1
|
Dantes G, Keane OA, Do L, Rumbika S, Ellis NH, Dutreuil VL, He Z, Bhatia AM. Clinical Predictors of Spontaneous Intestinal Perforation vs Necrotizing Enterocolitis in Extremely and Very Low Birth Weight Neonates. J Pediatr Surg 2024; 59:161608. [PMID: 39033072 DOI: 10.1016/j.jpedsurg.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/07/2024] [Accepted: 06/24/2024] [Indexed: 07/23/2024]
Abstract
PURPOSE Spontaneous intestinal perforation (SIP) and necrotizing enterocolitis (NEC) are distinct disease processes associated with significant morbidity and mortality. Initial treatment, laparotomy (LP) versus peritoneal drainage (PD), is disease specific however it can be difficult to distinguish these diagnoses preoperatively. We investigated clinical characteristics associated with each diagnosis and constructed a scoring algorithm for accurate preoperative diagnosis. METHODS A cohort of extreme and very low birth weight (<1500 g) neonates surgically treated for SIP or NEC between 07/2004-09/2022 were reviewed. Clinical characteristics included gestational age (GA), birth weight (BW), feeding history, physical exam, and laboratory/radiological findings. Intraoperative diagnosis was used to determine SIP vs NEC. Pre-drain diagnosis was used for patients treated with PD only. RESULTS 338 neonates were managed for SIP (n = 269, 79.6%) vs NEC (n = 69, 20.4%). PD was definitive treatment in 146 (43.2%) patients and 75 (22.2%) patients were treated with upfront LP. Characteristics associated with SIP included younger GA, younger age at initial laparotomy or drainage (ALD), and history of trophic or no feeds. Multivariate logistic regression determined pneumatosis, abdominal wall erythema, higher ALD and history of feeds to be highly predictive of NEC. A 0-8-point scale was designed based on these characteristics with the area under the receiver operating characteristic curve of 0.819 (95% CI 0.756-0.882) for the diagnosis of NEC. A threshold score of 1.5 had a 95.2% specificity for NEC. CONCLUSION Utilizing clinical characteristics associated with SIP & NEC we developed a scoring system designed to assist surgeons accurately distinguish SIP vs NEC in neonates. TYPE OF STUDY Retrospective Chart Review. LEVEL OF EVIDENCE Level III.
Collapse
MESH Headings
- Humans
- Infant, Newborn
- Enterocolitis, Necrotizing/diagnosis
- Enterocolitis, Necrotizing/surgery
- Intestinal Perforation/etiology
- Intestinal Perforation/diagnosis
- Intestinal Perforation/surgery
- Infant, Very Low Birth Weight
- Male
- Female
- Retrospective Studies
- Drainage
- Diagnosis, Differential
- Infant, Premature, Diseases/diagnosis
- Infant, Premature, Diseases/surgery
- Algorithms
- Laparotomy
- Gestational Age
- Infant, Extremely Low Birth Weight
- Spontaneous Perforation/diagnosis
Collapse
Affiliation(s)
- Goeto Dantes
- Department of Surgery, Emory University, Atlanta, GA, USA.
| | - Olivia A Keane
- Department of Surgery, Emory University, Atlanta, GA, USA
| | - Louis Do
- Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Savanah Rumbika
- Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Nathaniel H Ellis
- Division of Pediatric Surgery, Department of Surgery, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Valerie L Dutreuil
- Emory Department of Pediatrics, Emory University, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Zhulin He
- Emory Department of Pediatrics, Emory University, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Amina M Bhatia
- Division of Pediatric Surgery, Department of Surgery, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| |
Collapse
|
2
|
Dantes G, Keane OA, Raikot S, Do L, Rumbika S, He Z, Bhatia AM. Necrotizing enterocolitis following spontaneous intestinal perforation in very low birth weight neonates. J Perinatol 2024:10.1038/s41372-024-02155-3. [PMID: 39448869 DOI: 10.1038/s41372-024-02155-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/21/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
PURPOSE Necrotizing enterocolitis (NEC) and spontaneous intestinal perforation (SIP) are severe gastrointestinal complications of prematurity. The clinical presentation and treatment of NEC and SIP (peritoneal drain vs laparotomy) can overlap; however, the pathogenesis is distinct. Therefore, a patient initially treated for SIP can subsequently develop NEC. This phenomenon has only been described in case reports, and no risk factor evaluation exists. We evaluate clinical characteristics, risk factors, and outcomes of patients treated for a distinct episode of NEC after SIP. METHODS We performed a retrospective review of very low birth weight (<1500 g) neonates who presented with pneumoperitoneum between 07/2004 and 09/2022. Data was obtained from two separate neonatal intensive care units that were part of the same institution. Patients with an initial preoperative, intraoperative, or pathological diagnosis of NEC were excluded. Patients with an intraoperative diagnosis of SIP or preoperative diagnosis of SIP successfully treated with a peritoneal drain (PD) were evaluated. Patients subsequently treated (medically or surgically) for NEC after SIP were then compared to SIP-alone patients. Clinical characteristics included demographics, gestational age (GA), birth weight (BW), perinatal risk factors (chorioamnionitis, steroids, indomethacin), postoperative feeding regimen, and length of stay (LOS) were compared. RESULTS Of the 278 patients included, 31 (11.2%) patients had NEC after SIP. There was no difference in GA (25 weeks vs 25 weeks, p = 0.933) or BW (760 g vs 735 g, p = 0.370) between NEC after SIP vs SIP alone cohorts, respectively. Twenty (64%) of NEC after-SIP patients were previously treated with LP. NEC after SIP occurred with a median onset of 56 days. Pneumatosis was the most frequent (81%) presenting symptom and 12 (39%) patients had hematochezia. Four (12.9%) patients required LP for NEC and all had NEC intraoperatively and on pathology. A majority (77.4%) of patients were on breast milk (BM) at time of NEC diagnosis. NEC after SIP patients had lower maternal age at delivery (29.0 vs 25.0, p = 0.055) and the incidence of NEC after LP (primary or failed drain) was higher than PD alone (16.7% vs 6.2%, p = 0.007). NEC after SIP patients had longer LOS (135 vs 81, p < 0.001). CONCLUSION We report an 11.2% incidence of NEC at a median of 56 days following successful treatment of SIP, resulting in increased LOS. SIP patients are a high-risk cohort and protocols to prevent this phenomenon should be investigated.
Collapse
Affiliation(s)
- Goeto Dantes
- Department of Surgery, Emory University, Atlanta, GA, USA.
| | - Olivia A Keane
- Department of Surgery, Emory University, Atlanta, GA, USA
| | - Swathi Raikot
- Division of Pediatric Surgery, Department of Surgery, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Louis Do
- Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Savanah Rumbika
- Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Zhulin He
- Pediatric Biostatistics Core, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Amina M Bhatia
- Division of Pediatric Surgery, Department of Surgery, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| |
Collapse
|
3
|
Khattab M, Donnelly D, Blizzard A, Chabra S, David E, Stumpf KA, Nayak SP. Resuscitation strategies for surgical patients in the neonatal intensive care unit. Semin Perinatol 2024:151988. [PMID: 39462753 DOI: 10.1016/j.semperi.2024.151988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Resuscitation of an infant with surgical complications includes both perioperative and post-operative management. In most cases this requires a multidisciplinary approach to achieve the best outcomes. Challenges include immediate expert management at infant delivery with the use of protocolized care, available anesthesia expertise, close monitoring of electrolytes with prompt attention to fluid status, and meticulous pain management. In this review, we will address contemporary research and ongoing challenges associated with resuscitation. We will make recommendations for effective resuscitation of this vulnerable and unique population from premature infant to term infant with complex surgical needs.
Collapse
Affiliation(s)
- Mona Khattab
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.
| | - Darby Donnelly
- Department of General Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amanda Blizzard
- Department of General Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shilpi Chabra
- Department of Pediatrics, University of Washington and Seattle Children's Hospital, WA, USA
| | - Elmer David
- Department of Pediatrics, University of southern California, Keck school of medicine, CA, USA
| | - Katherine A Stumpf
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sujir Pritha Nayak
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
4
|
Shaikh H, Lyle ANJ, Oslin E, Gray MM, Weiss EM. Eligible Infants Included in Neonatal Clinical Trials and Reasons for Noninclusion: A Systematic Review. JAMA Netw Open 2024; 7:e2441372. [PMID: 39453652 DOI: 10.1001/jamanetworkopen.2024.41372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024] Open
Abstract
Importance Results of clinical trials can only represent included participants, and many neonatal trials fail due to insufficient participation. Infants not included in research may differ from those included in meaningful ways, biasing the sample and limiting the generalizability of findings. Objective To describe the proportion of eligible infants included in neonatal clinical trials and the reasons for noninclusion. Evidence Review A systematic search of Cochrane CENTRAL was performed by retrieving articles meeting the following inclusion criteria: full-length, peer-reviewed articles describing clinical trial results in at least 20 human infants from US neonatal intensive care units, published in English, and added to Cochrane CENTRAL between 2017 and 2022. Retrieved articles were screened for inclusion by 2 independent researchers. Findings In total 120 articles met inclusion criteria and 91 of these (75.8%) reported the number of infants eligible for participation, which totaled 26 854 in aggregate. Drawing from these, an aggregate of 11 924 eligible infants (44.4%) were included in reported results. Among all eligible infants, most reasons for noninclusion in results were classified as modifiable or potentially modifiable by the research team. Parents declining to participate (8004 infants [29.8%]) or never being approached (2507 infants [9.3%]) were the 2 predominant reasons for noninclusion. Other modifiable reasons included factors related to study logistics, such as failure to appropriately collect data on enrolled infants (859 of 26 854 infants [3.2%]) and other reasons (1907 of 26 854 infants [7.1%]), such as loss to follow-up or eligible participants that were unaccounted for. Nonmodifiable reasons, including clinical change or death, accounted for a small proportion of eligible infants who were not included (858 of 26 854 infants [3.2%]). Conclusions and Relevance This systematic review of reporting on eligible infants included and not included in neonatal clinical trials highlights the need for improved documentation on the flow of eligible infants through neonatal clinical trials and may also inform recruitment expectations for trialists designing future protocols. Improved adherence to standardized reporting may clarify which potential participants are being missed, improving understanding of the generalizability of research findings. Furthermore, these findings suggest that future work to understand why parents decline to participate in neonatal research trials and why some are never approached about research may help increase overall participation.
Collapse
Affiliation(s)
- Henna Shaikh
- Department of Pediatrics, University of Washington School of Medicine, Seattle
| | - Allison N J Lyle
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Medical Group-Neonatology, Louisville, Kentucky
| | - Ellie Oslin
- Department of Pediatrics, University of Washington School of Medicine, Seattle
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Medical Group-Neonatology, Louisville, Kentucky
| | - Megan M Gray
- Department of Pediatrics, University of Washington School of Medicine, Seattle
| | - Elliott Mark Weiss
- Department of Pediatrics, University of Washington School of Medicine, Seattle
- Treuman Katz Center for Pediatric Bioethics & Palliative Care, Seattle Children's Research Institute, Seattle, Washington
| |
Collapse
|
5
|
Butzer SK, Faust K, Oberthuer A, Kleindiek C, Kuehne B, Haertel C, Mehler K. Wide use of broad-spectrum antibiotics in very low birth weight infants with spontaneous focal intestinal perforation-is it really justified? Infection 2024; 52:1901-1910. [PMID: 38634989 PMCID: PMC11499452 DOI: 10.1007/s15010-024-02257-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE Very low birth weight (VLBW) infants are at a risk of spontaneous focal intestinal perforation (FIP). Treatment includes supportive care, antibiotics, and drainage with/without surgery. Broad-spectrum antibiotic agents like carbapenems are applied frequently, although their use is not well-supported by the limited evidence of causal pathogens. We hypothesize that the use of carbapenems may not be necessary in VLBW infants with FIP. Our primary objective was to evaluate the antimicrobial use in VLBW infants with FIP in a cohort of the German Neonatal Network (GNN). The secondary objective was to characterize a subset in detail as a benchmark for future targets of stewardship. METHODS Data on VLBW infants with FIP was collected prospectively within the GNN, a collaboration of 68 neonatal intensive care units (NICU). With regards to the primary objective, patient characteristics and antimicrobial treatment were extracted from the predefined GNN database. To address our secondary objective, an additional on-site assessment of laboratory and microbiological culture results were performed. RESULTS In the GNN cohort, 613/21,646 enrolled infants (2.8%) developed FIP requiring surgery. They were frequently treated with carbapenems (500/613 (81.6%)) and vancomycin (497/613 (81.1%)). In a subset of 124 VLBW infants, 77 (72.6%) had proof of gram-positive bacteria in the abdominal cavity, coagulase-negative staphylococci (CoNS) predominantly. Despite the low prevalence of gram-negative bacteria (n = 6 (4.8%)), the combination of meropenem and vancomycin was prescribed most frequently (n = 96 (78.0%)). CONCLUSION The use of carbapenems as broad-spectrum antimicrobials agents might not be justified in most VLBW infants with FIP. Knowledge on the development of the neonatal gut microbiota, local resistance patterns and individual microbiological findings should be taken into consideration when implementing antimicrobial stewardship programs (ASPs).
Collapse
Affiliation(s)
- Sarina K Butzer
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Faculty of Medicine, University of Cologne, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany.
| | - Kirstin Faust
- Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - André Oberthuer
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Faculty of Medicine, University of Cologne, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
| | | | - Benjamin Kuehne
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany
| | - Christoph Haertel
- Department of Pediatrics, University of Würzburg, Würzburg, Germany
- German Neonatal Network (GNN), Lübeck, Germany
| | - Katrin Mehler
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Faculty of Medicine, University of Cologne, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany.
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, University of Cologne, University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
6
|
Maselli KM, Shah NR, Amin SC, Wieczorek DN, Lutrzykowska ZL, Matusko N, Hirschl RB, Speck KE, Gadepalli SK. Is There Still a Role for Peritoneal Drains in Neonatal Pneumoperitoneum? A Single-Center Experience. J Surg Res 2024; 302:509-516. [PMID: 39178566 DOI: 10.1016/j.jss.2024.07.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 08/26/2024]
Abstract
INTRODUCTION Although pneumoperitoneum from necrotizing enterocolitis or spontaneous intestinal perforation is a surgical emergency, risk stratification to determine which neonates benefit from initial peritoneal drainage (PD) is lacking. METHODS Using a single-center retrospective review of very low birth weight neonates under 1500 g who underwent PD for pneumoperitoneum (January 2015 to December 2023) from necrotizing enterocolitis or spontaneous intestinal perforation, two cohorts were created: drain "responders" (patients managed definitively with PD; includes placement of a second drain) and "nonresponders" (patients who underwent subsequent laparotomy or died after PD). Antenatal/postnatal characteristics, periprocedural clinical data, and hospital outcomes were compared between responders and nonresponders using Student's t-test, chi-squared test, or Kruskal-Wallis test as appropriate, with P < 0.05 considered significant. RESULTS Fifty-six neonates were included: 31 (55%) drain responders and 25 (45%) nonresponders. Birth weight, gestational age, sex, ethnicity, use of postnatal steroids, and enteral feeds were similar between the cohorts. Nonresponders had higher base deficits (-3.4 versus -5.0, P = 0.032) and FiO2 (0.25 versus 0.52, P = 0.001) after drain placement. Drain responders had significantly shorter lengths of stay (89 versus 148 days, P = 0.014) and lower mortality (6.4% versus 56%, P < 0.001). A subgroup analysis of the nonresponders showed no differences in birth weight, vasopressor requirement, FiO2, or postdrain base deficit between nonresponders who had a drain alone versus laparotomy following drain placement. CONCLUSIONS PD remains a viable initial therapy for pneumoperitoneum in premature very low birth weight neonates (< 1500 g), demonstrating clinical response in more than half. Ongoing clinical assessment and judgment is imperative after drain placement to ensure continued clinical improvement.
Collapse
Affiliation(s)
- Kathryn M Maselli
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, Michigan Medicine, C.S. Mott Children's Hospital, Ann Arbor, Michigan.
| | - Nikhil R Shah
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, Michigan Medicine, C.S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Sharmi C Amin
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, Michigan Medicine, C.S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Daniel N Wieczorek
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, Michigan Medicine, C.S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Zuzanna L Lutrzykowska
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, Michigan Medicine, C.S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Niki Matusko
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, Michigan Medicine, C.S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Ronald B Hirschl
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, Michigan Medicine, C.S. Mott Children's Hospital, Ann Arbor, Michigan
| | - K Elizabeth Speck
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, Michigan Medicine, C.S. Mott Children's Hospital, Ann Arbor, Michigan
| | - Samir K Gadepalli
- Department of Surgery, Section of Pediatric Surgery, University of Michigan, Michigan Medicine, C.S. Mott Children's Hospital, Ann Arbor, Michigan
| |
Collapse
|
7
|
Speer AL, Lally KP, Pedroza C, Zhang Y, Poindexter BB, Chwals WJ, Hintz SR, Besner GE, Stevenson DK, Ohls RK, Truog WE, Stoll BJ, Rysavy MA, Das A, Tyson JE, Blakely ML. Surgical Necrotizing Enterocolitis and Spontaneous Intestinal Perforation Lead to Severe Growth Failure in Infants. Ann Surg 2024; 280:432-443. [PMID: 39264354 DOI: 10.1097/sla.0000000000006378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
OBJECTIVE We aimed to determine the incidence of growth failure in infants with necrotizing enterocolitis (NEC) or spontaneous intestinal perforation (SIP) and whether initial laparotomy versus peritoneal drainage (PD) impacted the likelihood of growth failure. SUMMARY BACKGROUND DATA Infants with surgical NEC and SIP have high mortality, and most have neurodevelopmental impairment and poor growth. Existing literature on growth outcomes for these infants is limited. METHODS This is a preplanned secondary study of the Necrotizing Enterocolitis Surgery Trial dataset. The primary outcome was growth failure (Z-score for weight <-2.0) at 18 to 22 months. We used logistic regression, including diagnosis and treatment, as covariates. Secondary outcomes were analyzed using the Fisher exact or Pearson χ2 test for categorical variables and the Wilcoxon rank sum test or one-way ANOVA for continuous variables. RESULTS Among 217 survivors, 207 infants (95%) had primary outcome data. Growth failure at 18 to 22 months occurred in 24/50 (48%) of NEC infants versus 65/157 (42%) SIP (P=0.4). The mean weight-for-age Z-score at 18 to 22 months in NEC infants was -2.05±0.99 versus -1.84±1.09 SIP (P=0.2), and the predicted mean weight-for-age Z-score SIP (Beta -0.27; 95% CI: -0.53, -0.01; P=0.041). Median declines in weight-for-age Z-score between birth and 18 to 22 months were significant in all infants but most severe (>2) in NEC infants (P=0.2). CONCLUSIONS This first ever prospective study of growth outcomes in infants with surgical NEC or SIP demonstrates that growth failure is very common, especially in infants with NEC, and persists at 18-22 months.
Collapse
Affiliation(s)
- Allison L Speer
- Department of Pediatric Surgery, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX
| | - Kevin P Lally
- Department of Pediatric Surgery, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX
| | - Claudia Pedroza
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX
| | - Yuxin Zhang
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX
| | - Brenda B Poindexter
- Department of Pediatrics, Division of Neonatology, Emory University School of Medicine, Atlanta, GA
| | - Walter J Chwals
- Department of Pediatric Surgery, Floating Hospital for Children, Tufts Medical Center, Boston, MA
| | - Susan R Hintz
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA
| | - Gail E Besner
- Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH
| | - David K Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA
| | - Robin K Ohls
- University of New Mexico Health Sciences Center, Albuquerque, NM
- Department of Pediatrics, Division of Neonatology, University of Utah School of Medicine, Salt Lake City, UT
| | - William E Truog
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO
| | - Barbara J Stoll
- Department of Pediatrics, Division of Neonatology, Emory University School of Medicine, Atlanta, GA
| | - Matthew A Rysavy
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX
| | - Abhik Das
- Social, Statistical and Environmental Sciences Unit, RTI International, Rockville, MD
| | - Jon E Tyson
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX
| | - Martin L Blakely
- Department of Pediatric Surgery, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX
| |
Collapse
|
8
|
Nayak SP, Sánchez-Rosado M, Reis JD, Brown LS, Mangona KL, Sharma P, Nelson DB, Wyckoff MH, Pandya S, Mir IN, Brion LP. Development of a Prediction Model for Surgery or Early Mortality at the Time of Initial Assessment for Necrotizing Enterocolitis. Am J Perinatol 2024; 41:1714-1727. [PMID: 38272063 DOI: 10.1055/a-2253-8656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
OBJECTIVE No available scale, at the time of initial evaluation for necrotizing enterocolitis (NEC), accurately predicts, that is, with an area under the curve (AUC) ≥0.9, which preterm infants will undergo surgery for NEC stage III or die within a week. STUDY DESIGN This is a retrospective cohort study (n = 261) of preterm infants with <33 weeks' gestation or <1,500 g birth weight with either suspected or with definite NEC born at Parkland Hospital between 2009 and 2021. A prediction model using the new HASOFA score (Hyperglycemia, Hyperkalemia, use of inotropes for Hypotension during the prior week, Acidemia, Neonatal Sequential Organ Failure Assessment [nSOFA] score) was compared with a similar model using the nSOFA score. RESULTS Among 261 infants, 112 infants had NEC stage I, 68 with NEC stage II, and 81 with NEC stage III based on modified Bell's classification. The primary outcome, surgery for NEC stage III or death within a week, occurred in 81 infants (surgery in 66 infants and death in 38 infants). All infants with pneumoperitoneum or abdominal compartment syndrome either died or had surgery. The HASOFA and the nSOFA scores were evaluated in 254 and 253 infants, respectively, at the time of the initial workup for NEC. Both models were internally validated. The HASOFA model was a better predictor of surgery for NEC stage III or death within a week than the nSOFA model, with greater AUC 0.909 versus 0.825, respectively, p < 0.001. Combining HASOFA at initial assessment with concurrent or later presence of abdominal wall erythema or portal gas improved the prediction surgery for NEC stage III or death with AUC 0.942 or 0.956, respectively. CONCLUSION Using this new internally validated prediction model, surgery for NEC stage III or death within a week can be accurately predicted at the time of initial assessment for NEC. KEY POINTS · No available scale, at initial evaluation, accurately predicts which preterm infants will undergo surgery for NEC stage III or die within a week.. · In this retrospective cohort study of 261 preterm infants with either suspected or definite NEC we developed a new prediction model (HASOFA score).. · The HASOFA-model had high discrimination (AUC: 0.909) and excellent calibration and was internally validated..
Collapse
Affiliation(s)
- Sujir P Nayak
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Mariela Sánchez-Rosado
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Neonatology, Joe DiMaggio Children's Hospital, Hollywood, Florida
| | - Jordan D Reis
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pediatrics, Baylor Scott and White, Dallas, Texas
| | - L Steven Brown
- Department of Research, Parkland Health and Hospital System, Dallas, Texas
| | - Kate L Mangona
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Priya Sharma
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pediatrics, Baylor Scott and White, Dallas, Texas
| | - David B Nelson
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, and Parkland Health, Dallas, Texas
| | - Myra H Wyckoff
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Samir Pandya
- Division of Pediatric Surgery, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Imran N Mir
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Luc P Brion
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
9
|
Li J, Zhou J, Weng J, Jin F, Shen Y, Qi Y, Jiang M, Hei M. Rapidly progressive necrotizing enterocolitis: Risk factors and a predictive model. Pediatr Res 2024:10.1038/s41390-024-03482-z. [PMID: 39147906 DOI: 10.1038/s41390-024-03482-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Rapidly progressive necrotizing enterocolitis (RP-NEC) is a particular subtype of NEC known for its rapid progression and high mortality rate. The objective of this study was to establish a predictive model for RP-NEC. METHODS This was a retrospective single-center cohort study. Patients were newborn infants with NEC (Bell's stage ≥ IIB) admitted from January 1, 2016 to December 31, 2023. The primary outcome was RP-NEC defined as the need for surgical intervention and/or death within 48 hours of the onset of NEC. RESULTS Totally 334 newborn infants were included, among which 82 (24.6%) were RP-NEC cases with a gestation age 34.1 (31.0, 37.0) weeks and birth weight 2100 (1413, 2800) g. Plasma sodium <135 mmol/L, C-reactive protein ≥10 mg/L, platelet count <100 × 109/L, lymphocyte count <1.5 × 109/L, pH <7.2 in blood gas, and ascites at NEC onset were identified as independent risk factors for RP-NEC. The model established presented an AUC value of 0.983 (95% CI 0.97-0.99). The calibration curve for validation was applied revealing a slope close to unity while the Hosmer-Lemeshow test yielded χ2 = 2.550 (p = 0.636). CONCLUSION The predictive model established on the above 6 items of RP-NEC is highly promising. IMPACT Currently, there is a paucity of research on this specific type of severe necrotizing enterocolitis (NEC) characterized by rapid progression. Our study was to investigate the risk factors associated with surgical intervention and/or death within 48 hours following onset in infants with NEC, establish a predictive model for infants with rapidly progressive NEC. The new data presented in this study was the ROC curve combining the above factors as well as hyponatremia.
Collapse
Affiliation(s)
- Jicheng Li
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Jingjing Zhou
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Jingwen Weng
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Fei Jin
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Yanhua Shen
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Yujie Qi
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
- National Center for Children's Health, Beijing, 100045, China
| | - Min Jiang
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China.
- National Center for Children's Health, Beijing, 100045, China.
| | - Mingyan Hei
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China.
- National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
10
|
Hair AB, Sullivan KM, Ahmad I, Zaniletti I, Acker SN, Premkumar MH, Reber K, Huff KA, Nayak SP, DiGeronimo R, Kim J, Roberts J, Markel TA, Brozanski B, Sharma J, Piazza AJ, Yanowitz TD. Initial surgery for spontaneous intestinal perforation in extremely low birth weight infants is not associated with mortality or in-hospital morbidities. J Perinatol 2024:10.1038/s41372-024-02037-8. [PMID: 38992239 DOI: 10.1038/s41372-024-02037-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 05/11/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024]
Abstract
OBJECTIVE Determine short-term outcomes following peritoneal drain (PD), laparotomy (LAP) after PD (PD-LAP), and LAP in extremely low birth weight (ELBW) infants with spontaneous intestinal perforation (SIP). STUDY DESIGN ELBW infants with SIP were identified using the Children's Hospitals Neonatal Database. Mortality and length of stay (LOS) were compared among groups. RESULTS Of 729 SIP infants from 6/2010-12/2016, 383(53%) received PD, 61(8%) PD-LAP, and 285(39%) LAP. PD infants had lower GA at birth, at SIP diagnosis and upon admission than PD-LAP or LAP; and higher sepsis rates than LAP. Bivariate analysis and Kaplan-Meier survival estimates suggested PD had increased mortality vs. PD-LAP and LAP (27%, 11.5%, and 15.8% respectively, p < 0.001). However, surgical approach was not significantly associated with mortality in multivariable analysis accounting for GA and illness severity. LOS did not differ by surgical approach. CONCLUSIONS In ELBW infants with SIP, mortality, and LOS are independent of the initial surgical approach.
Collapse
Affiliation(s)
- Amy B Hair
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.
| | - Kevin M Sullivan
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
- Nemours Children's Hospital, Wilmington, DE, USA
| | - Irfan Ahmad
- Children's Hospital of Orange County, Orange, CA, USA
| | | | - Shannon N Acker
- University of Colorado, Children's Hospital of Colorado, Aurora, CO, USA
| | | | - Kristina Reber
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Katie A Huff
- Indiana University School of Medicine, Riley Hospital for Children, Indianapolis, IN, USA
| | | | - Robert DiGeronimo
- Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - Jae Kim
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jessica Roberts
- Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Troy A Markel
- Indiana University School of Medicine, Riley Hospital for Children, Indianapolis, IN, USA
| | - Beverly Brozanski
- Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO, USA
| | - Jotishna Sharma
- Missouri University of Missouri Kansas City School of Medicine, Children's Mercy Hospital, Kansas City, MO, USA
| | - Anthony J Piazza
- Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Toby D Yanowitz
- University of Pittsburgh School of Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
11
|
Gipson DR, Chang AL, Lure AC, Mehta SA, Gowen T, Shumans E, Stevenson D, de la Cruz D, Aghaeepour N, Neu J. Reassessing acquired neonatal intestinal diseases using unsupervised machine learning. Pediatr Res 2024; 96:165-171. [PMID: 38413766 DOI: 10.1038/s41390-024-03074-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/11/2023] [Accepted: 01/02/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Acquired neonatal intestinal diseases have an array of overlapping presentations and are often labeled under the dichotomous classification of necrotizing enterocolitis (which is poorly defined) or spontaneous intestinal perforation, hindering more precise diagnosis and research. The objective of this study was to take a fresh look at neonatal intestinal disease classification using unsupervised machine learning. METHODS Patients admitted to the University of Florida Shands Neonatal Intensive Care Unit January 2013-September 2019 diagnosed with an intestinal injury, or had imaging findings of portal venous gas, pneumatosis, abdominal free air, or had an abdominal drain placed or exploratory laparotomy during admission were included. Congenital gastroschisis, omphalocele, intestinal atresia, malrotation were excluded. Data was collected via retrospective chart review with subsequent hierarchal, unsupervised clustering analysis. RESULTS Five clusters of intestinal injury were identified: Cluster 1 deemed the "Low Mortality" cluster, Cluster 2 deemed the "Mature with Inflammation" cluster, Cluster 3 deemed the "Immature with High Mortality" cluster, Cluster 4 deemed the "Late Injury at Full Feeds" cluster, and Cluster 5 deemed the "Late Injury with High Rate of Intestinal Necrosis" cluster. CONCLUSION Unsupervised machine learning can be used to cluster acquired neonatal intestinal injuries. Future study with larger multicenter datasets is needed to further refine and classify types of intestinal diseases. IMPACT Unsupervised machine learning can be used to cluster types of acquired neonatal intestinal injury. Five major clusters of acquired neonatal intestinal injury are described, each with unique features. The clusters herein described deserve future, multicenter study to determine more specific early biomarkers and tailored therapeutic interventions to improve outcomes of often devastating neonatal acquired intestinal injuries.
Collapse
Affiliation(s)
- Daniel R Gipson
- University of Florida College of Medicine, Department of Pediatrics, Division of Neonatology, Gainesville, FL, USA.
| | - Alan L Chang
- Stanford University School of Medicine, Department of Anesthesiology, Pain, and Perioperative Medicine, Department of Pediatrics, and Department of Biomedical Data Science, Stanford, CA, USA
| | - Allison C Lure
- Nationwide Children's Hospital, The Ohio State University College of Medicine, Department of Pediatrics, Division of Neonatology, Columbus, OH, USA
- University of Florida College of Medicine, Department of Pediatrics, Gainesville, FL, USA
| | - Sonia A Mehta
- University of Florida College of Medicine, Department of Pediatrics, Gainesville, FL, USA
- University of California, Irvine Medical Center, Department of Pediatrics, Division of Neonatology, Irvine, CA, USA
| | - Taylor Gowen
- University of Florida College of Medicine, Department of Pediatrics, Gainesville, FL, USA
- University of Florida College of Medicine, Department of Anesthesiology, Gainesville, FL, USA
| | - Erin Shumans
- University of Florida College of Medicine, Department of Pediatrics, Gainesville, FL, USA
| | - David Stevenson
- Stanford University School of Medicine, Department of Pediatrics, Division of Neonatology, Stanford, CA, USA
| | - Diomel de la Cruz
- University of Florida College of Medicine, Department of Pediatrics, Division of Neonatology, Gainesville, FL, USA
| | - Nima Aghaeepour
- Stanford University School of Medicine, Department of Anesthesiology, Pain, and Perioperative Medicine, Department of Pediatrics, and Department of Biomedical Data Science, Stanford, CA, USA
| | - Josef Neu
- University of Florida College of Medicine, Department of Pediatrics, Division of Neonatology, Gainesville, FL, USA
| |
Collapse
|
12
|
Klerk DH, van Varsseveld OC, Offringa M, Modi N, Lacher M, Zani A, Pakarinen MP, Koivusalo A, Jester I, Spruce M, Derikx JPM, Bakx R, Ksia A, Kooi EMW, Hulscher JBF. Core Outcome Set for Necrotizing Enterocolitis Treatment Trials. Pediatrics 2024; 153:e2023065619. [PMID: 38726575 DOI: 10.1542/peds.2023-065619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Variability in outcome reporting in necrotizing enterocolitis (NEC) treatment trials hinders conducting meta-analyses and implementing novel treatments. We aimed to develop a core outcome set (COS) for NEC treatment trials including outcome measures most relevant to patients and physicians, from NEC diagnosis to adulthood. METHODS Clinicians and/or researchers from low-middle- and high-income countries were approached based on their scientific contributions to NEC literature, and patients and parents through local organizations. We presented participants with 45 outcomes used in NEC research, identified through a systematic review. To achieve consensus, outcomes were rated on a scale of 1 to 9 in 3 online Delphi rounds, and discussed at a final consensus meeting. RESULTS Seventy-one participants from 25 countries completed all Delphi rounds, including 15 patients and family representatives. Thirteen outcomes reached consensus in one of the stakeholder groups and were included in the consensus meeting, 6 outcomes reached consensus in both groups. Twenty-seven participants from both high- and low-middle-income countries attended the online consensus meeting, including family representatives and NEC patients. After discussion and a final vote, 5 outcomes reached consensus to be included: mortality, NEC-related mortality, short bowel syndrome, quality of life, and neurodevelopmental impairment. CONCLUSIONS This NEC COS includes 5 predominantly long-term outcomes agreed upon by clinicians, patients, and family representatives. Use of this international COS will help standardize outcome selection in clinical trials, ensure these are relevant to those most affected by NEC care, and, ultimately, improve the care of infants with NEC.
Collapse
Affiliation(s)
| | - Otis C van Varsseveld
- Division of Pediatric Surgery, Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Martin Offringa
- Child Health Evaluative Sciences, The Hospital for Sick Children Research Institute
| | - Neena Modi
- Section of Neonatal Medicine, School of Public Health, Chelsea and Westminster Hospital campus, Imperial College London, London, United Kingdom
| | - Martin Lacher
- Department of Pediatric Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Augusto Zani
- Department of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Mikko P Pakarinen
- Department of Pediatric Surgery, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Antti Koivusalo
- Department of Pediatric Surgery, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Ingo Jester
- Departments of Paediatric Surgery, Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham, United Kingdom
| | - Marie Spruce
- NEC United Kingdom Charity, Nottingham, United Kingdom
| | - Joep P M Derikx
- Department of Pediatric Surgery, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, the Netherlands
| | - Roel Bakx
- Department of Pediatric Surgery, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, the Netherlands
| | - Amine Ksia
- Departments of Surgery and Pediatric Surgery, Fattouma Bourguiba Hospital, Monastir Medical School, Monastir University, Tunisia
| | | | - Jan B F Hulscher
- Division of Pediatric Surgery, Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
13
|
Massaro AN, Boyer B, Gill M, Kim D, Laughon M, Walsh M, Lemmon ME, Pilon B, Baer G. Measuring Clinical Benefit in Neonatal Randomized Clinical Trials: Challenges and Opportunities. J Pediatr 2024; 269:113972. [PMID: 38401783 PMCID: PMC11181158 DOI: 10.1016/j.jpeds.2024.113972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/01/2024] [Accepted: 02/16/2024] [Indexed: 02/26/2024]
Affiliation(s)
- An N Massaro
- Office of Pediatric Therapeutics, Office of the Commisioner, US FDA, Silver Spring, MD.
| | - Beth Boyer
- Duke-Margolis Center for Health Policy, Washington, DC
| | - Mira Gill
- Duke-Margolis Center for Health Policy, Washington, DC
| | - Dure Kim
- Duke-Margolis Center for Health Policy, Washington, DC
| | - Matthew Laughon
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Michele Walsh
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| | - Monica E Lemmon
- Duke-Margolis Center for Health Policy, Washington, DC; Departments of Pediatrics and Population Health Sciences, Duke University School of Medicine, Durham, NC
| | | | - Gerri Baer
- Office of Pediatric Therapeutics, Office of the Commisioner, US FDA, Silver Spring, MD
| |
Collapse
|
14
|
Wootton SH, Rysavy M, Davis P, Thio M, Romero-Lopez M, Holzapfel LF, Thrasher T, Wade JD, Owen L. Practical approaches for supporting informed consent in neonatal clinical trials. Acta Paediatr 2024; 113:923-930. [PMID: 38389165 PMCID: PMC11006570 DOI: 10.1111/apa.17165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/23/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
The survival and health of preterm and critically ill infants have markedly improved over the past 50 years, supported by well-conducted neonatal research. However, newborn research is difficult to undertake for many reasons, and obtaining informed consent for research in this population presents several unique ethical and logistical challenges. In this article, we explore methods to facilitate the consent process, including the role of checklists to support meaningful informed consent for neonatal clinical trials. CONCLUSION: The authors provide practical guidance on the design and implementation of an effective consent checklist tailored for use in neonatal clinical research.
Collapse
Affiliation(s)
- Susan H. Wootton
- Division of Infectious Diseases, Department of Pediatrics, McGovern Medical School at UTHealth Houston, Houston, Texas, USA
- Institute for Clinical Research and Learning Health Care, McGovern Medical School at UTHealth Houston, Houston, Texas, USA
- Children's Memorial Hermann Hospital, Houston, Texas, USA
| | - Matthew Rysavy
- Institute for Clinical Research and Learning Health Care, McGovern Medical School at UTHealth Houston, Houston, Texas, USA
- Children's Memorial Hermann Hospital, Houston, Texas, USA
- Division of Neonatology, Department of Pediatrics, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Peter Davis
- Newborn Research, Neonatal Services, Royal Women's Hospital, Melbourne, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Australia
| | - Marta Thio
- Newborn Research, Neonatal Services, Royal Women's Hospital, Melbourne, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Australia
- Gandel Simulation Service, Royal Women's Hospital, Melbourne, Australia
| | - Mar Romero-Lopez
- Institute for Clinical Research and Learning Health Care, McGovern Medical School at UTHealth Houston, Houston, Texas, USA
- Children's Memorial Hermann Hospital, Houston, Texas, USA
- Division of Neonatology, Department of Pediatrics, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Lindsay F. Holzapfel
- Children's Memorial Hermann Hospital, Houston, Texas, USA
- Division of Neonatology, Department of Pediatrics, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Tamara Thrasher
- Children's Memorial Hermann Hospital, Houston, Texas, USA
- March of Dimes NICU Family Support Program, Houston, Texas, USA
| | - Jaleesa D. Wade
- Division of Neonatology, Department of Pediatrics, McGovern Medical School at UTHealth, Houston, Texas, USA
| | - Louise Owen
- Newborn Research, Neonatal Services, Royal Women's Hospital, Melbourne, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Melbourne, Australia
- Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Australia
| |
Collapse
|
15
|
Sha C, Van Brunt T, Kudria J, Schmidt D, Yurovsky A, Bandovic J, Giarrizzo M, Lin J, Tsirka SA, Bialkowska AB, Wollmuth L, Speer E, Hsieh H. A graded neonatal mouse model of necrotizing enterocolitis demonstrates that mild enterocolitis is sufficient to activate microglia and increase cerebral cytokine expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.03.551849. [PMID: 38746118 PMCID: PMC11092491 DOI: 10.1101/2023.08.03.551849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Necrotizing enterocolitis (NEC) is an inflammatory gastrointestinal process that afflicts approximately 10% of preterm infants born in the United States each year, with a mortality rate of 30%. NEC severity is graded using Bell's classification system, from stage I mild NEC to stage III severe NEC. Over half of NEC survivors present with neurodevelopmental impairment during adolescence, a long-term complication that is poorly understood but can occur even after mild NEC. Although multiple animal models exist, none allow the experimenter to control nor represent the gradient of symptom severities seen in NEC patients. We bridge this knowledge gap by developing a graded murine model of NEC and studying its relationship with neuroinflammation across a range of NEC severities. Methods Postnatal day 3 (P3) C57BL/6 mice were fed a formula containing different concentrations (0% control, 0.25%, 1%, 2%, and 3%) of dextran sodium sulfate (DSS). P3 mice were fed every 3 hours for 72-hours. We collected data on weight gain and behavior (activity, response, body color) during feeding. At the end of the experiment, we collected tissues (intestine, liver, plasma, brain) for immunohistochemistry, immunofluorescence, and cytokine and chemokine analysis. Results Throughout NEC induction, mice fed higher concentrations of DSS died sooner, lost weight faster, and became sick or lethargic earlier. Intestinal characteristics (dilation, color, friability) were worse in mice fed with higher DSS concentrations. Histology revealed small intestinal disarray among mice fed all DSS concentrations, while higher DSS concentrations resulted in reduced small intestinal cellular proliferation and increased hepatic and systemic inflammation. In the brain, IL-2, G-CSF, and CXCL1 concentrations increased with higher DSS concentrations. Although the number of neurons and microglia in the CA1 hippocampal region did not differ, microglial branching was significantly reduced in DSS-fed mice. Conclusion We characterize a novel graded model of NEC that recapitulates the full range of NEC severities. We show that mild NEC is sufficient to initiate neuroinflammation and microglia activation. This model will facilitate studies on the neurodevelopmental effects of NEC.
Collapse
|
16
|
Halpern MD, Gupta A, Zaghloul N, Thulasingam S, Calton CM, Camp SM, Garcia JGN, Ahmed M. Extracellular Nicotinamide Phosphoribosyltransferase Is a Therapeutic Target in Experimental Necrotizing Enterocolitis. Biomedicines 2024; 12:970. [PMID: 38790933 PMCID: PMC11118767 DOI: 10.3390/biomedicines12050970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency of prematurity. Postulated mechanisms leading to inflammatory necrosis of the ileum and colon include activation of the pathogen recognition receptor Toll-like receptor 4 (TLR4) and decreased levels of transforming growth factor beta (TGFβ). Extracellular nicotinamide phosphoribosyltransferase (eNAMPT), a novel damage-associated molecular pattern (DAMP), is a TLR4 ligand and plays a role in a number of inflammatory disease processes. To test the hypothesis that eNAMPT is involved in NEC, an eNAMPT-neutralizing monoclonal antibody, ALT-100, was used in a well-established animal model of NEC. Preterm Sprague-Dawley pups delivered prematurely from timed-pregnant dams were exposed to hypoxia/hypothermia and randomized to control-foster mother dam-fed rats, injected IP with saline (vehicle) 48 h after delivery; control + mAB-foster dam-fed rats, injected IP with 10 µg of ALT-100 at 48 h post-delivery; NEC-orally gavaged, formula-fed rats injected with saline; and NEC + mAb-formula-fed rats, injected IP with 10 µg of ALT-100 at 48 h. The distal ileum was processed 96 h after C-section delivery for histological, biochemical, molecular, and RNA sequencing studies. Saline-treated NEC pups exhibited markedly increased fecal blood and histologic ileal damage compared to controls (q < 0.0001), and findings significantly reduced in ALT-100 mAb-treated NEC pups (q < 0.01). Real-time PCR in ileal tissues revealed increased NAMPT in NEC pups compared to pups that received the ALT-100 mAb (p < 0.01). Elevated serum levels of tumor necrosis factor alpha (TNFα), interleukin 6 (IL-6), interleukin-8 (IL-8), and NAMPT were observed in NEC pups compared to NEC + mAb pups (p < 0.01). Finally, RNA-Seq confirmed dysregulated TGFβ and TLR4 signaling pathways in NEC pups that were attenuated by ALT-100 mAb treatment. These data strongly support the involvement of eNAMPT in NEC pathobiology and eNAMPT neutralization as a strategy to address the unmet need for NEC therapeutics.
Collapse
Affiliation(s)
- Melissa D. Halpern
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Akash Gupta
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Nahla Zaghloul
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Senthilkumar Thulasingam
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Christine M. Calton
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Sara M. Camp
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL 33458, USA (J.G.N.G.)
| | - Joe G. N. Garcia
- Center for Inflammation Science and Systems Medicine, University of Florida Scripps Research Institute, Jupiter, FL 33458, USA (J.G.N.G.)
| | - Mohamed Ahmed
- Division of Neonatology, Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
17
|
Maiga AW, Snyder RA, Kao LS, Raval MV, Patel MB, Blakely ML. Advancing Randomized Clinical Trials in Surgery: Role of Exception From Informed Consent, Central Institutional Review Board, and Bayesian Approaches. J Surg Res 2024:S0022-4804(24)00167-7. [PMID: 38670847 DOI: 10.1016/j.jss.2024.03.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/16/2024] [Accepted: 03/12/2024] [Indexed: 04/28/2024]
Affiliation(s)
- Amelia W Maiga
- Division of Acute Care Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Critical Illness, Brain dysfunction, and Survivorship Center, Vanderbilt Center for Health Services Research, Vanderbilt Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Rebecca A Snyder
- Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lillian S Kao
- Division of Acute Care Surgery, Department of Surgery, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Mehul V Raval
- Division of Pediatric Surgery, Department of Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Mayur B Patel
- Division of Acute Care Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee; Critical Illness, Brain dysfunction, and Survivorship Center, Vanderbilt Center for Health Services Research, Vanderbilt Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee; Geriatric Research Education and Clinical Center, Surgical Services, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Martin L Blakely
- Department of Surgery, Institute for Clinical Research and Learning Health Care, Institute for Implementation Science, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
18
|
Epstein AA, Janos SN, Menozzi L, Pegram K, Jain V, Bisset LC, Davis JT, Morrison S, Shailaja A, Guo Y, Chao AS, Abdi K, Rikard B, Yao J, Gregory SG, Fisher K, Pittman R, Erkanli A, Gustafson KE, Carrico CWT, Malcolm WF, Inder TE, Cotten CM, Burt TD, Shinohara ML, Maxfield CM, Benner EJ. Subventricular zone stem cell niche injury is associated with intestinal perforation in preterm infants and predicts future motor impairment. Cell Stem Cell 2024; 31:467-483.e6. [PMID: 38537631 PMCID: PMC11129818 DOI: 10.1016/j.stem.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 02/11/2024] [Accepted: 03/01/2024] [Indexed: 04/07/2024]
Abstract
Brain injury is highly associated with preterm birth. Complications of prematurity, including spontaneous or necrotizing enterocolitis (NEC)-associated intestinal perforations, are linked to lifelong neurologic impairment, yet the mechanisms are poorly understood. Early diagnosis of preterm brain injuries remains a significant challenge. Here, we identified subventricular zone echogenicity (SVE) on cranial ultrasound in preterm infants following intestinal perforations. The development of SVE was significantly associated with motor impairment at 2 years. SVE was replicated in a neonatal mouse model of intestinal perforation. Examination of the murine echogenic subventricular zone (SVZ) revealed NLRP3-inflammasome assembly in multiciliated FoxJ1+ ependymal cells and a loss of the ependymal border in this postnatal stem cell niche. These data suggest a mechanism of preterm brain injury localized to the SVZ that has not been adequately considered. Ultrasound detection of SVE may serve as an early biomarker for neurodevelopmental impairment after inflammatory disease in preterm infants.
Collapse
Affiliation(s)
- Adrian A Epstein
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Sara N Janos
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Luca Menozzi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kelly Pegram
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Logan C Bisset
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Joseph T Davis
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Samantha Morrison
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Aswathy Shailaja
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Yingqiu Guo
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Agnes S Chao
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Khadar Abdi
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Blaire Rikard
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA; Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Kimberley Fisher
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Rick Pittman
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Al Erkanli
- Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Kathryn E Gustafson
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | | | - William F Malcolm
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Terrie E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - C Michael Cotten
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA
| | - Trevor D Burt
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA; Children's Health and Discovery Initiative, Duke University School of Medicine, Durham, NC, USA
| | - Mari L Shinohara
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Charles M Maxfield
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA.
| | - Eric J Benner
- Department of Pediatrics, Division of Neonatology, Duke University School of Medicine, Durham, NC, USA; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
19
|
Garg PM, Garg PP, Shenberger JS. Clinical utilization of intestinal pathology in the classification of NEC vs SIP cases and prognostication. J Perinatol 2024; 44:598-599. [PMID: 38480786 PMCID: PMC11003823 DOI: 10.1038/s41372-024-01932-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Affiliation(s)
- Parvesh Mohan Garg
- Department of Pediatrics/Neonatology, Atrium Health Wake Forest Baptist, Wake Forest School of Medicine, Winston Salem, NC, USA.
| | - Padma P Garg
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | | |
Collapse
|
20
|
Raba AA, Coleman J, Cunningham K. Evaluation of the management of intestinal perforation in very low birth infants, a 10-year review. Acta Paediatr 2024; 113:733-738. [PMID: 38182549 DOI: 10.1111/apa.17069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/17/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024]
Abstract
AIM The aim of this study was to assess outcomes of peritoneal drainage and laparotomy in the management of intestinal perforation secondary to necrotizing enterocolitis (NEC) and spontaneous intestinal perforation. METHODS A retrospective review of all preterm infants (birthweight ≤1500 g) who underwent surgical intervention (peritoneal drainage and/or laparotomy) for intestinal perforation between March 2010 and March 2020. RESULTS A total of 43 infants who underwent surgical intervention for intestinal perforation were included [19 (44%) with NEC and 24 (56%) with spontaneous intestinal perforation]. Peritoneal drainage was more commonly placed as the initial surgical procedure for management of spontaneous intestinal perforation compared with surgical NEC [23 (96%) vs. 11 (58%), p = 0.003]. Mortality was greater for infants who were initially managed with peritoneal drainage [11 (32%)] compared with those who underwent primary laparotomy [2 (22%), p = 0.5]. CONCLUSION Initial surgical management of intestinal perforation is more often according to underlying pathology. Our data support primary laparotomy for infants with perforated NEC.
Collapse
Affiliation(s)
- Ali Ahmed Raba
- Department of Neonatology, The Rotunda Hospital, Dublin, Ireland
| | - John Coleman
- Department of Neonatology, The Rotunda Hospital, Dublin, Ireland
| | - Katie Cunningham
- Department of Neonatology, The Rotunda Hospital, Dublin, Ireland
| |
Collapse
|
21
|
Keane OA, Dantes G, Dutreuil VL, Do L, Rumbika S, Sylvestre PB, Bhatia AM. Comparison of preoperative and intraoperative surgeon diagnosis and pathologic findings in spontaneous intestinal perforation vs necrotizing enterocolitis. J Perinatol 2024; 44:568-574. [PMID: 38263461 DOI: 10.1038/s41372-024-01876-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/28/2023] [Accepted: 01/05/2024] [Indexed: 01/25/2024]
Abstract
OBJECTIVE To investigate the accuracy of preoperative and intraoperative diagnosis via comparison to pathologic diagnosis in spontaneous intestinal perforation (SIP) vs. necrotizing enterocolitis (NEC). STUDY DESIGN A retrospective review of neonates <1500 g treated for pneumoperitoneum between 07/2004-09/2022 was conducted. Patients treated for NEC medically prior to diagnosis and those treated with drain only were excluded. Fleiss' Kappa analysis assessed agreement between all three diagnoses: preoperative, intraoperative, and pathologic. RESULT Overall, 125 patients were included with mean birthweight 834.2 g (SD:259.2) and mean gestational age 25.8 weeks (SD:2.2). Preoperative and intraoperative diagnoses agreed in 90.3%, intraoperative and pathologic agreed in 71.1%, and preoperative and pathologic agreed in 75.2% of patients. Fleiss' Kappa was 0.55 (95% CI:0.43,0.68), indicating moderate agreement between the three diagnoses. CONCLUSION Our study shows moderate agreement between preoperative, intraoperative, and pathologic diagnoses. Further studies investigating the clinical characteristics of SIP and NEC are needed to improve diagnostic accuracy and management.
Collapse
Affiliation(s)
- Olivia A Keane
- Department of Surgery, Emory University, Atlanta, GA, USA.
| | - Goeto Dantes
- Department of Surgery, Emory University, Atlanta, GA, USA
| | - Valerie L Dutreuil
- Pediatric Biostatistics Core, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Louis Do
- Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Savanah Rumbika
- Emory University School of Medicine, Emory University, Atlanta, GA, USA
| | - Pamela B Sylvestre
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Amina M Bhatia
- Division of Pediatric Surgery, Department of Surgery, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| |
Collapse
|
22
|
Blakely ML, Krzyzaniak A, Dassinger MS, Pedroza C, Weitkamp JH, Gosain A, Cotten M, Hintz SR, Rice H, Courtney SE, Lally KP, Ambalavanan N, Bendel CM, Bui KCT, Calkins C, Chandler NM, Dasgupta R, Davis JM, Deans K, DeUgarte DA, Gander J, Jackson CCA, Keszler M, Kling K, Fenton SJ, Fisher KA, Hartman T, Huang EY, Islam S, Koch F, Lainwala S, Lesher A, Lopez M, Misra M, Overbey J, Poindexter B, Russell R, Stylianos S, Tamura DY, Yoder BA, Lucas D, Shaul D, Ham PB, Fitzpatrick C, Calkins K, Garrison A, de la Cruz D, Abdessalam S, Kvasnovsky C, Segura BJ, Shilyansky J, Smith LM, Tyson JE. Effect of Early vs Late Inguinal Hernia Repair on Serious Adverse Event Rates in Preterm Infants: A Randomized Clinical Trial. JAMA 2024; 331:1035-1044. [PMID: 38530261 PMCID: PMC10966421 DOI: 10.1001/jama.2024.2302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/12/2024] [Indexed: 03/27/2024]
Abstract
Importance Inguinal hernia repair in preterm infants is common and is associated with considerable morbidity. Whether the inguinal hernia should be repaired prior to or after discharge from the neonatal intensive care unit is controversial. Objective To evaluate the safety of early vs late surgical repair for preterm infants with an inguinal hernia. Design, Setting, and Participants A multicenter randomized clinical trial including preterm infants with inguinal hernia diagnosed during initial hospitalization was conducted between September 2013 and April 2021 at 39 US hospitals. Follow-up was completed on January 3, 2023. Interventions In the early repair strategy, infants underwent inguinal hernia repair before neonatal intensive care unit discharge. In the late repair strategy, hernia repair was planned after discharge from the neonatal intensive care unit and when the infants were older than 55 weeks' postmenstrual age. Main Outcomes and Measures The primary outcome was occurrence of any prespecified serious adverse event during the 10-month observation period (determined by a blinded adjudication committee). The secondary outcomes included the total number of days in the hospital during the 10-month observation period. Results Among the 338 randomized infants (172 in the early repair group and 166 in the late repair group), 320 underwent operative repair (86% were male; 2% were Asian, 30% were Black, 16% were Hispanic, 59% were White, and race and ethnicity were unknown in 9% and 4%, respectively; the mean gestational age at birth was 26.6 weeks [SD, 2.8 weeks]; the mean postnatal age at enrollment was 12 weeks [SD, 5 weeks]). Among 308 infants (91%) with complete data (159 in the early repair group and 149 in the late repair group), 44 (28%) in the early repair group vs 27 (18%) in the late repair group had at least 1 serious adverse event (risk difference, -7.9% [95% credible interval, -16.9% to 0%]; 97% bayesian posterior probability of benefit with late repair). The median number of days in the hospital during the 10-month observation period was 19.0 days (IQR, 9.8 to 35.0 days) in the early repair group vs 16.0 days (IQR, 7.0 to 38.0 days) in the late repair group (82% posterior probability of benefit with late repair). In the prespecified subgroup analyses, the probability that late repair reduced the number of infants with at least 1 serious adverse event was higher in infants with a gestational age younger than 28 weeks and in those with bronchopulmonary dysplasia (99% probability of benefit in each subgroup). Conclusions and Relevance Among preterm infants with inguinal hernia, the late repair strategy resulted in fewer infants having at least 1 serious adverse event. These findings support delaying inguinal hernia repair until after initial discharge from the neonatal intensive care unit. Trial Registration ClinicalTrials.gov Identifier: NCT01678638.
Collapse
Affiliation(s)
- Martin L Blakely
- Department of Surgery, Institute for Clinical Research and Learning Healthcare and Institute for Implementation Science, University of Texas Health Science Center, Houston
| | | | - Melvin S Dassinger
- Division of Pediatric Surgery, University of Arkansas for Medical Sciences, Little Rock
| | - Claudia Pedroza
- Department of Pediatrics, Institute for Clinical Research and Learning Healthcare, University of Texas Health Science Center, Houston
| | | | - Ankush Gosain
- Division of Pediatric Surgery, University of Colorado, Aurora
| | - Michael Cotten
- Division of Neonatology, Duke University, Durham, North Carolina
| | - Susan R Hintz
- Division of Neonatology, Stanford University, Palo Alto, California
| | - Henry Rice
- Division of Pediatric Surgery, Duke University, Durham, North Carolina
| | - Sherry E Courtney
- Division of Neonatology, University of Arkansas for Medical Sciences, Little Rock
| | - Kevin P Lally
- Department of Pediatric Surgery, University of Texas Health Science Center, Houston
| | | | | | - Kim Chi T Bui
- Division of Neonatology, Kaiser Permanente, Los Angeles, California
| | - Casey Calkins
- Division of Pediatric Surgery, Medical College of Wisconsin, Milwaukee
| | - Nicole M Chandler
- Division of Pediatric Surgery, Johns Hopkins All Children's Hospital, St Petersburg, Florida
| | - Roshni Dasgupta
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jonathan M Davis
- Division of Neonatology, Tufts Medical Center, Boston, Massachusetts
| | - Katherine Deans
- Department of Pediatric Surgery, Nemours Children's Hospital, Wilmington, Delaware
| | - Daniel A DeUgarte
- Division of Pediatric Surgery, David Geffen School of Medicine, University of California, Los Angeles
| | - Jeffrey Gander
- Division of Pediatric Surgery, University of Virginia, Charlottesville
| | - Carl-Christian A Jackson
- Division of Pediatric Surgery, Alpert Medical School, Brown University, Providence, Rhode Island
| | - Martin Keszler
- Division of Neonatology, Alpert Medical School, Brown University, Providence, Rhode Island
| | - Karen Kling
- Rady Children's Hospital and Division of Pediatric Surgery, University of California, San Diego
| | - Stephen J Fenton
- Division of Pediatric Surgery, University of Utah, Salt Lake City
| | | | - Tyler Hartman
- Division of Neonatology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | - Eunice Y Huang
- Department of Pediatric Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Saleem Islam
- Division of Pediatric Surgery, University of Florida, Gainesville
- Department of Surgery, Aga Khan University, Sindh, Pakistan
| | - Frances Koch
- Division of Neonatology, Medical University of South Carolina, Charleston
| | - Shabnam Lainwala
- Division of Neonatology, Connecticut Children's Medical Center, Hartford
| | - Aaron Lesher
- Division of Pediatric Surgery, Medical University of South Carolina, Charleston
| | - Monica Lopez
- Department of Pediatric Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Meghna Misra
- Pediatric Surgery, Elliot Hospital, Manchester, New Hampshire
| | - Jamie Overbey
- Division of Neonatology, Naval Medical Center, San Diego, California
| | - Brenda Poindexter
- Division of Neonatology, School of Medicine, Emory University, Atlanta, Georgia
| | - Robert Russell
- Division of Pediatric Surgery, University of Alabama at Birmingham
| | - Steven Stylianos
- Division of Pediatric Surgery, Columbia University Medical Center, Morgan Stanley Children's Hospital, New York, New York
| | - Douglas Y Tamura
- Division of Pediatric Surgery, Valley Children's Hospital, Madera, California
| | | | - Donald Lucas
- F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Division of Pediatric Surgery, Naval Medical Center, San Diego, California
| | - Donald Shaul
- Division of Pediatric Surgery, Kaiser Permanente, Los Angeles, California
| | - P Ben Ham
- Division of Pediatric Surgery, University at Buffalo, Buffalo, New York
| | - Colleen Fitzpatrick
- Division of Pediatric Surgery, Cohen Children's Medical Center, Northwell Health, New Hyde Park, New York
| | - Kara Calkins
- Division of Neonatology, David Geffen School of Medicine, University of California, Los Angeles
| | - Aaron Garrison
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Diomel de la Cruz
- Division of Neonatology, School of Medicine, University of Florida, Gainesville
| | - Shahab Abdessalam
- Division of Neonatology, University of Nebraska Medical Center, Omaha
| | | | - Bradley J Segura
- Division of Pediatric Surgery, M Health Fairview University of Minnesota Masonic Children's Hospital, Minneapolis
| | - Joel Shilyansky
- Department of Pediatric Surgery, University of Iowa Stead Family Children's Hospital, Iowa City
| | | | - Jon E Tyson
- Department of Pediatrics, Institute for Clinical Research and Learning Healthcare, University of Texas Health Science Center, Houston
| |
Collapse
|
23
|
Garg PM, Lett K, Ansari MAY, Cunningham H, Ware J, Pittman I, Riddick R, Sawaya D, Berch B, Morris M, Varshney N, Shenberger JS, Taylor C, Reddy K, Hillegass W. Outcomes by disease onset, sex, and intervention in neonates with SIP and surgical NEC. Pediatr Res 2024; 95:1009-1021. [PMID: 37488302 DOI: 10.1038/s41390-023-02749-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 06/29/2023] [Accepted: 07/09/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Outcomes of infants following surgical necrotizing enterocolitis (NEC) and spontaneous intestinal perforation (SIP) categorized by the age of onset, interventions, and sex are not well defined. METHODS Retrospective comparison of infants categorized by age of onset (NEC at <10, 10-20, and >20 days) and SIP at <7 versus ≥7 days), sex, and intervention [Penrose Drain (PD) vs. laparotomy]. RESULTS A total of 114 infants had NEC and 37 had SIP. On multinomial logistic regression, infants with NEC/SIP onset >20 days had significantly lower odds of small bowel involvement (aOR = 0.07, 95% CI: 0.01-0.33, p = 0.001), higher necrosis (aOR = 3.59, 95% CI: 1.34-9.65, p = 0.012) and higher CRP (p = 0.004) than onset <10 days. Initial laparotomy was associated with more bowel loss (24.1 cm [12.3; 40.6] vs.12.1 [8.00; 23.2]; p = 0.001), small and large intestine involvement (47.1% vs 17.2%; p = 0.01), and ileocecal valve resection (42% vs. 19.4%; p = 0.036) than initial PD therapy. Females underwent fewer small bowel resections (52.3% vs 73.6%; p = 0.025) but had higher surgical morbidity (53.7% vs. 24.7%.; p = 0.001) than males. CONCLUSION Clinical, radiological, and histopathological presentation and outcomes in preterm infants with surgical NEC/SIP are associated with age of disease onset, sex, and initial intervention. IMPACT Neonates with surgical NEC onset >20 days had more severe necrosis, inflammation, kidney injury, and bowel loss than those with <10 days. Initial laparotomy was associated with later age onset, more bowel loss, and ileocecal valve resection compared to initial PD treatment, but not with differences in mortality or length of stay. Female sex was associated with lower maturity, more placental malperfusion, less often small bowel involvement, lower pre-NEC hematocrit as well as higher surgical morbidity than males. Whether the management of surgical NEC and SIP should differ by the age of onset requires further investigation.
Collapse
Affiliation(s)
- Parvesh Mohan Garg
- Department of Pediatrics/Neonatology, Wake Forest University, Winston-Salem, NC, USA.
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MI, USA.
| | - Katheryn Lett
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MI, USA
| | - Md Abu Yusuf Ansari
- Department of Data Sciences, University of Mississippi Medical Center, Jackson, MI, USA
| | - Helen Cunningham
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MI, USA
| | - Jennifer Ware
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MI, USA
| | - Isabella Pittman
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MI, USA
| | - Robin Riddick
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, MI, USA
| | - David Sawaya
- Department of Pediatric Surgery, University of Mississippi Medical Center, Jackson, MI, USA
| | - Berry Berch
- Department of Pediatric Surgery, University of Mississippi Medical Center, Jackson, MI, USA
| | - Michael Morris
- Department of Pathology, University of Mississippi Medical Center, Jackson, MI, USA
| | - Neha Varshney
- Department of Pathology, University of Mississippi Medical Center, Jackson, MI, USA
| | - Jeffrey S Shenberger
- Department of Pediatrics/Neonatology, Wake Forest University, Winston-Salem, NC, USA
| | - Charlotte Taylor
- Department of Radiology, University of Mississippi Medical Center, Jackson, MI, USA
| | - Kartik Reddy
- Department of Radiology, University of Mississippi Medical Center, Jackson, MI, USA
| | - William Hillegass
- Department of Data Sciences, University of Mississippi Medical Center, Jackson, MI, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, MI, USA
| |
Collapse
|
24
|
Nair J, Martin CR. Context of time on intestinal injury in preterm infants. Pediatr Res 2024; 95:873-874. [PMID: 37875727 DOI: 10.1038/s41390-023-02844-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 10/26/2023]
Affiliation(s)
- Jayasree Nair
- Division of Neonatology, Weill Cornell Medicine, New York, NY, USA
| | - Camilia R Martin
- Division of Neonatology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
25
|
Colaizy TT, Poindexter BB, McDonald SA, Bell EF, Carlo WA, Carlson SJ, DeMauro SB, Kennedy KA, Nelin LD, Sánchez PJ, Vohr BR, Johnson KJ, Herron DE, Das A, Crawford MM, Walsh MC, Higgins RD, Stoll BJ. Neurodevelopmental Outcomes of Extremely Preterm Infants Fed Donor Milk or Preterm Infant Formula: A Randomized Clinical Trial. JAMA 2024; 331:582-591. [PMID: 38497706 PMCID: PMC10828950 DOI: 10.1001/jama.2023.27693] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/15/2023] [Indexed: 03/19/2024]
Abstract
Importance Maternal milk feeding of extremely preterm infants during the birth hospitalization has been associated with better neurodevelopmental outcomes compared with preterm formula. For infants receiving no or minimal maternal milk, it is unknown whether donor human milk conveys similar neurodevelopmental advantages vs preterm formula. Objective To determine if nutrient-fortified, pasteurized donor human milk improves neurodevelopmental outcomes at 22 to 26 months' corrected age compared with preterm infant formula among extremely preterm infants who received minimal maternal milk. Design, Setting, and Participants Double-blind, randomized clinical trial conducted at 15 US academic medical centers within the Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network. Infants younger than 29 weeks 0 days' gestation or with a birth weight of less than 1000 g were enrolled between September 2012 and March 2019. Intervention Preterm formula or donor human milk feeding from randomization to 120 days of age, death, or hospital discharge. Main Outcomes and Measures The primary outcome was the Bayley Scales of Infant and Toddler Development (BSID) cognitive score measured at 22 to 26 months' corrected age; a score of 54 (score range, 54-155; a score of ≥85 indicates no neurodevelopmental delay) was assigned to infants who died between randomization and 22 to 26 months' corrected age. The 24 secondary outcomes included BSID language and motor scores, in-hospital growth, necrotizing enterocolitis, and death. Results Of 1965 eligible infants, 483 were randomized (239 in the donor milk group and 244 in the preterm formula group); the median gestational age was 26 weeks (IQR, 25-27 weeks), the median birth weight was 840 g (IQR, 676-986 g), and 52% were female. The birthing parent's race was self-reported as Black for 52% (247/478), White for 43% (206/478), and other for 5% (25/478). There were 54 infants who died prior to follow-up; 88% (376/429) of survivors were assessed at 22 to 26 months' corrected age. The adjusted mean BSID cognitive score was 80.7 (SD, 17.4) for the donor milk group vs 81.1 (SD, 16.7) for the preterm formula group (adjusted mean difference, -0.77 [95% CI, -3.93 to 2.39], which was not significant); the adjusted mean BSID language and motor scores also did not differ. Mortality (death prior to follow-up) was 13% (29/231) in the donor milk group vs 11% (25/233) in the preterm formula group (adjusted risk difference, -1% [95% CI, -4% to 2%]). Necrotizing enterocolitis occurred in 4.2% of infants (10/239) in the donor milk group vs 9.0% of infants (22/244) in the preterm formula group (adjusted risk difference, -5% [95% CI, -9% to -2%]). Weight gain was slower in the donor milk group (22.3 g/kg/d [95% CI, 21.3 to 23.3 g/kg/d]) compared with the preterm formula group (24.6 g/kg/d [95% CI, 23.6 to 25.6 g/kg/d]). Conclusions and Relevance Among extremely preterm neonates fed minimal maternal milk, neurodevelopmental outcomes at 22 to 26 months' corrected age did not differ between infants fed donor milk or preterm formula. Trial Registration ClinicalTrials.gov Identifier: NCT01534481.
Collapse
Affiliation(s)
- Tarah T. Colaizy
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City
| | - Brenda B. Poindexter
- Department of Pediatrics, Children’s Healthcare of Atlanta, School of Medicine, Emory University, Atlanta, Georgia
| | - Scott A. McDonald
- Social, Statistical, and Environmental Sciences Unit, RTI International, Research Triangle Park, North Carolina
| | - Edward F. Bell
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City
| | - Waldemar A. Carlo
- Division of Neonatology, Heersink School of Medicine, University of Alabama, Birmingham
| | - Susan J. Carlson
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City
| | - Sara B. DeMauro
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Kathleen A. Kennedy
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Leif D. Nelin
- Department of Pediatrics, Nationwide Children’s Hospital, College of Medicine, Ohio State University, Columbus
| | - Pablo J. Sánchez
- Department of Pediatrics, Nationwide Children’s Hospital, College of Medicine, Ohio State University, Columbus
| | - Betty R. Vohr
- Department of Pediatrics, Women and Infants Hospital, Brown University, Providence, Rhode Island
| | - Karen J. Johnson
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City
| | - Dianne E. Herron
- Department of Pediatrics, School of Medicine, Indiana University, Indianapolis
| | - Abhik Das
- Social, Statistical, and Environmental Sciences Unit, RTI International, Rockville, Maryland
| | - Margaret M. Crawford
- Social, Statistical, and Environmental Sciences Unit, RTI International, Rockville, Maryland
| | - Michele C. Walsh
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Rosemary D. Higgins
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
- College of Health and Human Services, George Mason University, Fairfax, Virginia
| | - Barbara J. Stoll
- Department of Pediatrics, Children’s Healthcare of Atlanta, School of Medicine, Emory University, Atlanta, Georgia
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston
| | | |
Collapse
|
26
|
Garg PP, Riddick R, Ansari MAY, Pittman I, Ladd MR, Porcelli P, Garg PM. Risk factors for postoperative complications in preterm infants with surgical necrotizing enterocolitis and associated outcomes. J Neonatal Perinatal Med 2024; 17:77-89. [PMID: 38217616 PMCID: PMC10939752 DOI: 10.3233/npm-230076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
BACKGROUND We aim to determine clinical risk factors for postoperative complications in preterm infants with surgical necrotizing enterocolitis (NEC) or spontaneous intestinal perforation (SIP). METHODS A retrospective cohort study of preterm infants with surgical NEC or SIP to compare clinical factors between those with and without postoperative complications. RESULTS 78/109 (71.5%) infants had any complication following surgical NEC. Adhesions (20/35, 57.1%) and wound infection (6/35, 17.1%) were the most common single surgical complications. Patients with a single surgical complication (35/66, 53%) were significantly less likely to be exposed to antenatal steroids, more frequently had a jejunostomy, needed a central line longer, and had a longer length of stay than those without any surgical complication. Infants with > 1 surgical complication (43/71, 60.5%) included mainly females, and had AKI more frequently at NEC onset, lower weight z-scores and lower weight for length z- scores at 36 weeks PMA than those without any complications.On multinomial logistic regression, antenatal steroids exposure (OR 0.23 [CI 0.06, 0.84]; p = 0.027) was independently associated with lower risk and jejunostomy 4.81 (1.29, 17.9) was independently associated with higher risk of developing a single complication. AKI following disease onset (OR 5.33 (1.38, 20.6), P = 0.015) was independently associated with > 1 complication in surgical NEC/SIP infants. CONCLUSION Infants with postoperative complications following surgical NEC were more likely to be female, have additional morbidities, and demonstrate growth failure at 36 weeks PMA than those without surgical complications. There was no difference in mortality between those with and without surgical complications.
Collapse
Affiliation(s)
- P P Garg
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - R Riddick
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - M A Y Ansari
- Department of Data Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - I Pittman
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - M R Ladd
- Department of General Surgery/Pediatric Surgery, Atrium Health Wake Forest Baptist, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - P Porcelli
- Department of Pediatrics/Neonatology, Atrium Health Wake Forest Baptist, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - P M Garg
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Pediatrics/Neonatology, Atrium Health Wake Forest Baptist, Wake Forest School of Medicine, Winston Salem, NC, USA
| |
Collapse
|
27
|
Rivero E, Javed F, Manzar S, Bhat R. Laparotomy or peritoneal drainage in preterm infants with spontaneous intestinal perforation or surgical necrotizing enterocolitis? J Perinatol 2024; 44:146-150. [PMID: 37660215 DOI: 10.1038/s41372-023-01770-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 09/04/2023]
Affiliation(s)
- Estephanie Rivero
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Faiza Javed
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Shabih Manzar
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Ramachandra Bhat
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA.
| |
Collapse
|
28
|
Lyle ANJ, Shaikh H, Oslin E, Gray MM, Weiss EM. Race and Ethnicity of Infants Enrolled in Neonatal Clinical Trials: A Systematic Review. JAMA Netw Open 2023; 6:e2348882. [PMID: 38127349 PMCID: PMC10739112 DOI: 10.1001/jamanetworkopen.2023.48882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/08/2023] [Indexed: 12/23/2023] Open
Abstract
Importance Representativeness of populations within neonatal clinical trials is crucial to moving the field forward. Although racial and ethnic disparities in research inclusion are well documented in other fields, they are poorly described within neonatology. Objective To describe the race and ethnicity of infants included in a sample of recent US neonatal clinical trials and the variability in this reporting. Evidence Review A systematic search of US neonatal clinical trials entered into Cochrane CENTRAL 2017 to 2021 was conducted. Two individuals performed inclusion determination, data extraction, and quality assessment independently with discrepancies adjudicated by consensus. Findings Of 120 studies with 14 479 participants that met the inclusion criteria, 75 (62.5%) included any participant race or ethnicity data. In the studies that reported race and ethnicity, the median (IQR) percentage of participants of each background were 0% (0%-1%) Asian, 26% (9%-42%) Black, 3% (0%-12%) Hispanic, 0% (0%-0%) Indigenous (eg, Alaska Native, American Indian, and Native Hawaiian), 0% (0%-0%) multiple races, 57% (30%-68%) White, and 7% (1%-21%) other race or ethnicity. Asian, Black, Hispanic, and Indigenous participants were underrepresented, while White participants were overrepresented compared with a reference sample of the US clinical neonatal intensive care unit (NICU) population from the Vermont Oxford Network. Many participants were labeled as other race or ethnicity without adequate description. There was substantial variability in terms and methods of reporting race and ethnicity data. Geographic representation was heavily skewed toward the Northeast, with nearly one-quarter of states unrepresented. Conclusions and Relevance These findings suggest that neonatal research may perpetuate inequities by underrepresenting Asian, Black, Hispanic, and Indigenous neonates in clinical trials. Studies varied in documentation of race and ethnicity, and there was regional variation in the sites included. Based on these findings, funders and clinical trialists are advised to consider a 3-point targeted approach to address these issues: prioritize identifying ways to increase diversity in neonatal clinical trial participation, agree on a standardized method to report race and ethnicity among neonatal clinical trial participants, and prioritize the inclusion of participants from all regions of the US in neonatal clinical trials.
Collapse
Affiliation(s)
- Allison N J Lyle
- Department of Pediatrics, University of Washington School of Medicine, Seattle
| | - Henna Shaikh
- Department of Pediatrics, University of Washington School of Medicine, Seattle
| | - Ellie Oslin
- Treuman Katz Center for Pediatric Bioethics and Palliative Care, Seattle Children's Research Institute, Seattle, Washington
| | - Megan M Gray
- Department of Pediatrics, University of Washington School of Medicine, Seattle
| | - Elliott Mark Weiss
- Department of Pediatrics, University of Washington School of Medicine, Seattle
- Treuman Katz Center for Pediatric Bioethics and Palliative Care, Seattle Children's Research Institute, Seattle, Washington
| |
Collapse
|
29
|
Lin X, Wu C. Identification and evaluation of probiotic potential of Bifidobacterium breve AHC3 isolated from chicken intestines and its effect on necrotizing enterocolitis (NEC) in newborn SD rats. PLoS One 2023; 18:e0287799. [PMID: 37917716 PMCID: PMC10621988 DOI: 10.1371/journal.pone.0287799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/13/2023] [Indexed: 11/04/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe intestinal disease of the newborn infants, associated with high morbidity and mortality. It has been reported that Bifidobacterium could protect the intestinal barrier function and reduce the risk of NEC. This study aimed to evaluate the probiotic potential of Bifidobacterium strains isolated from the chicken intestines and its effect on necrotizing enterocolitis in newborn SD rats. Out of 32 isolates, B. breve AHC3 not only exhibited excellent probiotic potential, including tolerance to artificial simulated gastric conditions, adhesion to HT-29 cells, antioxidant capacity and antibacterial activity, but also possessed reliable safety. Additionally, NEC model was established to further investigate the effect of B. breve AHC3 on necrotizing enterocolitis in newborn SD rats. It was illustrated that administration of B. breve AHC3 significantly not only reduced the incidence of NEC (from 81.25% to 34.38%) (P< 0.05), but also alleviated the severity of ileal injury (P< 0.05). Compared with NEC model, B. breve AHC3 could significantly decrease the level of proinflammatory factor TNF-α (P< 0.05) and increase the level of antiinflammatory factor IL-10 (P< 0.05) in the ileum of NEC rats. Through the intervention of B. breve AHC3, the gray value of inducible nitric oxide synthase (iNOS) in intestinal tissue of NEC rats was significantly reduced (P< 0.05). It was indicated that B. breve AHC3 exhibited prominent probiotic potential and reliable safety. In the neonatal SD rat model of NEC, B. breve AHC3 had an available protective effect on the intestinal injury of NEC, which might be related to reducing the inflammatory reaction in the ileum and inhibiting the expression of iNOS in intestinal tissue cells. B. breve AHC3 could be used as a potential treatment for human NEC.
Collapse
Affiliation(s)
- Xiaopei Lin
- Department of Pediatrics, Maternity and Child Health Care Hospital Affiliated to Anhui Medical University (Anhui Maternity and Child Health Care Hospital), Hefei, Anhui, China
| | - Changjun Wu
- Institute of Microbiology, Anhui Academy of Medical Sciences, Hefei, Anhui, China
| |
Collapse
|
30
|
Chen Y, Lan C, Zhong W, Song K, Ma Z, Huang L, Zhu Y, Xia H. Plasma anti-myosin autoantibodies in the diagnosis of necrotizing enterocolitis. Eur J Pediatr 2023; 182:5203-5210. [PMID: 37715022 PMCID: PMC10640473 DOI: 10.1007/s00431-023-05188-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/17/2023]
Abstract
We aimed to assess whether autoantibodies can be used as biomarkers for necrotizing enterocolitis (NEC) and applied for its early diagnosis. A prospective observational study was conducted in neonates with suspected NEC abdominal distension (the developmental study), which consisted of 50 neonates finally divided into NEC (n = 24) and non-NEC (n = 26) cohorts based on follow-up results. Serum samples were collected within 48 h of illness onset and used for screening NEC-associated plasma autoantibodies by autoantigen microarray. Additionally, we validated anti-myosin autoantibodies by enzyme-linked immunosorbent assay (ELISA) in an independent validation study, for which we selected plasma samples within 48 h of onset of NEC (n = 38) and samples of gestational age- and weight-matched controls (n = 13). Autoantigen microarray revealed that both IgG and IgM anti-myosin autoantibodies in plasma from neonates with NEC were significantly higher than those in neonates with other diagnoses. ELISA showed that plasma anti-myosin autoantibodies increased in the NEC cohort, with 1.5-fold higher levels than in the non-NEC cohort. Anti-myosin autoantibodies were able to distinguish NEC from non-NEC, achieving an area under the curve (AUC) of 0.8856 (95% confidence interval (CI): 0.7918-0.9795), with sensitivity of 81.58% and specificity of 76.93%. Plasma anti-myosin autoantibodies were significantly higher in all three subtypes of NEC (P < 0.0001 for NEC I; P = 0.0018 for NEC II; P = 0.0011 for NEC III), especially in NEC stage I than that in the non-NEC controls. CONCLUSION Anti-myosin autoantibodies may be applied as a promising diagnostic marker for NEC, especially for NEC stage I. WHAT IS KNOWN • Intestinal damage and self-antigen exposure may lead to increased autoantibodies, and they are widely used as biomarkers for diagnosing inflammatory bowel disease. • Necrotizing enterocolitis (NEC) is a devastating disease with overwhelming inflammation and immune dysregulation. WHAT IS NEW • Increased autoantibodies were present in patients with NEC, even before typical X-ray manifestations. • Anti-myosin autoantibodies may be applied as a promising diagnostic marker for NEC.
Collapse
Affiliation(s)
- Yuqiong Chen
- Department of Pediatrics, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Avenue, Tianhe District, Guangzhou, Guangdong, CN 510630, China
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China
- Department of Pediatrics, The First People's Hospital of Chenzhou, Chenzhou, Hunan, China
| | - Chaoting Lan
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China
| | - Weiyong Zhong
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China
| | - Kai Song
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China
| | - Zuyi Ma
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China
| | - Lihua Huang
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China.
| | - Yun Zhu
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China.
| | - Huimin Xia
- Department of Pediatrics, The First Affiliated Hospital of Jinan University, No. 613 West Huangpu Avenue, Tianhe District, Guangzhou, Guangdong, CN 510630, China.
- Provincial Key Laboratory of Research in Structure Birth Defect Diseaseand, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No.9 Jinsui Road, Zhujiang New Town, Tianhe District, Guangzhou, Guangdong, CN 510623, China.
| |
Collapse
|
31
|
Clyman RI, Hills NK. Prophylactic indomethacin, antenatal betamethasone, and the risk of intestinal perforation in infants <28 weeks' gestation. J Perinatol 2023; 43:1252-1261. [PMID: 36973384 DOI: 10.1038/s41372-023-01653-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/06/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
OBJECTIVE To determine if intestinal perforations before 14 days (either spontaneous (SIP) or necrotizing enterocolitis-induced) are increased when infants who received antenatal betamethasone shortly before birth are treated with prophylactic indomethacin (PINDO). STUDY DESIGN Observational study of 475 infants <28 week's gestation assigned to either a PINDO-protocol (n = 231) or expectant management protocol (n = 244) during consecutive protocol epochs. RESULTS Intestinal perforations before 14 days occurred in 33/475 (7%). In unadjusted and adjusted models, we found no associations between PINDO-protocol and intestinal perforations. PINDO-protocol did not increase intestinal perforations or SIP-alone even when given to infants who received betamethasone <7 or <2 days before delivery. 213/231 (92%) PINDO-protocol infants actually received indomethacin. The results were unchanged when examined just in those who received indomethacin. CONCLUSION In our study, early intestinal perforations and SIP-alone were not increased when PINDO was used by protocol in infants who received antenatal betamethasone shortly before birth.
Collapse
Affiliation(s)
- Ronald I Clyman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
- Department of Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA.
| | - Nancy K Hills
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
32
|
Gao J, Lai D, Tou J. Survey on surgical treatment of neonatal necrotizing enterocolitis in China 2022. WORLD JOURNAL OF PEDIATRIC SURGERY 2023; 6:e000588. [PMID: 37575368 PMCID: PMC10414103 DOI: 10.1136/wjps-2023-000588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Objective The aim of this study was to identify the state of surgical treatment of neonatal necrotizing enterocolitis (NEC) in China. Methods A total of 246 delegates (88.0% senior surgeons) completed a survey sent by the Neonatal Surgery Group of the Pediatric Surgery Branch of the Chinese Medical Association in 2022. Five centers were eliminated due to lack of experience. Results Generally, 38.2% of surgeons work in centers where more than 20 cases of surgical NEC are treated per year. A total of 81.3% of surgeons reported the use of ultrasonography; the most used biomarkers were white blood cell count (95.9%), C-reactive protein (93.8%), and procalcitonin (76.3%). Most surgeons (80.9%) used a combination of two (67.2%) antibiotics or single (29.5%) antibiotic for a treatment period of 7-14 days, and most used antibiotics were carbapenems (73.9%), penicillin and cephalosporins (56.0%). Patients are issued the fasting order for 5-7 days by nearly half surgeons (49.8%) for conservative treatment. 70.1% of surgeons deemed that the most difficult decision was to evaluate the optimal timing of surgery. Most surgeons (76.3%) performed diagnostic aspiration of peritoneal fluid. Laparoscopy was performed for the diagnosis and/or treatment of NEC by 40.2% of surgeons. A total of 53.5% of surgeons reported being able to identify localized intestinal necrosis preoperatively. Surgeons relied the most on pneumoperitoneum (94.2%) and failure of conservative treatment (88.8%) to evaluate the surgical indications. At laparotomy, surgical treatments vary according to NEC severity. Infants are fasted for 5-7 days by 55.2% of surgeons postoperatively. Most surgeons (91.7%) followed up with patients with NEC after discharge for up to 5 years (53.8%). Conclusions The most difficult aspect of surgical NEC is evaluating the timing of surgery, and surgeons in the children's specialized hospitals are experienced. The treatment of NEC totalis is controversial, and the indications for laparoscopy need to be further clarified. More multicenter prospective studies are needed to develop surgical guidelines in the future.
Collapse
Affiliation(s)
- Jiafang Gao
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dengming Lai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Neonatal Surgery Group of the Pediatric Surgery Branch, Chinese Medical Association, Beijing, China
| | - Jinfa Tou
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Neonatal Surgery Group of the Pediatric Surgery Branch, Chinese Medical Association, Beijing, China
| |
Collapse
|
33
|
Bethell GS, Hall NJ. Recent advances in our understanding of NEC diagnosis, prognosis and surgical approach. Front Pediatr 2023; 11:1229850. [PMID: 37583622 PMCID: PMC10424793 DOI: 10.3389/fped.2023.1229850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 07/21/2023] [Indexed: 08/17/2023] Open
Abstract
Necrotising enterocolitis (NEC) remains a devasting condition that has seen limited improvement in outcomes in recent years. The incidence of the disease is increasing as more extremely premature infants survive. NEC is responsible for 1 in 10 neonatal deaths and up to 61% of survivors have significant neurodevelopmental delay. The aim of this review is to highlight recent advances in diagnosis, prognosis and surgical approach in this condition. Many recent studies have reported novel methods of diagnosis of NEC with the aim of earlier and more accurate identification. These include imaging and machine learning techniques. Prognostication of NEC is particularly important to allow earlier escalation of therapy. Around 25% of infants with NEC will require surgery and recent data has shown that time from disease onset to surgery is greater in infants whose indication for surgery is failed medical management, rather than pneumoperitoneum. This indication was also associated with worse outcomes compared to pneumoperitoneum. Ongoing research has highlighted several new methods of disease prognostication which includes differentiating surgical from medical NEC. Finally, recent randomised controlled trials in surgical technique are discussed along with the implications of these for practice. Further, high quality research utilising multi-centre collaborations and high fidelity data from electronic patient records is needed to address the issues discussed and ultimately improve outcomes in NEC.
Collapse
Affiliation(s)
- George S Bethell
- University Surgical Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Nigel J Hall
- University Surgical Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
34
|
Li LT, Hebballi NB, Nguyen T, Morice C, Lally KP. Complication rates in very low and extremely low birth weight infants following laparotomy: a prospective study. Pediatr Surg Int 2023; 39:237. [PMID: 37477761 DOI: 10.1007/s00383-023-05520-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 07/22/2023]
Abstract
INTRODUCTION Surgical site occurrences (SSO), including surgical site infection, dehiscence, and incisional hernia, are complications following laparotomy. SSO rates in premature neonates are poorly understood. We hypothesize that SSO rates are higher among extremely low birth weight (ELBW) infants compared to very low birth weight (VLBW) infants and strive to determine the optimal abdominal closure method for these infants. METHODS We conducted a prospective observational study of infants < 1.5 kg (kg) undergoing laparotomy at two institutions from 1/1/2020 to 5/1/2022. Patients were grouped by weight and closure; SSO rates were computed and the association tested using Fisher's exact test. RESULTS We identified 59 patients and 104 total operations. At initial surgery, 37 patients weighed < 1 kg (ELBW); 22 patients weighed 1-1.5 kg (VLBW). Complication rate for ELBW was 6(16%) vs. 2(9%) in VLBW, but not significant (p = 0.45). More complications followed a single-layer compared to a two-layer closure (18 vs. 2), but not significant (p = 0.30). CONCLUSIONS SSO rates are higher for ELBW infants undergoing laparotomy, and fewer complications follow two-layer closure. However, these findings did not reach statistical significance. Further studies are needed to identify modifiable factors to reduce postoperative complications in these infants.
Collapse
Affiliation(s)
- Linda T Li
- Department of Surgery, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, 10029, USA.
| | - Nutan B Hebballi
- Department of Pediatric Surgery, McGovern Medical School at UTHealth Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Thian Nguyen
- Department of Pediatric Surgery, McGovern Medical School at UTHealth Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Christina Morice
- Department of Pediatric Surgery, McGovern Medical School at UTHealth Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Kevin P Lally
- Department of Pediatric Surgery, McGovern Medical School at UTHealth Houston, 6431 Fannin St., Houston, TX, 77030, USA
- Children's Memorial Hermann Hospital, 6411 Fannin St., Houston, TX, 77030, USA
| |
Collapse
|
35
|
Garg PM, Riddick R, Ansari MAY, Pittman I, Hillegass W. Clinical impact of timing of surgery on outcomes in preterm infants with surgical necrotizing enterocolitis. RESEARCH SQUARE 2023:rs.3.rs-3084887. [PMID: 37461487 PMCID: PMC10350193 DOI: 10.21203/rs.3.rs-3084887/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Background The clinical impact of the timing of surgery on outcomes in preterm infants with surgical necrotizing enterocolitis (NEC) is not well defined. Aim We sought to investigate the impact of the different timing of surgery from the day of NEC diagnosis on clinical outcomes in preterm infants with surgical NEC. Study Design Retrospective Cohort Study. Subjects Preterm 75 infants admitted between January 2013 and December 31, 2018, with an NEC (Bell stage III) diagnosis. Outcomes Comparison of clinical information by the timing of surgery at three different time points (less and more than 48 hours, 96 hours, and 168 hours) in preterm infants with surgical NEC. Results 75 infants were included in the analysis. Those who received surgery after 48 hours (n= 29/75) had lower median gestational age, lower birth weight, had less pneumoperitoneum, were out born less frequently, had higher acute kidney injury, were intubated and ventilated more frequently, and had higher hemorrhagic and reparative lesions on histopathology than those receiving surgery after 48 hours. Infants receiving surgery after 96 hours had similar trends expect had significantly lower hematocrit and more prolonged parenteral nutrition dependence than less than 96 hours group. The infants receiving surgery after one week had significantly lower birth weight and had higher reparative changes and cholestasis than those receiving surgery < 1 week.There was no significant impact of surgery timing on the length of bowel loss, surgical morbidity, Bronchopulmonary dysplasia, white matter injury, and mortality. Conclusion The infants receiving surgery later were young and smaller and received parenteral nutrition longer with no significant impact on morbidities and mortality. Our data point out that there are advantages of operating early with fewer morbidities which need further confirmation and evaluation in large multicentric prospective studies or clinical trials.
Collapse
|
36
|
Keane OA, Zamora AK, Ourshalimian S, Mahdi EM, Song AY, Kim E, Lakshmanan A, Kim ES, Kelley-Quon LI. Opioid and Methadone Use for Infants With Surgically Treated Necrotizing Enterocolitis. JAMA Netw Open 2023; 6:e2318910. [PMID: 37347485 PMCID: PMC10288332 DOI: 10.1001/jamanetworkopen.2023.18910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/02/2023] [Indexed: 06/23/2023] Open
Abstract
Importance Necrotizing enterocolitis (NEC) requiring surgical intervention is the most common reason for surgical procedures in preterm neonates. Opioids are used to manage postoperative pain, with some infants requiring methadone to treat physiologic opioid dependence or wean from nonmethadone opioid treatment during recovery. Objective To describe postoperative opioid use and methadone treatment for infants with surgically treated NEC and evaluate postoperative outcomes. Design, Setting, and Participants A cohort study of infants with surgically treated NEC admitted from January 1, 2013, to December 31, 2022, to 48 Children's Hospital Association hospitals contributing data to the Pediatric Health Information System (PHIS) was performed. Infants who received methadone preoperatively, were aged 14 days or less at the time of the operation, had a congenital heart disease-related operation, or died within 90 days of the operation were excluded. Mixed-effects multivariable logistic regression was used to evaluate thresholds for duration of opioid use after the operation associated with methadone treatment and clinical outcomes associated with methadone use were enumerated. Exposure Postoperative administration of nonmethadone opioids. Main Outcomes and Measures Methadone use and postoperative length of stay, ventilator days, and total parenteral nutrition (TPN) days. Results Of the 2037 infants with surgically treated NEC identified, the median birth weight was 920 (IQR, 700.0-1479.5) g; 1204 were male (59.1%), 911 were White (44.7%), and 343 were Hispanic (16.8%). Infants received nonmethadone opioids for a median of 15 (IQR, 6-30) days after the operation and 231 received methadone (11.3%). The median first day of methadone use was postoperative day 18 (IQR, days 9-64) and continued for 28 days (IQR, 14-73). Compared with infants who received nonmethadone opioids for 1 to 5 days, infants receiving 16 to 21 days of opioids were most likely to receive methadone treatment (odds ratio, 11.45; 95% CI, 6.31-20.77). Methadone use was associated with 21.41 (95% CI, 10.81-32.02) more days of postoperative length of stay, 10.80 (95% CI, 3.63-17.98) more ventilator days, and 16.21 (95% CI, 6.34-26.10) more TPN days. Conclusions and Relevance In this cohort study of infants with surgically treated NEC, prolonged use of nonmethadone opioids after the operation was associated with an increased likelihood of methadone treatment and increased postoperative length of stay, ventilation, and TPN use. Optimizing postoperative pain management for infants requiring an operation may decrease the need for methadone treatment and improve health care use.
Collapse
Affiliation(s)
- Olivia A. Keane
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, California
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles
| | - Abigail K. Zamora
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, California
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles
| | - Shadassa Ourshalimian
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, California
| | - Elaa M. Mahdi
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, California
| | - Ashley Y. Song
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Eugene Kim
- Department of Anesthesia, Children’s Hospital Los Angeles, Los Angeles, California
| | - Ashwini Lakshmanan
- Division of Neonatology, Children’s Hospital Los Angeles, Los Angeles, California
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles
| | - Eugene S. Kim
- Department of Surgery, Cedars-Sinai Health System, Los Angeles, California
| | - Lorraine I. Kelley-Quon
- Division of Pediatric Surgery, Children’s Hospital Los Angeles, Los Angeles, California
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles
| |
Collapse
|
37
|
Klerk DH, van Varsseveld OC, Offringa M, Modi N, Lacher M, Zani A, Pakarinen MP, Koivusalo A, Jester I, Spruce M, Derikx JPM, Bakx R, Ksia A, Vermeulen MJ, Kooi EMW, Hulscher JBF. Development of an international core outcome set for treatment trials in necrotizing enterocolitis-a study protocol. Trials 2023; 24:367. [PMID: 37259112 DOI: 10.1186/s13063-023-07413-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023] Open
Abstract
AIM Necrotizing enterocolitis (NEC) is the most lethal disease of the gastrointestinal tract of preterm infants. New and existing management strategies need clinical evaluation. Large heterogeneity exists in the selection, measurement, and reporting of outcome measures in NEC intervention studies. This hampers meta-analyses and the development of evidence-based management guidelines. We aim to develop a Core Outcome Set (COS) for NEC that includes the most relevant outcomes for patients and physicians, from moment of diagnosis into adulthood. This COS is designed for use in NEC treatment trials, in infants with confirmed NEC. METHODS This study is designed according to COS-STAD (Core Outcome Set-STAndards for Development) recommendations and the COMET (Core Outcome Measures in Effectiveness Trials) Initiative Handbook. We obtained a waiver from the Ethics Review Board and prospectively registered this study with COMET (Study 1920). We will approach 125 clinicians and/or researchers from low-middle and high-income countries based on their scientific output (using SCIVAL, a bibliometric tool). Patients and parents will be approached through local patient organisations. Participants will be separated into three panels, to assess differences in priorities between former patients and parents (1. lay panel), clinicians and researchers involved in the neonatal period (2. neonatal panel) and after the neonatal period (3. post-neonatal panel). They will be presented with outcomes currently used in NEC research, identified through a systematic review, in a Delphi process. Eligible outcome domains are also identified from the patients and parents' perspectives. Using a consensus process, including three online Delphi rounds and a final face-to-face consensus meeting, the COS will be finalised and include outcomes deemed essential to all stakeholders: health care professionals, parents and patients' representatives. The final COS will be reported in accordance with the COS-Standards for reporting (COS-STAR) statement. CONCLUSIONS Development of an international COS will help to improve homogeneity of outcome measure reporting in NEC, will enable adequate and efficient comparison of treatment strategies, and will help the interpretation and implementation of clinical trial results. This will contribute to high-quality evidence regarding the best treatment strategy for NEC in preterm infants.
Collapse
Affiliation(s)
- Daphne H Klerk
- Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Otis C van Varsseveld
- Department of Surgery, Division of Paediatric Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martin Offringa
- Division of Neonatology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Neena Modi
- Section of Neonatal Medicine, School of Public Health, Chelsea and Westminster Hospital campus, Imperial College London, London, UK
| | - Martin Lacher
- Department of Paediatric Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Augusto Zani
- Department of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Mikko P Pakarinen
- Department of Paediatric Surgery, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Antti Koivusalo
- Department of Paediatric Surgery, Children's Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Ingo Jester
- Departments of Paediatric Surgery, Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham, UK
| | | | - Joep P M Derikx
- Department of Paediatric Surgery, UMC, Emma Children's Hospital, Amsterdam, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Roel Bakx
- Department of Paediatric Surgery, UMC, Emma Children's Hospital, Amsterdam, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Amine Ksia
- Department of Surgery, Department of Paediatric Surgery, Monastir Medical School, Fattouma Bourguiba Hospital, Monastir University, Monastir, Tunisia
| | - Marijn J Vermeulen
- Care4Neo, Neonatal Patient and Parent Organization, Rotterdam, the Netherlands
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Elisabeth M W Kooi
- Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan B F Hulscher
- Department of Surgery, Division of Paediatric Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
38
|
Yang J, Shi Y. Paneth cell development in the neonatal gut: pathway regulation, development, and relevance to necrotizing enterocolitis. Front Cell Dev Biol 2023; 11:1184159. [PMID: 37266449 PMCID: PMC10231676 DOI: 10.3389/fcell.2023.1184159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Paneth cells (PCs) are intestinal epithelial cells (IECs) that contain eosinophilic granules, which are located in Lieberkühn crypts. An increasing number of animal and human experiments have indicated that PCs are involved in the progression of a variety of intestinal as well as systemic inflammatory responses including necrotizing enterocolitis (NEC). NEC is an enteric acquired disease with high mortality that usually occurs in premature infants and neonates, however the underlying mechanisms remain unclear. In this review, we summarize the features of PCs, including their immune function, association with gut microbiota and intestinal stem cells, and their mechanism of regulating IEC death to explore the possible mechanisms by which PCs affect NEC.
Collapse
|
39
|
Garg PM, Shenberger JS. Surgical NEC, Randomized Control Trials, Missed Opportunity. J Neonatal Perinatal Med 2023; 16:193-194. [PMID: 37270818 PMCID: PMC10511036 DOI: 10.3233/npm-230048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Affiliation(s)
- P M Garg
- Department of Pediatrics/Neonatology, Atrium Health Wake Forest Baptist, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - J S Shenberger
- Department of Pediatrics/Neonatology, Atrium Health Wake Forest Baptist, Wake Forest School of Medicine, Winston Salem, North Carolina
| |
Collapse
|
40
|
Muacevic A, Adler JR, Basamh HA, Aljohany MS, Bustangi NM. Primary Peritoneal Drainage Versus Laparotomy for Perforated Necrotizing Enterocolitis in Very-Low-Birth-Weight Infants: A Retrospective Cohort Study at an Academic Center in Saudi Arabia. Cureus 2023; 15:e33895. [PMID: 36819445 PMCID: PMC9934917 DOI: 10.7759/cureus.33895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2023] [Indexed: 01/19/2023] Open
Abstract
Background and objective Necrotizing enterocolitis (NEC) is a detrimental complication of the gastrointestinal tract among preterm infants with very low birth weight (VLBW) and is associated with high morbidity and mortality. About one-third of these cases require surgical intervention due to intestinal perforation. The preferred method for the surgical management of perforated NEC is still a matter of controversy. In light of this, we aimed to compare the outcomes of treating perforated NEC in VLBW infants with primary peritoneal drainage (PPD) versus laparotomy. Method We conducted a retrospective chart review of VLBW infants with perforated NEC treated at King Abdulaziz University Hospital between January 1, 2015, and March 31, 2020. Results Twenty-seven infants with perforated NEC were identified; 12 were managed initially with PPD, and 15 underwent laparotomy. There was no difference between groups in terms of postoperative outcomes, length of hospital stay, or mortality before discharge. Among infants managed with PPD, 50% (5/10) underwent second drainage and survived, while 33% (4/12) underwent laparotomy. Conclusion We identified no difference in postoperative outcomes and mortality between managing perforated NEC in VLBW infants with either PPD or laparotomy. However, randomized clinical trials with larger sample sizes and defined outcome measures are needed for reaching definitive conclusions.
Collapse
|
41
|
Manohar K, Mesfin FM, Liu J, Shelley WC, Brokaw JP, Markel TA. Gut-Brain cross talk: The pathogenesis of neurodevelopmental impairment in necrotizing enterocolitis. Front Pediatr 2023; 11:1104682. [PMID: 36873645 PMCID: PMC9975605 DOI: 10.3389/fped.2023.1104682] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating condition of multi-factorial origin that affects the intestine of premature infants and results in high morbidity and mortality. Infants that survive contend with several long-term sequelae including neurodevelopmental impairment (NDI)-which encompasses cognitive and psychosocial deficits as well as motor, vision, and hearing impairment. Alterations in the gut-brain axis (GBA) homeostasis have been implicated in the pathogenesis of NEC and the development of NDI. The crosstalk along the GBA suggests that microbial dysbiosis and subsequent bowel injury can initiate systemic inflammation which is followed by pathogenic signaling cascades with multiple pathways that ultimately lead to the brain. These signals reach the brain and activate an inflammatory cascade in the brain resulting in white matter injury, impaired myelination, delayed head growth, and eventual downstream NDI. The purpose of this review is to summarize the NDI seen in NEC, discuss what is known about the GBA, explore the relationship between the GBA and perinatal brain injury in the setting of NEC, and finally, highlight the existing research into possible therapies to help prevent these deleterious outcomes.
Collapse
Affiliation(s)
- Krishna Manohar
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Fikir M Mesfin
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Jianyun Liu
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - W Christopher Shelley
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - John P Brokaw
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Troy A Markel
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States.,Riley Hospital for Children, Indiana University Health, Indianapolis, IN, United States
| |
Collapse
|
42
|
Yan XL, Liu XC, Zhang YN, Du TT, Ai Q, Gao X, Yang JL, Bao L, Li LQ. Succinate aggravates intestinal injury in mice with necrotizing enterocolitis. Front Cell Infect Microbiol 2022; 12:1064462. [PMID: 36519131 PMCID: PMC9742382 DOI: 10.3389/fcimb.2022.1064462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
Background Necrotizing enterocolitis (NEC) is the most prevalent gastrointestinal disorder that predominantly threatens preterm newborns. Succinate is an emerging metabolic signaling molecule that was recently studied in relation to the regulation of intestinal immunity and homeostasis. We aimed to investigate the relationship between NEC and gut luminal succinate and preliminarily explored the effect of succinate on NEC pathogenesis. Methods Fecal samples from human neonates and mouse pups were analyzed by HPLC - MS/MS and 16S rRNA gene sequencing. C57BL/6 mice were randomly divided into four groups: control, NEC, Lsuc, and Hsuc. The mortality, weight gain, and intestinal pathological changes in four mouse groups were observed. Inflammatory cytokines and markers of macrophages were identified by quantitative real-time PCR. Succinate receptor 1 (SUCNR1) localization was visualized by immunohistochemistry. The protein levels of SUCNR1 and hypoxia-inducible factor 1a (HIF-1a) were quantified by western blotting. Results The levels of succinate in feces from NEC patients were higher than those in feces from non-NEC patients (P <0.05). In the murine models, succinate levels in intestinal content samples were also higher in the NEC group than in the control group (P <0.05). The change in succinate level was closely related to intestinal flora composition. In samples from human neonates, relative to the control group, the NEC group showed a higher abundance of Enterobacteriaceae and a lower abundance of Lactobacillaceae and Lactobacillus (P <0.05). In the murine models, relative to the control group, increased abundance was observed for Clostridiaceae, Enterococcaceae, Clostridium_sensu_stricto_1, and Enterococcus, whereas decreased abundance was observed for Lactobacillaceae and Lactobacillus (P <0.05). Increased succinate levels prevented mice from gaining weight, damaged their intestines, and increased their mortality; upregulated the gene expression of interleukin-1β (IL-1β), IL-6, IL-18 and tumor necrosis factor (TNF); and downregulated the gene expression of IL-10 and transforming growth factor (TGF)-β. Exogenous succinic acid increased inducible nitric oxide synthase (iNOS) gene expression but decreased Arginase-1 (Arg1) gene expression; and increased the protein expression of SUCNR1 and HIF-1a. Conclusion Succinate plays an important role in the development of necrotizing enterocolitis severity, and the activation of the HIF-1a signaling pathway may lead to disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lei Bao
- Department of Neonatology Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lu-Quan Li
- Department of Neonatology Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
43
|
Solis-Garcia G, Jasani B. Laparotomy vs. peritoneal drainage: the need for better evidence : Letter to editor on the manuscript "Initial surgical treatment of necrotizing enterocolitis: a meta-analysis of peritoneal drainage versus laparotomy". Eur J Pediatr 2022; 181:3559-3560. [PMID: 35821132 DOI: 10.1007/s00431-022-04556-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/10/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Gonzalo Solis-Garcia
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada.
- Division of Neonatology, Hospital for Sick Children, 666 University Avenue, Toronto, ON, Canada.
| | - Bonny Jasani
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
- Division of Neonatology, Hospital for Sick Children, 666 University Avenue, Toronto, ON, Canada
| |
Collapse
|
44
|
Li W, Tang J, Zhu Z, Tang W. Initial surgical treatment of necrotizing enterocolitis: a meta-analysis of peritoneal drainage versus laparotomy. Eur J Pediatr 2022; 181:2593-2601. [PMID: 35451633 DOI: 10.1007/s00431-022-04454-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 11/29/2022]
Abstract
Necrotizing enterocolitis (NEC) in premature infants is associated with high morbidity and mortality, and the optimal intervention remains uncertain. To compare the mortality of primary peritoneal drainage versus primary peritoneal laparotomy as initial surgical intervention for NEC. All data were extracted from PubMed, Embase, and the Cochrane Library. Studies published up to December 2021. Patients with NEC. Studies centered on primary peritoneal drainage and primary peritoneal laparotomy as the initial surgical treatment. Mortality outcomes were available for both interventions. Randomized controlled trials, retrospective cohort studies, and case series in peer-reviewed journals. Language limited to English. Odds ratio (OR) with 95% confidence intervals (CIs) was used to evaluate mortality outcome. Subgroup analyses and linear regression were performed to ascertain the association between mortality pre-specified factors. Data of 1062 patients received peritoneal drainage and 2185 patients received peritoneal laparotomy from five case series, five retrospective cohort studies, and three randomized controlled trials. Peritoneal drainage caused similar mortality (OR 1.49, 95% CI 0.99-2.26) compared with peritoneal laparotomy as initial surgical management for NEC infants. The subgroup analysis of study design, sample size, birth weight, and sex showed similar findings, but inconsistent results were found for country (USA: 1.47, 95% CI 0.90-2.41; Canada: 2.53, 95% CI 0.30-21.48; Australia: 10.29, 95% CI 1.03-102.75; Turkey: 0.09, 95% CI 0.01-0.63) and gestational age (age mean difference < 3: 1.23, 95% CI 0.72-2.11; age mean difference ≥ 3: 2.29, 95% CI 1.04-5.05). No statistically significance was found for the linear regression between mortality and sample size (P = 0.842), gestational age (P = 0.287), birth weight (P = 0.257), sex (P = 0.6). Small sample size, high heterogeneity, NEC, and spontaneous intestinal perforation (SIP) had to be analyzed together, lack of selection criteria for the future selection of an intervention, and no clear, standardized procedures. Conclusion: There was no significant difference in mortality between peritoneal drainage and laparotomy as initial surgical intervention. The results suggest that either intervention could be used in selected patients. What is Known: • Necrotizing enterocolitis (NEC) in premature infants is associated with high morbidity and mortality, and the optimal intervention remains uncertain. What is New: • No significant difference of mortality between peritoneal drainage and laparotomy as initial surgical intervention.
Collapse
Affiliation(s)
- Wei Li
- Department of Neonatal Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Tang
- Department of Neonatal Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongxian Zhu
- Department of Neonatal Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Weibing Tang
- Department of Neonatal Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
45
|
Bench to bedside - new insights into the pathogenesis of necrotizing enterocolitis. Nat Rev Gastroenterol Hepatol 2022; 19:468-479. [PMID: 35347256 DOI: 10.1038/s41575-022-00594-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2022] [Indexed: 12/12/2022]
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of death and disability from gastrointestinal disease in premature infants. Recent discoveries have shed light on a unifying theorem to explain the pathogenesis of NEC, suggesting that specific treatments might finally be forthcoming. A variety of experiments have highlighted how the interaction between bacterial signalling receptors on the premature intestine and an abnormal gut microbiota incites a pro-inflammatory response in the intestinal mucosa and its underlying endothelium that leads to NEC. Central amongst the bacterial signalling receptors implicated in NEC development is the lipopolysaccharide receptor Toll-like receptor 4 (TLR4), which is expressed at higher levels in the premature gut than in the full-term gut. The high prenatal intestinal expression of TLR4 reflects the role of TLR4 in the regulation of normal gut development, and supports additional studies indicating that NEC develops in response to signalling events that occur in utero. This Review provides new evidence explaining the pathogenesis of NEC, explores new findings indicating that NEC development has origins before birth, and discusses future questions and opportunities for discovery in this field.
Collapse
|
46
|
Swanson JR, Hair A, Clark RH, Gordon PV. Spontaneous intestinal perforation (SIP) will soon become the most common form of surgical bowel disease in the extremely low birth weight (ELBW) infant. J Perinatol 2022; 42:423-429. [PMID: 35177793 DOI: 10.1038/s41372-022-01347-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 01/25/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022]
Abstract
Recent data have revealed declines in the prevalence rates of NEC over the last decade in premature infants. In contrast, SIP has either remained steady or risen during the same epoch. These trends are consistent with our knowledge of the clinical arena. The ability to discern SIP contamination within NEC datasets has slowly improved. Additionally, quality improvement efforts are being utilized to reduce NEC through stewardship of antibiotics, acid inhibitors, central lines and blood products, as well as optimization of human milk diets. These forces are moving us to a new era, where NEC will no longer be the dominant surgical intestinal disease of the extremely preterm neonate. Indeed, in the extremely low birth weight (ELBW) population, SIP may already be the most prevalent reason for abdominal surgery. In this perspective, the reader will find supporting data and references for these assertions as well as predictions for the future.
Collapse
Affiliation(s)
- Jonathan R Swanson
- Division of Neonatology, University of Virginia Children's Hospital, Charlottesville, VA, USA.
| | - Amy Hair
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | - Reese H Clark
- Pediatrix-Obstetrix Center for Research and Education, Sunrise, FL, USA
| | | |
Collapse
|