Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Feb 1.
Published in final edited form as: Semin Perinatol. 2016 Nov 8;41(1):41–51. doi: 10.1053/j.semperi.2016.09.017

Impact of Probiotics on Necrotizing Enterocolitis

Mark A Underwood 1
PMCID: PMC5334136  NIHMSID: NIHMS828679  PMID: 27836423

Abstract

A large number of randomized placebo-controlled clinical trials and cohort studies have demonstrated a decrease in the incidence of necrotizing enterocolitis with administration of probiotic microbes. These studies have prompted many neonatologists to adopt routine prophylactic administration of probiotics while others await more definitive studies and/or probiotic products with demonstrated purity and stable numbers of live organisms. Cross-contamination and inadequate sample size limit the value of further traditional placebo-controlled randomized controlled trials. Key areas for future research include mechanisms of protection, optimum probiotic species or strains (or combinations thereof) and duration of treatment, interactions between diet and the administered probiotic, and the influence of genetic polymorphisms in the mother and infant on probiotic response. Next generation probiotics selected based on bacterial genetics rather than ease of production and large cluster-randomized clinical trials hold great promise for NEC prevention.


It has been more than 20 years since the first premature infants were enrolled in the first published cohort study of probiotic microbes for the prevention of necrotizing enterocolitis (NEC). In that landmark study, Dr. Angela Hoyos and her colleagues gave every baby admitted to the neonatal intensive care unit (NICU) in Hospital Simon Bolivar, Bogota, Colombia for a one year period, a probiotic formulation containing Lactobacillus acidophilus and Bifidobacterium infantis (Infloran, Swiss Serum and Vaccine Institute, Bern, Switzerland) at a dose of 2.5 × 108 of each organism once daily for the entire hospitalization. They then compared the incidence of NEC during the probiotic year and the previous year and found a dramatic decrease with administration of probiotics.1

Since that time, 35 randomized placebo controlled clinical trials with NEC, death and/or sepsis as a reported outcome (the first 33 of these trials have recently been summarized2 with two additional trials published subsequently3,4) and an additional 10 cohort studies comparing periods of time with universal treatment with probiotics to control periods2,5 have been published in English language journals. Tables 16-12 and 21,5,13-17 present the data for the largest of these trials and cohort studies (those with at least 200 infants per arm). When all 35 randomized controlled trials are combined a total of 5559 premature infants received probiotics and 5513 premature infants received either a placebo or a blinded non-treatment. Among those studies reporting stage 2 or greater NEC as an outcome, unweighted percentages were 3.3% in infants receiving probiotics and 6.1% in control infants. Among those studies reporting culture positive sepsis the unweighted percentages were 12% and 14%, and among those studies reporting death 5.1% and 7.2% in the probiotic and control groups respectively. When the 11 cohort studies are combined, the unweighted percentages are strikingly similar to the randomized trials: 7742 infants received probiotics (NEC 1.4%, sepsis 12%, and death 7.6%) and 7592 did not (NEC 4.4%, sepsis 14% and death 9.2%). Multiple English language meta-analyses of the randomized controlled trials have been performed (the most recent are summarized in Table 3),18-23 all with similar conclusions: treatment of premature infants with probiotics decreases the risk of NEC and decreases mortality. Many of the published studies also included data on feeding tolerance and length of hospitalization, both of which were improved in the probiotic groups. It is possible that routine administration of probiotics to premature infants is the most studied, safe, and effective preventive intervention ever to be rejected by U.S. neonatology. In this article we will review the mechanisms by which probiotics exert their protective effects, the risks and obstacles to probiotic administration, the data favoring specific probiotic species and strains, and promising future directions.

Table 1. Probiotic RCTs with 200 or more premature infants in each arm.

Author
Year
Country Probioti
c
Species
(strain)
Brand
(Compa
ny)
Popul
ation
Dose
×
durati
on
Number
enrolled
NEC
cases
Stage 2,
3
Culture
+ sepsis
cases
Deaths
Pr
ob
Co
nt
Pr
ob
Co
nt
Pr
ob
Co
nt
Pr
ob
Co
nt
No benefit
Costeloe 2015*6 U K B breve (BBG 001) NR (Yakult) GA 23-30 w 1.6e8 - 1.6e9/d until 36w 650 660 62 66 73 77 54 56
Oncel 20147 Turkey L reuteri (DSM 17938) NR (Biogaia AB) BW < 1500 g + GA < 32 w 1e8/d until discharge 200 200 8 10 13 25 15 20
Rojas 20128 Colombia L reuteri (DSM 17938) NR (Biogaia AB) BW < 2000 g 1e8/d until dischrge 372 378 9 15 24 17 22 28
Dani 20029 Italy L rhamnous (GG) Dicoflor (Dicofarm) BW < 1500 g and GA < 33 w 6e9/d until discharge 295 290 4 8 14 12 0 2
Benefit
Manzoni 201410 Italy + NZ L rhamnosus (GG) + Bovine lactoferrin NR BW < 1500 g 6e9/d until day 30 238 258 0 14 NR NR 9 26
Jacobs 2013**11 Australia + NZ B infantis (BB-02) + S thermophiles (TH-415957) + B lactis (BB-12 15954) ABC Dophilus (Solgar) BW < 1500 g + GA < 32 w 1e9/d until discharge 548 551 11 24 72 89 27 28
Lin 200812 Taiwan L acidophilus (NCDO 1748) + B bifidum (NCDO 1453) Infloran (Laboratorio Farmaceutico) BW < 1500 g + GA < 34 w 1e9 BID until 6 w 217 217 4 14 40 24 2 9
*

Subgroup analysis showed a decrease in NEC among infants with colonization with the probiotic compared to those not colonized.

**

Subgroup analysis showed a decrease in sepsis in infants > 28 weeks but not < 28 weeks.

Table 2. Probiotic cohort studies with 200 or more premature infants in each cohort.

Author
Year
Country Probiot
ic
Species
(strain)
Brand
(Compa
ny)
Popul
ation
Dose
×
dura
tion
Numbe
r
enrolle
d
NEC
cases
Stage
2, 3
Culture
+ sepsis
cases
Deaths
Pr
ob
Co
nt
Pr
ob
Co
nt
Pr
ob
Co
nt
Pr
ob
Co
nt
No benefit
Repa 2015*13 Austria L acidophilus + B infantis (NR) Infloran (Laboratorio Farmaceutico) GA < 34 w 2e9 BID until discharge or 37 w 230 233 16 24 60 78 16 30
Li 201314 U S B bifidum + B infantis + S thermophiles (NR) ABC Dophilus (NR) BW < 1500 g + GA < 33 w 5e8-1e9/d until discharge or 36 w 291 289 7 9 NR NR 4 3
Benefit
Hartel 2014**15 Germany L acidophilus + B infantis (NR) Infloran (Berna) BW < 1500 g + GA 23-31 w 1e9/d from d2 to d14 3789 1562 116 (3.6) 76 (4.8) 428 (11) 195 (12) 292 (7.7) 160 (10)
Guthmann 20165 Switzerland and Germany L acidophilus + B infantis (NR) Infloran (Laboratorio Farmaceutico) BW<1 500 g + GA <32 w 2e9 BID d1-3 to d10-14 591 633 8 33 NR NR 21 32
Janvier 201416 Canada B bifidum + B breve + B infantis + B (NR) + L rhamnosus (GG) FloraBABY (Renew Life) GA < 32 w 2e9/d until 34 w 294 317 16 31 54 57 20 31
Bonsante 201317 France L reuteri (LCR35) Lcr Restituo (Probionov) GA 24-31 w 2e8 BID until discharge or 36 w 347 783 4 41 37 130 8 38
Hoyos 1999***1 Colombia L acidophilus + B infantis (NR) Infloran Berna 7 (Swiss Serum and Vaccine Institute) All NICU patie nts 5e8/d until discharge 1237 1282 34 85 69 70 137 140
*

The probiotic cohort was smaller and more premature. Subgroup analysis showed a significant decrease in NEC among human milk fed infants but not formula fed infants.

**

percentages included in parentheses due to difference in size of the two cohorts.

***

included both term and premature infants.

Table 3. Recent meta-analyses of probiotic trials in premature infants.

Author Year Number of studies included RR for NEC (95% CI) RR for death (95% CI) Other analyses
Aceti 201618 26 0.47 (0.36-0.60), p < 0.00001 Probiotics containing Bifidobacterium: RR 0.24 (0.10-0.54), p = 0.0006 Probiotic combinations: RR 0.48 (0.37-0.62), p < 0.00001
Olsen 201619 12 0.55 (0.39-0.78), p = 0.0006 0.72, (0.61-0.85), p < 0.0001
Aggarwal S 201620 23 0.69 (0.56 and 0.86), p=0.0007
Lau 201521 20 0.51 (0.39-0.67), p<0.001 0.73 (0.58-0.93), p=0.009
Athalye-Jape 201522 8 (limited to L reuteri) 0.69 (0.47-1.01), 3 RCTs 0.79 (0.57-1.09), 3 RCTs
Al Faleh 201423 24 0.43 (0.33-0.56), 20 RCTs 0.65 (0.52-0.81), 17 RCTs Probiotics containing either Lactobacillus alone or in combination with Bifidobacterium were effective

Mechanisms of action

Probiotics are variously categorized as either dietary supplements or medications and are generally defined as containing live organisms that improve health. At the most basic level, probiotics are administered in an attempt to alter the composition of the intestinal microbiota. Most probiotic products contain one or more of the following bacterial or fungal genera: Lactobacillus, Bifidobacterium, Streptococcus, Escherichia, Enterococcus, Bacillus, or Saccharomyces. The capacity for an ingested probiotic microbe to reach the small intestine and impact the microbial community in the small and large bowel is determined by its resistance to oral and gastric enzymes and acids and to bile acids and its ability to effectively compete with other gut microbes without triggering an untoward host response. This review will focus on the probiotic genera that have been most studied in premature infants and NEC animal models: bifidobacteria and lactobacilli.

Colonization of the fetal intestinal tract begins in utero. Until recently, amniotic fluid was believed to be sterile until the time of rupture of the fetal membranes, however careful studies have demonstrated that the placenta and the amniotic fluid become colonized with microbes, predominantly from the maternal vagina or by hematogenous spread of maternal oral microbes even prior to obvious membrane rupture.24,25 Since vaginal dysbiosis and maternal periodontal disease are both associated with preterm labor,26,27 it is likely that many preterm infants are colonized in utero through swallowed amniotic fluid with both maternal commensals and pathobionts. At the time of rupture of membranes and delivery the fetus/neonate is exposed to large numbers of bacteria with this initial wave of colonists influenced by delivery type and antibiotic exposure. Healthy term infants who are born vaginally and breast-fed undergo a change in the fecal microbiota in the first weeks of life from a preponderance of vaginal microbes to a preponderance of organisms that are capable of consuming human milk oligosaccharides (HMOs). Of dozens of bacterial genera tested, only bacteroides and bifidobacteria are capable of consuming HMOs.28 In a cohort of predominantly breast-fed infants in Bangladesh, we found the dominant fecal bacteria to be Actinobacterium at the phylum level and Bifidobacterium infantis at the subspecies level. Furthermore we found that the infants with the highest numbers of B. infantis had better growth, larger thymus size by ultrasound, and higher T cell and IgG responses to several vaccines.29 Premature infants often have prolonged hospitalizations, periods of time without any enteral feedings, exposure to antibiotics and other agents with the potential to alter the gut microbiota (e.g. acid blocking agents) and therefore become colonized with a community of microbes that is dominated by Gram negative Proteobacteria and Gram positive Firmicutes at the phylum level and is heavily influenced by corrected gestational age.30 Most of the pathobionts that cause sepsis and NEC in premature infants are either Proteobacteria (e.g. E. coli, Klebsiella, Proteus, Serratia, Pseudomonas) or Firmicutes (enterococci, streptococci, staphylococci and clostridia). A bloom of Proteobacteria just prior to the onset of NEC has been demonstrated.31,32 Bifidobacteria are conspicuously absent in most very premature infants even those fed exclusively human milk.

Bifidobacteria

The primary mechanism of colonization of the intestinal tract by bifidobacteria in term infants is the species-specific capacity to consume HMOs as a food source. The bacterial genomes of many strains of bifidobacteria have been sequenced revealing a large variety of bacterial glycosidases with those species most commonly found in infant feces (infantis, bifidum, and breve) expressing the glycosidases necessary to digest HMOs and those species more commonly found in adult feces (animalis, longum, adolescentis, dentium, catenulatum, and pseudocatenulatum) expressing glycosidases necessary to digest plant oligosaccharides.33,34 Among the bifidobacterial species, B. infantis is unique in the number of fucosidases, sialidases, and other glycosyl hydrolases expressed in its genome. Thus B. infantis is able to utilize the full range of HMOs as a nutrient source, whereas B. bifidum is unable to utilize fucosylated and sialylated HMOs and B. breve predominantly utilizes neutral HMOs with capacity to utilize fucosylated and sialylated HMOs varying by strain.35-37 In premature infants, administration of probiotic B. infantis led to colonization while administration of probiotic B. animalis ssp lactis did not.38

In addition to an advantage in colonization in the presence of human milk, bifidobacterial species demonstrate other properties that may contribute to prevention of NEC. B. infantis binds to Caco-2 cells,39 produces a single bacteriocin with activity against other lactic acid bacilli as well as staphylococci, streptococci, Salmonella and E. coli,40 and secretes molecules with anti-inflammatory properties.39,41 In the rat model of NEC, B. infantis decreases the incidence and severity of NEC and decreases expression of IL6, IL8, TNFα, IL23, and iNOS.42 Culture medium from B. infantis decreases the severity of infection with C. sakazaki in the mouse with attenuation of three mechanisms of pathogenesis: induction of IL1β and TNFα, decrease in mucin production, and increase in apoptosis.43

B. bifidum produces two known bacteriocins40 and is a more abundant producer of short chain fatty acids than other bifidobacteria tested (the short chain fatty acids butyrate, propionate, and acetate are produced by commensal bacteria and serve as a primary nutrient source for host colonocytes).44 In the rat model of NEC, B. bifidum decreases the incidence and severity of NEC, attenuates induction of IL6, attenuates induction of trefoil factor 3 and several antimicrobial peptides, improves barrier function, activates TLR2, and decreases apoptosis.45-47

B. breve has also been tested in the rat with decreased incidence and severity of NEC, attenuated TLR4 signaling, increased TLR2 signaling and suppressed inflammation.48 In the rat weanling colitis model, B. breve ameliorated colitis with attenuations in IL1α, IL1β, IL10, TNFα, and TGFβ in the colon.49

Lactobacilli

While Lactobacillus species are found in abundance in the vagina, they are not typical colonizers of the newborn intestinal tract. Several species of Lactobacillus are utilized as food additives and sold as commercial probiotics and have been extensively characterized. Lactobacilli are not generally able to consume intact HMOs, however a species of L casei has recently been shown to contain a gene cluster capable of metabolizing lacto-N-biose, a key component of HMOs and a second cluster capable of metabolizing the HMO lacto-N-triose utilizing metabolic pathways that differ from those of bifidobacteria.50,51 In premature prolonged rupture of membranes, if the maternal cervical microbiota is dominated by lactobacilli, the incidences of microbial invasion of the amniotic cavity and histologic chorioamnionitis are decreased.52 In vitro studies demonstrate marked heterogeneity among species of Lactobacillus in the capacity to induce activation and maturation of dendritic cells with some species predominantly pro-inflammatory (e.g. L salivarius).53

L. acidophilus is a common food additive, is stable across a range of pH and temperature, and decreased disease severity in a rat NEC model.54 L acidophilus strains produce at least 27 bacteriocins with antibacterial activity against a wide range of gut microbes including other lactobacilli, Enterobacteriaceae, Bacillus sp,, Corynebacterium sp, Vibrio sp, staphylococci, and C. sakazaki55-57 L acidophilus secretes anti-inflammatory factors that inhibit induction of NFκB and IL-8 by platelet activating factor (PAF) an inflammatory pathway important in NEC.58 L acidophilus produces short chain fatty acids and also secretes factors that facilitate uptake of butyrate by the host colonocyte.59 A recent study demonstrated that L. acidophilus altered expression in human fetal intestinal cells of a variety of genes important in immune response, apoptosis and cell survival, cell adhesion, the cell cycle, development and angiogenesis.60

L. reuteri has direct antimicrobial activity through secretion of at least one bacteriocin (reuterin) and an antibiotic (reutericycline).61,62 In vitro studies have demonstrated strain specific anti-inflammatory effects via suppression of tumor necrosis factor (TNF) in activated human macrophages.63 In a rat model of NEC, L reuteri strains DSM 17938 and ATCC PTA 4659 decreased the incidence and severity of NEC, decreased mRNA expression of IL-6, TNF-α, TLR4, and NF-κB and protein expression of TLR4, TNF-α and IL-1β and increased numbers of regulatory T cells in the ileum.64,65 L reuteri also impacts intestinal motility which may be of importance in the premature infant given immaturity of peristaltic activity.66,67

A single strain of L. rhamnosus (GG ATCC 53103) is perhaps the most studied probiotic organism. This strain has antimicrobial activity that appears to be predominately related to lowering the luminal pH through production of lactic acid and other bacterial products.68 L rhamnosus GG has anti-inflammatory effects; the primary mechanism appears to be upregulation of the IL10R2 receptor subunit which decreases expression of TNFα and MIP2 and increases expression of the suppressors of cytokine signaling (SOCS) family.69 L rhamnosus GG also improves intestinal barrier function.70-72 In both the piglet and mouse models of NEC, a different strain of L. rhamnosus (HN001) decreased the incidence and severity of NEC by altering TLR9 signaling.73

Risks of probiotic administration

The risks and drawbacks of administration of probiotics fall into four categories. First, case reports clearly document occurrences of sepsis caused by the administered probiotic microbe. This risk is particularly relevant to premature infants given the immaturity of their intestinal barrier and increased risk of translocation of intestinal microbes into the lymphatic and/or systemic circulation. Among the probiotic products commonly administered to premature infants, there are reports of sepsis from Saccharomyces boulardii in one preterm infant and three term infants (two of these infants were in a bed adjacent to an infant receiving the probiotic),74-76 L. rhamnosus GG in several premature infants and term infants many of whom had either congenital heart disease, gastroschisis, or short gut syndrome,77-82 and in a few infants receiving probiotic bifidobacteria.83-85 It has been argued that the actual incidence of probiotic-induced sepsis is likely under-reported as anaerobic cultures may not be routinely performed in infants receiving probiotics, however among the clinical trials of premature infants reporting mortality and/or culture-negative clinical sepsis, the incidences of both are either decreased or unchanged suggesting that probiotic-induced sepsis is likely very rare.

In adults, bacteremia caused by Lactobacillus species occurs most commonly in the elderly, the immunocompromised, and those with central venous catheters with L. salivarius the most common species in Taiwan86 and L. rhamnosus the most common in Europe.87,88 In Finland, where a database of all positive blood cultures for the entire country is maintained, there was no increase in Lactobacillus bacteremia over a period of years in which use of probiotic lactobacilli increased dramatically.89 In Stockholm, the incidence of Lactobacillus bacteremia remained <1% of all positive blood cultures in spite of marked increases in probiotic consumption and none of the organisms isolated were identical to the probiotic strains commonly available there.90 In a Quebec hospital, administration of a probiotic containing three species of Lactobacillus to every adult patient receiving antibiotics (a total of 44,835 inpatients over a decade) resulted in a dramatic decrease in Clostridium difficile infection with no cases of Lactobacillus bacteremia.91 Infections with L. acidophilus appear to be less common than with L. rhamnosus (including occasional cases of endocarditis).92 Most lactobacilli are sensitive to erythromycin, penicillins, clindamycin and vancomycin and resistant to metronidazole and aminoglycosides, with the most notable exception that many L. rhamnosus strains are resistant to vancomycin.93,94

Bacteremia caused by Bifidobacterium species in adults is much less commonly reported, predominantly affects the immunocompromised, and often presents with fever and abdominal pain.95 It is possible that the incidence of Bifidobacterium bacteremia is underreported due to the challenges of growing this organism in standard culture media, though increased utilization of matrix-assisted laser desorption-ionization–time of flight mass spectrometry may clarify this. The importance of molecular diagnosis to confirm that an ingested probiotic is identical to the organism isolated from the infection has been stressed.96 Bifidobacteria are generally sensitive to ampicillin and vancomycin and resistant to aminoglycosides.97,98

Second, contamination of commercially available products with pathogenic organisms is a possibility that likely varies with the quality control methods employed by the manufacturer. The recent report of a premature infant who received the probiotic ABC Dophilus (containing B. lactis, Streptococcus thermophilus, and L rhamnosus) and developed NEC with evidence of intestinal ischemia from the esophagus to the rectum is particularly disturbing. Histopathologic analysis of resected necrotic bowel revealed invasive fungal infection and fungal DNA from the tissue block demonstrated Rhizopus oryzae. Unopened containers of ABC Dophilus of the same lot as that administered to the infant were found to be contaminated with the same species of fungus.99 The Health Advisory released by the Centers for Disease Control following the death of this infant emphasized that probiotic products available in the U.S. are viewed as dietary supplements and therefore are not evaluated for safety or efficacy by the Food and Drug Administration (FDA) (http://emergency.cdc.gov/HAN/han00373.asp accessed 5 May 2016). It is noteworthy that ABC Dophilus decreased the incidence of NEC in a large multi-center trial with no increase in the incidence of sepsis and no reports of probiotic-related sepsis or mucormycosis.11

Third, several studies have demonstrated that many commercially available probiotic products do not contain the numbers or strains of bacteria advertised on the product label, that there is significant variation between lots of a given product, and that species and strain changes in probiotics are common and often not reflected clearly on the label.100,101 This may be due in part to lack of oversight of probiotic manufacture and to challenges related to the changing taxonomy of probiotic microbes. The result is that it is difficult for the clinician to be certain of the viability and purity of available products and for those reviewing clinical trials to be certain which probiotic was administered. In the U.S., the risks of contamination or suboptimal purity or viability of probiotic microbes could be obviated by production of probiotic products that qualify as drugs rather than dietary supplements and by testing these products under the auspices of Investigational New Drug (IND) oversight by the FDA. U.S. probiotic manufacturers have been hesitant to adopt this approach due to concerns that the IND process would preclude them from selling the probiotic product as a dietary supplement (a much larger market). An appealing alternative would be to follow the lead of the Canadian government and create a new classification for oversight of probiotic products recognizing that probiotics differ significantly from dietary supplements, drugs, and vaccines. The result of the Canadian approach is that our neighbors to the north have access to probiotics that are tested for purity and viability.102

Fourth, probiotic organisms administered to one infant have been identified in the feces of infants in the NICU that did not receive the probiotic.76 The mechanisms of cross-contamination in the NICU have not been completely characterized but likely include transmission through contamination of NICU surfaces and the hands of caregivers. In a trial of B infantis vs B lactis, the former was identified in the stools of infants receiving the latter and confirmed at the strain level.38 The challenges of cross-contamination include both the potential impact on NICU infants and the blunting of any protective effects of probiotic organisms in clinical trials. The largest randomized clinical trial of probiotics in premature infants to date demonstrated cross contamination in all study sites with 49% of the placebo infants colonized with the study probiotic.6 These observations question the value of meta-analyses and of further randomized clinical trials of probiotics. The recent call for large cluster or cross-over cluster randomized trials in which the NICU is randomized rather than the infant is particularly applicable to probiotics and NEC.103

Which Probiotic to prevent NEC?

Clinical trials comparing probiotic strains, doses, and duration of administration are uncommon and have not been powered to detect differences in NEC incidence.104,105 Animal studies and meta-analyses of available trials have been somewhat helpful in comparing differing strains of probiotics and single organism products to multiple organism cocktails. The following general principles have emerged: combination products may have advantages over single organisms;106-108 if a single organism is administered bifidobacteria appear to be more effective than lactobacilli and both are more effective than Saccharomyces;18,106 among bifidobacteria B. infantis has advantages in colonization over other species and subspecies;37 among lactobacilli L acidophilus appears to have advantages over L. reuteri and L rhamnosus GG; and a short course of probiotics (10-14 days early in life with repeat courses following antibiotic administration) may be as useful in NEC prevention as longer courses particularly in the human milk-fed infant.5 Unfortunately comparisons of NEC prevention between studies suffer from the well-established observation that the incidence of NEC varies dramatically over time and from NICU to NICU for unclear reasons.

It is striking that much of the world has embraced routine administration of probiotic microbes to premature infants with positive outcomes while neonatologists in the U.S. and the American Academy of Pediatrics remain hesitant to embrace this approach (somewhat reminiscent of U.S. neonatologists being the last to accept room air resuscitation at birth). While we wait for more definitive studies, based on the conservative assumptions of 3000 NEC cases/year (about 5% of infants with birth weight < 1500 grams) and 600 NEC deaths/year (20% mortality) in the U.S. and a relative risk reduction of 0.5, it becomes feasible to estimate the savings per year of increased probiotic use in lives, meters of intestines, millions of dollars and billions of neurons109 (I come up with 300, 120, 250, and 12000 respectively).

If a neonatologist were to decide in conjunction with the parents after review of the evidence that the potential benefits of decreased risk of NEC, sepsis, death, feeding intolerance, and prolongation of hospital stay outweighed the risks of sepsis and contamination, what products currently available would be most appealing? FloraBaby (contains L. rhamnosus plus four strains of Bifidobacterium: infantis, longum, breve and bifidum) decreased the incidence of NEC in a cohort study and is produced in Canada under the auspices of Health Canada which means that the manufacturers follow good manufacturing practice and the strains are known.16 Infloran (the current version contains L. acidophilus and B. infantis) is produced in Europe and licensed in Switzerland as a drug and has perhaps the best success rate in randomized controlled trials. Natren Life Start powder contains a strain of B. infantis that has been shown to completely metabolize HMOs. Biogaia ProTectis contains a single strain of L. reuteri (the same strain described in the meta-analysis)22, is available as a liquid drop either with or without added Vitamin D, and has been shown in several studies of term infants to decrease colic.110 An alternative would be to encourage the motivated parents to take a probiotic product themselves in hopes of transmitting the desired microbes to their infant.111

Future Directions

Historically, most probiotic microbes were chosen based on ease of manufacture, stability and resistance to gastric acid. Advances in understanding of the “healthy” neonatal microbiota and the impact of changes in these communities of microbes over time in preterm and term infants and mechanisms of action of various probiotic species and strains suggest that next generation probiotics with specific properties may allow targeted manipulations of the gut microbiota. For instance “activated” probiotic microbes with specific desirable genes up-regulated during manufacture may improve colonization or increase specific anti-inflammatory or barrier strengthening effects. Currently probiotic products are limited to a few species of bacteria and a single fungal species. As ability to identify differences in intestinal archaea and viruses (both phages and human viruses) between health and disease develops, it is likely that a broad range of probiotics from multiple kingdoms may become available. Precision medicine is likely to play an important role in individualizing responses to dysbiosis. This will require more rapid and interpretable analysis of the intestinal microbiota or reliable microbial biomarkers of a disordered microbial community. For instance, recent descriptions of a microbiota for age z-score to identify dysmaturation of the intestinal microbiota in undernutrition or obesity may have applicability in the identification of the high risk premature infant microbiome.112 The recent identification of host genetic polymorphisms that influence the intestinal microbiota suggests the possibility of early identification of preterm infants at higher risk for intestinal dysbiosis and NEC (which may require differing probiotic and/or prebiotic strategies).113,114 Furthermore the marked heterogeneity of HMOs between mothers and in a given mother over time and differential effects on the infant intestinal microbiota suggest that some HMOs may have more beneficial effects than others.115 The capacity to measure HMOs in an individual milk sample and then select a paired intervention (augmentation of specific prebiotic HMOs in combination with a probiotic capable of consuming those HMOs) may eventually be feasible.

Conclusion

Probiotic administration has been shown to decrease mortality and NEC in several randomized clinical trials and in combination with human milk appears to represent a promising and relatively safe intervention. Many NICUs throughout the world routinely provide prophylactic probiotics to premature infants with good evidence of success. Improvement in quality control by manufacturers, oversight by governmental agencies in the U.S., and development of new strains of probiotic microbes are urgently needed. Cluster randomized clinical trials comparing commercially available probiotic strains sufficiently powered to determine differences in NEC incidence (including thousands of premature infants in each arm) are needed.

Acknowledgments

Supported in part by a grant from the NIH: HD059127

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Hoyos AB. Reduced incidence of necrotizing enterocolitis associated with enteral administration of Lactobacillus acidophilus and Bifidobacterium infantis to neonates in an intensive care unit. International journal of infectious diseases : IJID : official publication of the International Society for Infectious Diseases. 1999 Summer;3(4):197–202. doi: 10.1016/s1201-9712(99)90024-3. [DOI] [PubMed] [Google Scholar]
  • 2.Vongbhavit K, Underwood MA. Prevention of Necrotizing Enterocolitis Through Manipulation of the Intestinal Microbiota of the Premature Infant. Clinical therapeutics. 2016 Apr;38(4):716–732. doi: 10.1016/j.clinthera.2016.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Xu L, Wang Y, Wang Y, et al. A double-blinded randomized trial on growth and feeding tolerance with Saccharomyces boulardii CNCM I-745 in formula-fed preterm infants. Jornal de pediatria. 2016 Mar 3; doi: 10.1016/j.jped.2015.08.013. [DOI] [PubMed] [Google Scholar]
  • 4.Kanic Z, Micetic Turk D, Burja S, Kanic V, Dinevski D. Influence of a combination of probiotics on bacterial infections in very low birthweight newborns. Wiener klinische Wochenschrift. 2015 Dec;127(5):S210–215. doi: 10.1007/s00508-015-0845-0. [DOI] [PubMed] [Google Scholar]
  • 5.Guthmann F, Arlettaz Mieth RP, Bucher HU, Buhrer C. Short courses of dual-strain probiotics appear to be effective in reducing necrotising enterocolitis. Acta Paediatr. 2016 Mar;105(3):255–259. doi: 10.1111/apa.13280. [DOI] [PubMed] [Google Scholar]
  • 6.Costeloe K, Hardy P, Juszczak E, Wilks M, Millar MR Probiotics in Preterm Infants Study Collaborative G. Bifidobacterium breve BBG-001 in very preterm infants: a randomised controlled phase 3 trial. Lancet. 2016 Feb 13;387(10019):649–660. doi: 10.1016/S0140-6736(15)01027-2. [DOI] [PubMed] [Google Scholar]
  • 7.Oncel MY, Sari FN, Arayici S, et al. Lactobacillus Reuteri for the prevention of necrotising enterocolitis in very low birthweight infants: a randomised controlled trial. Archives of disease in childhood. 2014 Mar;99(2):F110–115. doi: 10.1136/archdischild-2013-304745. [DOI] [PubMed] [Google Scholar]
  • 8.Rojas MA, Lozano JM, Rojas MX, et al. Prophylactic probiotics to prevent death and nosocomial infection in preterm infants. Pediatrics. 2012 Nov;130(5):e1113–1120. doi: 10.1542/peds.2011-3584. [DOI] [PubMed] [Google Scholar]
  • 9.Dani C, Biadaioli R, Bertini G, Martelli E, Rubaltelli FF. Probiotics feeding in prevention of urinary tract infection, bacterial sepsis and necrotizing enterocolitis in preterm infants. A prospective double-blind study. Biology of the neonate. 2002 Aug;82(2):103–108. doi: 10.1159/000063096. [DOI] [PubMed] [Google Scholar]
  • 10.Manzoni P, Meyer M, Stolfi I, et al. Bovine lactoferrin supplementation for prevention of necrotizing enterocolitis in very-low-birth-weight neonates: a randomized clinical trial. Early human development. 2014 Mar;90(1):S60–65. doi: 10.1016/S0378-3782(14)70020-9. [DOI] [PubMed] [Google Scholar]
  • 11.Jacobs SE, Tobin JM, Opie GF, et al. Probiotic Effects on Late-onset Sepsis in Very Preterm Infants: A Randomized Controlled Trial. Pediatrics. 2013 Dec;132(6):1055–1062. doi: 10.1542/peds.2013-1339. [DOI] [PubMed] [Google Scholar]
  • 12.Lin HC, Hsu CH, Chen HL, et al. Oral probiotics prevent necrotizing enterocolitis in very low birth weight preterm infants: a multicenter, randomized, controlled trial. Pediatrics. 2008 Oct;122(4):693–700. doi: 10.1542/peds.2007-3007. [DOI] [PubMed] [Google Scholar]
  • 13.Repa A, Thanhaeuser M, Endress D, et al. Probiotics (Lactobacillus acidophilus and Bifidobacterium bifidum) prevent NEC in VLBW infants fed breast milk but not formula. Pediatric research. 2015 Feb;77(2):381–388. doi: 10.1038/pr.2014.192. [DOI] [PubMed] [Google Scholar]
  • 14.Li D, Rosito G, Slagle T. Probiotics for the prevention of necrotizing enterocolitis in neonates: an 8-year retrospective cohort study. Journal of clinical pharmacy and therapeutics. 2013 Dec;38(6):445–449. doi: 10.1111/jcpt.12084. [DOI] [PubMed] [Google Scholar]
  • 15.Hartel C, Pagel J, Rupp J, et al. Prophylactic use of Lactobacillus acidophilus/Bifidobacterium infantis probiotics and outcome in very low birth weight infants. The Journal of pediatrics. 2014 Aug;165(2):285–289. e281. doi: 10.1016/j.jpeds.2014.04.029. [DOI] [PubMed] [Google Scholar]
  • 16.Janvier A, Malo J, Barrington KJ. Cohort Study of Probiotics in a North American Neonatal Intensive Care Unit. The Journal of pediatrics. 2014 Jan 7;164(5):980–985. doi: 10.1016/j.jpeds.2013.11.025. [DOI] [PubMed] [Google Scholar]
  • 17.Bonsante F, Iacobelli S, Gouyon JB. Routine probiotic use in very preterm infants: retrospective comparison of two cohorts. American journal of perinatology. 2013 Jan;30(1):41–46. doi: 10.1055/s-0032-1321498. [DOI] [PubMed] [Google Scholar]
  • 18.Aceti A, Gori D, Barone G, et al. Probiotics for prevention of necrotizing enterocolitis in preterm infants: systematic review and meta-analysis. Italian journal of pediatrics. 2015;41:89. doi: 10.1186/s13052-015-0199-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Olsen R, Greisen G, Schroder M, Brok J. Prophylactic Probiotics for Preterm Infants: A Systematic Review and Meta-Analysis of Observational Studies. Neonatology. 2016;109(2):105–112. doi: 10.1159/000441274. [DOI] [PubMed] [Google Scholar]
  • 20.Aggarwal S, Topaloglu H, Kumar S. Probiotics Significantly Reduce Mortality In Preterm Newborns With Necrotising Enterocolitis: Results of Large Network Meta-Analysis. Value in health : the journal of the International Society for Pharmacoeconomics and Outcomes Research. 2015 Nov;18(7):A576–577. [Google Scholar]
  • 21.Lau CS, Chamberlain RS. Probiotic administration can prevent necrotizing enterocolitis in preterm infants: A meta-analysis. Journal of pediatric surgery. 2015 Aug;50(8):1405–1412. doi: 10.1016/j.jpedsurg.2015.05.008. [DOI] [PubMed] [Google Scholar]
  • 22.Athalye-Jape G, Rao S, Patole S. Lactobacillus reuteri DSM 17938 as a Probiotic for Preterm Neonates: A Strain-Specific Systematic Review. JPEN Journal of parenteral and enteral nutrition. 2015 Jun 9; doi: 10.1177/0148607115588113. [DOI] [PubMed] [Google Scholar]
  • 23.Alfaleh K, Anabrees J. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Cochrane database of systematic reviews (Online) 2014 Apr 10;4:CD005496. doi: 10.1002/14651858.CD005496.pub4. [DOI] [PubMed] [Google Scholar]
  • 24.Collado MC, Rautava S, Aakko J, Isolauri E, Salminen S. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Scientific reports. 2016;6:23129. doi: 10.1038/srep23129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Payne MS, Bayatibojakhi S. Exploring preterm birth as a polymicrobial disease: an overview of the uterine microbiome. Frontiers in immunology. 2014;5:595. doi: 10.3389/fimmu.2014.00595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ide M, Papapanou PN. Epidemiology of association between maternal periodontal disease and adverse pregnancy outcomes--systematic review. Journal of periodontology. 2013 Apr;84(4 Suppl):S181–194. doi: 10.1902/jop.2013.134009. [DOI] [PubMed] [Google Scholar]
  • 27.Donders GG, Van Calsteren K, Bellen G, et al. Predictive value for preterm birth of abnormal vaginal flora, bacterial vaginosis and aerobic vaginitis during the first trimester of pregnancy. BJOG : an international journal of obstetrics and gynaecology. 2009 Sep;116(10):1315–1324. doi: 10.1111/j.1471-0528.2009.02237.x. [DOI] [PubMed] [Google Scholar]
  • 28.Yu ZT, Chen C, Newburg DS. Utilization of major fucosylated and sialylated human milk oligosaccharides by isolated human gut microbes. Glycobiology. 2013 Nov;23(11):1281–1292. doi: 10.1093/glycob/cwt065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Huda MN, Lewis Z, Kalanetra KM, et al. Stool Microbiota and Vaccine Responses of Infants. Pediatrics. 2014 Aug;134(2):E362–E372. doi: 10.1542/peds.2013-3937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.La Rosa PS, Warner BB, Zhou Y, et al. Patterned progression of bacterial populations in the premature infant gut. Proceedings of the National Academy of Sciences of the United States of America. 2014 Aug 26;111(34):12522–12527. doi: 10.1073/pnas.1409497111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Mai V, Young CM, Ukhanova M, et al. Fecal microbiota in premature infants prior to necrotizing enterocolitis. PloS one. 2011;6(6):e20647. doi: 10.1371/journal.pone.0020647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Claud EC, Keegan KP, Brulc JM, et al. Bacterial community structure and functional contributions to emergence of health or necrotizing enterocolitis in preterm infants. Microbiome. 2013;1(1):20. doi: 10.1186/2049-2618-1-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sela DA, Mills DA. The marriage of nutrigenomics with the microbiome: the case of infant-associated bifidobacteria and milk. The American journal of clinical nutrition. 2014 Mar;99(3):697S–703S. doi: 10.3945/ajcn.113.071795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Arboleya S, Stanton C, Ryan CA, Dempsey E, Ross PR. Bosom Buddies: The Symbiotic Relationship Between Infants and Bifidobacterium longum ssp. longum and ssp. infantis. Genetic and Probiotic Features. Annual review of food science and technology. 2016;7:1–21. doi: 10.1146/annurev-food-041715-033151. [DOI] [PubMed] [Google Scholar]
  • 35.Garrido D, Ruiz-Moyano S, Lemay DG, Sela DA, German JB, Mills DA. Comparative transcriptomics reveals key differences in the response to milk oligosaccharides of infant gut-associated bifidobacteria. Scientific reports. 2015;5:13517. doi: 10.1038/srep13517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ruiz-Moyano S, Totten SM, Garrido DA, et al. Variation in consumption of human milk oligosaccharides by infant gut-associated strains of Bifidobacterium breve. Applied and environmental microbiology. 2013 Oct;79(19):6040–6049. doi: 10.1128/AEM.01843-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Underwood MA, German JB, Lebrilla CB, Mills DA. Bifidobacterium longum subspecies infantis: champion colonizer of the infant gut. Pediatric research. 2015 Jan;77(1-2):229–235. doi: 10.1038/pr.2014.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Underwood MA, Kalanetra KM, Bokulich NA, et al. A comparison of two probiotic strains of bifidobacteria in premature infants. The Journal of pediatrics. 2013 Dec;163(6):1585–1591. e1589. doi: 10.1016/j.jpeds.2013.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wickramasinghe S, Pacheco AR, Lemay DG, Mills DA. Bifidobacteria grown on human milk oligosaccharides downregulate the expression of inflammation-related genes in Caco-2 cells. BMC microbiology. 2015;15:172. doi: 10.1186/s12866-015-0508-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Martinez FA, Balciunas EM, Converti A, Cotter PD, de Souza Oliveira RP. Bacteriocin production by Bifidobacterium spp. A review. Biotechnology advances. 2013 Jul;31(4):482–488. doi: 10.1016/j.biotechadv.2013.01.010. [DOI] [PubMed] [Google Scholar]
  • 41.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. American journal of physiology Gastrointestinal and liver physiology. 2013 Jan 15;304(2):G132–141. doi: 10.1152/ajpgi.00142.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Underwood MA, Arriola J, Gerber CW, et al. Bifidobacterium longum subsp. infantis in experimental necrotizing enterocolitis: alterations in inflammation, innate immune response, and the microbiota. Pediatric research. 2014 Oct;76(4):326–333. doi: 10.1038/pr.2014.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Weng M, Ganguli K, Zhu W, Shi HN, Walker WA. Conditioned media from Bifidobacteria infantis protects against Cronobacter sakazakii-induced intestinal inflammation in newborn mice. American journal of physiology Gastrointestinal and liver physiology. 2014 Mar 13;306(9):6779–6787. doi: 10.1152/ajpgi.00183.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Arboleya S, Salazar N, Solis G, et al. Assessment of intestinal microbiota modulation ability of Bifidobacterium strains in in vitro fecal batch cultures from preterm neonates. Anaerobe. 2013 Feb;19:9–16. doi: 10.1016/j.anaerobe.2012.11.001. [DOI] [PubMed] [Google Scholar]
  • 45.Khailova L, Dvorak K, Arganbright KM, et al. Bifidobacterium bifidum improves intestinal integrity in a rat model of necrotizing enterocolitis. American journal of physiology Gastrointestinal and liver physiology. 2009 Nov;297(5):G940–949. doi: 10.1152/ajpgi.00141.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Khailova L, Mount Patrick SK, Arganbright KM, Halpern MD, Kinouchi T, Dvorak B. Bifidobacterium bifidum reduces apoptosis in the intestinal epithelium in necrotizing enterocolitis. American journal of physiology Gastrointestinal and liver physiology. 2010 Nov;299(5):G1118–1127. doi: 10.1152/ajpgi.00131.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Underwood MA, Kananurak A, Coursodon CF, et al. Bifidobacterium bifidum in a rat model of necrotizing enterocolitis: antimicrobial peptide and protein responses. Pediatric research. 2012 May;71(5):546–551. doi: 10.1038/pr.2012.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Satoh T, Izumi H, Iwabuchi N, et al. Bifidobacterium breve prevents necrotising enterocolitis by suppressing inflammatory responses in a preterm rat model. Beneficial microbes. 2015 Sep 30;:1–8. doi: 10.3920/BM2015.0035. [DOI] [PubMed] [Google Scholar]
  • 49.Izumi H, Minegishi M, Sato Y, Shimizu T, Sekine K, Takase M. Bifidobacterium breve alters immune function and ameliorates DSS-induced inflammation in weanling rats. Pediatric research. 2015 Oct;78(4):407–416. doi: 10.1038/pr.2015.115. [DOI] [PubMed] [Google Scholar]
  • 50.Bidart GN, Rodriguez-Diaz J, Monedero V, Yebra MJ. A unique gene cluster for the utilization of the mucosal and human milk-associated glycans galacto-N-biose and lacto-N-biose in Lactobacillus casei. Molecular microbiology. 2014 Aug;93(3):521–538. doi: 10.1111/mmi.12678. [DOI] [PubMed] [Google Scholar]
  • 51.Bidart GN, Rodriguez-Diaz J, Yebra MJ. The Extracellular Wall-Bound beta-N-Acetylglucosaminidase from Lactobacillus casei Is Involved in the Metabolism of the Human Milk Oligosaccharide Lacto-N-Triose. Applied and environmental microbiology. 2016 Jan;82(2):570–577. doi: 10.1128/AEM.02888-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kacerovsky M, Vrbacky F, Kutova R, et al. Cervical microbiota in women with preterm prelabor rupture of membranes. PloS one. 2015;10(5):e0126884. doi: 10.1371/journal.pone.0126884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Elawadli I, Brisbin JT, Mallard BA, Griffiths MW, Corredig M, Sharif S. Differential effects of lactobacilli on activation and maturation of mouse dendritic cells. Beneficial microbes. 2014 Sep;5(3):323–334. doi: 10.3920/BM2013.0066. [DOI] [PubMed] [Google Scholar]
  • 54.Goncalves FL, Soares LM, Figueira RL, Simoes AL, Gallindo RM, Sbragia L. Evaluation of the expression of I-FABP and L-FABP in a necrotizing enterocolitis model after the use of Lactobacillus acidophilus. Journal of pediatric surgery. 2015 Apr;50(4):543–549. doi: 10.1016/j.jpedsurg.2014.07.007. [DOI] [PubMed] [Google Scholar]
  • 55.Nandakumar R, Talapatra K. Quantitative profiling of bacteriocins present in dairy-free probiotic preparations of Lactobacillus acidophilus by nanoliquid chromatography-tandem mass spectrometry. Journal of dairy science. 2014;97(4):1999–2008. doi: 10.3168/jds.2013-7470. [DOI] [PubMed] [Google Scholar]
  • 56.Awaisheh SS, Al-Nabulsi AA, Osaili TM, Ibrahim S, Holley R. Inhibition of Cronobacter sakazakii by heat labile bacteriocins produced by probiotic LAB isolated from healthy infants. Journal of food science. 2013 Sep;78(9):M1416–1420. doi: 10.1111/1750-3841.12209. [DOI] [PubMed] [Google Scholar]
  • 57.Anjum N, Maqsood S, Masud T, Ahmad A, Sohail A, Momin A. Lactobacillus acidophilus: characterization of the species and application in food production. Critical reviews in food science and nutrition. 2014;54(9):1241–1251. doi: 10.1080/10408398.2011.621169. [DOI] [PubMed] [Google Scholar]
  • 58.Borthakur A, Bhattacharyya S, Kumar A, Anbazhagan AN, Tobacman JK, Dudeja PK. Lactobacillus acidophilus alleviates platelet-activating factor-induced inflammatory responses in human intestinal epithelial cells. PloS one. 2013;8(10):e75664. doi: 10.1371/journal.pone.0075664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kumar A, Alrefai WA, Borthakur A, Dudeja PK. Lactobacillus acidophilus counteracts enteropathogenic E. coli-induced inhibition of butyrate uptake in intestinal epithelial cells. American journal of physiology Gastrointestinal and liver physiology. 2015 Oct 1;309(7):G602–607. doi: 10.1152/ajpgi.00186.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Guo S, Guo Y, Ergun A, Lu L, Walker WA, Ganguli K. Secreted Metabolites of Bifidobacterium infantis and Lactobacillus acidophilus Protect Immature Human Enterocytes from IL-1beta-Induced Inflammation: A Transcription Profiling Analysis. PloS one. 2015;10(4):e0124549. doi: 10.1371/journal.pone.0124549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Spinler JK, Taweechotipatr M, Rognerud CL, Ou CN, Tumwasorn S, Versalovic J. Human-derived probiotic Lactobacillus reuteri demonstrate antimicrobial activities targeting diverse enteric bacterial pathogens. Anaerobe. 2008 Jun;14(3):166–171. doi: 10.1016/j.anaerobe.2008.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yang Y, Zhao X, Le MH, Zijlstra RT, Ganzle MG. Reutericyclin producing Lactobacillus reuteri modulates development of fecal microbiota in weanling pigs. Frontiers in microbiology. 2015;6:762. doi: 10.3389/fmicb.2015.00762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lin YP, Thibodeaux CH, Pena JA, Ferry GD, Versalovic J. Probiotic Lactobacillus reuteri suppress proinflammatory cytokines via c-Jun. Inflammatory bowel diseases. 2008 Aug;14(8):1068–1083. doi: 10.1002/ibd.20448. [DOI] [PubMed] [Google Scholar]
  • 64.Liu Y, Fatheree NY, Mangalat N, Rhoads JM. Lactobacillus reuteri strains reduce incidence and severity of experimental necrotizing enterocolitis via modulation of TLR4 and NF-kappaB signaling in the intestine. American journal of physiology Gastrointestinal and liver physiology. 2012 Mar 15;302(6):G608–617. doi: 10.1152/ajpgi.00266.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Liu Y, Fatheree NY, Dingle BM, Tran DQ, Rhoads JM. Lactobacillus reuteri DSM 17938 changes the frequency of Foxp3+ regulatory T cells in the intestine and mesenteric lymph node in experimental necrotizing enterocolitis. PloS one. 2013;8(2):e56547. doi: 10.1371/journal.pone.0056547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kunze WA, Mao YK, Wang B, et al. Lactobacillus reuteri enhances excitability of colonic AH neurons by inhibiting calcium-dependent potassium channel opening. Journal of cellular and molecular medicine. 2009 Aug;13(8B):2261–2270. doi: 10.1111/j.1582-4934.2009.00686.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wu RY, Pasyk M, Wang B, et al. Spatiotemporal maps reveal regional differences in the effects on gut motility for Lactobacillus reuteri and rhamnosus strains. Neurogastroenterology and motility : the official journal of the European Gastrointestinal Motility Society. 2013 Mar;25(3):e205–214. doi: 10.1111/nmo.12072. [DOI] [PubMed] [Google Scholar]
  • 68.Fayol-Messaoudi D, Berger CN, Coconnier-Polter MH, Lievin-Le Moal V, Servin AL. pH-, Lactic acid-, and non-lactic acid-dependent activities of probiotic Lactobacilli against Salmonella enterica Serovar Typhimurium. Applied and environmental microbiology. 2005 Oct;71(10):6008–6013. doi: 10.1128/AEM.71.10.6008-6013.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Mirpuri J, Sotnikov I, Myers L, et al. Lactobacillus rhamnosus (LGG) regulates IL-10 signaling in the developing murine colon through upregulation of the IL-10R2 receptor subunit. PloS one. 2012;7(12):e51955. doi: 10.1371/journal.pone.0051955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lin PW, Nasr TR, Berardinelli AJ, Kumar A, Neish AS. The probiotic Lactobacillus GG may augment intestinal host defense by regulating apoptosis and promoting cytoprotective responses in the developing murine gut. Pediatric research. 2008 Nov;64(5):511–516. doi: 10.1203/PDR.0b013e3181827c0f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Patel RM, Myers LS, Kurundkar AR, Maheshwari A, Nusrat A, Lin PW. Probiotic bacteria induce maturation of intestinal claudin 3 expression and barrier function. The American journal of pathology. 2012 Feb;180(2):626–635. doi: 10.1016/j.ajpath.2011.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yan F, Polk DB. Characterization of a probiotic-derived soluble protein which reveals a mechanism of preventive and treatment effects of probiotics on intestinal inflammatory diseases. Gut microbes. 2012 Jan-Feb;3(1):25–28. doi: 10.4161/gmic.19245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Good M, Sodhi CP, Ozolek JA, et al. Lactobacillus rhamnosus HN001 decreases the severity of necrotizing enterocolitis in neonatal mice and preterm piglets: evidence in mice for a role of TLR9. American journal of physiology Gastrointestinal and liver physiology. 2014 Jun 1;306(11):G1021–1032. doi: 10.1152/ajpgi.00452.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chioukh FZ, Ben Hmida H, Ben Ameur K, Toumi A, Monastiri K. Saccharomyces cerevisiae fungemia in a premature neonate treated receiving probiotics. Medecine et maladies infectieuses. 2013 Aug;43(8):359–360. doi: 10.1016/j.medmal.2013.06.008. [DOI] [PubMed] [Google Scholar]
  • 75.Belet N, Dalgic N, Oncel S, et al. Catheter-related fungemia caused by Saccharomyces cerevisiae in a newborn. The Pediatric infectious disease journal. 2005 Dec;24(12):1125. doi: 10.1097/01.inf.0000189984.51944.cd. [DOI] [PubMed] [Google Scholar]
  • 76.Perapoch J, Planes AM, Querol A, et al. Fungemia with Saccharomyces cerevisiae in two newborns, only one of whom had been treated with ultra-levura. European journal of clinical microbiology & infectious diseases : official publication of the European Society of Clinical Microbiology. 2000 Jun;19(6):468–470. doi: 10.1007/s100960000295. [DOI] [PubMed] [Google Scholar]
  • 77.Dani C, Coviello CC, Corsini II, Arena F, Antonelli A, Rossolini GM. Lactobacillus Sepsis and Probiotic Therapy in Newborns: Two New Cases and Literature Review. AJP reports. 2016 Mar;6(1):e25–29. doi: 10.1055/s-0035-1566312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Brecht M, Garg A, Longstaff K, Cooper C, Andersen C. Lactobacillus Sepsis following a Laparotomy in a Preterm Infant: A Note of Caution. Neonatology. 2016;109(3):186–189. doi: 10.1159/000441965. [DOI] [PubMed] [Google Scholar]
  • 79.Kunz AN, Noel JM, Fairchok MP. Two cases of Lactobacillus bacteremia during probiotic treatment of short gut syndrome. Journal of pediatric gastroenterology and nutrition. 2004 Apr;38(4):457–458. doi: 10.1097/00005176-200404000-00017. [DOI] [PubMed] [Google Scholar]
  • 80.Land MH, Rouster-Stevens K, Woods CR, Cannon ML, Cnota J, Shetty AK. Lactobacillus sepsis associated with probiotic therapy. Pediatrics. 2005 Jan;115(1):178–181. doi: 10.1542/peds.2004-2137. [DOI] [PubMed] [Google Scholar]
  • 81.De Groote MA, Frank DN, Dowell E, Glode MP, Pace NR. Lactobacillus rhamnosus GG bacteremia associated with probiotic use in a child with short gut syndrome. The Pediatric infectious disease journal. 2005 Mar;24(3):278–280. doi: 10.1097/01.inf.0000154588.79356.e6. [DOI] [PubMed] [Google Scholar]
  • 82.Sadowska-Krawczenko I, Paprzycka M, Korbal P, et al. Lactobacillus rhamnosus GG suspected infection in a newborn with intrauterine growth restriction. Beneficial microbes. 2014 Dec;5(4):397–402. doi: 10.3920/BM2013.0074. [DOI] [PubMed] [Google Scholar]
  • 83.Bertelli C, Pillonel T, Torregrossa A, et al. Bifidobacterium longum bacteremia in preterm infants receiving probiotics. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2015 Mar 15;60(6):924–927. doi: 10.1093/cid/ciu946. [DOI] [PubMed] [Google Scholar]
  • 84.Zbinden A, Zbinden R, Berger C, Arlettaz R. Case series of Bifidobacterium longum bacteremia in three preterm infants on probiotic therapy. Neonatology. 2015;107(1):56–59. doi: 10.1159/000367985. [DOI] [PubMed] [Google Scholar]
  • 85.Ohishi A, Takahashi S, Ito Y, et al. Bifidobacterium septicemia associated with postoperative probiotic therapy in a neonate with omphalocele. The Journal of pediatrics. 2010 Apr;156(4):679–681. doi: 10.1016/j.jpeds.2009.11.041. [DOI] [PubMed] [Google Scholar]
  • 86.Lee MR, Tsai CJ, Liang SK, Lin CK, Huang YT, Hsueh PR. Clinical characteristics of bacteraemia caused by Lactobacillus spp. and antimicrobial susceptibilities of the isolates at a medical centre in Taiwan, 2000-2014. International journal of antimicrobial agents. 2015 Oct;46(4):439–445. doi: 10.1016/j.ijantimicag.2015.06.017. [DOI] [PubMed] [Google Scholar]
  • 87.Gouriet F, Million M, Henri M, Fournier PE, Raoult D. Lactobacillus rhamnosus bacteremia: an emerging clinical entity. European journal of clinical microbiology & infectious diseases : official publication of the European Society of Clinical Microbiology. 2012 Sep;31(9):2469–2480. doi: 10.1007/s10096-012-1599-5. [DOI] [PubMed] [Google Scholar]
  • 88.Cannon JP, Lee TA, Bolanos JT, Danziger LH. Pathogenic relevance of Lactobacillus: a retrospective review of over 200 cases. European journal of clinical microbiology & infectious diseases : official publication of the European Society of Clinical Microbiology. 2005 Jan;24(1):31–40. doi: 10.1007/s10096-004-1253-y. [DOI] [PubMed] [Google Scholar]
  • 89.Salminen MK, Tynkkynen S, Rautelin H, et al. Lactobacillus bacteremia during a rapid increase in probiotic use of Lactobacillus rhamnosus GG in Finland. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2002 Nov 15;35(10):1155–1160. doi: 10.1086/342912. [DOI] [PubMed] [Google Scholar]
  • 90.Sullivan A, Nord CE. Probiotic lactobacilli and bacteraemia in Stockholm. Scandinavian journal of infectious diseases. 2006;38(5):327–331. doi: 10.1080/00365540500449826. [DOI] [PubMed] [Google Scholar]
  • 91.Maziade PJ, Pereira P, Goldstein EJ. A Decade of Experience in Primary Prevention of Clostridium difficile Infection at a Community Hospital Using the Probiotic Combination Lactobacillus acidophilus CL1285, Lactobacillus casei LBC80R, and Lactobacillus rhamnosus CLR2 (Bio-K+) Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2015 May 15;60(2):S144–147. doi: 10.1093/cid/civ178. [DOI] [PubMed] [Google Scholar]
  • 92.Salvana EM, Frank M. Lactobacillus endocarditis: case report and review of cases reported since 1992. The Journal of infection. 2006 Jul;53(1):e5–e10. doi: 10.1016/j.jinf.2005.10.005. [DOI] [PubMed] [Google Scholar]
  • 93.Goldstein EJ, Tyrrell KL, Citron DM. Lactobacillus species: taxonomic complexity and controversial susceptibilities. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2015 May 15;60(2):S98–107. doi: 10.1093/cid/civ072. [DOI] [PubMed] [Google Scholar]
  • 94.Zhou JS, Pillidge CJ, Gopal PK, Gill HS. Antibiotic susceptibility profiles of new probiotic Lactobacillus and Bifidobacterium strains. International journal of food microbiology. 2005 Feb 1;98(2):211–217. doi: 10.1016/j.ijfoodmicro.2004.05.011. [DOI] [PubMed] [Google Scholar]
  • 95.Weber E, Reynaud Q, Suy F, et al. Bifidobacterium species bacteremia: risk factors in adults and infants. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2015 Aug 1;61(3):482–484. doi: 10.1093/cid/civ347. [DOI] [PubMed] [Google Scholar]
  • 96.Aroutcheva A, Auclair J, Frappier M, et al. Importance of Molecular Methods to Determine Whether a Probiotic is the Source of Lactobacillus Bacteremia. Probiotics and antimicrobial proteins. 2016 Mar;8(1):31–40. doi: 10.1007/s12602-016-9209-y. [DOI] [PubMed] [Google Scholar]
  • 97.Xiao JZ, Takahashi S, Odamaki T, Yaeshima T, Iwatsuki K. Antibiotic susceptibility of bifidobacterial strains distributed in the Japanese market. Bioscience, biotechnology, and biochemistry. 2010;74(2):336–342. doi: 10.1271/bbb.90659. [DOI] [PubMed] [Google Scholar]
  • 98.Masco L, Van Hoorde K, De Brandt E, Swings J, Huys G. Antimicrobial susceptibility of Bifidobacterium strains from humans, animals and probiotic products. The Journal of antimicrobial chemotherapy. 2006 Jul;58(1):85–94. doi: 10.1093/jac/dkl197. [DOI] [PubMed] [Google Scholar]
  • 99.Vallabhaneni S, Walker TA, Lockhart SR, et al. Notes from the Field: Fatal Gastrointestinal Mucormycosis in a Premature Infant Associated with a Contaminated Dietary Supplement — Connecticut, 2014. Morbidity and Mortality Weekly Report. 2015;64(6):155–156. [PMC free article] [PubMed] [Google Scholar]
  • 100.Lewis ZT, Shani G, Masarweh CF, et al. Validating bifidobacterial species and subspecies identity in commercial probiotic products. Pediatric research. 2016 Mar;79(3):445–452. doi: 10.1038/pr.2015.244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Marcobal A, Underwood MA, Mills DA. Rapid determination of the bacterial composition of commercial probiotic products by terminal restriction fragment length polymorphism analysis. Journal of pediatric gastroenterology and nutrition. 2008 May;46(5):608–611. doi: 10.1097/MPG.0b013e3181660694. [DOI] [PubMed] [Google Scholar]
  • 102.Janvier A, Lantos J, Barrington K. The politics of probiotics: probiotics, necrotizing enterocolitis and the ethics of neonatal research. Acta Paediatr. 2013 Feb;102(2):116–118. doi: 10.1111/apa.12083. [DOI] [PubMed] [Google Scholar]
  • 103.Tarnow-Mordi W, Cruz M, Morris J. Design and conduct of a large obstetric or neonatal randomized controlled trial. Seminars in fetal & neonatal medicine. 2015 Dec;20(6):389–402. doi: 10.1016/j.siny.2015.10.001. [DOI] [PubMed] [Google Scholar]
  • 104.Hays S, Jacquot A, Gauthier H, et al. Probiotics and growth in preterm infants: A randomized controlled trial, PREMAPRO study. Clinical nutrition (Edinburgh, Scotland) 2015 Jul 16; doi: 10.1016/j.clnu.2015.06.006. [DOI] [PubMed] [Google Scholar]
  • 105.Underwood MA, Salzman NH, Bennett SH, et al. A randomized placebo-controlled comparison of 2 prebiotic/probiotic combinations in preterm infants: impact on weight gain, intestinal microbiota, and fecal short-chain fatty acids. Journal of pediatric gastroenterology and nutrition. 2009 Feb;48(2):216–225. doi: 10.1097/MPG.0b013e31818de195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Patel RM, Denning PW. Therapeutic use of prebiotics, probiotics, and postbiotics to prevent necrotizing enterocolitis: what is the current evidence? Clinics in perinatology. 2013 Mar;40(1):11–25. doi: 10.1016/j.clp.2012.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wu SF, Chiu HY, Chen AC, Lin HY, Lin HC, Caplan M. Efficacy of different probiotic combinations on death and necrotizing enterocolitis in a premature rat model. Journal of pediatric gastroenterology and nutrition. 2013 Jul;57(1):23–28. doi: 10.1097/MPG.0b013e3182929210. [DOI] [PubMed] [Google Scholar]
  • 108.Shiou SR, Yu Y, Guo Y, et al. Synergistic protection of combined probiotic conditioned media against neonatal necrotizing enterocolitis-like intestinal injury. PloS one. 2013;8(5):e65108. doi: 10.1371/journal.pone.0065108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Barrington KJ, Janvier A. Reply: To PMID 24411521. The Journal of pediatrics. 2015 Feb;166(2):502–503. doi: 10.1016/j.jpeds.2014.10.063. [DOI] [PubMed] [Google Scholar]
  • 110.Schreck Bird A, Gregory PJ, Jalloh MA, Risoldi Cochrane Z, Hein DJ. Probiotics for the Treatment of Infantile Colic: A Systematic Review. Journal of pharmacy practice. 2016 Mar 2; doi: 10.1177/0897190016634516. [DOI] [PubMed] [Google Scholar]
  • 111.Benor S, Marom R, Ben Tov A, Armoni Domany K, Zaidenberg-Israeli G, Dollberg S. Probiotic supplementation in mothers of very low birth weight infants. American journal of perinatology. 2014 Jun;31(6):497–504. doi: 10.1055/s-0033-1353490. [DOI] [PubMed] [Google Scholar]
  • 112.Blanton LV, Charbonneau MR, Salih T, et al. Gut bacteria that prevent growth impairments transmitted by microbiota from malnourished children. Science. 2016 Feb 19;351(6275) doi: 10.1126/science.aad3311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Morrow AL, Meinzen-Derr J, Huang P, et al. Fucosyltransferase 2 non-secretor and low secretor status predicts severe outcomes in premature infants. The Journal of pediatrics. 2011 May;158(5):745–751. doi: 10.1016/j.jpeds.2010.10.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Hartel C, Hartz A, Pagel J, et al. NOD2 Loss-of-Function Mutations and Risks of Necrotizing Enterocolitis or Focal Intestinal Perforation in Very Low-birth-weight Infants. Inflammatory bowel diseases. 2016 Feb;22(2):249–256. doi: 10.1097/MIB.0000000000000658. [DOI] [PubMed] [Google Scholar]
  • 115.Underwood MA, Gaerlan S, De Leoz ML, et al. Human milk oligosaccharides in premature infants: absorption, excretion, and influence on the intestinal microbiota. Pediatric research. 2015 Dec;78(6):670–677. doi: 10.1038/pr.2015.162. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES