 20 children
Group GB: 21 children Group GARLB: 23 children
Group GB: S. boulardii + ORS
Group GARLB: combined probiotic product (L. acidophilus, L. rhamnosus, B. longum and S. boulardii)
PlaceboDosage:
For S. boulardii—4 × 1010 lyophilized cells/dose 2×/day for 5 days
For combined probiotic product—(L. acidophilus 6.625 × 107 lyophilized cells/dose, L. rhamnosus 3.625 × 107 lyophilized cells/dose, B. longum 8.75 × 106 lyophilized cells/dose and S. boulardii) 1.375 × 107 lyophilized cells/dose) 2×/day for 5 days
Statistically significant reduction in diarrhea duration in both study groups versus control; S. boulardii alone performed better than the combined product in this regard
Significant reduction in diarrhea and vomiting duration when comparing the merged intervention groups versus placebo
The study included only rotavirus positive patients
Corrêa et al., 2011 [39]Double-blind
RCT
176 patients with ages: 6–48 months
90 in intervention group
86 in control group
S. boulardiiPlacebo—excipientsDosage: 4 × 109 viable cells 2×/day for 5 daysStatistically significant decrease in frequency and duration of diarrhea if S. boulardii was administered during the first three days of gastroenteritis evolutionRotavirus positive patients—statistically significant decrease in frequency of diarrhea after 3 days of intervention
Mourey et al., 2020 [41]Double-blind
RCT
100 children with ages: 3–36 months
49 in intervention group
51 in control group
S. boulardii + ORS + ZnPlacebo + ORS + ZnDosage: S. boulardii 5 × 109 living cells 2×/day for 5 days
Zn—10 mg 1×/day in infants < 1 year; 20 mg 1×/day in children > 1 year
Significant increase in stool consistency starting from day three of intervention
Significant decrease in time of recovery from diarrhea in the intervention group
Rotavirus positive patients: absence of fecal virus detection after 5 days of treatment in old patients belonging to the intervention group
Legend: CFU—colony forming units; L. acidophilusLactobacillus acidophilus; L. caseiLactobacillus casei; LF—lactose-free formula; ORS—oral rehydration solution; RCT—randomized controlled trial; S. boulardiiSaccharomyces boulardii; Zn—zinc.

3.3. Lactobacillus acidophilus

L. acidophilus has been one of the bacteria widely used alone or as part of combined products for the treatment of acute diarrhea in children, in spite of insufficient evidence available [2]. The suppositions surrounding the potential beneficial effects of this strain emerged from evidence supporting its involvement in inhibition of bacterial invasion of epithelial intestinal cells, a consistent characteristic of other Lactobacilli as well [42]. L. acidophilus can also augment specific immune reactions and is stable at high temperatures, so it is unsurprising that several compounds appeared on international markets, which claimed to deliver relief of gastroenteritis-associated symptoms [43]. However, one study and a meta-analysis reported no benefit regarding its complementary use in those pediatric gastroenteritis cases in which an etiological factor was identified from the analyzed stool specimens [44,45]. In similar fashion, one randomized controlled trial reported no clear advantage of L. acidophilus over placebo in cases where rotavirus or norovirus was identified, neither in terms of reducing symptom duration, nor in terms of diminishing stool viral load [46]. Some evidence emerged regarding the advantages of L. acidophilus in shortening hospitalization duration or number of days with fever, but only in patients with unknown etiology of the diarrhea or in whom a rotavirus infection was excluded, as detailed in Table 3 [44,47,48]. Salazar-Lindo et al. also reported a lower number of stools in those children supplemented with L. acidophilus suffering from mild diarrhea, but in their study cohort an infectious agent (namely rotavirus) was identified in only a very limited number of patients [49]. Given the inconsistent results reported by various studies, a meta-analysis conducted by authors belonging to the Working Group on Probiotics and Prebiotics of the ESPGHAN concluded that there is insufficient evidence available to support the administration of L. acidophilus in childhood acute gastroenteritis [50].

3.4. Lactobacillus reuteri

L. reuteri strains are able to modulate lipopolysaccharide-induced intestinal inflammation and reduce mucosal damage caused by enteric infections in rats [51]. Both anti-bacterial and anti-inflammatory effects have been associated with their use, as demonstrated in a murine model in which the administration of L. reuteri antagonized the extension of enteral mucosa inflammation and C. difficile infection [52]. Moreover, two in vitro studies also confirmed L. reuteri’s potential in combating E. coli growth [53], modulating immune response against Salmonella typhimurium [54] and another study conducted on rats related its use with the development of mucosal rotavirus-specific IgA production [55]. A randomized controlled trial conducted in pre-school children asserted the protective effect of L. reuteri against infectious diarrhea, which lasted through a follow-up period of 3 months after previous administration of the product for the same amount of time (Table 3) [56].
Shornikova et al. reported amelioration of diarrhea in a small pediatric cohort which received L. reuteri, as opposed to placebo, with most of the patients belonging to the intention-to-treat group not experiencing any watery stools after two days of probiotic supplementation. Given that rotavirus infection was identified in 75% of the study population, the authors proposed L. reuteri as a potential therapeutic adjuvant in rotavirus enteritis. However, the study failed to identify any significant increase in specific rotavirus IgA antibody titer in the L. reuteri recipient group [57]. Another research study by these authors, performed on a rotavirus cohort in children aged between 6 and 36 months, confirmed the significant reduction in the duration of watery diarrhea associated with L. reuteri administration, as well as a good restoration of the enteral microbiota in this setting (Table 3) [58].
A Botswanian pediatric randomized controlled trial which included subjects vaccinated against rotavirus found no significant association between the administration of L. reuteri and acute gastroenteritis outcome, whether viral or bacterial. No notable replication inhibition of bacteria such as Shigella, Campylobacter, Enteropathogenic E. coli or Cryptosporidium was reported, but the study did not separately evaluate the subpopulation in which enteric viral infections were found in feces samples, as most of those children also presented another viral/bacterial co-infection [59]. Moreover, a Polish study found no benefit of L. reuteri on diarrhea-related outcomes, regardless of rotavirus vaccination status, but the infectious etiology was unknown in most of the patients enrolled [60].

3.5. Lactobacillus plantarum

Lactic acid bacteria have the ability to attach to cellular monolayers, exerting protection against rotavirus, as well as against the transmissible gastroenteritis virus (TGEV), according to Maragkoudakis et al. [61]. Out of all the Lactobacillus species studied, L. plantarum had the highest attachment ability and was found to have an antiviral effect against both viruses, as proven by the same experimental study [61]. Another study performed on mice proved that exopolysaccharides released by L. plantarum effectively reduce the number of diarrheic episodes and rotaviral shedding [62]. The decrease in rotavirus load in subjects supplemented with a L. plantarum product was later confirmed by research conducted in children, which also showed a significant decrease in diarrhea duration and in its severity (assessed through daily changes in Vesikari score—Table 3 [32]) [63].
Table 3. Characteristics of clinical studies which assessed overall and pathogen specific impact of probiotic L. acidophilus, L. reuterii and L. plantarum in childhood diarrhea.
Table 3. Characteristics of clinical studies which assessed overall and pathogen specific impact of probiotic L. acidophilus, L. reuterii and L. plantarum in childhood diarrhea.
Reference (Author, Year)Type of StudyPopulation and Study Group AssignmentType of InterventionMain OutcomePathogen Specific Benefits
Intervention/Study GroupControl GroupDuration of Treatment/Dosage
Pinto et al., 2016, 2016 [44,47]Retrospective cohort study290 children with ages: 6–60 months
65 in study group
225 in control group
L. acidophilus mixture (80% L. acidophilus, 10% L. bulgaricus, 5% B. bifidum, 5% S. thermophilusStandard treatmentDosage: 1 × 109 CFU/g 2×/day (no information regarding duration of treatment)Significant decrease in diarrhea duration of IV rehydration among the study group
No influence of the probiotic supplementation upon length of hospital stay
Patients with positive stool studies (positive stool culture, positive stool for C. difficile toxin, positive rotavirus antigen)—no benefits from probiotic use as opposed to those with negative stool studies
Khanna et al., 2005 [46]Double-blind
RCT
98 children with ages: 6 months–12 years
48 in intervention group
50 in control group
L. acidophilus + ORSORSDosage: 1.5 × 1010 tyndalized cells 1×/day for 3 daysNo significant benefit of probiotic products in reducing mean diarrhea duration, mean ORS requirement, mean rehydration period and duration of hospital stayRotavirus positive patients—no beneficial effect in the intervention group
Hong Chau et al., 2018 [46]Double-blind
RCT
290 children with ages: 9–60 months
143 in intervention group
147 in control group
L. acidophilus + ORS + Zn + antimicrobials were neededPlacebo + ORS + Zn + antimicrobials were neededDosage: 1 × 108 CFU cells 2×/day for 5 daysNo significant reduction in diarrhea and hospitalization duration, nor in stool frequency Rotavirus/Norovirus positive patients—no significant decrease in stool load and no influence of intervention upon symptom duration
Shornikova et al., 1997 [57]Double-blind
RCT
40 children with ages: 6–36 months
19 in intervention group
21 in control group
L. reuteri + ORSPlacebo + ORSDosage: 1010–1011 CFU/g 1×/day for 5 days/hospitalization period (if shorter than 5 days)Shorter mean duration of diarrhea during hospital stay in the intervention group
No influence on dehydration correction and electrolyte levels of the probiotic supplementation
Significant reduction in vomiting after the second day of treatment in the intervention group
No influence of the intervention treatment upon rotavirus IgA antibody production
Shornikova et al., 1997 [58]Double-blind
RCT
66 children with ages: 6–36 months
20 in group 1
21 in group 2
25 in control group
Group 1: L. reuteri—small dose + ORS
Group 2: L. reuteri—large dose + ORS
Placebo + ORSDosage:
Small dose: 2.5 × 106–5 × 106 CFU/mL 1×/day for a maximum of 5 days
Large dose: 5 × 108–2.5 × 109 CFU/mL 1×/day for 5 days
Large dose of L. reuteri yielded a significant reduction in duration and frequency of diarrhea after 2 days of treatment
Good restoration of enteral microbiota in both intervention groups
The study included only rotavirus positive patients
No influence of L. reuteri upon rotavirus IgA and IgG antibody titers
Pernica et al., 2022 [59]Multicentric double-blind
RCT
272 children with ages: 2–60 months
135 in intervention group
137 in control group
L. reuteri + ORS/parenteral IV rehydration + antibiotic treatment (were needed) Placebo + ORS/parenteral IV rehydration + antibiotic treatment (were needed)Dosage: 1 × 108 CFU/g 1×/day for 60 daysNo significant impact of the probiotic strain upon recurrence of diarrhea, length of hospital stay, developmental of sepsis, 7-day and 60-day mortality rate during the follow-up period of 60 daysPatients with positive stool culture for Shigella, Campylobacter, enteropathogenic E. Coli or Cryptosporidium: no notable replication inhibition of the identified bacteria
Szymański et al., 2019 [60]Double-blind
RCT
91 children with ages: <5 years
44 in intervention group
47 in control group
L. reuteri + standard rehydration therapyPlacebo + standard rehydration therapyDosage: 2 × 108 CFU
1×/day for 5 days
Significant decrease in duration of hospitalization in the intervention group
No influence of intervention therapy upon diarrhea duration, frequency and recurrence, duration of IV rehydration, nor upon improvement of Vesikari scale
Children not vaccinated against rotavirus—significant reduction in hospital stay in the intervention group; no influence of intervention therapy upon diarrhea duration
Children vaccinated against rotavirus—similar primary and secondary outcome in both groups
Shin et al., 2020 [63]RCT50 children with ages: 1–69 months
15 in group 1
8 in group 2
27 in group 3
Group I: L. plantarum LRCC5310
Group 3: Saccharomyces
species
Group 2: Placebo + standard
treatment
Dosage: not specifiedSignificant decrease in stool frequency and diarrhea duration between group I and placebo in day 3 of hospitalization
Significant decrease in diarrhea duration and in changes of Vesikari scale when comparing merged intervention groups with placebo
The study included only rotavirus positive patients
Significant reduction in rotavirus fecal titer in group 1 versus placebo
Legend: CFU—colony forming units; IV—intravenous, L. acidophilusLactobacillus acidophilus; L. plantarumLactobacillus plantarum; L. reuteriLactobacillus reuteri; ORS—oral rehydration solution; RCT—randomized controlled trial; S. thermophilusStreptococus thermophilus; Zn—zinc.

3.6. Combinations of Multiple Probiotic Strains

Table 4 provides insights regarding the specific beneficial use of combined probiotic products in childhood diarrhea. A double-blind randomized controlled trial reported effective reduction of diarrhea duration in children who were administered twice daily a combined suspension, consisting of 6 different strains: B. longum, B. lactis, L. acidophilus, L. rhamnosus, L. plantarum and Pediococcus pentosaceus. The study also established that each individual component of the combined aforementioned suspension can inhibit in vitro rotavirus infection, without cytotoxic effects on the population of cells analyzed [64]. A B. longum and L. acidophilus mixture can also significantly shorten diarrhea duration, according to a Korean study [65]. Another similar study demonstrated significant reduction in hospitalization duration in children infected with rotavirus who received a mixed probiotic preparation consisting of L. acidophilus, L. rhamnosus, B. longum and S. boulardii for at least 5 days as opposed to oral/parenteral rehydration alone [66]. A preparation containing Bacillus mesentericus, Clostridium butyricum and Enterococcus faecalis also seems to effectively decrease diarrhea severity in combination with symptomatic and rehydration therapy after 3 days of daily two-dose administration in patients infected with rotavirus, but not in those infected with Salmonella [67]. Similarly, the administration of a suspension containing L. casei, L. rhamnosus, Streptococcus thermophilus (S. thermophilus), B. breve, L. acidophilus, L. bulgaricus and B. infantis twice daily for a minimum of 5 days reduced the number of hospitalization days related to rotaviral diarrhea [68]. A combination of 8 probiotic strains (L. acidophilus, L. paracasei, L. bulgaricus, L. plantarum, Bifidobacterium breve, Bifidobacterium infantis, Bifidobacterium longum and S. thermophilus) has also been proposed as an adjuvant therapeutic agent in rotavirus diarrhea, as it promoted a shorter recovery time and significantly decreased the requirement of oral rehydration therapy and stool volume loss after just two days of administration [69]. Furthermore, the simultaneous administration of L. rhamnosus and L. reuteri supposedly reduces the detection rate of rotavirus fecal antigen after 5 days of treatment, which suggests that these two probiotics positively interfere with viral elimination time. Still, this evidence is supported by a study with very limited follow-up time [70].

3.7. Synbiotic Products

Prebiotic supplements in the form of pectin-derived acidic oligosaccharides, short-chain galacto-oligosaccharides, long-chain oligosaccharides added to a fermented milk concentrate have been proven to enhance protection against rotavirus infection in suckling rats, through a presumed interaction with viral particle attachment to the enterocyte, which consequently inhibits viral replication [71,72]. Synbiotic products, containing both prebiotics and probiotics seem to also stimulate immune response against enteric viruses through modulation of the interferon signaling pathway [73]. Hence, synbiotics have been proposed as additional products to classical therapy in severe rotavirus enteritis by a review mainly based on in vitro studies [74]. The authors acknowledged, however, that the paucity of randomized controlled trials mandated future research investigating the preventive and therapeutic benefits of synbiotics in rotavirus diarrhea [74].
Evaluation of etiology-based performance of synbiotics in the management of diarrhea in children has been so far based on two randomized-controlled trials, one of them including only patients in whom stool samples were positive for rotavirus and the other one conducting a separate analysis on children with diarrhea triggered by the same etiological agent [75,76]. As detailed in Table 4, Dewi et al. proved that a combination of L. casei, L. rhamnosus, S. thermophilus, B. breve, L. acidophilus, B. infantis and L. bulgaricus and fructo-oligosaccharides (FOS) can aid in decreasing diarrhea duration in children with rotaviral gastroenteritis, whereas Islek et al. reported the augmentation of the same shortening in diarrhea timespan in the study subgroup with rotavirus infection in which B. lactis together with inulin were administered, as opposed to children infected with adenovirus or Entamoeba histolytica, in whom the synbiotic produced no significant difference [75,76]. Another three studies have also supported products consisting of multiple probiotic strains and FOS as complementary agents to standard treatment that can yield a shorter recovery of stool consistency and overall decrease in duration of diarrhea [77,78,79]. Moreover, L. paracasei combined with arabinogalactan and xylo-oligosaccharides can improve stool output frequency, positively impact hospital stay and decrease parental working days absences, according to an Australian study [80]. However, all these four studies have provided no information regarding diarrhea etiology for any of the pediatric patients enrolled [77,78,79,80].
Table 4. Characteristics of clinical studies which assessed overall and pathogen specific impact of combined probiotic strains and synbiotic supplementation in childhood diarrhea.
Table 4. Characteristics of clinical studies which assessed overall and pathogen specific impact of combined probiotic strains and synbiotic supplementation in childhood diarrhea.
Reference (Author, Year)Type of StudyPopulation and Study Group AssignmentType of InterventionMain OutcomePathogen Specific Benefits
Intervention/Study GroupControl GroupDuration of Treatment/Dosage
Lee et al., 2015 [64]Double-blind
RCT
29 children with ages: 3 months–7 years
13 in intervention group
16 in control group
Combined product: B. longum, B. lactis, L. acidophilus, L. rhamnosus, L. plantarum and Pediococcus pentosaceusPlaceboDosage: 109 CFU/g (108 CFU/each strain) 1×/day for 1 weekSignificant decrease in diarrhea duration after 7 days of treatment
No significant difference in abdominal pain, vomiting, fever duration, nor in paraclinical data between the two groups at the end of treatment
Rotavirus positive patients—significant decrease in diarrhea and vomiting duration after 7 days of treatment; no significant difference in abdominal pain, fever duration, nor in paraclinical data between the two groups at the end of treatment
Park et al., 2017 [65]Double-blind
RCT
57 children with ages: 9–16 months
28 in intervention group
29 in control group
Combined product: B. longum, L. acidophilusPlaceboDosage: 2.2 × 109 CFU/g of probiotic mixture of B. longum 2 × 1010 CFU/g and L. acidophilus 2 × 109 CFU/g
2×/day for 3 days
Significant reduction in diarrhea duration in the intervention group
No impact of the combined product upon fever duration, diarrhea and vomiting frequency
The study included only rotavirus positive patients
Teran et al., 2009 [66]Single-blind RCT75 children with ages: 28 days–24 months
25 in group 1 (nitazoxanide group)
25 in group 2 (probiotic group)
25 in control group
Group 1: Nitazoxanide
Group 2: combined product—L. acidophilus, L. rhamnosus, B. longum and S. boulardii
Standard
treatment
Dosage:
Nitazoxanide—15 mg/kg/day divided into 2 doses for 3 days
Combined product—1.25 × 108 CFU/g
2×/day for 5 days
Significant reduction in duration of diarrhea and hospitalization period in both intervention groups versus placeboThe study included only rotavirus positive patients
Huang et al., 2014 [67]Open Label
RCT
159 children with ages: 3 months–14 years
82 in intervention group
77 in control group
Combined product: E. faecalis, C. butyricum, B. mesentericusSuportive treatmentDosage: a tablet of the combined probiotic product contains: 3.48 × 108 CFU of a mixture of E. faecalis (3.17 × 108 CFU), C. butyricum (2 × 107 CFU) and B.mesentericus (1.1 × 107 CFU)
children < 6 years—2 × 1 tabl/day
children 6–12 years—2 × 2 tabl/day
children > 12 years—2 × 3 tabl/day
for 7 days
Significant reduction in diarrhea duration and Vesikari score in the intervention group
No influence of intervention therapy upon mild stool frequency and hospitalization duration
Rotavirus positive patients: significant reduction in diarrhea severity after 3 days of probiotic treatment
Salmonella positive patients: no significant reduction in diarrhea severity in patients belonging to intervention group
Sobouti et al., 2016 [68]Single-blind RCT60 patients with ages: 3 months–7 years
32 in intervention group
28 in control group
Combined product: L. casei, L. rhamnosus, S. termophilus, B. breve, L. acidophilus, L. bulgaricus, B. infantis + ORS/IV rehydration therapyORS/IV rehydration therapyDosage: 1 × 109 CFU/g 2×/day for 5 daysSignificant decrease in hospitalization duration in the intervention group
No association was found between probiotic treatment and degree of dehydration
The study included only rotavirus positive patients
Dubey et al., 2008 [69]Double-blind
RCT
224 children with ages: 6 months–2 years
113 in intervention group
111 in control group
Combined product: L. acidophilus, L. paracasei, L. bulgaricus, L. plantarum, B. breve, B. infantis, B. longum, S. thermophilus + ORS/IV rehydration therapyPlacebo + ORS/IV rehydration therapyDosage: each sachet of the combined probiotic product contains 9 × 1010 CFU/g
children < 5 kg—2 sachets/day for 4 days
children—5–10 kg—4 sachets/day for 4 days
Significant reduction in recovery time in the intervention group
Significant decrease in stool frequency starting from the second day of treatment
No significant difference between the two groups in term of volume of ORS/parenteral solutions administered
The study included only rotavirus positive patients
Rosenfeldt et al., 2002 [70]Double-blind
RCT
69 children with ages: 6–36 months
30 in intervention group
39 in control group
Combined product: L. rhamnosus, L. reuteri + ORS/IV rehydration therapyPlacebo + ORS/IV rehydration therapyDosage: 2.2 × 1010 CFU of a mixture of L. rhamnosus (1.7 × 1010 CFU), L. reuteri (0.5 × 1010 CFU) 2×/day for 5 daysSignificant decrease in hospital stay duration, duration of diarrhea after start of treatment and in reduction of loose stool frequency on the 5th day of follow-up in intervention versus control group
No significant difference between the two groups in term of fever duration
Significant reduction in fecal rotavirus antigen positivity rate on day 5 among the intervention group
Dewi et al., 2015 [75]Double-blind RCT69 children with ages: 6–59 months
34 in intervention group
35 in control group
Synbiotic product: L. casei, L. rhamnosus, S. thermophilus, B. breve, L. acidophilus, B. infantis, L. bulgaricus, FOS + ORS/IV rehydration therapy + ZnPlacebo + ORS/IV rehydration therapy + ZnDosage: a sachet of the synbiotic product contains 1 × 109 CFU/dose of a mixture of L. casei (4 × 108 CFU), L. rhamnosus (3.5 × 108 CFU), S. thermophilus (1 × 108 CFU), B. breve (5 × 107 CFU), L. acidophilus (5 × 107 CFU), B. infantis (4 × 107 CFU), L. bulgaricus (1 × 107 CFU) and FOS (990 mg) 1×/day for 5 daysSignificant decrease in diarrhea recovery time
No impact upon hospital stay
The study included only rotavirus positive patients
Islek et al., 2014 [76]Double-blind RCT156 children with ages: 2–60 months
79 in intervention group
77 in control group
Synbiotic product: B. lactis, inulin + standard treatmentsPlacebo + standard treatmentDosage: a sachet of the synbiotic product contains a mixture of B. lactis (5 × 1010 CFU) and inulin (900 mg)
1×/day for 5 days
Significant decrease in diarrhea duration and stool frequency, especially in those who started the intervention treatment within the first 24 h of symptom onset
No impact on vomiting nor on fever duration
Rotavirus positive patients: the shortest diarrhea duration among those patients belonging to the intervention group
Adenovirus positive patients: no influence on symptom duration
E. histolytica: no influence on symptom duration
Legend: B. bifidumBifidobacterium bifidum; B. breveBifidobacterium breve; B. lactisBifidobacterium lactis; B. infantisBifidobacterium infantis; B. longumBifidobacterium longum; B. mesentericusBacilus mesentericus; CFU—colony forming units; C. butyricumClostridium butyricum; C. difficileClostridium difficile; E. ColiEscherichia Coli; E. faecalisEnterococcus faecalis; E. histolyticaEntamoeba histolytica; FOS—fructo-oligosaccharides: IV—intravenous, L. acidophilusLactobacillus acidophilus; L. bulgaricusLactobacillus bulgaricus; L. caseiLactobacillus casei; L. helveticusLactobacillus helveticus; L. paracaseiLactobacillus paracasei; L. plantarumLactobacillus plantarum; L. rhamnosusLactobacillus rhamnosus; L. reuteriLactobacillus reuteri; LF—lactose-free formula; ORS—oral rehydration solution; RCT—randomized controlled trial; S. boulardiiSaccharomyces boulardii; S. thermophilusStreptococus thermophilus; Zn—zinc.

4. Discussion

The latest position paper of the Working Group on Probiotics and Prebiotics of the ESPGHAN, published in 2022, provides weak recommendations for the use of L. rhamnosus and S. boulardii in the management of acute pediatric gastroenteritis, based on low evidence [68]. An even lower level of evidence sustained the administration of L. reuteri for the same purpose, whereas a recommendation was issued against the use of Bacillus clausii and against the combination of L. reuteri and L. helveticus in childhood diarrhea, similarly to their previous publication [18,81]. The authors have provided no specific advice for diarrhea with an established etiology and have also mentioned the potential impact of rotavirus vaccine coverage in influencing their analysis of previously published data [81].
This review tried to add novelty to the existing literature data by assessing the impact of probiotic and synbiotic use upon symptom duration, severity and immune response in both viral and bacterial gastroenteritis in children. Most of the studies conducted so far which assessed the impact of probiotic use upon childhood enteric infections have focused solely on rotavirus diarrhea and provided the basis for two previous meta-analyses [82,83], as rotavirus still remains the leading cause of diarrhea-associated complications, especially in underdeveloped countries [84,85]. However, norovirus seems to overcome rotavirus as the etiological diarrheic agent which most commonly requires medical care in children, in those countries with high anti-rotavirus vaccination coverage, such as the United States [86]. Several theories have emerged regarding the possible inhibition of replication exerted by probiotics upon enteric viruses, and probiotic supplementation has been recommended for prevention of rotaviral diarrhea, as well as for diminishing symptom duration [82,85]. As previously presented and detailed in Table 1, Table 2, Table 3 and Table 4, several strains have proven their benefits in rotavirus-associated diarrhea, including individual strains, as well as combined probiotic preparations. The majority of these studies involved supplementation with either L. rhamnosus or S. boulardii, and combined suspensions included at least one of these two strains, which still represent the most studied and validated probiotics in the pediatric literature [18,87]. One study which analyzed a combined probiotic product was omitted from Table 4, due to the lack of information regarding the exact content of the product used, as only a commercial name was provided in the manuscript abstract and main text [88]. However, very few studies separately analyzed the impact of probiotic use upon recovery from bacterial diarrhea [23,30,59]. This might be explained by the small study populations included within randomized controlled trials, and the even more size-limited sub-cohorts in which a bacterial etiology was identified. Experimental research studies have suggested that probiotics such as S. boulardii and several Lactobacillus strains show antimicrobial effects, but with limited sensitivity against the analyzed pathogens [89,90].
On the other hand, several studies have evaluated the impact of probiotic strains such as L. reuteri or of a product consisting of B. lactis, L. rhamnosus, and L. acidophilus on diarrhea severity and duration, in spite of not reporting any information regarding the source of infection, which might have greatly influenced the analyzed outcome parameters [91,92,93]. In similar fashion, one Indian study asserted that S. boulardii reduces time until resolution of watery stools and duration of diarrhea after 5 days of treatment, but conducted no separate etiology-based evaluation, due to the very limited number of cases in which the source of diarrhea (rotavirus/Vibrio cholerae) was known [94]. Synbiotics containing FOS or inulin might also positively influence childhood gastroenteritis outcome, but there is insufficient data to assess the true impact of their use in particular infectious settings [95,96]. Consequently, the most recently published Cochrane systematic review underlines that there is insufficient data to allow pathogen-specific analysis of the role of probiotic supplementation in acute gastroenteritis, due to the paucity of the studies which have identified etiological agents of diarrhea or have conducted a separate statistical evaluation, focused on a certain pathogen [97]. Furthermore, the question of adequate, safety/efficacy balanced dosage of even the most widely used strains still remains into place and experts in the field suggest conducting trials comparing administration of different dosages of the same/various strains [98].
It is also worth mentioning that most of the studies conducted so far on the matter of probiotic efficacy in acute pediatric gastroenteritis included hospitalized children, and a very limited number of them were performed in outpatient settings or included follow-up of patients’ outcome after discharge from the hospital [30,31]. A decrease in the duration of diarrhea in relation to L. reuteri use was reported within a Turkish study performed in an ambulatory care unit [92]. Still, as represented in Table 1, Table 2, Table 3 and Table 4, most of the studies which evaluated pathogen-particular benefits of probiotics have only evaluated their efficacy after 5–7 days of administration.
Recent literature data supports the administration of probiotics also as a prophylactic measure against several enteric infections. Two studies conducted on a murine model showed that S. boulardii can effectively inhibit tissue inflammation and bacterial translocation, which provide protection against Salmonella [99,100]. Prevention of rotavirus gastroenteritis seems on the other hand to be enhanced by L. rhamnosus administration, especially in hospitalized children [101], as well as by supplemented formula feeding, containing strains such as Bifidobacterium animalis subspecies lactis BB12 alone/in combination with S. thermophilus [102]. L. acidophilus also enhanced protection against rotavirus, according to a meta-analysis conducted by Di et al. [103]. L. reuteri on the other hand has been regarded as a possible prophylactic agent against diarrhea in pre-school children [104], helping maintain the integrity of the intestinal mucosa epithelial barrier, offering protection against its disruption, often caused by enterotoxigenic E. coli infection [105]. Moreover, L. plantarum interferes with binding of enteropathogenic E. coli to the intestinal epithelial cells [106]. The results obtained among in vitro studies still require confirmation among large-cohort studies and as Depoorter et al. suggest in their review, there is overall insufficient proof to routinely recommend probiotics for prevention of acute gastroenteritis in children [107].

5. Conclusions

Rotavirus infection remains the most frequently evaluated etiology in relation to probiotic efficacy, with multiple studies proposing a decrease in its related diarrhea and hospitalization duration with widely used probiotics such as S. boulardii, L. reuteri and L. rhamnosus, as well as synbiotic products containing at least one of these three strains, as complementary treatment to supportive therapy. There is, however, insufficient data on the impact of probiotics in relation to rotavirus immunization status or their influence on the diarrhea episodes treated in outpatient settings. Clinical reports are scarce regarding potential probiotic benefit in bacterial diarrhea, but some strains have been shown to inhibit in vitro growth of certain microbial pathogens. Future studies, conducted on larger cohorts, are warranted to assess the individual and additive effects of probiotic strains on outcome of pediatric gastroenteritis with identified causative agents.

Author Contributions

M.O.S. and C.O.M. conceptualized and designed the study, conducted literature search and drafted the initial manuscript. M.O.S., C.O.M. and L.E.M. reviewed and revised the manuscript. H.A. participated in collecting literature data and helped in drafting the manuscript tables. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bernaola Aponte, G.; Bada Mancilla, C.A.; Carreazo, N.Y.; Rojas Galarza, R.A. Probiotics for Treating Persistent Diarrhoea in Children. Cochrane Database Syst. Rev. 2013, 2013, CD007401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Thomas, D.W.; Greer, F.R.; American Academy of Pediatrics Committee on Nutrition; American Academy of Pediatrics Section on Gastroenterology, Hepatology and Nutrition. Probiotics and Prebiotics in Pediatrics. Pediatrics 2010, 126, 1217–1231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kechagia, M.; Basoulis, D.; Konstantopoulou, S.; Dimitriadi, D.; Gyftopoulou, K.; Skarmoutsou, N.; Fakiri, E.M. Health Benefits of Probiotics: A Review. ISRN Nutr. 2013, 2013, 481651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Tissier, H. Traitement des Infections Intestinales par la Méthode de Transformation de la Flore Bactérienne de l’Intestin. Compt. Rend. Soc. Biol. 1906, 60, 359–361. [Google Scholar]
  5. Salminen, S.J.; Gueimonde, M.; Isolauri, E. Probiotics That Modify Disease Risk. J. Nutr. 2005, 135, 1294–1298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Pineiro, M.; Stanton, C. Probiotic Bacteria: Legislative Framework—Requirements to Evidence Basis. J. Nutr. 2007, 137, 850S–853S. [Google Scholar] [CrossRef] [Green Version]
  7. Weng, M.; Walker, W.A. Bacterial Colonization, Probiotics, and Clinical Disease. J. Pediatr. 2006, 149, S107–S114. [Google Scholar] [CrossRef]
  8. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. 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. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
  9. Anirban, M.; Puneet, S. Probiotics for Diarrhea in Children. J. Med. Res. Innov. 2017, 1, AV5–AV12. [Google Scholar] [CrossRef] [Green Version]
  10. Castagliuolo, I.; Riegler, M.F.; Valenick, L.; LaMont, J.T.; Pothoulakis, C. Saccharomyces boulardii Protease Inhibits the Effects of Clostridium Difficile Toxins A and B in Human Colonic Mucosa. Infect. Immun. 1999, 67, 302–307. [Google Scholar] [CrossRef] [Green Version]
  11. De Vrese, M.; Stegelmann, A.; Richter, B.; Fenselau, S.; Laue, C.; Schrezenmeir, J. Probiotics—Compensation for Lactase Insufficiency. Am. J. Clin. Nutr. 2001, 73, 421S–429S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Do Carmo, M.S.; Santos, C.I.D.; Araújo, M.C.; Girón, J.A.; Fernandes, E.S.; Monteiro-Neto, V. Probiotics, Mechanisms of Action, and Clinical Perspectives for Diarrhea Management in Children. Food Funct. 2018, 9, 5074–5095. [Google Scholar] [CrossRef] [PubMed]
  13. Yang, B.; Lu, P.; Li, M.-X.; Cai, X.-L.; Xiong, W.-Y.; Hou, H.-J.; Ha, X.-Q. A Meta-Analysis of the Effects of Probiotics and Synbiotics in Children with Acute Diarrhea. Medicine 2019, 98, e16618. [Google Scholar] [CrossRef] [Green Version]
  14. Wang, F.; Zhao, T.; Wang, W.; Dai, Q.; Ma, X. Meta-Analysis of the Efficacy of Probiotics to Treat Diarrhea. Medicine 2022, 101, e30880. [Google Scholar] [CrossRef] [PubMed]
  15. Lanata, C.F.; Fischer-Walker, C.L.; Olascoaga, A.C.; Torres, C.X.; Aryee, M.J.; Black, R.E. Child Health Epidemiology Reference Group of the World Health Organization and UNICEF Global Causes of Diarrheal Disease Mortality in Children < 5 Years of Age: A Systematic Review. PLoS ONE 2013, 8, e72788. [Google Scholar] [CrossRef] [Green Version]
  16. Guarino, A.; Lo Vecchio, A.; Dias, J.A.; Berkley, J.A.; Boey, C.; Bruzzese, D.; Cohen, M.B.; Cruchet, S.; Liguoro, I.; Salazar-Lindo, E.; et al. Universal Recommendations for the Management of Acute Diarrhea in Nonmalnourished Children. J. Pediatr. Gastroenterol. Nutr. 2018, 67, 586–593. [Google Scholar] [CrossRef] [Green Version]
  17. Shane, A.L.; Mody, R.K.; Crump, J.A.; Tarr, P.I.; Steiner, T.S.; Kotloff, K.; Langley, J.M.; Wanke, C.; Warren, C.A.; Cheng, A.C.; et al. 2017 Infectious Diseases Society of America Clinical Practice Guidelines for the Diagnosis and Management of Infectious Diarrhea. Clin. Infect. Dis 2017, 65, 1963–1973. [Google Scholar] [CrossRef]
  18. Szajewska, H.; Guarino, A.; Hojsak, I.; Indrio, F.; Kolacek, S.; Orel, R.; Salvatore, S.; Shamir, R.; van Goudoever, J.B.; Vandenplas, Y.; et al. Use of Probiotics for the Management of Acute Gastroenteritis in Children: An Update. J. Pediatr. Gastroenterol. Nutr. 2020, 71, 261. [Google Scholar] [CrossRef]
  19. Hojsak, I.; Fabiano, V.; Pop, T.L.; Goulet, O.; Zuccotti, G.V.; Çokuğraş, F.C.; Pettoello-Mantovani, M.; Kolaček, S. Guidance on the Use of Probiotics in Clinical Practice in Children with Selected Clinical Conditions and in Specific Vulnerable Groups. Acta Paediatr. 2018, 107, 927–937. [Google Scholar] [CrossRef]
  20. Meliț, L.E.; Mărginean, C.O.; Săsăran, M.O. The Challenges of Eradicating Pediatric Helicobacter Pylori Infection in the Era of Probiotics. Children 2022, 9, 795. [Google Scholar] [CrossRef]
  21. Freedman, S.B.; Xie, J.; Nettel-Aguirre, A.; Pang, X.-L.; Chui, L.; Williamson-Urquhart, S.; Schnadower, D.; Schuh, S.; Sherman, P.M.; Lee, B.E.; et al. A Randomized Trial Evaluating Virus-Specific Effects of a Combination Probiotic in Children with Acute Gastroenteritis. Nat. Commun. 2020, 11, 2533. [Google Scholar] [CrossRef] [PubMed]
  22. Freedman, S.B.; Finkelstein, Y.; Pang, X.L.; Chui, L.; Tarr, P.I.; VanBuren, J.M.; Olsen, C.; Lee, B.E.; Hall-Moore, C.A.; Sapien, R.; et al. Pathogen-Specific Effects of Probiotics in Children with Acute Gastroenteritis Seeking Emergency Care: A Randomized Trial. Clin. Infect. Dis. 2022, 75, 55–64. [Google Scholar] [CrossRef] [PubMed]
  23. Basu, S.; Chatterjee, M.; Ganguly, S.; Chandra, P.K. Effect of Lactobacillus rhamnosus GG in Persistent Diarrhea in Indian Children: A Randomized Controlled Trial. J. Clin. Gastroenterol. 2007, 41, 756–760. [Google Scholar] [CrossRef] [PubMed]
  24. Guandalini, S.; Pensabene, L.; Zikri, M.A.; Dias, J.A.; Casali, L.G.; Hoekstra, H.; Kolacek, S.; Massar, K.; Micetic-Turk, D.; Papadopoulou, A.; et al. Lactobacillus GG Administered in Oral Rehydration Solution to Children with Acute Diarrhea: A Multicenter European Trial. J. Pediatr. Gastroenterol. Nutr. 2000, 30, 54–60. [Google Scholar] [CrossRef] [PubMed]
  25. Szymański, H.; Pejcz, J.; Jawień, M.; Chmielarczyk, A.; Strus, M.; Heczko, P.B. Treatment of Acute Infectious Diarrhoea in Infants and Children with a Mixture of Three Lactobacillus rhamnosus Strains—A Randomized, Double-Blind, Placebo-Controlled Trial. Aliment. Pharm. 2006, 23, 247–253. [Google Scholar] [CrossRef]
  26. Guarino, A.; Canani, R.B.; Spagnuolo, M.I.; Albano, F.; Di Benedetto, L. Oral Bacterial Therapy Reduces the Duration of Symptoms and of Viral Excretion in Children with Mild Diarrhea. J. Pediatr. Gastroenterol. Nutr. 1997, 25, 516–519. [Google Scholar] [CrossRef]
  27. Fang, S.-B.; Lee, H.-C.; Hu, J.-J.; Hou, S.-Y.; Liu, H.-L.; Fang, H.-W. Dose-Dependent Effect of Lactobacillus rhamnosus on Quantitative Reduction of Faecal Rotavirus Shedding in Children. J. Trop. Pediatr. 2009, 55, 297–301. [Google Scholar] [CrossRef] [Green Version]
  28. Aggarwal, S.; Upadhyay, A.; Shah, D.; Teotia, N.; Agarwal, A.; Jaiswal, V. Lactobacillus GG for Treatment of Acute Childhood Diarrhoea: An Open Labelled, Randomized Controlled Trial. Indian J. Med. Res. 2014, 139, 379–385. [Google Scholar]
  29. Lo Vecchio, A.; Nunziata, F.; Bruzzese, D.; Conelli, M.L.; Guarino, A. Rotavirus Immunisation Status Affects the Efficacy of Lacticaseibacillus Rhamnosus GG for the Treatment of Children with Acute Diarrhoea: A Meta-Analysis. Benef. Microbes 2022, 13, 283–294. [Google Scholar] [CrossRef]
  30. Sindhu, K.N.C.; Sowmyanarayanan, T.V.; Paul, A.; Babji, S.; Ajjampur, S.S.R.; Priyadarshini, S.; Sarkar, R.; Balasubramanian, K.A.; Wanke, C.A.; Ward, H.D.; et al. Immune Response and Intestinal Permeability in Children with Acute Gastroenteritis Treated with Lactobacillus rhamnosus GG: A Randomized, Double-Blind, Placebo-Controlled Trial. Clin. Infect. Dis. 2014, 58, 1107–1115. [Google Scholar] [CrossRef]
  31. Grenov, B.; Namusoke, H.; Lanyero, B.; Nabukeera-Barungi, N.; Ritz, C.; Mølgaard, C.; Friis, H.; Michaelsen, K.F. Effect of Probiotics on Diarrhea in Children with Severe Acute Malnutrition: A Randomized Controlled Study in Uganda. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 396–403. [Google Scholar] [CrossRef] [PubMed]
  32. Schnadower, D.; Tarr, P.I.; Gorelick, M.H.; O’Connell, K.; Roskind, C.G.; Powell, E.C.; Rao, J.; Bhatt, S.; Freedman, S.B. Validation of the Modified Vesikari Score in Children with Gastroenteritis in 5 US Emergency Departments. J. Pediatr. Gastroenterol. Nutr. 2013, 57, 514–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Li, Z.; Zhu, G.; Li, C.; Lai, H.; Liu, X.; Zhang, L. Which Probiotic Is the Most Effective for Treating Acute Diarrhea in Children? A Bayesian Network Meta-Analysis of Randomized Controlled Trials. Nutrients 2021, 13, 4319. [Google Scholar] [CrossRef] [PubMed]
  34. Vineeth, S.; Saireddy, S.; Keerthi, T.; Mantada, P.K. Efficacy of Bacillus Clausii and Saccharomyces boulardii in Treatment of Acute Rotaviral Diarrhea in Pediatric Patients. Indones. J. Clin. Pharm. 2017, 6, 91–98. [Google Scholar] [CrossRef] [Green Version]
  35. Gaón, D.; García, H.; Winter, L.; Rodríguez, N.; Quintás, R.; González, S.N.; Oliver, G. Effect of Lactobacillus Strains and Saccharomyces boulardii on Persistent Diarrhea in Children. Medicina 2003, 63, 293–298. [Google Scholar]
  36. Das, S.; Gupta, P.K.; Das, R.R. Efficacy and Safety of Saccharomyces boulardii in Acute Rotavirus Diarrhea: Double Blind Randomized Controlled Trial from a Developing Country. J. Trop. Pediatr. 2016, 62, 464–470. [Google Scholar] [CrossRef] [Green Version]
  37. Dalgic, N.; Sancar, M.; Bayraktar, B.; Pullu, M.; Hasim, O. Probiotic, Zinc and Lactose-Free Formula in Children with Rotavirus Diarrhea: Are They Effective? Pediatr. Int. 2011, 53, 677–682. [Google Scholar] [CrossRef] [PubMed]
  38. Grandy, G.; Medina, M.; Soria, R.; Terán, C.G.; Araya, M. Probiotics in the Treatment of Acute Rotavirus Diarrhoea. A Randomized, Double-Blind, Controlled Trial Using Two Different Probiotic Preparations in Bolivian Children. BMC Infect. Dis. 2010, 10, 253. [Google Scholar] [CrossRef] [Green Version]
  39. Corrêa, N.B.O.; Penna, F.J.; Lima, F.M.L.S.; Nicoli, J.R.; Filho, L.A.P. Treatment of Acute Diarrhea with Saccharomyces boulardii in Infants. J. Pediatr. Gastroenterol. Nutr. 2011, 53, 497–501. [Google Scholar] [CrossRef]
  40. Bennett, A.; Pollock, L.; Jere, K.C.; Pitzer, V.E.; Lopman, B.; Bar-Zeev, N.; Iturriza-Gomara, M.; Cunliffe, N.A. Duration and Density of Fecal Rotavirus Shedding in Vaccinated Malawian Children with Rotavirus Gastroenteritis. J. Infect. Dis. 2019, 222, 2035–2040. [Google Scholar] [CrossRef]
  41. Mourey, F.; Sureja, V.; Kheni, D.; Shah, P.; Parikh, D.; Upadhyay, U.; Satia, M.; Shah, D.; Troise, C.; Decherf, A. A Multicenter, Randomized, Double-Blind, Placebo-Controlled Trial of Saccharomyces boulardii in Infants and Children with Acute Diarrhea. Pediatr. Infect. Dis. J. 2020, 39, e347–e351. [Google Scholar] [CrossRef]
  42. Coconnier, M.H.; Bernet, M.F.; Kernéis, S.; Chauvière, G.; Fourniat, J.; Servin, A.L. Inhibition of Adhesion of Enteroinvasive Pathogens to Human Intestinal Caco2 Cells by Lactobacillus acidophilus Strain LB Decreases Bacterial Invasion. FEMS Microbiol. Lett. 1993, 110, 299–305. [Google Scholar] [CrossRef] [PubMed]
  43. Coconnier, M.H.; Liévin, V.; Bernet-Camard, M.F.; Hudault, S.; Servin, A.L. Antibacterial Effect of the Adhering Human Lactobacillus acidophilus Strain LB. Antimicrob. Agents Chemother. 1997, 41, 1046–1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Pinto, J.M.; Petrova, A. Lactobacillus acidophilus Mixture in Treatment of Children Hospitalized with Acute Diarrhea. Clin. Pediatr. 2016, 55, 1202–1209. [Google Scholar] [CrossRef]
  45. Cheng, H.; Ma, Y.; Liu, X.; Tian, C.; Zhong, X.; Zhao, L. A Systematic Review and Meta-Analysis: Lactobacillus acidophilus for Treating Acute Gastroenteritis in Children. Nutrients 2022, 14, 682. [Google Scholar] [CrossRef] [PubMed]
  46. Hong Chau, T.T.; Minh Chau, N.N.; Hoang Le, N.T.; Chung The, H.; Voong Vinh, P.; Nguyen To, N.T.; Ngoc, N.M.; Tuan, H.M.; Chau Ngoc, T.L.; Kolader, M.-E.; et al. A Double-Blind, Randomized, Placebo-Controlled Trial of Lactobacillus acidophilus for the Treatment of Acute Watery Diarrhea in Vietnamese Children. Pediatr. Infect. Dis. J. 2018, 37, 35–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Khanna, V.; Alam, S.; Malik, A.; Malik, A. Efficacy of Tyndalized Lactobacillus acidophilus in Acute Diarrhea. Indian J. Pediatr. 2005, 72, 935–938. [Google Scholar] [CrossRef] [PubMed]
  48. Liévin-Le Moal, V.; Sarrazin-Davila, L.E.; Servin, A.L. An Experimental Study and a Randomized, Double-Blind, Placebo-Controlled Clinical Trial to Evaluate the Antisecretory Activity of Lactobacillus acidophilus Strain LB against Nonrotavirus Diarrhea. Pediatrics 2007, 120, e795–e803. [Google Scholar] [CrossRef]
  49. Salazar-Lindo, E.; Figueroa-Quintanilla, D.; Caciano, M.I.; Reto-Valiente, V.; Chauviere, G.; Colin, P. Lacteol Study Group Effectiveness and Safety of Lactobacillus LB in the Treatment of Mild Acute Diarrhea in Children. J. Pediatr. Gastroenterol. Nutr. 2007, 44, 571–576. [Google Scholar] [CrossRef]
  50. Szajewska, H.; Ruszczyński, M.; Kolaček, S. Meta-Analysis Shows Limited Evidence for Using Lactobacillus acidophilus LB to Treat Acute Gastroenteritis in Children. Acta Paediatr. 2014, 103, 249–255. [Google Scholar] [CrossRef]
  51. Liu, Y.; Fatheree, N.Y.; Mangalat, N.; Rhoads, J.M. Human-Derived Probiotic Lactobacillus reuteri Strains Differentially Reduce Intestinal Inflammation. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 299, G1087–G1096. [Google Scholar] [CrossRef] [Green Version]
  52. Ragan, M.V.; Wala, S.J.; Goodman, S.D.; Bailey, M.T.; Besner, G.E. Next-Generation Probiotic Therapy to Protect the Intestines From Injury. Front. Cell Infect. Microbiol. 2022, 12, 863949. [Google Scholar] [CrossRef] [PubMed]
  53. Fijan, S.; Šulc, D.; Steyer, A. Study of the in vitro Antagonistic Activity of Various Single-Strain and Multi-Strain Probiotics against Escherichia Coli. Int. J. Env. Res. Public Health 2018, 15, 1539. [Google Scholar] [CrossRef] [Green Version]
  54. Kiššová, Z.; Tkáčiková, Ľ.; Mudroňová, D.; Bhide, M.R. Immunomodulatory Effect of Lactobacillus reuteri (Limosilactobacillus Reuteri) and Its Exopolysaccharides Investigated on Epithelial Cell Line IPEC-J2 Challenged with Salmonella Typhimurium. Life 2022, 12, 1955. [Google Scholar] [CrossRef] [PubMed]
  55. Preidis, G.A.; Saulnier, D.M.; Blutt, S.E.; Mistretta, T.-A.; Riehle, K.P.; Major, A.M.; Venable, S.F.; Barrish, J.P.; Finegold, M.J.; Petrosino, J.F.; et al. Host Response to Probiotics Determined by Nutritional Status of Rotavirus-Infected Neonatal Mice. J. Pediatr. Gastroenterol. Nutr. 2012, 55, 299–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Gutierrez-Castrellon, P.; Lopez-Velazquez, G.; Diaz-Garcia, L.; Jimenez-Gutierrez, C.; Mancilla-Ramirez, J.; Estevez-Jimenez, J.; Parra, M. Diarrhea in Preschool Children and Lactobacillus reuteri: A Randomized Controlled Trial. Pediatrics 2014, 133, e904–e909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Shornikova, A.V.; Casas, I.A.; Isolauri, E.; Mykkänen, H.; Vesikari, T. Lactobacillus reuteri as a Therapeutic Agent in Acute Diarrhea in Young Children. J. Pediatr. Gastroenterol. Nutr. 1997, 24, 399–404. [Google Scholar] [CrossRef]
  58. Shornikova, A.V.; Casas, I.A.; Mykkänen, H.; Salo, E.; Vesikari, T. Bacteriotherapy with Lactobacillus reuteri in Rotavirus Gastroenteritis. Pediatr. Infect. Dis. J. 1997, 16, 1103–1107. [Google Scholar] [CrossRef]
  59. Pernica, J.M.; Arscott-Mills, T.; Steenhoff, A.P.; Mokomane, M.; Moorad, B.; Bapabi, M.; Lechiile, K.; Mangwegape, O.; Batisani, B.; Mawoko, N.; et al. Optimising the Management of Childhood Acute Diarrhoeal Disease Using a Rapid Test-and- Treat Strategy and/or Lactobacillus reuteri DSM 17938: A Multicentre, Randomised, Controlled, Factorial Trial in Botswana. BMJ Glob. Health 2022, 7, e007826. [Google Scholar] [CrossRef]
  60. Szymański, H.; Szajewska, H. Lack of Efficacy of Lactobacillus reuteri DSM 17938 for the Treatment of Acute Gastroenteritis: A Randomized Controlled Trial. Pediatr. Infect. Dis. J. 2019, 38, e237–e242. [Google Scholar] [CrossRef]
  61. Maragkoudakis, P.A.; Chingwaru, W.; Gradisnik, L.; Tsakalidou, E.; Cencic, A. Lactic Acid Bacteria Efficiently Protect Human and Animal Intestinal Epithelial and Immune Cells from Enteric Virus Infection. Int. J. Food Microbiol. 2010, 141, S91–S97. [Google Scholar] [CrossRef] [PubMed]
  62. Kim, K.; Lee, G.; Thanh, H.D.; Kim, J.-H.; Konkit, M.; Yoon, S.; Park, M.; Yang, S.; Park, E.; Kim, W. Exopolysaccharide from Lactobacillus plantarum LRCC5310 Offers Protection against Rotavirus-Induced Diarrhea and Regulates Inflammatory Response. J. Dairy Sci. 2018, 101, 5702–5712. [Google Scholar] [CrossRef] [PubMed]
  63. Shin, D.Y.; Yi, D.Y.; Jo, S.; Lee, Y.M.; Kim, J.-H.; Kim, W.; Park, M.r.; Yoon, S.m.; Kim, Y.; Yang, S.; et al. Effect of a New Lactobacillus plantarum Product, LRCC5310, on Clinical Symptoms and Virus Reduction in Children with Rotaviral Enteritis. Medicine 2020, 99, e22192. [Google Scholar] [CrossRef] [PubMed]
  64. Lee, D.K.; Park, J.E.; Kim, M.J.; Seo, J.G.; Lee, J.H.; Ha, N.J. Probiotic Bacteria, B. longum and L. acidophilus Inhibit Infection by Rotavirus in vitro and Decrease the Duration of Diarrhea in Pediatric Patients. Clin. Res. Hepatol. Gastroenterol. 2015, 39, 237–244. [Google Scholar] [CrossRef] [PubMed]
  65. Park, M.S.; Kwon, B.; Ku, S.; Ji, G.E. The Efficacy of Bifidobacterium Longum BORI and Lactobacillus acidophilus AD031 Probiotic Treatment in Infants with Rotavirus Infection. Nutrients 2017, 9, 887. [Google Scholar] [CrossRef] [Green Version]
  66. Teran, C.G.; Teran-Escalera, C.N.; Villarroel, P. Nitazoxanide vs. Probiotics for the Treatment of Acute Rotavirus Diarrhea in Children: A Randomized, Single-Blind, Controlled Trial in Bolivian Children. Int. J. Infect. Dis. 2009, 13, 518–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Huang, Y.-F.; Liu, P.-Y.; Chen, Y.-Y.; Nong, B.-R.; Huang, I.-F.; Hsieh, K.-S.; Chen, K.-T. Three-Combination Probiotics Therapy in Children with Salmonella and Rotavirus Gastroenteritis. J. Clin. Gastroenterol. 2014, 48, 37–42. [Google Scholar] [CrossRef]
  68. Sobouti, B.; Noorbakhsh, S.; Ashraf, H.; Ashraf -Talesh, S. Use of Probiotic for the Treatment of Acute Rotavirus Diarrhea in Children: A Randomized Single-Blind Controlled Trial. Int. J. Child. Adolesc. 2016, 2, 5–9. [Google Scholar]
  69. Dubey, A.P.; Rajeshwari, K.; Chakravarty, A.; Famularo, G. Use of VSL[Sharp]3 in the Treatment of Rotavirus Diarrhea in Children: Preliminary Results. J. Clin. Gastroenterol. 2008, 42, S126–S129. [Google Scholar] [CrossRef]
  70. Rosenfeldt, V.; Michaelsen, K.F.; Jakobsen, M.; Larsen, C.N.; Møller, P.L.; Pedersen, P.; Tvede, M.; Weyrehter, H.; Valerius, N.H.; Paerregaard, A. Effect of Probiotic Lactobacillus Strains in Young Children Hospitalized with Acute Diarrhea. Pediatr. Infect. Dis. J. 2002, 21, 411–416. [Google Scholar] [CrossRef]
  71. Rigo-Adrover, M.; Pérez-Berezo, T.; Ramos-Romero, S.; van Limpt, K.; Knipping, K.; Garssen, J.; Knol, J.; Franch, À.; Castell, M.; Pérez-Cano, F.J. A Fermented Milk Concentrate and a Combination of Short-Chain Galacto-Oligosaccharides/Long-Chain Fructo-Oligosaccharides/Pectin-Derived Acidic Oligosaccharides Protect Suckling Rats from Rotavirus Gastroenteritis. Br. J. Nutr. 2017, 117, 209–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Rigo-Adrover, M.; Saldaña-Ruíz, S.; van Limpt, K.; Knipping, K.; Garssen, J.; Knol, J.; Franch, A.; Castell, M.; Pérez-Cano, F.J. A Combination of ScGOS/LcFOS with Bifidobacterium Breve M-16V Protects Suckling Rats from Rotavirus Gastroenteritis. Eur. J. Nutr. 2017, 56, 1657–1670. [Google Scholar] [CrossRef] [PubMed]
  73. Ishizuka, T.; Kanmani, P.; Kobayashi, H.; Miyazaki, A.; Soma, J.; Suda, Y.; Aso, H.; Nochi, T.; Iwabuchi, N.; Xiao, J.; et al. Immunobiotic Bifidobacteria Strains Modulate Rotavirus Immune Response in Porcine Intestinal Epitheliocytes via Pattern Recognition Receptor Signaling. PLoS ONE 2016, 11, e0152416. [Google Scholar] [CrossRef] [PubMed]
  74. Gonzalez-Ochoa, G.; Flores-Mendoza, L.K.; Icedo-Garcia, R.; Gomez-Flores, R.; Tamez-Guerra, P. Modulation of Rotavirus Severe Gastroenteritis by the Combination of Probiotics and Prebiotics. Arch. Microbiol. 2017, 199, 953–961. [Google Scholar] [CrossRef] [Green Version]
  75. Dewi, M.R.; Soenarto, Y.; Karyana, I.P.G. Efficacy of Synbiotic Treatment in Children with Acute Rotavirus Diarrhea. Paediatr. Indones. 2015, 55, 74–78. [Google Scholar] [CrossRef]
  76. Islek, A.; Sayar, E.; Yılmaz, A.; Baysan, B.; Mutlu, D.; Artan, R. The Role of Bifidobacterium Lactis B94 plus Inulin in the Treatment of Acute Infectious Diarrhea in Children. Turk. J. Gastroenterol. J. Turk. Soc. Gastroenterol. 2014, 25, 628–633. [Google Scholar] [CrossRef]
  77. Kluijfhout, S.; Trieu, T.-V.; Vandenplas, Y. Efficacy of the Probiotic Probiotical Confirmed in Acute Gastroenteritis. Pediatr. Gastroenterol. Hepatol. Nutr. 2020, 23, 464–471. [Google Scholar] [CrossRef]
  78. Yazar, A.S.; Güven, Ş.; Dinleyici, E.Ç. Effects of Zinc or Synbiotic on the Duration of Diarrhea in Children with Acute Infectious Diarrhea. Turk. J. Gastroenterol. 2016, 27, 537–540. [Google Scholar] [CrossRef]
  79. Vandenplas, Y.; De Hert, S.G.; PROBIOTICAL–Study Group. Randomised Clinical Trial: The Synbiotic Food Supplement Probiotical vs. Placebo for Acute Gastroenteritis in Children. Aliment. Pharm. 2011, 34, 862–867. [Google Scholar] [CrossRef]
  80. Passariello, A.; Terrin, G.; Cecere, G.; Micillo, M.; De Marco, G.; Di Costanzo, M.; Cosenza, L.; Leone, L.; Nocerino, R.; Canani, R.B. Randomised Clinical Trial: Efficacy of a New Synbiotic Formulation Containing Lactobacillus paracasei B21060 plus Arabinogalactan and Xilooligosaccharides in Children with Acute Diarrhoea. Aliment. Pharm. 2012, 35, 782–788. [Google Scholar] [CrossRef]
  81. Szajewska, H.; Canani, R.B.; Domellöf, M.; Guarino, A.; Hojsak, I.; Indrio, F.; Lo Vecchio, A.; Mihatsch, W.A.; Mosca, A.; Orel, R.; et al. Probiotics for the Management of Pediatric Gastrointestinal Disorders: Position Paper of the ESPGHAN Special Interest Group on Gut Microbiota and Modifications. J. Pediatr. Gastroenterol. Nutr. 2022. [Google Scholar] [CrossRef] [PubMed]
  82. Steyer, A.; Mičetić-Turk, D.; Fijan, S. The Efficacy of Probiotics as Antiviral Agents for the Treatment of Rotavirus Gastrointestinal Infections in Children: An Updated Overview of Literature. Microorganisms 2022, 10, 2392. [Google Scholar] [CrossRef] [PubMed]
  83. Ahmadi, E.; Alizadeh-Navaei, R.; Rezai, M.S. Efficacy of Probiotic Use in Acute Rotavirus Diarrhea in Children: A Systematic Review and Meta-Analysis. Casp. J. Intern. Med. 2015, 6, 187–195. [Google Scholar]
  84. Burnett, E.; Parashar, U.D.; Tate, J.E. Rotavirus Infection, Illness, and Vaccine Performance in Malnourished Children: A Review of the Literature. Pediatr. Infect. Dis. J. 2021, 40, 930–936. [Google Scholar] [CrossRef]
  85. Colbère-Garapin, F.; Martin-Latil, S.; Blondel, B.; Mousson, L.; Pelletier, I.; Autret, A.; François, A.; Niborski, V.; Grompone, G.; Catonnet, G.; et al. Prevention and Treatment of Enteric Viral Infections: Possible Benefits of Probiotic Bacteria. Microbes Infect. 2007, 9, 1623–1631. [Google Scholar] [CrossRef]
  86. Payne, D.C.; Vinjé, J.; Szilagyi, P.G.; Edwards, K.M.; Staat, M.A.; Weinberg, G.A.; Hall, C.B.; Chappell, J.; Bernstein, D.I.; Curns, A.T.; et al. Norovirus and Medically Attended Gastroenteritis in U.S. Children. N. Engl. J. Med. 2013, 368, 1121–1130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. McFarland, L.V.; Srinivasan, R.; Setty, R.P.; Ganapathy, S.; Bavdekar, A.; Mitra, M.; Raju, B.; Mohan, N. Specific Probiotics for the Treatment of Pediatric Acute Gastroenteritis in India: A Systematic Review and Meta-Analysis. JPGN Rep. 2021, 2, e079. [Google Scholar] [CrossRef]
  88. Narayanappa, D. Randomized Double Blinded Controlled Trial to Evaluate the Efficacy and Safety of Bifilac in Patients with Acute Viral Diarrhea. Indian J. Pediatr. 2008, 75, 709–713. [Google Scholar] [CrossRef]
  89. Ali, S.I.; Naqvi, S.B.S.; Yousuf, R.I. Antidiarrheal Potential of Lactobacillus Strains Isolated from Pharmaceutical Formulations for the Treatment of Pediatric Diarrhea. Pak. J. Pharm. Sci. 2020, 33, 1073–1078. [Google Scholar]
  90. Quraishi, F.; Fatima, G.; Shaheen, S.; Memon, Z.; Kainat, S.; Agha, F. In-Vitro Comparison of Antimicrobial Actions of Probiotics (Lactobacilli Species and Saccharomyces boulardii) with Standard Antibiotics for the Treatment of Diarrhea in Pediatric Population. Int. J. Clin. Med. 2018, 09, 827–840. [Google Scholar] [CrossRef] [Green Version]
  91. Maragkoudaki, M.; Chouliaras, G.; Moutafi, A.; Thomas, A.; Orfanakou, A.; Papadopoulou, A. Efficacy of an Oral Rehydration Solution Enriched with Lactobacillus reuteri DSM 17938 and Zinc in the Management of Acute Diarrhoea in Infants: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2018, 10, 1189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Dinleyici, E.C.; Dalgic, N.; Guven, S.; Metin, O.; Yasa, O.; Kurugol, Z.; Turel, O.; Tanir, G.; Yazar, A.S.; Arica, V.; et al. Lactobacillus reuteri DSM 17938 Shortens Acute Infectious Diarrhea in a Pediatric Outpatient Setting. J. Pediatr. 2015, 91, 392–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Chen, K.; Xin, J.; Zhang, G.; Xie, H.; Luo, L.; Yuan, S.; Bu, Y.; Yang, X.; Ge, Y.; Liu, C. A Combination of Three Probiotic Strains for Treatment of Acute Diarrhoea in Hospitalised Children: An Open Label, Randomised Controlled Trial. Benef. Microbes 2020, 11, 339–346. [Google Scholar] [CrossRef] [PubMed]
  94. Riaz, M.; Alam, S.; Malik, A.; Ali, S.M. Efficacy and Safety of Saccharomyces boulardii in Acute Childhood Diarrhea: A Double Blind Randomised Controlled Trial. Indian J. Pediatr. 2012, 79, 478–482. [Google Scholar] [CrossRef]
  95. Dinleyici, E.C.; Dalgic, N.; Guven, S.; Ozen, M.; Kara, A.; Arica, V.; Metin-Timur, O.; Sancar, M.; Kurugol, Z.; Tanir, G.; et al. The Effect of a Multispecies Synbiotic Mixture on the Duration of Diarrhea and Length of Hospital Stay in Children with Acute Diarrhea in Turkey: Single Blinded Randomized Study. Eur. J. Pediatr. 2013, 172, 459–464. [Google Scholar] [CrossRef]
  96. Gundogdu, Z. Effect of a Synbiotic on Infantile Acute Gastroenteritis. Benef. Microbes 2013, 4, 231–235. [Google Scholar] [CrossRef]
  97. Collinson, S.; Deans, A.; Padua-Zamora, A.; Gregorio, G.V.; Li, C.; Dans, L.F.; Allen, S.J. Probiotics for Treating Acute Infectious Diarrhoea. Cochrane Database Syst. Rev. 2020, 12, CD003048. [Google Scholar] [CrossRef]
  98. Vandenplas, Y. Probiotics and Prebiotics in Infectious Gastroenteritis. Best Pr. Res. Clin. Gastroenterol. 2016, 30, 49–53. [Google Scholar] [CrossRef]
  99. Martins, F.S.; Vieira, A.T.; Elian, S.D.A.; Arantes, R.M.E.; Tiago, F.C.P.; Sousa, L.P.; Araújo, H.R.C.; Pimenta, P.F.; Bonjardim, C.A.; Nicoli, J.R.; et al. Inhibition of Tissue Inflammation and Bacterial Translocation as One of the Protective Mechanisms of Saccharomyces boulardii against Salmonella Infection in Mice. Microbes Infect. 2013, 15, 270–279. [Google Scholar] [CrossRef]
  100. Mahzounieh, M.; Karimi, I.; Zahraei Salehi, T.; Marjanian, R. The Preventive Effect of Sacharomyces boulardii in Pathogenesis of Salmonella Typhimurium in Experimentally Infected Rats. Pak. J. Biol. Sci. 2006, 9, 632–635. [Google Scholar] [CrossRef]
  101. Szajewska, H.; Wanke, M.; Patro, B. Meta-Analysis: The Effects of Lactobacillus rhamnosus GG Supplementation for the Prevention of Healthcare-Associated Diarrhoea in Children. Aliment. Pharm. 2011, 34, 1079–1087. [Google Scholar] [CrossRef] [PubMed]
  102. Phuapradit, P.; Varavithya, W.; Vathanophas, K.; Sangchai, R.; Podhipak, A.; Suthutvoravut, U.; Nopchinda, S.; Chantraruksa, V.; Haschke, F. Reduction of Rotavirus Infection in Children Receiving Bifidobacteria-Supplemented Formula. J. Med. Assoc. Thai. 1999, 82, S43–S48. [Google Scholar] [PubMed]
  103. Di, J.-B.; Gai, Z.-T. Protective Efficacy of Probiotics on the Treatment of Acute Rotavirus Diarrhea in Children: An Updated Meta-Analysis. Eur. Rev. Med. Pharm. Sci. 2020, 24, 9675–9683. [Google Scholar] [CrossRef]
  104. Urbańska, M.; Szajewska, H. The Efficacy of Lactobacillus reuteri DSM 17938 in Infants and Children: A Review of the Current Evidence. Eur. J. Pediatr. 2014, 173, 1327–1337. [Google Scholar] [CrossRef] [PubMed]
  105. Karimi, S.; Jonsson, H.; Lundh, T.; Roos, S. Lactobacillus reuteri Strains Protect Epithelial Barrier Integrity of IPEC-J2 Monolayers from the Detrimental Effect of Enterotoxigenic Escherichia Coli. Physiol. Rep. 2018, 6, e13514. [Google Scholar] [CrossRef] [Green Version]
  106. Michail, S.; Abernathy, F. Lactobacillus plantarum Reduces the in vitro Secretory Response of Intestinal Epithelial Cells to Enteropathogenic Escherichia Coli Infection. J. Pediatr. Gastroenterol. Nutr. 2002, 35, 350–355. [Google Scholar] [CrossRef]
  107. Depoorter, L.; Vandenplas, Y. Probiotics in Pediatrics. A Review and Practical Guide. Nutrients 2021, 13, 2176. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Săsăran, M.O.; Mărginean, C.O.; Adumitrăchioaiei, H.; Meliț, L.E. Pathogen-Specific Benefits of Probiotic and Synbiotic Use in Childhood Acute Gastroenteritis: An Updated Review of the Literature. Nutrients 2023, 15, 643. https://0-doi-org.brum.beds.ac.uk/10.3390/nu15030643

AMA Style

Săsăran MO, Mărginean CO, Adumitrăchioaiei H, Meliț LE. Pathogen-Specific Benefits of Probiotic and Synbiotic Use in Childhood Acute Gastroenteritis: An Updated Review of the Literature. Nutrients. 2023; 15(3):643. https://0-doi-org.brum.beds.ac.uk/10.3390/nu15030643

Chicago/Turabian Style

Săsăran, Maria Oana, Cristina Oana Mărginean, Heidrun Adumitrăchioaiei, and Lorena Elena Meliț. 2023. "Pathogen-Specific Benefits of Probiotic and Synbiotic Use in Childhood Acute Gastroenteritis: An Updated Review of the Literature" Nutrients 15, no. 3: 643. https://0-doi-org.brum.beds.ac.uk/10.3390/nu15030643

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop