Neonatal Necrotizing Enterocolitis




Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in the neonatal intensive care unit (NICU). It is primarily a disease of prematurity, with only 10% of affected infants born after 36 weeks of gestation. NEC contributes significantly to short- and long-term morbidity as well as mortality of preterm infants. It is estimated that between 14% and 43% of all causes of intestinal failure are the result of sequelae of NEC. Although the pathogenesis of the disease was first described more than 30 years ago, the mortality rate for NEC has decreased minimally. Despite decades of vigorous research efforts, many unanswered questions remain. A number of potential contributing factors have been identified; however, the precise etiology of this multifactorial, complex disease process remains elusive. Preventive measures have been of limited success, and therapy is mostly supportive, consisting of medical stabilization and efforts to prevent progression of the disease.


Epidemiology


Despite significant advances in neonatal intensive care, necrotizing enterocolitis is associated with high morbidity and mortality. It is estimated that $5 billion is spent annually in the care of patients with NEC and the morbidity associated with it.


NEC affects about 1% to 5% of all newborns admitted to NICUs, with an incidence of 7% to 14% in very low-birth-weight infants. It is noteworthy that a significant intercenter variability has been reported. The only consistent epidemiologic precursors of NEC are prematurity and enteral alimentation, although other risk factors have been identified. According to the data from the Vermont-Oxford Network encompassing about two-thirds of all extremely low-birth-weight (ELBW) infants born in the United States, the risk of NEC increases with lower gestational age and is as high as 12% in ELBW (501 to 750 g) compared to 3.3% in infants with birth weights between 1251 and 1500 g. Because the number of ELBW infants treated in NICUs is relatively small, the majority of cases occur in the category of 30 to 32 weeks of gestation. Data from the National Institute of Child Health and Human Development (NICHD) Neonatal Research Network in three time periods from 1987 to 2000 show no change over time, with an incidence of 6% to 7% among infants with a birth weight of less than 1500 g. There is no consistent association between gender, socioeconomic status, or seasonal variability and the occurrence of necrotizing enterocolitis.


NEC is primarily a disease of the convalescent preterm infant with a peak incidence at 2 to 3 weeks postnatal age when the newborn has recovered from the acute period and is enterally fed. Earlier initiation of enteral feedings is associated with an earlier onset of NEC, which may account for the fact that NEC occurs significantly earlier in more mature infants than in extremely preterm infants, as illustrated in Figure 45-1 . In infants who have never been fed, NEC may be associated with maternal chorioamnionitis and intrauterine exposure to cytokines. In general, cases of NEC are sporadic, although some centers have reported episodic outbreaks that occur more commonly in crowded nurseries. Most affected newborns are preterm (PT) infants; however, 10% of cases occur in full-term (FT) babies. Full-term infants tend to develop the disease earlier, at a mean age of younger than 5 days, and are more likely to have predisposing factors. Some of the risk factors implicated include a history of abnormal antenatal Doppler studies, gastroschisis, perinatal hypoxia, multiple gestation, history of umbilical artery catheter, polycythemia, sepsis, and congenital heart disease, all of which could potentially compromise intestinal blood flow. In a retrospective chart review by Ostlie et al., predisposing factors were lacking in 38% of NEC cases in FT infants, although most cases occur in the ICU setting. Term infants who develop NEC are generally formula fed, and the need for surgical intervention or survival does not seem to differ significantly from that of preterm infants.




Figure 45-1


Necrotizing enterocolitis in infants over a 3-year period. The horizontal axis shows the number of infants at increasing gestational ages. The vertical axis shows the postnatal age of onset of necrotizing enterocolitis. This graph demonstrates that there is an inverse relationship between age of onset and gestational age.

(From Neu J, Bernstein H. Update on host defense and immunonutrients. Clin Perinatol 2002; 29 (1):41–64. with permission.)


The reported incidence of NEC in infants with congenital heart disease is 3.3% to 6.8%, which is 10- to 100-fold higher than rates described for the entire late preterm/term newborn population. In infants with congenital heart disease, NEC is generally associated with anomalies that result in compromised mesenteric blood flow, such as left-sided obstructive lesions or single ventricle physiology.


Episodes of low cardiac output, shock, and cardiopulmonary bypass also represent significant risk factors. Independent of the anatomic defect, the presence of a left-to-right shunt at the level of the ductus arteriosus with a persistent diastolic flow reversal in the descending aorta (diastolic steal) and mesenteric hypoperfusion are postulated as the main pathogenic mechanisms. The presence of an atrial septal defect (ASD) or a ventricular septal defect (VSD), both left-to-right intracardiac shunts, was also associated with a significant increase in NEC in a large cohort of preterm infants with a birth weight less than 1500 g at birth. In a retrospective review, NEC was not associated with a particular feeding pattern, the overall mortality for patients with congenital heart disease and NEC was 24.4%.


In recent years, epidemiologic data have emerged, suggesting that interventions and therapies frequently used in the care of preterm infants may also be associated with an increased incidence of NEC. Particularly, the administration of prolonged courses of antibiotics and H2 receptor antagonists, which result in altered microbial colonization and selection of pathogenic organisms, seem to have a detrimental effect, potentially increasing long-term morbidity and mortality.


The first case of transfusion-associated NEC was published in 2006. Since then, there have been multiple reports on the potential association of NEC and blood transfusions, which prompted the term “transfusion-associated NEC” defined by an antecedent packed red blood cells (PRBC) transfusion within 48 hours before onset of symptoms. The mechanism for this association remains unclear. Transfusion-related acute gut injury, or TRAGI, has been proposed as a final pathway leading to NEC in this subset of patients. A decrease in the incidence of transfusion-associated NEC was observed after the implementation of a protocol that includes withholding enteral feeds during the time of transfusion.


A potential association of respiratory support with continuous positive airway pressure (CPAP) and NEC has not been confirmed.




Pathology


The characteristic gross pathologic features of NEC are signs of ischemia and patchy necrosis, typically of the distal small intestine as well as the hepatic and splenic flexures of the colon ( Figure 45-2 ). Involvement of both the small and large intestine is present in about 44% of cases, whereas 26% have only colonic compromise, and in 30%, lesions are limited to small intestine. The severity of bowel wall necrosis ranges from a small localized mucosal necrosis of a bowel segment to transmural necrosis of the entire small intestine and colon. In cases of fulminant presentation, the entire gastrointestinal tract may be involved, which is referred to as NEC totalis. Other pathologic findings include the presence of gastrointestinal bleeding, peritonitis, intestinal distension, pneumatosis intestinalis, and portal venous gas. Intraabdominal fluid collection or a pneumoperitoneum, the latter indicating the presence of intestinal perforation, may also be found.




Figure 45-2


Intraoperative photograph of necrotizing enterocolitis (NEC) demonstrating dilated bowel loops with areas of pneumatosis, bowel wall ischemia, and necrosis.

(Courtesy J. DiFiore, MD, and O. Soldes, MD.)


Classical microscopic features ( Figure 45-3 ) include areas of ulceration of the mucosa and submucosa in association with full-thickness necrosis, thrombosis of blood vessels, and a prominent influx of inflammatory cells (macrophages, neutrophils, and monocytes) into the submucosal layer. Coagulation necrosis, suggestive of an ischemic insult, is generally the predominant finding. Inflammatory changes and evidence of intestinal cell apoptosis are also characteristic microscopic features. Of interest, in a series of pathologic observations, reparative changes including epithelial regeneration and granulation tissue fibrosis were reported in two-thirds of cases. In situ hybridization studies in intestinal tissue from NEC patients demonstrated increased matrix metalloproteinase (MMP) messenger RNA (mRNA) expression, indicating an upregulation of many members of the MMP family including MMP1, 3 (stromelysin-1), 7, 12, and 24. These zinc-dependent endopeptidases capable of degrading extracellular matrix have been shown to play a key role in tissue damage. In conjunction with stromelysin upregulation, an increased transcription of TIMP-1, a natural tissue inhibitor of metalloproteinases, was also described, suggesting an active feedback loop to counteract ongoing tissue destruction. Interleukin (IL)-1β and tumor necrosis factor α (TNF)α, both important inflammatory mediators in NEC, stimulate the production of MMPs, whereas TNFα downregulates TIMP-1. These data indicate that TNFα plays a crucial role in tissue destruction in patients with NEC. In pathologic specimens of patients undergoing intestinal resection for NEC, intestinal cell apoptosis and expression of the inducible nitric oxide synthase (iNOS) have been demonstrated, as well as increased tissue transcripts for TNFα, IL-8, and IL-11, and decreased interferon γ (IFN)γ.




Figure 45-3


Microscopic evidence of severe submucosal gas-filled cysts (pneumatosis intestinalis) in a preterm infant with necrotizing enterocolitis (NEC). Note the marked hemorrhage throughout the bowel wall and the inflammatory exudate on the surface of the mucosa. Hematoxylin and eosin stain, ×56.

(Courtesy Beverly Dahms, MD.)




Pathogenesis


The medical syndrome of NEC is well recognized; however, its pathogenesis remains poorly understood. Because of the multifactorial origin of the disease and the inherent limitations in experimental models, attempts to develop early diagnostic tools and preventive measures have been largely unsuccessful. Several animal models for the study of NEC have been developed. These experimental models yield pathologic and cytokine profiles similar to those observed in human newborns; however, they fail to reproduce the intrinsic and complex aspects of prematurity, thus limiting to some degree the extrapolation of results to clinical NEC. The roles of factors involved in regulation of epithelial function, cytokine production, bacterial colonization, protective and toxic effects of nitric oxide (NO), intestinal mucosal restitution, and immaturity of protective mechanisms as contributors to the initiation and/or progression of NEC continue to be explored. The influence of genetics on the individual susceptibility to the development of NEC is still emerging. Recent studies have shown that a carrier state of genetic polymorphisms may be associated with perinatal morbidity, including NEC. The identification of a genetic marker with high sensitivity and specificity for the prediction of NEC could potentially lead to the implementation of effective preventive measures.


A generally accepted hypothesis of the pathophysiology of NEC involves an initial insult, such as ischemia, contributing to loss of intestinal barrier integrity. Once formula feedings are introduced, they serve as substrate for bacterial proliferation. Pathogenic enteric organisms then invade the injured mucosa, thereby promoting the production of proinflammatory cytokines. Infants, particularly those born prematurely, may have a reduced ability to counterregulate this surge in proinflammatory mediators, leading to additional injury, eventual breach of the mucosal barrier, and bacterial translocation. A systemic inflammatory reaction syndrome (SIRS) results in a clinical picture of septic shock.


Ischemic Injury


Ischemic injury is thought to play a central role in the cascade of events leading to the development of NEC. Several investigators have used animal models of NEC that rely on the induction of intermittent ischemia and reperfusion of the intestine, typically through temporary occlusion of the blood supply to the small intestine. Although such models may provide valuable information regarding the biology of intestinal ischemia reperfusion injury, it is noteworthy that a clear ischemic–reperfusion injury is often absent or very remote in patients in whom NEC develops during the convalescence phase of their hospitalization.


Neonatal asphyxia, recurrent episodes of hypoxia–bradycardia, systemic arterial hypotension, patent ductus arteriosus, congenital heart disease, and polycythemia are some of the mechanisms that may lead to intestinal ischemia. In states of hypoperfusion, the “diving reflex” is activated as a protective mechanism to ensure adequate blood supply to vital organs, including the brain and the heart, while shunting blood from the splanchnic circulation, thus worsening gut ischemia. Ischemic injury of the intestinal mucosa ensues, followed by activation of the inflammatory cascade, reperfusion injury, and gut barrier dysfunction with bacterial translocation and the systemic effects associated with it. Basal intestinal vascular resistance is maintained by a dynamic balance between constrictor (endothelin-1 [ET-1]) and dilator (NO) stimuli, and in newborns, this balance favors NO-dependent vasodilation, leading to a low vascular resistance ensuring adequate blood flow to the rapidly growing intestine. Disruption of intestinal endothelial cell function could alter this balance favoring ET-1-dependent vasoconstriction and thus lead to significant intestinal ischemia. Doppler studies of the superior mesenteric artery in premature infants who later developed NEC demonstrated a high resistance pattern suggestive of abnormal intestinal blood flow. Decreased resistance to blood flow was found in patients with established NEC. It has been postulated that this apparent discrepancy may actually represent a biphasic response of the mesenteric vessels, with an initial phase (high resistance to flow/ischemic injury), followed by rebound hyperemia (low resistance to flow/reperfusion injury), due to the release of inflammatory mediators. It is possible that alterations in the autoregulation of intestinal blood flow, mediated by a balance between vasoconstrictor and vasodilator molecules, play a role in the development and progression of NEC. In addition, intestinal tissue removed from infants with NEC demonstrates abnormalities in expression and regulation of these important vasoactive mediators.


Gut Barrier


In mature animals, the intestine has many physical barriers to bacteria, including peristalsis, gastric acidity, proteolytic enzymes, intestinal mucus, cell-surface gly­coconjugates, and tight junctions between intestinal epithelial cells. These protective mechanisms limit the bacterial microenvironment to the gut lumen and prevent attachment and translocation across the intestinal epithelium. However, experimental data demonstrate that pathogenic organisms adhere to and translocate across the intestine to a greater extent in immature compared to mature animals. Abnormal peristaltic activity and hypomotility in immature infants may increase bacterial adherence, allowing for bacterial overgrowth. In the NICU, the use of narcotics for sedation and pain management may accentuate this phenomenon. In preterm infants, the repertoire of cell-surface glycoconjugates, which serve as adhesion sites for a variety of microbes, have a different pattern of carbohydrate residues. This functional difference may in part account for differences in bacterial colonization patterns in these infants. Colonization of the intestine by commensal microorganisms is a key step in intestinal maturation and maintenance of the gut barrier. Alternatively, colonization with pathogenic organisms may trigger an inappropriate reaction by the immature intestine with activation of the inflammatory cascade, which promotes disruption of the gut barrier resulting in NEC. Human defensins (or cryptidins) produced and secreted from Paneth cells protect against bacterial translocation; however, this mechanism is impaired in premature infants, particularly those with NEC. Trefoil factor peptides (TFFs, 1 to 3) are part of the protective mechanism operating in the intestinal mucosa and play a fundamental role in epithelial protection, repair, and restitution. These secreted peptides have been identified in a site-specific pattern in the gastrointestinal mucosa, and their expression has been shown to be upregulated in early stages of mucosal repair. A study by Lin et al. demonstrated that TFF3 mRNA and protein expression are deficient in immature rats. Impaired mucosal regeneration in part due to failure of upregulation of TFF peptide expression may contribute to the pathogenesis of NEC. Tight junctions between epithelial cells maintain the semipermeable properties of the intestinal epithelium, limiting the passage of bacteria and other macromolecules. Immaturity in the composition and function of the tight junctions through the interactions of structural proteins (claudins and occludins) may explain the increased permeability of the immature intestine. Furthermore, changes in expression of genes coding for these structural proteins have been described during the lesional and reparative phases of NEC.


In premature infants, there is immaturity of the functional barrier that limits growth of bacteria, the immunologic host–defense mechanism, and various biochemical factors involved in gut barrier protection. It is known that the numbers of intestinal B and T lymphocytes are decreased in neonates and do not approach adult levels until 3 to 4 weeks of life. Newborns also have reduced levels of secretory immunoglobulin A (IgA) in salivary samples, presumably reflecting decreased activity in the intestine. Furthermore, the intestinal lamina propria is devoid of IgA-secreting cells, rendering the newborn more susceptible to infection on the mucosal surface.


Bacterial Colonization


The role of bacterial colonization in the pathogenesis of NEC has long been recognized. In utero, the intestinal tract is free of bacteria, but it becomes rapidly colonized during the first days of life. Mode of delivery, treatment with antibiotics, and type of enteral feedings affect the timing and patterns of colonization. Infants delivered vaginally have earlier colonization with both Bifidobacterium and Lactobacillus , whereas infants delivered by cesarean section can have colonization with these beneficial organisms delayed by up to 30 days. After birth and during the first week of life, the normal breast-fed newborn gut is colonized by predominantly protective anaerobe bacteria including lactobacilli and bifidobacteria. In formula-fed infants, similar numbers of Bacteroides and Bifidobacterium are found with minor components of the more pathogenic Staphylococcus , Escherichia coli , and Clostridium . In contrast, the preterm infant’s gut is exposed to a flora with a predominance of potentially pathogenic gram-negative species, with a lesser degree of bacterial diversity and fewer protective anaerobe species. The use of broad-spectrum antibiotics during the immediate neonatal period may select a population of resistant organisms, thereby aggravating this situation. In the presence of other risk factors, alteration of the intestinal microbiome may set the stage for the later development of NEC. Despite the description of outbreaks of NEC in some nurseries, no single specific organism can be identified as the etiologic agent. Klebsiella , E. coli, Clostridium species, and Staphylococcus epidermidis, as well as a variety of viruses and fungal species, have all been isolated from patients with NEC. Particular interest has been placed on Enterobacter sakazakii (ES), which can cause sepsis and meningitis in low-birth-weight infants and has been identified in the hospital environment in association with infant bottle brushes and food preparation equipment. Ingestion of ES-contaminated infant formula has been implicated in cases of necrotizing enterocolitis. In vitro and in vivo studies suggest that ES adheres to the enterocyte and promotes cell necrosis and apoptosis in a dose-dependent manner. In 2002, the U.S. Food and Drug Administration (FDA) published a warning regarding the presence of ES in baby formula.


Feedings and NEC


The contribution of different feeding practices to the pathogenesis of NEC has been extensively studied. Timing of introduction of feeds, type, volume, and rate of advance have all been the subject of clinical studies. Experimental data suggest that introduction of feeds is important to establish intestinal barrier function. In a rat model, starvation reduces gut mucosal barrier function; conversely, early feeds improved intestinal mucosal growth in piglets. It has been shown that infants who were never fed and developed NEC were more likely to rapidly progress to severe disease. It is possible that the early introduction of breast-milk feedings prepares the intestinal barrier to better respond to noxious stress later on.


In several randomized controlled trials, rapid advancement of enteral feeds was associated with a shorter time to full feeds and more rapid attainment of birth weight without an increased incidence of NEC. These results are tempered by another trial where rapid progression of enteral feeds was associated with NEC. There have been several Cochrane reviews addressing timing of feeding introduction, rate of feeding advancements, and enteral fasting versus introduction of early trophic feeds. None of those interventions seems to decrease the incidence of NEC, whereas more conservative feeding schedules result in delays to achieve full feeds and to regain birth weight. Breast milk or donor breast milk is preferred for initiation of enteral alimentation for preterm infants when available. A protocol of enteral feedings containing at least 50% of human milk in the first 14 days of life was associated with a sixfold decrease in the incidence of NEC. Of interest, the benefits of breast-milk feedings may follow a dose-related beneficial effect on the risk of NEC and mortality. In addition to the well-known immunologic properties in breast milk, the presence of other factors such as epithelial growth factor (EGF), erythropoietin, insulin-like growth factor (IGF), and the anti-inflammatory cytokine IL-10 may account for its protective effects. The currently available data support the promotion of breast milk as the initial nutrient of choice for the premature infant. It is well known that feeding practices vary among centers, but in general, judicious introduction of gut-priming feeds followed by a steady rate of advancement with close attention paid to feeding intolerance seems to be the preponderant approach. The use of standardized feeding protocols has been shown to be protective. The effects of other enterally administered supplements (e.g., vitamins, iron, fortifiers) on the incidence of NEC are unclear.


Abnormal Inflammatory Response


The modulation of the inflammatory response seems to be developmentally regulated, and it requires an appropriate balance between proinflammatory and anti-inflammatory signaling in order to maintain a healthy intestinal homeostatic environment.


The inability of the premature newborn to distinguish between and respond appropriately to commensal and pathogenic microorganisms and the aberrant regulation of other mediators such as platelet-activating factor (PAF) lead to a proinflammatory environment that could contribute to the pathology seen in NEC. Human fetal intestinal epithelial cells demonstrate an exaggerated production of inflammatory cytokines in response to pathogenic and commensal bacteria, as well as to endogenous inflammatory mediators such as TNFα and IL. Several proinflammatory cytokines that mediate inflammatory cell recruitment through activation and amplification of the immune response in local host defense have been implicated in NEC, including TNFα, IL-1β, IL-6, IL-8, IL-12, and IL-18. Anti-inflammatory cytokines modulate the host’s inflammatory response; a lack of upregulation of these molecules to counteract the effects of proinflammatory mediators results in more severe tissue injury. Both anti-inflammatory cytokines, IL-4 and IL-10, have been implicated in NEC.


Lipopolysaccharide (LPS), which is a principal component of the outer cell wall of gram-negative bacteria, recognizes and binds to Toll-like receptor 4 (TLR4). Circulating LPS is increased in patients with NEC, which inhibits epithelial restitution and initiates inflammatory signaling cascades within the enterocyte, including activation of the transcription factor nuclear factor (NF)-κB. NF-κB proteins activate transcription of a wide variety of genes important in inflammatory and immune responses. In the resting state, NF-κB dimers are bound in the cytoplasm to inhibitory κB (IκB) proteins. By activation of an IKK kinase, NF-κB is freed to translocate to the nucleus, where it triggers gene transcription. An autoregulatory feedback loop exists in which NF-κB activation leads to IκBa synthesis, which in turn can terminate the NF-κB response. However, it has been shown that immature enterocytes have increased NF-κB activity associated with decreased baseline expression of IκB isoforms. In addition, there is more rapid IκBa degradation and delayed resynthesis of IκBa in immature enterocytes. These data suggest that the increased inflammatory cytokine production in the immature intestine may result from the combined effect of increased NF-κB activity and translocation to the nucleus because of decreased IκB expression, and decreased inhibition of NF-κB due to an accelerated degradation and delayed resynthesis of IκBa.


The pathogenesis of NEC is complex, and our understanding of it is still incomplete. Multiple pathogenic mechanisms affecting the premature infant, such as perinatal ischemia, feedings and bacterial colonization, and inflammation, converge into a final common pathway leading to intestinal barrier failure, tissue damage, and organ system failure ( Figure 45-4 ). Increasing our understanding of the mechanisms involved in NEC through well-designed clinical and basic research will allow us to develop and implement interventions aimed at early diagnosis and prevention. Currently, introduction of enteral feedings with breast milk is the most readily available way to decrease the incidence of the disease.




Figure 45-4


The complex interplay of various pathogenic mechanisms affecting the premature infant and converging to a final common pathway leading to necrotizing enterocolitis (NEC).




Clinical Manifestations


The early stages of NEC present with nonspecific signs that may occur in an otherwise stable preterm infant or may represent alternative pathologies such as sepsis ( Box 45-1 ). Temperature instability, lethargy, and apneic spells may precede abdominal symptoms. The majority of preterm infants develop NEC during their convalescent phase, generally while on or close to full enteral feeds. A sudden increase in gastric residuals or episodes of emesis, which may be bilious or nonbilious, is suggestive of a disease process. The abdominal examination may reveal decreased or absent bowel sounds, distension, and tenderness. Although the presence of occult fecal blood is nonspecific, a grossly bloody stool in a high-risk infant is suggestive of necrotizing enterocolitis and is the most common presenting sign in term infants.



Box 45-1

Presentation of Necrotizing Enterocolitis


Clinical Signs and Symptoms





  • Apnea and bradycardia



  • Lethargy



  • Temperature instability (need to increase environmental temperature)



  • Abdominal distension/tenderness/guarding



  • Discoloration of abdominal skin



  • Bloody stools (grossly or occult)



  • Tachycardia



  • Hypotension



  • Poor perfusion



  • Respiratory failure



Laboratory Findings





  • Increased WBC count with or without left shift



  • Neutropenia



  • Coagulation abnormalities—DIC (↑ PT/PTT, ↓ Fibrinogen)



  • Anemia



  • Thrombocytopenia or thrombocytosis (less common)



  • Increased inflammatory markers (C-reactive protein)



  • Hyperglycemia



  • Electrolyte abnormalities/hyponatremia



  • Acidosis (metabolic and/or respiratory)



Radiographic Findings





  • Dilated bowel loops with “stacked” appearance



  • Thickened bowel walls



  • Fixed bowel loops



  • Pneumatosis intestinalis



  • Portal venous air



  • Free intraabdominal air



  • Ascites



  • Gasless abdomen



Intraoperative Findings





  • Pneumatosis intestinalis



  • Intestinal inflammation



  • Bowel ischemia



  • Bowel necrosis



  • Perforation



DIC, Disseminated intravascular coagulation; PT, prothrombin time; PTT, partial thromboplastin time; WBC, white blood cell count.



Abdominal distension may progress rapidly and be accompanied by abdominal skin discoloration. As the disease evolves, the infant may develop cardiovascular instability and respiratory failure. Endotracheal intubation and mechanical ventilation may be necessary because of either severe apnea or significant abdominal distension compromising pulmonary function. A significant increase in intraabdominal pressure may lead to decreased venous return to the heart and thus contribute to hypotension and tachycardia. The infant may develop features of SIRS with evidence of cardiac dysfunction and septic shock, culminating in multiorgan system failure and death. The severity and progression of the disease are variable. Approximately one-half of infants with NEC will recover fully with bowel rest and antibiotics, whereas up to 40% will develop severe disease requiring urgent surgical intervention. An observational cohort study by Moss et al. found that the median and mean time interval from the diagnosis of NEC to severe disease resulting in surgical intervention were 1 and 6 days, respectively. The highest risk for bowel perforation is during the first 72 hours after diagnosis. Furthermore, 63% of the patients required surgical intervention within the first 2 days and 80% within the first week. Symptoms of bowel perforation and peritonitis include marked abdominal distension with inability to ventilate, erythematous or blue discoloration of abdominal skin, and clinical deterioration. Laboratory findings consist of neutropenia or neutrophilia, thrombocytopenia, increased inflammatory markers (C-reactive protein), electrolyte imbalances, and abnormal coagulation panel consistent with disseminated intra­vascular coagulation (DIC) (see Box 45-1 ). A progressive decrease in absolute granulocyte counts and thrombocytopenia suggest increasing severity of disease. Persistent metabolic acidosis and refractory thrombocytopenia may indicate intestinal necrosis and the need for surgical exploration. Some patients will progress rapidly to shock, DIC, multisystem organ failure, and death, despite maximized medical and/or surgical therapy. Unfortunately, there are no early clinical or laboratory markers available to identify patients at risk for rapid progression.


To classify the severity of disease, Bell’s staging criteria as modified by Kliegman ( Table 45-1 ) may be used. Stage I is nonspecific; symptoms may be present with the sepsis syndrome without intestinal involvement. Stage II is NEC as diagnosed by the presence of pneumatosis intestinalis in addition to systemic symptoms with increasing severity as outlined previously. Stage IIIA is defined by severe involvement with cardiorespiratory instability and stage IIIB by the presence of bowel perforation. Bell’s classification has been used widely in research to stratify infants according to severity of disease.


Jul 24, 2019 | Posted by in GASTROENTEROLOGY | Comments Off on Neonatal Necrotizing Enterocolitis

Full access? Get Clinical Tree

Get Clinical Tree app for offline access