Abstract
In spite of four decades of research, necrotizing enterocolitis (NEC) remains the most common gastrointestinal complication in premature infants with high mortality and long-term morbidity. The composition of the intestinal microbiota of the premature infant differs dramatically from that of the healthy term infant and appears to be an important risk factor for NEC. Promising NEC prevention strategies that alter the intestinal microbiota include probiotics, prebiotics, synbiotics, lacteroferrin, and human milk feeding.
Keywords: human milk, probiotic, prebiotic, synbiotic, lactoferrin, necrotizing enterocolitis, premature infant
Introduction
Necrotizing enterocolitis (NEC) is a common and devastating disease of premature infants. It affects approximately 7% of infants weighing between 500 and 1,500 g with mortality rates as high as 30%1. The pathophysiology of NEC has been an area of active study for four decades. Current thinking suggests that NEC is not a single disease or infection but the final pathway of a variety of insults. Risk factors include prematurity of the innate and adaptive immune responses (e.g. a poorly regulated inflammatory responses and alterations in intestinal permeability, motility, apoptosis, and autophagy), enteral feeding, an altered intestinal microbiota and variation in intestinal perfusion1-3. The current clinical staging of NEC was initially proposed by Bell4 and modified by Walsh5 and has endured for three decades. The challenges of this classification include disagreements among experts as to the clinical relevance of stage 1 NEC (resulting in variation in inclusion of stage 1 NEC in reports of clinical trials and cohort studies), the lack of distinction between NEC and spontaneous ileal perforation without necrosis, and a lack of evidence regarding applicability to term infants with NEC. Treatment of NEC has changed little over the decades: bowel rest, broad-spectrum antibiotics, parenteral nutrition, with support of ventilation and blood pressure and either peritoneal drainage or resection of necrotic bowel in severe cases. There is significant short-term morbidity, including abnormal bowel function, prolonged parenteral nutrition requiring central line placement, and longer lengths of stay in hospital with significantly higher costs6. Long term morbidity includes poor growth, malabsorption and delays in neurodevelopment7.
Two compelling observations shed light on the pathogenesis of NEC. First, the onset of NEC is generally at 2-6 weeks of life and tends to occur later in the most premature infants with the highest risk of NEC at 29-33 weeks corrected gestational age8. This observation supports the hypothesis that a certain level of “maturation” of the immune system is required for NEC pathogenesis. It is likely not coincidental that the Paneth cells of the small intestine become functional at about this time. These sentinels of the crypts of Lieberkuhn shape the composition of the intestinal microbiota and protect the intestinal stem cells from injury9, 10. Second, small but carefully performed studies demonstrate alterations in the intestinal microbiota prior to the onset of NEC. The term dysbiosis implies an alteration in the composition of the intestinal microbiota and/or microbiome related to disease. Independent investigators have demonstrated that an early predominance of Firmicutes (particularly Clostridiaceae) in the first weeks of life predisposes to NEC and that a sudden bloom of Proteobacteria (particularly Enterobacteriaceae) is common in the days just prior to the onset of NEC11-14. The latter observation is particularly compelling in light of the capacity of several Enterobacteriaceae to trigger an inflammatory response and then outcompete other commensal bacteria by selective consumption of the products of inflammation15. In this article, we will touch briefly on the causes of dysbiosis in the premature infant and review the efficacy of attempts to prevent NEC by dietary interventions designed to correct dysbiosis including probiotics, prebiotics, synbiotics, lactoferrin, and human milk.
Gut colonization and dysbiosis in premature infants
For many years, accepted dogma maintained that the in utero environment was sterile and that the intestinal tract of the fetus was not colonized with bacteria until the time of rupture of membranes. Recent studies suggest that the fetal membranes are not impermeable to bacteria and that many fetuses are exposed to microbes in the amniotic fluid before delivery16-18. The impact of this early exposure is unclear. While early colonizers of the infant gut are heavily influenced by mode of delivery19, the “second wave” of colonists in term infants is mostly determined by feeding type with breast fed infants dominated by bifidobacteria and bacteroides and formula fed infants dominated by streptococci, staphylococci and lactobacilli20. The “second wave” of gut colonists in premature infants is less influenced by type of feeding and differs markedly from that of term infants with high numbers of Clostridiaceae and Enterobacteriacea and relatively low numbers of Bifidobacteriaceae and Bacteroidetes21-25. Perhaps the most important influence on the composition of the premature gut microbiota is degree of prematurity26. The use of acid suppressive medication delays intestinal transit time, alters the intestinal microbiota27 and increases the risk of NEC28. In addition, antibiotic administration leads to changes in the composition of the gut microbiota, suppressing growth of both commensal and pathogenic bacteria, and increases the risk of NEC25, 29, 30. In spite of (or perhaps in part related to) aggressive cleaning protocols, the NICU environment is an important source of pathogenic organisms and influences intestinal colonization of infants with prolonged hospitalizations31. Other potential influences on the intestinal microbiota of premature infants include duration of feeding tubes, kangaroo skin-to-skin care, periods of gut rest, administration of colostrum to the buccal mucosa32 and genetic factors (e.g. common mutations in the FUT2 gene)33, 34. Among the many factors predisposing to dysbiosis in premature infants, those with clear associations with NEC include degree of prematurity26, formula feeding35, antibiotics29, 36, and acid-blocking agents28. The concept of altering the intestinal microbiota or correcting dysbiosis to decrease risk of NEC is promising. We will review five overlapping strategies: probiotics, prebiotics, synbiotics, lactoferrin and human milk (Figure 1).
Figure 1.
Dietary and supplemental strategies for altering the intestinal microbiota of the premature infant. Shading represents areas of overlap.
Probiotics
Probiotics are biological formulations or dietary supplements containing living microorganisms, most commonly one or more of the following genera: Bifidobacterium, Lactobacillus, Streptococcus, Escherichia, or Saccharomyces37, 38. Most currently available probiotics were selected because of their ease of production, stability, or food-preservative properties, rather than based on a specific mechanism of disease prevention. Mounting evidence suggests that in addition to influencing the composition and diversity of the intestinal microbiota, probiotic microbes influence the host innate and adaptive immune systems through a variety of mechanisms. Many of these mechanisms appear to be species, subspecies, or even strain specific. For example, three species of Bifidobacterium decrease incidence and severity of NEC in animal models39-41 but appear to utilize different mechanisms: B. longum subsp infantis attenuates induction of IL6, IL8, TNFα and IL23 in the rat NEC model39, decreases IL1β induced IL8 and IL6 expression in immature human gut xenografts42, and has a competitive advantage over other gut microbes in the presence of human milk oligosaccharides43; B. bifidum improves barrier function44, decreases apoptosis45 and attenuates IL6 induction in the rat NEC model40 and alters short chain fatty acid production in vitro in feces from premature infants46; and B. breve decreases inflammation in the rat NEC model41 and alters butyrate production46 and increases serum levels of TGFβ expression in premature infants47. Lactobacilli also show diversity of function with 3 species that decrease NEC in animal models48-50 with different mechanisms: L. acidophilus secretes one or more molecules that inhibit induction of inflammation by platelet activating factor51 and alters expression of hundreds of genes important in apoptosis, angiogenesis, and immune response52; L. reuteri decreases expression of IL6 and TNFα and increases ileal regulatory T cells in the rat NEC model53 and increases intestinal motility54; L. rhamnosus (strain GG, ATCC 53103) decreases expression of TNFα and MIP2 through upregulation of the IL10 receptor55 and decreases intestinal permeability56 through both increased expression of tight junction proteins57 and decreased apoptosis58, while a different strain (HN001) decreases incidence and severity of NEC in both a mouse and a piglet model, through alterations in TLR9 signaling49.
Multiple randomized placebo-controlled clinical trials (RCT) and cohort studies of probiotics in premature infants have been performed. A recent meta-analysis of 20 RCTs found probiotics to decrease the risk of NEC (OR 0.43, 95% CI 0.31-0.56) and death (OR 0.65, 95% CI 0.52-0.81) in this high risk population37. A meta-analysis of 12 cohort studies including more than 10,000 premature infants found similar rates of protection (RR for NEC 0.55, 95% CI 0.39-0.78 and RR for death 0.72, 95% CI 0.61-0.85)59. Tables 147, 60-91 and 292-101 summarize English language RCTs and cohort studies in premature infants that included NEC, sepsis, or death as a reported outcome. The recent publication of the much awaited PiPS trial60 which showed no improvement in NEC, sepsis, or mortality in 1315 premature infants with gestational age 23-30 weeks randomized to receive either B. breve (strain BBG-001) or placebo underscores the importance of determining the best species and strain of probiotic for NEC prevention and that this choice may differ based on populations and genetics. Clinical trials comparing probiotic species or strains in premature infants are needed. Given the significant challenges, including the large required sample size and the high rates of cross-contamination, cluster-randomized trials may be of particular value69.
Table 1.
Summary of probiotic RCTs in premature infants
| Author Year |
Country | Probiotic Species (strain) |
Brand (Company) |
Population | Dose × duration |
Number enrolled |
NEC cases Stage 2, 3 |
Culture + sepsis cases |
Deaths | ||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Prob | Cont | Prob | Cont | Prob | Cont | Prob | Cont | ||||||
| Costeloe 201560 | UK | B breve (BBG 001) | NR (Yakult) | GA 23-30w | 1.6e8-1.6e9/d until 36w | 650 | 660 | 62 | 66 | 73 | 77 | 54 | 56 |
| Dilli 201561 | Turkey | B lactis (NR) | Maflor (Mamse l) | BW < 1500g + GA < 32w | 5e9/d × 8w | 100 | 100 | 2 | 18 | 8 | 13 | 3 | 12 |
| Saengtawes in 201562 | Thailand | L acidophilus + B bifida (NR) | Infloran (NR) | BW < 1500g + GA < 34w | 2e9 BID × 6w | 31 | 29 | 1 | 1 | 2 | 1 | 0 | 0 |
| Tewari 201563 | India | Bacillus clausii (NR) | Enterogermina (Sanofi Aventis) | GA < 34w | 8e8 TID × 3-5w | 123 | 121 | 0 | 0 | 20 | 25 | 12 | 14 |
| Hays 201564 | France | B lactis + B longum(NR) | NR | BW 700-1600g + GA 25-31w | 1e9/d × 4-6w | 48 | 52 | 5 | 3 | 8 | 10 | 1 | 1 |
| B lactis (NR) | 50 | 2 | 9 | 1 | |||||||||
| B longum (NR) | 49 | 1 | 8 | 3 | |||||||||
| Van Niekirk 201565 | South Africa | B infantis (NR) + L rhamnosus (GG) | Pro-B2 (C Pharm) | BW < 1250g | 7e8/d × 28d | 91 | 93 | 0 | 4 | 15 | 10 | 5 | 7 |
| Dutta 201566 | India | L acidophilus + L rhamnos | NR (Aristo) | GA 27-33 w | 1e10 BID × 21d us + B longum + S boulardii (NR) | 38 | 35 | 1 | 0 | 3 | 6 | 3 | 2 |
| 1e10 BID × 14d | 38 | 3 | 1 | 3 | |||||||||
| 1e9 BID × 21d | 38 | 2 | 6 | 2 | |||||||||
| Patole 201467 | Australia | B breve (M-16V) | NR (Morinaga) | BW < 1500g + GA < 33w | 3e9/d until 37w | 79 | 80 | 0 | 1 | 17 | 12 | 0 | 0 |
| Oncel 201468 | Turkey | L reuteri (DSM 17938) | NR (Biogaia AB) | BW < 1500g + GA < 32w | 1e8/d until discharge | 200 | 200 | 8 | 10 | 13 | 25 | 15 | 20 |
| Totsu 201469 | Japan | B bifidum (OLB 6378) | NR (Meiji) | BW < 1500g | 1.25e 9 BID until > 2kg | 153 | 130 | 0 | 0 | 6 | 10 | 2 | 0 |
| Jacobs 201370 | Australia + NZ | B infantis (BB-02) + S thermophiles (TH-415957) + B lactis (BB-1215954) | ABC Dophilus (Solgar) | BW < 1500g + GA < 32w | 1e9/d until discharge | 548 | 551 | 11 | 24 | 72 | 89 | 27 | 28 |
| Serce 201371 | Turkey | Saccharomyces boulardii (NR) | Reflor (Biocodex) | BW < 1500g + GA < 32w | 5e8/kg BID until discharge | 104 | 104 | 7 | 7 | 72 | 89 | 27 | 28 |
| Demirel 201372 | Turkey | Saccharomyces boulardii (NR) | Reflor (Biocodex) | BW < 1500g + GA < 32w | 5e9/kg BID until discharge | 135 | 136 | 6 | 7 | 20 | 21 | 5 | 5 |
| Rojas 201273 | Colombia | L reuteri(DSM 17938) | NR (Biogaia AB) | BW < 2000g | 1e8/d until discharge | 372 | 378 | 9 | 15 | 24 | 17 | 22 | 28 |
| Fernandez-Carrocera 201274 | Mexico | L acidophilus + L rhamnosus + L casei + L plantarum + B infantis + S thermophiles (NR) | 107M96 y 106M96 (Italmex) | BW < 1500g | 3e9/d (NR) | 75 | 75 | 6 | 12 | NR | NR | 1 | 7 |
| Al-Hosni 201275 | USA | B infantis (NR) + L rhamnosus (GG) | Align (Proctor and Gamble) + Culturelle (Amerifit) | BW 501-1000g | 1e9/d until 34w | 50 | 50 | 2 | 2 | 13 | 16 | 3 | 4 |
| Sari 201176 | Turkey | L sporogenes (NR) | NR (DMG Italia) | BW < 1500g or GA <33w | 3.5e8/d (NR) | 110 | 111 | 6 | 10 | NR | NR | 3 | 4 |
| Braga 201177 | Brazil | L casei + B breve (NR) | NR (Yakult) | BW 750-1499g | 3.5e7-3.5e9/d × 28d | 119 | 112 | 0 | 4 | NR | NR | 26 | 27 |
| Romeo 201178 | Italy | L reuteri (ATCC 55730) | NR | BW < 2500g + GA < 37w | 1e8/d until discharge | 83 | 83 | NR | NR | 1 | 9 | NR | NR |
| L rhamnosus (ATCC 53103 | NR | 6e9/d until discharge | 83 | NR | 2 | NR | |||||||
| Awad 201079 | Egypt | L rhamnosus (GG) | Lacteol Fort (Axcan) | GA 28-41w | 6e9 BID until discharge | 60 | 30 | 0 | 5 | NR | NR | NR | NR |
| Killed L rhamnosus (GG) | 60 | 1 | NR | NR | |||||||||
| Mihatsch 201080 | Germany | B lactis (BB12) | NR (Nestle) | BW < 1500g + GA < 30w | 2e9/kg q 4h × 6w | 91 | 89 | 2 | 4 | 28 | 29 | 2 | 1 |
| Samanta 200981 | India | B longum + B infantis + B bifidum + L acidophilus (NR) | NR | BW < 1500g + GA < 32w | 1e10 BID until discharge | 91 | 95 | 5 | 15 | 13 | 28 | 4 | 14 |
| Rouge 200982 | France | B longum (BB536) + L rhamnosus (GG) | NR (Morina + Valio) | BW < 1500g + GA < 32w | 1e8 QID until discharge | 45 | 49 | 2 | 1 | 15 | 13 | 2 | 4 |
| Lin 200883 | Taiwan | L acidophilus (NCDO 1748) + B bifidum (NCDO 1453) | Infloran (Laboratorio Farmaceutico) | BW < 1500g + GA < 34w | 1e9 BID until 6w | 217 | 217 | 4 | 14 | 40 | 24 | 2 | 9 |
| Stratiki 200784 | Greece | B lactis (NR) | Prenan (Nestle) | GA 27-37w | varied with feeding volume × 30d | 41 | 36 | 0 | 3 | 0 | 3 | NR | NR |
| Wang 200785 | Japan | B breve (M-16V) | NR (Morinaga) | GA 23-36w | 1.6e8 BID until discharge | 33 | 33 | 0* | 0* | 0 | 0 | NR | NR |
| Manzoni 200686 | Italy | L rhamnosus (GG) | Dicoflor 60 (Dicofarm) | BW < 1500g | 6e9/d until 6w | 39 | 41 | 1 | 3 | 19 | 22 | 5 | 6 |
| Fujii 200647 | Japan | B breve (M-16V) | NR (Morinaga) | GA 31w (SD 3w) | 1e9 BID until discharge | 11 | 8 | 0 | 0 | 1 | 1 | NR | NR |
| Lin 200587 | Taiwan | L acidophilus + B infantis (NR) | Infloran (Swiss Serum and Vaccine Institute) | BW < 1500g | 1e9 BID until discharge | 180 | 187 | 2 | 10 | 22 | 36 | 7 | 20 |
| Bin-Nun 200588 | Israel | B infantis + S thermophilus + B lactis (NR) | ABC Dophilus (Solgar) | BW < 1500g | 1e9/d until 36w | 72 | 72 | 1 | 10 | NR | NR | 3 | 8 |
| Costalos 200389 | Greece | Saccharomyces boulardii (NR) | NR | GA 28-32w | 1e9/d × 30d | 51 | 36 | 5 | 6 | 3 | 3 | NR | NR |
| Dani 200290 | Italy | L rhamnosus (GG) | Dicoflor (Dicofarm) | BW < 1500g + GA < 33w | 6e9/d until discharge | 295 | 290 | 4 | 8 | 14 | 12 | 0 | 2 |
| Reuman 198691 | USA | L acidophilus (NR) | NR | BW < 2000g | 1e8 BID until discharge | 15 | 15 | NR | NR | NR | NR | 1 | 3 |
| Total | 4668 | 4298 | 161 | 263 | 548 | 601 | 244 | 310 | |||||
| % of reported | 100 | 100 | 3.59 | 6.26 | 13.18 | 15.48 | 5.75 | 7.62 | |||||
NR: not reported, BW: birth weight, GA: gestational age, g: grams, w: weeks, d: days
1.6e8: 1.6×108 organisms
number of NEC cases obtained by personal communication with the author
Table 2.
Summary of probiotic cohort studies in premature infants
| Author Year | Country | Probiotic Species (strain) | Brand (Company) | Population | Dose × duration | Number enrolled | NEC cases Stage 2, 3 | Culture + sepsis cases | Deaths | ||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Prob | Cont | Prob | Cont | Prob | Cont | Prob | Cont | ||||||
| Dang 201592 | USA | L rhamnosus (GG) + B infantis (NR) | Culturelle (Amerifit) + Align (Proctor and Gamble) | BW < 1250g and/or GA < 28w | 1e9/d until 34w | 128 | 135 | 2 | 8 | NR | NR | 19 | 21 |
| Repa 201593 | Austria | L acidophilus + B infantis(NR) | Infloran (Laboratoriofarmaceutico) | BW < 1500g | 2e9 BID (NR) | 230 | 233 | 16 | 24 | 60 | 78 | 16 | 30 |
| Hartel 201494 | Germany | L acidophilus + B infantis(NR) | Infloran (Berna) | BW < 1500g + GA < 32w | 1e9/d × 14d | 3789 | 1562 | 116* | 76* | 428 | 195 | 292 | 160 |
| Janvier 201495 | Canada | B bifidum + B breve + B infantis + B longum + L rhamnosus (NR + GG) | FloraBA BY (Renew Life) | GA < 32w | 2e9/d until 34w | 294 | 317 | 16 | 31 | 54 | 57 | 20 | 31 |
| Bonsante 201396 | France | L rhamnosus (LCR35) | Lcr Restituo (Probionov) | GA 24-31w | 2e8 BID until 36w | 347 | 783 | 4 | 41 | 37 | 130 | 8 | 38 |
| Li 201397 | USA | B bifidum + B infantis + S thermophilus | NR | BW < 1500g | NR | 291 | 289 | 7 | 8 | NR | NR | 4 | 3 |
| Hunter 201298 | USA | L reuteri (DSM 17938) | BioGaia (BioGaia) | BW < 1000g | 5.5e7/d until 40w | 79 | 232 | 2 | 35 | 19 | 72 | NR | NR |
| Luoto 201099 | Finland | L rhamnosus (GG) | NR | BW < 1500g | 6e9/d until discharge | 418 | 1900 | 19 | 61 | NR | NR | NR | NR |
| Yamashiro 2010100 | Japan | B breve (M-16 V) | NR | BW < 1500g | 1e9/d (NR) | 338 | 226 | 0 | 6 | 70 | 65 | 39 | 38 |
| Hoyos 1999101 | Colombia | L acidophilus + B infantis | Infloran | All NICU admit | 5e8/d (NR) | 1237 | 1282 | 34 | 85 | 69 | 70 | 137 | 140 |
| Total | 7151 | 6959 | 100 | 299 | 737 | 667 | 535 | 461 | |||||
| % of reported | 100 | 100 | 1.40 | 4.30 | 11.67 | 14.37 | 8.04 | 9.55 | |||||
NR: not reported, BW: birth weight, GA: gestational age, g: grams, w: weeks, d: days
2e9: 2×109 organisms, Prob: probiotic, Cont: control
Only surgical NEC reported
Probiotics are not without risk, particularly in vulnerable populations such as premature infants. Oversight of production of probiotic products varies from country to country. In the U.S. most commercial probiotics are marketed as dietary supplements with no claims of prevention, treatment, or mitigation of disease. Several studies have demonstrated that most commercial products have limited reliability in terms of purity, composition and numbers of live organisms102, 103. Observations of cross-contamination among infants within a NICU suggest that results of RCTs may be blunted by colonization of the probiotic in the placebo infants104, 105. Even more concerning are rare reports of contamination of commercial probiotics with pathogenic microbes; a recent such case resulted in the death of a premature infant106. Sepsis cases resulting from translocation of ingested probiotics into the systemic circulation are rare but have been reported for many probiotic species107-109.
Prebiotics
Prebiotics are non-digestible dietary products that selectively stimulate the growth or activity of beneficial commensal bacteria110, 111. The most commonly administered prebiotics include lactulose, inulin, polydextrose, short-chain (sc) and long-chain (lc) fructo-oligosaccharides and galacto-oligosaccharides, and combinations of the above. The potential complexity of the prebiotic approach to altering the gut microbiota is exemplified by the observations that different isomers of GOS are preferentially consumed by different species of Bifidobacterium112 and that some gut pathogens (e.g. E coli EHEC and C. perfringens) are able to consume some isomers of GOS113. Studies in premature infants demonstrate that prebiotics increase fecal Bifidobacteria114-116, decrease fecal pH115, 117, 118, reduce stool viscosity118, improve gastric motility117, 119, decrease feeding intolerance117, 119, alter production of protective short chain fatty acids120, enhance immune response121, and increase secretory IgA119, 122, 123.
RCTs of prebiotics in premature infants that reported NEC, sepsis or death are summarized in Table 361, 124-127. Most studies randomized infants and initiated therapy with the first feed or before the third day of life, and duration of therapy was typically until hospital discharge. A meta-analysis including 7 trials in premature infants found that supplementation with prebiotics increased fecal Bifidobacteria and Lactobacilli, but did not improve the outcomes of NEC, sepsis or time to full enteral feeding114. Limited follow-up studies of premature infants treated with prebiotic supplements show no significant decrease in allergic or infectious diseases or vaccine response at 12 months of age128, 129. Two possible explanations for the limited efficacy of prebiotics in premature infants include 1) lack of specificity of commercial prebiotics (i.e. both commensal and potentially pathogenic bacteria are able to use some commercial prebiotics as a food source113) and 2) provision of a targeted prebiotic (food source for a limited number of species) without an inoculation of the associated probiotic commensals may be ineffective in cases of severe dysbiosis as seen in premature infants.
Table 3.
Summary of prebiotic RCTs in premature infants
| Author Year | Country | Prebiotic (composition) | Brand (Company) | Population | Dose × duration | Number enrolled | NEC cases Stage 2, 3 | Culture + sepsis cases | Deaths | ||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Preb | Cont | Preb | Cont | Preb | Cont | Preb | Cont | ||||||
| Dilli 201561 | Turkey | Inulin | Maflor (Mamsel) | BW < 1500g + GA < 32w | 900mg/d until discharge | 100 | 100* | 12 | 18* | 10 | 13* | 2 | 12* |
| Armanian 2014124 | Iran | scGOS:lcF OS(9:1) | NR (Nutricia MMP) | BW < 1500g + GA < 34w | varied with feeding volume | 25 | 50 | 1 | 11 | 4 | 17 | 1 | 1 |
| Riskin 2010125 | Israel | 1% lactulose | Laevo lac, (Fresenius Kabi) | GA 23-34w | 1g/100 ml each feed until discharge | 15 | 13 | 1 | 2 | 2 | 4 | 0 | 1 |
| Modi 2010126 | UK | scGOS:lcF OS (9:1) | NR (Danone) | GA < 33w | 0.8g/10 ml(of formula feedings only) | 73 | 81 | 2 | 1 | 9 | 10 | 2 | 1 |
| Westerbeek 2010127 | Netherlands | 80% scGOS/lcF OS + 20% AOS | NR | BW < 1500g and/or GA < 32w | 1.5g/kg/d × 28d | 55 | 58 | 10 | 6 | 32 | 48 | 2 | 3 |
| total | 268 | 302 | 26 | 38 | 57 | 92 | 7 | 18 | |||||
| % of reported | 100 | 100 | 9.70 | 12.58 | 21.27 | 30.46 | 2.61 | 5.96 | |||||
NR: not reported, BW: birth weight, GA: gestational age, g: grams, w: weeks, d: days
scGOS: short-chain galacto-oligosaccharides, lcFOS: long-chain fructo-oligosaccharides, AOS: acidic oligosaccharides
Same control group as reported in Table 1
Synbiotics
A synbiotic is a product that contains both a probiotic microbe and a prebiotic substrate. This combination is particularly compelling as competition for food often determines the composition of the microbiota in a given anatomic niche. The challenge in administration of an effective synbiotic may be in the careful selection of both the prebiotic and the probiotic, with the ideal combination likely including a prebiotic that is consumable by specific commensal gut microbes and not by pathogens or pathobionts and a probiotic with desirable mechanisms of protection. RCTs of synbiotics in premature infants are summarized in Table 461, 130, 131. Future studies of highly specific synbiotic combinations are needed. Human milk is discussed separately, but may represent the quintessential synbiotic given the presence of both prebiotic human milk oligosaccharides and live bacteria.
Table 4.
Summary of synbiotic RCTs in premature infants
| Author Year | Country | Synbiotic | Brand (Company) | Population | Dose × duration | Number enrolled | NEC cases Stage 2, 3 | Culture + sepsis cases | Deaths | ||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Synb | Cont | Synb | Cont | Synb | Cont | Synb | Cont | ||||||
| Dilli 201561 | Turk | Inulin + B lactis | Maflor (Mamsel) | BW < 1500g + GA < 32w | inulin 900 mg + 5e9/d × 8w | 100 | 100* | 4 | 18* | 8 | 13* | 3 | 12* |
| Nandhini 2015130 | India | Inulin + L acidophilus + LGG + L casei + L plantarum + L bulgaricus + B infantis + B breve + B longum | Prepro HS (Fourrts) | BW > 1000g + GA 28-34w | 100 mg/d + 7×109 4×109 3×109 3×109 3×109 3×109 3×109 4×109 BID × 7d | 108 | 110 | 0 | 3 | 4 | 4 | 10 | 9 |
| Underwood 2009131 | USA | Inulin + LGG | ProBio Plus (UAS) | BW 750-2000g + GA < 35w | 5e8 BID × 28d or until discharge | 30 | 29 | 1 | 1 | 6 | 5 | 0 | 0 |
| Inulin + L acidophilus + B longum + B bifidum + B infantis | 31 | 1 | 2 | 0 | |||||||||
| total | 269 | 239 | 6 | 22 | 20 | 22 | 13 | 21 | |||||
| % of reported | 100 | 100 | 2.23 | 9.20 | 7.43 | 9.20 | 4.83 | 8.79 | |||||
LGG: L rhamnosus (GG), NR: not reported, BW: birth weight, GA: gestational age, g: grams, w: weeks, d: days
5e9: 5×109 organisms
Same control group as reported in Table 1
Lactoferrin
Lactoferrin is a complex molecule found in abundance in human milk with prebiotic132, antimicrobial133, and anti-inflammatory properties134. In addition lactoferrin may influence the intestinal microbiota by sequestering iron (the competition for iron in the intestinal lumen is fierce as evidenced by the complexity of bacterial products that facilitate iron recruitment135). Both bovine lactoferrin and recombinant human lactoferrin have been studied in RCTs in premature infants with and without a probiotic with mixed results (Table 5136-140). A recent meta-analysis reported that oral lactoferrin supplementation decreased late onset sepsis (number needed to treat for an additional beneficial (NNTB) 11), NEC (NNTB 20) and all-cause mortality (NNTB 20). Supplementation with both lactoferrin and a probiotic decreased late onset sepsis (NNTB 8) and NEC (NNTB 20) but not all-cause mortality. Oral lactoferrin with or without probiotics decreased fungal sepsis but did not decrease chronic lung disease or length of hospital stay141. Most reports supported administration of lactoferrin as safe in preterm infants136, 138. Some researchers have excluded infants with a family history of cow's milk allergy from trials of bovine lactoferrin142.
Table 5.
Summary of lactoferrin RCTs in premature infants
| Author Year | Country | Lactoferrin | Brand (Company) | Population | Dose × duration | Number enrolled | NEC cases Stage 2, 3 | Culture + sepsis cases | Deaths | ||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Lact | Cont | Lact | Cont | Lact | Cont | Lact | Cont | ||||||
| Kaur 2015136 | India | bLF | apolactoferrin (NR) | BW < 2000g | varied with BW × 28d | 63 | 67 | NR | NR | 2 | 9 | 0 | 5 |
| Ochoa 2015137 | Peru | bLF | NR (Tatua) | BW < 2500g | 200mg/kg/d devided 3 doses × 28d | 95 | 95 | NR | NR | 4 | 4 | 7 | 3 |
| Akin 2014138 | Turk | bLF | LF100 (Dicofarm) | BW < 1500g + GA < 32w | 200mg/d until discharge | 22 | 25 | 0 | 5 | 4 | 8 | 0 | 1 |
| Manzoni 2014139 | Italy | bLF | LF100 (Dicofarm) | BW < 1500g | 100mg/d × 30d BW > 1000g × 45d BW < 1000g | 247 | 258 | 5 | 14 | NR | NR | 5 | 18 |
| bLF + LGG | Dicoflor 60 (Dicofarm) | 100mg/d + 6e9/d | 238 | 0 | NR | 9 | |||||||
| Sherman 2013*140 | USA | talactoferrin | NR | 750-1500g | 150 mg/kg BID | 60 | 60 | NR | NR | 4 | 4 | 1 | 1 |
| total | 725 | 505 | 5 | 19 | 14 | 25 | 22 | 28 | |||||
| % of reported | 100 | 100 | 0.99 | 6.71 | 5.83 | 10.12 | 3.03 | 5.54 | |||||
NR: not reported, BW: birth weight, GA: gestational age, g: grams, w: weeks, d: days
bLF: bovine lactoferrin , LGG: L rhamnosus (GG)
Abstract only
Human milk
Human milk has been described as a tissue (similar to plasma) rather than simply a food source given its incredible complexity. Human milk contains secretory immunoglobulin A, lactoferrin, lysozyme, bile salt-stimulating lipase, growth factors, and human milk oligosaccharides (HMOs), all of which provide protective benefits that could potentially contribute to a reduction of NEC. The decrease in NEC with provision of human milk seems to be dose related35. As we have already addressed lactoferrin and data on specific activity of most other human milk components are limited, we will focus on HMOs and human milk bacteria.
HMOs are abundant complex sugar molecules that are not digestable by the human intestinal tract due to the lack of glycosidases necessary to cleave the specific linkages that characterize these molecules. The obvious question is why a mother expends tremendous energy at great cost to herself, even in times of famine, to produce molecules that are not a food source for her infant. The partial answer to this compelling question is that HMOs are a potential food source for intestinal microbes (ie a prebiotic)143. Testing of a wide variety of gut microbes in culture media with HMOs as the only carbon source has revealed that HMOs are highly specific: only a relatively few species of bifidobacteria and bacteroides are able to consume HMOs144-146. Evaluation of bacterial genomes has confirmed that only these few species encode the complex array of glycosidases necessary to transport and digest HMOs147, 148. In other words, HMOs and the few bacteria that are able to consume them either represent a marvelously complex co-evolution of human lactation and a select group of commensal bacteria or incredibly clever design. The complexity of HMO production (with variability from woman to woman and within a given woman over time in the numbers and types of HMOs) allows a mother to shape the microbiota of her offspring149.
Recent studies suggest that some HMO structures are more readily consumed by gut microbes than others150, 151 and that some HMOs are absorbed from the gut into the bloodstream and can be detected in plasma152 with a subset of these structures filtered by the kidneys and detectable in the urine153. In addition, mothers who deliver prematurely have a higher degree of variability in production of fucosylated HMOs than mothers who deliver at term154. This variation among the more than 100 HMOs characterized to date suggests that some HMOs may be more important in shaping the gut microbiota than others. As an example, about 20% of the North American population is homozygous for a common deletion in the FUT2 gene. These individuals are unable to produce a fucosyl transferase that is essential to creation of α1-2 fucosyl linkages in secreted glycans and have been historically referred to as non-secretors. Non-secretor individuals are at higher risk for some inflammatory diseases of the intestinal tract (e.g. Crohn's disease and celiac disease) and at lower risk for some intestinal infectious diseases (e.g. norovirus and rotavirus)155-158. Non-secretor mothers are unable to create specific fucosylated HMOs (e.g. 2’fucosyllactose) which appears to influence the intestinal microbiota of their infants159.
In a rat model, a specific HMO, disialyllacto-N-tetraose (DSLNT) appears to be protective against NEC160, 161. One clinical study indicated that low concentrations of DSLNT in 4-day mother's milk were associated with increased risk of NEC in VLBW premature infants with HIV-infected mothers (p < 0.05)162. These observations suggest that the protective effect of HMOs against NEC may be highly structure-specific.
The questions of whether human milk contains live bacteria and the origin of these bacteria may have particular relevance to the intestinal microbiota of the premature infant. Historically milk was thought to be sterile until contaminated by bacteria from the mother's skin and the baby's oral cavity. However recent studies of the milk microbiota suggest that some of the microbes present in human milk originate in the mother's gut with transfer likely occurring through the fecal-skin route or through the maternal lymphatic system with gut microbes being shuttled to the breast by dendritic cells or macrophages163. Much is yet to be discovered in this area, however studies of manipulation of the mother's intestinal microbiota to improve the health of her infant are promising, particularly in the prevention of allergies and atopic disease164, necrotizing enterocolitis165 and preterm labor166.
Conclusion
The premature infant is particularly vulnerable to NEC and sepsis likely due to the combination of immature immune responses and dysbiosis. Manipulating the composition of the intestinal microbiota and expression of gut microbial genes is a promising strategy which impacts both of these factors. Among the interventions reviewed, human milk, probiotics, and lactoferrin are currently the most promising. Second generation probiotics, selected based on specific mechanisms of action and/or bacterial genomic sequence and produced at high standards of purity and viability are high priorities. Given that none of the current approaches completely eliminates NEC, further clinical trials and cohort studies of novel probiotics or probiotic combinations for mother and/or baby, combinations of lactoferrin and novel probiotics, and individualized supplementation of human milk with deficient components (e.g. specific HMO or sIgA molecules) in premature infants are indicated.
Acknowledgements
MAU has received funding support from the Eunice Kennedy Shriver National Institute of Child Health & Human Development of the National Institutes of Health, Bethesda Maryland (grant number R01HD059127) and the National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda Maryland (grant number UL1 TR000002). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
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.
Conflicts of interest: none
References
- 1.Neu J, Walker WA. Necrotizing enterocolitis. N Engl J Med. 2011;364:255–64. doi: 10.1056/NEJMra1005408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Dominguez KM, Moss RL. Necrotizing enterocolitis. Clin Perinatol. 2012;39:387–401. doi: 10.1016/j.clp.2012.04.011. [DOI] [PubMed] [Google Scholar]
- 3.McElroy SJ, Underwood MA, Sherman MP. Paneth cells and necrotizing enterocolitis: a novel hypothesis for disease pathogenesis. Neonatology. 2013;103:10–20. doi: 10.1159/000342340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Bell MJ, Ternberg JL, Feigin RD, et al. Neonatal necrotizing enterocolitis. Therapeutic decisions based upon clinical staging. Ann Surg. 1978;187:1–7. doi: 10.1097/00000658-197801000-00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Walsh MC, Kliegman RM. Necrotizing enterocolitis: treatment based on staging criteria. Pediatr Clin North Am. 1986;33:179–201. doi: 10.1016/S0031-3955(16)34975-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Johnson TJ, Patel AL, Bigger HR, Engstrom JL, Meier PP. Cost savings of human milk as a strategy to reduce the incidence of necrotizing enterocolitis in very low birth weight infants. Neonatology. 2015;107:271–6. doi: 10.1159/000370058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Shah TA, Meinzen-Derr J, Gratton T, et al. Hospital and neurodevelopmental outcomes of extremely low-birth-weight infants with necrotizing enterocolitis and spontaneous intestinal perforation. J Perinatol. 2012;32:552–8. doi: 10.1038/jp.2011.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Yee WH, Soraisham AS, Shah VS, et al. Incidence and timing of presentation of necrotizing enterocolitis in preterm infants. Pediatrics. 2012;129:e298–304. doi: 10.1542/peds.2011-2022. [DOI] [PubMed] [Google Scholar]
- 9.Salzman NH, Bevins CL. Dysbiosis--a consequence of Paneth cell dysfunction. Semin Immunol. 2013;25:334–41. doi: 10.1016/j.smim.2013.09.006. [DOI] [PubMed] [Google Scholar]
- 10.Clevers HC, Bevins CL. Paneth cells: maestros of the small intestinal crypts. Annu Rev Physiol. 2013;75:289–311. doi: 10.1146/annurev-physiol-030212-183744. [DOI] [PubMed] [Google Scholar]
- 11.Morrow AL, Lagomarcino AJ, Schibler KR, et al. Early microbial and metabolomic signatures predict later onset of necrotizing enterocolitis in preterm infants. Microbiome. 2013;1:13. doi: 10.1186/2049-2618-1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mai V, Young CM, Ukhanova M, et al. Fecal microbiota in premature infants prior to necrotizing enterocolitis. PLoS One. 2011;6:e20647. doi: 10.1371/journal.pone.0020647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Berrington JE, Stewart CJ, Cummings SP, Embleton ND. The neonatal bowel microbiome in health and infection. Curr Opin Infect Dis. 2014;27:236–43. doi: 10.1097/QCO.0000000000000061. [DOI] [PubMed] [Google Scholar]
- 14.Cassir N, Benamar S, Khalil JB, et al. Clostridium butyricum Strains and Dysbiosis Linked to Necrotizing Enterocolitis in Preterm Neonates. Clin Infect Dis. 2015;61:1107–15. doi: 10.1093/cid/civ468. [DOI] [PubMed] [Google Scholar]
- 15.Winter SE, Baumler AJ. Dysbiosis in the inflamed intestine: chance favors the prepared microbe. Gut Microbes. 2014;5:71–3. doi: 10.4161/gmic.27129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ardissone AN, de la Cruz DM, Davis-Richardson AG, et al. Meconium microbiome analysis identifies bacteria correlated with premature birth. PLoS One. 2014;9:e90784. doi: 10.1371/journal.pone.0090784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.DiGiulio DB, Gervasi M, Romero R, et al. Microbial invasion of the amniotic cavity in preeclampsia as assessed by cultivation and sequence-based methods. J Perinat Med. 2010;38:503–13. doi: 10.1515/JPM.2010.078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.DiGiulio DB, Gervasi MT, Romero R, et al. Microbial invasion of the amniotic cavity in pregnancies with small-for-gestational-age fetuses. J Perinat Med. 2010;38:495–502. doi: 10.1515/JPM.2010.076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Dominguez-Bello MG, Costello EK, Contreras M, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci U S A. 2010;107:11971–5. doi: 10.1073/pnas.1002601107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Arboleya S, Ang L, Margolles A, et al. Deep 16S rRNA metagenomics and quantitative PCR analyses of the premature infant fecal microbiota. Anaerobe. 2012;18:378–80. doi: 10.1016/j.anaerobe.2012.04.013. [DOI] [PubMed] [Google Scholar]
- 21.Arboleya S, Binetti A, Salazar N, et al. Establishment and development of intestinal microbiota in preterm neonates. FEMS Microbiol Ecol. 2012;79:763–72. doi: 10.1111/j.1574-6941.2011.01261.x. [DOI] [PubMed] [Google Scholar]
- 22.Gomez-Llorente C, Plaza-Diaz J, Aguilera M, et al. Three main factors define changes in fecal microbiota associated with feeding modality in infants. J Pediatr Gastroenterol Nutr. 2013;57:461–6. doi: 10.1097/MPG.0b013e31829d519a. [DOI] [PubMed] [Google Scholar]
- 23.Berrington JE, Stewart CJ, Embleton ND, Cummings SP. Gut microbiota in preterm infants: assessment and relevance to health and disease. Arch Dis Child Fetal Neonatal Ed. 2013;98:F286–90. doi: 10.1136/archdischild-2012-302134. [DOI] [PubMed] [Google Scholar]
- 24.Jakobsson HE, Abrahamsson TR, Jenmalm MC, et al. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut. 2014;63:559–66. doi: 10.1136/gutjnl-2012-303249. [DOI] [PubMed] [Google Scholar]
- 25.Greenwood C, Morrow AL, Lagomarcino AJ, et al. Early empiric antibiotic use in preterm infants is associated with lower bacterial diversity and higher relative abundance of Enterobacter. J Pediatr. 2014;165:23–9. doi: 10.1016/j.jpeds.2014.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.La Rosa PS, Warner BB, Zhou Y, et al. Patterned progression of bacterial populations in the premature infant gut. Proc Natl Acad Sci U S A. 2014;111:12522–7. doi: 10.1073/pnas.1409497111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gupta RW, Tran L, Norori J, et al. Histamine-2 receptor blockers alter the fecal microbiota in premature infants. J Pediatr Gastroenterol Nutr. 2013;56:397–400. doi: 10.1097/MPG.0b013e318282a8c2. [DOI] [PubMed] [Google Scholar]
- 28.Guillet R, Stoll BJ, Cotten CM, et al. Association of H2-blocker therapy and higher incidence of necrotizing enterocolitis in very low birth weight infants. Pediatrics. 2006;117:e137–42. doi: 10.1542/peds.2005-1543. [DOI] [PubMed] [Google Scholar]
- 29.Weintraub AS, Ferrara L, Deluca L, et al. Antenatal antibiotic exposure in preterm infants with necrotizing enterocolitis. J Perinatol. 2012;32:705–9. doi: 10.1038/jp.2011.180. [DOI] [PubMed] [Google Scholar]
- 30.Kuppala VS, Meinzen-Derr J, Morrow AL, Schibler KR. Prolonged initial empirical antibiotic treatment is associated with adverse outcomes in premature infants. J Pediatr. 2011;159:720–5. doi: 10.1016/j.jpeds.2011.05.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Brooks B, Firek BA, Miller CS, et al. Microbes in the neonatal intensive care unit resemble those found in the gut of premature infants. Microbiome. 2014;2:1. doi: 10.1186/2049-2618-2-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Sohn K, Kalanetra K, Mills DA, et al. Buccal administration of human colostrum: impact on the oral microbiota. J Perinatol. 2015 doi: 10.1038/jp.2015.157. In press. [DOI] [PubMed] [Google Scholar]
- 33.Wacklin P, Tuimala J, Nikkila J, et al. Faecal microbiota composition in adults is associated with the FUT2 gene determining the secretor status. PLoS One. 2014;9:e94863. doi: 10.1371/journal.pone.0094863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Morrow AL, Meinzen-Derr J, Huang P, et al. Fucosyltransferase 2 non-secretor and low secretor status predicts severe outcomes in premature infants. J Pediatr. 2011;158:745–51. doi: 10.1016/j.jpeds.2010.10.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Meinzen-Derr J, Poindexter B, Wrage L, et al. Role of human milk in extremely low birth weight infants' risk of necrotizing enterocolitis or death. J Perinatol. 2009;29:57–62. doi: 10.1038/jp.2008.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Underwood MA. Intestinal dysbiosis: novel mechanisms by which gut microbes trigger and prevent disease. Prev Med. 2014;65:133–7. doi: 10.1016/j.ypmed.2014.05.010. [DOI] [PubMed] [Google Scholar]
- 37.AlFaleh K, Anabrees J. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Evid Based Child Health. 2014;9:584–671. doi: 10.1002/ebch.1976. [DOI] [PubMed] [Google Scholar]
- 38.Thomas DW, Greer FR. Probiotics and prebiotics in pediatrics. Pediatrics. 2010;126:1217–31. doi: 10.1542/peds.2010-2548. [DOI] [PubMed] [Google Scholar]
- 39.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. Pediatr Res. 2014;76:326–33. doi: 10.1038/pr.2014.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Underwood MA, Kananurak A, Coursodon CF, et al. Bifidobacterium bifidum in a rat model of necrotizing enterocolitis: antimicrobial peptide and protein responses. Pediatr Res. 2012;71:546–51. doi: 10.1038/pr.2012.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Satoh T, Izumi H, Iwabuchi N, et al. Bifidobacterium breve prevents necrotising enterocolitis by suppressing inflammatory responses in a preterm rat model. Benef Microbes. 2015:1–8. doi: 10.3920/BM2015.0035. [DOI] [PubMed] [Google Scholar]
- 42.Ganguli K, Meng D, Rautava S, et al. Probiotics prevent necrotizing enterocolitis by modulating enterocyte genes that regulate innate immune-mediated inflammation. Am J Physiol Gastrointest Liver Physiol. 2013;304:G132–41. doi: 10.1152/ajpgi.00142.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Underwood MA, German JB, Lebrilla CB, Mills DA. Bifidobacterium longum subspecies infantis: champion colonizer of the infant gut. Pediatr Res. 2015;77:229–35. doi: 10.1038/pr.2014.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Khailova L, Dvorak K, Arganbright KM, et al. Bifidobacterium bifidum improves intestinal integrity in a rat model of necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol. 2009;297:G940–9. doi: 10.1152/ajpgi.00141.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Khailova L, Mount Patrick SK, Arganbright KM, et al. Bifidobacterium bifidum reduces apoptosis in the intestinal epithelium in necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol. 2010;299:G1118–27. doi: 10.1152/ajpgi.00131.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.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;19:9–16. doi: 10.1016/j.anaerobe.2012.11.001. [DOI] [PubMed] [Google Scholar]
- 47.Fujii T, Ohtsuka Y, Lee T, et al. Bifidobacterium breve enhances transforming growth factor beta1 signaling by regulating Smad7 expression in preterm infants. J Pediatr Gastroenterol Nutr. 2006;43:83–8. doi: 10.1097/01.mpg.0000228100.04702.f8. [DOI] [PubMed] [Google Scholar]
- 48.Goncalves FL, Soares LM, Figueira RL, et al. Evaluation of the expression of I-FABP and L-FABP in a necrotizing enterocolitis model after the use of Lactobacillus acidophilus. J Pediatr Surg. 2015;50:543–9. doi: 10.1016/j.jpedsurg.2014.07.007. [DOI] [PubMed] [Google Scholar]
- 49.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. Am J Physiol Gastrointest Liver Physiol. 2014;306:G1021–32. doi: 10.1152/ajpgi.00452.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.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. Am J Physiol Gastrointest Liver Physiol. 2012;302:G608–17. doi: 10.1152/ajpgi.00266.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Borthakur A, Bhattacharyya S, Kumar A, et al. Lactobacillus acidophilus alleviates platelet-activating factor-induced inflammatory responses in human intestinal epithelial cells. PLoS One. 2013;8:e75664. doi: 10.1371/journal.pone.0075664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Biersmith B, Wang ZH, Geisbrecht ER. Fine-Tuning of the Actin Cytoskeleton and Cell Adhesion During Drosophila Development by the Unconventional Guanine Nucleotide Exchange Factors Myoblast City and Sponge. Genetics. 2015;200:551–67. doi: 10.1534/genetics.115.177063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.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:e56547. doi: 10.1371/journal.pone.0056547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kunze WA, Mao YK, Wang B, et al. Lactobacillus reuteri enhances excitability of colonic AH neurons by inhibiting calcium-dependent potassium channel opening. J Cell Mol Med. 2009;13:2261–70. doi: 10.1111/j.1582-4934.2009.00686.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.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:e51955. doi: 10.1371/journal.pone.0051955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.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. Pediatr Res. 2008;64:511–6. doi: 10.1203/PDR.0b013e3181827c0f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Patel RM, Myers LS, Kurundkar AR, et al. Probiotic bacteria induce maturation of intestinal claudin 3 expression and barrier function. Am J Pathol. 2012;180:626–35. doi: 10.1016/j.ajpath.2011.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.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;3:25–8. doi: 10.4161/gmic.19245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Olsen R, Greisen G, Schroder M, Brok J. Prophylactic Probiotics for Preterm Infants: A Systematic Review and Meta-Analysis of Observational Studies. Neonatology. 2015;109:105–12. doi: 10.1159/000441274. [DOI] [PubMed] [Google Scholar]
- 60.Costeloe K, Hardy P, Juszczak E, Wilks M, Millar MR. Bifidobacterium breve BBG-001 in very preterm infants: a randomised controlled phase 3 trial. Lancet. 2015 doi: 10.1016/S0140-6736(15)01027-2. [DOI] [PubMed] [Google Scholar]
- 61.Dilli D, Aydin B, Fettah ND, et al. The propre-save study: effects of probiotics and prebiotics alone or combined on necrotizing enterocolitis in very low birth weight infants. J Pediatr. 2015;166:545–51. e1. doi: 10.1016/j.jpeds.2014.12.004. [DOI] [PubMed] [Google Scholar]
- 62.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 Med Assoc Thai. 2014;97(Suppl 6):S20–5. [PubMed] [Google Scholar]
- 63.Tewari VV, Dubey SK, Gupta G. Bacillus clausii for Prevention of Late-onset Sepsis in Preterm Infants: A Randomized Controlled Trial. J Trop Pediatr. 2015;61:377–85. doi: 10.1093/tropej/fmv050. [DOI] [PubMed] [Google Scholar]
- 64.Hays S, Jacquot A, Gauthier H, et al. Probiotics and growth in preterm infants: A randomized controlled trial, PREMAPRO study. Clin Nutr. 2015 doi: 10.1016/j.clnu.2015.06.006. doi: 101016/j.clnu.2015.06.006. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 65.Van Niekerk E, Nel DG, Blaauw R, Kirsten GF. Probiotics Reduce Necrotizing Enterocolitis Severity in HIV-exposed Premature Infants. J Trop Pediatr. 2015;61:155–64. doi: 10.1093/tropej/fmv004. [DOI] [PubMed] [Google Scholar]
- 66.Dutta S, Ray P, Narang A. Comparison of stool colonization in premature infants by three dose regimes of a probiotic combination: a randomized controlled trial. Am J Perinatol. 2015;32:733–40. doi: 10.1055/s-0034-1395473. [DOI] [PubMed] [Google Scholar]
- 67.Patole S, Keil AD, Chang A, et al. Effect of Bifidobacterium breve M-16V supplementation on fecal bifidobacteria in preterm neonates--a randomised double blind placebo controlled trial. PLoS One. 2014;9:e89511. doi: 10.1371/journal.pone.0089511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.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. Arch Dis Child Fetal Neonatal Ed. 2014;99:F110–5. doi: 10.1136/archdischild-2013-304745. [DOI] [PubMed] [Google Scholar]
- 69.Totsu S, Yamasaki C, Terahara M, Uchiyama A, Kusuda S. Bifidobacterium and enteral feeding in preterm infants: cluster-randomized trial. Pediatr Int. 2014;56:714–9. doi: 10.1111/ped.12330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Jacobs SE, Tobin JM, Opie GF, et al. Probiotic effects on late-onset sepsis in very preterm infants: a randomized controlled trial. Pediatrics. 2013;132:1055–62. doi: 10.1542/peds.2013-1339. [DOI] [PubMed] [Google Scholar]
- 71.Serce O, Benzer D, Gursoy T, Karatekin G, Ovali F. Efficacy of Saccharomyces boulardii on necrotizing enterocolitis or sepsis in very low birth weight infants: a randomised controlled trial. Early Hum Dev. 2013;89:1033–6. doi: 10.1016/j.earlhumdev.2013.08.013. [DOI] [PubMed] [Google Scholar]
- 72.Demirel G, Erdeve O, Celik IH, Dilmen U. Saccharomyces boulardii for prevention of necrotizing enterocolitis in preterm infants: a randomized, controlled study. Acta Paediatr. 2013;102:e560–5. doi: 10.1111/apa.12416. [DOI] [PubMed] [Google Scholar]
- 73.Rojas MA, Lozano JM, Rojas MX, et al. Prophylactic probiotics to prevent death and nosocomial infection in preterm infants. Pediatrics. 2012;130:e1113–20. doi: 10.1542/peds.2011-3584. [DOI] [PubMed] [Google Scholar]
- 74.Fernandez-Carrocera LA, Solis-Herrera A, Cabanillas-Ayon M, et al. Double-blind, randomised clinical assay to evaluate the efficacy of probiotics in preterm newborns weighing less than 1500 g in the prevention of necrotising enterocolitis. Arch Dis Child Fetal Neonatal Ed. 2013;98:F5–9. doi: 10.1136/archdischild-2011-300435. [DOI] [PubMed] [Google Scholar]
- 75.Al-Hosni M, Duenas M, Hawk M, et al. Probiotics-supplemented feeding in extremely low-birth-weight infants. J Perinatol. 2012;32:253–9. doi: 10.1038/jp.2011.51. [DOI] [PubMed] [Google Scholar]
- 76.Sari FN, Dizdar EA, Oguz S, et al. Oral probiotics: Lactobacillus sporogenes for prevention of necrotizing enterocolitis in very low-birth weight infants: a randomized, controlled trial. Eur J Clin Nutr. 2011;65:434–9. doi: 10.1038/ejcn.2010.278. [DOI] [PubMed] [Google Scholar]
- 77.Braga TD, da Silva GA, de Lira PI, 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 Nutr. 2011;93:81–6. doi: 10.3945/ajcn.2010.29799. [DOI] [PubMed] [Google Scholar]
- 78.Romeo MG, Romeo DM, Trovato L, et al. 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;31:63–9. doi: 10.1038/jp.2010.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Awad H, Mokhtar H, Imam SS, et al. Comparison between killed and living probiotic usage versus placebo for the prevention of necrotizing enterocolitis and sepsis in neonates. Pak J Biol Sci. 2010;13:253–62. doi: 10.3923/pjbs.2010.253.262. [DOI] [PubMed] [Google Scholar]
- 80.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;98:156–63. doi: 10.1159/000280291. [DOI] [PubMed] [Google Scholar]
- 81.Samanta M, Sarkar M, Ghosh P, et al. Prophylactic probiotics for prevention of necrotizing enterocolitis in very low birth weight newborns. J Trop Pediatr. 2009;55:128–31. doi: 10.1093/tropej/fmn091. [DOI] [PubMed] [Google Scholar]
- 82.Rouge C, Piloquet H, Butel MJ, et al. Oral supplementation with probiotics in very-low-birth-weight preterm infants: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr. 2009;89:1828–35. doi: 10.3945/ajcn.2008.26919. [DOI] [PubMed] [Google Scholar]
- 83.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;122:693–700. doi: 10.1542/peds.2007-3007. [DOI] [PubMed] [Google Scholar]
- 84.Stratiki Z, Costalos C, Sevastiadou S, et al. The effect of a bifidobacter supplemented bovine milk on intestinal permeability of preterm infants. Early Hum Dev. 2007;83:575–9. doi: 10.1016/j.earlhumdev.2006.12.002. [DOI] [PubMed] [Google Scholar]
- 85.Wang C, Shoji H, Sato H, et al. Effects of oral administration of bifidobacterium breve on fecal lactic acid and short-chain fatty acids in low birth weight infants. J Pediatr Gastroenterol Nutr. 2007;44:252–7. doi: 10.1097/01.mpg.0000252184.89922.5f. [DOI] [PubMed] [Google Scholar]
- 86.Manzoni P, Mostert M, Leonessa ML, et al. Oral supplementation with Lactobacillus casei subspecies rhamnosus prevents enteric colonization by Candida species in preterm neonates: a randomized study. Clin Infect Dis. 2006;42:1735–42. doi: 10.1086/504324. [DOI] [PubMed] [Google Scholar]
- 87.Lin HC, Su BH, Chen AC, et al. 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. [DOI] [PubMed] [Google Scholar]
- 88.Bin-Nun A, Bromiker R, Wilschanski M, et al. Oral probiotics prevent necrotizing enterocolitis in very low birth weight neonates. J Pediatr. 2005;147:192–6. doi: 10.1016/j.jpeds.2005.03.054. [DOI] [PubMed] [Google Scholar]
- 89.Costalos C, Skouteri V, Gounaris A, et al. Enteral feeding of premature infants with Saccharomyces boulardii. Early Hum Dev. 2003;74:89–96. doi: 10.1016/s0378-3782(03)00090-2. [DOI] [PubMed] [Google Scholar]
- 90.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. Biol Neonate. 2002;82:103–8. doi: 10.1159/000063096. [DOI] [PubMed] [Google Scholar]
- 91.Reuman PD, Duckworth DH, Smith KL, et al. Lack of effect of Lactobacillus on gastrointestinal bacterial colonization in premature infants. Pediatr Infect Dis. 1986;5:663–8. doi: 10.1097/00006454-198611000-00013. [DOI] [PubMed] [Google Scholar]
- 92.Dang S, Shook L, Garlitz K, Hanna M, Desai N. Nutritional outcomes with implementation of probiotics in preterm infants. J Perinatol. 2015;35:447–50. doi: 10.1038/jp.2014.234. [DOI] [PubMed] [Google Scholar]
- 93.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. Pediatr Res. 2015;77:381–8. doi: 10.1038/pr.2014.192. [DOI] [PubMed] [Google Scholar]
- 94.Hartel C, Pagel J, Rupp J, et al. Prophylactic use of Lactobacillus acidophilus/Bifidobacterium infantis probiotics and outcome in very low birth weight infants. J Pediatr. 2014;165:285–9. e1. doi: 10.1016/j.jpeds.2014.04.029. [DOI] [PubMed] [Google Scholar]
- 95.Janvier A, Malo J, Barrington KJ. Cohort study of probiotics in a North American neonatal intensive care unit. J Pediatr. 2014;164:980–5. doi: 10.1016/j.jpeds.2013.11.025. [DOI] [PubMed] [Google Scholar]
- 96.Bonsante F, Iacobelli S, Gouyon JB. Routine probiotic use in very preterm infants: retrospective comparison of two cohorts. Am J Perinatol. 2013;30:41–6. doi: 10.1055/s-0032-1321498. [DOI] [PubMed] [Google Scholar]
- 97.Li D, Rosito G, Slagle T. Probiotics for the prevention of necrotizing enterocolitis in neonates: an 8-year retrospective cohort study. J Clin Pharm Ther. 2013;38:445–9. doi: 10.1111/jcpt.12084. [DOI] [PubMed] [Google Scholar]
- 98.Hunter C, Dimaguila MA, Gal P, et al. Effect of routine probiotic, Lactobacillus reuteri DSM 17938, use on rates of necrotizing enterocolitis in neonates with birthweight < 1000 grams: a sequential analysis. BMC Pediatr. 2012;12:142. doi: 10.1186/1471-2431-12-142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Luoto R, Matomaki J, Isolauri E, Lehtonen L. Incidence of necrotizing enterocolitis in very-low-birth-weight infants related to the use of Lactobacillus GG. Acta Paediatr. 2010;99:1135–8. doi: 10.1111/j.1651-2227.2010.01795.x. [DOI] [PubMed] [Google Scholar]
- 100.Yamashiro Y, Nagata S. Beneficial microbes for premature infants, and children with malignancy undergoing chemotherapy. Benef Microbes. 2010;1:357–65. doi: 10.3920/BM2010.0035. [DOI] [PubMed] [Google Scholar]
- 101.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;3:197–202. doi: 10.1016/s1201-9712(99)90024-3. [DOI] [PubMed] [Google Scholar]
- 102.Lewis ZT, Shani G, Masarweh C, et al. Validating bifidobacterial species and subspecies identity in commercial probiotic products. Pediatr Res. 2015 doi: 10.1038/pr.2015.244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Marcobal A, Underwood MA, Mills DA. Rapid determination of the bacterial composition of commercial probiotic products by terminal restriction fragment length polymorphism analysis. J Pediatr Gastroenterol Nutr. 2008;46:608–11. doi: 10.1097/MPG.0b013e3181660694. [DOI] [PubMed] [Google Scholar]
- 104.Hickey L, Garland SM, Jacobs SE, O'Donnell CP, Tabrizi SN. Cross-colonization of infants with probiotic organisms in a neonatal unit. J Hosp Infect. 2014;88:226–9. doi: 10.1016/j.jhin.2014.09.006. [DOI] [PubMed] [Google Scholar]
- 105.Underwood MA, Kalanetra KM, Bokulich NA, et al. A comparison of two probiotic strains of bifidobacteria in premature infants. J Pediatr. 2013;163:1585–91. e9. doi: 10.1016/j.jpeds.2013.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.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. MMWR Morb Mortal Wkly Rep. 2015;64:155–6. [PMC free article] [PubMed] [Google Scholar]
- 107.Bertelli C, Pillonel T, Torregrossa A, et al. Bifidobacterium longum bacteremia in preterm infants receiving probiotics. Clin Infect Dis. 2015;60:924–7. doi: 10.1093/cid/ciu946. [DOI] [PubMed] [Google Scholar]
- 108.Ohishi A, Takahashi S, Ito Y, et al. Bifidobacterium septicemia associated with postoperative probiotic therapy in a neonate with omphalocele. J Pediatr. 2010;156:679–81. doi: 10.1016/j.jpeds.2009.11.041. [DOI] [PubMed] [Google Scholar]
- 109.Thompson C, McCarter YS, Krause PJ, Herson VC. Lactobacillus acidophilus sepsis in a neonate. J Perinatol. 2001;21:258–60. doi: 10.1038/sj.jp.7200509. [DOI] [PubMed] [Google Scholar]
- 110.Panigrahi P. Probiotics and prebiotics in neonatal necrotizing enterocolitis: New opportunities for translational research. Pathophysiology. 2014;21:35–46. doi: 10.1016/j.pathophys.2013.11.016. [DOI] [PubMed] [Google Scholar]
- 111.Hutkins RW, Krumbeck JA, Bindels LB, et al. Prebiotics: why definitions matter. Curr Opin Biotechnol. 2015;37:1–7. doi: 10.1016/j.copbio.2015.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Peacock KS, Ruhaak LR, Tsui MK, Mills DA, Lebrilla CB. Isomer-specific consumption of galactooligosaccharides by bifidobacterial species. J Agric Food Chem. 2013;61:12612–9. doi: 10.1021/jf403789r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Makelainen H, Saarinen M, Stowell J, Rautonen N, Ouwehand AC. Xylo oligosaccharides and lactitol promote the growth of Bifidobacterium lactis and Lactobacillus species in pure cultures. Benef Microbes. 2010;1:139–48. doi: 10.3920/BM2009.0029. [DOI] [PubMed] [Google Scholar]
- 114.Srinivasjois R, Rao S, Patole S. Prebiotic supplementation in preterm neonates: updated systematic review and meta-analysis of randomised controlled trials. Clin Nutr. 2013;32:958–65. doi: 10.1016/j.clnu.2013.05.009. [DOI] [PubMed] [Google Scholar]
- 115.Westerbeek EA, Slump RA, Lafeber HN, et al. 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 Infect Dis. 2013;32:269–76. doi: 10.1007/s10096-012-1739-y. [DOI] [PubMed] [Google Scholar]
- 116.Barrett E, Deshpandey AK, Ryan CA, et al. The neonatal gut harbours distinct bifidobacterial strains. Arch Dis Child Fetal Neonatal Ed. 2015;100:F405–10. doi: 10.1136/archdischild-2014-306110. [DOI] [PubMed] [Google Scholar]
- 117.Indrio F, Riezzo G, Raimondi F, et al. Prebiotics improve gastric motility and gastric electrical activity in preterm newborns. J Pediatr Gastroenterol Nutr. 2009;49:258–61. doi: 10.1097/MPG.0b013e3181926aec. [DOI] [PubMed] [Google Scholar]
- 118.Westerbeek EA, Hensgens RL, Mihatsch WA, et al. The effect of neutral and acidic oligosaccharides on stool viscosity, stool frequency and stool pH in preterm infants. Acta Paediatr. 2011;100:1426–31. doi: 10.1111/j.1651-2227.2011.02295.x. [DOI] [PubMed] [Google Scholar]
- 119.Roberfroid M, Gibson GR, Hoyles L, et al. Prebiotic effects: metabolic and health benefits. Br J Nutr. 2010;104(Suppl 2):S1–63. doi: 10.1017/S0007114510003363. [DOI] [PubMed] [Google Scholar]
- 120.Fukuda S, Toh H, Hase K, et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature. 2011;469:543–7. doi: 10.1038/nature09646. [DOI] [PubMed] [Google Scholar]
- 121.Seifert S, Watzl B. Inulin and oligofructose: review of experimental data on immune modulation. J Nutr. 2007;137:2563S–7S. doi: 10.1093/jn/137.11.2563S. [DOI] [PubMed] [Google Scholar]
- 122.Vandenplas Y, Zakharova I, Dmitrieva Y. Oligosaccharides in infant formula: more evidence to validate the role of prebiotics. Br J Nutr. 2015;113:1339–44. doi: 10.1017/S0007114515000823. [DOI] [PubMed] [Google Scholar]
- 123.Holscher HD, Faust KL, Czerkies LA, et al. Effects of prebiotic-containing infant formula on gastrointestinal tolerance and fecal microbiota in a randomized controlled trial. JPEN J Parenter Enteral Nutr. 2012;36:95S–105S. doi: 10.1177/0148607111430087. [DOI] [PubMed] [Google Scholar]
- 124.Armanian AM, Sadeghnia A, Hoseinzadeh M, et al. The Effect of Neutral Oligosaccharides on Reducing the Incidence of Necrotizing Enterocolitis in Preterm infants: A Randomized Clinical Trial. Int J Prev Med. 2014;5:1387–95. [PMC free article] [PubMed] [Google Scholar]
- 125.Riskin A, Hochwald O, Bader D, et al. The effects of lactulose supplementation to enteral feedings in premature infants: a pilot study. J Pediatr. 2010;156:209–14. doi: 10.1016/j.jpeds.2009.09.006. [DOI] [PubMed] [Google Scholar]
- 126.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;68:440–5. doi: 10.1203/PDR.0b013e3181f1cd59. [DOI] [PubMed] [Google Scholar]
- 127.Westerbeek EA, van den Berg JP, Lafeber HN, et al. Neutral and acidic oligosaccharides in preterm infants: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr. 2010;91:679–86. doi: 10.3945/ajcn.2009.28625. [DOI] [PubMed] [Google Scholar]
- 128.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;172:317–23. doi: 10.1007/s00431-012-1886-2. [DOI] [PubMed] [Google Scholar]
- 129.van den Berg JP, Westerbeek EA, van der Klis FR, et al. Neutral and acidic oligosaccharides supplementation does not increase the vaccine antibody response in preterm infants in a randomized clinical trial. PLoS One. 2013;8:e70904. doi: 10.1371/journal.pone.0070904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Nandhini LP, Biswal N, Adhisivam B, et al. Synbiotics for decreasing incidence of necrotizing enterocolitis among preterm neonates - a randomized controlled trial. J Matern Fetal Neonatal Med. 2015:1–5. doi: 10.3109/14767058.2015.1019854. [DOI] [PubMed] [Google Scholar]
- 131.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. J Pediatr Gastroenterol Nutr. 2009;48:216–25. doi: 10.1097/MPG.0b013e31818de195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Tian H, Maddox IS, Ferguson LR, Shu Q. Influence of bovine lactoferrin on selected probiotic bacteria and intestinal pathogens. Biometals. 2010;23:593–6. doi: 10.1007/s10534-010-9318-0. [DOI] [PubMed] [Google Scholar]
- 133.Lonnerdal B, Iyer S. Lactoferrin: molecular structure and biological function. Annu Rev Nutr. 1995;15:93–110. doi: 10.1146/annurev.nu.15.070195.000521. [DOI] [PubMed] [Google Scholar]
- 134.Sherman MP, Miller MM, Sherman J, Niklas V. Lactoferrin and necrotizing enterocolitis. Curr Opin Pediatr. 2014;26:146–50. doi: 10.1097/MOP.0000000000000068. [DOI] [PubMed] [Google Scholar]
- 135.Holden VI, Bachman MA. Diverging roles of bacterial siderophores during infection. Metallomics. 2015;7:986–95. doi: 10.1039/c4mt00333k. [DOI] [PubMed] [Google Scholar]
- 136.Kaur G, Gathwala G. Efficacy of Bovine Lactoferrin Supplementation in Preventing Late-onset Sepsis in low Birth Weight Neonates: A Randomized Placebo-Controlled Clinical Trial. J Trop Pediatr. 2015;61:370–6. doi: 10.1093/tropej/fmv044. [DOI] [PubMed] [Google Scholar]
- 137.Ochoa TJ, Zegarra J, Cam L, et al. Randomized controlled trial of lactoferrin for prevention of sepsis in peruvian neonates less than 2500 g. Pediatr Infect Dis J. 2015;34:571–6. doi: 10.1097/INF.0000000000000593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Akin IM, Atasay B, Dogu F, et al. Oral lactoferrin to prevent nosocomial sepsis and necrotizing enterocolitis of premature neonates and effect on T-regulatory cells. Am J Perinatol. 2014;31:1111–20. doi: 10.1055/s-0034-1371704. [DOI] [PubMed] [Google Scholar]
- 139.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 Hum Dev. 2014;90(Suppl 1):S60–5. doi: 10.1016/S0378-3782(14)70020-9. [DOI] [PubMed] [Google Scholar]
- 140.Sherman MP Niklas V, Radmacher P, et al. A clinical trial of talactoferrin to reduce infection in very preterm infants: safety and efficacy. Pediatric Academic Societies; Washington, D.C.: May 4-7, 2013. [Google Scholar]
- 141.Pammi M, Abrams SA. Oral lactoferrin for the prevention of sepsis and necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev. 2015;2:CD007137. doi: 10.1002/14651858.CD007137.pub4. [DOI] [PubMed] [Google Scholar]
- 142.Turin CG, Zea-Vera A, Pezo A, et al. Lactoferrin for prevention of neonatal sepsis. Biometals. 2014;27:1007–16. doi: 10.1007/s10534-014-9754-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Smilowitz JT, Lebrilla CB, Mills DA, German JB, Freeman SL. Breast milk oligosaccharides: structure-function relationships in the neonate. Annu Rev Nutr. 2014;34:143–69. doi: 10.1146/annurev-nutr-071813-105721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Ruiz-Moyano S, Totten SM, Garrido DA, et al. Variation in consumption of human milk oligosaccharides by infant gut-associated strains of Bifidobacterium breve. Appl Environ Microbiol. 2013;79:6040–9. doi: 10.1128/AEM.01843-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Garrido D, Dallas DC, Mills DA. Consumption of human milk glycoconjugates by infant-associated bifidobacteria: mechanisms and implications. Microbiology. 2013;159:649–64. doi: 10.1099/mic.0.064113-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Kim JH, An HJ, Garrido D, et al. Proteomic analysis of Bifidobacterium longum subsp. infantis reveals the metabolic insight on consumption of prebiotics and host glycans. PLoS One. 2013;8:e57535. doi: 10.1371/journal.pone.0057535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Sela DA, Garrido D, Lerno L, et al. Bifidobacterium longum subsp. infantis ATCC 15697 alpha-fucosidases are active on fucosylated human milk oligosaccharides. Appl Environ Microbiol. 2012;78:795–803. doi: 10.1128/AEM.06762-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.LoCascio RG, Desai P, Sela DA, Weimer B, Mills DA. Broad conservation of milk utilization genes in Bifidobacterium longum subsp. infantis as revealed by comparative genomic hybridization. Appl Environ Microbiol. 2010;76:7373–81. doi: 10.1128/AEM.00675-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Underwood MA. Human milk for the premature infant. Pediatr Clin North Am. 2013;60:189–207. doi: 10.1016/j.pcl.2012.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Underwood MA, Gaerlan S, De Leoz ML, et al. Human milk oligosaccharides in premature infants: absorption, excretion, and influence on the intestinal microbiota. Pediatr Res. 2015 doi: 10.1038/pr.2015.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Garrido D, Ruiz-Moyano S, Lemay DG, et al. Comparative transcriptomics reveals key differences in the response to milk oligosaccharides of infant gut-associated bifidobacteria. Sci Rep. 2015;5:13517. doi: 10.1038/srep13517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Ruhaak LR, Stroble C, Underwood MA, Lebrilla CB. Detection of milk oligosaccharides in plasma of infants. Anal Bioanal Chem. 2014;406:5775–84. doi: 10.1007/s00216-014-8025-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.De Leoz ML, Wu S, Strum JS, et al. A quantitative and comprehensive method to analyze human milk oligosaccharide structures in the urine and feces of infants. Anal Bioanal Chem. 2013;405:4089–105. doi: 10.1007/s00216-013-6817-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.De Leoz ML, Gaerlan SC, Strum JS, et al. Lacto-N-tetraose, fucosylation, and secretor status are highly variable in human milk oligosaccharides from women delivering preterm. J Proteome Res. 2012;11:4662–72. doi: 10.1021/pr3004979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Tong M, McHardy I, Ruegger P, et al. Reprograming of gut microbiome energy metabolism by the FUT2 Crohn's disease risk polymorphism. ISME J. 2014;8:2193–206. doi: 10.1038/ismej.2014.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Parmar AS, Alakulppi N, Paavola-Sakki P, et al. Association study of FUT2 (rs601338) with celiac disease and inflammatory bowel disease in the Finnish population. Tissue Antigens. 2012;80:488–93. doi: 10.1111/tan.12016. [DOI] [PubMed] [Google Scholar]
- 157.Payne DC, Currier RL, Staat MA, et al. Epidemiologic Association Between FUT2 Secretor Status and Severe Rotavirus Gastroenteritis in Children in the United States. JAMA Pediatr. 2015;169:1040–5. doi: 10.1001/jamapediatrics.2015.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Currier RL, Payne DC, Staat MA, et al. Innate Susceptibility to Norovirus Infections Influenced by FUT2 Genotype in a United States Pediatric Population. Clin Infect Dis. 2015;60:1631–8. doi: 10.1093/cid/civ165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Lewis ZT, Totten SM, Smilowitz JT, et al. Maternal fucosyltransferase 2 status affects the gut bifidobacterial communities of breastfed infants. Microbiome. 2015;3:13. doi: 10.1186/s40168-015-0071-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Jantscher-Krenn E, Zherebtsov M, Nissan C, et al. The human milk oligosaccharide disialyllacto-N-tetraose prevents necrotising enterocolitis in neonatal rats. Gut. 2012;61:1417–25. doi: 10.1136/gutjnl-2011-301404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Yu H, Lau K, Thon V, et al. Synthetic disialyl hexasaccharides protect neonatal rats from necrotizing enterocolitis. Angew Chem Int Ed Engl. 2014;53:6687–91. doi: 10.1002/anie.201403588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Van Niekerk E, Autran CA, Nel DG, et al. Human milk oligosaccharides differ between HIV-infected and HIV-uninfected mothers and are related to necrotizing enterocolitis incidence in their preterm very-low-birth-weight infants. J Nutr. 2014;144:1227–33. doi: 10.3945/jn.113.187799. [DOI] [PubMed] [Google Scholar]
- 163.Jeurink PV, van Bergenhenegouwen J, Jimenez E, et al. Human milk: a source of more life than we imagine. Benef Microbes. 2013;4:17–30. doi: 10.3920/BM2012.0040. [DOI] [PubMed] [Google Scholar]
- 164.Rautava S, Kainonen E, Salminen S, Isolauri E. Maternal probiotic supplementation during pregnancy and breast-feeding reduces the risk of eczema in the infant. J Allergy Clin Immunol. 2012;130:1355–60. doi: 10.1016/j.jaci.2012.09.003. [DOI] [PubMed] [Google Scholar]
- 165.Benor S, Marom R, Ben Tov A, et al. Probiotic supplementation in mothers of very low birth weight infants. Am J Perinatol. 2014;31:497–504. doi: 10.1055/s-0033-1353490. [DOI] [PubMed] [Google Scholar]
- 166.Jacob JA. Another Frontier in Microbiome Research: Preterm Birth. JAMA. 2015;314:1550–1. doi: 10.1001/jama.2015.11563. [DOI] [PubMed] [Google Scholar]

