Next Article in Journal
High-Calorie Diet Exacerbates the Crosstalk Between Gestational Diabetes and Youth-Onset Diabetes in Female Offspring Through Disrupted Estrogen Signaling
Previous Article in Journal
Evolving Research Focus on Diet and Cardiovascular Disease: A Systematic Review of 298 Cohort Studies Published from 2019 to 2024
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Safety and Tolerance of Bifidobacterium longum subsp. Infantis YLGB-1496 in Toddlers with Respiratory Symptoms

1
Department of Nutrition and Food Hygiene, School of Public Health, Peking University Health Science Center, Beijing 100191, China
2
School of Industrial Technology, Universiti Sains Malaysia, Gelugor 11800, Malaysia
3
Pediatric & Palliative Care, Hospital Pakar Universiti Sains Malaysia, Kota Bharu 16150, Malaysia
4
Kepala Batas Health Clinic, Kepala Batas 13200, Malaysia
5
Cluster of Regenerative Medicine, Advanced Medical & Dental Institute, Universiti Sains Malaysia, Bertam 13200, Malaysia
6
School of Nursing, Peking University, Beijing 100191, China
7
Department of Pediatrics, UKM Medical Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
*
Authors to whom correspondence should be addressed.
Nutrients 2025, 17(13), 2127; https://doi.org/10.3390/nu17132127
Submission received: 24 May 2025 / Revised: 23 June 2025 / Accepted: 24 June 2025 / Published: 26 June 2025
(This article belongs to the Section Prebiotics and Probiotics)

Abstract

Objective: The aim of this study was to examine the safety and tolerance of Bifidobacterium longum subsp. infantis YLGB-1496 (B. infantis YLGB-1496) in toddlers with respiratory illness. Methods: In this randomized controlled trial, 120 toddlers with respiratory illness were randomly assigned to the probiotic (YLGB-1496) or control group for a 12-week intervention. Follow-up examinations were conducted at baseline (week 0) and at weeks 6 and 12 of the intervention. Toddlers’ height and weight were measured by trained personnel, and defecation characteristics and gastrointestinal symptoms were recorded by parents or guardians. Stool samples were collected to determine the fecal pH, fecal calprotectin (FC) concentration, and fecal α1-antitrypsin (AAT) concentration. Results: A total of 115 toddlers completed the 12-week intervention (58 in the YLGB-1496 group and 57 in the control group). The height-for-age Z score (HAZ) in the YLGB-1496 group was significantly greater than that in the control group (p = 0.006). The weight-for-age Z score (WAZ) in the YLGB-1496 group increased between weeks 6 and 12, whereas the WAZ in the control group continuously decreased during the intervention. No differences in the frequency or consistency of defecation between the groups were observed. Toddlers in the YLGB-1496 group had lower incidences of poor appetite, nausea, vomiting, stomachache, lower abdominal pain, diarrhea, and dehydration (p < 0.05) but higher fecal AAT concentrations (p = 0.008) than did those in the control group. No differences in the fecal pH or FC concentration were observed between the groups. Conclusions: B. infantis YLGB-1496 demonstrated excellent safety and tolerability in toddlers and effectively reduced the gastrointestinal discomfort associated with respiratory illnesses.

1. Introduction

Bifidobacterium was first isolated from the feces of breastfed infants by Tissier [1]. Substantial clinical evidence supports the use of Bifidobacterium as a therapeutic option for both preterm and healthy full-term infants [2]. These bacteria are inherently vital components of a healthy infant gut microbiome, thus indicating significant potential for clinical application. While the majority of Lactobacillus and Bifidobacterium strains are recognized as probiotics, Bifidobacterium longum subsp. Infantis (B. infantis) may be particularly important during early life. Previous studies have demonstrated that B. infantis supplementation promotes the development of the immune system, intestinal maturation, gut microbiome balance, and digestive system absorption in infants and young children [3,4,5,6].
Research indicates that B. infantis increases the proliferation and differentiation of intestinal epithelial cells and stem cells. By modulating the expression of transmembrane proteins [7] and strengthening tight junctions between epithelial cells [8], B. infantis improves the function of the intestinal mucosal barrier [9]. In the assessment of intestinal maturity in infants and young children, fecal calprotectin (FC) and α1-antitrypsin (AAT) are commonly used biomarkers. Since AAT is neither degraded by digestive enzymes nor reabsorbed by the intestines, its levels correlate positively with the extent of serum protein loss in the gut, making it a useful indicator for evaluating gastrointestinal development and inflammatory status [10]. Studies have confirmed that B. infantis reduces FC concentrations in infants [11], suggesting that probiotics may improve intestinal barrier function by modulating gut permeability.
Several intervention trials have demonstrated that specific strains of B. infantis can modulate stool consistency, reducing the incidence of both constipation and diarrhea in infants and toddlers [12,13].
This manuscript presents a secondary analysis of data derived from our previously published randomized controlled trial (NCT05794815) [14], which established the efficacy of Bifidobacterium longum subsp. infantis YLGB-1496 (B. infantis YLGB-1496) in reducing respiratory symptoms in toddlers. However, the safety profile, tolerability, and specific impacts of this probiotic intervention on growth metrics and gastrointestinal (GI) health in toddlers with respiratory illnesses remained unexplored in that primary report. Therefore, the specific aims of this secondary analysis are: (a) to rigorously evaluate the safety and tolerability of B. infantis YLGB-1496 in toddlers (aged 1–3 years) experiencing respiratory illnesses and (b) to explore its effects on the intestinal health of this specific pediatric population. This work thus addresses critical gaps by focusing on these previously unreported variables from the original RCT dataset.

2. Materials and Methods

2.1. Study Design

Briefly, 120 toddlers with at least 2 kinds of respiratory symptoms were randomized 1:1 to receive B. infantis YLGB-1496 1 × 1010 colony-forming units (CFUs)/day or placebo (maltodextrin) in a 12-week intervention (Figure 1). According to previous studies, a high dose of B. infantis (1.8–2.8 × 1010 CFUs/day) is safe and well-tolerated in healthy infants [13,15].

2.2. Outcomes and Measurements

The outcomes of this analysis were physical development, gastrointestinal symptoms, defecation characteristics (stool hardness and defecation frequency), fecal pH, and FC and AAT concentrations.

2.2.1. Questionnaires

All questionnaires used in the study were validated for reliability and validity and translated into Malay [16]. Questionnaires primarily consisted of the following sections:
(a)
Sociodemographic questionnaire.
(b)
Stool characteristics: Stool consistency (evaluated using the Bristol Stool Scale (BSS)) and frequency.
(c)
Gastrointestinal health: Frequency of adverse gastrointestinal symptoms (poor appetite, nausea, vomiting, stomachache, anal discomfort, diarrhea, and dehydration).
(d)
Dietary intake: Consumption rates of breast milk, formula, yogurt, other dairy products, grains and tubers, vegetables, fruits, protein sources (meat, eggs, and seafood), beverages, snacks, and various nutritional supplements.
(e)
Adverse events (AEs) and serious adverse events (SAEs).

2.2.2. Body Measurements

At weeks 0, 6, and 12 of the intervention, professionally trained nurses, who had undergone uniform training, measured the height and weight of the infants using standardized measuring beds and scales. World Health Organization (WHO) Anthro software (Version 3.2) was used to calculate the growth and development Z scores of the infants on the basis of the onsite measurements of their height/length and weight data. These Z scores included the height-for-age Z score (HAZ), weight-for-age Z score (WAZ), weight-for-height Z score (WHZ), and body-mass-index-for-age Z score (BAZ).

2.2.3. Sample Collection and Analysis

Stool samples were collected at weeks 0, 6, and 12 of the intervention. The pH of the stool samples was measured using a solid and semisolid pH meter. The concentrations of AAT and FC in the stool were determined using enzyme-linked immunosorbent assay (ELISA) kits (Sunlong Biotech, Hangzhou, China).

2.3. Statistical Analysis

All analyses followed the intention-to-treat principle, consistent with the primary trial [14]. Continuous variables are presented as the mean value ± the standard deviation (mean ± SD) unless otherwise stated. Categorical variables are presented as frequencies and percentages (n, %). Differences in continuous variables between groups or across different time points were compared using Student’s t-test. Differences in count variables were analyzed using Poisson regression.
A mixed-effects model was employed to analyze the effects of intervention methods and intervention duration on various outcome measures. Baseline values, group, intervention time, and the interaction term between group and time were included as fixed effects, whereas individuals were treated as random effects. Confounding factors that differed between the two groups were considered in different models, and linear mixed-effects models were constructed. If the interaction term was significant, post hoc tests were conducted; if the interaction term was not significant, it was removed from the model for further analysis. For count variables as outcome measures, a generalized mixed-effects model with family = Poisson was used for analysis. All tests were two-sided, with p < 0.05 considered to indicate statistical significance, and were performed via R software version 4.2.3.

3. Results

3.1. Baseline Characteristics

Among the 120 screened participants, 63 were randomized to the YLGB-1496 group and 57 were randomized to the control group. Five participants in the YLGB-1496 group withdrew prior to intervention initiation. Consequently, 115 participants were ultimately included in the analysis (58 in the YLGB-1496 group and 57 in the control group); see the CONSORT flow diagram in the published paper [14]. The cesarean section rate was marginally higher in the YLGB-1496 group than in the control group (p = 0.049). Statistically significant between-group differences were observed in family caregiving methods (p = 0.003). Significant intergroup differences were not observed for any other outcomes.
At baseline, the breastfeeding rate was significantly higher in the YLGB-1496 group than the placebo group (p = 0.001). Additionally, vegetable consumption rates in the YLGB-1496 group remained lower than those in the control group at both baseline and week 12 (p < 0.05). No statistically significant differences in the consumption rates of other food categories or nutritional supplements were observed between the groups (complete data are available in the published paper [14]).

3.2. Growth and Development

The growth and development Z scores for the children in both groups during the intervention period are presented in Table 1. The emmeans package was employed to obtain estimated marginal means and contrasts of Z score changes in growth parameters (Figure 2). After adjusting for confounders, including dietary factors, through linear mixed-effects models, children in the YLGB-1496 group demonstrated significantly greater HAZ than did those in the control group. Additionally, WAZ in the YLGB-1496 group moderately increased from 6–12 weeks, whereas the WAZ in the control group continuously decreased throughout the intervention period, indicating divergent temporal trends between the groups (Figure 2b). No significant differences in the WHZ or BAZ were observed between the groups.

3.3. Defecation Characteristics

A summary of the changes in stool consistency and daily defecation frequency during the intervention is presented in Table 2 and Figure 3. Both groups maintained a daily defecation frequency of 1–2 times, with BSS scores of approximately 4 points. Although the YLGB-1496 group had a marginally greater defecation frequency and slightly lower BSS scores than the control group did, these differences were not significant in either the intergroup comparisons or the temporal analyses after adjusting for baseline values and dietary intake.

3.4. Gastrointestinal Symptoms

At baseline, some children had gastrointestinal symptoms, including poor appetite and diarrhea alongside respiratory discomfort. A summary of the incidence and occurrence rates of various gastrointestinal adverse symptoms during the intervention is presented in Table 3. In the YLGB-1496 group, the incidence rates of most gastrointestinal symptoms progressively decreased over time, whereas in the control group, incidence rates remained relatively stable. Generalized mixed-effects model analysis (Table 4) revealed significantly lower incidence rates of poor appetite, nausea, vomiting, abdominal pain, intestinal colic, diarrhea, and dehydration symptoms in the YLGB-1496 group than in the control group.

3.5. Biochemical Indicators

After adjusting for potential confounders, linear mixed-effects models revealed significantly higher AAT concentrations in the YLGB-1496 group. No significant intergroup differences were detected in stool pH or FC concentrations throughout the intervention period (Table 5).

4. Discussion

In our previous study, the effects of B. infantis YLGB-1496 intervention on respiratory symptoms in young children were elucidated [14]. Building on these findings, in this secondary analysis, we focused on evaluating the safety and tolerability of the intervention, particularly its impact on growth, development, and intestinal health.
In this study, we found that the HAZ in the YLGB-1496 group was significantly greater than that in the control group. Additionally, the WAZ in the YLGB-1496 group moderately increased during the later stages of the intervention, whereas that in the control group continuously decreased. Given the strong metabolic capacity of B. infantis for human milk oligosaccharides (HMOs), it is hypothesized that this strain promotes the production of short-chain fatty acids in the infant gut, thereby improving digestion and absorption and potentially regulating physical growth [17,18]. Animal studies have demonstrated that B. infantis CCFM1269 promotes osteoblast differentiation and the expression of bone-formation-related genes by modulating the gut microbiota and metabolites, significantly promoting bone formation and increasing the length of the femur and tibia [19]. Population trials have also revealed faster HAZ growth in infants receiving B. infantis supplements [20]. However, a meta-analysis on the impact of B. infantis on infant growth revealed that, while B. infantis supplementation does not negatively affect growth, it also does not significantly promote growth [21]. Furthermore, infants receiving probiotic supplementation alone showed greater increases in length and weight than those receiving synbiotics [22].
Stool frequency and consistency were assessed among the participants. Although the YLGB-1496 group presented slightly greater stool frequency and lower BSS scores than the control group, these differences were not statistically significant after adjustments were made for potential confounders. Previous studies on other B. infantis strains have revealed that B. infantis supplementation reduces stool softness in infants and that higher doses may increase the frequency of pasty or watery stools, whereas lower doses tend to reduce the number of formed stools and increase the number of soft, unformed stools [15,23]. A randomized controlled trial in healthy full-term infants revealed that B. infantis supplementation significantly reduced the incidence of both diarrhea and constipation [12]. These findings suggest that B. infantis may have a bidirectional regulatory effect on stool characteristics, adjusting stool frequency and consistency toward an optimal range. In this study, the mean BSS score of toddlers was approximately 4, indicating soft, formed stools, which may explain the lack of significant differences observed.
The intervention significantly reduced gastrointestinal discomfort, including poor appetite, nausea, vomiting, stomachache, anal discomfort, and diarrhea. These results are consistent with findings from trials involving other B. infantis strains [24]. Compared with lower doses, higher doses of probiotics are associated with faster symptom relief [15]. Similar conclusions were drawn from a study evaluating infant formula supplemented with B. infantis [25]. Meta-analyses have indicated that probiotics positively impact irritable bowel syndrome, abdominal pain, and bloating in children [26] and provide moderate protection against antibiotic-associated diarrhea, reducing its duration [27]. However, some studies have revealed no significant differences in gastrointestinal symptom outcomes between different probiotic doses [28].
The gastrointestinal tract of infants and young children is still developing, with immature tight junctions between intestinal epithelial cells and relatively high intestinal permeability. In this study, the concentration of AAT was significantly greater in the YLGB-1496 group than in the control group. No significant effects of B. infantis YLGB-1496 on the FC concentration or fecal pH were observed. The impact of probiotics on fecal pH appears to depend on the intervention dose and population, with higher doses and exclusively breastfed infants often exhibiting more pronounced and sustained reductions in fecal pH [15,17].
Biomarkers for assessing gastrointestinal maturity in infants and young children are limited, and previous findings have been inconsistent. Animal studies have shown that B. infantis FJSYZ1M3 improves the integrity of intestinal tight junctions [29]. Some animal studies have revealed that neither probiotics nor prebiotics alone affect FC concentrations [30], although B. longum has been shown to reduce intestinal damage in rats with inflammatory bowel disease [31]. A randomized controlled trial in adults with gastrointestinal symptoms using B. longum CECT7347 revealed no significant differences in FC concentrations between groups [32]. Previous studies have shown that mixed probiotics can reduce intestinal AAT concentrations [10,33]. However, few studies have demonstrated that B. infantis supplementation lowers AAT concentrations in infants and young children. Conversely, a study in which high and low doses of B. infantis were administered to newborns revealed that both doses increased AAT concentrations, with more pronounced effects in exclusively breastfed infants [15]. Data indicate that breastfed infants have higher AAT concentrations than formula-fed infants do [34], which may partially explain these findings. The unexpected elevation of AAT in the YLGB-1496 group warrants critical interpretation, as it may indicate mucosal stimulation rather than inflammatory pathology. Meanwhile, recently, AAT has been shown to promote tissue remodeling and inflammatory resolution [35,36]. Given the consistently low baseline AAT concentrations observed in children with respiratory illnesses throughout this cohort, these elevated values do not support the conclusion that B. infantis YLGB-1496 supplementation increased intestinal permeability.
The main strength of this study is that it is the first in which the safety and tolerance of B. infantis YLGB-1496 application in toddlers were evaluated. We measured changes in FC and AAT concentrations while also considering the influence of dietary factors during data analysis. However, this study has notable limitations. The incidence of gastrointestinal symptoms was primarily self-reported by parents, which may have introduced reporting or recall bias into the statistical results. In addition, group differences were observed in baseline characteristics such as delivery mode and primary caregiving methods. While these factors were included as covariates in our multivariate regression models to mitigate confounding, they may still exert some influence on the observed intervention effects. Ethical approval mandated exclusive use of non-invasive fecal sampling. However, fecal biomarker levels are substantially influenced by environmental factors, limiting their representativeness and potentially obscuring intervention effects. To determine the effect and value of B. infantis YLGB-1496 in clinical practice, more studies with serum biomarkers, larger sample sizes, different probiotic doses, and longer interventions are still needed.

5. Conclusions

B. infantis YLGB-1496 demonstrated excellent safety and tolerability in toddlers. B. infantis YLGB-1496 effectively reduced gastrointestinal discomfort associated with respiratory illnesses and promoted intestinal health.

Author Contributions

Methodology, P.L.; Software, P.L.; Formal Analysis, P.L.; Investigation, M.U.M., F.T., T.H.K., A.Y., I.J.A.H. and M.-T.L.; Data Curation, M.U.M.; Writing—Original Draft Preparation, P.L.; Writing—Review and Editing, M.U.M., F.T., T.H.K., A.Y., I.J.A.H., H.J., M.-T.L., A.A. and Y.Z.; Visualization, P.L.; Supervision, A.A. and Y.Z.; Project Administration, H.J., M.-T.L., A.A. and Y.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was conducted according to the guidelines of the Declaration of Helsinki, and all procedures involving human subjects were approved by the UKM Research Ethics Committee (No. UKM/PPI/111/8/JEP-2023-074, 2 March 2023) and the Medical Ethics Research Board of Peking University (No. IRB00001052-22166, 10 February 2023). The trial was registered at ClinicalTrials.gov (identifier number NCT05794815).

Informed Consent Statement

Informed consent was obtained from the parents or legal guardians of all the participants involved in the study.

Data Availability Statement

The data are not publicly available due to ethical restrictions.The data presented in this study are available on request from the corresponding author.

Acknowledgments

We acknowledge all the families that participated in this study. We would like to thank the National Center of Technology Innovation for Dairy for providing the study products. The authors extend their sincere appreciation to Hanglian Lan and Weilian Hung for their valuable support of this project. Their contributions were instrumental to our work.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. O’Neill, I.; Schofield, Z.; Hall, L.J. Exploring the role of the microbiota member Bifidobacterium in modulating immune-linked diseases. Emerg. Top. Life Sci. 2017, 1, 333–349. [Google Scholar] [CrossRef] [PubMed]
  2. Bajorek, S.; Duar, R.M.; Corrigan, M.; Matrone, C.; Winn, K.A.; Norman, S.; Mitchell, R.D.; Cagney, O.; Aksenov, A.A.; Melnik, A.V.; et al. B. infantis EVC001 Is Well-Tolerated and Improves Human Milk Oligosaccharide Utilization in Preterm Infants in the Neonatal Intensive Care Unit. Front. Pediatr. 2021, 9, 795970. [Google Scholar] [CrossRef] [PubMed]
  3. Chichlowski, M.; Shah, N.; Wampler, J.L.; Wu, S.S.; Vanderhoof, J.A. Bifidobacterium longum Subspecies infantis (B. infantis) in Pediatric Nutrition: Current State of Knowledge. Nutrients 2020, 12, 1581. [Google Scholar] [CrossRef]
  4. Moreno-Muñoz, J.A.; Martín-Palomas, M.; Jiménez López, J. Bifidobacterium longum subsp. infantis CECT 7210 (B. infantis IM-1®) shows activity against intestinal pathogens. Nutr. Hosp. 2022, 39, 65–68. [Google Scholar] [CrossRef]
  5. Barratt, M.J.; Nuzhat, S.; Ahsan, K.; Frese, S.A.; Arzamasov, A.A.; Sarker, S.A.; Islam, M.M.; Palit, P.; Islam, M.R.; Hibberd, M.C.; et al. Bifidobacterium infantis treatment promotes weight gain in Bangladeshi infants with severe acute malnutrition. Sci. Transl. Med. 2022, 14, eabk1107. [Google Scholar] [CrossRef] [PubMed]
  6. Lopez-Santamarina, A.; Lamas, A.; Del Carmen Mondragón, A.; Cardelle-Cobas, A.; Regal, P.; Rodriguez-Avila, J.A.; Miranda, J.M.; Franco, C.M.; Cepeda, A. Probiotic Effects against Virus Infections: New Weapons for an Old War. Foods 2021, 10, 130. [Google Scholar] [CrossRef]
  7. Ewaschuk, J.B.; Diaz, H.; Meddings, L.; Diederichs, B.; Dmytrash, A.; Backer, J.; Looijer-van Langen, M.; Madsen, K.L. Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 295, G1025–G1034. [Google Scholar] [CrossRef]
  8. Delacour, D.; Salomon, J.; Robine, S.; Louvard, D. Plasticity of the brush border—The yin and yang of intestinal homeostasis. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 161–174. [Google Scholar] [CrossRef]
  9. La Fata, G.; Weber, P.; Mohajeri, M.H. Probiotics and the Gut Immune System: Indirect Regulation. Probiotics Antimicrob Proteins 2018, 10, 11–21. [Google Scholar] [CrossRef]
  10. Castanet, M.; Costalos, C.; Haiden, N.; Hascoet, J.M.; Berger, B.; Sprenger, N.; Grathwohl, D.; Brüssow, H.; De Groot, N.; Steenhout, P.; et al. Early Effect of Supplemented Infant Formulae on Intestinal Biomarkers and Microbiota: A Randomized Clinical Trial. Nutrients 2020, 12, 1481. [Google Scholar] [CrossRef]
  11. Henrick, B.M.; Chew, S.; Casaburi, G.; Brown, H.K.; Frese, S.A.; Zhou, Y.; Underwood, M.A.; Smilowitz, J.T. Colonization by B. infantis EVC001 modulates enteric inflammation in exclusively breastfed infants. Pediatr. Res. 2019, 86, 749–757. [Google Scholar] [CrossRef] [PubMed]
  12. Escribano, J.; Ferre, N.; Gispert-Llaurado, M.; Luque, V.; Rubio-Torrents, C.; Zaragoza-Jordana, M.; Polanco, I.; Codoner, F.M.; Chenoll, E.; Morera, M.; et al. Bifidobacterium longum subsp infantis CECT7210-supplemented formula reduces diarrhea in healthy infants: A randomized controlled trial. Pediatr. Res. 2018, 83, 1120–1128. [Google Scholar] [CrossRef]
  13. Smilowitz, J.T.; Moya, J.; Breck, M.A.; Cook, C.; Fineberg, A.; Angkustsiri, K.; Underwood, M.A. Safety and tolerability of Bifidobacterium longum subspecies infantis EVC001 supplementation in healthy term breastfed infants: A phase I clinical trial. BMC Pediatr. 2017, 17, 133. [Google Scholar] [CrossRef]
  14. Li, P.; Mageswary, U.; Ali, A.; Taib, F.; Koo, T.H.; Yusof, A.; Jiang, H.; Lan, H.; Hung, W.; Liong, M.T.; et al. Clinical effects of Bifidobacterium longum Subsp. infantis YLGB-1496 on children with respiratory symptoms. Front. Nutr. 2025, 12, 1537610. [Google Scholar] [CrossRef]
  15. Capeding, M.R.Z.; Phee, L.C.M.; Ming, C.; Noti, M.; Vidal, K.; Le Carrou, G.; Frézal, A.; Moll, J.M.; Vogt, J.K.; Myers, P.N.; et al. Safety, efficacy, and impact on gut microbial ecology of a Bifidobacterium longum subspecies infantis LMG11588 supplementation in healthy term infants: A randomized, double-blind, controlled trial in the Philippines. Front. Nutr. 2023, 10, 1319873. [Google Scholar] [CrossRef]
  16. Lau, A.S.Y.; Yusoff, M.S.B.; Lee, Y.Y.; Choi, S.B.; Rashid, F.; Wahid, N.; Xiao, J.Z.; Liong, M.T.; School of Industrial Technology; Universiti Sains Malaysia Pulau Pinang Malaysia; et al. Development, translation and validation of questionnaires for diarrhea and respiratory-related illnesses during probiotic administration in children. Educ. Med. J. 2017, 9, 19–30. [Google Scholar] [CrossRef]
  17. Frese, S.A.; Hutton, A.A.; Contreras, L.N.; Shaw, C.A.; Palumbo, M.C.; Casaburi, G.; Xu, G.; Davis, J.C.C.; Lebrilla, C.B.; Henrick, B.M.; et al. Persistence of Supplemented Bifidobacterium longum subsp. infantis EVC001 in Breastfed Infants. mSphere 2017, 2, e00501–e00517. [Google Scholar] [CrossRef] [PubMed]
  18. Larke, J.A.; Kuhn-Riordon, K.; Taft, D.H.; Sohn, K.; Iqbal, S.; Underwood, M.A.; Mills, D.A.; Slupsky, C.M. Preterm Infant Fecal Microbiota and Metabolite Profiles Are Modulated in a Probiotic Specific Manner. J. Pediatr. Gastroenterol. Nutr. 2022, 75, 535–542. [Google Scholar] [CrossRef]
  19. Ding, M.; Li, B.; Chen, H.; Liang, D.; Ross, R.P.; Stanton, C.; Zhao, J.; Chen, W.; Yang, B. Human breastmilk-derived Bifidobacterium longum subsp. infantis CCFM1269 regulates bone formation by the GH/IGF axis through PI3K/AKT pathway. Gut Microbes 2023, 16, 2290344. [Google Scholar] [CrossRef]
  20. Huda, M.N.; Lewis, Z.; Kalanetra, K.M.; Rashid, M.; Ahmad, S.M.; Raqib, R.; Qadri, F.; Underwood, M.A.; Mills, D.A.; Stephensen, C.B. Stool microbiota and vaccine responses of infants. Pediatrics 2014, 134, e362–e372. [Google Scholar] [CrossRef]
  21. Guo, H.; Fan, M.; Hou, T.; Li, Y.; Wang, S.; Wang, X.; Peng, H.; Wang, M.; Wu, T.; Zhang, Y. Efficacy and Safety of Bifidobacterium longum Supplementation in Infants: A Meta-Analysis of Randomized Controlled Trials. Foods 2023, 12, 4451. [Google Scholar] [CrossRef]
  22. Nuzhat, S.; Hasan, S.M.T.; Palit, P.; Islam, M.R.; Mahfuz, M.; Islam, M.M.; Alam, M.A.; Flannery, R.L.; Kyle, D.J.; Sarker, S.A.; et al. Effects of probiotic and synbiotic supplementation on ponderal and linear growth in severely malnourished young infants in a randomized clinical trial. Sci. Rep. 2023, 13, 1845. [Google Scholar] [CrossRef]
  23. Manzano, S.; De Andres, J.; Castro, I.; Rodriguez, J.M.; Jimenez, E.; Espinosa-Martos, I. Safety and tolerance of three probiotic strains in healthy infants: A multi-centre randomized, double-blind, placebo-controlled trial. Benef. Microbes 2017, 8, 569–578. [Google Scholar] [CrossRef]
  24. Dimitratos, S.M.; Brown, H.; Shafizadeh, T.; Kazi, S.; Altmann, T.; Ostrer, B. Symptomatic relief from at-home use of activated Bifidobacterium infantis EVC001 probiotic in infants: Results from a consumer survey on the effects on diaper rash, colic symptoms, and sleep. Benef. Microbes 2021, 12, 27–34. [Google Scholar] [CrossRef] [PubMed]
  25. Dupont, C.; Rivero, M.; Grillon, C.; Belaroussi, N.; Kalindjian, A.; Marin, V. Alpha-lactalbumin-enriched and probiotic-supplemented infant formula in infants with colic: Growth and gastrointestinal tolerance. Eur. J. Clin. Nutr. 2010, 64, 765–767. [Google Scholar] [CrossRef]
  26. Ford, A.C.; Quigley, E.M.; Lacy, B.E.; Lembo, A.J.; Saito, Y.A.; Schiller, L.R.; Soffer, E.E.; Spiegel, B.M.; Moayyedi, P. Efficacy of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and chronic idiopathic constipation: Systematic review and meta-analysis. Am. J. Gastroenterol. 2014, 109, 1547–1561, quiz 1546, 1562. [Google Scholar] [CrossRef] [PubMed]
  27. Guo, Q.; Goldenberg, J.Z.; Humphrey, C.; El Dib, R.; Johnston, B.C. Probiotics for the prevention of pediatric antibiotic-associated diarrhea. Cochrane Database Syst. Rev. 2019, 4, Cd004827. [Google Scholar] [CrossRef] [PubMed]
  28. Zhang, T.; Zhang, C.; Zhang, J.; Sun, F.; Duan, L. Efficacy of Probiotics for Irritable Bowel Syndrome: A Systematic Review and Network Meta-Analysis. Front. Cell. Infect. Microbiol. 2022, 12, 859967. [Google Scholar] [CrossRef] [PubMed]
  29. Li, M.; Ding, J.; Stanton, C.; Ross, R.P.; Zhao, J.; Yang, B.; Chen, W. Bifidobacterium longum subsp. infantis FJSYZ1M3 ameliorates DSS-induced colitis by maintaining the intestinal barrier, regulating inflammatory cytokines, and modifying gut microbiota. Food Funct. 2023, 14, 354–368. [Google Scholar] [CrossRef]
  30. Li, Y.; Hintze, K.J.; Ward, R.E. Effect of supplemental prebiotics, probiotics and bioactive proteins on the microbiome composition and fecal calprotectin in C57BL6/j mice. Biochimie 2021, 185, 43–52. [Google Scholar] [CrossRef]
  31. Fornai, M.; Pellegrini, C.; Benvenuti, L.; Tirotta, E.; Gentile, D.; Natale, G.; Ryskalin, L.; Colucci, R.; Piccoli, E.; Ghelardi, E.; et al. Protective effects of the combination Bifidobacterium longum plus lactoferrin against NSAID-induced enteropathy. Nutrition 2020, 70, 110583. [Google Scholar] [CrossRef] [PubMed]
  32. Naghibi, M.; Pont-Beltran, A.; Lamelas, A.; Llobregat, L.; Martinez-Blanch, J.F.; Rojas, A.; Álvarez, B.; López Plaza, B.; Arcos Castellanos, L.; Chenoll, E.; et al. Effect of Postbiotic Bifidobacterium longum CECT 7347 on Gastrointestinal Symptoms, Serum Biochemistry, and Intestinal Microbiota in Healthy Adults: A Randomised, Parallel, Double-Blind, Placebo-Controlled Pilot Study. Nutrients 2024, 16, 3952. [Google Scholar] [CrossRef]
  33. Viljanen, M.; Kuitunen, M.; Haahtela, T.; Juntunen-Backman, K.; Korpela, R.; Savilahti, E. Probiotic effects on faecal inflammatory markers and on faecal IgA in food allergic atopic eczema/dermatitis syndrome infants. Pediatr. Allergy Immunol. Off. Publ. Eur. Soc. Pediatr. Allergy Immunol. 2005, 16, 65–71. [Google Scholar] [CrossRef] [PubMed]
  34. Oswari, H.; Prayitno, L.; Dwipoerwantoro, P.G.; Firmansyah, A.; Makrides, M.; Lawley, B.; Kuhn-Sherlock, B.; Cleghorn, G.; Tannock, G.W. Comparison of stool microbiota compositions, stool alpha1-antitrypsin and calprotectin concentrations, and diarrhoeal morbidity of Indonesian infants fed breast milk or probiotic/prebiotic-supplemented formula. J. Paediatr. Child Health 2013, 49, 1032–1039. [Google Scholar] [CrossRef] [PubMed]
  35. El-Saied, S.; Amar, A.; Kaplan, D.M.; Shitrit, R.; Kaminer, B.M.; Keshet, A.; Lewis, E.C. Local Alpha1-Antitrypsin Accelerates the Healing of Tympanic Membrane Perforation in Mice. Laryngoscope 2024, 134, 3802–3806. [Google Scholar] [CrossRef]
  36. Schukfeh, N.; Sivaraman, K.; Schmidt, A.; Vieten, G.; Dingemann, J.; Weidner, J.; Olmer, R.; Janciauskiene, S. Alpha-1-antitrypsin improves anastomotic healing in intestinal epithelial cells model. Pediatr. Surg. Int. 2024, 40, 258. [Google Scholar] [CrossRef]
Figure 1. Experimental Procedure.
Figure 1. Experimental Procedure.
Nutrients 17 02127 g001
Figure 2. Growth and Development Z Scores in Toddlers (estimated marginal means and contrasts): (a) The height-for-age Z score (HAZ), (b) The weight-for-age Z score (WAZ), (c) The body-mass-index-for-age Z score (BAZ), and (d) The weight-for-height Z score (WHZ).
Figure 2. Growth and Development Z Scores in Toddlers (estimated marginal means and contrasts): (a) The height-for-age Z score (HAZ), (b) The weight-for-age Z score (WAZ), (c) The body-mass-index-for-age Z score (BAZ), and (d) The weight-for-height Z score (WHZ).
Nutrients 17 02127 g002
Figure 3. Stool Frequency and Consistency in Toddlers (estimated marginal means and contrasts): (a) Stool frequency per day, (b) Bristol Stool Scale scores.
Figure 3. Stool Frequency and Consistency in Toddlers (estimated marginal means and contrasts): (a) Stool frequency per day, (b) Bristol Stool Scale scores.
Nutrients 17 02127 g003
Table 1. Effect of YLGB-1496 on Growth and Development Z Scores in Toddlers (Mean ± SD).
Table 1. Effect of YLGB-1496 on Growth and Development Z Scores in Toddlers (Mean ± SD).
VariablesControlYLGB-1496aModel b c
WeekGroupWeek × Group
HAZ
Baseline−0.81 ± 2.78−0.14 ± 2.490.18410.0070.1650.820
6 weeks−1.00 ± 2.91−0.14 ± 2.480.09620.0070.075
12 weeks−0.77 ± 3.010.20 ± 2.580.06930.0070.006
WAZ
Baseline−0.63 ± 1.54−0.20 ± 1.480.13510.1940.1620.029
6 weeks−0.66 ± 1.37−0.35 ± 1.380.23020.1940.243
12 weeks−0.80 ± 1.35−0.17 ± 1.240.01030.1940.329
BAZ
Baseline−0.08 ± 2.12−0.58 ± 2.660.26710.3820.5140.068
6 weeks0.09 ± 2.36−0.64 ± 2.520.11620.3790.414
12 weeks−0.33 ± 2.55−0.63 ± 2.460.52430.3800.194
WHZ
Baseline−0.20 ± 1.89−0.23 ± 1.950.93710.1260.6200.097
6 weeks−0.07 ± 2.00−0.39 ± 1.920.39720.1260.530
12 weeks−0.44 ± 2.14−0.34 ± 1.840.78130.1260.293
SD: standard deviation; HAZ: height-for-age Z score; WAZ: weight-for-age Z score; WHZ: weight-for-height Z score; and BAZ: body-mass-index-for-age Z score. a p value obtained via Student’s t-test. b Model 1: adjusted for baseline values, time, group, and interaction terms comprising time and group; Model 2: additionally adjusted for delivery and primary caregiver to correct for differences between groups; Model 3: additionally adjusted for breastmilk and vegetable intake during the intervention. c p values for the intervention were obtained from the linear mixed model.
Table 2. Effect of YLGB-1496 on Defecation Frequency and Stool Consistency in Study Participants (Mean ± SD).
Table 2. Effect of YLGB-1496 on Defecation Frequency and Stool Consistency in Study Participants (Mean ± SD).
VariablesControlYLGB-1496aModel bc
WeekGroupWeek × Group
Defecation frequency (times/day)
Baseline1.08 ± 0.821.37 ± 0.960.08210.1940.2530.110
6 weeks1.01 ± 0.721.34 ± 0.830.02620.1940.448
12 weeks1.02 ± 0.711.25 ± 0.790.11530.1940.211
Bristol Stool Scale scores
Baseline4.07 ± 1.023.92 ± 0.850.37710.2140.0180.942
6 weeks4.07 ± 0.943.74 ± 0.510.02120.2140.073
12 weeks4.13 ± 1.023.79 ± 0.470.02430.2140.232
SD: standard deviation. a p value obtained via Student’s t-test. b Model 1: adjusted for baseline values, time, group, and interaction terms comprising time and group; Model 2: additionally adjusted for delivery and primary caregiver to correct for differences between groups; Model 3: additionally adjusted for breastmilk and vegetable intake during the intervention. c p values for the intervention were obtained from the linear mixed model.
Table 3. Incidence of Gastrointestinal Symptoms during the Intervention.
Table 3. Incidence of Gastrointestinal Symptoms during the Intervention.
VariablesWeeksControlYLGB-1496IRR (95% CI)a
NIR (SE)NIR (SE)
Poor appetite0430.754 (0.165)300.517 (0.128)0.686 (0.426, 1.088)0.113
6430.754 (0.165)60.103 (0.053)0.137 (0.052, 0.298)<0.001
12420.737 (0.165)00 (0)————
Nausea0150.263 (0.073)130.224 (0.065)0.852 (0.399, 1.793)0.672
6140.246 (0.058)40.069 (0.042)0.281 (0.080, 0.783)0.025
12130.228 (0.056)00 (0)————
Vomiting0180.316 (0.076)190.328 (0.131)1.037 (0.542, 1.991)0.911
6160.281 (0.060)60.103 (0.047)0.369 (0.132, 0.896)0.037
12150.263 (0.059)20.034 (0.024)0.131 (0.021, 0.464)0.007
Stomachache0230.404 (0.082)240.414 (0.085)1.025 (0.577, 1.826)0.931
6220.386 (0.078)90.155 (0.064)0.402 (0.176, 0.845)0.021
12200.351 (0.077)20.034 (0.024)0.098 (0.016, 0.336)0.002
Anal discomfort0220.386 (0.105)140.241 (0.079)0.625 (0.313, 1.210)0.170
6240.421 (0.109)20.034 (0.024)0.082 (0.013, 0.276)0.001
12240.421 (0.109)00 (0)————
Diarrhea0480.842 (0.203)370.638 (0.125)0.758 (0.491, 1.160)0.204
6631.105 (0.237)150.259 (0.100)0.234 (0.128, 0.399)<0.001
12530.930 (0.222)70.121 (0.061)0.130 (0.054, 0.267)<0.001
Dehydration0420.737 (0.165)190.328 (0.111)0.445 (0.253, 0.753)0.003
6420.737 (0.165)30.052 (0.038)0.070 (0.017, 0.193)<0.001
12410.719 (0.166)00 (0)————
IR: incidence rate; SE: standard error; IRR: incidence rate ratio; CI: confidence interval. a p value obtained via Poisson regression.
Table 4. Generalized Mixed-Effects Model Analysis of the Impact of B. infantis on Gastrointestinal Symptoms (p values).
Table 4. Generalized Mixed-Effects Model Analysis of the Impact of B. infantis on Gastrointestinal Symptoms (p values).
Poor AppetiteNauseaVomitingStomachacheAnal DiscomfortDiarrheaDehydration
Model 1
week0.4630.3710.4250.2190.7770.1260.666
group<0.0010.0050.0020.001<0.001<0.0010.001
week × group1.0000.9990.2460.0940.9990.2330.910
Model 2
week0.4640.3710.4250.2530.7770.1260.666
group<0.001<0.0010.0040.0070.0010.001<0.001
Model 3
week0.4640.3710.4890.1820.7770.1300.666
group<0.001<0.0010.0160.0100.0040.001<0.001
Model 1: adjusted for baseline values, time, group, and interaction terms comprising time and group. Model 2: additionally adjusted for delivery and primary caregiver to correct for differences between groups. Model 3: additionally adjusted for breastmilk and vegetable intake during the intervention.
Table 5. Effect of YLGB-1496 on Biochemical Parameters in Stool Samples from Study Participants (Mean ± SD).
Table 5. Effect of YLGB-1496 on Biochemical Parameters in Stool Samples from Study Participants (Mean ± SD).
VariablesControlYLGB-1496aModel bc
WeekGroupWeek × Group
pH
06.26 ± 1.316.70 ± 1.150.06610.4760.0720.246
66.54 ± 1.197.06 ± 0.910.00920.4760.117
126.58 ± 1.096.87 ± 0.780.10030.4760.263
FC (mg/g)
01.30 ± 0.711.33 ± 0.590.77610.1230.1370.595
61.02 ± 0.741.48 ± 0.850.00320.1220.114
121.42 ± 2.751.67 ± 1.440.53730.1220.225
AAT (mg/g)
00.28 ± 1.030.41 ± 0.470.39910.251<0.0010.580
60.09 ± 0.110.42 ± 0.760.00120.251<0.001
120.11 ± 0.120.48 ± 0.62<0.00130.2510.008
SD: standard deviation, FC: fecal calprotectin, AAT: α1-antitrypsin. a p value obtained via Student’s t-test. b Model 1: adjusted for baseline values, time, group, and interaction terms comprising time and group; Model 2: additionally adjusted for delivery and primary caregiver to correct for differences between groups; Model 3: additionally adjusted for breastmilk and vegetable intake during the intervention. c p values for the intervention were obtained from the linear mixed model.
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

Li, P.; Uma Mageswary, M.; Taib, F.; Koo, T.H.; Yusof, A.; Hamid, I.J.A.; Jiang, H.; Liong, M.-T.; Ali, A.; Zhang, Y. Safety and Tolerance of Bifidobacterium longum subsp. Infantis YLGB-1496 in Toddlers with Respiratory Symptoms. Nutrients 2025, 17, 2127. https://doi.org/10.3390/nu17132127

AMA Style

Li P, Uma Mageswary M, Taib F, Koo TH, Yusof A, Hamid IJA, Jiang H, Liong M-T, Ali A, Zhang Y. Safety and Tolerance of Bifidobacterium longum subsp. Infantis YLGB-1496 in Toddlers with Respiratory Symptoms. Nutrients. 2025; 17(13):2127. https://doi.org/10.3390/nu17132127

Chicago/Turabian Style

Li, Pin, Mageswaran Uma Mageswary, Fahisham Taib, Thai Hau Koo, Azianey Yusof, Intan Juliana Abd Hamid, Hua Jiang, Min-Tze Liong, Adli Ali, and Yumei Zhang. 2025. "Safety and Tolerance of Bifidobacterium longum subsp. Infantis YLGB-1496 in Toddlers with Respiratory Symptoms" Nutrients 17, no. 13: 2127. https://doi.org/10.3390/nu17132127

APA Style

Li, P., Uma Mageswary, M., Taib, F., Koo, T. H., Yusof, A., Hamid, I. J. A., Jiang, H., Liong, M.-T., Ali, A., & Zhang, Y. (2025). Safety and Tolerance of Bifidobacterium longum subsp. Infantis YLGB-1496 in Toddlers with Respiratory Symptoms. Nutrients, 17(13), 2127. https://doi.org/10.3390/nu17132127

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