1,944
Views
0
CrossRef citations to date
0
Altmetric
Review

Microbiome-targeting therapies in the neonatal intensive care unit: safety and efficacy

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Article: 2221758 | Received 06 Dec 2022, Accepted 25 May 2023, Published online: 26 Jun 2023

ABSTRACT

Microbiome-targeting therapies have received great attention as approaches to prevent disease in infants born preterm, but their safety and efficacy remain uncertain. Here we summarize the existing literature, focusing on recent meta-analyses and systematic reviews that evaluate the performance of probiotics, prebiotics, and/or synbiotics in clinical trials and studies, emphasizing interventions for which the primary or secondary outcomes were prevention of necrotizing enterocolitis, late-onset sepsis, feeding intolerance, and/or reduction in hospitalization length or all-cause mortality. Current evidence suggests that probiotics and prebiotics are largely safe but conclusions regarding their effectiveness in the neonatal intensive care unit have been mixed. To address this ambiguity, we evaluated publications that collectively support benefits of probiotics with moderate to high certainty evidence in a recent comprehensive network meta-analysis, highlighting limitations in these trials that make it difficult to support with confidence the routine, universal administration of probiotics to preterm infants.

Introduction

The gut microbiome is a dynamic community that is seeded during infancy, and which trains the immune system and supports healthy growth and development of its host.Citation1 Microbial assembly in the infant gut is influenced by numerous factors, including the maternal microbiota, delivery mode (cesarean vs. vaginal birth), diet (breastfeeding vs. formula feeding), and postmenstrual age at birth. Infants born preterm (gestational age (GA) <37 weeks) have an immature intestine characterized by deficient mucosal immunity and often experience perturbations that interfere with microbial community maturation.Citation2 Specifically, preterm infants are typically hospitalized for prolonged periods in the neonatal intensive care unit (NICU), experience variable durations and intensities of enteral feeding and maternal milk consumption, are exposed to frequent courses of antibiotics, and reside in microbially controlled environments.Citation3 Early-life hospitalization of preterm infants has been associated with persistent enrichment of antibiotic resistance genes in the gut and altered community profiles.Citation4 Compared to age-matched infants born after full-term gestations, infants born preterm have lower gut microbial richness and diversity,Citation4 which may impact intestinal development and increase the risk of immune and metabolic diseases later in life.Citation2 Gut microbial community composition and diversity are also associated with several pathologies and comorbidities afflicting preterm infants.Citation2 Gut microbiome modulation is, therefore, an appealing approach to preventing disease in infants born preterm and a strategy that is particularly timely given our expanding insights into human gut microbial communities.

Neonates have morbidity and mortality rates proportional to the degree of prematurity at birth.Citation5 Infants born after shorter gestations (i.e., <32 weeks) are especially vulnerable to necrotizing enterocolitis (NEC), a devastating necroinflammatory event that affects ~ 7% of very low birthweight (VLBW) infants (<1,500 g at birth) and has a mortality rate approximating 25%.Citation6 NEC often requires surgery to remove nonviable bowel, and survivors are prone to lifelong complications such as short-bowel syndrome and impaired neurodevelopment.Citation7 The precise etiology of NEC is unknown, but signatures of antecedent aberrant pre-NEC gut microbial communities prompt speculation that gut microbial populations could initiate organ injury.Citation8,Citation9 Preterm birth is also a risk factor for late-onset sepsis (LOS), which develops after the first 72 hours of life and affects > 35% of extremely preterm infants (i.e., those born at <28 weeks GA).Citation10 Preterm infants have greater intestinal permeability than term infants,Citation11 which may facilitate translocation of enteric bacteria into the bloodstream. Indeed, data demonstrate that bloodstream pathogens can be cultured from stool prior to systemic invasion,Citation12 whereas the presence of Bifidobacterium in the preterm gut is associated with protection from LOS.Citation13 Given these findings, the infant gut microbiome has been proposed as a therapeutic target for the prevention of NEC and LOS. Finally, preterm infants commonly experience feeding intolerance (FI), defined as an inability to digest enteral feeds and which delays progression to full enteral feeding, prolonging hospitalization.Citation14 As gut bacteria affect intestinal homeostasis, microbiome-directed therapies may also present opportunities for improved nutritional support within NICUs.

While neonatal care has advanced significantly, convincing evidence in support of therapies that reduce morbidity and mortality by manipulating the gut microbiome composition or function in this vulnerable population remains elusive. Probiotics and prebiotics are microbiome-targeting therapeutics that have attracted intense research in neonatology in recent years.Citation15 However, altering the composition and function of the microbiome, while appealing, must be approached with caution. In this review, we provide a landscape analysis of the current state of the field, focusing on meta-analyses and systematic reviews published between 2017 and 2022 that have evaluated the performance of probiotics and prebiotics in clinical trials and studies. We study three different interventions to alter gut microbial content and/or function (probiotics, prebiotics, and synbiotics), and focus on studies in which the primary or secondary outcomes were prevention of NEC, LOS, FI, and/or reduction in hospitalization length or all-cause mortality.

Probiotics

Background

Probiotics are defined as live microorganisms which, when administered in adequate amounts, confer health benefit(s) on the host.Citation16 For vulnerable preterm infants, probiotics include bacteria and, to a lesser extent, yeast, that have been associated with healthy intestinal development and immune function. Bifidobacteria and lactobacilli have been the most studied, followed by Streptococcus thermophilus and the yeast Saccharomyces boulardii.Citation15

Probiotic bacteria ferment indigestible carbohydrates such as human milk oligosaccharides (HMOs) abundant in human milk into short-chain fatty acids (SCFAs), which have beneficial effects on gut health.Citation17 Some bifidobacteria are especially robust producers of acetate, which enhances gut barrier function and lowers luminal pH, general attributes of a healthy gut.Citation18,Citation19 Other commensals convert acetate and lactate produced by lactobacilli and bifidobacteria into butyrate, which fuels colonocytes and is considered anti-inflammatory.Citation20–22 In vitro studies demonstrate that some probiotics inhibit pathogen adherence to intestinal epithelial cellsCitation23,Citation24 and may have beneficial immunomodulatory (i.e., anti-inflammatory) properties.Citation25 Several trials have reported that preterm infants receiving probiotic supplementation have increased gut microbiome diversity and/or reduced abundance of disease-associated pathobionts, presumably via competitive exclusion.Citation19,Citation26–29

Given these theoretically beneficial properties, the concept of using probiotics to support healthy infant development and prevent disease is attractive. Indeed, many studies have been performed to determine if probiotics improve clinical outcomes in preterm infants, though considerable heterogeneity exists between patient cohorts (e.g., average GA and birthweight (BW)) and probiotic formulations used in these trials, a challenge that has been emphasized in recent network meta-analyses.Citation30–32 In the following section, we analyze the existing evidence for the effect of probiotic administration on disease outcomes among preterm infants, highlighting consensus recommendations and safety considerations. In our review, we prioritize randomized controlled trials (RCTs).

Single- versus multiple-strain probiotics in infants born preterm – general considerations

It remains unclear how multi-strain probiotics may exhibit additive or synergistic effects that outperform single-strain probiotics.Citation33 Additionally, some mechanisms, including HMO utilization, are strain-specific, which has implications for the selection of probiotic strains for clinical use in the NICU.Citation34,Citation35 Bifidobacterium species (spp.) have been the most common single-strain probiotic intervention tested, and combined Bifidobacterium spp. and Lactobacillus spp. comprise the most common multi-strain formulations.Citation36 Most single and combination probiotics have been studied few times in preterm infants, complicating comparisons of their efficacy between trials; indeed, a 2018 network meta-analysis found that a minority of the 25 studied probiotic strains or strain combinations showed any effect in reducing mortality or morbidity.Citation30 Although some studies report no difference between the effects of single- and multi-strain mixtures,Citation37,Citation38 the consensus favors multi-strain formulations,Citation31,Citation32,Citation39–41 with specific outcomes discussed in further detail below.

Necrotizing enterocolitis (NEC)

The first cohort study investigating the effects of probiotics on NEC and NEC-related death in preterm infants was conducted in 1999 in Colombia by Hoyos et al.Citation42 In a pre-post analysis of 1,237 newborns, the administration of a combination of organisms (250 million live L. acidophilus and 250 million B. longum subspecies (subsp.) infantis (B. infantis) per dose) was associated with significantly decreased rates of NEC and NEC-related mortality compared to the prior year. Many subsequent RCTs, observational studies, and meta-analyses that combine these study results have tried to demonstrate whether probiotics prevent severe NEC (Bell stage ≥ IICitation43) and associated mortality in preterm infants. A 2020 Cochrane Database systematic review by Sharif et al. reported results in favor of probiotic treatment in VLBW infants, calculating a reduced risk of NEC for trials at low risk of bias (16 trials; N = 4,597; risk ratio (RR): 0.70; 95% confidence interval (CI): 0.55, 0.89; study heterogeneity (I2)) 25%).Citation36 Similar findings were reported in a 2017 meta-analysis, which concluded that probiotics resulted in a 45% reduction in NEC incidence in VLBW infants (25 RCTs; N = 8,492; 95% CI: 0.43, 0.70; p < 0.001).Citation44 A combination of Lactobacillus spp. and Bifidobacterium spp. showed the strongest evidence of effect against NEC (RR: 0.36; 95% CI: 0.23, 0.59) with no heterogeneity in meta-analysis (I2 = 0)Citation36. This finding was replicated in a 2020 meta-analysis conducted by Morgan et al. (odds ratio (OR): 0.35; 95% CI: 0.20, 0.59; I2 = 0).Citation31 Despite such evidence of significantly reduced NEC incidence following probiotic administration, most benefit accrues to infants with BW > 1,000 g.Citation31,Citation40,Citation45–50 For instance, in a large, well-conducted RCT (ProPrem trial, N = 1,099) including VLBW infants with GA at birth <32 weeks, administration of a daily probiotic combination (B. infantis BB−02 300, S. thermophilus TH−4 350, and B. lactis BB−12 350, containing 1 × 109 total organisms) significantly reduced rates of NEC (RR: 0.46; 95% CI: 0.23, 0.93; p = 0.03).Citation45 However, a subgroup analysis of extremely low BW (ELBW) infants (<1,000 g) revealed that probiotics did not reduce NEC rates (RR: 0.73), with a non-significant interaction between probiotic treatment and GA or BW subgroup determined by logistic regression (p = 0.08).

However, probiotics have not uniformly reduced NEC rates in RCTs. The Probiotics for Preterm Infants (PiPS) trial, the largest probiotic RCT (N = 1,315) conducted to date to prevent NEC, allocated infants born between 23- and 30-weeks GA to receive B. breve BBG−001 (6.7 × 107 to 6.7 × 109 colony-forming units (CFUs)) or placebo. No difference in the primary outcomes of NEC or mortality was discerned.Citation51 Two observational studies even reported that probiotics were associated with a higher incidence of NEC, though both studies used historical controls prior to routine probiotic use at a single center.Citation52,Citation53 Furthermore, as mentioned above, there is a paucity of safety and efficacy data for probiotics in preventing NEC in ELBW infants, the subgroup at highest risk for NEC and other prematurity-related disorders.Citation47,Citation54 Though some studies indicate significant reductions in NEC in probiotic-treated ELBW infants,Citation46,Citation55 most conclude that probiotics do not prevent NEC in this lowest BW subgroup,Citation36,Citation45,Citation47,Citation54,Citation56,Citation57 as in the rigorous ProPrem trial described above.Citation45 Sharif et al. amalgamated these studies after a subgroup analysis focusing on ELBW infants and arrived at the same conclusion (N = 1,712; RR: 0.90; 95% CI: 0.68, 1.21; low certainty of evidence).Citation36 The less developed gut physiology as well as higher rates and duration of antibiotic exposure in ELBW infants possibly contribute to differences in observed efficacy according to BW.Citation2,Citation58

Inconsistent case definitions of NEC among probiotic trials might have contributed to the non-uniform results.Citation59,Citation60 Most papers specify the case definition as Bell stage ≥ II,Citation43 and do not include spontaneous intestinal perforation or Bell stage I cases. However, not all of them use the same Bell stage definition or apply additional clarification of Bell clinical diagnostic criteria such as the Vermont Oxford Network (VON) or the Centers for Disease Control and Prevention (CDC) definition.Citation59,Citation60 To ensure comparability and generalizability of study results, there is a need for a more consistent NEC case definition. Case fatality rates may be one metric that can indirectly address sensitivity and specificity of definitions used by relating NEC severity between studies.

Late-onset sepsis (LOS)

Several studies and systematic reviews report that probiotics significantly reduce LOS rates in infants born preterm or with low BW (<2,500 g),Citation55,Citation61 including VLBW infants.Citation46,Citation53,Citation54,Citation62 For example, Sharif et al.Citation36 reported that probiotics probably reduce LOS (N = 9,762; RR: 0.89; 95% CI: 0.82, 0.97; moderate certainty of evidence) in VLBW infants. Most, however, report no significant reduction of LOS after probiotic supplementation in infants born preterm.Citation31,Citation36,Citation45,Citation55,Citation63–66 Specifically, the large ProPremCitation45 and PiPSCitation51 studies reported no difference in rates of LOS between the probiotic and control groups.Citation67 Such findings in large and rigorous RCTs suggest that probiotic prophylaxis does not consistently prevent LOS in infants born preterm. Despite this lack of apparent efficacy, it is notable that probiotics do not considerably increase sepsis risk in the NICU, a safety concern given the increased intestinal permeability observed in preterm infantsCitation11; reports of probiotic-induced bloodstream infection are rare and respond well to antimicrobials,Citation68 though they highlight the need for active surveillance in probiotic trials.

Feeding intolerance (FI)

Probiotics have demonstrated mixed efficacy in improving feeding intolerance (FI) in preterm infants. A recent RCT found that a multi-strain probiotic supplementation of five Lactobacillus and Bifidobacterium spp. strains (B. breve HA−129 (1.2 billion CFUs), B. bifidum HA−132 (800 million CFUs), B. longum subsp. infantis HA−116 (600 million CFUs), B. longum subsp. longum HA−135 (400 million CFUs), and L. rhamnosus HA−111 (1.0 billion CFUs)) in 62 ELBW infants significantly decreased time to reach full enteral feeds compared to controls.Citation56 A recent meta-analysis of nine studies and over 1,200 infants born preterm reports that probiotics significantly reduced the incidence of FI and other related outcomes.Citation69 However, other studies,Citation44,Citation49,Citation66 including a 2021 meta-analysis,Citation55 found no such benefit.

The protean features of FI include abdominal distension, blood in stool, vomiting, bilious aspirate, time to reach full enteral feeds, and gastric residual volume. Significant variability in case definitions of FI makes it difficult to interpret the efficacy of interventions targeting this clinical entity.Citation70,Citation71 A recent systematic review of current definitions used to diagnose FI identifies multiple different combinations and descriptions of these variables.Citation70 There is a clear need for a standardized definition of FI to gain clarity about the value of probiotics in RCTs that attempt to prevent or reduce the frequency of this clinical entity.

All-cause mortality

Lastly, we summarize the effects of probiotics on all-cause mortality, a categorical and important outcome. A 2017 meta-analysis reported results in favor of probiotic treatment in VLBW infants, calculating 22% reductions in mortality (21 RCTs; N = 7,332; 95% CI: 0.66%, 0.93%; p = 0.01).Citation44 At the species level, Morgan et al.Citation31 reported that a combination of 1 or more Lactobacillus spp. and 1 or more Bifidobacterium spp. reduced all-cause mortality (52 studies; N = 14,003; OR: 0.56; 95% CI: 0.39, 0.80; risk difference (RD): −2.2%; 95% CI: −3.1, −0.1; high certainty of evidence), as discussed in detail below. Consistent with these findings, at the strain level, van den Akker et al.Citation30 found that 3/25 of their tested strain combinations showed significant reductions in mortality rates: B. bifidum NCDO 1453 and L. acidophilus NCDO 1748 (two studies, N = 494); B. bifidum, B. infantis, B. longum, and L. acidophilus (one study, N = 186); and B. infantis, L. acidophilus, L. casei, L. plantarum, L. rhamnosus, and S. thermophilus (one study, N = 150). While these results are encouraging, they underscore the fact that not all probiotic formulations are equivalent, and robust evidence is needed at the strain-level to support evidence-based clinical recommendations of probiotic treatment for preterm infants.

Safety

Probiotics in the U.S. are considered dietary supplements and therefore are not subject to the degree of oversight that the Food and Drug Administration (FDA)Citation72 provides to drugs. In 2015, a premature infant died of a rare fungal infection after being administered a probiotic contaminated with Rhizopus oryzae,Citation73 raising concerns over possible contamination during manufacturing.Citation72 In another study, only one of 16 commercial probiotics matched its label organism for organism.Citation74 For this reason, it is advised that probiotics be evaluated for purity and viability before administration and again during a trial, but this practice has not been universally applied.Citation75 Also, there are few assessments of safety beyond the administration period, though this is a general limitation of many therapeutic interventions.

Ensuring product purity is especially important for preterm infants whose compromised immune systems render them susceptible to outgrowth of even commensal bacterial species.Citation76,Citation77 In a recent systematic review of probiotic sepsis in preterm infants including reports published through January 2022, Kulkarni et al. found 16 studies with 32 total cases of probiotic-related sepsis, of which 25 cases were confirmed to be caused by the administered probiotic strain after full genomic analysis.Citation68 Further, they note that while there were no episodes of probiotic sepsis reported in the 2020 network meta-analysis by Morgan et al. (63 RCTs, N = 15,712),Citation31 only 12 RCTs conducted active and extended surveillance for such an event.Citation68 Many probiotics require anaerobic growth conditions, and that requires extra blood, incubated appropriately, to identify extra-intestinal dissemination accurately. Thus, although some studies have reported adverse event rates (most commonly bloodstream infections with the probiotic strain,Citation77–79) uniform large-scale monitoring of such events and long-term effects are needed to evaluate the safety of the intervention more thoroughly.Citation64 Lastly, commercial probiotics can carry transferable antibiotic resistance genes (ARGs),Citation80 which was linked to a vancomycin-resistant Enterococcus outbreak among probiotic-treated VLBW infants in a Turkish NICU.Citation81 As preterm infants receive frequent courses of antibiotics that apply selective pressure to commensal microbes and are at particularly high risk of infection,Citation3 care should be taken to select probiotic strains that minimize the potential of ARG dissemination to the existing microbiome.

Limitations to published studies of probiotics in preterm

Infants

Marked heterogeneity among studies prompts caution when endorsing probiotics as standard of care for premature infants.Citation47,Citation72,Citation82,Citation83 Studies often evaluate different probiotic species and strains that can have quite different theoretical efficacy profiles, thereby lending bias to outcome measurements.Citation84 Differences in probiotic dose, length of treatment, and viability may further contribute to conflicting findings.Citation36,Citation47 For example, in their network meta-analysis, Morgan et al. report substantial heterogeneity (I2 = 53.6%) among studies comparing the effect of Bifidobacterium spp. and Streptococcus salivarius subsp. thermophilus with placebo on NEC.Citation31 Substantial heterogeneity (I2 > 50%) was also reported for the comparisons of L reuteri; Lactobacillus spp., Bifidobacterium spp., and Saccharomyces boulardii; Lactobacillus spp., Bifidobacterium spp., and Enterococcus spp.; Lactobacillus spp. and Bifidobacterium spp.; and Bifidobacterium spp. and S. salivarius subsp. thermophilus vs. placebo on culture-proven LOS. It is worth noting, however, that the reported statistical heterogeneity was zero (I2 = 0) for studies that evaluated the effect of the probiotic combination of Lactobacillus spp. and Bifidobacterium spp., which has shown the most evidence of having a protective effect against NEC based on GRADE certainty of evidence.Citation31,Citation36 This suggests that at least for the combination of Lactobacillus spp. and Bifidobacterium spp. and NEC, there is low heterogeneity between trial results.

Several studies reported colonization of placebo infants which may have confounded negative results.Citation51,Citation85,Citation86 For example, the PiPS trial is one of the few RCTs that monitored rates of cross-contamination between the probiotic (B. breve BBG−001) and placebo arm. The authors report that B. breve BBG−001 was detected by culture in 49% of infants in the placebo group by 36 weeks postmenstrual age despite measures to minimize the possibility of cross-contamination such as cleaning all working surfaces after completing intervention preparations for each baby.Citation51 This underscores the need for more active surveillance for cross-colonization in probiotic RCTs.

Current recommendations

It is notable that professional societies have recently issued conflicting guidance regarding probiotic prophylaxis in preterm infants. While the American Gastroenterological Association (AGA)Citation87 and World Health Organization (WHO)Citation88 have given conditional recommendations for the use of probiotics in preterm infants based on moderate/high certainty evidence, the European Society for Paediatric Gastroenterology, Hepatology, and Nutrition (ESPGHAN) has taken an intermediate stance,Citation89 and the American Academy of Pediatrics (AAP) cautioned against the routine, universal use of probiotics in the NICU, especially for ELBW infants.Citation47

The new 2022 WHO guidelinesCitation88 broadly recommend probiotics for human-milk-fed very preterm infants based on the Cochrane review by Sharif et al.Citation36 These recommendations are conditional upon shared decision-making with parents,Citation88 emphasizing the need for clear evidence of benefits and safety that can be communicated to families when probiotics are offered. The AGACitation87 recommend certain single- and multi-strain combinations of Lactobacillus spp. and Bifidobacterium spp. for preterm infants based on findings from the network meta-analysis by Morgan et al.Citation31 discussed in detail in a later section. The ESPGHANCitation89 conditionally recommends specific strains to reduce NEC based on low certainty of evidence, while recommending against certain strains based on lack of efficacy and safety concerns. In all cases of conditional positive recommendations, the dosage, timing, and duration of treatment are not specified due to lack of data on optimal treatment course. In contrast, the AAPCitation47 supports a more cautionary approach based on the current lack of pharmaceutical-grade probiotics, variability in tested formulations, limited long-term safety information, and less convincing findings in ELBW infants. Unless compelling data – double-blind RCTs that are adequately powered to assess effects in ELBW infants, ensure purity and viability of the multi-strain probiotic compound (Lactobacillus spp. and Bifidobacterium spp.) prior to supplementation, and closely monitor stool samples for cross-contamination – demonstrate significant benefits in predeclared primary outcomes and long-term safety profiles,Citation90 many experts will likely continue to recommend against routine administration of probiotics to preterm infants. Considering these limitations, we believe there are insufficient data to currently endorse the routine, universal administration of probiotics to preterm infants to prevent NEC, mortality, LOS, or FI.

Prebiotics

Background

Prebiotics, i.e., substrates used by host microbes to confer health benefit(s),Citation91 are another approach to targeting the preterm infant microbiome for disease prevention. Prebiotics are most commonly oligosaccharides that can be fermented by commensal bacteria to promote their colonization and growth in the intestine. Human milk provides an abundance of prebiotics in the form of HMOs.Citation92 These complex glycans resist degradation by host enzymes and arrive in the colon intact, where they serve as carbon sources for bifidobacteria (especially B. infantis), Bacteroides, and lactobacilliCitation93–96. Bacterial fermentation of HMOs produces SCFAs, which promote epithelial barrier integrity and have anti-inflammatory activity in vitro.Citation97,Citation98 Bacterial strains in the infant gut differ in their ability to ferment HMOs, and by-products generated by HMO metabolism can support the growth of other commensals through cross-feeding.Citation34,Citation93 Besides promoting the growth of beneficial bacteria, HMOs have potentially immunomodulatory properties,Citation92 and some HMO-derived oligosaccharides can prevent Group B Streptococcus growth in vitro.Citation99,Citation100 HMOs can also function as soluble decoy receptors, preventing adherence of pathogens to host cells in vitro.Citation101

The most frequently studied prebiotic substrates include short-chain (sc) galacto-oligosaccharides (GOS), long-chain (lc) fructo-oligosaccharides (FOS), lactulose, and acidic oligosaccharides (AOS).Citation102 AOS comprise 12–14% of the total HMO fraction in human milk and pectin-derived AOS (pAOS) are included in some prebiotic mixtures.Citation103,Citation104 As over 200 structurally unique HMOs have been described, commercially produced substrates cannot fully mimic the complexity of HMOs in human milk.Citation95 In turn, human milk-derived fortifiers containing donor-derived HMOs have been used to increase protein intake as part of an exclusive human milk diet.Citation95 However, these are not reviewed here as they also contain other bioactive human milk-derived compounds and evidence supporting their usage is currently limited. Plant-derived prebiotic oligosaccharides are thus low-cost alternatives to HMOs that approximate their beneficial effects. Preterm infants receiving prebiotics have increased bifidobacteria and reduced abundance of coliforms and potential pathogens in stool.Citation105–109 Considerably fewer trials have been conducted with preterm infants to evaluate the efficacy and safety of prebiotic supplements compared to probiotics, limiting the generalizability of results supporting their use. Nonetheless, prebiotics are appealing because they do not involve the administration of live bacteria, which carries a small risk of sepsis.Citation68 It is also probably easier to standardize prebiotic than probiotic product quality. In the following section, we review the findings from existing studies in preterm infants assessing the effects of prebiotics on the aforementioned outcomes.

NEC

Infants fed partially or exclusively with human milk are at decreased risk for NEC.Citation110 Among human milk’s many bioactive compounds, HMOs are thought to be beneficial by modulating endogenous gut microbes and host defenses,Citation92 and commercial prebiotics may simulate these protective effects. Armanian et al. reported that infants receiving human milk supplemented with a 9:1 scGOS/lcGOS mixture had significantly less risk of suspected, but not proven, NEC in an RCT; however, providers were not blinded to an infant’s treatment.Citation111 In contrast, four studies of preterm infants fed partially or exclusively with prebiotic-supplemented formula found no reduced NEC riskCitation112–115. A 2019 meta-analysis concluded that based on pooled effects from 6 trials, prebiotics did not significantly reduce NEC morbidity (N = 737; RR: 0.79; 95% CI: 0.44, 1.44; p = 0.44).Citation116 Taken together, current data do not favor using prebiotics to prevent NEC. While these data are perhaps surprising given the presumed protection from NEC afforded by HMOs, they underscore the need to better understand the effects of prebiotic administration on bacterial function in the already perturbed preterm gut.

LOS

Prebiotics promote commensal bacteria growth and reduce pathogen adherence in vitro.Citation34,Citation94,Citation101 However, only one RCT, the ProPre-Save study, reported a lower frequency of LOS among preterm infants who received a prebiotic.Citation114 Other RCTs showed no benefit or only a statistically insignificant trend toward efficacy.Citation111–113,Citation115,Citation117 A 2019 meta-analysis by Chi et al. including 11 RCTs reported that LOS rates were significantly less in preterm infants treated with prebiotics (N = 1,106; RR: 0.64; 95% CI: 0.51, 0.78; p < 0.001).Citation116 A subgroup analysis showed that this effect was stronger for prebiotics added to human milk (p < 0.001) rather than distilled water (p = 0.28) and for those containing pAOS (p < 0.001), which have reported antibacterial properties,Citation99 compared to those without pAOS (p = 0.13). These conflicting results highlight the need for further research to understand if, and to what extent, prebiotic supplementation reduces LOS risk in infants born preterm.

FI

Several studies have reported that prebiotic supplementation softened stool and increased defecation frequency in preterm infants.Citation105,Citation107,Citation113,Citation114,Citation118 Additionally, one RCT of preterm infants given a 9:1 GOS/FOS mixture demonstrated that treated infants had decreased gastrointestinal transit time.Citation118 However, other studies found no significant changes in stool characteristics in infants born preterm.Citation109,Citation112,Citation117 The different prebiotic compositions, feeding modes, treatment dosages, and definitions of FI likely contribute to these mixed observations. When examining FI as a primary or secondary outcome, many studies report that prebiotics are well tolerated by preterm infants but do not necessarily improve or prevent this condition,Citation113,Citation117,Citation118 though a shortened timeframe to achieving full enteral feeds has been observed in some infants.Citation111,Citation114 Interestingly, Modi et al. found a small but significant improvement in enteral tolerance among preterm infants given formula supplemented with 9:1 scGOS/lcFOS, an effect that was more pronounced at lower GA.Citation112 The meta-analysis by Chi et al. reported that prebiotics were overall associated with a shortened time to full enteral feeding (six trials; N = 576; mean difference (MD): −0.99; 95% CI: −1.15, 0.83; p < 0.001) but did not influence the degree of FI (four trials; N = 413; RR: 0.87; 95% CI: 0.52, 1.45; p = 0.6).Citation116 These conclusions warrant further study to determine if prebiotics enhance enteral tolerance, particularly in the most premature infants. However, as for probiotics, variable case definitions make it difficult to conclude that prebiotics can prevent or lessen the severity of FI.

Safety and efficacy

In adults, prebiotics can cause symptoms such as flatulence and abdominal bloating, especially at high doses.Citation119 However, prebiotics in preterm infants appear to be safe and well-tolerated, as adverse effects have not been documented. The long-term effects of prebiotic supplementation in preterm infants are also not well understood, but a follow-up study over the first year of life did not report adverse effects related to prebiotic treatment.Citation120

Concerns have been raised that prebiotics could promote the growth of enteropathogens.Citation121 However, dietary prebiotics that resemble HMOs are preferentially metabolized by bifidobacteria and lactobacilli, a process that bolsters protection from pathogens.Citation18 Given preterm infants’ especially vulnerable immune defenses, a better understanding of how prebiotics affect bacterial growth in the infant gut is needed to fully evaluate their safety within this population.

A 2019 meta-analysis suggested that prebiotics overall may benefit preterm infants by decreasing the rate of LOS and mortality and shortening the length of stay and time to reach full enteral feeds.Citation113 However, there was no significant effect on NEC incidence. Prebiotics may be low-cost interventions that benefit preterm infants and augment the effects of probiotics. Nonetheless, more trials are necessary to confirm or refute the postulated benefit of the most promising prebiotics on outcomes in infants born preterm. Until then, existing data suggest that while prebiotics are safe and well-tolerated by preterm infants, their benefits are modest at best.

Synbiotics

Synbiotics combine probiotics and prebiotics in single administrations. Their combination enhances probiotic colonization and promotes therapeutic effects. Healthy term infants treated with a combination of L. plantarum and FOS for 7 days had high colonization rates of the probiotic for several months after treatment.Citation122 Most impressively, a large RCT of 4,556 term infants in rural India receiving this synbiotic reported reduced rates of LOS and mortality in the treatment group during the first 60 days of life.Citation123 Synbiotics have been evaluated in several RCTs in preterm infants. An RCT of exclusively breastfed preterm infants (N = 220) found that a consortium of Lactobacillus and Bifidobacterium spp. strains combined with FOS significantly reduced time to full enteral feeds, and there was a trend toward reduced NEC.Citation124 However, the intervention did not lower the risk of LOS. In comparison, the ProPre-Save study of 400 VLBW infants evaluated whether administration of a probiotic (B. lactis), prebiotic (inulin, a plant-derived lcFOS), or synbiotic (B. lactis plus inulin) reduced NEC risk compared to placebo.Citation114 Infants receiving the probiotic or synbiotic had a similarly reduced NEC risk, but the prebiotic alone did not have a significant effect. A 2022 Cochrane Database systematic review found low-certainty evidence that synbiotics reduce the risk of NEC (six trials; N = 907; RR: 0.18; 95% CI: 0.09, 0.40; p < 0.0001) and all-cause mortality (six trials; N = 925; RR: 0.53; 95% CI: 0.33, 0.85; p = 0.008), and very low-certainty evidence about their effect on LOS (five trials; N = 707; RR: 0.84; 95% CI: 0.58, 1.21; p = 0.34).Citation125 More specifically, a 2021 network meta-analysis including both probiotic and synbiotic intervention trials among preterm infants found that Lactobacillus spp. plus prebiotic was associated with a lower incidence of NEC (RR: 0.06; 95% CI: 0.01, 0.41) and LOS (RR: 0.18; 95% CI: 0.06, 0.44) compared to placebo, and Bifidobacterium spp. plus prebiotic was most strongly associated with lower mortality rates (surface under the cumulative ranking curve 83.94%) of the tested interventions.Citation32 Synbiotics may offer synergy between the protection afforded by probiotics and prebiotics individually. However, more RCTs that include larger numbers of preterm infants are necessary to rigorously determine the efficacy of synbiotic formulations on preterm neonatal health and their safety profile.

Probiotic efficacy: a closer look

Professional societies recently issued differing recommendations for using probiotics in infants born preterm to prevent NEC,Citation47,Citation87–89 based on the same meta-analyses that we reviewed. Such variations in interpretations of the existing literature prompted us to review a subset of the primary literature cited on probiotics in preterm infants.

To do this, we used as a starting point the comprehensive and well-annotated network meta-analysis of Morgan et al.Citation31 These authors analyzed 63 publications reporting the use of probiotics in RCTs in which 15,712 preterm infants were enrolled. We focused on 21 of these publications (20 papers, one meeting abstract), because they were considered to provide, in the aggregate, moderate- to high-certainty evidence and statistically significant differences between the treatment and at least one other treatment and placebo in pooled analyses. These “extreme of effect” studies underlie the forest green cells representing four outcomes of interest (all-cause mortality, NEC occurrence, reduction in days to reach full feeds, and reduction in duration of hospitalization (DOH)) in Figure 2 in the meta-analysis by Morgan et alCitation31 as summarized in .

Table 1. Probiotic interventions with efficacy against selected outcomes as supported by moderate to high certainty evidence and statistically significant differences between the treatment and at least one other treatment and placebo in pooled analyses (from Figure 2 in Morgan et al.Citation31

Morgan et al. concluded (1) Lactobacillus spp. and Bifidobacterium spp. prevented all-cause mortality and NEC, (2) B. animalis subsp. lactis prevented NEC and reduced DOH, (3) L. reuteri reduced DOH, and (4) a combination of Lactobacillus spp., Bifidobacterium spp., and Saccharomyces boulardii reduced days to reach full feeds.Citation31 Two authors (AD, JR) examined the methodology and primary data in these primary papers, independently scored risk of bias in domains of sequence generation, allocation concealment, and blinding of data collectors/outcome assessors, noted if the study was registered at a public website such as ClinicalTrials.Gov or presented in a protocol paper, and assessed if the outcome referenced in Morgan et al.Citation31 was the primary goal of the study (). Clinical content experts PIT and BBW were available to adjudicate discordant assessments, but agreement on all papers reviewed by AD and JR obviated the need for resolution.

Table 2. Effects of Bifidobacterium animalis subsp. lactis on NEC risk and/or duration of hospitalization (DOH).

Table 3. Effects of Lactobacillus reuteri on duration of hospitalization (DOH).

Table 4. Effects of a combination of Lactobacillus spp., Bifidobacterium spp., and Saccharomyces boulardii on days to reach full feed.

Table 5. Effects of a combination of Lactobacillus spp. and Bifidobacterium spp. on NEC occurrence and/or all-cause mortality.

Of the 21 publications, one (Hariharan et al.Citation138) was an abstract that contained insufficient data and information to analyze. Only 12 studies relevant to the outcomes of interest were registered with clearly declared primary and secondary outcomes, so the reader can determine if the outcomes were pre-specified, or exploratory. While 17 of the 21 publications provided sufficient information to enable the reader to conclude that there was low risk of bias in treatment sequence generation and allocation concealment, only seven provided unambiguous information to conclude that there was low risk of bias by data collectors or outcome assessors. In 12 of the 21 studies, none of the outcomes of interest was a primary outcome of the study or designated either as a secondary outcome. Only six of the 21 studies met all criteria by being pre-registered and providing sufficient assurance that methods of sequence generation, allocation concealment, data collection, and outcome assessment had low risks of bias. Of these six studies, none showed a statistically significant benefit of probiotics for a primary outcome, and two showed a benefit for a secondary outcome. The papers cited in support of the value of probiotics in the five outcomes of interest enrolled their last participant a median of 11.5 years ago, and the most recent of the studies enrolled its last participant in 2016 ().

Table 6. Year at which the last participant of the study was enrolled.

Even though there is little evidence in favor of probiotics improving the five outcomes of interest, it is possible that there was benefit to probiotics despite the non-significant differences between treatment and control groups. Specifically, two sources of type II error (failing to find a benefit to probiotics when one actually exists) may have been present in many trials. First, the propensity of probiotics to colonize controls in the same NICUs raises the possibility that the non-treated group might have been inadvertently treated by nosocomial spread in some studies, thereby reducing the apparent benefit of the intervention.Citation51,Citation85,Citation86 Second, the probiotics might have lost viability, and treatment groups were not administered a bioactive intervention. However, as many studies evaluated neither for colonization of controls nor for viability of the intervention, such exculpatory interpretations remain speculative. We also note that for the prevention of NEC, a subset of studies suggests that probiotics are effective, and the effects are reproduced in children with BW > 1,000 g, a commonality that lends some biological credence to the concept that microbial therapeutics might reduce NEC risk, though regrettably the benefit is demonstrated in those who are less likely to experience this outcome. We also wish to note that the primary studies were well-intentioned attempts to prevent profound consequences of preterm birth, and some shortcomings, such as non-registration of the trials, were more common in past decades than they are currently.

Meta-analyses attempt to amalgamate data from multiple studies to detect trends that can be employed in clinical settings, but which are not apparent in smaller studies. Morgan et al.Citation31 and the other five meta-analyses we used as sources of dataCitation30,Citation32,Citation36,Citation44,Citation55 for our review were conducted to combine evidence into point estimates (with confidence intervals) of the effects of interventions combined across multiple studies. These reviews graded and reported the quality of evidence (chiefly risks of bias relative to observed magnitude of effect) according to standard criteria. However, while meta-analyses often identify limitations of primary papers, conclusions do not always convey circumspection warranted by the quality of the data. Hence, while the AGA’s recommendationCitation87 of specific combinations of probiotics “ … for prevention of NEC over no and other probiotics” seems commensurate with the reviewCitation31 on which it was based, most studies at the root of the meta-analyses, as demonstrated in , rarely met criteria for trials that would qualify probiotics interventions as drugs. The presentation of data in a graded fashion in meta-analyses to illustrate the effects of risks of bias on assessment of the certainty of the evidenceCitation145 might generate more circumspect recommendations.

Conclusions

It is understandable that gut bacterial communities be considered as targets for beneficial manipulation in infants born preterm. There is increasing evidence that variations in gut microbial makeup are associated with undesirable outcomes in these infants, and such outcomes can be devastating. In the case of probiotics, cogent arguments have been offered in support of these interventions to prevent NEC,Citation146 especially in view of the difficulties conducting large and convincing RCTs.Citation147 For centers choosing to use probiotics, the administration of a live biotherapeutic product that is not yet approved by a regulatory authority for this indication is a complex process.Citation148 However, if current standards expected of drugs are employed, the interventions discussed in this review do not yet offer sufficient evidence to recommend their routine use in preterm infants.

Acknowledgments

We would like to thank David Zahrah from the Dantas Laboratory for confirming translations from Spanish language publications. P.I.T. is a holder of equity in, a member of the Scientific Advisory Board of, and a consultant to, MediBeacon Inc., which is developing technology to measure human intestinal permeability. He is also the inventor of intellectual property that could earn royalties if this technology results in a clinical product.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

This work is supported by the National Institute of General Medical Sciences (R01 GM099538 to G. D.), the National Institute of Diabetes and Digestive and Kidney Diseases (5P30 DK052574 to P. I. T. Administrative Core), the Eunice Kennedy Shriver National Institute of Child Health and Human Development (R01 HD092414 to G. D., B. B. W., and P. I. T.), and the Children’s Discovery Institute at St Louis Children’s Hospital and Washington University School of Medicine in St Louis (B. B. W. and P. I. T.). A. D. is supported by the Cellular & Molecular Biology Training Grant (T32GM139774).

References

  • Moore RE, Townsend SD. Temporal development of the infant gut microbiome. Open Biol. 2019;9(9):190128. doi:10.1098/rsob.190128.
  • Healy DB, Ryan CA, Ross RP, Stanton C, Dempsey EM. Clinical implications of preterm infant gut microbiome development. Nature Microbiol. 2022;7(1):22–24. doi:10.1038/s41564-021-01025-4.
  • Thänert R, Sawhney SS, Schwartz DJ, Dantas G. The resistance within: antibiotic disruption of the gut microbiome and resistome dynamics in infancy. Cell Host & Microbe. 2022;30(5):675–683. doi:10.1016/j.chom.2022.03.013.
  • Gasparrini AJ, Wang B, Sun X, Kennedy EA, Hernandez-Leyva A, Ndao IM, Tarr PI, Warner BB, Dantas G. Persistent metagenomic signatures of early-life hospitalization and antibiotic treatment in the infant gut microbiota and resistome. Nature Microbiol. 2019;4:2285–2297. doi:10.1038/s41564-019-0550-2.
  • Manuck TA, Rice MM, Bailit JL, Grobman WA, Reddy UM, Wapner RJ, Thorp JM, Caritis SN, Prasad M, Tita ATN, et al. Preterm neonatal morbidity and mortality by gestational age: a contemporary cohort. Am J Obstet Gynecol. 2016;215(1):.e103.1–.e103.14. doi:10.1016/j.ajog.2016.01.004.
  • Alsaied A, Islam N, Thalib L. Global incidence of Necrotizing Enterocolitis: a systematic review and Meta-analysis. BMC Pediatr. 2020;20:344. doi:10.1186/s12887-020-02231-5.
  • Hackam DJ, Sodhi CP. Bench to bedside — new insights into the pathogenesis of necrotizing enterocolitis. Nat Rev Gastroentero. 2022;1–12. doi:10.1038/s41575-022-00594-x.
  • Warner BB, Deych E, Zhou Y, Hall-Moore C, Weinstock GM, Sodergren E, Shaikh N, Hoffmann JA, Linneman LA, Hamvas A, et al. Gut bacteria dysbiosis and necrotising enterocolitis in very low birthweight infants: a prospective case-control study. Lancet. 2016;387(10031):1928–1936. doi:10.1016/S0140-6736(16)00081-7.
  • Pammi M, Cope J, Tarr PI, Warner BB, Morrow AL, Mai V, Gregory KE, Kroll JS, McMurtry V, Ferris MJ, et al. Intestinal dysbiosis in preterm infants preceding necrotizing enterocolitis: a systematic review and meta-analysis. Microbiome. 2017;5(1):31. doi:10.1186/s40168-017-0248-8.
  • Dong Y, Speer CP. Late-onset neonatal sepsis: recent developments. Arch Dis Child Fetal Neonatal Ed. 2015;100(3):F257–63. doi:10.1136/archdischild-2014-306213.
  • Mank E, van Harskamp D, van Toledo L, van Goudoever JB, Schierbeek H. Simultaneous assessment of intestinal permeability and lactase activity in human-milk-fed preterm infants by sugar absorption test: clinical implementation and analytical method. Clin Nutr. 2021;40(3):1413–1419. doi:10.1016/j.clnu.2020.08.034.
  • Carl MA, Ndao IM, Springman AC, Manning SD, Johnson JR, Johnston BD, Burnham C-A, Weinstock ES, Weinstock GM, Wylie TN, et al. Sepsis from the Gut: the enteric habitat of bacteria that cause late-onset neonatal bloodstream infections. Clin Infect Dis: Official Publ Infect Dis Society America. 2014;58(9):1211–1218. doi:10.1093/cid/ciu084.
  • Stewart CJ, Embleton ND, Marrs ECL, Smith DP, Fofanova T, Nelson A, Skeath T, Perry JD, Petrosino JF, Berrington JE, et al. Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls. Microbiome. 2017;5(1):75. doi:10.1186/s40168-017-0295-1.
  • Fanaro S. Feeding intolerance in the preterm infant. Early Hum Dev. 2013;89:S13–20. doi:10.1016/j.earlhumdev.2013.07.013.
  • Murphy K, Ross RP, Ryan CA, Dempsey EM, Stanton C. Probiotics, prebiotics, and synbiotics for the prevention of necrotizing enterocolitis. Front Nutr. 2021;8:667188. doi:10.3389/fnut.2021.667188.
  • Food and Agricultural Organization of the United Nations and World Health Organization. Health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria [Internet]. 2001 [accessed 2023 Apr 18]: https://www.iqb.es/digestivo/pdfs/probioticos.pdf.
  • Yu Z-T, Chen C, Newburg DS. Utilization of major fucosylated and sialylated human milk oligosaccharides by isolated human gut microbes. Glycobiology. 2013;23:1281–1292. doi:10.1093/glycob/cwt065.
  • Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke JM, Topping DL, Suzuki T, et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature. 2011;469(7331):543–547. doi:10.1038/nature09646.
  • Alcon-Giner C, Dalby MJ, Caim S, Ketskemety J, Shaw A, Sim K, Lawson MAE, Kiu R, Leclaire C, Chalklen L, et al. Microbiota supplementation with bifidobacterium and lactobacillus modifies the preterm infant gut microbiota and metabolome: an observational study. Cell Rep Med. 2020;1(5):100077. doi:10.1016/j.xcrm.2020.100077.
  • Falony G, Vlachou A, Verbrugghe K, Vuyst LD. Cross-feeding between bifidobacterium longum BB536 and acetate-converting, butyrate-producing colon bacteria during growth on oligofructose. Appl Environ Microb. 2006;72(12):7835–7841. doi:10.1128/AEM.01296-06.
  • Rivière A, Gagnon M, Weckx S, Roy D, Vuyst LD, Schloss PD. Mutual cross-feeding interactions between bifidobacterium longum subsp. longum NCC2705 and eubacterium rectale ATCC 33656 explain the bifidogenic and butyrogenic effects of arabinoxylan oligosaccharides. Appl Environ Microbiol. 2015;81(22):7767–7781. doi:10.1128/AEM.02089-15.
  • Duncan SH, Louis P, Flint HJ. Lactate-utilizing bacteria, isolated from human feces, that produce butyrate as a major fermentation product. Appl Environ Microbiol. 2004;70(10):5810–5817. doi:10.1128/AEM.70.10.5810-5817.2004.
  • Mack DR, Michail S, Wei S, McDougall L, Hollingsworth MA. Probiotics inhibit enteropathogenic E. coliadherence in vitro by inducing intestinal mucin gene expression. Am J Physiol-Gastr L. 1999;276(4):G941–50. doi:10.1152/ajpgi.1999.276.4.G941.
  • Bernet MF, Brassart D, Neeser JR, Servin AL. Adhesion of human bifidobacterial strains to cultured human intestinal epithelial cells and inhibition of enteropathogen-cell interactions. Appl Environ Microbiol. 1993;59(12):4121–4128. doi:10.1128/aem.59.12.4121-4128.1993.
  • Ganguli K, Meng D, Rautava S, Lu L, Walker WA, Nanthakumar N. Probiotics prevent necrotizing enterocolitis by modulating enterocyte genes that regulate innate immune-mediated inflammation. Am J Physiol-Gastr L. 2013;304(2):G132–41. doi:10.1152/ajpgi.00142.2012.
  • Martí M, Spreckels JE, Ranasinghe PD, Wejryd E, Marchini G, Sverremark-Ekström E, Jenmalm MC, Abrahamsson T. Effects of Lactobacillus reuteri supplementation on the gut microbiota in extremely preterm infants in a randomized placebo-controlled trial. Cell Rep Med. 2021;2:100206. doi:10.1016/j.xcrm.2021.100206.
  • Manzoni P, Mostert M, Leonessa ML, Priolo C, Farina D, Monetti C, Latino MA, Gomirato G. Oral supplementation with lactobacillus casei subspecies rhamnosus prevents enteric colonization by Candida species in preterm neonates: a randomized study. Clin Infect Dis. 2006;42(12):1735–1742. doi:10.1086/504324.
  • Samara J, Moossavi S, Alshaikh B, Ortega VA, Pettersen VK, Ferdous T, Hoops SL, Soraisham A, Vayalumkal J, Dersch-Mills D, et al. Supplementation with a probiotic mixture accelerates gut microbiome maturation and reduces intestinal inflammation in extremely preterm infants. Cell Host & Microbe. 2022;30(5):696–711.e5. doi:10.1016/j.chom.2022.04.005.
  • Beck LC, Masi AC, Young GR, Vatanen T, Lamb CA, Smith R, Coxhead J, Butler A, Marsland BJ, Embleton ND, et al. Strain-specific impacts of probiotics are a significant driver of gut microbiome development in very preterm infants. Nature Microbiol. 2022:1–11. doi:10.1038/s41564-022-01213-w
  • van den Akker CHP, van Goudoever JB, Szajewska H, Embleton ND, Hojsak I, Reid D, Shamir R. ESPGHAN Working Group for Probiotics, Prebiotics & Committee on Nutrition. Probiotics for preterm infants: a strain-specific systematic review and network meta-analysis. J Pediatr Gastroenterol Nutr. 2018;67(1):103–122. doi:10.1097/MPG.0000000000001897.
  • Morgan RL, Preidis GA, Kashyap PC, Weizman AV, Sadeghirad B, Group MP Prebiotic, and Synbiotic Work, Chang Y, Florez ID, Foroutan F, Shahid S, et al. Probiotics reduce mortality and morbidity in preterm, low-birth-weight infants: a systematic review and network meta-analysis of randomized trials. Gastroenterology. 2020;159(2):467–480. doi:10.1053/j.gastro.2020.05.096.
  • Chi C, Li C, Buys N, Wang W, Yin C, Sun J. Effects of probiotics in preterm infants: a network meta-analysis. Pediatrics. 2021;147:e20200706. doi:10.1542/peds.2020-0706.
  • Ouwehand AC, Invernici MM, Furlaneto FAC, Messora MR. Effectiveness of multi-strain versus single-strain probiotics. J Clin Gastroenterol. 2018;52(Supplement 1):S35–40. doi:10.1097/MCG.0000000000001052.
  • Lawson MAE, O’Neill IJ, Kujawska M, Javvadi SG, Wijeyesekera A, Flegg Z, Chalklen L, Hall LJ. Breast milk-derived human milk oligosaccharides promote Bifidobacterium interactions within a single ecosystem. Isme J. 2020;14:635–648. doi:10.1038/s41396-019-0553-2.
  • Barratt MJ, Nuzhat S, Ahsan K, Frese SA, Arzamasov AA, Sarker SA, Islam MM, Palit P, Islam MR, Hibberd MC, et al. Bifidobacterium infantis treatment promotes weight gain in Bangladeshi infants with severe acute malnutrition. Sci Transl Med. 2022;14(640):eabk1107. doi:10.1126/scitranslmed.abk1107.
  • Sharif S, Meader N, Oddie SJ, Rojas-Reyes MX, McGuire W. Probiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Db Syst Rev. 2020;2020(10): CD005496. doi:10.1002/14651858.CD005496.pub5.
  • Athalye-Jape G, Esvaran M, Patole S, Simmer K, Nathan E, Doherty D, Keil A, Rao S, Chen L, Chandrasekaran L, et al. Effect of single versus multistrain probiotic in extremely preterm infants: a randomised trial. Bmj Open Gastroenterol. 2022;9(1):e000811. doi:10.1136/bmjgast-2021-000811.
  • Priyadarshi A, Lowe G, Saddi V, Trivedi A, Luig M, Tracy M. Clinical outcomes of single vs. two-strain probiotic prophylaxis for prevention of necrotizing enterocolitis in preterm infants. Frontiers Pediatrics. 2021;9:729535. doi:10.3389/fped.2021.729535.
  • Jiao X, Fu M-D, Wang Y-Y, Xue J, Zhang Y. Bifidobacterium and Lactobacillus for preventing necrotizing enterocolitis in very-low-birth-weight preterm infants: a systematic review and meta-analysis. World J Pediatr. 2020;16:135–142. doi:10.1007/s12519-019-00297-5.
  • Thomas JP, Raine T, Reddy S, Belteki G. Probiotics for the prevention of necrotising enterocolitis in very low‐birth‐weight infants: a meta‐analysis and systematic review. Acta Paediatr. 2017;106:1729–1741. doi:10.1111/apa.13902.
  • Chang H-Y, Chen J-H, Chang J-H, Lin H-C, Lin C-Y, Peng C-C, Hills RK. Multiple strains probiotics appear to be the most effective probiotics in the prevention of necrotizing enterocolitis and mortality: an updated meta-analysis. Plos One. 2017;12(2):e0171579. doi:10.1371/journal.pone.0171579.
  • Hoyos AB. Reduced incidence of necrotizing enterocolitis associated with enteral administration of Lactobacillus acidophilus and Bifidobacterium infantis to neonates in an intensive care unit. Int J Infect Dis. 1999;3(4):197–202. doi:10.1016/S1201-9712(99)90024-3.
  • Walsh MC, Kliegman RM. Necrotizing enterocolitis: treatment based on staging criteria. Pediatr Clin N Am. 1986;33(1):179–201. doi:10.1016/S0031-3955(16)34975-6.
  • Sun J, Marwah G, Westgarth M, Buys N, Ellwood D, Gray PH. Effects of probiotics on necrotizing enterocolitis, sepsis, intraventricular hemorrhage, mortality, length of hospital stay, and weight gain in very preterm infants: a meta-analysis. Adv Nutrition Int Rev J. 2017;8(5):749–763. doi:10.3945/an.116.014605.
  • Jacobs SE, Tobin JM, Opie GF, Donath S, Tabrizi SN, Pirotta M, Morley CJ, Garland SM, Group PS. Probiotic effects on late-onset sepsis in very preterm infants: a randomized controlled trial. Pediatrics. 2013;132(6):1055–1062. doi:10.1542/peds.2013-1339.
  • Denkel LA, Schwab F, Garten L, Geffers C, Gastmeier P, Piening B, Kollmann TR. Protective effect of dual-strain probiotics in preterm infants: a multi-center time series analysis. Plos One. 2016;11(6):e0158136. doi:10.1371/journal.pone.0158136.
  • Poindexter B, Cummings J, Hand I, Adams-Chapman I, Aucott SW, Puopolo KM, Goldsmith JP, Kaufman D, Martin C, Mowitz M. AAP COMMITTEE on FETUS and NEWBORN. Use of probiotics in preterm infants. Pediatrics. 2021;147(6):e2021051485. doi:10.1542/peds.2021-051485.
  • Tobias J, Olyaei A, Laraway B, Jordan BK, Dickinson SL, Golzarri-Arroyo L, Fialkowski E, Owora A, Scottoline B. Bifidobacterium longum subsp. infantis EVC001 administration is associated with a significant reduction in the incidence of necrotizing enterocolitis in very low birth weight infants. J Pediatrics. 2022;244:64–71.e2. doi:10.1016/j.jpeds.2021.12.070.
  • Al-Hosni M, Duenas M, Hawk M, Stewart LA, Borghese RA, Cahoon M, Atwood L, Howard D, Ferrelli K, Soll R. Probiotics-supplemented feeding in extremely low-birth-weight infants. J Perinatol. 2012;32:253–259. doi:10.1038/jp.2011.51.
  • Singh B, Shah PS, Afifi J, Simpson CD, Mitra S, Dow K, El-Naggar W, Investigators CNN. Probiotics for preterm infants: a national retrospective cohort study. J Perinatol. 2019;39(4):533–539. doi:10.1038/s41372-019-0315-z.
  • Costeloe K, Bowler U, Brocklehurst P, Hardy P, Heal P, Juszczak E, King A, Panton N, Stacey F, Whiley A, et al. A randomised controlled trial of the probiotic Bifidobacterium breve BBG-001 in preterm babies to prevent sepsis, necrotising enterocolitis and death: the Probiotics in Preterm infantS (PiPS) trial. Health Technol Asses. 2016;20(66):1–194. doi:10.3310/hta20660.
  • Kane AF, Bhatia AD, Denning PW, Shane AL, Patel RM. Routine supplementation of lactobacillus rhamnosus GG and risk of necrotizing enterocolitis in very low birth weight infants. J Pediatrics. 2018;195:73–79.e2. doi:10.1016/j.jpeds.2017.11.055.
  • Escribano E, Zozaya C, Madero R, Sánchez L, van Goudoever J, Rodríguez JM, de Pipaon MS. Increased incidence of necrotizing enterocolitis associated with routine administration of Infloran™ in extremely preterm infants. Benef Microbes. 2018;9(5):683–690. doi:10.3920/BM2017.0098.
  • Dermyshi E, Wang Y, Yan C, Hong W, Qiu G, Gong X, Zhang T. The “golden age” of probiotics: a systematic review and meta-analysis of randomized and observational studies in preterm infants. Neonatol. 2017;112:9–23. doi:10.1159/000454668.
  • Deshmukh M, Patole S. Prophylactic probiotic supplementation for preterm neonates—A systematic review and meta-analysis of nonrandomized studies. Adv Nutr. 2021;12(4):1411–1423. doi:10.1093/advances/nmaa164.
  • Alshaikh B, Samara J, Moossavi S, Ferdous T, Soraisham A, Dersch-Mills D, Arrieta M-C, Amin H. Multi-strain probiotics for extremely preterm infants: a randomized controlled trial. Pediatr Res. 2022;92(6):1663–1670. doi:10.1038/s41390-022-02004-z.
  • Juber BA, Boly TJ, Pitcher GJ, McElroy SJ. Routine administration of a multispecies probiotic containing bifidobacterium and lactobacillus to very low birth weight infants had no significant impact on the incidence of necrotizing enterocolitis. Frontiers Pediatr. 2021;9:757299. doi:10.3389/fped.2021.757299.
  • Flannery DD, Ross RK, Mukhopadhyay S, Tribble AC, Puopolo KM, Gerber JS. Temporal trends and center variation in early antibiotic use among premature infants. JAMA Netw Open. 2018;1:e180164–e180164. doi:10.1001/jamanetworkopen.2018.0164.
  • Battersby C, Santhalingam T, Costeloe K, Modi N. Incidence of neonatal necrotising enterocolitis in high-income countries: a systematic review. Arch Dis Child Fetal Neonatal Ed. 2018;103(2):F182. doi:10.1136/archdischild-2017-313880.
  • Patel RM, Ferguson J, McElroy SJ, Khashu M, Caplan MS. Defining necrotizing enterocolitis: current difficulties and future opportunities. Pediatr Res. 2020;88:10–15. doi:10.1038/s41390-020-1074-4.
  • Meyer MP, Chow SSW, Alsweiler J, Bourchier D, Broadbent R, Knight D, Lynn AM, Patel H. Probiotics for prevention of severe necrotizing enterocolitis: experience of New Zealand neonatal intensive care units. Frontiers Pediatrics. 2020;8:119. doi:10.3389/fped.2020.00119.
  • Aceti A, Maggio L, Beghetti I, Gori D, Barone G, Callegari ML, Fantini MP, Indrio F, Meneghin F, Morelli L, et al. Probiotics prevent late-onset sepsis in human milk-fed, very low birth weight preterm infants: systematic review and meta-analysis. Nutrients. 2017;9(8):904. doi:10.3390/nu9080904.
  • Sawh SC, Deshpande S, Jansen S, Reynaert CJ, Jones PM. Prevention of necrotizing enterocolitis with probiotics: a systematic review and meta-analysis. PeerJ. 2016;4:e2429. doi:10.7717/peerj.2429.
  • Olsen R, Greisen G, Schrøder M, Brok J. Prophylactic probiotics for preterm infants: a systematic review and meta-analysis of observational studies. Neonatology. 2016;109:105–112. doi:10.1159/000441274.
  • AlFaleh K, Anabrees J. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Évid Based Child Heal Cochrane Rev J. 2014;9(3):584–671. doi:10.1002/ebch.1976.
  • Wejryd E, Marchini G, Frimmel V, Jonsson B, Abrahamsson T. Probiotics promoted head growth in extremely low birthweight infants in a double‐blind placebo‐controlled trial. Acta Paediatr. 2019;108:62–69. doi:10.1111/apa.14497.
  • Jacobs SE, Hickey L, Opie G, Donath S, Opie GF, Anderson PJ, Garland SM, Cheong JLY, Jacobs S, Hickey L, et al. Probiotics, prematurity and neurodevelopment: follow-up of a randomised trial. Bmj Paediatr Open. 2017;1(1):e000176. doi:10.1136/bmjpo-2017-000176.
  • Kulkarni T, Majarikar S, Deshmukh M, Ananthan A, Balasubramanian H, Keil A, Patole S. Probiotic sepsis in preterm neonates—a systematic review. Eur J Pediatr. 2022;181(6):2249–2262. doi:10.1007/s00431-022-04452-5.
  • Zhang W, Wang S, Xing Y, Wang H, Fu B, Long M, Cao J. Clinical efficacy of probiotics on feeding intolerance in preterm infants: a systematic review and meta-analysis. Transl Pediatrics. 2022;11:229–238. doi:10.21037/tp-21-624.
  • Weeks CL, Marino LV, Johnson MJ. A systematic review of the definitions and prevalence of feeding intolerance in preterm infants. Clin Nutr. 2021;40(11):5576–5586. doi:10.1016/j.clnu.2021.09.010.
  • Eveleens RD, Joosten KFM, de Koning BAE, Hulst JM, Verbruggen SCAT. Definitions, predictors and outcomes of feeding intolerance in critically ill children: a systematic review. Clin Nutr. 2020;39(3):685–693. doi:10.1016/j.clnu.2019.03.026.
  • Baranowski JR, Claud EC. Necrotizing enterocolitis and the preterm infant microbiome. Adv Exp Med Biol. 2019;1125:25–36.
  • Vallabhaneni S, Walker TA, Lockhart SR, Ng D, Chiller T, Melchreit R, Brandt ME, Smith RM. Centers for Disease Control and prevention (CDC). Notes from the Field. Mmwr Morbidity Mortal Wkly Rep. 2015;64:155–156.
  • Lewis ZT, Shani G, Masarweh CF, Popovic M, Frese SA, Sela DA, Underwood MA, Mills DA. Validating bifidobacterial species and subspecies identity in commercial probiotic products. Pediatr Res. 2016;79:445–452. doi:10.1038/pr.2015.244.
  • Underwood MA. Probiotics and the prevention of necrotizing enterocolitis. J Pediatr Surg. 2019;54(3):405–412. doi:10.1016/j.jpedsurg.2018.08.055.
  • Bertelli C, Pillonel T, Torregrossa A, Prod’hom G, Fischer CJ, Greub G, Giannoni E. Bifidobacterium longum bacteremia in preterm infants receiving probiotics. Clin Infect Dis. 2015;60(6):924–927. doi:10.1093/cid/ciu946.
  • Dani C, CC C, IC I, Arena F, Antonelli A, Rossolini GM. Lactobacillus sepsis and probiotic therapy in newborns: two new cases and literature review. Am J Perinatol Reports. 2015;6(01):e25–9. doi:10.1055/s-0035-1566312.
  • Chiang M-C, Chen C-L, Feng Y, Chen C-C, Lien R, Chiu C-H. Lactobacillus rhamnosus sepsis associated with probiotic therapy in an extremely preterm infant: pathogenesis and a review for clinicians. J Microbiol Immunol Infect. 2021;54(4):575–580. doi:10.1016/j.jmii.2020.03.029.
  • Esaiassen E, Cavanagh P, Hjerde E, Simonsen GS, Støen R, Klingenberg C. Bifidobacterium longum subspecies infantis bacteremia in 3 extremely preterm infants receiving probiotics. Emerg Infect Dis. 2016;22(9):1664–1666. doi:10.3201/eid2209.160033.
  • Sharma P, Tomar SK, Goswami P, Sangwan V, Singh R. Antibiotic resistance among commercially available probiotics. Food Res Int. 2014;57:176–195. doi:10.1016/j.foodres.2014.01.025.
  • Topcuoglu S, Gursoy T, Ovalı F, Serce O, Karatekin G. A new risk factor for neonatal vancomycin-resistant Enterococcus colonisation: bacterial probiotics. J Maternal-Fetal Neonatal Medicine. 2015;28(12):1491–1494. doi:10.3109/14767058.2014.958462.
  • van den Akker CHP, van Goudoever JB, Shamir R, Domellöf M, Embleton ND, Hojsak I, Lapillonne A, Mihatsch WA, Canani RB, Bronsky J, et al. Probiotics and preterm infants: a position paper by the european society for paediatric gastroenterology hepatology and nutrition committee on nutrition and the european society for paediatric gastroenterology hepatology and nutrition working group for probiotics and prebiotics. J Pediatric Gastroenterology Nutrition. 2020;70:664–680.
  • Thomas DW, Greer FR. American academy of pediatrics committee on nutrition, section on gastroenterology, hepatology, and nutrition. Probiotics and prebiotics in pediatrics. Pediatrics. 2010;126(6):1217–1231. doi:10.1542/peds.2010-2548.
  • McFarland LV, Evans CT, Goldstein EJC. Strain-specificity and disease-specificity of probiotic efficacy: a systematic review and meta-analysis. Frontiers Med. 2018;5:124. doi:10.3389/fmed.2018.00124.
  • Underwood MA, Kalanetra KM, Bokulich NA, Lewis ZT, Mirmiran M, Tancredi DJ, Mills DA. A comparison of two probiotic strains of bifidobacteria in premature infants. J Pediatrics. 2013;163(6):1585–1591.e9. doi:10.1016/j.jpeds.2013.07.017.
  • Deshpande G, Rao S, Athalye-Jape G, Conway P, Patole S. Probiotics in very preterm infants: the PiPS trial. Lancet. 2016;388(10045):655. doi:10.1016/S0140-6736(16)31271-5.
  • Su GL, Ko CW, Bercik P, Falck-Ytter Y, Sultan S, Weizman AV, Morgan RL. AGA clinical practice guidelines on the role of probiotics in the management of gastrointestinal disorders. Gastroenterology. 2020;159:697–705. doi:10.1053/j.gastro.2020.05.059.
  • WHO. Recommendations for care of the preterm or low-birth-weight infant. Geneva: World Health Organization; 2022. Licence: CC BY-NC-SA 3.0 IGO.
  • Szajewska H, Canani RB, Domellöf M, Guarino A, Hojsak I, Indrio F, Vecchio AL, Mihatsch WA, 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. 2023;76(2):232–247. doi:10.1097/MPG.0000000000003633.
  • Merenstein D, Pot B, Leyer G, Ouwehand AC, Preidis GA, Elkins CA, Hill C, Lewis ZT, Shane AL, Zmora N, et al. Emerging issues in probiotic safety: 2023 perspectives. Gut Microbes. 2023;15(1):2185034. doi:10.1080/19490976.2023.2185034.
  • Gibson GR, Hutkins R, Sanders ME, Prescott SL, Reimer RA, Salminen SJ, Scott K, Stanton C, Swanson KS, Cani PD, et al. Expert consensus document: the International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroentero. 2017;14(8):491–502. doi:10.1038/nrgastro.2017.75.
  • Bode L. Human milk oligosaccharides: every baby needs a sugar mama. Glycobiology. 2012;22:1147–1162. doi:10.1093/glycob/cws074.
  • Schwab C, Gänzle M. Lactic acid bacteria fermentation of human milk oligosaccharide components, human milk oligosaccharides and galactooligosaccharides. FEMS Microbiol Lett. 2011;315(2):141–148. doi:10.1111/j.1574-6968.2010.02185.x.
  • Thongaram T, Hoeflinger JL, Chow J, Miller MJ. Human milk oligosaccharide consumption by probiotic and human-associated bifidobacteria and lactobacilli. J Dairy Sci. 2017;100(10):7825–7833. doi:10.3168/jds.2017-12753.
  • Abbas S, Keir AK, Makrides M, Klein LD, Grzeskowiak LE, McPhee AJ, Rumbold AR. Tailoring human milk oligosaccharides to prevent necrotising enterocolitis among preterm infants. Frontiers Nutr. 2021;8:702888. doi:10.3389/fnut.2021.702888.
  • Duar RM, Casaburi G, Mitchell RD, Scofield LNC, Ramirez CAO, Barile D, Henrick BM, Frese SA. Comparative genome analysis of bifidobacterium longum subsp. infantis strains reveals variation in human milk oligosaccharide utilization genes among commercial probiotics. Nutrients. 2020;12:3247. doi:10.3390/nu12113247.
  • Zheng N, Gao Y, Zhu W, Meng D, Walker WA, Nie D. Short chain fatty acids produced by colonizing intestinal commensal bacterial interaction with expressed breast milk are anti-inflammatory in human immature enterocytes. Plos One. 2020;15(2):e0229283. doi:10.1371/journal.pone.0229283.
  • Chichlowski M, Lartigue GD, German JB, Raybould HE, Mills DA. Bifidobacteria isolated from infants and cultured on human milk oligosaccharides affect intestinal epithelial function. J Pediatr Gastroenterol Nutr. 2012;55(3):321–327. doi:10.1097/MPG.0b013e31824fb899.
  • Eiwegger T, Stahl B, Haidl P, Schmitt J, Boehm G, Dehlink E, Urbanek R, Szépfalusi Z. Prebiotic oligosaccharides: in vitro evidence for gastrointestinal epithelial transfer and immunomodulatory properties. Pediatr Allergy Immu. 2010;21(8):1179–1188. doi:10.1111/j.1399-3038.2010.01062.x.
  • Lin AE, Autran CA, Szyszka A, Escajadillo T, Huang M, Godula K, Prudden AR, Boons G-J, Lewis AL, Doran KS, et al. Human milk oligosaccharides inhibit growth of group B Streptococcus. J Biol Chem. 2017;292(27):11243–11249. doi:10.1074/jbc.M117.789974.
  • Laucirica DR, Triantis V, Schoemaker R, Estes MK, Ramani S. Milk oligosaccharides inhibit human rotavirus Infectivity in MA104 cells. J Nutrition. 2017;147(9):1709–1714. doi:10.3945/jn.116.246090.
  • Wilson B, Whelan K. Prebiotic inulin‐type fructans and galacto‐oligosaccharides: definition, specificity, function, and application in gastrointestinal disorders. J Gastroen Hepatol. 2017;32:64–68. doi:10.1111/jgh.13700.
  • Wiciński M, Sawicka E, Gębalski J, Kubiak K, Malinowski B. Human milk oligosaccharides: health benefits, potential applications in infant formulas, and pharmacology. Nutr. 2020;12:266. doi:10.3390/nu12010266.
  • Westerbeek EAM, Slump RA, Lafeber HN, Knol J, Georgi G, Fetter WPF, van Elburg RM. The effect of enteral supplementation of specific neutral and acidic oligosaccharides on the faecal microbiota and intestinal microenvironment in preterm infants. Eur J Clin Microbiol. 2013;32(2):269–276. doi:10.1007/s10096-012-1739-y.
  • Boehm G, Lidestri M, Casetta P, Jelinek J, Negretti F, Stahl B, Marini A. Supplementation of a bovine milk formula with an oligosaccharide mixture increases counts of faecal bifidobacteria in preterm infants. Archives Dis Child - Fetal Neonatal Ed. 2002;86(3):178F–181. doi:10.1136/fn.86.3.F178.
  • Knol J, Boehm G, Lidestri M, Negretti F, Jelinek J, Agosti M, Stahl B, Marini A, Mosca F. Increase of faecal bifidobacteria due to dietary oligosaccharides induces a reduction of clinically relevant pathogen germs in the faeces of formula‐fed preterm infants. Acta Paediatr. 2005;94:31–33. doi:10.1111/j.1651-2227.2005.tb02152.x.
  • Kapiki A, Costalos C, Oikonomidou C, Triantafyllidou A, Loukatou E, Pertrohilou V. The effect of a fructo-oligosaccharide supplemented formula on gut flora of preterm infants. Early Hum Dev. 2007;83:335–339. doi:10.1016/j.earlhumdev.2006.07.003.
  • Armanian A-M, Sadeghnia A, Hoseinzadeh M, Mirlohi M, Feizi A, Salehimehr N, Torkan M, Shirani Z. The effect of neutral oligosaccharides on fecal microbiota in premature infants fed exclusively with breast milk: a randomized clinical trial. J Res Pharm Pract. 2016;5:27–34. doi:10.4103/2279-042X.176558.
  • Yu X, Xing Y, Liu H, Chang Y, You Y, Dou Y, Liu B, Wang Q, Ma D, Chen L, et al. Effects of a formula with scGOS/lcFOS (9: 1) and Glycomacropeptide (GMP) supplementation on the gut microbiota of very preterm infants. Nutrients. 2022;14(9):1901. doi:10.3390/nu14091901.
  • Lucas A, Cole TJ. Breast milk and neonatal necrotising enterocolitis. Lancet. 1990;336:1519–1523. doi:10.1016/0140-6736(90)93304-8.
  • Armanian A-M, Sadeghnia A, Hoseinzadeh M, Mirlohi M, Feizi A, Salehimehr N, Saee N, Nazari J. The effect of neutral oligosaccharides on reducing the incidence of necrotizing enterocolitis in preterm infants: a randomized clinical trial. Int J Prev Medicine. 2014;5:1387–1395.
  • Modi N, Uthaya S, Fell J, Kulinskaya E. A randomized, double-blind, controlled trial of the effect of prebiotic oligosaccharides on enteral tolerance in preterm infants (ISRCTN77444690). Pediatr Res. 2010;68:440–445. doi:10.1203/PDR.0b013e3181f1cd59.
  • Dasopoulou M, Briana DD, Boutsikou T, Karakasidou E, Roma E, Costalos C, Malamitsi‐Puchner A. Motilin and gastrin secretion and lipid profile in preterm neonates following prebiotics supplementation. Jpen-Parenter Enter. 2015;39(3):359–368. doi:10.1177/0148607113510182.
  • Dilli D, Aydin B, Fettah ND, Özyazıcı E, Beken S, Zenciroğlu A, Okumuş N, Özyurt BM, İ̇pek MŞ, Akdağ A, et al. The ProPre-save study: effects of probiotics and prebiotics alone or combined on necrotizing enterocolitis in very low birth weight infants. J Pediatrics. 2015;166(3):545–551.e1. doi:10.1016/j.jpeds.2014.12.004.
  • Westerbeek EA, van den Berg JP, Lafeber HN, Fetter WP, Boehm G, Twisk JW, van Elburg RM. Neutral and acidic oligosaccharides in preterm infants: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutrition. 2009;91(3):679–686. doi:10.3945/ajcn.2009.28625.
  • Chi C, Buys N, Li C, Sun J, Yin C. Effects of prebiotics on sepsis, necrotizing enterocolitis, mortality, feeding intolerance, time to full enteral feeding, length of hospital stay, and stool frequency in preterm infants: a meta-analysis. Eur J Clin Nutr. 2019;73:657–670. doi:10.1038/s41430-018-0377-6.
  • Riskin A, Hochwald O, Bader D, Srugo I, Naftali G, Kugelman A, Cohen E, Mor F, Kaufman B, Shaoul R. The effects of lactulose supplementation to enteral feedings in premature infants: a pilot study. J Pediatrics. 2010;156(2):209–214. doi:10.1016/j.jpeds.2009.09.006.
  • Mihatsch WA, Hoegel J, Pohlandt F. Prebiotic oligosaccharides reduce stool viscosity and accelerate gastrointestinal transport in preterm infants. Acta Paediatr. 2006;95(7):843–848. doi:10.1080/08035250500486652.
  • Marteau P, Seksik P. Tolerance of Probiotics and Prebiotics. J Clin Gastroenterol. 2004;38(Supplement 2):S67–9. doi:10.1097/01.mcg.0000128929.37156.a7.
  • Niele N, van Zwol A, Westerbeek EA, Lafeber HN, van Elburg RM. Effect of non-human neutral and acidic oligosaccharides on allergic and infectious diseases in preterm infants. Eur J Pediatr. 2013;172:317–323. doi:10.1007/s00431-012-1886-2.
  • Sherman PM, Cabana M, Gibson GR, Koletzko BV, Neu J, Veereman-Wauters G, Ziegler EE, Walker WA. Potential roles and clinical utility of prebiotics in newborns, infants, and children: proceedings from a global prebiotic summit meeting, New York City, June 27-28, 2008. J Pediatrics. 2009;155(5):S61–70. doi:10.1016/j.jpeds.2009.08.022.
  • Panigrahi P, Parida S, Pradhan L, Mohapatra SS, Misra PR, Johnson JA, Chaudhry R, Taylor S, Hansen NI, Gewolb IH. Long-term colonization of a lactobacillus plantarum synbiotic preparation in the neonatal gut. J Pediatr Gastroenterol Nutr. 2008;47(1):45–53. doi:10.1097/MPG.0b013e31815a5f2c.
  • Panigrahi P, Parida S, Nanda NC, Satpathy R, Pradhan L, Chandel DS, Baccaglini L, Mohapatra A, Mohapatra SS, Misra PR, et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017;548(7668):407–412. doi:10.1038/nature23480.
  • Nandhini LP, Biswal N, Adhisivam B, Mandal J, VBhat B, Mathai B. Synbiotics for decreasing incidence of necrotizing enterocolitis among preterm neonates – a randomized controlled trial. J Maternal-Fetal Neonatal Medicine. 2015;29(5):821–825. doi:10.3109/14767058.2015.1019854.
  • Sharif S, Heath PT, Oddie SJ, McGuire W. Synbiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Db Syst Rev. 2022;2022(3) :CD014067. doi:10.1002/14651858.CD014067.pub2.
  • Lin H-C, Su B-H, Chen A-C, Lin T-W, Tsai C-H, Yeh T-F, Oh W. Oral probiotics reduce the incidence and severity of necrotizing enterocolitis in very low birth weight infants. Pediatrics. 2005;115:1–4. doi:10.1542/peds.2004-1463.
  • Lin H-C, Hsu C-H, Chen H-L, Chung M-Y, Hsu J-F, Lien R, Tsao L-Y, Chen C-H, Su B-H. Oral probiotics prevent necrotizing enterocolitis in very low birth weight preterm infants: a multicenter, randomized, controlled trial. Pediatrics. 2008;122:693–700. doi:10.1542/peds.2007-3007.
  • Samanta M, Sarkar M, Ghosh P, Ghosh JK, Sinha MK, Chatterjee S. Prophylactic probiotics for prevention of necrotizing enterocolitis in very low birth weight newborns. J Trop Pediatrics. 2009;55(2):128–131. doi:10.1093/tropej/fmn091.
  • Rougé C, Piloquet H, Butel M-J, Berger B, Rochat F, Ferraris L, Robert CD, Legrand A, de la Cochetière M-F, N’Guyen J-M, et al. Oral supplementation with probiotics in very-low-birth-weight preterm infants: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutrition. 2009;89(6):1828–1835. doi:10.3945/ajcn.2008.26919.
  • Braga TD, da Silva GAP, de Lira PIC, de Carvalho Lima M. Efficacy of Bifidobacterium breve and Lactobacillus casei oral supplementation on necrotizing enterocolitis in very-low-birth-weight preterm infants: a double-blind, randomized, controlled trial. Am J Clin Nutrition. 2010;93(1):81–86. doi:10.3945/ajcn.2010.29799.
  • Roy A, Chaudhuri J, Sarkar D, Ghosh P, Chakraborty S. Role of enteric supplementation of probiotics on late-onset sepsis by candida species in preterm low birth weight neonates: a randomized, double blind, placebo-controlled trial. North Am J Medical Sci. 2014;6(1):50–57. doi:10.4103/1947-2714.125870.
  • Saengtawesin V, Tangpolkaiwalsak R, Kanjanapattankul W. Effect of oral probiotics supplementation in the prevention of necrotizing enterocolitis among very low birth weight preterm infants. J Medical Assoc Thail Chotmaihet Thangphaet. 2014;97:S20–5.
  • Niekerk EV, Nel DG, Blaauw R, Kirsten GF. Probiotics reduce necrotizing enterocolitis severity in HIV-exposed premature infants. J Trop Pediatrics. 2015;61(3):155–164. doi:10.1093/tropej/fmv004.
  • Mihatsch WA, Vossbeck S, Eikmanns B, Hoegel J, Pohlandt F. Effect of Bifidobacterium lactis on the incidence of nosocomial infections in very-low-birth-weight infants: a randomized controlled trial. Neonatology. 2010;98(2):156–163. doi:10.1159/000280291.
  • Stratiki Z, Costalos C, Sevastiadou S, Kastanidou O, Skouroliakou M, Giakoumatou A, Petrohilou V. The effect of a bifidobacter supplemented bovine milk on intestinal permeability of preterm infants. Early Hum Dev. 2007;83(9):575–579. doi:10.1016/j.earlhumdev.2006.12.002.
  • Hays S, Jacquot A, Gauthier H, Kempf C, Beissel A, Pidoux O, Jumas-Bilak E, Decullier E, Lachambre E, Beck L, et al. Probiotics and growth in preterm infants: a randomized controlled trial, PREMAPRO study. Clin Nutr. 2016;35(4):802–811. doi:10.1016/j.clnu.2015.06.006.
  • Shashidhar A, Rao PNS, Nesargi S, Bhat S, Chandrakala BS. Probiotics for promoting feed tolerance in very low birth weight neonates — a randomized controlled trial. Indian Pediatr. 2017;54:363–367. doi:10.1007/s13312-017-1106-2.
  • Hariharan D, Balasubramanian L, Kannappan V, Veluswami G. 49th annual meeting of the european society for paediatric gastroenterology, hepatology and nutrition. Probiotic supplementation in VLBW preterm infants improves feeding tolerance and reduces risk of gram negative sepsis. J Pediatr Gastroenterol Nutr. 2016;62:655.
  • Arora S, Khurana MS, Saini R. To study the role of probiotics in the prevention of necrotizing enterocolitis in preterm neonates. Int J Contemp Pediatrics. 2017;4:1792–1797. doi:10.18203/2349-3291.ijcp20173787.
  • Romeo MG, Romeo DM, Trovato L, Oliveri S, Palermo F, Cota F, Betta P. Role of probiotics in the prevention of the enteric colonization by Candida in preterm newborns: incidence of late-onset sepsis and neurological outcome. J Perinatol. 2011;31:63–69. doi:10.1038/jp.2010.57.
  • Rojas MA, Lozano JM, Rojas MX, Rodriguez VA, Rondon MA, Bastidas JA, Perez LA, Rojas C, Ovalle O, Garcia-Harker JE, et al. Prophylactic probiotics to prevent death and nosocomial infection in preterm infants. Pediatrics. 2012;130(5):e1113–20. doi:10.1542/peds.2011-3584.
  • Indrio F, Riezzo G, Tafuri S, Ficarella M, Carlucci B, Bisceglia M, Polimeno L, Francavilla R. Probiotic supplementation in preterm: feeding intolerance and hospital cost. Nutrients. 2017;9:965. doi:10.3390/nu9090965.
  • Oncel MY, Sari FN, Arayici S, Guzoglu N, Erdeve O, Uras N, Oguz SS, Dilmen U. Lactobacillus Reuteri for the prevention of necrotising enterocolitis in very low birthweight infants: a randomised controlled trial. Archives Dis Child - Fetal Neonatal Ed. 2014;99(2):F110. doi:10.1136/archdischild-2013-304745.
  • Hernández-Enríquez NP, Rosas-Sumano AB, Monzoy-Ventre MA, Galicia-Flores L. Lactobacillus reuteri DSM 17938 en la prevención de enterocolitis necrosante en recién nacidos prematuros. Estudio piloto de eficacia y seguridad. Revista Mexicana de Pediatría. 2016;83:37–43.
  • Freedman SB, Xie J, Neufeld MS, Hamilton WL, Hartling L, Tarr PI, Nettel-Aguirre A, Chuck A, Lee B, Alberta Provincial Pediatric Enteric Infection Team (APPETITE), et al. Shiga toxin–producing escherichia coli infection, antibiotics, and risk of developing hemolytic uremic syndrome: a meta-analysis. Clin Infect Dis. 2016;62(10):1251–1258. doi:10.1093/cid/ciw099.
  • Barbian ME, Patel RM. Probiotics for prevention of necrotizing enterocolitis: where do we stand? Semin Perinatol. 2023;47(1):151689. doi:10.1016/j.semperi.2022.151689.
  • Jarrett P, Meczner A, Costeloe K, Fleming P. Historical aspects of probiotic use to prevent necrotising enterocolitis in preterm babies. Early Hum Dev. 2019;135:51–57. doi:10.1016/j.earlhumdev.2019.05.015.
  • Barbian ME, Buckle R, Denning PW, Patel RM. To start or not: factors to consider when implementing routine probiotic use in the NICU. Early Hum Dev. 2019;135:66–71. doi:10.1016/j.earlhumdev.2019.05.009.