Sie sind auf Seite 1von 5

Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Fetal and Neonatal Medicine


journal homepage: www.elsevier.com/locate/siny

Enteral feeding composition and necrotizing enterocolitis


Diomel de la Cruz∗, Catalina Bazacliu
University of Florida, Department of Pediatrics, Division of Neonatology, Gainesville, FL, USA

A R T I C LE I N FO A B S T R A C T

Keywords: Enteral feeding and composition play a chief role in the prevention and treatment of necrotizing enterocolitis
Necrotizing enterocolitis (NEC). In the face of decades of research on this fatal disease, the exact mechanism of disease is still poorly
Human milk understood. There is established evidence that providing mother's own breast milk and standardization of
Donor breast milk feeding regimens leads to a decreased risk for NEC. More recent studies have focused on the provision of donor
Lactoferrin
human milk or an exclusive human milk diet in the endeavor to prevent NEC while still maintaining adequate
Human milk oligosaccharides
nutrition to the premature infant. There is growing literature on the provision of specific human milk compo-
nents and its effect on the incidence of NEC.

1. Introduction nutrition [12]. The benefits of human milk are attributed to the pre-
sence of bioactive substances that act with bactericidal and im-
Necrotizing enterocolitis (NEC) is the most common, life-threa- munomodulating properties. Evidence shows that exclusive human
tening gastrointestinal emergency in the neonatal period affecting ap- milk appears to decrease the risk of late-onset sepsis and feeding in-
proximately 7% of premature very low birth weight (VLBW) infants. tolerance and other complications of prematurity such as retinopathy of
Mortality rates range from 15% to 30%, depending on the need for prematurity [13,14]. Furthermore, provision of human milk to pre-
surgical intervention or disease severity [1,2]. NEC significantly in- mature infants is associated with improved neurodevelopmental out-
creases medical care costs, especially if surgery is required [3]. Total comes, shorter NICU stay, decreased hospital readmissions and lower
hospital costs for NEC are estimated at US$7 million per year [4,5]. The obesity and insulin resistance in adolescence [15–18].
death toll and the long-term sequelae make prevention and manage-
ment of this disease very important. Despite being studied for more 2.1. Exclusive human milk
than six decades, the pathophysiology of NEC is still incompletely
deciphered and poorly understood. The underlying cause is considered A large amount of data supports the concept that exclusive human
multifactorial, an association of intestinal and immune system im- milk intake may reduce the incidence and severity of NEC [19–22].
maturity, intestinal mucosal injury, bacterial dysbiosis, and type of Lucas et al. [23] demonstrated, in a landmark paper in 1990, that
feeds, being widely accepted [6–11]. We know that a vast majority of human breast milk has protective effects against NEC when compared
cases of NEC develop after the initiation of enteral feeding. Despite the with formula. Abrams et al. [24] performed a meta-analysis and re-
persistent challenges in the diagnosis and prevention of NEC, there has ported a lower risk of NEC in those who received an exclusive human
been considerable evidence for the past few decades that two feeding milk diet.
interventions decrease its incidence: provision of breastmilk and stan- One challenge that confronts an exclusive human milk diet is the
dardized feeding regimens. Specific human milk fractions and compo- matter of fortification. Human milk does not contain sufficient amounts
nents seemed promising for prevention of NEC in preclinical studies, of protein, calcium, phosphorous and calories for the ideal growth of
but there is scarce clinical evidence of significant impact on disease the premature infant [12]. Most neonatal intensive care units use bo-
prevention and treatment for any single one. vine-based fortifier that may promote an environment conducive to
NEC. The most recent meta-analysis looking at 14 trials on bovine
2. Human milk and necrotizing enterocolitis fortification of breastmilk reports that human breastmilk fortification
results in slightly improved growth and alkaline phosphatase, without
Human milk is established to be the most favorable form of infant increasing the risk of NEC [25]. Further studies on human milk-based


Corresponding author. University of Florida, Department of Pediatrics, Division of Neonatology, 1600 SW Archer Road, PO Box 100296, Gainesville, FL 32610-
0296, USA.
E-mail address: ddelacruz@ufl.edu (D.d.l. Cruz).

https://doi.org/10.1016/j.siny.2018.08.003

1744-165X/ © 2018 Published by Elsevier Ltd.

Please cite this article as: Cruz, D.d.l., Seminars in Fetal and Neonatal Medicine (2018), https://doi.org/10.1016/j.siny.2018.08.003
D.d.l. Cruz, C. Bazacliu Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

human milk fortifier are still needed. or even a more aggressive daily advancement of enteral feeds
(> 20 mL/kg/day) [39,40].
2.2. Donor human milk
3.2. Early fortification of feeds
Questions remain regarding whether donor human milk has the
same protective effect as mother's own milk. The evidence is less per- It is generally recognized that fortification of human milk is re-
suasive, given that most studies do not randomize infants to purely quired to deliver adequate calories, calcium and phosphorous and
donor breast milk versus formula. More recent meta-analyses have protein to the preterm infant [25,44]. However, early fortification and
concluded that infants fed donor breastmilk had poor growth but less increasing osmolality of feeds has raised questions on its safety [45].
NEC compared to those fed formula [26,27]. Notably, in both meta- Two RCTs have shown that early fortification of enteral feeds did not
analyses, the benefit of preventing NEC was lost when donor human increase the risk of NEC or feeding intolerance while presenting the
milk was used to supplement mother's own milk. Furthermore, most of benefit of improved growth and protein delivery [46,47].
the studies used in the Cochrane review used studies published in the
1980s and excluded infants at the highest risk, namely the smallest 4. Colostrum
infants. Two randomized controlled trials (RCTs) have failed to de-
monstrate a reduction in NEC, whether on donor human milk or for- Produced in the first few days postpartum, colostrum has a higher
mula, when mother's own milk was unavailable [21,28]. We found no concentration of proteins and immunoactive components than the
clinical trial directly comparing the impact of donor milk with mother's mature milk [48]. The application of both bovine and human colostrum
own milk on the development of NEC. Given the current knowledge, an had been studied recently.
RCT may even be considered unethical. Recently, the DoMINO trial Studies in a preterm piglet NEC model suggest that bovine colos-
demonstrated no cognitive benefits when infants were randomized to trum protects against NEC improves the intestinal architecture and
donor breastmilk versus formula [29]. Donor human milk differs from decreases inflammation [49]. Based on these data, a feasibility and
mother's own breast milk with respect to caloric content and macro- tolerability pilot safety study on bovine colostrum administration in
nutrient composition, as well as the amount of bioactive ingredients. preterm infants was initiated in Denmark and China. The researchers
Holder pasteurization, performed to decrease bacteria and viruses, also found no difference in feeding tolerance or clinical adverse outcomes
diminishes or inactivates protective elements of mother's own milk, during the first two weeks of life, except a transient elevation of plasma
including secretory IgA, lactoferrin, commensal bacteria, and lysozymes tyrosine on day 7 in infants receiving bovine colostrum, an effect at-
[30,31]. tributed to their increased protein intake [50].
Several studies on the effect of oral human colostrum administration
2.3. Dosing of human milk in preterm infants and its relationship with the immune system con-
cluded with contradictory results. In a retrospective cohort, Seigel et al.
Multiple studies examined the effect of different doses of human demonstrated that colostrum could be safely administered orophar-
milk on the incidence of NEC. In the secondary analysis of a meta- yngeally to ELBW infants in the first two days of life and might provide
analysis, Abrams et al. [24] reported that the percentage of diet other the benefit of a better weight gain at 36 weeks [51]. This study was
than exclusive human milk was a significant predictor of NEC. The risk followed by an RCT of oral colostrum priming that showed significant
for NEC increased by 11–21% for every 10% increase in non-human reduction in the median length of hospitalization as compared to in-
milk in the diet. Two RCTs [21,32] reported that the incidence of NEC fants who did not receive oral colostrum, but no impact on salivary
appeared to decrease in those infants who received more than 50% of immune peptides and oral microbiota in preterm infants receiving oral
their total feeds made up of mother's own milk. The basis of this see- colostrum versus the control group [52]. Two other RCTs in preterm
mingly dose-dependent response is unclear and may be related to more infants demonstrated increase in salivary secretory IgA [53] and cir-
protection provided by a greater intake of human milk, or less formula. culating secretory immunoglobulin A [54]. Lee et al. noted a significant
decrease in clinical sepsis, as well as an increase in immunoprotective
3. Standardized feeding regimens and necrotizing enterocolitis factors and a decrease in pro-inflammatory cytokines in the colostrum
group [54].
The preterm gut is especially at risk for feeding intolerance. This is
most likely due to an immature gut barrier, microbial dysbiosis, limited 5. Human milk components
intestinal function and motility. A linear trajectory of feeding ad-
vancement is believed to be the most intuitive. Several studies have 5.1. Lactoferrin
shown that standardized feeding regimens result in a decreased in-
cidence of NEC [33–35]. Known for its antimicrobial properties, lactoferrin is the most
abundant protein in human milk whey [48] and may be effective in
3.1. Rate of advancement of feeds preventing NEC. Bovine lactoferrin (BLF) has a 69% similarity with the
human lactoferrin [48] but has a much lower concentration in bovine
At the present time, the subject of trophic feeding has divided milk. Animal studies demonstrated that lactoferrin might prevent in-
clinicians into those who delay the initiation of nutritive feeding, ex- testinal inflammation and NEC by interfering with activation of the pro-
tended the duration of trophic feeding for a few days to a week, and inflammatory cascade. It may compete with LPS in binding on Toll-like
those who advance enteral feedings without offering trophic feeding. receptor-4, inhibiting the activation of nuclear factor-κβ-regulated
There are clinical data suggesting that trophic feeding for a few days genes [55].
lead to a decreased risk of NEC when compared to early progressive A large multicenter trial comparing BLF, bovine lactoferrin with
feeding [36,37]. Conversely, prolonged trophic feeds have been shown Lactobacillus rhamnosus (BLF + LGG) and placebo in VLBW infants
to a delay time to full enteral feeding [38,39]. A delay in achievement showed a statistically significant reduction in NEC in the BLF + LGG
of full enteral feeds increases risk for nosocomial sepsis and infection, group and a trend towards decreasing NEC in BLF in comparison with
prolongs parenteral nutrition and central lines, extends length of hos- controls [56]. Of note, the incidence of NEC in the control group was
pital stay, and increases risk for postnatal growth restriction [38–43]. relatively low, at 5.3%. Another small trial from Turkey showed de-
Most current meta-analyses have not revealed any increased risk for creased sepsis but no difference in NEC, even though there was no NEC
NEC or feeding intolerance with early progression from trophic feeds, case in the lactoferrin-treated group [57]. A Cochrane review of

2
D.d.l. Cruz, C. Bazacliu Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

lactoferrin in preterm infants concluded that lactoferrin decreased NEC own milk combined with bovine milk fortifier, but the current evidence
stage II or greater by 30%, but the quality of evidence was marginal is insufficient to support this practice. Due to composition changes in-
[58]. Administration of recombinant human lactoferrin had no effect on duced by pasteurization and thawing to which human donor milk is
NEC rates, but significantly reduced in hospital-acquired infection in subjected, it does not seem to have the same protective effects against
the treatment group [59]. The results of the “Enteral lactoferrin in NEC without the presence of mother's own milk. Due to its undeniable
neonates” (ELFIN), a large RCT of lactoferrin in infants less than 32 benefits, extensive research aimed to identify protective components
weeks gestation that finished enrolling in 2017, is expected to shed has been done. Even though animal studies demonstrated that a variety
more light on the role lactoferrin may play in NEC prevention in the of human milk components seem to be effective in preventing and de-
future (www.npeu.ox.ac.uk). creasing severity of NEC, the few tested in human trials fail to show a
significant improvement. More research in this area is needed, though it
5.2. Human milk oligosaccharides faces a few problems. The animal models for NEC do not completely
recapitulate the premature infants' disease and it is likely that the array
Human milk oligosaccharides (HMOs) are a major bioactive com- of bioactive components from human milk act synergistically to
ponent of human milk that varies with maternal genetics and stage of dampen the immune system response and to nourish and protect the
lactation [60]. HMOs play an important role in microbial colonization intestinal epithelium. Clinically, NEC is a disease with an overall low
of the gut after birth, as suggested by the increase in fucosyltransferase incidence, making clinical trials require a high number of patients to
activity during the first postnatal weeks [61], and subsequently serve as enroll to adequately power for effect.
nutritional substrate for “good” bacteria influencing the development of
intestinal microbiome [60]. HMOs also inhibit bacterial pathogen ad-
herence to mucosal surfaces, protecting breast-fed infants against var- 6.1. Practice points
ious infections [62]. A sialated isomer, disialyllacto-N-tetraose, that
confers protection from NEC was identified in animal models by Jant- • Two feeding interventions decrease the risk for necrotizing en-
scher-Krenn et al. [63]. Sialylated galacto-oligosaccharides and 2′-fu- terocolitis: provision of mother's breast milk and standardization of
cosyllactose were also shown to reduce NEC in neonatal rodents [64]. feeding regimens.
Mechanistically, 2′-fucosyllactose improves intestinal perfusion by up- • Early progressive feeding and fortification are safe with the benefit
regulating endothelial nitric oxide synthase in a mouse model [65]. of better growth and shorter time to full feeds.
Colostrum HMOs can also be protective by attenuating the in- • Donor human breastmilk and human milk-based human milk for-
flammatory response and promoting the maturation of the intestinal tifier may contribute to NEC prevention.
mucosal immune system in ex-vivo immature intestinal cells culture • A higher dose of human breast milk (> 50% of feeds) reduces the
[66]. A large amount of fucosylated oligosaccharides, including se- incidence of NEC.
cretor antigen (H) are found in milk. The secretory status of the mother • Human milk components could not be recommended for prevention
determines the fucosylation of glycans in the milk [60] which may or treatment of NEC at this time.
further increase the risk of developing NEC in low or non-secretory
infants, as Morrow et al. demonstrated that premature infants with
6.2. Research directions
fucosyltransferase-2 low secretor phenotype have a ten-fold increase in
NEC [67].
• The role of human milk-based human milk fortification in the nu-
trition of the preterm infant and the prevention of NEC.
5.3. Other milk components
• The role of donor human milk in the nutrition of the extremely
premature infant.
A few other human milk components that have been studied for
their potential in preventing NEC are listed in Table 1.
• The role of oral colostrum in modulation of immune system and
microbiota.

6. Summary
• The role of individual or combination of human milk component in
prevention and treatment of disease.

Enteral feeds with mother's own milk and consistent feeding prac-
tices are the only proven and modifiable interventions that help prevent Conflicts of interest
a significant number of NEC cases. Exclusive human milk diet could,
theoretically, be better than the more customary combined mother's None declared.

Table 1
Milk components contributing to necrotizing enterocolitis (NEC) prevention.
Component Proposed mechanism Human studies

Transforming growth factor- • Bound to glycans: bioavailability is increased by • Highest in colostrum, decreased during lactation [69]
β2 low pH [48] • small
Low TGF-β2 in the milk of mothers whose infants developed NEC, but the study had a
• Decreases
[68]
inflammation and promotes repair sample size and was not powered for NEC [69]

Immunoglobulins • Trials used IgG, IgG/IgA and IgG/IgA/IgM


• Cochrane review of oral immunoglobulins for NEC prophylaxis concluded that oral Ig
did not result in significant reduction of NEC [70]
• No trial for IgA alone was done
Glutamine • Decreases gut permeability and preserves villi
structure in mature human intestine [71]
• Cochrane review meta-analysis of glutamine supplementation oral or enteral to
prevent mortality and morbidity included 12 studies did not show any differences in
mortality or morbidities, including NEC [72]
Arginine • Plasma concentration lower in infants who • Three small trials with contradictory results
developed NEC [73] • Cochrane review found some reduction in NEC stage I and III, but concluded that there
is insufficient evidence to support supplementation [74]

3
D.d.l. Cruz, C. Bazacliu Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

Funding sources very low birth weight infants during the first 60 days of life. Neonatology
2012;102:276–81.
[33] Wiedmeier S, Henry E, Baer V, et al. Center differences in NEC within one health-
None. care system may depend on feeding protocol. Am J Perinatol 2008;25:5–11.
[34] Patole SK, de Klerk N. Impact of standardised feeding regimens on incidence of
References neonatal necrotising enterocolitis: a systematic review and meta-analysis of ob-
servational studies. Arch Dis Child Fetal Neonatal Ed 2005;90:F147–51.
[35] Jasani B, Patole S. Standardized feeding regimen for reducing necrotizing en-
[1] Neu J, Walker WA. Necrotizing enterocolitis. N Engl J Med 2011;364:255–64. terocolitis in preterm infants: an updated systematic review. J Perinatol
[2] Horbar JD, Carpenter JH, Badger GJ, et al. Mortality and neonatal morbidity among 2017;37:827–33.
infants 501 to 1500 grams from 2000 to 2009. Pediatrics 2012;129:1019–26. [36] Berseth CL, Bisquera JA, Paje VU. Prolonging small feeding volumes early in life
[3] Ganapathy V, Hay JW, Kim JH. Costs of necrotizing enterocolitis and cost-effec- decreases the incidence of necrotizing enterocolitis in very low birth weight infants.
tiveness of exclusively human milk-based products in feeding extremely premature Pediatrics 2003;111:529–34.
infants. Breastfeed Med 2012;7:29–37. [37] Henderson G, Craig S, Brocklehurst P, McGuire W. Enteral feeding regimens and
[4] Ganapathy V, Hay JW, Kim JH, Lee ML, Rechtman DJ. Long term healthcare costs of necrotising enterocolitis in preterm infants: a multicentre case–control study. Arch
infants who survived neonatal necrotizing enterocolitis: a retrospective longitudinal Dis Child Fetal Neonatal Ed 2009;94:F120–3.
study among infants enrolled in Texas Medicaid. BMC Pediatr 2013;13:127. [38] Leaf A, Dorling J, Kempley S, et al. Early or delayed enteral feeding for preterm
[5] Bisquera JA, Cooper TR, Berseth CL. Impact of necrotizing enterocolitis on length of growth-restricted infants: a randomized trial. Pediatrics 2012;129:e1260–8.
stay and hospital charges in very low birth weight infants. Pediatrics [39] Morgan J, Young L, Mcguire W. Delayed introduction of progressive enteral feeds to
2002;109:423–8. prevent necrotising enterocolitis in very low birth weight infants. Cochrane
[6] Neu J. Necrotizing enterocolitis: the mystery goes on. Neonatology Database Syst Rev 2014;12. CD001970.
2014;106:289–95. [40] Morgan J, Young L, Mcguire W. Slow advancement of enteral feed volumes to
[7] Torrazza RM, Neu J. The altered gut microbiome and necrotizing enterocolitis. Clin prevent necrotising enterocolitis in very low birth weight infants. Cochrane
Perinatol 2013;40:93–108. Database Syst Rev 2014;12. CD001241.
[8] Patole S. Prevention and treatment of necrotising enterocolitis in preterm neonates. [41] Stoll BJ, Hansen NI, Adams-Chapman I, et al. Neurodevelopmental and growth
Early Hum Dev 2007;83:635–42. impairment among extremely low-birth-weight infants with neonatal infection.
[9] Morowitz MJ, Poroyko V, Caplan M, Alverdy J, Liu DC. Redefining the role of in- J Am Med Assoc 2004;292:2357.
testinal microbes in the pathogenesis of necrotizing enterocolitis. Pediatrics [42] Karagol BS, Zenciroglu A, Okumus N, Polin RA. Randomized controlled trial of slow
2010;125:777–85. vs rapid enteral feeding advancements on the clinical outcomes of preterm infants
[10] Carlisle EM, Morowitz MJ. The intestinal microbiome and necrotizing enterocolitis. with birth weight 750–1250 g. J Parenter Enteral Nutr 2013;37:223–8.
Curr Opin Pediatr 2013;25:382–7. [43] Ehrenkranz RA, Dusick AM, Vohr BR, Wright LL, Wrage LA, Poole WK. Growth in
[11] Lin PW, Stoll BJ. Necrotising enterocolitis. Lancet 2006;368(9543):1271–83. the neonatal intensive care unit influences neurodevelopmental and growth out-
[12] Section on Breastfeeding. Breastfeeding and the use of human milk. Pediatrics comes of extremely low birth weight infants. Pediatrics 2006;117:1253–61.
2012;129:e827–41. [44] Picaud J-C, Houeto N, Buffin R, Loys C-M, Godbert I, Haÿs S. Additional protein
[13] Bharwani SK, Green BF, Pezzullo JC, Bharwani SS, Bharwani SS, Dhanireddy R. fortification is necessary in extremely low-birth-weight infants fed human milk.
Systematic review and meta-analysis of human milk intake and retinopathy of J Pediatr Gastroenterol Nutr 2016;63:103–5.
prematurity: a significant update. J Perinatol 2016;36:913–20. [45] Rosas R, Sanz M, Fernández-Calle P, et al. Experimental study showed that adding
[14] Patel AL, Johnson TJ, Engstrom JL, et al. Impact of early human milk on sepsis and fortifier and extra-hydrolysed proteins to preterm infant mothers' milk increased
health-care costs in very low birth weight infants. J Perinatol 2013;33:514–9. osmolality. Acta Paediatr 2016;105:e555–60.
[15] Lechner BE, Vohr BR. Neurodevelopmental outcomes of preterm infants fed human [46] Shah SD, Dereddy N, Jones TL, Dhanireddy R, Talati AJ. Early versus delayed
milk: a systematic review. Clin Perinatol 2017;44:69–83. human milk fortification in very low birth weight infants – a randomized controlled
[16] Singhal A, Cole TJ, Fewtrell M, Lucas A. Breastmilk feeding and lipoprotein profile trial. J Pediatr 2016;174. 126–31.e1.
in adolescents born preterm: follow-up of a prospective randomised study. Lancet [47] Mukhopadhyay K, Narnag A, Mahajan R. Effect of human milk fortification in ap-
2004;363(9421):1571–8. propriate for gestation and small for gestation preterm babies: a randomized con-
[17] Singhal A, Cole TJ, Lucas A. Early nutrition in preterm infants and later blood trolled trial. Indian Pediatr 2007;44:286–90.
pressure: two cohorts after randomised trials. Lancet 2001;357(9254):413–9. [48] Ballard O, Morrow AL. Human milk composition. Pediatr Clin 2013;60:49–74.
[18] Isaacs EB, Fischl BR, Quinn BT, Chong WK, Gadian DG, Lucas A. Impact of breast [49] Shen RL, Thymann T, Østergaard MV, et al. Early gradual feeding with bovine
milk on intelligence quotient, brain size, and white matter development. Pediatr colostrum improves gut function and NEC resistance relative to infant formula in
Res 2010;67:357–62. preterm pigs. Am J Physiol Gastrointest Liver Physiol 2015;309:G310–23.
[19] Sullivan S, Schanler RJ, Kim JH, et al. An exclusively human milk-based diet is [50] Juhl SM, Ye X, Zhou P, et al. Bovine colostrum for preterm infants in the first days of
associated with a lower rate of necrotizing enterocolitis than a diet of human milk life. J Pediatr Gastroenterol Nutr 2018;66:471–8.
and bovine milk-based products. J Pediatr 2010;156. 562–567.e1. [51] Seigel JK, Smith PB, Ashley PL, et al. Early administration of oropharyngeal co-
[20] Cristofalo EA, Schanler RJ, Blanco CL, et al. Randomized trial of exclusive human lostrum to extremely low birth weight infants. Breastfeed Med 2013;8:491–5.
milk versus preterm formula diets in extremely premature infants. J Pediatr [52] Romano-Keeler J, Azcarate-Peril MA, Weitkamp J-H, et al. Oral colostrum priming
2013;163. 1592–5.e1. shortens hospitalization without changing the immunomicrobial milieu. J Perinatol
[21] Schanler RJ, Lau C, Hurst NM, Smith EO. Randomized trial of donor human milk 2017;37:36–41.
versus preterm formula as substitutes for mothers' own milk in the feeding of ex- [53] Glass K, Greecher C, Doheny K. Oropharyngeal administration of colostrum in-
tremely premature infants. Pediatrics 2005;116:400–6. creases salivary secretory IgA levels in very low-birth-weight infants. Am J
[22] Meinzen-Derr J, Poindexter B, Wrage L, Morrow AL, Stoll B, Donovan EF. Role of Perinatol 2017;34:1389–95.
human milk in extremely low birth weight infants' risk of necrotizing enterocolitis [54] Lee J, Kim H-S, Jung YH, et al. Oropharyngeal colostrum administration in ex-
or death. J Perinatol 2009;29:57–62. tremely premature infants: an RCT. Pediatrics 2015;135:e357–66.
[23] Lucas A, Cole TJ. Breast milk and neonatal necrotising enterocolitis. Lancet [55] Actor JK, Hwang S-A, Kruzel ML. Lactoferrin as a natural immune modulator. Curr
336(8730):1519–1523. Pharmaceut Des 2009;15:1956–73.
[24] Abrams SA, Schanler RJ, Lee ML, Rechtman DJ. Greater mortality and morbidity in [56] Manzoni P, Meyer M, Stolfi I, et al. Bovine lactoferrin supplementation for pre-
extremely preterm infants fed a diet containing cow milk protein products. vention of necrotizing enterocolitis in very-low-birth-weight neonates: a rando-
Breastfeed Med 2014;9:281–5. mized clinical trial. Early Hum Dev 2014;90:S60–5.
[25] Brown JV, Embleton ND, Harding JE, McGuire W. Multi-nutrient fortification of [57] Akin IM, Atasay B, Dogu F, et al. Oral lactoferrin to prevent nosocomial sepsis and
human milk for preterm infants. Cochrane Database Syst Rev 2016;5. CD000343. necrotizing enterocolitis of premature neonates and effect on T-regulatory cells. Am
[26] Quigley M, McGuire W. Formula versus donor breast milk for feeding preterm or J Perinatol 2014;31:1111–20.
low birth weight infants. Cochrane Database Syst Rev 2014;4. CD002971. [58] Pammi M, Abrams SA. Oral lactoferrin for the prevention of sepsis and necrotizing
[27] Boyd CA, Quigley MA, Brocklehurst P. Donor breast milk versus infant formula for enterocolitis in preterm infants. Cochrane Database Syst Rev 2015;2. CD007137.
preterm infants: systematic review and meta-analysis. Arch Dis Child Fetal Neonatal [59] Sherman MP, Adamkin DH, Niklas V, et al. Randomized controlled trial of ta-
Ed 2007;92:F169–75. lactoferrin oral solution in preterm infants. J Pediatr 2016;175. 68–73.e3.
[28] Corpeleijn WE, de Waard M, Christmann V, et al. Effect of donor milk on severe [60] Underwood MA, Gaerlan S, De Leoz MLA, et al. Human milk oligosaccharides in
infections and mortality in very low-birth-weight infants. JAMA Pediatr premature infants: absorption, excretion and influence on the intestinal microbiota.
2016;170:654. Pediatr Res 2015;78:670–7.
[29] O'Connor DL, Gibbins S, Kiss A, et al. Effect of supplemental donor human milk [61] Nanthakumar NN, Dai D, Newburg DS, Walker WA. The role of indigenous mi-
compared with preterm formula on neurodevelopment of very low-birth-weight croflora in the development of murine intestinal fucosyl- and sialyltransferases.
infants at 18 months. J Am Med Assoc 2016;316:1897. Faseb J 2003;17:44–6.
[30] Peila C, Moro GE, Bertino E, et al. The effect of holder pasteurization on nutrients [62] Morrow AL, Ruiz-Palacios GM, Jiang X, Newburg DS. Human-milk glycans that
and biologically-active components in donor human milk: a review. Nutrients inhibit pathogen binding protect breast-feeding infants against infectious diarrhea.
2016;8(8). J Nutr 2005;135:1304–7.
[31] Ewaschuk JB, Unger S, O'Connor DL, et al. Effect of pasteurization on selected [63] Jantscher-Krenn E, Zherebtsov M, Nissan C, et al. The human milk oligosaccharide
immune components of donated human breast milk. J Perinatol 2011;31:593–8. disialyllacto-N-tetraose prevents necrotising enterocolitis in neonatal rats. Gut
[32] Corpeleijn WE, Kouwenhoven SMP, Paap MC, et al. Intake of own mother's milk 2012;61:1417–25.
during the first days of life is associated with decreased morbidity and mortality in [64] Autran CA, Schoterman MHC, Jantscher-Krenn E, Kamerling JP, Bode L. Sialylated

4
D.d.l. Cruz, C. Bazacliu Seminars in Fetal and Neonatal Medicine xxx (xxxx) xxx–xxx

galacto-oligosaccharides and 2′-fucosyllactose reduce necrotising enterocolitis in [69] Frost BL, Jilling T, Lapin B, Maheshwari A, Caplan MS. Maternal breast milk
neonatal rats. Br J Nutr 2016;116:294–9. transforming growth factor-beta and feeding intolerance in preterm infants. Pediatr
[65] Good M, Sodhi CP, Yamaguchi Y, et al. The human milk oligosaccharide 2′-fuco- Res 2014;76:386–93.
syllactose attenuates the severity of experimental necrotising enterocolitis by en- [70] Foster JP, Seth R, Cole MJ. Oral immunoglobulin for preventing necrotizing en-
hancing mesenteric perfusion in the neonatal intestine. Br J Nutr terocolitis in preterm and low birth weight neonates. Cochrane Database Syst Rev
2016;116:1175–87. 2016;4. CD001816.
[66] He Y, Liu S, Leone S, Newburg DS. Human colostrum oligosaccharides modulate [71] van der Hulst RR, van Kreel BK, von Meyenfeldt MF, et al. Glutamine and the
major immunologic pathways of immature human intestine. Mucosal Immunol preservation of gut integrity. Lancet 1993;341(8857):1363–5.
2014;7:1326–39. [72] Moe-Byrne T, Brown JVE, McGuire W. Glutamine supplementation to prevent
[67] Morrow AL, Meinzen-Derr J, Huang P, et al. Fucosyltransferase 2 non-secretor and morbidity and mortality in preterm infants. Cochrane Database Syst Rev 2016;1.
low secretor status predicts severe outcomes in premature infants. J Pediatr CD001457.
2011;158:745–51. [73] Becker RM, Wu G, Galanko JA, et al. Reduced serum amino acid concentrations in
[68] Maheshwari A, Kelly DR, Nicola T, et al. TGF-β2 suppresses macrophage cytokine infants with necrotizing enterocolitis. J Pediatr 2000;137:785–93.
production and mucosal inflammatory responses in the developing intestine. [74] Shah PS, Shah VS, Kelly LE. Arginine supplementation for prevention of necrotising
Gastroenterology 2011;140:242–53. enterocolitis in preterm infants. Cochrane Database Syst Rev 2017;4. CD004339.

Das könnte Ihnen auch gefallen