ArticlePDF Available

Neonatal Necrotizing Enterocolitis

Authors:

Abstract and Figures

Akhil Maheshwari1, Laura L Corbin2, Robert L Schelonka21Division of Neonatology and Center for Neonatal and Pediatric Gastrointestinal Disease, Department of Pediatrics, University of Illinois at Chicago, IL, 2Division of Neonatology, Department of Pediatrics, Oregon Health and Sciences University, Portland, OR, USAAbstract: Necrotizing enterocolitis is the most common gastrointestinal emergency in preterm neonates and a major cause of morbidity and mortality in premature infants born before 32 weeks of gestation or with a birth weight less than 1500 g. In this review, we discuss predisposing factors, clinical manifestations, and the quality of evidence for various preventive and therapeutic strategies.Keywords: necrotizing enterocolitis, inflammation, mucosa, pneumatosis, neonate
Content may be subject to copyright.
© 2011 Maheshwari et al, publisher and licensee Dove Medical Press Ltd. This is an Open Access article
which permits unrestricted noncommercial use, provided the original work is properly cited.
Research and Reports in Neonatology 2011:1 39–53
Research and Reports in Neonatology Dovepress
submit your manuscript | www.dovepress.com
Dovepress
39
REVIEW
open access to scientific and medical research
Open Access Full Text Article
http://dx.doi.org/10.2147/RRN.S23459
Neonatal necrotizing enterocolitis
Akhil Maheshwari
1
Laura L Corbin
2
Robert L Schelonka
2
1
Division of Neonatology and
Center for Neonatal and Pediatric
Gastrointestinal Disease, Department
of Pediatrics, University of Illinois at
Chicago, IL,
2
Division of Neonatology,
Department of Pediatrics, Oregon
Health and Sciences University,
Portland, OR, USA
Correspondence: Akhil Maheshwari
Center for Neonatal and Pediatric
Gastrointestinal Disease, Children’s
Hospital of University of Illinois,
University of Illinois at Chicago,
840 S Wood St, CSB 1257, UIC
m/c 856, Chicago, IL 60612, USA
Tel +1 312 996 4185
Fax +1 312 355 5548
Email akhil1@uic.edu
Abstract: Necrotizing enterocolitis is the most common gastrointestinal emergency in
preterm neonates and a major cause of morbidity and mortality in premature infants born
before 32 weeks of gestation or with a birth weight less than 1500 g. In this review, we discuss
predisposing factors, clinical manifestations, and the quality of evidence for various preventive
and therapeutic strategies.
Keywords: necrotizing enterocolitis, inflammation, mucosa, pneumatosis, neonate
Introduction
Necrotizing enterocolitis (NEC), an inflammatory bowel necrosis of infants,
1,2
is the
most common gastrointestinal emergency in preterm neonates and a major cause of
morbidity and mortality in neonatal intensive care units throughout the world.
3,4
In
this review, we discuss pathophysiological factors that may predispose the developing
intestine to NEC, describe the clinical manifestations of this disease, and provide a
critical appraisal of therapeutic strategies.
Incidence and epidemiology
The incidence of NEC is estimated to be 1–3 per 1000 live births, with more than 90%
of all cases occurring in preterm infants.
1
NEC occurs in 4%–11% of all premature
infants born with very low birth weight (,1500 g), and the frequency in this subgroup
is also inversely related to birth weight and gestational age.
5,6
In the National Institute
of Child Health and Development cohort at neonatal research network centers, NEC
was recorded in 11.5%, 9%, 6%, and 4% of infants weighing 401–750 g, 751–1000 g,
1001–1250 g, and 1251–1500 g, respectively.
7
The incidence of NEC varies significantly between neonatal intensive care units.
8–11
Cases occur in each individual neonatal intensive care unit at an “endemic” rate specific
for that unit, which may show some seasonal fluctuation and may be punctuated by
minor epidemics.
7,12–15
Although the reasons for these center differences are unclear,
plausible explanation(s) include biological differences in patient populations and
distribution of birth weights, infectious milieu in the neonatal intensive care units,
and consistency in labeling of cases that recover without requiring significant medical
or surgical intervention.
15
Despite improvements in neonatal intensive care and increased overall survival of
critically ill premature neonates, mortality rates from NEC can reach 50%.
5,16,17
Most
Number of times this article has been viewed
This article was published in the following Dove Press journal:
Research and Reports in Neonatology
23 August 2011
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
40
Maheshwari et al
deaths occur in extremely low birth weight infants, who
frequently develop severe disease and require surgery.
5
Pathophysiology
Although the etiopathogenesis of NEC remains unclear,
current epidemiological and experimental evidence
18,19
identifies several diverse risk factors and supports a multi-
factorial model of disease (summarized in Figure 1).
Prematurity is the most important predictor of NEC.
Immaturity of the gastrointestinal tract, particularly in
the context of its motility, digestion, perfusion, barrier
function, and immune defense, is a major predisposing
factor for NEC.
20–22
The pathophysiological importance of
prematurity is evident from the near exclusive occurrence
of NEC in preterm infants, even though events generally
considered to be critical in the pathogenesis of NEC, such
as gut mucosal injury, altered barrier function, and bacterial
translocation, are recorded frequently in critically ill patients
of all ages.
23–25
Evidence for genetic predisposition to NEC is modest.
Bhandari et al
26
recorded NEC in one or both twins in nine of
63 (14%) pairs of monozygotic twins and in 29 of 189 (15%)
pairs of dizygotic twins. After controlling for covariates,
genetic factors did not account for any variance in liability
for NEC. NEC has been associated with single nucleotide
polymorphisms in the interleukin (IL)-4 receptor (+1902G,
protective),
27
IL-18 (-607A, increased severity),
28
vascular
endothelial growth factor (+450C, increased risk),
29
and the
carbamoyl-phosphate synthetase 1 genes (T450N, increased
risk).
30
In contrast, NEC is not associated with most single
nucleotide polymorphisms that have been linked with
Crohn’s disease and/or ulcerative colitis, such as those in the
genetic sequences of tumor necrosis factor-alpha (TNF-α),
IL-1, IL-4, IL-6, IL-8, and IL-10, CD14, toll-like receptor
4, caspase-recruitment domain 15, and nucleotide-binding
oligomerization domain containing 2.
31–33
NEC usually occurs in infants who are receiving enteral
feedings. Although NEC can occur in neonates who have
never been fed, 90%–95% of cases occur in infants with
a history of recent volume advancement or reinitiation
of enteral feedings.
34,35
Besides the risk of direct osmotic
injury to the gut mucosa, feedings may also alter splanchnic
blood flow and increase the risk of ischemic injury in
underperfused regions by increasing local oxygen needs.
In addition, immaturity of motility and digestion in the
developing intestine may leave undigested food in the lumen
for prolonged periods, promoting bacterial overgrowth and
translocation.
36
Products of bacterial fermentation, such as
short chain fatty acids, can also injure the immature gut
mucosa.
37,38
Infants receiving formula feedings are at increased
risk of NEC compared with exclusively breastfed
neonates.
39–46
Formula lacks both cellular as well as soluble
immunoprotective factors, such as IgA and various natural
antimicrobials, and also has a propensity to alter the normal
postnatal gut bacterial colonization.
47–49
Recent studies
indicate that formula feeding in newborn animals may
directly induce inflammatory changes in the gut mucosa.
50
Host susceptibility
Prematurity, immature barrier
function, male gender, ethnic
predisposition
Bacterial flora
Luminal bacteria, endotoxins
altered bacterial colonization
Enteral feedings
Hypertonic feeds/meds,
H
2
-blockers, malabsorption,
H
2
productiion, endotoxins,
volatile organic acids
Gut mucosal injury
Inflammatory response
Inflammatory cells, PAF, TNFα,
leukotrienes, interleukins
single nucleotide polymorphisms in
IL4R, IL18, VEGF
Ischemia
Placental insufficiency,
maternal cocaine, prenatal indocin,
polycythemia, hypoxic/ischemic injury,
cyanotic heart disease, anemia/blood
transfusions, single nucleotide
polymorphisms in CPS1
Figure 1 Current epidemiological and experimental information on necrotizing enterocolitis supports a multifactorial model of disease. Clinical and histopathological features
indicate that tissue ischemia, bacterial ora, a dysregulated inammatory response, and enteral feedings may contribute to the pathogenesis of necrotizing enterocolitis in
premature infants.
Abbreviations: IL, interleukin; VEGF, vascular endothelial growth factor; PAF, plasminogen activating factor; TNFα, tumor necrosis factor alpha.
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
41
Neonatal necrotizing enterocolitis
In spite of a large body of data from physiological and
retrospective studies, a direct association between specific
feeding regimens and/or the rapidity of advancement of feed
volumes and NEC has not been conclusively proven.
45,51
Several observational studies have suggested that delaying
the introduction of enteral feeds beyond the first few days
after birth, and using standardized regimes to increase the
volume of feeds by less than about 24 mL/kg body weight
each day may be associated with a lower risk of NEC.
35,52–56
In the National Institute of Child Health and Development
neonatal research network, the incidence of NEC was
higher at centers where enteral feeding was introduced
earlier and feeding volumes advanced rapidly.
57
In a recent
retrospective study from a multihospital system,
58
fulminant
NEC characterized by massive bowel necrosis and rapid
progression to death within 48 hours was associated with
advancement of feedings by more than 20 mL/kg/day and/or
an increase in concentration of human milk fortifier within
48 hours before developing NEC. However, the association
between aggressive enteral feeding and NEC has not been
evident in randomized controlled trials comparing slow
versus rapid advancement of feedings.
51,59–61
A meta-analysis
based on these studies showed that cautiously advanced
enteral feedings are not only safe, but may also reduce
other morbidities associated with prematurity.
62
Similarly,
current trial data do not provide evidence that delayed
introduction of progressive enteral feeds reduces the risk
of NEC in very low birth weight infants. In a meta-analysis
of five randomized controlled trials (600 infants, delayed
introduction of feedings defined as later than 5–7 days after
birth, and early introduction as less than 4 days after birth),
delayed introduction of feedings did not reduce the risk
of NEC (relative risk [RR] 0.89, 95% confidence interval
[CI] 0.58–1.37) or all-cause mortality (RR 0.93, 95% CI
0.53–1.64). Infants who had delayed introduction of enteral
feeds took significantly longer to establish full enteral feeding
(reported median difference 3 days).
Mucosal injury may be an early event. Gut epithelial
injury is believed to be an early event in NEC. Although the
causes of this initial epithelial injury remain unclear, primary
apoptotic and autophagic mechanisms have been invoked.
63,64
This disruption of the epithelial barrier is presumed to
allow bacterial translocation, which in turn triggers a local
inflammatory response.
63,65,66
Ischemia may play a role in NEC. Coagulative necrosis,
which is typically associated with ischemia, is a prominent
histopathological finding in NEC.
67–69
The predilection
for the ileocecal region, a watershed area supplied by
end-arteries,
70
also indicates that ischemia may be an
important pathophysiological event in NEC. Infants with
NEC may have decreased endothelial nitric oxide synthase
activity and decreased arteriolar nitric oxide production,
which can place the developing intestine at a higher risk of
ischemic injury.
71
However, this association between hypoxia-
ischemia and NEC has not been clearly established in
clinical studies in preterm infants.
72
Although minor transient
episodes of hypoxia and/or hypotension are not uncommon
in premature neonates, major ischemic events are obvious in
only a minority of preterm infants with NEC,
6,9,72,73
and tend
to occur early in the neonatal period rather than in postnatal
weeks 2–4 when NEC occurs.
73,74
In full-term infants, NEC tends to occur at an earlier
postnatal age than in preterm infants, and is more obviously
associated with factors that may conceivably cause splanchnic
hypoperfusion. Many term or near-term infants with NEC
have a history of placental insufficiency and absence/reversal
of end-diastolic blood flow in the umbilical vessels in utero,
perinatal asphyxia, polycythemia, episodes of low cardiac
output or clinical shock, and congenital cyanotic heart
disease.
80,83,84
NEC is characterized by a severe, unregulated inflam-
matory response. It is characterized by a prominent leu-
kocyte infiltrate comprised of activated macrophages and
neutrophils.
63,65,75
Human and animal studies have demon-
strated increased tissue expression of TNFα and platelet acti-
vating factor (PAF), which may propagate ongoing mucosal
injury by triggering a cascade of inflammatory mediators,
including IL-1, IL-6, IL-8, IL-10, IL-12, and IL-18.
36,76–81
Activation of the complement and coagulation cascades,
cytokines, reactive oxygen species, and nitric oxide further
amplify the mucosal injury.
36
In infants with NEC, increased
expression of PAF may be coupled with reduced levels of
PAF acetylhydrolase (the enzyme which degrades PAF),
further augmenting its local inflammatory effects.
36,77,82,83
In
experimental animals, attempts to regulate the inflammatory
response by depletion of neutrophils or by using anti-TNFα
antibodies have successfully reduced the severity of tissue
damage.
84,85
Bacteria play an essential role in the pathogenesis of
NEC. Several lines of evidence emphasize the importance of
bacterial flora in the pathogenesis of NEC: NEC occurs only
after postnatal bacterial colonization of the gastrointestinal
tract; intestinal injury prior to colonization may cause
strictures or atresia, but not NEC;
86
pneumatosis intestinalis,
the pathognomonic finding in NEC, reflects the entrapment
of the gaseous products of bacterial fermentation in affected
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
42
Maheshwari et al
tissues;
87
enterally administered aminoglycosides can reduce
the incidence of NEC;
88
and NEC-like lesions do not develop
in germ-free animals.
16,89
Bacteria play a key role in NEC
by activating the immune system in the mucosa and causing
inflammatory injury.
66
Bacterial products such as short chain
fatty acids (acetate, butyrate) can also directly damage the
epithelial barrier.
38,90
Cases of NEC are often temporally and spatially
clustered in neonatal intensive care units, suggesting that
NEC may be caused by a transmissible agent.
91
However,
most studies, whether based on culture techniques or on
polymerase chain reaction amplification of 16S ribosomal
RNA,
92,93
have failed to consistently implicate a single agent.
Cultures of blood and other sterile fluids from infants with
NEC usually yield microorganisms that typically colonize
critically ill preterm infants and the neonatal intensive care
unit microenvironment. Because these microorganisms are
not unique to neonatal intensive care units, the interaction of
bacteria and bacterial products with the immature intestine
tends to receive greater emphasis in the pathogenesis of NEC
than the presence of specific bacterial pathogens.
65
However,
some recent studies suggest that early duodenal colonization
with specific Enterobacteriaceae and Clostridia may predict
later development of NEC.
93,94
Similarly, in a polymerase
chain reaction-based comparison of fecal microbiota from
preterm infants with and without NEC, Wang et al
95
found a
marked reduction in bacterial diversity and also an abnormal
pattern of bacterial colonization, with a relative abundance
of gammaproteobacteria (which include Enterobacteriaceae
and Pseudomonadaceae) and reduced numbers of Firmicutes.
The oligoclonality of gut microbiota and the disproportionate
representation of Gram-negative bacilli may be related to
administration of broad-spectrum antibiotics, delayed or
interrupted feedings, and exposure to selected multidrug-
resistant nursery flora.
96,97
Delayed or altered acquisition of
intestinal microbiota by early prolonged antibiotic treatment
increases the risk for NEC. This was shown in a recent
cohort analysis of 4039 extremely low birth weight infants.
Infants who received at least 4 days of initial empirical
antibiotic treatment, with sterile body fluid cultures, had an
increased risk of NEC with an odds ratio [OR] of 1.34 (95%
CI 1.04–1.73).
98
The association of specific bacterial groups
with NEC is intriguing and merits further evaluation.
Immaturity of intestinal barrier function may promote
bacterial translocation and increase the risk of NEC. Tight
junctions and the glycoprotein mucin layer secreted by goblet
cells comprise the structural component of the intestinal
barrier, whereas IgA, lysozyme, phospholipase A2, and
antimicrobial peptides, such as defensins and cathelicidins,
are components of the biochemical barrier.
Immaturity of Paneth cells, specialized crypt cells that
produce natural antimicrobials (such as enteric human
defensins 5 and 6) and MD2 (a key component of the
lipopolysaccharide receptor complex), has been suggested to
contribute to the risk of NEC.
99–102
Acute NEC is associated
with a low number of Paneth cells, which show weak
immunoreactivity or complete absence of lysozyme.
103,104
During recovery from NEC, Paneth cell hyperplasia/
metaplasia has been observed with increased expression of
enteric defensins.
99,101
Secretory IgA (sIgA) antibodies are an important host
defense mechanism, preventing luminal antigens and
microorganisms from entering the mucosa. In an adult human
subject, 70%–80% of all Ig-producing cells in the body are
located in the intestinal mucosa and most of these cells produce
IgA.
105,106
In contrast, neonates lack IgA immunocytes at birth
and the first sIgA may not appear in mucosal secretions until
sometime between postnatal week 2 and 8.
107
This deficiency
can be partially offset in breastfed infants by sIgA present in
colostrum/milk;
108
breastfed infants receive about 0.5–1.0 g/
day of antibodies in milk throughout lactation, which is
comparable with the 2.5 g daily production of antibodies by a
65 kg adult, and a source of passive immunity against antigens
“seen” by the mother-infant dyad.
109
In formula-fed premature
infants, the absence of milk-borne sIgA is a significant
immunological disadvantage and a likely contributor to the
increased risk of NEC.
46
Compared with term infants, premature neonates have
increased gut mucosal permeability, and this permeability
is even greater in infants who subsequently develop NEC.
110
Studies on human tissue samples and in rodent models show
that the intestinal epithelium is breached early during NEC
through apoptosis or necrosis.
63
In this process, excessive
nitric oxide production, either directly or through its
reactive nitrogen derivative, peroxynitrite, may accentuate
epithelial injury through membrane oxidation, induction
of apoptosis, and direct mitochondrial damage.
111–113
In
preterm infants, a deficiency in the mucus layer may promote
bacterial adherence and increase gut mucosal permeability,
predisposing to mucosal injury.
114
The developmental
deficiency of epithelial trophic factors, such as the epidermal
growth factor and heparin-binding epidermal growth factor-
like growth factor, may further increase the risk of injury to
the epithelial barrier.
115–117
Do red blood cell transfusions predispose to NEC? In
convalescing preterm infants, red blood cell transfusions have
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
43
Neonatal necrotizing enterocolitis
been temporally associated with NEC.
118,119
Red blood cell
transfusions can dampen the normal postprandial increase in
mesenteric blood flow in premature infants, particularly in those
with a birth weight ,1250 g.
120
Immaturity of vascular auto-
regulation in extremely preterm infants is linked to defects in
endothelial nitric oxide synthesis
68
and could plausibly explain
a higher risk of mucosal injury following transfusions.
Pathology
The disease is commonly localized to the ileocolic region,
although the colon may be frequently involved in term
infants.
121
Some infants with severe, aggressive disease may
develop total gut necrosis (NEC totalis). The four major histo-
pathological findings in NEC are coagulative necrosis, bacte-
rial overgrowth, pneumatosis intestinalis, and inflammation.
Clinical features
The presenting signs of NEC are protean and may be insidious
in onset or sudden and catastrophic. NEC typically presents
at 2–4 weeks after birth, although the onset may be as late
as 3 months in some infants.
16,72,122
The age of onset of NEC
correlates inversely with gestational age at birth in a nonlinear
(log-normal) relationship, where infants born at ,28 weeks’
gestation tend to develop NEC at a disproportionately greater
postnatal age than their more mature counterparts.
123,124
NEC often presents with nonspecific systemic signs, such
as tachycardia, apnea, lethargy, and temperature instability.
Gastrointestinal signs may include increased prefeed residuals
or delayed gastric emptying, emesis, abdominal distention,
tenderness, and/or ileus with hypoactive bowel sounds. Grossly
bloody stools are seen in approximately 25% of infants.
Clinical progression of NEC is commonly staged using the
modified Bell’s criteria (Table 1).
125
Characteristically, NEC
follows an initial early stage of systemic inflammatory
response, followed by a definite stage of localized peritonitis,
and finally, an advanced stage of generalized peritonitis.
In a recent study, Clark et al
126
described the clinical
characteristics of infants who died of NEC. Compared with
5594 infants who recovered from NEC and were discharged
home, there were 1505 infants diagnosed with NEC who
died. In multivariate analysis, lower gestational age, lower
birth weight, treatment with assisted ventilation on the day
of diagnosis of NEC, treatment with vasopressors at the
time of diagnosis, and African-American ethnicity were
associated with mortality. In another study,
58
fulminant
NEC, characterized by massive bowel necrosis and rapid
progression to death within 48 hours, was recorded in
7%–10% of all cases and was associated with lower birth
weight (1088 ± 545 g versus 1652 ± 817 g), earlier gestational
age (27.5 ± 3.3 weeks versus 31.1 ± 4.4 weeks), radiographic
evidence of portal venous air, hematocrit ,22%, a history
of advancement in feeding volume .20 mL/kg/day, an
immature to total neutrophil ratio .0.5, blood lymphocyte
count ,4000/µL, and a history of increased concentration of
Table 1 Modied Bell’s staging criteria for necrotizing enterocolitis
125
Stage Classication System signs Intestinal signs Radiological signs
IA Suspected NEC Temperature instability, apnoea,
bradycardia, lethargy
Increased pregavage residuals,
mild abdominal distention,
emesis, guaiac-positive stool
Normal or intestinal
dilation, mild ileus
IB Suspected NEC Same as above Bright-red blood from rectum Same as above
IIA Proven NEC – mildly ill Same as above Same as above, plus absent bowel
sounds, with or without
abdominal tenderness
Intestinal dilation, ileus,
pneumatosis intestinalis
IIB Proven NEC – moderately ill Same as above, plus mild
metabolic acidosis and mild
thrombocytopenia
Same as above, plus absent bowel
sounds, denite abdominal tenderness,
with or without abdominal cellulitis
or right lower quadrant mass
Same as IIA, plus
portal vein gas, with
or without ascites
IIIA Advanced NEC – severely ill,
bowel intact
Same as lllB, plus hypotension
bradycardia, severe apnoea,
combined respiratory and
metabolic acidosis, disseminated
intravascular coagulation,
and neutropenia
Same as above, plus signs of
generalized peritonitis, marked
tenderness, and distention of
abdomen
Same as IIB, plus
denite ascites
IIIB Advanced NEC – severely ill,
bowel perforated
Same as IIIA Same as IIIA Same as IIB, plus
pneumoperitoneum
Note: This table was published in Pediatric Clinics of North America, Vol 33, MC Walsh, RM Kliegman, Necrotizing enterocolitis: Treatment based on staging criteria, Pages
179–201, © Copyright Elsevier 1986.
Abbreviation: NEC, necrotizing enterocolitis.
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
44
Maheshwari et al
human milk fortifier within 48 hours before developing NEC.
In another study of NEC totalis,
127
breast milk feeding was
noted to have a protective effect.
NEC in full-term infants differs from that seen in pre-
mature infants. Approximately 10% of infants with NEC
are born at term. Unlike preterm infants who develop NEC
in the second or third week of life (median 12 days), most
term cases are seen within the first week (median 2 days) and
often have colonic involvement.
121,128,129
NEC in term infants
is usually secondary, associated with conditions such as birth
asphyxia, polycythemia, congenital heart disease, rotavirus
infections, and Hirschsprung’s disease.
121,128–134
Outcomes
are generally better than in preterm neonates, with mortality
rates of 0%–13%.
121,135,136
Diagnosis
A high index of suspicion in diagnosing at-risk infants is
crucial. Most clinical antecedents prior to Bell stage III
NEC are nonspecific for gastrointestinal pathology and
may not provide sufficient time to the clinician for early
institution of treatment measures. In a recent retrospective
study, Christensen et al
137
reviewed the medical records of
118 infants with stage III NEC. The earliest recognized
antecedents of NEC were nonspecific, including apnea/
bradycardia, skin mottling, and irritability, which were first
noted at a mean of 2.8 ± 2.1, 4.5 ± 3.1, and 5.4 ± 3.7 hours,
respectively, prior to the diagnosis of NEC. The most
frequently identified gastrointestinal antecedents were
blood in the stools, increased abdominal girth, and elevated
prefeeding gastric residuals or emesis, identified 2.0 ± 1.9,
2.8 ± 3.1, and 4.9 ± 4.0 hours before NEC was recognized.
No consistent laboratory antecedents were discovered.
Radiographic features remain the mainstay of definitive
diagnosis. The pathognomonic sign for NEC is pneumatosis
intestinalis (Figure 2).
138
These radiolucent shadows have a
bubbly appearance when air is submucosal and become linear
when subserosal. Portal venous gas has been associated with
a poor prognosis, although this association has recently been
questioned.
139
Sonographic appearance of portal air is an early
sign. In a prospective cohort study, sonographic portal air
had a specificity of 86% for advanced NEC ($stage II), and
the sensitivity was lower at 45%.
140
It is not known if portal
venous gas visualized by ultrasound or on plain radiographs
has the same prognostic significance. Sonographic detection
of echoic free fluid and bowel wall thinning may also be more
sensitive for intestinal perforation than plain radiography.
141
Serial radiographs are invaluable in following the progression
of NEC, particularly in the first 48 hours after onset of disease.
Although intestinal perforation may occur within a few hours
to as late as 8 days following the onset of NEC,
142
more than
two-thirds of all perforations occur within 30–48 hours.
143
In some infants who present with bloody stools but minimal
systemic signs, pneumatosis may be limited to the colon and
may indicate a relatively benign course.
144
Most patients with NEC develop leukocytosis and
neutrophilia, although neutropenia can occur in advanced
disease due to the migration of neutrophils into the peritoneal
cavity.
145
Blood cultures may grow organisms typically
associated with late-onset sepsis. Thrombocytopenia may
occur in stage II and III, and patients with advanced NEC
may have evidence of disseminated intravascular coagulation.
Breath hydrogen testing was initially heralded as a diagnostic
tool, but subsequent studies have shown it as lacking
discriminant value.
146
The differential diagnosis of NEC includes infections
(systemic or intestinal), gastrointestinal obstruction, volvulus,
and isolated intestinal perforation. Idiopathic focal intestinal
perforations can occur spontaneously or in association with
deficiency of the muscularis propria,
147
early use of postnatal
corticosteroids alone
148
or with indomethacin,
149
and with
occult candidal infections.
150
Pneumoperitoneum develops
in such patients, but they are usually less ill than those with
NEC. Some experts believe that isolated perforations may
not be related to NEC.
AB
CD
Figure 2 Abdominal radiographs with characteristic ndings of necrotizing
enterocolitis. (A) Diffuse pneumatosis intestinalis with a linear distribution in the
left upper and middle quadrants. (B) Characteristic arborization pattern of portal
venous gas. (C) Free peritoneal gas with the falciform ligament visible (arrow).
(D) Free peritoneal gas seen in a left lateral decubitus view.
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
45
Neonatal necrotizing enterocolitis
Treatment
Medical management
Rapid initiation of therapy is necessary for suspected as well
as proven cases of NEC. There is no definitive treatment
for established NEC, and therefore treatment is directed at
supportive care and prevention of further injury with cessation
of feeding, nasogastric decompression, and administration of
intravenous fluids (see Table 2). Infants are usually made nil
per os for a variable period of time, depending on the severity
of disease. Parenteral antibiotics are widely used for the
Table 2 Treatment and prevention of necrotizing enterocolitis
Therapeutic intervention Current status Evidence level Recommendation level
Treatment
Gastric/Intestinal
decompression and bowel rest
Provide supportive care and prevent further injury with cessation
of feeding, nasogastric decompression, and administration of
intravenous uids. Infants stay nil per os for 3–5 days in stage I,
and 10–14 days in stages II and III.
III B
Parenteral antibiotics Broad spectrum antibiotics should be administered based on
local antibiotic sensitivity patterns. Anaerobic coverage should
be considered in infants with stage III NEC.
II-3 C
Primary peritoneal drainage
versus exploratory laparotomy
Choices for surgical management in infants with NEC include
peritoneal drain placement and exploratory laparotomy.
In unstable premature infants with perforated NEC, peritoneal
drainage can be cautiously considered as an alternative to
exploratory laparotomy, although the best surgical approach
in these infants remains unresolved.
I C
Prevention
Antenatal corticosteroids Small benecial effect of antenatal steroids for reducing risk of NEC. I A
Minimal enteral (trophic)
feedings
Infants receiving trophic feedings take less time to tolerate full
enteral feeds and have a shorter duration of hospital stay,
without an effect on the incidence of necrotizing enterocolitis.
I C
Slow advancement of feedings No evidence to suggest that slow advancement of enteral feed
volumes reduces the risk of NEC in very low birth weight infants.
I D
Breast milk Although the mechanism of protection is not completely
understood, there is strong evidence favoring the use of human
milk to reduce the risk of NEC in premature infants.
II-2 A
Oral immunoglobulins Data from available trials do not support oral administration of
immunoglobulin for the prevention of NEC.
I D
Enteral antibiotics Enteral antibiotic treatment leads to a small reduction in
NEC risk; however, increase in antimicrobial-resistant intestinal
microbiota precludes routine use of this therapy.
I D
Amino acid supplementation Data are insufcient at present to support supplemental
administration of parenteral L-arginine or glutamine to reduce
the risk of NEC.
I C
Recombinant cytokines and
growth factors
Epidermal growth factor is a promising agent in preclinical studies.
In early clinical studies, enteral administration of a synthetic
amniotic uid-like solution containing erythropoietin and
granulocyte-colony stimulating factor has shown an encouraging
safety and efcacy prole.
III I
Probiotics Probiotics may reduce the risk of severe NEC and related
mortality; however, important questions remain regarding
optimal choice of agent(s) and dose.
I C
Prebiotics Recent nonhuman animal experimental data suggest that
oligofructose prebiotics may be useful in protecting against
experimental NEC.
NA* NA*
Notes: *Insufcient human data to determine evidence level or recommendation. Levels of evidence: I, Evidence obtained from at least one properly designed randomized,
controlled trial; II, evidence obtained from well-designed controlled trials without randomization (II 1), cohort or case-control analytic studies, (II 2) evidence obtained
from multiple time series with or without the intervention (II 3); III, opinions of respected authorities based on clinical experience, descriptive studies, or reports of expert
committees. Levels of recommendations for clinical use: A, Good scientic evidence suggests that the benets substantially outweigh the potential risks; B, at least fair
scientic evidence suggests that the benets outweigh the potential risks; C, at least fair scientic evidence suggests that there are benets provided, but the balance between
benets and risks are too close for making general recommendations; D, at least fair scientic evidence suggests that the risks outweigh potential benets; I, scientic evidence
is lacking, of poor quality, or conicting, such that the risk–benet balance cannot be assessed.
Abbreviation: NEC, necrotizing enterocolitis.
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
46
Maheshwari et al
treatment of NEC, but there is surprisingly sparse evidence
guiding the choice of antimicrobial agent and duration of
therapy. One study comparing alternative treatment regimens
that included 90 infants with definite NEC, treated 46 cases
with ampicillin and gentamicin, while 44 cases received
cefotaxime and vancomycin. Infants $2200 g birthweight
had similar outcomes with either regimen. Smaller infants
given cefotaxime and vancomycin had a lower risk of
culture-positive peritonitis (P = 0.01), and were less likely
to die (P = 0.048) or develop thrombocytopenia (P = 0.004).
These data suggest that carefully chosen antibiotic regimens
can improve the outcome of NEC.
151
Antibiotic coverage
for anaerobes should be considered for infants with stage
III NEC.
Surgical management
Approximately 20%–40% of patients with pneumatosis
intestinalis will require surgical management. Indications
for surgery include evidence of perforation seen on
abdominal radiographs or positive abdominal paracentesis
(stool or organism on Gram stain from peritoneal uid).
Failure of medical management, a single fixed bowel loop
on radiographs, abdominal wall erythema, or a palpable
mass are all relative indications for surgery. In rare cases,
the entire intestine can be involved, precluding surgical
intervention. Ideally, surgery should be performed after the
development of bowel necrosis, but before perforation and
peritonitis occurs.
In unstable premature infants with perforated NEC,
peritoneal drainage can be cautiously considered as an
alternative to exploratory laparotomy, although the best
surgical approach in these infants remains unresolved. In
the NECSTEPS trial,
152
there was no statistically significant
difference in 90-day survival, dependence on parenteral
nutrition, or length of hospital stay in 117 very low birth
weight infants randomly assigned to peritoneal drainage or
laparotomy. However, other studies have raised important
concerns about the routine use of peritoneal drainage. In
the NET trial,
153
69 extremely low birth weight patients
were randomized to peritoneal drainage or laparotomy,
and no signicant differences were noted in survival,
length of hospital stay, ventilator dependence, or need
for parenteral nutrition. However, peritoneal drainage
was effective as a definitive treatment in only 4/35 (11%)
surviving neonates, and the rest either required a delayed
laparotomy (26/34, 74%) or died. In a recent meta-analysis,
Rao et al
154
reviewed data from the NET and NECSTEPS
trials and detected no significant differences in mortality
within 28 days of peritoneal drainage or laparotomy
(28/90 versus 30/95; typical RR 0.99, 95% CI 0.64–1.52;
n = 185), mortality by 90 days after the primary procedure
(typical RR 1.05, 95% CI 0.71–1.55; n = 185) and the
number of infants needing total parenteral nutrition for
more than 90 days (typical RR 1.18, 95% CI 0.72–1.95;
n = 116). Nearly 50% of infants in the peritoneal drainage
group could avoid the need for laparotomy during the
study period (44/90 versus 95/96; typical RR 0.49, 95%
CI 0.39–0.61; n = 186). One study found that the time to
attain full enteral feeds in infants #1000 g was prolonged
in the peritoneal drainage group (mean difference 20.77,
95% CI 3.62–37.92).
Although the immediate outcome following peritoneal
drainage or laparotomy appears to be similar, there are
concerns about the risk of neurodevelopmental impairment
following peritoneal drainage. In a recent multicenter trial,
peritoneal drainage was associated with increased risk
of death or neurodevelopmental impairment.
155
A meta-
analysis of three prospective observational studies and two
randomized controlled trials suggested a significant excess
mortality of 55% associated with peritoneal drainage.
156
There is a need for better identification of patients who are
less likely to tolerate laparotomy and who may benefit from
peritoneal drainage as a temporizing strategy.
Prevention
The existing evidence shows a small beneficial effect of
antenatal steroids in reducing the risk of NEC (see Table 2).
157
This may be accomplished by accelerating maturation of the
gut epithelial barrier and by reducing the overall severity
of illness via prevention of lung disease. When analyzed
together, eight randomized controlled comparisons of ante-
natal corticosteroid administration with placebo or with no
treatment, including 1675 infants, showed a risk reduction
in NEC of 0.46 (95% CI 0.29–0.74).
157
Multiple courses of
antenatal steroids do not appear to reduce the risk of NEC
further. In a randomized trial of one to four weekly treat-
ments of antenatal steroids or placebo that included 1858
pregnant women, and the outcomes of 2304 infants, the rate
of NEC, ie, 1%, was similar in both groups.
158
Minimal enteral (trophic) feedings
Initiating feeds by using small amounts of milk or formula
may promote the maturation of peristaltic activity and
enzymatic systems, release of digestive hormones, and
augment intestinal blood flow.
3,43,159–162
However, in a
meta-analysis of nine randomized trials including 754
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
47
Neonatal necrotizing enterocolitis
very low birth weight infants,
163
early trophic feedings did
not affect feed tolerance, growth rates, or the risk of NEC
(RR 1.07, 95% CI 0.67–1.70; risk difference 0.01, 95%
CI -0.04–0.05).
Slow advancement of feedings
Meta-analyses of three randomized controlled trials in which
a total of 396 infants were included found no significant
effects of feeding advancement on the risk of NEC (RR 0.96,
95% CI 0.48–1.92) or all-cause mortality (RR 1.40, 95%
CI 0.71–2.80).
45
Infants who had slow rates of feed volume
advancement took longer to regain birth weight (median
difference 2–5 days) and to establish full enteral feeding
(median difference 3–5 days). No statistically significant
effect on total duration of hospital stay was detected. The
currently available data do not provide evidence that slow
advancement of enteral feed volumes reduces the risk of
NEC in very low birth weight infants. Of note, few partici-
pants were extremely low birth weight or growth restricted,
so conclusions about infants at greatest risk for NEC cannot
be drawn from the available data.
Breast milk
Experimental and clinical studies show a protective effect
of human milk feeds against NEC when compared with
formula.
39–44,48,164,165
The protective effects of breast milk
against NEC are retained even in pasteurized, banked
donor milk. In meta-analysis of data from five randomized
trials,
46
formula-fed infants were at higher risk of NEC than
infants who received donor milk (RR 2.5, 95% CI 1.2–5.1;
risk difference 0.03, 95% CI 0.01–0.06; number needed to
harm 33, 95% CI 17–100). More recently, Sullivan et al
166
showed that an exclusively human milk-based diet protected
extremely premature infants against NEC and surgical
NEC when compared with a mother’s milk-based diet that
included bovine milk-derived human milk fortifier and
preterm formula.
Oral immunoglobulins
Three trials, including a total of 2095 neonates, were reviewed
together. Oral administration of IgG or an IgG/IgA combina-
tion did not result in a significant reduction in incidence of
definite NEC (RR 0.84, 95% CI 0.57–1.25), suspected NEC
(RR 0.84, 95% CI 0.49–1.46), need for surgery (RR 0.21,
95% CI 0.02–1.75), or death from NEC (RR 1.10, 95% CI
0.47–2.59).
49
Based on the available trials, the evidence does
not support the administration of oral immunoglobulin for
the prevention of NEC.
Enteral antibiotics
To determine the effect of enteral antibiotic prophylaxis and
subsequent development of NEC, five randomized controlled
trials involving 456 infants were compared. Enteral antibiotic
administration resulted in a significant risk reduction for NEC
(RR 0.47, CI 0.28–0.78; risk difference -0.10, 0.16 to -0.04);
number needed to treat 10 [6–25]). There was a statisti-
cally significant reduction in NEC-related deaths (RR 0.32,
0.10–0.96; risk difference -0.07, CI -0.13–0.01) and number
needed to treat of 14 (8–100). However, concerns about the
development of resistant bacteria remain, and meta-analysis
revealed a borderline increase in antimicrobial-resistant intes-
tinal microbiota with enteral antibiotic treatment.
88
Amino acid supplementation
Nitric oxide augments gastrointestinal perfusion, barrier
function, and mucosal repair.
167
The supplementation of
L-arginine, a major substrate for nitric oxide production,
appears promising in small cohorts in reducing NEC but
the data are insufficient at present to support a practice
recommendation.
168–170
Similarly, glutamine promotes gut
epithelial proliferation and barrier function in animal stud-
ies, but a larger multicenter trial of parenteral glutamine
supplementation did not show a beneficial effect in reducing
the incidence of NEC in preterm infants.
171
Probiotics
Probiotics are living microorganisms which, when ingested,
can exert a health benefit beyond basic nutrition. Probiotics
improve intestinal defense mechanisms, including mucosal
IgA secretion, intestinal epithelial cell proliferation, and
barrier function, decrease inflammation and epithelial cell
apoptosis,
172
and may be useful in preventing NEC.
173,174
Alfaleh et al
175
analyzed 16 eligible trials including
2842 infants.
174,176–190
Included trials were highly variable with
regard to enrollment criteria such as birth weight and gesta-
tional age, baseline risk of NEC in the control groups, timing,
dose, formulation of the probiotics, and feeding regimens.
Enteral probiotics significantly reduced the incidence of
severe NEC (stage II or more, typical RR 0.35, 95% CI 0.24–
0.52) and mortality (typical RR 0.40, 95% CI 0.27–0.60).
Deshpande et al
191
selected 11 of these trials
174,178–181,184–189
involving 2176 neonates and reported a similar reduction
in NEC. The frequency of NEC decreased from 6.56% (71
of 1082) in the control group to 2.37% (26 of 1094) in the
probiotics-treated group. Meta-analysis using a fixed-effects
model showed reduction in risk of NEC in the probiotics-
treated group (RR 0.35, 95% CI 0.23–0.55; P , 0.00001).
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
48
Maheshwari et al
Only four of these trials reported a significantly higher risk
for NEC in the control group.
174,179,180,189
The number needed
to treat with probiotics to prevent one case of NEC was 25
(95% CI 17–34). Current evidence indicates that enteral
supplementation with probiotics can prevent severe NEC
and decrease all-cause mortality in preterm infants. However,
further study is needed before routine supplementation using
infant formulas with probiotics and also to determine the
safety and efficacy of probiotic formulations in extremely
low birth weight infants.
175,192
Prebiotics
Prebiotics are nondigestible dietary supplements (usually
carbohydrates or mucins) which promote proliferation
of beneficial commensal bacteria like Lactobacillus and
Bifidobacterium. Recent experimental data suggest that
oligofructose prebiotics may be protective against NEC.
193–196
Recombinant cytokines
Epidermal growth factor, an important component of gut
secretions, human milk, and amniotic fluid, promotes
epithelial proliferation, migration, and mucosal repair
following injury.
197
Oral administration of recombinant
epidermal growth factor protects experimental animals against
NEC-like lesions.
198
Other studies have similarly evaluated the
role of hematopoietic growth factors, such as erythropoietin
and granulocyte-colony stimulating factor. We have recently
shown that transforming growth factor-β
2
may protect mouse
pups against NEC-like injury.
66
These cytokines are present in
amniotic fluid and human milk, are swallowed by the fetus in
large amounts,
199,200
and have a demonstrated an in vitro and
in vivo protective effect on gut mucosa.
201–206
Lactoferrin
As an addition to antibiotics, lactoferrin has been considered
by some to enhance the response of the immune system
when faced with sepsis. Although immune enhancement
may play a role in the treatment of NEC, current data do
not support the use of lactoferrin as a single agent at this
time. Manzoni et al
182
randomized 472 very low birth weight
infants to receive either lactoferrin alone or in combination
with Lactobacillus rhamnosus GG. Prophylaxis with oral
lactoferrin alone did not reduce the incidence of NEC
(RR 0.33, 95% CI 0.09–1.17; risk difference -0.04, 95%
CI -0.0–0.00), but a significant reduction in NEC was noted
when lactoferrin was combined with L. rhamnosus GG (RR
0.05, 95% CI 0.0–0.90; risk difference -0.06, 95% CI -0.10
to -0.02; number needed to treat 17, 95% CI 10–50).
Prognosis
Mortality rates range between 20% and 50%. Approximately
27%–63% of affected infants may require surgery,
1,16
and as many as 50% infants may die in the postoperative
period.
1,15,17
Subacute complications include strictures,
dysmotility, malabsorption, and short gut syndrome.
15
Severe NEC has been associated with growth delay
that can persist beyond infancy into childhood and poor
neurodevelopmental outcome at a corrected gestational age
of 18–22 months.
207
Summary
Despite advances in the diagnosis and management of many
neonatal diseases, NEC remains a devastating condition
for many infants. While it is established that very low
birth weight infants are at greatest risk for development of
NEC, human milk-feeding appears to be the single most
effective strategy to reduce, but not eliminate, this disease.
Current medical management of NEC is largely supportive
and likely does not modify the etiopathogenesis of the
disease. Controversies remain regarding optimal surgical
management for this condition. Although there are important
gaps in our understanding of NEC, future research should
focus on prevention of the disease and early recognition that
occurs well before the onset of intestinal necrosis.
Acknowledgment
AM is supported by a National Institutes of Health award.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Henry MC, Moss RL. Necrotizing enterocolitis. Annu Rev Med. 2009;60:
111–124.
2. Frost BL, Jilling T, Caplan MS. The importance of pro-inflammatory
signaling in neonatal necrotizing enterocolitis. Semin Perinatol.
2008;32(2):100–106.
3. Thompson AM, Bizzarro MJ. Necrotizing enterocolitis in newborns:
Pathogenesis, prevention and management. Drugs. 2008;68(9):
1227–1238.
4. Neu J, Walker WA. Necrotizing enterocolitis. N Engl J Med. 2011;364(3):
255–264.
5. Lee JS, Polin RA. Treatment and prevention of necrotizing enterocolitis.
Semin Neonatol. 2003;8(6):449–459.
6. Holman RC, Stoll BJ, Clarke MJ, Glass RI. The epidemiology of
necrotizing enterocolitis infant mortality in the United States. Am J
Public Health. 1997;87(12):2026–2031.
7. 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(2):e137–e142.
8. Horbar JD, Badger GJ, Carpenter JH, et al. Trends in mortality and
morbidity for very low birth weight infants, 1991–1999. Pediatrics.
2002;110(1 Pt 1):143–151.
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
49
Neonatal necrotizing enterocolitis
9. Lemons JA, Bauer CR, Oh W, et al. Very low birth weight outcomes of
the National Institute of Child health and human development neonatal
research network, January 1995 through December 1996. NICHD
Neonatal Research Network. Pediatrics. 2001;107(1):E1.
10. Wiedmeier SE, Henry E, Baer VL, et al. Center differences in NEC
within one health-care system may depend on feeding protocol. Am J
Perinatol. 2008;25(1):5–11.
11. Sankaran K, Puckett B, Lee DS, et al. Variations in incidence of
necrotizing enterocolitis in Canadian neonatal intensive care units.
J Pediatr Gastroenterol Nutr. 2004;39(4):366–372.
12. Snyder CL, Hall M, Sharma V, St Peter SD. Seasonal variation in the
incidence of necrotizing enterocolitis. Pediatr Surg Int. 2010;26(9):
895–898.
13. Guinan M, Schaberg D, Bruhn FW, Richardson CJ, Fox WW. Epidemic
occurrence of neonatal necrotizing enterocolitis. Am J Dis Child. 1979;
133(6):594–597.
14. Gerber AR, Hopkins RS, Lauer BA, Curry-Kane AG, Rotbart HA.
Increased risk of illness among nursery staff caring for neonates with
necrotizing enterocolitis. Pediatr Infect Dis. 1985;4(3):246–249.
15. Stoll BJ. Epidemiology of necrotizing enterocolitis. Clin Perinatol.
1994;21(2):205–218.
16. Lin PW, Stoll BJ. Necrotising enterocolitis. Lancet. 2006;368(9543):
1271–1283.
17. Blakely ML, Lally KP, McDonald S, et al. Postoperative outcomes of
extremely low birth-weight infants with necrotizing enterocolitis or
isolated intestinal perforation: A prospective cohort study by the NICHD
Neonatal Research Network. Ann Surg. 2005;241(6):984–989.
18. Harrell FE. Regression Modeling Strategies with Applications to Linear
Models, Logistic Regression, and Survival Analysis. New York, NY:
Springer-Verlag; 2001.
19. Taylor JM, Ankerst DP, Andridge RR. Validation of biomarker-based
risk prediction models. Clin Cancer Res. 2008;14(19):5977–5983.
20. Chandler JC, Hebra A. Necrotizing enterocolitis in infants with very
low birth weight. Semin Pediatr Surg. 2000;9(2):63–72.
21. Snyder CL, Gittes GK, Murphy JP, Sharp RJ, Ashcraft KW,
Amoury RA. Survival after necrotizing enterocolitis in infants weighing
less than 1,000 g: 25 years’ experience at a single institution. J Pediatr
Surg. 1997;32(3):434–437.
22. Rowe MI, Reblock KK, Kurkchubasche AG, Healey PJ. Necrotizing
enterocolitis in the extremely low birth weight infant. J Pediatr Surg.
1994;29(8):987–990.
23. Gatt M, Reddy BS, MacFie J. Review article: Bacterial translocation
in the critically ill evidence and methods of prevention. Aliment
Pharmacol Ther. 2007;25(7):741–757.
24. MacFie J, Reddy BS, Gatt M, Jain PK, Sowdi R, Mitchell CJ.
Bacterial translocation studied in 927 patients over 13 years. Br J Surg.
2006;93(1):87–93.
25. Stechmiller JK, Treloar D, Allen N. Gut dysfunction in critically ill
patients: A review of the literature. Am J Crit Care. 1997;6(3):204–209.
26. Bhandari V, Bizzarro MJ, Shetty A, et al. Familial and genetic
susceptibility to major neonatal morbidities in preterm twins. Pediatrics.
2006;117(6):1901–1906.
27. Treszl A, Heninger E, Kalman A, Schuler A, Tulassay T, Vasarhelyi B.
Lower prevalence of IL-4 receptor alpha-chain gene G variant in
very-low-birth-weight infants with necrotizing enterocolitis. J Pediatr
Surg. 2003;38(9):1374–1378.
28. Heninger E, Treszl A, Kocsis I, Derfalvi B, Tulassay T, Vasarhelyi B.
Genetic variants of the interleukin-18 promoter region (-607) influence
the course of necrotising enterocolitis in very low birth weight neonates.
Eur J Pediatr. 2002;161(7):410–411.
29. Banyasz I, Bokodi G, Vasarhelyi B, et al. Genetic polymorphisms
for vascular endothelial growth factor in perinatal complications. Eur
Cytokine Netw. 2006;17(4):266–270.
30. M o on en RM , Pa ul us s en A D, Sou r en NY, Kess e ls AG,
Rubio-Gozalbo ME, Villamor E. Carbamoyl phosphate synthetase
polymorphisms as a risk factor for necrotizing enterocolitis. Pediatr
Res. 2007; 62(2):188–190.
31. Henderson G, Craig S, Baier RJ, Helps N, Brocklehurst P, McGuire W.
Cytokine gene polymorphisms in preterm infants with necrotising
enterocolitis: Genetic association study. Arch Dis Child Fetal Neonatal
Ed. 2009;94(2):F124–F128.
32. Habib Z, Arnaud B, Pascal DL, et al. CARD15/NOD2 is not a
predisposing factor for necrotizing enterocolitis. Dig Dis Sci. 2005;
50(9):1684–1687.
33. Szebeni B, Szekeres R, Rusai K, et al. Genetic polymorphisms of
CD14, toll-like receptor 4, and caspase-recruitment domain 15 are
not associated with necrotizing enterocolitis in very low birth weight
infants. J Pediatr Gastroenterol Nutr. 2006;42(1):27–31.
34. Grylack LJ. Neonatal necrotizing enterocolitis revisited. Perinatal
Press. 1986;9:146–148.
35. McKeown RE, Marsh TD, Amarnath U, et al. Role of delayed feeding
and of feeding increments in necrotizing enterocolitis. J Pediatr. 1992;
121(5 Pt 1):764–770.
36. Hsueh W, Caplan MS, Qu XW, Tan XD, De Plaen IG, Gonzalez-
Crussi F. Neonatal necrotizing enterocolitis: Clinical considerations
and pathogenetic concepts. Pediatr Dev Pathol. 2003;6(1):6–23.
37. Di Lorenzo M, Bass J, Krantis A. An intraluminal model of necrotizing
enterocolitis in the developing neonatal piglet. J Pediatr Surg. 1995;
30(8):1138–1142.
38. Nafday SM, Chen W, Peng L, Babyatsky MW, Holzman IR, Lin J.
Short-chain fatty acids induce colonic mucosal injury in rats with
various postnatal ages. Pediatr Res. 2005;57(2):201–204.
39. Lucas A, Cole TJ. Breast milk and neonatal necrotising enterocolitis.
Lancet. 1990;336(8730):1519–1523.
40. Meinzen-Derr J, Poindexter B, Wrage L, Morrow AL, Stoll B,
Donovan EF. Role of human milk in extremely low birth weight infants’
risk of necrotizing enterocolitis or death. J Perinatol. 2009;29(1):
57–62.
41. Sisk PM, Lovelady CA, Dillard RG, Gruber KJ, O’Shea TM. Early
human milk feeding is associated with a lower risk of necrotizing
enterocolitis in very low birth weight infants. J Perinatol. 2007;27(7):
428–433.
42. Updegrove K. Necrotizing enterocolitis: The evidence for use of human
milk in prevention and treatment. J Hum Lact. 2004;20(3):335–339.
43. Sangild PT, Siggers RH, Schmidt M, et al. Diet- and colonization-
dependent intestinal dysfunction predisposes to necrotizing enterocolitis
in preterm pigs. Gastroenterology. 2006;130(6):1776–1792.
44. Caplan MS, Amer M, Jilling T. The role of human milk in necrotizing
enterocolitis. Adv Exp Med Biol. 2002;503:83–90.
45. McGuire W, Bombell S. Slow advancement of enteral feed volumes
to prevent necrotising enterocolitis in very low birth weight infants.
Cochrane Database Syst Rev. 2008;2:CD001241.
46. 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.
47. Emami CN, Petrosyan M, Giuliani S, et al. Role of the host defense
system and intestinal microbial flora in the pathogenesis of necrotizing
enterocolitis. Surg Infect (Larchmt). 2009;10(5):407–417.
48. Siggers RH, Siggers J, Thymann T, Boye M, Sangild PT. Nutritional
modulation of the gut microbiota and immune system in preterm
neonates susceptible to necrotizing enterocolitis. J Nutr Biochem. 2011;
22(6):511–521.
49. Foster J, Cole M. Oral immunoglobulin for preventing necrotizing
enterocolitis in preterm and low birth-weight neonates. Cochrane
Database Syst Rev. 2004;1:CD001816.
50. Hang P, Sangild PT, Sit WH, et al. Temporal proteomic analysis of
intestine developing necrotizing enterocolitis following enteral formula
feeding to preterm pigs. J Proteome Res. 2009;8(1):72–81.
51. Rayyis SF, Ambalavanan N, Wright L, Carlo WA. Randomized trial of
“slow” versus “fast” feed advancements on the incidence of necrotizing
enterocolitis in very low birth weight infants. J Pediatr. 1999;134(3):
293–297.
52. Brown EG, Sweet AY. Preventing necrotizing enterocolitis in neonates.
JAMA. 1978;240(22):2452–2454.
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
50
Maheshwari et al
53. Patole S. Safety of enteral feed volumes in neonates at risk for necrotizing
enterocolitis: The never-ending story. Pediatrics. 2004;114(1):327.
54. Patole SK, de Klerk N. Impact of standardised feeding regimens on
incidence of neonatal necrotising enterocolitis: A systematic review and
meta-analysis of observational studies. Arch Dis Child Fetal Neonatal
Ed. 2005;90(2):F147–F151.
55. Premji SS, Chessell L, Paes B, Pinelli J, Jacobson K. A matched cohort
study of feeding practice guidelines for infants weighing less than
1,500 g. Adv Neonatal Care. 2002;2(1):27–36.
56. Kuzma-O’Reilly B, Duenas ML, Greecher C, et al. Evaluation,
development, and implementation of potentially better practices
in neonatal intensive care nutrition. Pediatrics. 2003;111(4 Pt 2):
e461–e470.
57. Uauy RD, Fanaroff AA, Korones SB, Phillips EA, Phillips JB,
Wright LL. Necrotizing enterocolitis in very low birth weight infants:
Biodemographic and clinical correlates. National Institute of Child
Health and Human Development Neonatal Research Network. J Pediatr.
1991;119(4):630–638.
58. Lambert DK, Christensen RD, Baer VL, et al. Fulminant necrotizing
enterocolitis in a multihospital healthcare system. J Perinatol. May 12,
2011. [Epub ahead of print.]
59. Caple J, Armentrout D, Huseby V, et al. Randomized, controlled trial
of slow versus rapid feeding volume advancement in preterm infants.
Pediatrics. 2004;114(6):1597–1600.
60. Salhotra A, Ramji S. Slow versus fast enteral feed advancement in very
low birth weight infants: A randomized control trial. Indian Pediatr.
2004;41(5):435–441.
61. Krishnamurthy S, Gupta P, Debnath S, Gomber S. Slow versus rapid
enteral feeding advancement in preterm newborn infants 1000–1499 g:
A randomized controlled trial. Acta Paediatr. 2010;99(1):42–46.
62. 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.
63. Jilling T, Lu J, Jackson M, Caplan MS. Intestinal epithelial apoptosis
initiates gross bowel necrosis in an experimental rat model of neonatal
necrotizing enterocolitis. Pediatr Res. 2004;55(4):622–629.
64. Richardson WM, Dai S, Dyer M, et al. Toll like receptor-4 activation
links enterocyte autophagy with apoptosis via the stress response
gene ATG 16 in the pathogenesis of necrotizing enterocolitis. J Surg
Res;158(2):209.
65. Nanthakumar NN, Fusunyan RD, Sanderson I, Walker WA. Inflamma-
tion in the developing human intestine: A possible pathophysiologic
contribution to necrotizing enterocolitis. Proc Natl Acad Sci U S A.
2000;97(11):6043–6048.
66. Maheshwari A, Kelly DR, Nicola T, et al. TGF-beta(2) suppresses
macrophage cytokine production and mucosal inflammatory responses
in the developing intestine. Gastroenterology. 2011;140(1):242–253.
67. Nowicki P. Intestinal ischemia and necrotizing enterocolitis. J Pediatr.
1990;117(1 Pt 2):S14–S19.
68. Nowicki PT. Ischemia and necrotizing enterocolitis: Where, when, and
how. Semin Pediatr Surg. 2005;14(3):152–158.
69. Nowicki PT, Nankervis CA. The role of the circulation in the
pathogenesis of necrotizing enterocolitis. Clin Perinatol. 1994;21(2):
219–234.
70. Rist CB, Watts JC, Lucas RJ. Isolated ischemic necrosis of the cecum
in patients with chronic heart disease. Dis Colon Rectum. 1984;27(8):
548–551.
71. Reber KM, Nankervis CA, Nowicki PT. Newborn intestinal circulation.
Physiology and pathophysiology. Clin Perinatol. 2002;29(1):23–39.
72. Neu J. The ‘myth’ of asphyxia and hypoxia-ischemia as primary causes
of necrotizing enterocolitis. Biol Neonate. 2005;87(2):97–98.
73. Neu J, Mshvildadze M, Mai V. A roadmap for understanding and
preventing necrotizing enterocolitis. Curr Gastroenterol Rep. 2008;
10(5):450–457.
74. Neu J, Chen M, Beierle E. Intestinal innate immunity: How does it
relate to the pathogenesis of necrotizing enterocolitis. Semin Pediatr
Surg. 2005;14(3):137–144.
75. Pender SL, Braegger C, Gunther U, Monteleone G, Meuli M,
Schuppan G. Matrix metalloproteinases in necrotising enterocolitis.
Pediatr Res. 2003;54(2):160–164.
76. Caplan MS, Sun XM, Hseuh W, Hageman JR. Role of platelet
activating factor and tumor necrosis factor-alpha in neonatal necrotizing
enterocolitis. J Pediatr. 1990;116(6):960–964.
77. Caplan MS, Simon D, Jilling T. The role of PAF, TLR, and the
inflammatory response in neonatal necrotizing enterocolitis. Semin
Pediatr Surg. 2005;14(3):145–151.
78. Viscardi RM, Lyon NH, Sun CC, Hebel JR, Hasday JD. Inflammatory
cytokine mRNAs in surgical specimens of necrotizing enterocolitis
and normal newborn intestine. Pediatr Pathol Lab Med. 1997;17(4):
547–559.
79. Halpern MD, Holubec H, Dominguez JA, et al. Up-regulation of IL-18
and IL-12 in the ileum of neonatal rats with necrotizing enterocolitis.
Pediatr Res. 2002;51(6):733–739.
80. Halpern MD, Holubec H, Dominguez JA, et al. Hepatic inflammatory
mediators contribute to intestinal damage in necrotizing enterocolitis.
Am J Physiol Gastrointest Liver Physiol. 2003;284(4):G695–G6702.
81. Nanthakumar N, Meng D, Goldstein AM, et al. The mechanism of
excessive intestinal inflammation in necrotizing enterocolitis: An
immature innate immune response. PLoS One. 2011;6(3):e17776.
82. Edelson MB, Bagwell CE, Rozycki HJ. Circulating pro- and counter-
inflammatory cytokine levels and severity in necrotizing enterocolitis.
Pediatrics. 1999;103(4 Pt 1):766–771.
83. Ng PC, Li K, Wong RP, et al. Proinflammatory and anti-inflammatory
cytokine responses in preterm infants with systemic infections. Arch
Dis Child Fetal Neonatal Ed. 2003;88(3):F209–F213.
84. Musemeche C, Caplan M, Hsueh W, Sun X, Kelly A. Experimental
necrotizing enterocolitis: The role of polymorphonuclear neutrophils.
J Pediatr Surg. 1991;26(9):1047–1049.
85. Halpern MD, Clark JA, Saunders TA, et al. Reduction of experimental
necrotizing enterocolitis with anti-TNF-alpha. Am J Physiol Gastrointest
Liver Physiol. 2006;290(4):G757–G764.
86. Hsueh W, Caplan MS, Tan X, MacKendrick W, Gonzalez-Crussi F.
Necrotizing enterocolitis of the newborn: Pathogenetic concepts in
perspective. Pediatr Dev Pathol. 1998;1(1):2–16.
87. Neu J, Weiss MD. Necrotizing enterocolitis: Pathophysiology and
prevention. JPEN J Parenter Enteral Nutr. 1999;23(5 Suppl):S13–S17.
88. Bury RG, Tudehope D. Enteral antibiotics for preventing necrotizing
enterocolitis in low birthweight or preterm infants. Cochrane Database
Syst Rev. 2001;1:CD000405.
89. Chan KL, Ng SP, Chan KW, Wo YH, Tam PK. Pathogenesis of neonatal
necrotizing enterocolitis: A study of the role of intraluminal pressure, age
and bacterial concentration. Pediatr Surg Int. 2003;19(8):573–577.
90. Lin J, Nafday SM, Chauvin SN, et al. Variable effects of short chain fatty
acids and lactic acid in inducing intestinal mucosal injury in newborn
rats. J Pediatr Gastroenterol Nutr. 2002;35(4):545–550.
91. Kliegman RM, Walker WA, Yolken RH. Necrotizing enterocolitis:
Research agenda for a disease of unknown etiology and pathogenesis.
Pediatr Res. 1993;34(6):701–708.
92. Millar MR, Linton CJ, Cade A, Glancy D, Hall M, Jalal H. Application
of 16S rRNA gene PCR to study bowel flora of preterm infants with
and without necrotizing enterocolitis. J Clin Microbiol. 1996;34(10):
2506–2510.
93. Hoy CM, Wood CM, Hawkey PM, Puntis JW. Duodenal microflora in
very-low-birth-weight neonates and relation to necrotizing enterocolitis.
J Clin Microbiol. 2000;38(12):4539–4547.
94. de la Cochetiere MF, Piloquet H, des Robert C, Darmaun D,
Galmiche JP, Roze JC. Early intestinal bacterial colonization and
necrotizing enterocolitis in premature infants: The putative role of
Clostridium. Pediatr Res. 2004;56(3):366–370.
95. Wang Y, Hoenig JD, Malin KJ, et al. 16S rRNA gene-based analysis
of fecal microbiota from preterm infants with and without necrotizing
enterocolitis. ISME J. 2009;3(8):944–954.
96. Van Camp JM, Tomaselli V, Coran AG. Bacterial translocation in the
neonate. Curr Opin Pediatr. 1994;6(3):327–333.
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
51
Neonatal necrotizing enterocolitis
97. Claud EC, Walker WA. Hypothesis: Inappropriate colonization of
the premature intestine can cause neonatal necrotizing enterocolitis.
FASEB J. 2001;15(8):1398–1403.
98. Cotten CM, Taylor S, Stoll B, et al. Prolonged duration of initial
empirical antibiotic treatment is associated with increased rates of
necrotizing enterocolitis and death for extremely low birth weight
infants. Pediatrics. 2009;123(1):58–66.
99. Salzman NH, Polin RA, Harris MC, et al. Enteric defensin expression
in necrotizing enterocolitis. Pediatr Res. 1998;44(1):20–26.
100. Wolfs TG, Derikx JP, Hodin CM, et al. Localization of the lipopoly-
saccharide recognition complex in the human healthy and inflamed
premature and adult gut. Inflamm Bowel Dis. 2010;16(1):68–75.
101. Puiman PJ, Burger-Van Paassen N, Schaart MW, et al. Paneth cell
hyperplasia and metaplasia in necrotizing enterocolitis. Pediatr Res.
2011;69(3):217–223.
102. Bevins CL, Salzman NH. Paneth cells, antimicrobial peptides and
maintenance of intestinal homeostasis. Nat Rev Microbiol. 2011;9(5):
356–368.
103. Coutinho HB, da Mota HC, Coutinho VB, et al. Absence of lysozyme
(muramidase) in the intestinal Paneth cells of newborn infants with
necrotising enterocolitis. J Clin Pathol. 1998;51(7):512–514.
104. Schaart MW, de Bruijn AC, Bouwman DM, et al. Epithelial functions
of the residual bowel after surgery for necrotising enterocolitis in
human infants. J Pediatr Gastroenterol Nutr. 2009;49(1):31–41.
105. Brandtzaeg P, Halstensen TS, Kett K, et al. Immunobiology and
immunopathology of human gut mucosa: Humoral immunity and intra-
epithelial lymphocytes. Gastroenterology. 1989;97(6):1562–1584.
106. Van der Heijden PJ, Stok W, Bianchi AT. Contribution of
immunoglobulin-secreting cells in the murine small intestine to the
total ‘background’ immunoglobulin production. Immunology. 1987;
62(4):551–555.
107. Brandtzaeg P. Overview of the mucosal immune system. Curr Top
Microbiol Immunol. 1989;146:13–25.
108. Ogra PL, Losonsky GA, Fishaut M. Colostrum-derived immunity and
maternal-neonatal interaction. Ann N Y Acad Sci. 1983;409:82–95.
109. Hanson LA, Korotkova M. The role of breastfeeding in prevention of
neonatal infection. Semin Neonatol. 2002;7(4):275–281.
110. Piena-Spoel M, Albers MJ, ten Kate J, Tibboel D. Intestinal permeabil-
ity in newborns with necrotizing enterocolitis and controls: Does the
sugar absorption test provide guidelines for the time to (re-)introduce
enteral nutrition? J Pediatr Surg. 2001;36(4):587–592.
111. Hackam DJ, Upperman JS, Grishin A, Ford HR. Disordered enterocyte
signaling and intestinal barrier dysfunction in the pathogenesis of
necrotizing enterocolitis. Semin Pediatr Surg. 2005;14(1):49–57.
112. Upperman JS, Potoka D, Grishin A, Hackam D, Zamora R, Ford HR.
Mechanisms of nitric oxide-mediated intestinal barrier failure in
necrotizing enterocolitis. Semin Pediatr Surg. 2005;14(3):159–166.
113. Chokshi NK, Guner YS, Hunter CJ, Upperman JS, Grishin A, Ford HR.
The role of nitric oxide in intestinal epithelial injury and restitution
in neonatal necrotizing enterocolitis. Semin Perinatol. 2008;32(2):
92–99.
114. Clark JA, Doelle SM, Halpern MD, et al. Intestinal barrier failure
during experimental necrotizing enterocolitis: Protective effect of
EGF treatment. Am J Physiol Gastrointest Liver Physiol. 2006;291(5):
G938–G949.
115. Shin CE, Falcone RA Jr, Stuart L, Erwin CR, Warner BW. Diminished
epidermal growth factor levels in infants with necrotizing enterocolitis.
J Pediatr Surg. 2000;35(2):173–176.
116. Feng J, El-Assal ON, Besner GE. Heparin-binding EGF-like growth
factor (HB-EGF) and necrotizing enterocolitis. Semin Pediatr Surg.
2005;14(3):167–174.
117. Warner BB, Ryan AL, Seeger K, Leonard AC, Erwin CR, Warner BW.
Ontogeny of salivary epidermal growth factor and necrotizing
enterocolitis. J Pediatr. 2007;150(4):358–363.
118. Mally P, Golombek SG, Mishra R, et al. Association of necrotizing
enterocolitis with elective packed red blood cell transfusions in stable,
growing, premature neonates. Am J Perinatol. 2006;23(8):451–458.
119. Christensen RD, Lambert DK, Henry E, et al. Is transfusion-
associated necrotizing enterocolitis” an authentic pathogenic entity?
Transfusion. 2010;50(5):1106–1112.
120. Krimmel GA, Baker R, Yanowitz TD. Blood transfusion alters the
superior mesenteric artery blood flow velocity response to feeding in
premature infants. Am J Perinatol. 2009;26(2):99–105.
121. Maayan-Metzger A, Itzchak A, Mazkereth R, Kuint J. Necrotizing
enterocolitis in full-term infants: case-control study and review of the
literature. J Perinatol. 2004;24(8):494–499.
122. Caplan MS, Jilling T. New concepts in necrotizing enterocolitis. Curr
Opin Pediatr. 2001;13(2):111–115.
123. González-Rivera R, Culverhouse RC, Hamvas A, Tarr PI, Warner BB.
The age of necrotizing enterocolitis onset: an application of Sartwell’s
incubation period model. J Perinatol. 2011;31:519–523.
124. Sharma R, Hudak ML, Tepas JJ, 3rd et al. Impact of gestational age
on the clinical presentation and surgical outcome of necrotizing
enterocolitis. J Perinatol. 2006;26(6):342–347.
125. Walsh MC, Kliegman RM. Necrotizing enterocolitis: Treatment based
on staging criteria. Pediatr Clin North Am. 1986;33(1):179–201.
126. Clark RH, Gordon P, Walker WM, Laughon M, Smith PB, Spitzer AR.
Characteristics of patients who die of necrotizing enterocolitis.
J Perinatol. May 19, 2011. [Epub ahead of print.]
127. Thompson A, Bizzarro M, Yu S, Diefenbach K, Simpson BJ, Moss RL.
Risk factors for necrotizing enterocolitis totalis: A case-control study.
J Perinatol. March 24, 2011. [Epub ahead of print.]
128. Siahanidou T, Mandyla H, Anagnostakis D, Papandreou E. Twenty-six
full-term (FT) neonates with necrotizing enterocolitis (NEC). J Pediatr
Surg. 2004;39(5):791.
129. Ostlie DJ, Spilde TL, St Peter SD, et al. Necrotizing enterocolitis in
full-term infants. J Pediatr Surg. 2003;38(7):1039–1042.
130. Bolisetty S, Lui K. Necrotizing enterocolitis in full-term neonates.
J Paediatr Child Health. 2001;37(4):413–414.
131. Ruangtrakool R, Laohapensang M, Sathornkich C, Talalak P.
Necrotizing enterocolitis: A comparison between full-term and
pre-term neonates. J Med Assoc Thai. 2001;84(3):323–331.
132. Wiswell TE, Robertson CF, Jones TA, Tuttle DJ. Necrotizing entero-
colitis in full-term infants. A case-control study. Am J Dis Child.
1988;142(5):532–535.
133. Martinez-Tallo E, Claure N, Bancalari E. Necrotizing enterocolitis in
full-term or near-term infants: Risk factors. Biol Neonate. 1997;71(5):
292–298.
134. Rodin AE, Nichols MM, Hsu FL. Necrotizing enterocolitis occurring
in full-term neonates at birth. Arch Pathol. 1973;96(5):335–338.
135. Lambert DK, Christensen RD, Henry E, et al. Necrotizing enterocolitis
in term neonates: Data from a multihospital health-care system.
J Perinatol. 2007;27(7):437–443.
136. Raboei EH. Necrotizing enterocolitis in full-term neonates: is it
aganglionosis? Eur J Pediatr Surg. 2009;19(2):101–104.
137. Christensen RD, Wiedmeier SE, Baer VL, et al. Antecedents of
Bell stage III necrotizing enterocolitis. J Perinatol. 2010;30(1):
54–57.
138. Buonomo C. The radiology of necrotizing enterocolitis. Radiol Clin
North Am. 1999;37(6):1187–1198, vii.
139. Sharma R, Tepas JJ 3rd, Hudak ML, et al. Portal venous gas and
surgical outcome of neonatal necrotizing enterocolitis. J Pediatr Surg.
2005;40(2):371–376.
140. Dordelmann M, Rau GA, Bartels D, et al. Evaluation of portal venous
gas detected by ultrasound examination for diagnosis of necrotising
enterocolitis. Arch Dis Child Fetal Neonatal Ed. 2009;94(3):
F183–F187.
141. Dilli D, Suna Oguz S, Erol R, Ozkan-Ulu H, Dumanli H, Dilmen U.
Does abdominal sonography provide additional information over
abdominal plain radiography for diagnosis of necrotizing enterocolitis
in neonates? Pediatr Surg Int. 2011;27(3):321–327.
142. Kliegman RM, Fanaroff AA. Neonatal necrotizing enterocolitis in the
absence of pneumatosis intestinalis. Am J Dis Child. 1982;136(7):
618–620.
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
52
Maheshwari et al
143. Frey EE, Smith W, Franken EA, Jr, Wintermeyer KA. Analysis of
bowel perforation in necrotizing enterocolitis. Pediatr Radiol. 1987;
17(5):380–382.
144. Travadi JN, Patole SK, Gardiner K. Pneumatosis coli, a benign form
of necrotising enterocolitis. Indian Pediatr. 2003;40(4):349–351.
145. Maheshwari A. Practical approaches to the neutropenic neonate.
In: Ohls RK, Yoder MC, editors. Neonatology: Questions and
Controversies Series. Vo l . Hematology, Immunology, Infectious
Disease. Philadelphia, PA: WB Saunders Company; 2008.
146. Cheu HW, Brown DR, Rowe MI. Breath hydrogen excretion as a
screening test for the early diagnosis of necrotizing enterocolitis. Am
J Dis Child. 1989;143(2):156–159.
147. Holland AJ, Shun A, Martin HC, Cooke-Yarborough C, Holland J.
Small bowel perforation in the premature neonate: Congenital or
acquired? Pediatr Surg Int. 2003;19(6):489–494.
148. Gordon PV, Price WA, Stiles AD, Rutledge JC. Early postnatal
dexamethasone diminishes transforming growth factor alpha
localization within the ileal muscularis propria of newborn mice and
extremely low-birth-weight infants. Pediatr Dev Pathol. 2001;4(6):
532–537.
149. Paquette L, Friedlich P, Ramanathan R, Seri I. Concurrent use of
indomethacin and dexamethasone increases the risk of spontaneous
intestinal perforation in very low birth weight neonates. J Perinatol.
2006;26(8):486–492.
150. Coates EW, Karlowicz MG, Croitoru DP, Buescher ES. Distinctive
distribution of pathogens associated with peritonitis in neonates with
focal intestinal perforation compared with necrotizing enterocolitis.
Pediatrics. 2005;116(2):e241–e246.
151. Scheifele DW, Ginter GL, Olsen E, Fussell S, Pendray M. Comparison
of two antibiotic regimens for neonatal necrotizing enterocolitis.
J Antimicrob Chemother. 1987;20(3):421–429.
152. Moss RL, Dimmitt RA, Barnhart DC, et al. Laparotomy versus
peritoneal drainage for necrotizing enterocolitis and perforation.
N Engl J Med. 2006;354(21):2225–2234.
153. Rees CM, Eaton S, Kiely EM, Wade AM, McHugh K, Pierro A.
Peritoneal drainage or laparotomy for neonatal bowel perforation?
A randomized controlled trial. Ann Surg. 2008;248(1):44–51.
154. Rao SC, Basani L, Simmer K, Samnakay N, Deshpande G. Peritoneal
drainage versus laparotomy as initial surgical treatment for perfo-
rated necrotizing enterocolitis or spontaneous intestinal perforation
in preterm low birth weight infants. Cochrane Database Syst Rev.
2011; 6:CD006182.
155. Blakely ML, Tyson JE, Lally KP, et al. Laparotomy versus peritoneal
drainage for necrotizing enterocolitis or isolated intestinal perforation
in extremely low birth weight infants: Outcomes through 18 months
adjusted age. Pediatrics. 2006;117(4):e680–e687.
156. Sola JE, Tepas JJ 3rd, Koniaris LG. Peritoneal drainage versus
laparotomy for necrotizing enterocolitis and intestinal perforation:
A meta-analysis. J Surg Res. 2010;161(1):95–100.
157. Roberts D, Dalziel S. Antenatal corticosteroids for accelerating fetal
lung maturation for women at risk of preterm birth. Cochrane Database
Syst Rev. 2006;3:CD004454.
158. Murphy KE, Hannah ME, Willan AR, et al. Multiple courses of
antenatal corticosteroids for preterm birth (MACS): A randomised
controlled trial. Lancet. 2008;372(9656):2143–2151.
159. Hay WW Jr. Strategies for feeding the preterm infant. Neonatology.
2008;94(4):245–254.
160. Tyson JE, Kennedy KA, Lucke JF, Pedroza C. Dilemmas initiating
enteral feedings in high risk infants: How can they be resolved? Semin
Perinatol. 2007;31(2):61–73.
161. Schmolzer G, Urlesberger B, Haim M, et al. Multi-modal approach to
prophylaxis of necrotizing enterocolitis: Clinical report and review of
literature. Pediatr Surg Int. 2006;22(7):573–580.
162. Newell SJ. Enteral feeding of the micropremie. Clin Perinatol. 2000;
27(1):221–234, viii.
163. Bombell S, McGuire W. Early trophic feeding for very low birth weight
infants. Cochrane Database Syst Rev. 2009;3:CD000504.
164. [No authors listed]. Breastfeeding and the use of human milk.
American Academy of Pediatrics. Work Group on Breastfeeding.
Pediatrics. 1997;100(6):1035–1039.
165. Gartner LM, Morton J, Lawrence RA, et al. Breastfeeding and the use
of human milk. Pediatrics. 2005;115(2):496–506.
166. Sullivan S, Schanler RJ, Kim JH, 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(4):562–567.
167. Ziegler TR, Evans ME, Fernandez-Estivariz C, Jones DP. Trophic
and cytoprotective nutrition for intestinal adaptation, mucosal repair,
and barrier function. Annu Rev Nutr. 2003;23:229–261.
168. Amin HJ, Zamora SA, McMillan DD, et al. Arginine supplementation
prevents necrotizing enterocolitis in the premature infant. J Pediatr.
2002;140(4):425–431.
169. Shah P, Shah V. Arginine supplementation for prevention of necro-
tising enterocolitis in preterm infants. Cochrane Database Syst Rev.
2007;3:CD004339.
170. Seitz G, Warmann SW, Guglielmetti A, et al. Protective effect of
tumor necrosis factor alpha antibody on experimental necrotizing
enterocolitis in the rat. J Pediatr Surg. 2005;40(9):1440–1445.
171. Tubman TR, Thompson SW, McGuire W. Glutamine supplementation
to prevent morbidity and mortality in preterm infants. Cochrane
Database Syst Rev. 2008;1:CD001457.
172. Kelly N, Friend K, Boyle P, et al. The role of the glutathione antioxidant
system in gut barrier failure in a rodent model of experimental
necrotizing enterocolitis. Surgery. 2004;136(3):557–566.
173. Lin HC, Su BH, Oh W. Oral probiotics prevent necrotizing enterocolitis.
J Pediatr. 2006;148(6):849.
174. Lin HC, Hsu CH, Chen HL, et al. Oral probiotics prevent necro-
tizing enterocolitis in very low birth weight preterm infants:
A multicenter, randomized, controlled trial. Pediatrics. 2008;122(4):
693–700.
175. 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.
176. Li Y, Shimizu T, Hosaka A, Kaneko N, Ohtsuka Y, Yamashiro Y. Effects
of bifidobacterium breve supplementation on intestinal flora of low
birth weight infants. Pediatr Int. 2004;46(5):509–515.
177. Reuman PD, Duckworth DH, Smith KL, Kagan R, Bucciarelli RL,
Ayoub EM. Lack of effect of Lactobacillus on gastrointestinal bacterial
colonization in premature infants. Pediatr Infect Dis. 1986;5(6):
663–668.
178. Kitajima H, Sumida Y, Tanaka R, Yuki N, Takayama H, Fujimura M.
Early administration of Bifidobacterium breve to preterm infants:
Randomised controlled trial. Arch Dis Child Fetal Neonatal Ed.
1997;76(2):F101–F107.
179. Bin-Nun A, Bromiker R, Wilschanski M, et al. Oral probiotics prevent
necrotizing enterocolitis in very low birth weight neonates. J Pediatr.
2005;147(2):192–196.
180. 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):1–4.
181. 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(12):1735–1742.
182. Manzoni P, Rinaldi M, Cattani S, et al. Bovine lactoferrin
supplementation for prevention of late-onset sepsis in very low-
birth-weight neonates: A randomized trial. JAMA. 2009;302(13):
1421–1428.
183. Millar MR, Bacon C, Smith SL, Walker V, Hall MA. Enteral feeding
of premature infants with Lactobacillus GG. Arch Dis Child. 1993;69
(5 Spec No):483–487.
184. Costalos C, Skouteri V, Gounaris A, et al. Enteral feeding of premature
infants with Saccharomyces boulardii. Early Hum Dev. 2003;74(2):
89–96.
Research and Reports in Neonatology
Publish your work in this journal
Submit your manuscript here: http://www.dovepress.com/research-and-reports-in-neonatology-journal
Research and Reports in Neonatology is an international, peer-reviewed,
open access journal publishing original research, reports, editorials,
reviews and commentaries on neonatal health. The manuscript manage-
ment system is completely online and includes a very quick and fair
peer-review system. Visit http://www.dovepress.com/testimonials.php
to read real quotes from published authors.
Research and Reports in Neonatology 2011:1
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
Dovepress
53
Neonatal necrotizing enterocolitis
185. Mohan R, Koebnick C, Schildt J, Mueller M, Radke M, Blaut M.
Effects of Bifidobacterium lactis Bb12 supplementation on body
weight, fecal pH, acetate, lactate, calprotectin, and IgA in preterm
infants. Pediatr Res. 2008;64(4):418–422.
186. 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(9):575–579.
187. 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(2):103–108.
188. 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(6):
1828–1835.
189. Samanta M, Sarkar M, Ghosh P, Ghosh J, Sinha M, Chatterjee S.
Prophylactic probiotics for prevention of necrotizing enterocolitis
in very low birth weight newborns. J Trop Pediatr. 2009;55(2):
128–1231.
190. Sari FN, Dizdar EA, Oguz S, Erdeve O, Uras N, Dilmen U. 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(4):434–439.
191. Deshpande G, Rao S, Patole S, Bulsara M. Updated meta-analysis of
probiotics for preventing necrotizing enterocolitis in preterm neonates.
Pediatrics. 2010;125(5):921–930.
192. Braegger C, Chmielewska A, Decsi T, et al. Supplementation of
infant formula with probiotics and/or prebiotics: a systematic review
and comment by the ESPGHAN committee on nutrition. J Pediatr
Gastroenterol Nutr. 2011;52(2):238–250.
193. Sanderson IR. Dietary modulation of GALT. J Nutr. 2007;137
(11 Suppl):2557S–2562S.
194. Lewis S, Burmeister S, Brazier J. Effect of the prebiotic oligofructose
on relapse of Clostridium difficile-associated diarrhea: A randomized,
controlled study. Clin Gastroenterol Hepatol. 2005;3(5):442–448.
195. Butel MJ, Waligora-Dupriet AJ, Szylit O. Oligofructose and
experimental model of neonatal necrotising enterocolitis. Br J Nutr.
2002;87 Suppl 2:S213–S219.
196. Catala I, Butel MJ, Bensaada M, et al. Oligofructose contributes
to the protective role of bifidobacteria in experimental necrotising
enterocolitis in quails. J Med Microbiol. 1999;48(1):89–94.
197. Maheshwari A. Role of cytokines in human intestinal villous
development. Clin Perinatol. 2004;31(1):143–155.
198. Dvorak B, Halpern MD, Holubec H, et al. Epidermal growth factor
reduces the development of necrotizing enterocolitis in a neonatal
rat model. Am J Physiol Gastrointest Liver Physiol. 2002;282(1):
G156–G164.
199. Calhoun DA, Christensen RD. Hematopoietic growth factors in
neonatal medicine: The use of enterally administered hematopoietic
growth factors in the neonatal intensive care unit. Clin Perinatol. 2004;
31(1):169–182.
200. Campbell EL, Louis NA, Tomassetti SE, et al. Resolvin E1 promotes
mucosal surface clearance of neutrophils: A new paradigm for
inflammatory resolution. FASEB J. 2007;21(12):3162–3170.
201. Christensen RD, Havranek T, Gerstmann DR, Calhoun DA. Enteral
administration of a simulated amniotic fluid to very low birth weight
neonates. J Perinatol. 2005;25(6):380–385.
202. Barney CK, Purser N, Christensen RD. A phase 1 trial testing an
enteral solution patterned after human amniotic fluid to treat feeding
intolerance. Adv Neonatal Care. 2006;6(2):89–95.
203. Barney CK, Lambert DK, Alder SC, Scoffield SH, Schmutz N,
Christensen RD. Treating feeding intolerance with an enteral solution
patterned after human amniotic fluid: A randomized, controlled,
masked trial. J Perinatol. 2007;27(1):28–31.
204. Sullivan SE, Calhoun DA, Maheshwari A, et al. Tolerance of simulated
amniotic fluid in premature neonates. Ann Pharmacother. 2002;36(10):
1518–1524.
205. Gersting JA, Christensen RD, Calhoun DA. Effects of enterally
administering granulocyte colony-stimulating factor to suckling mice.
Pediatr Res. 2004;55(5):802–806.
206. Juul SE, Zhao Y, Dame JB, Du Y, Hutson AD, Christensen RD.
Origin and fate of erythropoietin in human milk. Pediatr Res.
2000;48(5):660–667.
207. Hintz SR, Kendrick DE, Stoll BJ, et al. Neurodevelopmental
and growth outcomes of extremely low birth weight infants after
necrotizing enterocolitis. Pediatrics. 2005;115(3):696–703.
208. Tyson JE, Kennedy KA. Trophic feedings for parenterally fed infants.
Cochrane Database Syst Rev. 2005;3:CD000504.
... We have described how the pro-inflammatory characteristics of still-maturing intestinal macrophages in the preterm intestine can increase the risk of NEC and refer the reader to the articles listed in the references. 121,[168][169][170][171][172][173]200,226,[229][230][231][232][233][234][235][236][237] Several genetic and epigenetic factors may also alter the susceptibility to NEC and/or its severity, its clinical and histopathological manifestations, and if surgery is needed, of an adverse postoperative course and outcome. 227,238 conclusions Recent years have brought considerable progress in our understanding of the heterogeneity and inflammatory immaturity of gut macrophages in premature and full-term newborn infants; particularly in the context of diseases such as NEC. ...
Article
Full-text available
Macrophages are large highly motile phagocytic leukocytes that appear early during embryonic development and have been conserved during evolution. The developmental roles of macrophages were first described nearly a century ago, at about the time these cells were being identified as central effectors in phagocytosis and elimination of microbes. Since then, we have made considerable progress in understanding the development of various subsets of macrophages and the diverse roles these cells play in both physiology and disease. This article reviews the phylogeny and the ontogeny of macrophages with a particular focus on the gastrointestinal tract, and the role of these mucosal macrophages in immune surveillance, innate immunity, homeostasis, tissue remodeling, angiogenesis, and repair of damaged tissues. We also discuss the importance of these macrophages in the inflammatory changes in neonatal necrotizing enterocolitis (NEC). This article presents a combination of our own peer-reviewed clinical and preclinical studies, with an extensive review of the literature using the databases PubMed, EMBASE, and Scopus. Macrophages were first described at the beginning of the previous century by Paul Ehrlich and Ilya Metchnikoff as important mediators of innate immunity.1 The name "macrophages" or "big eaters" came from the Greek words, "makros" or large, and "phagein" or eat.2 Macrophages are large cells with an irregular cell shape, oval- or kidney-shaped nucleus, cytoplasmic vesicles, central nucleus, and high cytoplasm-to-nucleus ratio.3 These cells are highly phagocytic and motile, and modulate immune responses by releasing various mediators.4 The term mononuclear phagocyte includes lineage-committed bone marrow precursors, circulating monocytes, resident macrophages, and dendritic cells (DCs).5 In this review, we have focused on the macrophage lineage as it expands and matures first, in utero, and plays an important role in the innate immune responses of newborn infants.
... There is some hypothetical mechanism for the pathogenesis of TANEC such as immune mechanisms as responsible for the acute mucosal injury of the intestine by passive transfusion of biological response mediators from donor or activation of T-cell antigens of red blood cells (9). The other effective factors consist of the age of the blood products, the severity of anemia in the neonate, and the low levels of nitric oxides in stored red blood cells resulted in the vasoconstriction of the lower mesenteric vessels and decreased blood ow and resulted in mucosal injury (7). ...
Preprint
Full-text available
Background: The correlation between necrotizing enteroculitis (NEC) and packed cell transfusion (PCT) has recently been identified. Based on some research, 25–35% of NEC has been linked to transfusion, we planned this study to determine, the association between PCT and feeding tolerance in well preterm newborns. Method: Our study was a clinical trial study in preterm infants admitted to NICU of Mofid Children's Hospital from April 2017 to May 2018.Seventy well premature babies with a birth weight of <1500 grams and gestational age <32 weeks with enteral feeding, who need PCT were enrolled. The eligible patients divided by simple randomization to two groups, in the intervention group (35 patients) the baby's breastfeeding withholding just during the time of PCT and continue as usual after that, but in control groups (35 patients) feeding of neonates is given as usual regardless of PCT. The feeding tolerance during the first 72 hours after transfusion was compared between the two groups. Sick neonates exclude from the study. Data analysis was performed in SPSS version 20. Results: The mean gestational age, birth weights, and postnatal age in the intervention group were 30.13 weeks, 1245.71grams, and 17 days respectively and in the control group were 29.97weeks, 1169.43grams and 15.46 days respectively without any statistically significant difference between them. Except for hemoglobin and hematocrit pre-transfusion, other characteristics of patients were similar. In the evaluation of feeding tolerance after transfusion during 24, 48 and 72 hours, 32(91.2%), 33(94.73%), 34(97.1%) of both groups, had feeding tolerance with no significant difference There were no statistically significant differences between neonates with and without the feeding tolerance in the patients of each group. Conclusion: Our research showed that in well preterm neonates with a good general condition, during PCT, withholding of feeding, isn’t necessary and continued breastfeeding seems to be safe. Trial registration: All ethical considerations of the study were approved by the institutional review board and the research ethics committee at Shahid Beheshti University of Medical Sciences, Tehran, Iran (IR. SBMU.RETECH.1395.1010) and granted ethical approval and the Iranian Registry of Clinical Trial code are IRCT20200419047136N1. Approved by Iranian Registry of Clinical Trials Trial Id: 47347 IRCT Id: IRCT20200419047136N1 Registration date: 2020-05-04, 1399/02/15 (The link directly for trial registration: https://en.irct.ir/trial/47347) Approved by Research Ethics Committee: IR. SBMU.RETECH.1395.1010
Article
Full-text available
Antecedentes: La enterocolitis necrotizante (ECN) es un trastorno intestinal grave que afecta principalmente a los recién nacidos prematuros o gravemente enfermos. Se caracteriza por necrosis de la mucosa intestinal, que puede dar lugar a complicaciones graves. Reporte de caso: Este estudio de caso se trata de un recién nacido a término con varios factores de riesgo para desarrollar ECN, entre ellos bajo peso al nacer y alto riesgo de transmisión vertical del VIH por infección de la madre. La madre, de 36 años, tenía antecedentes de VIH, pero no recibió terapia antirretroviral durante el embarazo. Además, solo asistió a dos controles prenatales durante todo el embarazo. El bebé nació por cesárea debido a la enfermedad infecciosa de la madre y la presentación de nalgas. Después de ingresar al hospital, el bebé recibió fórmula especial para bebés prematuros, pero al octavo día de alimentación enteral, el bebé desarrolló signos de ECN. Conclusiones: Es importante señalar que los tratamientos con zidovudina en recién nacidos aumentan el riesgo de desarrollar ECN, especialmente si se administran por vía oral. Las evaluaciones clínicas y radiológicas periódicas son críticas para los pacientes diagnosticados con ECN, y se debe brindar un manejo médico oportuno y adecuado para cada caso.
Article
Establishing full enteral nutrition in critically ill preterm infants with immature gastrointestinal function is challenging. In this article, we will summarize emerging clinical evidence from randomized clinical trials suggesting the feasibility and efficacy of feeding interventions targeting the early establishment of full enteral nutrition. We will also examine trial outcomes of higher volume feedings after the establishment of full enteral nutrition. Only data from randomized clinical trials will be discussed extensively. Future opportunities for clinical research will also be presented.
Article
Background: Necrotizing enterocolitis (NEC) and postnatal growth restriction are significant clinical dilemmas that contribute to short- and long-term morbidities for the most premature infants. Purpose: After a rise in NEC rates in a regional neonatal intensive care unit (NICU), improvement practices were implemented by an interdisciplinary quality improvement (QI) work group whose focus was initially on nutrition and growth. QI work was refocused to address both NEC and growth concurrently. Methods: Through various QI initiatives and with evolving understanding of NEC and nutrition, the work group identified and implemented multiple practices changes over 2-decade time span. A standardized tool was used to review each case of NEC and outcomes were continually tracked to guide QI initiatives. Local findings: Focused QI work contributed to a significant reduction in NEC rates from 16.2% in 2007 to 0% in 2018 for inborn infants. Exclusive human milk diet was a critical part of the success. Postnatal growth outcomes initially declined after initial NEC improvement work. Improvement work that focused jointly on NEC and nutrition resulted in improved growth outcomes without impacting NEC. Implications for practice: Use of historical perspective along with evolving scientific understanding can guide local improvement initiatives. Work must continue to optimize lactation during NICU hospitalization. More research is needed to determine impact of care practices on gastrointestinal inflammation including medication osmolality, probiotics, and noninvasive respiratory support.
Article
Full-text available
Background: Recently, the correlation between necrotizing enterocolitis (NEC) and packed cell transfusion (PCT) has been identified. Evidence shows that 25-35% of NEC cases are associated with PCT. Objectives: this study aimed to determine the association between PCT and feeding tolerance in healthy preterm newborns. Methods: Materials and Methods: This clinical trial was performed on preterm infants, admitted to the neonatal intensive care unit (NICU) of Mofid Children's Hospital, Tehran, Iran, from April 2017 to May 2018. A total of 70 healthy premature infants (birth weight < 1500 g and gestational age < 32 weeks) with enteral feeding, who required PCT, were included in this study. The eligible infants were divided into two groups by block randomization. In the intervention group (n = 35), breastfeeding was withheld only during PCT and then continued as usual. On the other hand, in the control group (n = 35), feeding was performed as usual, regardless of PCT. Feeding tolerance within the first 72 hours post-transfusion was compared between the two groups. Sick newborns were excluded from the study. Data analysis was performed in SPSS version 20. Results: The mean gestational age, birth weight, and postnatal age of the neonates were 30.13 weeks, 1245.71 g, and 17 days in the intervention group and 29.97 weeks, 1169.43 g, and 15.46 days in the control group, respectively; there was no significant difference between the two groups. Except for pre-transfusion hemoglobin and hematocrit levels, other characteristics of the two groups were similar. Feeding tolerance was reported in 32 (91.2%), 33 (94.73%), and 34 (97.1%) newborns at 24, 48, and 72 hours post-transfusion in both groups, without any significant difference. There was no significant difference between neonates with and without feeding tolerance in either of the groups. Conclusions: According to the present results, withholding feeding during PCT is not necessary in healthy preterm neonates with a good general condition, and continued breastfeeding seems to be a safe option.
Article
In the clinic of the Volga Research Medical University on the basis of Nizhny Novgorod Regional Children’s Clinical Hospital” and “Children’s City Clinical Hospital № 1” from 2014 to 2018 82 newborns including 53 boys (65%) and 29 girls (35%) at the various stages of necrotizing enterocolitis were observed. There were 15 full-term infants (18%), and 67 premature newborns (82%). Symptoms of the disease were manifested in terms from 1 day to 25 days of life. During hospitalization, children were examined by neonatologists and hospital surgeons with the following by instrumental examinations (radiography, ultrasound). The severity of the condition and severity of the process were determined accordingly to the classification by to M.J. Bell (1978). The correlation between the clinical and epidemiological characteristics of newborns and the development of the pathological process was studied using the coefficient of conjugation of characters (φ), on the Wilcoxon, Van der Warden criteria and the sign criterion. Using mathematical analysis, conditionally “weak”, “medium” and “strong” risk factors for the development of NEC are identified. When assessing the occurrence of predictors in different stages of the disease, somatic diseases of the mother, chronic fetal hypoxia, fetal infections (IUI), chronic placental insufficiency (HFPN), weakness of labor, a body weight of the child <1500g were noted to be most often detected
Chapter
We now have an increased understanding of the genetics, cell biology, and physiology of electrolyte transport processes in the mammalian intestine, due to the availability of sophisticated methodologies ranging from genome wide association studies to CRISPR‐CAS technology, stem cell‐derived organoids, 3D microscopy, electron cryomicroscopy, single cell RNA sequencing, transgenic methodologies, and tools to manipulate cellular processes at a molecular level. This knowledge has simultaneously underscored the complexity of biological systems and the interdependence of multiple regulatory systems. In addition to the plethora of mammalian neurohumoral factors and their cross talk, advances in pyrosequencing and metagenomic analyses have highlighted the relevance of the microbiome to intestinal regulation. This article provides an overview of our current understanding of electrolyte transport processes in the small and large intestine, their regulation in health and how dysregulation at multiple levels can result in disease. Intestinal electrolyte transport is a balance of ion secretory and ion absorptive processes, all exquisitely dependent on the basolateral Na⁺/K⁺ ATPase; when this balance goes awry, it can result in diarrhea or in constipation. The key transporters involved in secretion are the apical membrane Cl⁻ channels and the basolateral Na⁺‐K⁺‐2Cl⁻ cotransporter, NKCC1 and K⁺ channels. Absorption chiefly involves apical membrane Na⁺/H⁺ exchangers and Cl⁻/HCO3⁻ exchangers in the small intestine and proximal colon and Na⁺ channels in the distal colon. Key examples of our current understanding of infectious, inflammatory, and genetic diarrheal diseases and of constipation are provided. © 2019 American Physiological Society. Compr Physiol 9:947‐1023, 2019.
Article
Full-text available
Necrotising enterocolitis (NEC) is one of the leading causes of neonatal morbidity, mortality and surgical emergencies. As the survival rate of extremely low birth weight (ELBW) infants is rising, so is the risk of NEC. The aim of this study was to compare diagnostics parameters like clinical and radiological findings and laboratory indicators and the treatment and outcome of NEC patients from 2000 till 2007 (Group 1) and from 2008 till 2016 (Group 2) treated in Neonatology Clinic (NC) of Children’s Clinical University Hospital (CCUH). In the rectrospective study, 277 newborns were divided among Group I and Group II – 105 and 172 patients, respectively. There were no statistically significant differences between both study groups in mean gestational age and birth weight. In both groups the first signs of NEC appeared on average eight days after birth. Differences in the diagnostic method used in both groups were not statistically significant; specific radiological findings were seen in approximately 1/3 of the cases. There were statistically significant differences in the management of NEC and patient mortality. Conservative therapy was applied in 70.0% of patients in both study groups. Over time, peritoneal drainage (PPD) as the sole surgical treatment decreased by 6.4%, but PPD with following enterostomy decreased by 8.9%. In Group 2 mortality of NEC patients decreased by 17.4%. Mortality among surgically treated NEC patients decreased as well, by 9.0%.
Article
Full-text available
Reply to questions raised regarding my review article entitled: NECROTIZING ENTEROCOLITIS IN THE FULL-TERM NEONATE (DOI: 10.1046/j.1440-1754.2001.00584.x)
Article
Background: Immaturity, ischemia, and disturbances in gut mucosal integrity due to infections or hyperosmolar feeds are some of the suspected mechanisms in the development of necrotising enterocolitis (NEC) in preterm infants. Decreased concentration of nitric oxide is proposed as one of the possible cellular mechanisms for NEC. Plasma arginine concentrations were found to be lower in infants who developed NEC. Arginine can act as a substrate for the production of nitric oxide in the tissues and arginine supplementation may help in preventing NEC. Objectives: This review examines the efficacy and safety of arginine supplementation in decreasing the incidence of NEC among preterm neonates. Search strategy: A literature search was performed using the following databases: MEDLINE (1966 - June 2004), EMBASE (1980 - June 2004), CINAHL (1982 - June 2004), Cochrane Controlled Trials Register (Issue 2, 2004 of Cochrane Library) and abstracts from the annual meetings of the Society for Pediatric Research, American Pediatric Society and Pediatric Academic Societies published in Pediatric Research (1991-2004). No language restrictions were applied. Selection criteria: Study design: randomized or quasi-randomized controlled trials. Population: preterm neonates. Intervention: enteral or parenteral arginine supplementation (in addition to what an infant may be receiving from enteral or parenteral source), compared to placebo or no treatment; arginine administered orally or parenterally for at least 7 days in order to achieve adequate plasma arginine levels (145 umol/l). Outcomes: any of the following outcomes - NEC, death prior to discharge, death due to NEC, surgery for NEC, duration of total parenteral nutrition, plasma concentrations of arginine and glutamine at baseline and seven days after intervention, side effects of arginine. Data collection and analysis: The methodological quality of the trials was assessed using the information provided in the studies and by personal communication with the author. Data on relevant outcomes were extracted and the effect size was estimated and reported as relative risk (RR), risk difference (RD) and mean difference (MD) as appropriate. Main results: Only one eligible study was identified. The methodological quality of the included study was good. There was a statistically significant reduction in the risk of developing NEC (any stage) in the arginine group compared with the placebo group (RR 0.24 [95% CI 0.10, 0.61], RD -0.21 [95% CI -0.32, -0.09]). No significant side effects directly attributable to arginine were observed. Reviewers' conclusions: The data are insufficient at present to support a practice recommendation. A multicentre randomized controlled study of arginine supplementation in preterm neonates is needed, focusing on the incidence of NEC, particularly stage 2 or 3.