Warning: fopen(/home/virtual/pediatrics/journal/upload/ip_log/ip_log_2024-04.txt) [function.fopen]: failed to open stream: Permission denied in /home/virtual/pediatrics/journal/ip_info/view_data.php on line 82

Warning: fwrite(): supplied argument is not a valid stream resource in /home/virtual/pediatrics/journal/ip_info/view_data.php on line 83
An update on necrotizing enterocolitis: pathogenesis and preventive strategies

An update on necrotizing enterocolitis: pathogenesis and preventive strategies

Article information

Korean J Pediatr. 2011;54(9):368-372
Publication date (electronic) : 2011 September 30
doi : https://doi.org/10.3345/kjp.2011.54.9.368
Department of Pediatrics, Ajou University School of Medicine, Suwon, Korea.
Corresponding author: Jang Hoon Lee, MD. Department of Pediatrics, Ajou University School of Medicine, San 5 Woncheon-dong, Yeongtong-gu, Suwon 443-721, Korea. Tel: +82-31-219-5165, Fax: +82-31-219-5169, neopedlee@gmail.com
Received 2011 July 29; Accepted 2011 August 28.

Abstract

Necrotizing enterocolitis (NEC) is one of the most critical morbidities in preterm infants. The incidence of NEC is 7% in very-low-birth-weight infants, and its mortality is 15 to 30%. Infants who survive NEC have various complications, such as nosocomial infection, malnutrition, growth failure, bronchopulmonary dysplasia, retinopathy of prematurity, and neurodevelopmental delays. The most important etiology in the pathogenesis of NEC is structural and immunological intestinal immaturity. In preterm infants with immature gastrointestinal tracts, development of NEC may be associated with a variety of factors, such as colonization with pathogenic bacteria, secondary ischemia, genetic polymorphisms conferring NEC susceptibility, anemia with red blood cell transfusion, and sensitization to cow milk proteins. To date, a variety of preventive strategies has been accepted or attempted in clinical practice with regard to the pathogenesis of NEC. These strategies include the use of breast feeding, various feeding strategies, probiotics, prebiotics, glutamine and arginine, and lactoferrin. There is substantial evidence for the efficacy of breast feeding and the use of probiotics in infants with birth weights above 1,000 g, and these strategies are commonly used in clinical practice. Other preventive strategies, however, require further research to establish their effect on NEC.

Introduction

Necrotizing enterocolitis (NEC) is one of the most critical morbidities occurring in preterm infants. In spite of improvements in neonatal intensive care, the incidence of NEC has increased to 7% in very-low-birth-weight (VLBW) infants1). Mortality from NEC is 15 to 30% and is especially high in infants with lower birth weight, earlier gestation, and surgical interventions2). Infants who have recovered from NEC are more susceptible to nosocomial infection, malnutrition, growth failure, bronchopulmonary dysplasia, retinopathy of prematurity, and longer hospitalization3). In addition, severe NEC requiring surgical intervention is a significant risk factor for adverse neurodevelopmental outcomes4). Although advances in NEC research have been achieved, much remains to be elucidated on disease pathophysiology. NEC arises from multifactorial origins, including intestinal immaturity, infection, and ischemia.

Prevention is important for the management of NEC, because treatment strategies may be ineffective in some cases of clinically significant NEC. In this article, the author reviews the current literature on factors predisposing infants to NEC and strategies for its prevention and management.

Pathogenesis

1. Intestinal immaturity

An underdeveloped gastrointestinal tract in preterm infants may trigger the development of NEC. Decreased intestinal peristalsis may result in extended exposure of the intestinal epithelium to noxious substances. Immature mucus coatings and incompletely formed tight junctions also contribute to disease pathogenesis5,6). Additionally, the gastrointestinal tract's immunological functions are too immature in preterm infants to adequately respond to colonization by pathogenic bacteria7). For example, Toll-like Receptor-4 expression is down-regulated in the mature intestinal epithelium upon stimulation by gram-negative lipopolysaccharide but is increased in the immature intestinal epithelium, eliciting an exaggerated pro-inflammatory response through up-regulation of the NF-kB pathway8,9).

2. Infection and colonization by pathogenic bacteria

Prolonged antibiotic exposure is associated with an increased risk of NEC. This association persisted in multivariate analyses that excluded confounding factors, such as gestational age, birth weight, and sepsis10). Prolonged antibiotic exposure may not only delay beneficial colonization by normal gastrointestinal flora, but may also promote colonization by pathogenic bacteria11). In some instances, the replication rates of pathogens facilitate their outcompetition of normal flora, predisposing preterm infants to NEC, especially in conjunction with intestinal barrier impairment12).

3. Ischemia

Preterm infants frequently experience ischemic events, such as hypotension, hypothermia, anemia, and patent ductus arteriosus during intensive care. Although experimental and clinical studies have excluded ischemia as a major trigger for NEC13,14), it may play a secondary role in the pathogenesis. Ischemia can disrupt endothelial cell function and alter the endothelin-1/nitric oxide balance in favor of vasoconstriction, causing expansion of ischemic intestinal lesions15,16).

4. Additional contributing factors

Small studies have identified contributions from specific genetic polymorphisms, such as mutations in carbamoyl phosphate synthetase17), vascular endothelial growth factor18), and interleukin-10 and 1219) in the development of NEC. There are also concerns that NEC may be associated with anemia with red blood cell (RBC) transfusion20,21). Although an exact mechanism remains undefined, it is suggested that severe anemia results in insufficient oxygen to meet the increased requirements of mesenteric vessels after enteral feeding. RBC transfusion may also interrupt the mesenteric vascular tone via an imbalance of nitric oxide and endothelin-1, stimulating the production of pro-inflammatory cytokines as occurs during multiple organ failure22). Recently, several in vitro studies have reported that sensitization to cow milk proteins may be involved in NEC pathogenesis23,24). At present, however, data are insufficient to determine the involvement of these factors in the pathogenesis of NEC.

Preventive strategies

1. Enteral feeding strategies

Human breast milk may protect against NEC by inhibiting gut colonization by pathogenic flora, enhancing maturation of the intestinal barrier, and controlling the pro-inflammatory response. A meta-analysis of a few small randomized controlled trials concluded that human breast milk confers a protective effect against NEC25,26). However, these trials varied in their definitions of breast milk and in trial design parameters, such as maternal vs. donor milk, term vs. preterm, fortified vs. unfortified, and feeding exclusively with human breast milk vs. supplementation with formula. A recent randomized controlled trial reported that an exclusively human-milk-based diet (i.e., human breast milk and a human-based fortifier) significantly reduced the incidence of NEC and surgical NEC27). In a metaanalysis comparing donor breast milk to infant formula in low-birthweight infants, formula feeding was correlated with a higher incidence of NEC (typical relative risk [RR]. 2.5; 95% confidence interval [CI], 1.2 to 5.1), although it increased short term growth rate28).

Berseth et al.29) reported that prolonging small feeding volumes early in life decreased the incidence of NEC in VLBW infants compared with feeding advancement (1.4% vs. 10%, P=0.03). Nevertheless, slow advancement of enteral feeding (15 to 20 mL/kg/day) did not reduce NEC incidence compared with rapid feeding advancement in VLBW infants (30 to 35 mL/kg/day)30). Early trophic feeding in VLBW infants did not increase the incidence of NEC31,32). Meanwhile, continuous nasogastric tube feeding did not decrease NEC compared with bolus milk feeding33). However, these findings are limited by insufficient randomized controlled trials.

2. Probiotics

Immediately after birth, colonization of the intestine in healthy, term infants begins with maternal vaginal flora. Bifidobacterium species are usually dominant among the complex intestinal flora that develops during the first few post-natal weeks in breast-fed infants. Preterm infants may have abnormal colonization by intestinal flora due to exposure to pathogenic bacteria in the intensive care unit, antibiotic use, prolonged fasting, and less feeding with breast milk.

Probiotics may protect against the development of NEC through enhancement of the barrier preventing bacterial migration across the intestinal mucosa34), competition with pathogenic flora35), and modification of the host's response to microbial products36).

A recent meta-analysis reported the effect of probiotics on the prevention of NEC37). Enteral probiotic supplementation significantly reduced the incidence of severe NEC (stage II or higher; typical RR, 0.35; 95% CI, 0.24 to 0.52) and mortality (typical RR, 0.40; 95% CI, 0.27 to 0.60). Probiotic supplementation also significantly decreased NEC incidence in subgroup analyses of VLBW infants (typical RR, 0.34; 95% CI, 0.23 to 0.50). There have been no reports of systemic infections with probiotic supplementation in eligible trials. However, there are limitations to the meta-analysis; Birth weight and gestational age of subjects in trials were highly variable, and probiotic supplementation differed widely with regard to timing, dosage, and species of probiotics. Most importantly, no data were reported for efficacy and safety of probiotics in extremely-low-birth-weight infants, the most vulnerable patients.

3. Prebiotics

Prebiotics are indigestible substances that selectively promote the growth and colonization of probiotic lactobacilli or bifidobacteria38,39). Prebiotics can be classified as either natural substances in human breast milk or synthetic substances, such as a mixture of galactooligosaccharides and fructo-oligosaccharides. Riskin et al.40) reported that a low dose of lactulose promotes enteral feeding and has a trend toward a lower incidence of NEC. Although randomized controlled trials of prebiotic supplementation in preterm infants have not conclusively proven their preventive effect on NEC41,42), they may be efficacious in this population.

4. Glutamine and arginine

In critical patients, Glutamine can be an essential amino acid. It plays an important role as a metabolic fuel43) and also maintains the functional integrity of the gut44). A meta-analysis of 5 randomized controlled trials on the effects of glutamine supplementation on NEC showed no statistically effect on its incidence of NEC45), but a recent randomized trial reported its statistically significant reduction of NEC incidence46).

Arginine is an amino acid important for the formation of nitric oxide47). Compared to control infants, preterm infants with NEC have significantly decreased levels of plasma arginine48). Amin et al.49) reported that parenteral arginine supplementation increases plasma arginine levels and decreases NEC incidence. However, interpretation of this study is confounded by its inclusion of many infants with low grade NEC (stage I) in the control group.

5. Additional preventive strategies

Lactoferrin is a major whey protein in mammalian milk and has an important function in the innate immune system. There is high amino acid homology (77%) between human and bovine lactoferrin50). In a randomized controlled trial, enteral bovine lactoferrin supplementation decreased the incidence of NEC when combination with Lactobacillus rhamnosus GG51). In addition to the preventive strategies reviewed here, epidermal growth factor (EGF)52), heparin-binding EGF53), endothelin-1 receptor antagonist54), and ω-3 fatty acids55) may be candidates for NEC prevention, but confirmatory evidence is required.

Conclusion

Despite improvements in neonatal intensive care, NEC remains a critical disease in preterm infants and confers a high incidence of mortality and many serious complications. In preterm infants with insufficient maturation of the gastrointestinal tract, development of NEC may be associated with a variety of factors, including colonization by pathogenic bacteria, secondary ischemia, genetic polymorphisms conferring susceptibility to NEC, anemia with RBC transfusion, and sensitization to cow milk proteins. To date, a variety of preventive strategies have been developed or used in clinical practice, including the use of breast feeding, various feeding strategies, probiotics, prebiotics, glutamine and arginine, and lactoferrin. Breast feeding and probiotic use in infants with birth weights above 1,000 g have been proven effective in clinical practice. However, other strategies currently lack sufficient evidence for routine use as preventive measures. Therefore, clinical trials enrolling large numbers of infants and employing rigorous controls will be essential to prove the efficacy of other strategies in preventing NEC.

References

1. Holman RC, Stoll BJ, Curns AT, Yorita KL, Steiner CA, Schonberger LB. Necrotising enterocolitis hospitalisations among neonates in the United States. Paediatr Perinat Epidemiol 2006;20:498–506. 17052286.
2. Guthrie SO, Gordon PV, Thomas V, Thorp JA, Peabody J, Clark RH. Necrotizing enterocolitis among neonates in the United States. J Perinatol 2003;23:278–285. 12774133.
3. Leviton A, Dammann O, Engelke S, Allred E, Kuban KC, O'Shea TM, et al. The clustering of disorders in infants born before the 28th week of gestation. Acta Paediatr 2010;99:1795–1800. 20712837.
4. Rees CM, Pierro A, Eaton S. Neurodevelopmental outcomes of neonates with medically and surgically treated necrotizing enterocolitis. Arch Dis Child Fetal Neonatal Ed 2007;92:F193–F198. 16984980.
5. Allen A, Bell A, Mantle M, Pearson JP. The structure and physiology of gastrointestinal mucus. Adv Exp Med Biol 1982;144:115–133. 7044056.
6. Muresan Z, Paul DL, Goodenough DA. Occludin 1B, a variant of the tight junction protein occludin. Mol Biol Cell 2000;11:627–634. 10679019.
7. Hunter CJ, Upperman JS, Ford HR, Camerini V. Understanding the susceptibility of the premature infant to necrotizing enterocolitis (NEC). Pediatr Res 2008;63:117–123. 18091350.
8. Lotz M, Gütle D, Walther S, Ménard S, Bogdan C, Hornef MW. Postnatal acquisition of endotoxin tolerance in intestinal epithelial cells. J Exp Med 2006;203:973–984. 16606665.
9. Claud EC, Zhang X, Petrof EO, Sun J. Developmentally regulated tumor necrosis factor-alpha induced nuclear factor-kappaB activation in intestinal epithelium. Am J Physiol Gastrointest Liver Physiol 2007;292:G1411–G1419. 17307728.
10. Alexander VN, Northrup V, Bizzarro MJ. Antibiotic exposure in the newborn intensive care unit and the risk of necrotizing enterocolitis. J Pediatr 2011;159:392–397. 21489560.
11. Wang Y, Hoenig JD, Malin KJ, Qamar S, Petrof EO, Sun J, et al. 16S rRNA gene-based analysis of fecal microbiota from preterm infants with and without necrotizing enterocolitis. ISME J 2009;3:944–954. 19369970.
12. Claud EC, Walker WA. Hypothesis: inappropriate colonization of the premature intestine can cause neonatal necrotizing enterocolitis. FASEB J 2001;15:1398–1403. 11387237.
13. Caplan MS, Hedlund E, Adler L, Hsueh W. Role of asphyxia and feeding in a neonatal rat model of necrotizing enterocolitis. Pediatr Pathol 1994;14:1017–1028. 7855004.
14. Stoll BJ. Epidemiology of necrotizing enterocolitis. Clin Perinatol 1994;21:205–218. 8070222.
15. Nowicki PT. Ischemia and necrotizing enterocolitis: where, when, and how. Semin Pediatr Surg 2005;14:152–158. 16084402.
16. Nankervis CA, Giannone PJ, Reber KM. The neonatal intestinal vasculature: contributing factors to necrotizing enterocolitis. Semin Perinatol 2008;32:83–91. 18346531.
17. Moonen RM, Paulussen AD, Souren NY, Kessels AG, Rubio-Gozalbo ME, Villamor E. Carbamoyl phosphate synthetase polymorphisms as a risk factor for necrotizing enterocolitis. Pediatr Res 2007;62:188–190. 17597649.
18. Bányász I, Bokodi G, Vásráhelyi B, Treszl A, Derzbach L, Szabó A, et al. Genetic polymorphisms for vascular endothelial growth factor in perinatal complications. Eur Cytokine Netw 2006;17:266–270. 17353160.
19. Treszl A, Kaposi A, Hajdú J, Szabó M, Tulassay T, Vásárhelyi B. The extent to which genotype information may add to the prediction of disturbed perinatal adaptation: none, minor, or major? Pediatr Res 2007;62:610–614. 17805198.
20. El-Dib M, Narang S, Lee E, Massaro AN, Aly H. Red blood cell transfusion, feeding and necrotizing enterocolitis in preterm infants. J Perinatol 2011;31:183–187. 21252964.
21. Singh R, Visintainer PF, Frantz ID 3rd, Shah BL, Meyer KM, Favila SA, et al. Association of necrotizing enterocolitis with anemia and packed red blood cell transfusions in preterm infants. J Perinatol 2011;31:176–182. 21273983.
22. Reber KM, Nankervis CA, Nowicki PT. Newborn intestinal circulation. Physiology and pathophysiology. Clin Perinatol 2002;29:23–39. 11917738.
23. Chuang SL, Hayes PJ, Ogundipe E, Haddad M, MacDonald TT, Fell JM. Cow's milk protein-specific T-helper type I/II cytokine responses in infants with necrotizing enterocolitis. Pediatr Allergy Immunol 2009;20:45–52. 18298426.
24. Abdelhamid AE, Chuang SL, Hayes P, Fell JM. In vitro cow's milk protein-specific inflammatory and regulatory cytokine responses in preterm infants with necrotizing enterocolitis and sepsis. Pediatr Res 2011;69:165–169. 20975616.
25. Boyd CA, Quigley MA, Brocklehurst P. Donor breast milk versus infant formula for preterm infants: systematic review and meta-analysis. Arch Dis Child Fetal Neonatal Ed 2007;92:F169–F175. 16556615.
26. McGuire W, Anthony MY. Donor human milk versus formula for preventing necrotising enterocolitis in preterm infants: systematic review. Arch Dis Child Fetal Neonatal Ed 2003;88:F11–F14. 12496220.
27. Sullivan S, Schanler RJ, Kim JH, Patel AL, Trawöger R, Kiechl-Kohlendorfer U, et al. An exclusively human milk-based diet is associated with a lower rate of necrotizing enterocolitis than a diet of human milk and bovine milk-based products. J Pediatr 2010;156:562–567.e1. 20036378.
28. Quigley MA, Henderson G, Anthony MY, McGuire W. Formula milk versus donor breast milk for feeding preterm or low birth weight infants. Cochrane Database Syst Rev 2007;(4):CD002971. 17943776.
29. Berseth CL, Bisquera JA, Paje VU. Prolonging small feeding volumes early in life decreases the incidence of necrotizing enterocolitis in very low birth weight infants. Pediatrics 2003;111:529–534. 12612232.
30. Morgan J, Young L, McGuire W. Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants. Cochrane Database Syst Rev 2011;(3):CD001241. 21412870.
31. Bombell S, McGuire W. Early trophic feeding for very low birth weight infants. Cochrane Database Syst Rev 2009;(3):CD000504. 19588318.
32. Jeon GW, Park SE, Choi CW, Hwang JH, Chang YS, Park WS. The effects of early enteral feeding in extremely low birth-weight infants. Korean J Pediatr 2005;48:711–715.
33. Premji S, Chessell L. Continuous nasogastric milk feeding versus intermittent bolus milk feeding for premature infants less than 1500 grams. Cochrane Database Syst Rev 2003;(1):CD001819. 12535414.
34. Orrhage K, Nord CE. Factors controlling the bacterial colonization of the intestine in breastfed infants. Acta Paediatr Suppl 1999;88:47–57. 10569223.
35. Reid G, Howard J, Gan BS. Can bacterial interference prevent infection? Trends Microbiol 2001;9:424–428. 11553454.
36. Duffy LC. Interactions mediating bacterial translocation in the immature intestine. J Nutr 2000;130(2S Suppl):432S–436S. 10721921.
37. Alfaleh K, Anabrees J, Bassler D, Al-Kharfi T. Probiotics for prevention of necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev 2011;(3):CD005496. 21412889.
38. Kapiki A, Costalos C, Oikonomidou C, Triantafyllidou A, Loukatou E, Pertrohilou V. The effect of a fructo-oligosaccharide supplemented formula on gut flora of preterm infants. Early Hum Dev 2007;83:335–339. 16978805.
39. Agostoni C, Axelsson I, Goulet O, Koletzko B, Michaelsen KF, Puntis JW, et al. Prebiotic oligosaccharides in dietetic products for infants: a commentary by the ESPGHAN Committee on Nutrition. J Pediatr Gastroenterol Nutr 2004;39:465–473. 15572882.
40. Riskin A, Hochwald O, Bader D, Srugo I, Naftali G, Kugelman A, et al. The effects of lactulose supplementation to enteral feedings in premature infants: a pilot study. J Pediatr 2010;156:209–214. 19879595.
41. Westerbeek EA, van den Berg JP, Lafeber HN, Fetter WP, Boehm G, Twisk JW, et al. Neutral and acidic oligosaccharides in preterm infants: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr 2010;91:679–686. 20032496.
42. Indrio F, Riezzo G, Raimondi F, Francavilla R, Montagna O, Valenzano ML, et al. Prebiotics improve gastric motility and gastric electrical activity in preterm newborns. J Pediatr Gastroenterol Nutr 2009;49:258–261. 19561548.
43. Windmueller HG, Spaeth AE. Respiratory fuels and nitrogen metabolism in vivo in small intestine of fed rats. Quantitative importance of glutamine, glutamate, and aspartate. J Biol Chem 1980;255:107–112. 7350142.
44. Li N, Lewis P, Samuelson D, Liboni K, Neu J. Glutamine regulates Caco-2 cell tight junction proteins. Am J Physiol Gastrointest Liver Physiol 2004;287:G726–G733. 15130874.
45. Tubman TR, Thompson SW, McGuire W. Glutamine supplementation to prevent morbidity and mortality in preterm infants. Cochrane Database Syst Rev 2008;(1):CD001457. 18253992.
46. Sevastiadou S, Malamitsi-Puchner A, Costalos C, Skouroliakou M, Briana DD, Antsaklis A, et al. The impact of oral glutamine supplementation on the intestinal permeability and incidence of necrotizing enterocolitis/septicemia in premature neonates. J Matern Fetal Neonatal Med 2011;24:1294–1300. 21463215.
47. Castillo L, DeRojas-Walker T, Yu YM, Sanchez M, Chapman TE, Shannon D, et al. Whole body arginine metabolism and nitric oxide synthesis in newborns with persistent pulmonary hypertension. Pediatr Res 1995;38:17–24. 7478791.
48. Zamora SA, Amin HJ, McMillan DD, Kubes P, Fick GH, Bützner JD, et al. Plasma L-arginine concentrations in premature infants with necrotizing enterocolitis. J Pediatr 1997;131:226–232. 9290608.
49. Amin HJ, Zamora SA, McMillan DD, Fick GH, Butzner JD, Parsons HG, et al. Arginine supplementation prevents necrotizing enterocolitis in the premature infant. J Pediatr 2002;140:425–431. 12006956.
50. Buccigrossi V, de Marco G, Bruzzese E, Ombrato L, Bracale I, Polito G, et al. Lactoferrin induces concentration-dependent functional modulation of intestinal proliferation and differentiation. Pediatr Res 2007;61:410–414. 17515863.
51. Manzoni P, Rinaldi M, Cattani S, Pugni L, Romeo MG, Messner H, et al. Bovine lactoferrin supplementation for prevention of late-onset sepsis in very low-birth-weight neonates: a randomized trial. JAMA 2009;302:1421–1428. 19809023.
52. Dvorak B, Halpern MD, Holubec H, Williams CS, McWilliam DL, Dominguez JA, et al. Epidermal growth factor reduces the development of necrotizing enterocolitis in a neonatal rat model. Am J Physiol Gastrointest Liver Physiol 2002;282:G156–G164. 11751169.
53. Feng J, El-Assal ON, Besner GE. Heparin-binding epidermal growth factor-like growth factor decreases the incidence of necrotizing enterocolitis in neonatal rats. J Pediatr Surg 2006;41:144–149. 16410124.
54. Nowicki PT, Dunaway DJ, Nankervis CA, Giannone PJ, Reber KM, Hammond SB, et al. Endothelin-1 in human intestine resected for necrotizing enterocolitis. J Pediatr 2005;146:805–810. 15973323.
55. Ohtsuka Y, Okada K, Yamakawa Y, Ikuse T, Baba Y, Inage E, et al. ω-3 fatty acids attenuate mucosal inflammation in premature rat pups. J Pediatr Surg 2011;46:489–495. 21376198.

Article information Continued