1
|
Quigley M, Embleton ND, Meader N, McGuire W. Donor human milk for preventing necrotising enterocolitis in very preterm or very low-birthweight infants. Cochrane Database Syst Rev 2024; 9:CD002971. [PMID: 39239939 PMCID: PMC11378496 DOI: 10.1002/14651858.cd002971.pub6] [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] [Indexed: 09/07/2024]
Abstract
BACKGROUND When sufficient maternal milk is not available, donor human milk or formula are the alternative forms of enteral nutrition for very preterm or very low-birthweight (VLBW) infants. Donor human milk may retain the non-nutritive benefits of maternal milk and has been proposed as a strategy to reduce the risk of necrotising enterocolitis (NEC) and associated mortality and morbidity in very preterm or VLBW infants. OBJECTIVES To assess the effectiveness of donor human milk compared with formula for preventing NEC and associated morbidity and mortality in very preterm or VLBW infants when sufficient maternal milk is not available. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, the Maternity and Infant Care (MIC) database, and the Cumulative Index to Nursing and Allied Health Literature (CINAHL), from the earliest records to February 2024. We searched clinical trials registries and examined the reference lists of included studies. SELECTION CRITERIA Randomised or quasi-randomised controlled trials comparing feeding with donor human milk versus formula in very preterm (< 32 weeks' gestation) or VLBW (< 1500 g) infants. DATA COLLECTION AND ANALYSIS Two review authors evaluated the risk of bias in the trials, extracted data, and synthesised effect estimates using risk ratio, risk difference, and mean difference, with associated 95% confidence intervals. The primary outcomes were NEC, late-onset invasive infection, and all-cause mortality before hospital discharge. The secondary outcomes were growth parameters and neurodevelopment. We used the GRADE approach to assess the certainty of the evidence for our primary outcomes. MAIN RESULTS Twelve trials with a total of 2296 infants fulfilled the inclusion criteria. Most trials were small (average sample size was 191 infants). All trials were performed in neonatal units in Europe or North America. Five trials were conducted more than 40 years ago; the remaining seven trials were conducted in the year 2000 or later. Some trials had methodological weaknesses, including concerns regarding masking of investigators and selective reporting. Meta-analysis showed that donor human milk reduces the risk of NEC (risk ratio (RR) 0.53, 95% confidence interval (CI) 0.37 to 0.76; I² = 4%; risk difference (RD) -0.03, 95% CI -0.05 to -0.01; 11 trials, 2261 infants; high certainty evidence). Donor human milk probably has little or no effect on late-onset invasive infection (RR 1.12, 0.95 to 1.31; I² = 27%; RD 0.03, 95% CI -0.01 to -0.07; 7 trials, 1611 infants; moderate certainty evidence) or all-cause mortality (RR 1.00, 95% CI 0.76 to 1.31; I² = 0%; RD -0.00, 95% CI -0.02 to 0.02; 9 trials, 2116 infants; moderate certainty evidence). AUTHORS' CONCLUSIONS The evidence shows that donor human milk reduces the risk of NEC by about half in very preterm or VLBW infants. There is probably little or no effect on late-onset invasive infection or all-cause mortality before hospital discharge.
Collapse
Key Words
- humans
- infant, newborn
- bias
- enteral nutrition
- enteral nutrition/methods
- enterocolitis, necrotizing
- enterocolitis, necrotizing/epidemiology
- enterocolitis, necrotizing/prevention & control
- infant formula
- infant, extremely premature
- infant, premature
- infant, premature, diseases
- infant, premature, diseases/mortality
- infant, premature, diseases/prevention & control
- infant, very low birth weight
- milk, human
- randomized controlled trials as topic
Collapse
MESH Headings
- Humans
- Infant, Newborn
- Bias
- Enteral Nutrition/methods
- Enterocolitis, Necrotizing/epidemiology
- Enterocolitis, Necrotizing/prevention & control
- Infant Formula
- Infant, Extremely Premature
- Infant, Premature
- Infant, Premature, Diseases/prevention & control
- Infant, Premature, Diseases/mortality
- Infant, Very Low Birth Weight
- Milk, Human
- Randomized Controlled Trials as Topic
Collapse
Affiliation(s)
- Maria Quigley
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Nicholas D Embleton
- Newcastle Neonatal Service , Newcastle Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | | | - William McGuire
- Centre for Reviews and Dissemination , University of York, York, UK
| |
Collapse
|
2
|
Getahun BA, Mulatu S, Workie HM. Time to Reach Full Enteral Feeding and Its Predictors among Very Low Birth Weight Neonates Admitted in the Neonatal Intensive Care Unit: A Follow-Up Cohort Study. J Nutr Metab 2024; 2024:9384734. [PMID: 38957371 PMCID: PMC11217576 DOI: 10.1155/2024/9384734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/18/2024] [Accepted: 06/08/2024] [Indexed: 07/04/2024] Open
Abstract
Background Feeding is crucial for very low birth weight neonates to grow and develop properly. This study aims to determine the time to achieve full enteral feeding and predictors among neonates admitted at Felege Hiwot Comprehensive Specialized Hospital. Methods An institutional retrospective follow-up study design was conducted among 332 very low birth weight neonates from July 1, 2018, to June 30, 2021. Samples were selected through a computer-generated simple random sampling method, and the data were entered into Epi data version 4.6 and then exported to STATA version 16 for analysis. Kaplan-Meier with the log-rank test was used to test for the presence of difference in survival among predictor variables. Model goodness of fit and assumptions were checked by the Cox-Snell residual and the global test, respectively. Variables with p value <0.25 in the bi-variable analysis were fitted to the multivariable Cox-proportional hazard model. Finally, the adjusted hazard ratio (AHR) with 95% CI was computed, and variables with a p value less than 0.05 in the multivariable Cox regression analysis were considered significant predictors of time to reach full enteral feeding. Results A total of 332 neonates were followed for 2,132 person days of risk time and 167 (50.3%) of very low birth weight neonates started full enteral feeding. The overall incidence rate of full enteral feeding was 7.8 per 100 person day observations. The median survival time was 7 days. Very low birth weight neonates delivered from pregnancy-induced hypertension-free mothers (AHR: 2.1; 95% CI: 1.12, 3.94), gestational age of ≥33 weeks (AHR: 5,; 95% CI: 2.29, 11.13), kangaroo mother care initiated (AHR: 1.4; 95% CI: 1.01, 2.00), avoiding prefeed residual aspiration (AHR: 1.42; 95% CI: 1.002-2.03), and early enteral feeding (AHR: 1.5; 95% CI: 1.03, 2.35) were significant predictors of full enteral feeding. Conclusions According to this study, the time to achieve full enteral feeding was relatively short. Therefore, healthcare professionals should emphasize achieving full enteral feeding and address hindering factors to save the lives of VLBW neonates.
Collapse
Affiliation(s)
| | - Sileshi Mulatu
- Bahir Dar UniversityCollege of Medical and Health SciencesDepartment of PCHN, Bahir Dar, Ethiopia
| | | |
Collapse
|
3
|
Esubalew H, Messelu MA, Tarekegn BT, Admasu AT, Abrha NN, Terefe B. Time to full enteral feeding and its predictors among very low birth weight (VLBW) neonates admitted to the neonatal intensive care units (NICU) in comprehensive specialized hospitals in Northwest Ethiopia. BMC Pediatr 2024; 24:366. [PMID: 38807061 PMCID: PMC11131264 DOI: 10.1186/s12887-024-04719-w] [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: 12/16/2023] [Accepted: 03/22/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Time to full enteral feeding is the time when neonates start to receive all of their prescribed nutrition as milk feeds. Delayed to achieve full enteral feeding had resulted in short- and long-term physical and neurological sequelae. However, there are limited studies to assess the time to full enteral feeding and its predictors among very low birth-weight neonates in Ethiopia. Therefore, this study aimed to assess the time to full enteral feeding and its predictors among very low birth-weight neonates admitted to comprehensive specialized hospitals in Northwest Ethiopia. METHODS A multi-center institutional-based retrospective follow-up study was conducted among 409 VLBW neonates from March 1, 2019 to February 30, 2023. A simple random sampling method was used to select study participants. Data were entered into EpiData version 4.2 and then exported into STATA version 16 for analysis. The Kaplan-Meier survival curve together with the log-rank test was fitted to test for the presence of differences among groups. Proportional hazard assumptions were checked using a global test. Variables having a p- value < 0.25 in the bivariable Cox-proportional hazard model were candidates for multivariable analysis. An adjusted Hazard Ratio (AHR) with 95% Confidence Intervals (CI) was computed to report the strength of association, and variables having a P-value < 0.05 at the 95% confidence interval were considered statistically significant predictor variables. RESULT The median time to full enteral feeding was 10 (CI: 10-11) days. Very Low Birth-Weight (VLBW) neonates who received a formula feeding (AHR: 0.71, 95% CI: 0.53, 0.96), gestational age of 32-37 weeks (AHR: 1.66, 95% CI: 1.23, 2.23), without Necrotizing Enterocolitis (NEC) (AHR: 2.16, 95% CI: 1.65, 2.84), and single birth outcome (AHR: 1.42, 95% CI: 1.07, 1.88) were statistically significant variables with time to full enteral feeding. CONCLUSION AND RECOMMENDATIONS This study found that the median time to full enteral feeding was high. Type of feeding, Necrotizing Enterocolitis (NEC), Gestational Age (GA) at birth, and birth outcome were predictor variables. Special attention and follow-up are needed for those VLBW neonates with NEC, had a GA of less than 32 weeks, and had multiple birth outcomes.
Collapse
Affiliation(s)
- Hilena Esubalew
- Felege Hiwot Comprehensive Specialized Hospital, Bahir Dar, Ethiopia.
| | - Mengistu Abebe Messelu
- College of Medicine and Health Sciences, Department of Nursing, Debre Markos University, Debre Markos, Ethiopia
| | - Bethelihem Tigabu Tarekegn
- College of Medicine and Health Sciences, School of Nursing, Department of Pediatrics and Child Health, University of Gondar, Gondar, Ethiopia
| | - Aster Tefera Admasu
- College of Medicine and health Sciences, School of Medicine, Department of Biomedical Sciences, University of Gondar, Gondar, Ethiopia
| | - Nega Nigussie Abrha
- College of Medicine and Health Sciences, School of Nursing, Department of Emergency and Critical Care Nursing, Univiersity of Gondar, Gondar, Ethiopia
| | - Bewuketu Terefe
- College of Medicine and Health Sciences, School of Nursing, Department of Community Health Nursing, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
4
|
Healy D, Wang S, Grimaud G, Warda AK, Ross P, Stanton C, Dempsey EM. Longitudinal observational study protocol - Preterm Infants: Microbiome Establishment, Neuro-CrossTalk and Origins (PIMENTO). BMJ Open 2023; 13:e075060. [PMID: 37748849 PMCID: PMC10533688 DOI: 10.1136/bmjopen-2023-075060] [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: 04/25/2023] [Accepted: 08/17/2023] [Indexed: 09/27/2023] Open
Abstract
INTRODUCTION Very preterm infants are at risk of abnormal microbiome colonisation in the first weeks to months of life. Several important associated factors have been identified including gestational age, mode of delivery, antibiotic exposure and feeding. Preterm infants are at risk of a number of pathologies for which the microbiome may play a central role, including necrotising enterocolitis and sepsis. The objective of this study is to determine detailed microbiome changes that occur around implementation of different management practices including empiric antibiotic use, advancement of feeds and administration of probiotics during admission to the neonatal intensive care unit. METHODS AND ANALYSIS A single-site, longitudinal observational study of infants born less than 32 weeks gestation, including collection of maternal samples around delivery and breastmilk and infant samples from admission through discharge from the neonatal unit. ETHICS AND DISSEMINATION The protocol was approved by the Clinical Research Ethics Committee of the Cork Teaching Hospitals.The findings from this study will be disseminated in peer-reviewed journals, during scientific conferences, and directly to the study participants. Sequencing data will be deposited in public databases. TRIAL REGISTRATION NUMBER NCT05803577.
Collapse
Affiliation(s)
- David Healy
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Shuo Wang
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Alicja Katarzyna Warda
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre Moorepark, Moorepark, Ireland
| | - Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre Moorepark, Moorepark, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre Moorepark, Moorepark, Ireland
| | - Eugene M Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
| |
Collapse
|
5
|
Quitadamo PA, Zambianco F, Palumbo G, Copetti M, Gentile MA, Mondelli A. Trend and Predictors of Breastmilk Feeding among Very-Low-Birth-Weight Infants in NICU and at Discharge. Nutrients 2023; 15:3314. [PMID: 37571252 PMCID: PMC10421341 DOI: 10.3390/nu15153314] [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: 04/30/2023] [Revised: 07/10/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Mothers' own milk (MOM) for premature babies is considered a life-saving drug for its proven protective action against the complications of prematurity and for effects on outcome in the short and long term, especially neurological ones. We studied the use of MOM for infants weighing <1500 g for a period of 5 years, evaluating the trend over time and the impact of some variables on human milk feeding performance. Statistical comparisons concerned the rate of feeding with breast milk during a stay in an NICU and at discharge with respect to two types of variables: (1) maternal and neonatal characteristics (gestational age, birth weight, type of pregnancy (whether single or twin), maternal age) and (2) feeding characteristics (time of the start of minimal enteral feeding and availability of MOM, days until the achievement of full enteral feeding). Group comparisons were performed using ANOVA or t-test for continuous variables and Pearson chi-squared test or Fisher exact test for categorical variables. We observed an increase, between 2017 and 2021, in MOM use (p = 0.003). The availability of the own mothers' milk occurred, on average, on the fourth day of life and improved over the years. The start of minimal enteral feeding (MEF) with human milk averaged 1.78 days, and 54.3% of VLBWs received MEF with donor milk on the first day of life. The average percentage of feeding with the mothers' milk at discharge was 47.6%, with 36.1% of exclusive MOM and an increase from 45.8% in 2017 (33.3% exclusive) to 58.82% (41.18% exclusive) in 2021. The mean average daily growth of the weight improved (p < 0.001) during this period, and there was no statistical difference between infants fed with maternal milk and those fed with bank milk. Older maternal age, early-start feeding with maternal milk and low gestational age had a statistically significant impact on feeding with MOM at discharge.
Collapse
Affiliation(s)
- Pasqua Anna Quitadamo
- Neonatal Intensive Care Unit, Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (G.P.); (M.A.G.); (A.M.)
- HMB, Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Federica Zambianco
- San Raffaele Faculty of Medicine, University of San Raffaele Vita-Salute, 20132 Milano, Italy;
| | - Giuseppina Palumbo
- Neonatal Intensive Care Unit, Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (G.P.); (M.A.G.); (A.M.)
- HMB, Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Massimiliano Copetti
- Statistical Department, Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Maria Assunta Gentile
- Neonatal Intensive Care Unit, Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (G.P.); (M.A.G.); (A.M.)
- HMB, Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Antonio Mondelli
- Neonatal Intensive Care Unit, Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (G.P.); (M.A.G.); (A.M.)
- HMB, Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| |
Collapse
|
6
|
Sharif S, Meader N, Oddie SJ, Rojas-Reyes MX, McGuire W. Probiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Database Syst Rev 2023; 7:CD005496. [PMID: 37493095 PMCID: PMC10370900 DOI: 10.1002/14651858.cd005496.pub6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
BACKGROUND Intestinal dysbiosis may contribute to the pathogenesis of necrotising enterocolitis (NEC) in very preterm or very low birth weight (VLBW) infants. Dietary supplementation with probiotics to modulate the intestinal microbiome has been proposed as a strategy to reduce the risk of NEC and associated mortality and morbidity in very preterm or VLBW infants. OBJECTIVES To determine the effect of supplemental probiotics on the risk of NEC and associated mortality and morbidity in very preterm or very low birth weight infants. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, the Maternity and Infant Care database, and CINAHL from inception to July 2022. We searched clinical trials databases and conference proceedings, and examined the reference lists of retrieved articles. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs comparing probiotics with placebo or no probiotics in very preterm infants (born before 32 weeks' gestation) and VLBW infants (weighing less than 1500 g at birth). DATA COLLECTION AND ANALYSIS Two review authors independently evaluated risk of bias of the trials, extracted data, and synthesised effect estimates using risk ratios (RRs), risk differences (RDs), and mean differences (MDs), with associated 95% confidence intervals (CIs). The primary outcomes were NEC and all-cause mortality; secondary outcome measures were late-onset invasive infection (more than 48 hours after birth), duration of hospitalisation from birth, and neurodevelopmental impairment. We used the GRADE approach to assess the certainty of the evidence. MAIN RESULTS We included 60 trials with 11,156 infants. Most trials were small (median sample size 145 infants). The main potential sources of bias were unclear reporting of methods for concealing allocation and masking caregivers or investigators in about half of the trials. The formulation of the probiotics varied across trials. The most common preparations contained Bifidobacterium spp., Lactobacillus spp., Saccharomyces spp., andStreptococcus spp., alone or in combination. Very preterm or very low birth weight infants Probiotics may reduce the risk of NEC (RR 0.54, 95% CI 0.46 to 0.65; I² = 17%; 57 trials, 10,918 infants; low certainty). The number needed to treat for an additional beneficial outcome (NNTB) was 33 (95% CI 25 to 50). Probiotics probably reduce mortality slightly (RR 0.77, 95% CI 0.66 to 0.90; I² = 0%; 54 trials, 10,484 infants; moderate certainty); the NNTB was 50 (95% CI 50 to 100). Probiotics probably have little or no effect on the risk of late-onset invasive infection (RR 0.89, 95% CI 0.82 to 0.97; I² = 22%; 49 trials, 9876 infants; moderate certainty). Probiotics may have little or no effect on neurodevelopmental impairment (RR 1.03, 95% CI 0.84 to 1.26; I² = 0%; 5 trials, 1518 infants; low certainty). Extremely preterm or extremely low birth weight infants Few data were available for extremely preterm or extremely low birth weight (ELBW) infants. In this population, probiotics may have little or no effect on NEC (RR 0.92, 95% CI 0.69 to 1.22, I² = 0%; 10 trials, 1836 infants; low certainty), all-cause mortality (RR 0.92, 95% CI 0.72 to 1.18; I² = 0%; 7 trials, 1723 infants; low certainty), or late-onset invasive infection (RR 0.93, 95% CI 0.78 to 1.09; I² = 0%; 7 trials, 1533 infants; low certainty). No trials provided data for measures of neurodevelopmental impairment in extremely preterm or ELBW infants. AUTHORS' CONCLUSIONS Given the low to moderate certainty of evidence for the effects of probiotic supplements on the risk of NEC and associated morbidity and mortality for very preterm or VLBW infants, and particularly for extremely preterm or ELBW infants, there is a need for further large, high-quality trials to provide evidence of sufficient validity and applicability to inform policy and practice.
Collapse
Key Words
- female
- humans
- infant
- infant, newborn
- enterocolitis, necrotizing
- enterocolitis, necrotizing/epidemiology
- fetal growth retardation
- infant, extremely premature
- infant, premature, diseases
- infant, premature, diseases/etiology
- infant, premature, diseases/prevention & control
- infant, very low birth weight
- probiotics
Collapse
Affiliation(s)
- Sahar Sharif
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Nicholas Meader
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sam J Oddie
- Centre for Reviews and Dissemination, University of York, York, UK
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Maria X Rojas-Reyes
- Institut d'Recerca Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- Evaluation Unit of the Canary Islands Health Service (SESCS), Tenerife, Spain
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
7
|
Sharif S, Oddie SJ, Heath PT, McGuire W. Prebiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Database Syst Rev 2023; 6:CD015133. [PMID: 37262358 PMCID: PMC10234253 DOI: 10.1002/14651858.cd015133.pub2] [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] [Indexed: 06/03/2023]
Abstract
BACKGROUND Dietary supplementation with prebiotic oligosaccharides to modulate the intestinal microbiome has been proposed as a strategy to reduce the risk of necrotising enterocolitis (NEC) and associated mortality and morbidity in very preterm or very low birth weight (VLBW) infants. OBJECTIVES To assess the benefits and harms of enteral supplementation with prebiotics (versus placebo or no treatment) for preventing NEC and associated morbidity and mortality in very preterm or VLBW infants. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, the Maternity and Infant Care database and the Cumulative Index to Nursing and Allied Health Literature (CINAHL), from the earliest records to July 2022. We searched clinical trials databases and conference proceedings, and examined the reference lists of retrieved articles. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs comparing prebiotics with placebo or no prebiotics in very preterm (< 32 weeks' gestation) or VLBW (< 1500 g) infants. The primary outcomes were NEC and all-cause mortality, and the secondary outcomes were late-onset invasive infection, duration of hospitalisation since birth, and neurodevelopmental impairment. DATA COLLECTION AND ANALYSIS Two review authors separately evaluated risk of bias of the trials, extracted data, and synthesised effect estimates using risk ratio (RR), risk difference (RD), and mean difference (MD), with associated 95% confidence intervals (CIs). The primary outcomes of interest were NEC and all-cause mortality; our secondary outcome measures were late-onset (> 48 hours after birth) invasive infection, duration of hospitalisation, and neurodevelopmental impairment. We used the GRADE approach to assess the level of certainty of the evidence. MAIN RESULTS We included seven trials in which a total of 705 infants participated. All the trials were small (mean sample size 100). Lack of clarity on methods to conceal allocation and mask caregivers or investigators were potential sources of bias in three of the trials. The studied prebiotics were fructo- and galacto-oligosaccharides, inulin, and lactulose, typically administered daily with enteral feeds during birth hospitalisation. Meta-analyses of data from seven trials (686 infants) suggest that prebiotics may result in little or no difference in NEC (RR 0.97, 95% CI 0.60 to 1.56; RD none fewer per 1000, 95% CI 50 fewer to 40 more; low-certainty evidence), all-cause mortality (RR 0.43, 95% CI 0.20 to 0.92; 40 per 1000 fewer, 95% CI 70 fewer to none fewer; low-certainty evidence), or late-onset invasive infection (RR 0.79, 95% CI 0.60 to 1.06; 50 per 1000 fewer, 95% CI 100 fewer to 10 more; low-certainty evidence) prior to hospital discharge. The certainty of this evidence is low because of concerns about the risk of bias in some trials and the imprecision of the effect size estimates. The data available from one trial provided only very low-certainty evidence about the effect of prebiotics on measures of neurodevelopmental impairment (Bayley Scales of Infant Development (BSID) Mental Development Index score < 85: RR 0.84, 95% CI 0.25 to 2.90; very low-certainty evidence; BSID Psychomotor Development Index score < 85: RR 0.24, 95% 0.03 to 2.00; very low-certainty evidence; cerebral palsy: RR 0.35, 95% CI 0.01 to 8.35; very low-certainty evidence). AUTHORS' CONCLUSIONS The available trial data provide low-certainty evidence about the effects of prebiotics on the risk of NEC, all-cause mortality before discharge, and invasive infection, and very low-certainty evidence about the effect on neurodevelopmental impairment for very preterm or VLBW infants. Our confidence in the effect estimates is limited; the true effects may be substantially different. Large, high-quality trials are needed to provide evidence of sufficient validity to inform policy and practice decisions.
Collapse
Key Words
- humans
- infant, newborn
- enterocolitis, necrotizing
- enterocolitis, necrotizing/etiology
- enterocolitis, necrotizing/prevention & control
- infant, extremely premature
- infant, premature, diseases
- infant, premature, diseases/etiology
- infant, premature, diseases/prevention & control
- infant, very low birth weight
- infections
Collapse
Affiliation(s)
- Sahar Sharif
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sam J Oddie
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Paul T Heath
- Division of Child Health and Vaccine Institute, St. George's, University of London, London, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
8
|
Yoon SA, Lee MH, Chang YS. Impact of time to full enteral feeding on long-term neurodevelopment without mediating by postnatal growth failure in very-low-birth-weight-infants. Sci Rep 2023; 13:2990. [PMID: 36804430 PMCID: PMC9941577 DOI: 10.1038/s41598-023-29646-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
This study aimed to determine if time to achieve full enteral feeding (TFF) directly impacted long-term neurodevelopmental delay (NDD) and whether long-term postnatal growth failure (PGF) was a mediator of this association in very-low-birth-weight (VLBW) infants. Using prospectively collected cohort data from the Korean Neonatal Network, we included eligible VLBW infants who achieved TFF at least once and classified enrolled infants into four groups using exposure severity (P1 to P4 as TFF < 16, 16-30, 31-45, and > 45 postnatal days, respectively). After adjusting for confounding variables, survival without NDD was significantly decreased in P4 infants compared with that in P2 infants. P1 infants had a lower risk of weight and height PGF than P2 infants; however, P4 infants had higher risks of height and head circumference PGF than P2 infants. Weight and height PGF were significantly associated with an increased risk of NDD. In mediation analysis, early and delayed TFF revealed direct positive and negative impacts, respectively, on the risk of NDD without mediation by PGF. TFF impacted survival without NDD, and PGF did not mediate this association in VLBW infants. Additionally, these results can be translated into evidence-based quality improvement practice.
Collapse
Affiliation(s)
- Shin Ae Yoon
- grid.254229.a0000 0000 9611 0917Department of Pediatrics, Chungbuk National University Hospital, Chungbuk National University School of Medicine, 1 Sunhwan ro 776, Seowon-gu, Cheongju, 28644 Republic of Korea
| | - Myung Hee Lee
- Research and Statistical Center, Social Information Research Institute, Seoul, Republic of Korea ,MEDITOS, Institute of Biomedical and Clinical Research, Seoul, Republic of Korea
| | - Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea. .,Samsung Medical Center, Cell and Gene Therapy Institute, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Embleton ND, Berrington JE. Milk-Based Bionutrient Trials to Improve Outcomes in Preterm Infants: Challenges and Opportunities. Am J Perinatol 2022; 39:S68-S72. [PMID: 36470294 DOI: 10.1055/s-0042-1758857] [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: 12/12/2022]
Abstract
OBJECTIVE Bionutrients (or immunonutrients) are dietary components present in milk, or supplements that could be added to milk diets, that impact health and disease. With few exceptions, most of these are present in human breastmilk and the majority are also present in amniotic fluid. STUDY DESIGN Bionutrients can be proteins and peptides including enzymes, hormones, immunoglobulins, and growth factors and can also be molecules such as human milk oligosaccharides, amino acids, or lipids such as docosahexaenoic acid. Many of these have ancient origins, are found in other species, and existed before mammalian lactation evolved. Bionutrients may act in diverse ways when administered enterally: they may impact gut bacterial communities or epithelial cell metabolism, or they may pass into the lamina propria where they interact with the gut and systemic immune systems. Clinical trials have often used bovine analogs such as lactoferrin or may use artificially synthesized or recombinant compounds including insulin, bile salt stimulated lipase, or oligosaccharides. RESULTS Challenges arise because the bioactivity of proteins, such as lactoferrin, may be affected by processing and pasteurization meaning that the impacts of commercial products may differ. The challenge of determining the optimal bioactivity of any single preparation may be even greater in complex compounds such as milk fat globule membrane. It is also possible that bioactivity is affected by the milk matrix, that is, may differ between formula and human milk. CONCLUSION Finally, it is important to appreciate that nutrients do not function in isolation, and most will not act like drugs, that is, they may take several days or longer to exert an affect. KEY POINTS · Breastmilk contains high concentrations of bionutrients and provides more than macro- and micronutrients.. · Bionutrients can be proteins (e.g. enzymes, hormones, or immunoglobulins) or molecules (e.g. human milk oligosaccharides or amino acids).. · Bionutrients can be added to milk feeds but high quality trials are needed..
Collapse
Affiliation(s)
- Nicholas D Embleton
- Newcastle Hospitals National Health Service Trust, Newcastle upon Tyne, United Kingdom.,Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Janet E Berrington
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.,Translational Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
10
|
Young G, Berrington JE, Cummings S, Dorling J, Ewer AK, Frau A, Lett L, Probert C, Juszczak E, Kirby J, Beck LC, Renwick VL, Lamb C, Lanyon CV, McGuire W, Stewart C, Embleton N. Mechanisms affecting the gut of preterm infants in enteral feeding trials: a nested cohort within a randomised controlled trial of lactoferrin. Arch Dis Child Fetal Neonatal Ed 2022; 108:272-279. [PMID: 36396443 PMCID: PMC10176413 DOI: 10.1136/archdischild-2022-324477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To determine the impact of supplemental bovine lactoferrin on the gut microbiome and metabolome of preterm infants. DESIGN Cohort study nested within a randomised controlled trial (RCT). Infants across different trial arms were matched on several clinical variables. Bacteria and metabolite compositions of longitudinal stool and urine samples were analysed to investigate the impact of lactoferrin supplementation. SETTING Thirteen UK hospitals participating in a RCT of lactoferrin. PATIENTS 479 infants born <32 weeks' gestation between June 2016 and September 2017. RESULTS 10 990 stool and 22 341 urine samples were collected. Analyses of gut microbiome (1304 stools, 201 infants), metabolites (171 stools, 83 infants; 225 urines, 90 infants) and volatile organic compounds (314 stools, 117 infants) were performed. Gut microbiome Shannon diversity at 34 weeks corrected age was not significantly different between infants in the lactoferrin (mean=1.24) or placebo (mean=1.06) groups (p=0.11). Lactoferrin receipt explained less than 1% variance in microbiome compositions between groups. Metabolomic analysis identified six discriminative features between trial groups. Hospital site (16%) and postnatal age (6%) explained the greatest variation in microbiome composition. CONCLUSIONS This multiomic study identified minimal impacts of lactoferrin but much larger impacts of hospital site and postnatal age. This may be due to the specific lactoferrin product used, but more likely supports the findings of the RCT in which this study was nested, which showed no impact of lactoferrin on reducing rates of sepsis. Multisite mechanistic studies nested within RCTs are feasible and help inform trial interpretation and future trial design.
Collapse
Affiliation(s)
- Greg Young
- Applied Sciences, Northumbria University Faculty of Health and Life Sciences, Newcastle upon Tyne, England, UK.,Microbial Environments, Hub for Biotechnology in the Built Environment, Newcastle upon Tyne, England, UK
| | - Janet E Berrington
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK .,Newcastle Neonatal Service, Ward 35 Neonatal Unit, Royal Victoria Infirmary, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Stephen Cummings
- School of Health and Life Sciences, Teesside University, Middlesbrough, North Yorkshire, UK
| | - Jon Dorling
- Department of Neonatal Medicine, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Andrew K Ewer
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Alessandra Frau
- Gastroenterology Research Unit, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Lauren Lett
- Gastroenterology Research Unit, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Chris Probert
- Gastroenterology Research Unit, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Ed Juszczak
- School of Medicine, University of Nottingham School of Medicine, Nottingham, Notts, UK
| | - John Kirby
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Lauren C Beck
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Victoria L Renwick
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Christopher Lamb
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Clare V Lanyon
- Applied Sciences, Northumbria University Faculty of Health and Life Sciences, Newcastle upon Tyne, England, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, North Yorkshire, UK
| | - Christopher Stewart
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Nicholas Embleton
- Newcastle Neonatal Service, Ward 35 Neonatal Unit, Royal Victoria Infirmary, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK.,Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
11
|
Strobel NA, Adams C, McAullay DR, Edmond KM. Mother's Own Milk Compared With Formula Milk for Feeding Preterm or Low Birth Weight Infants: Systematic Review and Meta-analysis. Pediatrics 2022; 150:188643. [PMID: 35921674 DOI: 10.1542/peds.2022-057092d] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES We assessed the effect of feeding preterm or low birth weight infants with infant formula compared with mother's own milk on mortality, morbidity, growth, neurodevelopment, and disability. METHODS We searched Medline (Ovid), Embase (Ovid), and Cochrane Central Register of Controlled Studies to October 1, 2021. RESULTS Forty-two studies enrolling 89 638 infants fulfilled the inclusion criteria. We did not find evidence of an effect on mortality (odds ratio [OR] 1.26, 95% confidence interval [CI] 0.91-1.76), infection (OR 1.52, 95% CI 0.98-2.37), cognitive neurodevelopment (standardized mean difference -1.30, 95% CI -3.53 to 0.93), or on growth parameters. Formula milk feeding increased the risk of necrotizing enterocolitis (OR 2.99, 95% CI 1.75-5.11). The Grading of Recommendations Assessment, Development, and Evaluation certainty of evidence was low for mortality and necrotizing enterocolitis, and very low for neurodevelopment and growth outcomes. CONCLUSIONS In preterm and low birth weight infants, low to very low-certainty evidence indicates that feeding with infant formula compared with mother's own milk has little effect on all-cause mortality, infection, growth, or neurodevelopment, and a higher risk of developing necrotizing enterocolitis.
Collapse
|
12
|
Sharif S, Heath PT, Oddie SJ, McGuire W. Synbiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Database Syst Rev 2022; 3:CD014067. [PMID: 35230697 PMCID: PMC8887627 DOI: 10.1002/14651858.cd014067.pub2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Intestinal dysbiosis may contribute to the pathogenesis of necrotising enterocolitis (NEC) in very preterm or very low birth weight (VLBW) infants. Dietary supplementation with synbiotics (probiotic micro-organisms combined with prebiotic oligosaccharides) to modulate the intestinal microbiome has been proposed as a strategy to reduce the risk of NEC and associated mortality and morbidity. OBJECTIVES To assess the effect of enteral supplementation with synbiotics (versus placebo or no treatment, or versus probiotics or prebiotics alone) for preventing NEC and associated morbidity and mortality in very preterm or VLBW infants. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials, MEDLINE, Embase, the Maternity and Infant Care database and CINAHL, from earliest records to 17 June 2021. We searched clinical trials databases and conference proceedings, and examined the reference lists of retrieved articles. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs comparing prophylactic synbiotics supplementation with placebo or no synbiotics in very preterm (< 32 weeks' gestation) or very low birth weight (< 1500 g) infants. DATA COLLECTION AND ANALYSIS Two review authors separately performed the screening and selection process, evaluated risk of bias of the trials, extracted data, and synthesised effect estimates using risk ratio (RR), risk difference (RD), and mean difference, with associated 95% confidence intervals (CIs). We used the GRADE approach to assess the level of certainty for effects on NEC, all-cause mortality, late-onset invasive infection, and neurodevelopmental impairment. MAIN RESULTS We included six trials in which a total of 925 infants participated. Most trials were small (median sample size 200). Lack of clarity on methods used to conceal allocation and mask caregivers or investigators were potential sources of bias in four of the trials. The studied synbiotics preparations contained lactobacilli or bifidobacteria (or both) combined with fructo- or galacto-oligosaccharides (or both). Meta-analyses suggested that synbiotics may reduce the risk of NEC (RR 0.18, 95% CI 0.09 to 0.40; RD 70 fewer per 1000, 95% CI 100 fewer to 40 fewer; number needed to treat for an additional beneficial outcome (NNTB) 14, 95% CI 10 to 25; six trials (907 infants); low certainty evidence); and all-cause mortality prior to hospital discharge (RR 0.53, 95% CI 0.33 to 0.85; RD 50 fewer per 1000, 95% CI 120 fewer to 100 fewer; NNTB 20, 95% CI 8 to 100; six trials (925 infants); low-certainty evidence). Synbiotics may have little or no effect on late-onset invasive infection, but the evidence is very uncertain (RR 0.84, 95% CI 0.58 to 1.21; RD 20 fewer per 1000, 95% CI 70 fewer to 30 more; five trials (707 infants); very low-certainty evidence). None of the trials assessed neurodevelopmental outcomes. In the absence of high levels of heterogeneity, we did not undertake any subgroup analysis (including the type of feeding). AUTHORS' CONCLUSIONS The available trial data provide only low-certainty evidence about the effects of synbiotics on the risk of NEC and associated morbidity and mortality for very preterm or very low birth weight infants. Our confidence in the effect estimates is limited; the true effects may be substantially different from these estimates. Large, high-quality trials would be needed to provide evidence of sufficient validity and applicability to inform policy and practice.
Collapse
Affiliation(s)
- Sahar Sharif
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Paul T Heath
- Division of Child Health and Vaccine Institute, St. George's, University of London, London, UK
| | - Sam J Oddie
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
13
|
Young L, Oddie SJ, McGuire W. Delayed introduction of progressive enteral feeds to prevent necrotising enterocolitis in very low birth weight infants. Cochrane Database Syst Rev 2022; 1:CD001970. [PMID: 35049036 PMCID: PMC8771918 DOI: 10.1002/14651858.cd001970.pub6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Enteral feeding for very preterm or very low birth weight (VLBW) infants is often delayed for several days after birth due to concern that early introduction of feeding may not be tolerated and may increase the risk of necrotising enterocolitis. Concerns exist, however, that delaying enteral feeding may diminish the functional adaptation of the gastrointestinal tract and prolong the need for parenteral nutrition with its attendant infectious and metabolic risks. OBJECTIVES To determine the effects of delayed introduction of progressive enteral feeds on the risk of necrotising enterocolitis, mortality and other morbidities in very preterm or VLBW infants. SEARCH METHODS Search strategies were developed by an information specialist in consultation with the review authors. The following databases were searched in October 2021 without date or language restrictions: CENTRAL (2021, Issue 10), MEDLINE via OVID (1946 to October 2021), Embase via OVID (1974 to October 2021), Maternity and Infant Care via OVID (1971 to October 2021), CINAHL (1982 to October 2021). We also searched for eligible trials in clinical trials databases, conference proceedings, previous reviews, and reference lists of retrieved articles. SELECTION CRITERIA Randomised controlled trials that assessed the effects of delayed (four or more days after birth) versus earlier introduction of progressive enteral feeds on necrotising enterocolitis, mortality and other morbidities in very preterm or VLBW infants. DATA COLLECTION AND ANALYSIS Two review authors separately evaluated trial risk of bias, extracted data, and synthesised effect estimates using risk ratio (RR), risk difference (RD), and mean difference. We used the GRADE approach to assess the certainty of evidence for effects on necrotising enterocolitis, mortality, feed intolerance, and invasive infection. MAIN RESULTS We included 14 trials in which a total of 1551 infants participated. Potential sources of bias were lack of clarity on methods to generate random sequences and conceal allocation in half of the trials, and lack of masking of caregivers or investigators in all of the trials. Trials typically defined delayed introduction of progressive enteral feeds as later than four to seven days after birth and early introduction as four days or fewer after birth. Infants in six trials (accounting for about half of all of the participants) had intrauterine growth restriction or circulatory redistribution demonstrated by absent or reversed end-diastolic flow velocities in the fetal aorta or umbilical artery. Meta-analyses showed that delayed introduction of progressive enteral feeds may not reduce the risk of necrotising enterocolitis (RR 0.81, 95% confidence interval (CI) 0.58 to 1.14; RD -0.02, 95% CI -0.04 to 0.01; 13 trials, 1507 infants; low-certainty evidence due risk of bias and imprecision) nor all-cause mortality before hospital discharge (RR 0.97, 95% CI 0.70 to 1.36; RD -0.00, 95% CI -0.03 to 0.03; 12 trials, 1399 infants; low-certainty evidence due risk of bias and imprecision). Delayed introduction of progressive enteral feeds may slightly reduce the risk of feed intolerance (RR 0.81, 95% CI 0.68 to 0.97; RD -0.09, 95% CI -0.17 to -0.02; number needed to treat for an additional beneficial outcome = 11, 95% CI 6 to 50; 6 trials, 581 infants; low-certainty evidence due to risk of bias and imprecision) and probably increases the risk of invasive infection (RR 1.44, 95% CI 1.15 to 1.80; RD 0.10, 95% CI 0.04 to 0.15; number needed to treat for a harmful outcome = 10, 95% CI 7 to 25; 7 trials, 872 infants; moderate-certainty evidence due to risk of bias). AUTHORS' CONCLUSIONS: Delaying the introduction of progressive enteral feeds beyond four days after birth (compared with earlier introduction) may not reduce the risk of necrotising enterocolitis or death in very preterm or VLBW infants. Delayed introduction may slightly reduce feed intolerance, and probably increases the risk of invasive infection.
Collapse
Affiliation(s)
- Lauren Young
- Department of Neonatal Medicine, Trevor Mann Baby Unit, Royal Alexandra Children's Hospital, Brighton, UK
| | - Sam J Oddie
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
14
|
Renwick VL, Stewart CJ. Exploring functional metabolites in preterm infants. Acta Paediatr 2022; 111:45-53. [PMID: 34626496 DOI: 10.1111/apa.16146] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/08/2021] [Indexed: 12/26/2022]
Abstract
AIM Metabolomics is the study of small molecules that represent the functional end points of cellular reactions that can impact health. Necrotising enterocolitis (NEC) and late onset sepsis (LOS) are the main cause of death in preterm infants surviving the initial days of life. METHODS This review will explore and summarise the current literature exploring metabolomics in preterm infants. RESULTS There are a relatively limited number of studies investigating metabolomics in preterm infants with NEC and/or LOS and matched controls. Nonetheless, it is evident across longitudinally age-related metabolomic studies that there are significant changes in metabolite profiles post-partum and over the first year of life. Existing studies have reported associations between the metabolite profiles of serum, urine and stool in health and disease in preterm infants. Although some studies have found selected metabolites are associated with disease, the specific metabolites vary from study to study, and larger studies are required. Excitingly, recent work has also begun to untangle how microbially produced metabolites can impact immunoregulation of the infant. CONCLUSION Metabolic exploration is an emerging research area with huge potential for developing novel biomarkers and better understanding disease processes in preterm infants.
Collapse
Affiliation(s)
- Victoria L. Renwick
- Clinical and Translational Research Institute Newcastle University Newcastle upon Tyne UK
| | - Christopher J. Stewart
- Clinical and Translational Research Institute Newcastle University Newcastle upon Tyne UK
| |
Collapse
|
15
|
Healy DB, Ryan CA, Ross RP, Stanton C, Dempsey EM. Clinical implications of preterm infant gut microbiome development. Nat Microbiol 2022; 7:22-33. [PMID: 34949830 DOI: 10.1038/s41564-021-01025-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
Perturbations to the infant gut microbiome during the first weeks to months of life affect growth, development and health. In particular, assembly of an altered intestinal microbiota during infant development results in an increased risk of immune and metabolic diseases that can persist into childhood and potentially into adulthood. Most research into gut microbiome development has focused on full-term babies, but health-related outcomes are also important for preterm babies. The systemic physiological immaturity of very preterm gestation babies (born earlier than 32 weeks gestation) results in numerous other microbiome-organ interactions, the mechanisms of which have yet to be fully elucidated or in some cases even considered. In this Perspective, we compare assembly of the intestinal microbiome in preterm and term infants. We focus in particular on the clinical implications of preterm infant gut microbiome composition and discuss the prospects for microbiome diagnostics and interventions to improve the health of preterm babies.
Collapse
Affiliation(s)
- David B Healy
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.
| | - C Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Eugene M Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,INFANT Research Centre, Cork University Hospital, Cork, Ireland
| |
Collapse
|
16
|
Xu S, Zhao C, Jia L, Ma Z, Zhang X, Shi H. Relationship between preterm, low birth weight, and development defects of enamel in the primary dentition: A meta-analysis. Front Pediatr 2022; 10:975340. [PMID: 36440332 PMCID: PMC9684462 DOI: 10.3389/fped.2022.975340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/12/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND This study aimed to systematically analyze the relationship between preterm (PT), low birth weight (LBW), and developmental defects of enamel (DDE) in the primary dentition. METHODS Following the retrieval of the databases, case-control studies, cross-sectional studies, and cohort studies on the relationship between PT, LBW and DDE, which had been published in English or Chinese up to January 2022 were included. The data about odds ratio (OR) and 95% confidence interval (95% CI) were extracted and calculated using STATA 12.0 Software. Case-control studies were evaluated using the Newcastle-Ottawa Scale (NOS), while cross-sectional studies and cohort studies were evaluated using the JBI scale. The heterogeneity of each study was evaluated using the Q test. RESULTS A total of 15 studies were included, of which 8 studied the relationship between PT and DDE, and 13 explored the relationship between LBW and DDE including three about the relationship between very low birth weight (VLBW) and DDE. Seven studies explored the relationship between PT, LBW, and DDE. The results of this meta-analysis showed that both PT and LBW especially VLBW (OR = 7.19, 95% CI: 4.98-10.38) were risk factors for DDE in the primary dentition (OR = 2.33, 95% CI: 1.55-3.51) (OR = 1.67, 95% CI: 1.08-2.59). The subgroup results showed that PT and LBW were both associated with the occurrence of enamel hypoplasia (EHP) (OR = 6.89, 95% CI: 3.33-14.34; OR = 2.78, 95% CI: 2.10-3.68) rather than enamel opacity (OR = 0.94, 95% CI: 0.55-1.61; OR = 1.03, 95% CI: 0.66-1.61). There was no publication bias about the included studies (P = 0.75 > 0.05; P = 0.47 > 0.05). CONCLUSION This meta-analysis demonstrated that both PT and LBW especially VLBW are associated with a higher risk of DDE in the primary dentition. PT and LBW are both related to the occurrence of EHP. However, the relationship between PT, LBW, and enamel opacity has not been verified. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/display_record.php?, identifier: CRD42021262761.
Collapse
Affiliation(s)
- Shan Xu
- Department of Pediatric Dentistry, Hospital of Stomatology & Hebei Provincial Key Laboratory of Stomatology, Hebei Medical University, Shijiazhuang, China
| | - Caiyun Zhao
- Department of Pediatric Dentistry, Hospital of Stomatology & Hebei Provincial Key Laboratory of Stomatology, Hebei Medical University, Shijiazhuang, China
| | - Liying Jia
- Department of Preventive Dentistry, Hospital of Stomatology & Hebei Provincial Key Laboratory of Stomatology, Hebei Medical University, Shijiazhuang, China
| | - Zhe Ma
- Department of Preventive Dentistry, Hospital of Stomatology & Hebei Provincial Key Laboratory of Stomatology, Hebei Medical University, Shijiazhuang, China
| | - Xiaolin Zhang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Province Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, China
| | - Hong Shi
- Department of Pediatric Dentistry, Hospital of Stomatology & Hebei Provincial Key Laboratory of Stomatology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
17
|
Oddie SJ, Young L, McGuire W. Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants. Cochrane Database Syst Rev 2021; 8:CD001241. [PMID: 34427330 PMCID: PMC8407506 DOI: 10.1002/14651858.cd001241.pub8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Early enteral feeding practices are potentially modifiable risk factors for necrotising enterocolitis (NEC) in very preterm or very low birth weight (VLBW) infants. Observational studies suggest that conservative feeding regimens, including slowly advancing enteral feed volumes, reduce the risk of NEC. However, it is unclear whether slow feed advancement may delay establishment of full enteral feeding, and if it could be associated with infectious morbidities secondary to prolonged exposure to parenteral nutrition. OBJECTIVES To determine the effects of slow rates of enteral feed advancement on the risk of NEC, mortality, and other morbidities in very preterm or VLBW infants. SEARCH METHODS We searched CENTRAL (2020, Issue 10), Ovid MEDLINE (1946 to October 2020), Embase via Ovid (1974 to October 2020), Maternity and Infant Care database (MIDIRS) (1971 to October 2020), CINAHL (1982 to October 2020), and clinical trials databases and reference lists of retrieved articles for eligible trials. SELECTION CRITERIA We included randomised or quasi-randomised controlled trials that assessed effects of slow (up to 24 mL/kg/d) versus faster rates of advancement of enteral feed volumes on the risk of NEC in very preterm or VLBW infants. DATA COLLECTION AND ANALYSIS Two review authors separately evaluated trial risk of bias, extracted data, and synthesised effect estimates using risk ratio (RR), risk difference (RD), and mean difference. We used the GRADE approach to assess the certainty of evidence. Outcomes of interest were NEC, all-cause mortality, feed intolerance, and invasive infection. MAIN RESULTS We included 14 trials involving a total of 4033 infants (2804 infants participated in one large trial). None of the trials masked parents, caregivers, or investigators. Risk of bias was otherwise low. Most infants were stable very preterm or VLBW infants of birth weight appropriate for gestation. About one-third of all infants were extremely preterm or extremely low birth weight (ELBW), and about one-fifth were small for gestational age, growth-restricted, or compromised as indicated by absent or reversed end-diastolic flow velocity in the foetal umbilical artery. Trials typically defined slow advancement as daily increments of 15 to 24 mL/kg, and faster advancement as daily increments of 30 to 40 mL/kg. Meta-analyses showed that slow advancement of enteral feed volumes probably has little or no effect on the risk of NEC (RR 1.06, 95% confidence interval (CI) 0.83 to 1.37; RD 0.00, 95% CI -0.01 to 0.02; 14 trials, 4026 infants; moderate-certainty evidence) or all-cause mortality prior to hospital discharge (RR 1.13, 95% CI 0.91 to 1.39; RD 0.01, 95% CI -0.01 to 0.02; 13 trials, 3860 infants; moderate-certainty evidence). Meta-analyses suggested that slow advancement may slightly increase feed intolerance (RR 1.18, 95% CI 0.95 to 1.46; RD 0.05, 95% CI -0.02 to 0.12; 9 trials, 719 infants; low-certainty evidence) and may slightly increase the risk of invasive infection (RR 1.14, 95% CI 0.99 to 1.31; RD 0.02, 95% CI -0.00 to 0.05; 11 trials, 3583 infants; low-certainty evidence). AUTHORS' CONCLUSIONS The available trial data indicate that advancing enteral feed volumes slowly (daily increments up to 24 mL/kg) compared with faster rates probably does not reduce the risk of NEC, death, or feed intolerance in very preterm or VLBW infants. Advancing the volume of enteral feeds at a slow rate may slightly increase the risk of invasive infection.
Collapse
Affiliation(s)
- Sam J Oddie
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Lauren Young
- Department of Neonatal Medicine, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
18
|
Sharif S, Oddie SJ, Heath PT, McGuire W. Prebiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Hippokratia 2021. [DOI: 10.1002/14651858.cd015133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Sahar Sharif
- Centre for Reviews and Dissemination; University of York; York UK
| | - Sam J Oddie
- Bradford Neonatology; Bradford Teaching Hospitals NHS Foundation Trust; Bradford UK
| | - Paul T Heath
- Division of Child Health and Vaccine Institute; St. George's, University of London; London UK
| | - William McGuire
- Centre for Reviews and Dissemination; University of York; York UK
| |
Collapse
|
19
|
李 颖, 吴 繁. [Enteral feeding in extremely low birth weight infants with a gestational age of <28 weeks versus ≥ 28 weeks]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:690-695. [PMID: 34266525 PMCID: PMC8292661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/03/2021] [Indexed: 09/21/2023]
Abstract
OBJECTIVE To investigate the condition of enteral feeding and the incidence of feeding intolerance (FI) in extremely low birth weight (ELBW) infants with a gestational age (GA) of < 28 weeks versus ≥ 28 weeks. METHODS A retrospective analysis was performed for the ELBW infants who were admitted to the Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, from January 2018 to January 2020. According to GA, they were divided into two groups:GA < 28 weeks (n=41) and GA ≥ 28 weeks (n=38). The two groups were compared in terms of enteral feeding and incidence rate of FI during hospitalization. RESULTS Among the 41 infants in the GA < 28 weeks group, 2 (5%) were small-for-gestational-age (SGA) infants, while among the 38 infants in the GA ≥ 28 weeks group, 24 (63%) were SGA infants. Compared with the GA ≥ 28 weeks group, the GA < 28 weeks group had significantly longer initial and final time to full enteral feeding (P < 0.05). The GA < 28 weeks group had a significantly higher recurrence rate of FI than the GA ≥ 28 weeks group (54% vs 32%, P < 0.05). Compared with the GA < 28 weeks group, the GA ≥ 28 weeks group had a significantly lower incidence rate of FI within 1-2 weeks after birth (5% vs 29%, P < 0.05) and a significantly shorter duration of FI (P < 0.05). The ELBW infants in the GA ≥ 28 weeks group had a higher velocity of weight gain, a shorter length of hospital stay, and a lower incidence rate of bronchopulmonary dysplasia, but a higher incidence rate of extrauterine growth retardation (EUGR) than the GA < 28 weeks group at discharge (P < 0.05). Conclusions: Compared with the ELBW infants with a GA of < 28 weeks, the ELBW infants with a GA of ≥ 28 weeks have a lower recurrence rate of FI and can achieve full enteral feeding earlier, but with a higher incidence rate of EUGR, suggesting that enteral feeding needs to be more active for the ELBW infants with a GA of ≥ 28 weeks.
Collapse
Affiliation(s)
- 颖 李
- />广州医科大学附属第三医院新生儿科, 广东广州 510150Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - 繁 吴
- />广州医科大学附属第三医院新生儿科, 广东广州 510150Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| |
Collapse
|
20
|
李 颖, 吴 繁. [Enteral feeding in extremely low birth weight infants with a gestational age of <28 weeks versus ≥ 28 weeks]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23:690-695. [PMID: 34266525 PMCID: PMC8292661 DOI: 10.7499/j.issn.1008-8830.2101006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/03/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To investigate the condition of enteral feeding and the incidence of feeding intolerance (FI) in extremely low birth weight (ELBW) infants with a gestational age (GA) of < 28 weeks versus ≥ 28 weeks. METHODS A retrospective analysis was performed for the ELBW infants who were admitted to the Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, from January 2018 to January 2020. According to GA, they were divided into two groups:GA < 28 weeks (n=41) and GA ≥ 28 weeks (n=38). The two groups were compared in terms of enteral feeding and incidence rate of FI during hospitalization. RESULTS Among the 41 infants in the GA < 28 weeks group, 2 (5%) were small-for-gestational-age (SGA) infants, while among the 38 infants in the GA ≥ 28 weeks group, 24 (63%) were SGA infants. Compared with the GA ≥ 28 weeks group, the GA < 28 weeks group had significantly longer initial and final time to full enteral feeding (P < 0.05). The GA < 28 weeks group had a significantly higher recurrence rate of FI than the GA ≥ 28 weeks group (54% vs 32%, P < 0.05). Compared with the GA < 28 weeks group, the GA ≥ 28 weeks group had a significantly lower incidence rate of FI within 1-2 weeks after birth (5% vs 29%, P < 0.05) and a significantly shorter duration of FI (P < 0.05). The ELBW infants in the GA ≥ 28 weeks group had a higher velocity of weight gain, a shorter length of hospital stay, and a lower incidence rate of bronchopulmonary dysplasia, but a higher incidence rate of extrauterine growth retardation (EUGR) than the GA < 28 weeks group at discharge (P < 0.05). Conclusions: Compared with the ELBW infants with a GA of < 28 weeks, the ELBW infants with a GA of ≥ 28 weeks have a lower recurrence rate of FI and can achieve full enteral feeding earlier, but with a higher incidence rate of EUGR, suggesting that enteral feeding needs to be more active for the ELBW infants with a GA of ≥ 28 weeks.
Collapse
Affiliation(s)
- 颖 李
- />广州医科大学附属第三医院新生儿科, 广东广州 510150Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - 繁 吴
- />广州医科大学附属第三医院新生儿科, 广东广州 510150Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| |
Collapse
|
21
|
Frau A, Lett L, Slater R, Young GR, Stewart CJ, Berrington J, Hughes DM, Embleton N, Probert C. The Stool Volatile Metabolome of Pre-Term Babies. Molecules 2021; 26:3341. [PMID: 34199338 PMCID: PMC8199543 DOI: 10.3390/molecules26113341] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/13/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022] Open
Abstract
The fecal metabolome in early life has seldom been studied. We investigated its evolution in pre-term babies during their first weeks of life. Multiple (n = 152) stool samples were studied from 51 babies, all <32 weeks gestation. Volatile organic compounds (VOCs) were analyzed by headspace solid phase microextraction gas chromatography mass spectrometry. Data were interpreted using Automated Mass Spectral Deconvolution System (AMDIS) with the National Institute of Standards and Technology (NIST) reference library. Statistical analysis was based on linear mixed modelling, the number of VOCs increased over time; a rise was mainly observed between day 5 and day 10. The shift at day 5 was associated with products of branched-chain fatty acids. Prior to this, the metabolome was dominated by aldehydes and acetic acid. Caesarean delivery showed a modest association with molecules of fungal origin. This study shows how the metabolome changes in early life in pre-term babies. The shift in the metabolome 5 days after delivery coincides with the establishment of enteral feeding and the transition from meconium to feces. Great diversity of metabolites was associated with being fed greater volumes of milk.
Collapse
Affiliation(s)
- Alessandra Frau
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3GE, UK; (L.L.); (R.S.); (C.P.)
| | - Lauren Lett
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3GE, UK; (L.L.); (R.S.); (C.P.)
| | - Rachael Slater
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3GE, UK; (L.L.); (R.S.); (C.P.)
| | - Gregory R. Young
- Hub for Biotechnology in the Built Environment, Northumbria University, Newcastle upon Tyne NE1 8ST, UK;
| | - Christopher J. Stewart
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 8ST, UK; (C.J.S.); (J.B.)
| | - Janet Berrington
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 8ST, UK; (C.J.S.); (J.B.)
- Department of Neonatology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 8ST, UK;
| | - David M. Hughes
- Department of Health Data Science, University of Liverpool, Liverpool, Merseyside L69 3GA, UK;
| | - Nicholas Embleton
- Department of Neonatology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 8ST, UK;
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne NE1 8ST, UK
| | - Chris Probert
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 3GE, UK; (L.L.); (R.S.); (C.P.)
| |
Collapse
|
22
|
Abstract
This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To evaluate the effect of enteral supplementation with synbiotics (versus placebo or no treatment, or versus probiotics or prebiotics alone) on the risk of necrotising enterocolitis and associated morbidity and mortality in very preterm or very low birth weight infants.
Collapse
|
23
|
Beck LC, Granger CL, Masi AC, Stewart CJ. Use of omic technologies in early life gastrointestinal health and disease: from bench to bedside. Expert Rev Proteomics 2021; 18:247-259. [PMID: 33896313 DOI: 10.1080/14789450.2021.1922278] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: At birth, the gastrointestinal (GI) tract is colonized by a complex community of microorganisms, forming the basis of the gut microbiome. The gut microbiome plays a fundamental role in host health, disorders of which can lead to an array of GI diseases, both short and long term. Pediatric GI diseases are responsible for significant morbidity and mortality, but many remain poorly understood. Recent advancements in high-throughput technologies have enabled deeper profiling of GI morbidities. Technologies, such as metagenomics, transcriptomics, proteomics and metabolomics, have already been used to identify associations with specific pathologies, and highlight an exciting area of research. However, since these diseases are often complex and multifactorial by nature, reliance on a single experimental approach may not capture the true biological complexity. Therefore, multi-omics aims to integrate singular omic data to further enhance our understanding of disease.Areas covered: This review will discuss and provide an overview of the main omic technologies that are used to study complex GI pathologies in early life.Expert opinion: Multi-omic technologies can help to unravel the complexities of several diseases during early life, aiding in biomarker discovery and enabling the development of novel therapeutics and augment predictive models.
Collapse
Affiliation(s)
- Lauren C Beck
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Claire L Granger
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK.,Newcastle Neonatal Service, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| | - Andrea C Masi
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Christopher J Stewart
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
24
|
Bushati C, Chan B, Harmeson Owen A, Woodbury A, Yang M, Fung C, Lechtenberg E, Rigby M, Baserga M. Challenges in Implementing Exclusive Human Milk Diet to Extremely Low-Birth-Weight Infants in a Level III Neonatal Intensive Care Unit. Nutr Clin Pract 2021; 36:1198-1206. [PMID: 33624353 DOI: 10.1002/ncp.10625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PROBLEM Extremely low-birth-weight (ELBW) infants require fortification of human milk (HM) to prevent growth failure. Bovine milk-based fortifiers (BOV-f) may be associated with feeding intolerance and necrotizing enterocolitis. Evidence suggests that an exclusive HM diet (EHMD) using HM-based fortifier (HM-f) may improve these outcomes. INTERVENTION EHMD was introduced as a quality improvement project to improve feeding tolerance in ELBW infants. METHOD Implementation included establishing EHMD feeding protocol and growth monitoring. We compared infants receiving HM with BOV-f (n = 49) with infants receiving an EHMD (n = 15). The primary outcome was a reduction of no oral intake days due to feeding intolerance. RESULTS The EHMD cohort had a more advanced GA (28 vs 26 weeks; P = .03), more males (66.7% vs 42.9%; P = .02), and higher incidence of SGA (40.0% vs 18.4%; P = .16) compared with the HM-f group. The EHMD cohort had fewer days with no oral intake (2 vs 5; P < .005), which is insignificant when adjusted for small for gestational age (SGA) (P = .26). The EHMD cohort vs. the HM-f cohort had a significant decrease in weight and length z-scores from birth to discharge (-1.09 vs -0.26 [P = .002]; -1.76 vs -0.83 [P = .02]). Inadequate weight gain persisted after adjustment for SGA. Interventions were performed to increase caloric intake and institute milk preparation changes. CONCLUSION EHMD improved feeding tolerance in our ELBW infants. Observed growth failure might be skewed by SGA prevalence. We highlight that implementation of EHMD requires close growth assessment, especially for SGA infants.
Collapse
Affiliation(s)
- Charity Bushati
- Women's and Children's Services, University of Utah Hospital, Salt Lake City, Utah, USA
| | - Belinda Chan
- Neonatology Division, University of Utah, Salt Lake City, Utah, USA
| | | | - Anne Woodbury
- Nutrition Care Services, University of Utah, Salt Lake City, Utah, USA
| | - Michelle Yang
- Neonatology Division, University of Utah, Salt Lake City, Utah, USA
| | - Camille Fung
- Neonatology Division, University of Utah, Salt Lake City, Utah, USA
| | - Ellen Lechtenberg
- Lactation Service, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Marilyn Rigby
- Neonatology APC Group, Intermountain Health Care, Salt Lake City, Utah, USA
| | - Mariana Baserga
- Neonatology Division, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
25
|
Wright H, Bannaga AS, Iriarte R, Mahmoud M, Arasaradnam RP. Utility of volatile organic compounds as a diagnostic tool in preterm infants. Pediatr Res 2021; 89:263-268. [PMID: 32120380 DOI: 10.1038/s41390-020-0828-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/29/2020] [Accepted: 02/12/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Volatile organic compounds (VOCs) are hydrocarbons that originate within different healthy and diseased tissues. VOCs can be secreted into the circulation and then excreted in the urine and faeces. In the lungs, VOCs are locally produced and can be detected in exhaled breath. VOCs can be identified using non-invasive techniques, which make their use in preterm infants safe and desirable. METHODS A systematic search of the literature in PubMed, Embase and Web of Science was conducted looking for VOCs techniques and diagnostic performance in preterm infants. A total of 50 articles identified with only seven papers were included in the final analysis in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). RESULTS VOCs could diagnose necrotising enterocolitis up to 4 days before a clinical diagnosis; for late onset sepsis, up to 3 days before; and for bronchopulmonary dysplasia, up to 2 weeks before. In addition to these diagnostic uses, VOCs analysis could also distinguish breastfed from formula-fed preterm neonates in the first month of life. CONCLUSION VOCs analysis is a non-invasive tool that makes the use in preterm infants of preference. VOCs analytic techniques require more research and consensus between researchers to overcome their limitations. IMPACT Volatile organic compounds are hydrocarbons that can separate between healthy and diseased states in preterm infants. Biomarker panels developed from volatile organic compounds are potential diagnostic tools. The non-invasive nature of acquiring volatile organic compounds markers make it desirable in the paediatric patients. Research into exact chemical components of the volatile organic compounds can inform about the pathophysiology of disease in preterm infants. More robust longitudinal studies with repeated experiments are required before volatile organic compounds can be applied in clinical practice.
Collapse
Affiliation(s)
- Harriet Wright
- Warwick Medical School, Warwick University, Coventry, UK
| | - Ayman S Bannaga
- Warwick Medical School, Warwick University, Coventry, UK.,University Hospital Coventry & Warwickshire, Coventry, UK
| | | | | | - Ramesh P Arasaradnam
- Warwick Medical School, Warwick University, Coventry, UK. .,University Hospital Coventry & Warwickshire, Coventry, UK. .,Applied Health Sciences, University of Coventry, Coventry, UK. .,School of Health Sciences, University of Leicester, Leicester, UK.
| |
Collapse
|
26
|
Richards J, Rankin J, Juszczak E, Dorling J, McGuire W, Embleton ND. Parental experiences of being approached to join multiple neonatal clinical trials: qualitative study (PARENT). Arch Dis Child Fetal Neonatal Ed 2021; 106:84-87. [PMID: 32737064 DOI: 10.1136/archdischild-2020-319031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To explore parents' perceptions and experience of being approached for enrolment of their preterm infant in more than one trial or study. DESIGN A qualitative study involving 17 in-depth semistructured interviews, with parents who had been approached for multiple studies and who subsequently consented for their infant(s) to join at least one. Parents who declined all studies were not approached. SETTING AND PARTICIPANTS Parents of preterm infants receiving care at one of three neonatal intensive care units in the north of England. FINDINGS Most parents did not view concurrent participation in multiple trials or studies as a significant issue within the wider context of their infant's care. Most parents did not feel pressured into enrolling their infant into more than one study, but some suggested that participation in several provided justification for the subsequent refusal to join others, articulating feeling of guilt at saying 'no', and others appeared fatigued by multiple approaches. Parents focused on the perceived risks and benefits of each individual study and, while acknowledging that making a fully informed decision was not possible, largely agreed due to their belief in the benefits of research, trust in the health professionals caring for their baby and a range of complex personal motivations. CONCLUSIONS Parents valued the autonomy to make decisions about participation and felt, with hindsight, that their decisions were right. Research teams could be more aware of parental feelings of guilt or gratitude that may motivate them to give consent. Similarly, the capacity of parents to fully remember details of multiple studies when they are stressed, and their infant is sick, should be taken into consideration, and continued efforts should be made to ensure ongoing consent to participation.
Collapse
Affiliation(s)
- Judy Richards
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Judith Rankin
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ed Juszczak
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jon Dorling
- Division of Neonatal-Perinatal Medicine, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Nicholas D Embleton
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK .,Newcastle Neonatal Service, Newcastle Hospitals NHS Trust, Newcastle upon Tyne, UK
| |
Collapse
|
27
|
Walsh V, Brown JVE, Copperthwaite BR, Oddie SJ, McGuire W. Early full enteral feeding for preterm or low birth weight infants. Cochrane Database Syst Rev 2020; 12:CD013542. [PMID: 33368149 PMCID: PMC8094920 DOI: 10.1002/14651858.cd013542.pub2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The introduction and advancement of enteral feeds for preterm or low birth weight infants is often delayed because of concerns that early full enteral feeding will not be well tolerated or may increase the risk of necrotising enterocolitis. Early full enteral feeding, however, might increase nutrient intake and growth rates; accelerate intestinal physiological, metabolic, and microbiomic postnatal transition; and reduce the risk of complications associated with intravascular devices for fluid administration. OBJECTIVES: To determine how early full enteral feeding, compared with delayed or progressive introduction of enteral feeds, affects growth and adverse events such as necrotising enterocolitis, in preterm or low birth weight infants. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search Cochrane Central Register of Controlled Trials; MEDLINE Ovid, Embase Ovid, Maternity & Infant Care Database Ovid, the Cumulative Index to Nursing and Allied Health Literature, and clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials to October 2020. SELECTION CRITERIA Randomised controlled trials that compared early full enteral feeding with delayed or progressive introduction of enteral feeds in preterm or low birth weight infants. DATA COLLECTION AND ANALYSIS We used the standard methods of Cochrane Neonatal. Two review authors separately assessed trial eligibility, evaluated trial quality, extracted data, and synthesised effect estimates using risk ratios (RR), risk differences, and mean differences (MD) with 95% confidence intervals (CI). We used the GRADE approach to assess the certainty of evidence. MAIN RESULTS We included six trials. All were undertaken in the 2010s in neonatal care facilities in India. In total, 526 infants participated. Most were very preterm infants of birth weight between 1000 g and 1500 g. Trials were of good methodological quality, but a potential source of bias was that parents, clinicians, and investigators were not masked. The trials compared early full feeding (60 mL/kg to 80 mL/kg on day one after birth) with minimal enteral feeding (typically 20 mL/kg on day one) supplemented with intravenous fluids. Feed volumes were advanced daily as tolerated by 20 mL/kg to 30 mL/kg body weight to a target steady-state volume of 150 mL/kg to 180 mL/kg/day. All participating infants were fed preferentially with maternal expressed breast milk, with two trials supplementing insufficient volumes with donor breast milk and four supplementing with preterm formula. Few data were available to assess growth parameters. One trial (64 participants) reported a slower rate of weight gain (median difference -3.0 g/kg/day), and another (180 participants) reported a faster rate of weight gain in the early full enteral feeding group (MD 1.2 g/kg/day). We did not meta-analyse these data (very low-certainty evidence). None of the trials reported rate of head circumference growth. One trial reported that the mean z-score for weight at hospital discharge was higher in the early full enteral feeding group (MD 0.24, 95% CI 0.06 to 0.42; low-certainty evidence). Meta-analyses showed no evidence of an effect on necrotising enterocolitis (RR 0.98, 95% CI 0.38 to 2.54; 6 trials, 522 participants; I² = 51%; very low-certainty evidence). AUTHORS' CONCLUSIONS Trials provided insufficient data to determine with any certainty how early full enteral feeding, compared with delayed or progressive introduction of enteral feeds, affects growth in preterm or low birth weight infants. We are uncertain whether early full enteral feeding affects the risk of necrotising enterocolitis because of the risk of bias in the trials (due to lack of masking), inconsistency, and imprecision.
Collapse
Affiliation(s)
- Verena Walsh
- Centre for Reviews and Dissemination, University of York, York, UK
| | | | | | - Sam J Oddie
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
28
|
Sharif S, Meader N, Oddie SJ, Rojas-Reyes MX, McGuire W. Probiotics to prevent necrotising enterocolitis in very preterm or very low birth weight infants. Cochrane Database Syst Rev 2020; 10:CD005496. [PMID: 33058137 PMCID: PMC8094746 DOI: 10.1002/14651858.cd005496.pub5] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Intestinal dysbiosis may contribute to the pathogenesis of necrotising enterocolitis (NEC) in very preterm or very low birth weight infants. Dietary supplementation with probiotics to modulate the intestinal microbiome has been proposed as a strategy to reduce the risk of NEC and associated mortality and morbidity. OBJECTIVES: To determine the effect of supplemental probiotics on the risk of NEC and mortality and morbidity in very preterm or very low birth weight infants. SEARCH METHODS We searched Cochrane Central Register of Controlled Trials (CENTRAL; 2020, Issue 2) in the Cochrane Library; MEDLINE Ovid (1946 to 17 Feb 2020), Embase Ovid (1974 to 17 Feb 2020), Maternity & Infant Care Database Ovid (1971 to 17 Feb 2020), the Cumulative Index to Nursing and Allied Health Literature (1982 to 18 Feb 2020). We searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomised controlled trials (RCTs) and quasi-RCTs. SELECTION CRITERIA We included RCTs and quasi-RCTs comparing probiotic supplementation with placebo or no probiotics in very preterm or very low birth weight infants. DATA COLLECTION AND ANALYSIS We used the standard methods of Cochrane Neonatal. Two review authors separately evaluated trial quality, extracted data, and synthesised effect estimates using risk ratio (RR), risk difference (RD), and mean difference. We used the GRADE approach to assess the certainty of evidence for effects on NEC, all-cause mortality, late-onset infection, and severe neurodevelopmental impairment. MAIN RESULTS We included 56 trials in which 10,812 infants participated. Most trials were small (median sample size 149). Lack of clarity on methods to conceal allocation and mask caregivers or investigators were the main potential sources of bias in about half of the trials. Trials varied by the formulation of the probiotics. The most commonly used preparations contained Bifidobacterium spp., Lactobacillus spp., Saccharomyces spp., and Streptococcus spp. alone or in combinations. Meta-analysis showed that probiotics may reduce the risk of NEC: RR 0.54, 95% CI 0.45 to 0.65 (54 trials, 10,604 infants; I² = 17%); RD -0.03, 95% CI -0.04 to -0.02; number needed to treat for an additional beneficial outcome (NNTB) 33, 95% CI 25 to 50. Evidence was assessed as low certainty because of the limitations in trials design, and the presence of funnel plot asymmetry consistent with publication bias. Sensitivity meta-analysis of trials at low risk of bias showed a reduced risk of NEC: RR 0.70, 95% CI 0.55 to 0.89 (16 trials, 4597 infants; I² = 25%); RD -0.02, 95% CI -0.03 to -0.01; NNTB 50, 95% CI 33 to 100. Meta-analyses showed that probiotics probably reduce mortality (RR 0.76, 95% CI 0.65 to 0.89; (51 trials, 10,170 infants; I² = 0%); RD -0.02, 95% CI -0.02 to -0.01; NNTB 50, 95% CI 50 to 100), and late-onset invasive infection (RR 0.89, 95% CI 0.82 to 0.97; (47 trials, 9762 infants; I² = 19%); RD -0.02, 95% CI -0.03 to -0.01; NNTB 50, 95% CI 33 to 100). Evidence was assessed as moderate certainty for both these outcomes because of the limitations in trials design. Sensitivity meta-analyses of 16 trials (4597 infants) at low risk of bias did not show an effect on mortality or infection. Meta-analysis showed that probiotics may have little or no effect on severe neurodevelopmental impairment (RR 1.03, 95% CI 0.84 to 1.26 (five trials, 1518 infants; I² = 0%). The certainty on this evidence is low because of limitations in trials design and serious imprecision of effect estimate. Few data (from seven of the trials) were available for extremely preterm or extremely low birth weight infants. Meta-analyses did not show effects on NEC, death, or infection (low-certainty evidence). AUTHORS' CONCLUSIONS Given the low to moderate level of certainty about the effects of probiotic supplements on the risk of NEC and associated morbidity and mortality for very preterm or very low birth weight infants, and particularly for extremely preterm or extremely low birth weight infants, further, large, high-quality trials are needed to provide evidence of sufficient quality and applicability to inform policy and practice.
Collapse
Affiliation(s)
- Sahar Sharif
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Nicholas Meader
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sam J Oddie
- Centre for Reviews and Dissemination, University of York, York, UK
- Bradford Neonatology, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Maria Ximena Rojas-Reyes
- Department of Clinical Epidemiology and Public Health, Institut de Recerca Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| |
Collapse
|
29
|
Berrington JE, McGuire W, Embleton ND. ELFIN, the United Kingdom preterm lactoferrin trial: interpretation and future questions 1. Biochem Cell Biol 2020; 99:1-6. [PMID: 32830532 DOI: 10.1139/bcb-2020-0073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Results from previous studies have suggested that supplemental bovine lactoferrin (BLF) given to preterm infants (<32 weeks gestation) reduces late-onset sepsis (LOS) and necrotising enterocolitis (NEC). The Enteral Lactoferrin in Neonates (ELFIN) study, performed in the UK, aimed to further address this issue with a well powered double-blind placebo controlled trial of >2200 preterm infants. The results from ELFIN did not demonstrate a reduction in LOS or NEC, or several other clinically important measures. Of the 1093 infants, 316 (29%) in the intervention group developed late-onset sepsis versus 334 (31%) of 1089 in the control group, with an adjusted risk ratio of 0.95 (95% CI = 0.86-1.04; p = 0.233). Reasons for the differences in ELFIN trial results and other studies may include population differences, the routine use of antifungal prophylaxis in the UK, timing of administration of the lactoferrin in relation to disease onset, or specific properties of the lactoferrin used in the different trials. The UK National Institutes for Health Research funded "Mechanisms Affecting the Guts of Preterm Infants in Enteral feeding trials" (MAGPIE) study is further exploring the use of lactoferrin, and the results should be available soon.
Collapse
Affiliation(s)
- Janet Elizabeth Berrington
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, and Translational and Clinical Medicine, Newcastle University, Newcastle, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York YO10 5DD, UK
| | - Nicholas David Embleton
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, and Population Health Sciences, Newcastle University, Newcastle, UK
| |
Collapse
|
30
|
Bovine Lactoferrin Supplementation Does Not Disrupt Microbiota Development in Preterm Infants Receiving Probiotics. J Pediatr Gastroenterol Nutr 2020; 71:216-222. [PMID: 32404742 DOI: 10.1097/mpg.0000000000002734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVE The aim of the study was to assess whether bovine lactoferrin (bLf) supplementation disrupts intestinal microbiota development in preterm infants less than 31 weeks gestational age receiving prophylactic probiotic administration. METHODS Subjects were recruited from the LACUNA trial (ISRCTN66482337), designed to assess bLf safety. These subjects were randomized to daily receive either probiotic supplements or probiotics supplemented with 100 mg bLf mixed with their feeds (human milk or formula). Stools were collected weekly from enrolled infants for 1 month and the microbiota characterized using V6-16S rRNA gene amplicon profiling. RESULTS Infants' microbiomes did not increase in alpha diversity over time in both feeding interventions. Infants receiving bLf supplementation had overall higher species richness as compared with those not receiving these supplements and lactoferrin supplementation had differing effects on infant microbiota species richness depending on the infant's gestational age. Principal co-ordinate analysis revealed that the infant microbiotas did not separate by intervention group, gestational age bracket at birth or sampling time and the main factor dictating sample clustering was infant identity. There were very few detectable differences in taxa relative abundance or functional gene content between the microbiotas in the 2 study groups. CONCLUSIONS Bovine lactoferrin supplementation has minimal impact on microbiota composition/function in preterm infants receiving probiotics, and therefore, is unlikely to disrupt microbiota development.
Collapse
|
31
|
Dorling J, Hewer O, Hurd M, Bari V, Bosiak B, Bowler U, King A, Linsell L, Murray D, Omar O, Partlett C, Rounding C, Townend J, Abbott J, Berrington J, Boyle E, Embleton N, Johnson S, Leaf A, McCormick K, McGuire W, Patel M, Roberts T, Stenson B, Tahir W, Monahan M, Richards J, Rankin J, Juszczak E. Two speeds of increasing milk feeds for very preterm or very low-birthweight infants: the SIFT RCT. Health Technol Assess 2020; 24:1-94. [PMID: 32342857 DOI: 10.3310/hta24180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Observational data suggest that slowly advancing enteral feeds in preterm infants may reduce necrotising enterocolitis but increase late-onset sepsis. The Speed of Increasing milk Feeds Trial (SIFT) compared two rates of feed advancement. OBJECTIVE To determine if faster (30 ml/kg/day) or slower (18 ml/kg/day) daily feed increments improve survival without moderate or severe disability and other morbidities in very preterm or very low-birthweight infants. DESIGN This was a multicentre, two-arm, parallel-group, randomised controlled trial. Randomisation was via a web-hosted minimisation algorithm. It was not possible to safely and completely blind caregivers and parents. SETTING The setting was 55 UK neonatal units, from May 2013 to June 2015. PARTICIPANTS The participants were infants born at < 32 weeks' gestation or a weight of < 1500 g, who were receiving < 30 ml/kg/day of milk at trial enrolment. INTERVENTIONS When clinicians were ready to start advancing feed volumes, the infant was randomised to receive daily feed increments of either 30 ml/kg/day or 18 ml/kg/day. In total, 1400 infants were allocated to fast feeds and 1404 infants were allocated to slow feeds. MAIN OUTCOME MEASURES The primary outcome was survival without moderate or severe neurodevelopmental disability at 24 months of age, corrected for gestational age. The secondary outcomes were mortality; moderate or severe neurodevelopmental disability at 24 months corrected for gestational age; death before discharge home; microbiologically confirmed or clinically suspected late-onset sepsis; necrotising enterocolitis (Bell's stage 2 or 3); time taken to reach full milk feeds (tolerating 150 ml/kg/day for 3 consecutive days); growth from birth to discharge; duration of parenteral feeding; time in intensive care; duration of hospital stay; diagnosis of cerebral palsy by a doctor or other health professional; and individual components of the definition of moderate or severe neurodevelopmental disability. RESULTS The results showed that survival without moderate or severe neurodevelopmental disability at 24 months occurred in 802 out of 1224 (65.5%) infants allocated to faster increments and 848 out of 1246 (68.1%) infants allocated to slower increments (adjusted risk ratio 0.96, 95% confidence interval 0.92 to 1.01). There was no significant difference between groups in the risk of the individual components of the primary outcome or in the important hospital outcomes: late-onset sepsis (adjusted risk ratio 0.96, 95% confidence interval 0.86 to 1.07) or necrotising enterocolitis (adjusted risk ratio 0.88, 95% confidence interval 0.68 to 1.16). Cost-consequence analysis showed that the faster feed increment rate was less costly but also less effective than the slower rate in terms of achieving the primary outcome, so was therefore found to not be cost-effective. Four unexpected serious adverse events were reported, two in each group. None was assessed as being causally related to the intervention. LIMITATIONS The study could not be blinded, so care may have been affected by knowledge of allocation. Although well powered for comparisons of all infants, subgroup comparisons were underpowered. CONCLUSIONS No clear advantage was identified for the important outcomes in very preterm or very low-birthweight infants when milk feeds were advanced in daily volume increments of 30 ml/kg/day or 18 ml/kg/day. In terms of future work, the interaction of different milk types with increments merits further examination, as may different increments in infants at the extremes of gestation or birthweight. TRIAL REGISTRATION Current Controlled Trials ISRCTN76463425. FUNDING This project was funded by the National Institute for Health Research Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 24, No. 18. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Jon Dorling
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Oliver Hewer
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Madeleine Hurd
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Vasha Bari
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Beth Bosiak
- Women's College Hospital, Toronto, ON, Canada
| | - Ursula Bowler
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Andrew King
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Louise Linsell
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - David Murray
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Omar Omar
- Birmingham Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | | | - Catherine Rounding
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - John Townend
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | | | - Janet Berrington
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Elaine Boyle
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Nicholas Embleton
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Samantha Johnson
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Alison Leaf
- National Institute for Health Research Southampton Biomedical Research Centre Department of Child Health, University of Southampton, Southampton, UK
| | - Kenny McCormick
- John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| | | | - Tracy Roberts
- School of Health and Population Sciences, University of Birmingham, Birmingham, UK
| | - Ben Stenson
- The Simpson Centre for Reproductive Health, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Warda Tahir
- School of Health and Population Sciences, University of Birmingham, Birmingham, UK
| | - Mark Monahan
- School of Health and Population Sciences, University of Birmingham, Birmingham, UK
| | - Judy Richards
- Institute of Health & Society, Newcastle University, Newcastle upon Tyne, UK
| | - Judith Rankin
- Institute of Health & Society, Newcastle University, Newcastle upon Tyne, UK
| | - Edmund Juszczak
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
32
|
Walsh V, Brown JVE, Copperthwaite BR, Oddie SJ, McGuire W. Early full enteral feeding for preterm or low birth weight infants. Cochrane Database Syst Rev 2020; 2020:CD013542. [PMCID: PMC7067362 DOI: 10.1002/14651858.cd013542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/01/2024]
Abstract
This is a protocol for a Cochrane Review (Intervention). The objectives are as follows: We aim to assess the benefits and harms of early full enteral nutrition versus progressive introduction of enteral feeds in preterm or low birth weight (LBW) infants. Where data are available, we will undertake subgroup analyses of very preterm or very low birth weight (VLBW) infants (versus infants born after a longer gestation or with higher birth weight), infants who are 'small for gestational age' at birth (versus those deemed 'appropriate for gestation'), infants fed with human milk only (versus formula‐fed infants), and trials set in low‐ or middle‐income countries (versus high‐income countries).
Collapse
Affiliation(s)
- Verena Walsh
- University of YorkCentre for Reviews and DisseminationYorkUKY010 5DD
| | | | | | - Sam J Oddie
- Bradford Royal InfirmaryDuckworth LaneBradfordUKBD9 6RJ
| | - William McGuire
- University of YorkCentre for Reviews and DisseminationYorkUKY010 5DD
| | | |
Collapse
|
33
|
Thai JD, Gregory KE. Bioactive Factors in Human Breast Milk Attenuate Intestinal Inflammation during Early Life. Nutrients 2020; 12:E581. [PMID: 32102231 PMCID: PMC7071406 DOI: 10.3390/nu12020581] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/15/2020] [Accepted: 02/16/2020] [Indexed: 12/14/2022] Open
Abstract
Human breast milk is well known as the ideal source of nutrition during early life, ensuring optimal growth during infancy and early childhood. Breast milk is also the source of many unique and dynamic bioactive components that play a key role in the development of the immune system. These bioactive components include essential microbes, human milk oligosaccharides (HMOs), immunoglobulins, lactoferrin and dietary polyunsaturated fatty acids. These factors all interact with intestinal commensal bacteria and/or immune cells, playing a critical role in establishment of the intestinal microbiome and ultimately influencing intestinal inflammation and gut health during early life. Exposure to breast milk has been associated with a decreased incidence and severity of necrotizing enterocolitis (NEC), a devastating disease characterized by overwhelming intestinal inflammation and high morbidity among preterm infants. For this reason, breast milk is considered a protective factor against NEC and aberrant intestinal inflammation common in preterm infants. In this review, we will describe the key microbial, immunological, and metabolic components of breast milk that have been shown to play a role in the mechanisms of intestinal inflammation and/or NEC prevention.
Collapse
Affiliation(s)
- Julie D. Thai
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Katherine E. Gregory
- Department of Pediatric Newborn Medicine, Department of Nursing, Brigham and Women’s Hospital, Boston, MA 02115, USA;
| |
Collapse
|
34
|
Abstract
Late-onset sepsis (LOS) and necrotising enterocolitis (NEC) account for the highest number of deaths in premature infants and often cause severe morbidity in survivors. NEC is an inflammatory mediated condition, but its pathophysiology remains poorly understood. There is increasing evidence that in LOS the causative organism most often translocates from the gut. No causative microorganism has been consistently associated with either LOS or NEC, but an aberrant gut microbiome development could play a pivotal role. A low bacterial diversity and a delay in anaerobic bacteria colonization may predispose preterm infants to disease development. Conversely, a predominance of Bifidobacterium species and breast milk feeding might help to prevent disease onset. With numerous studies reporting conflicting results, further research is needed to better understand the role of microorganisms and type of feeding in the health status of preterm infants.
Collapse
Affiliation(s)
- Andrea C Masi
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | |
Collapse
|
35
|
Brown JVE, Walsh V, McGuire W. Formula versus maternal breast milk for feeding preterm or low birth weight infants. Cochrane Database Syst Rev 2019; 8:CD002972. [PMID: 31452191 PMCID: PMC6710607 DOI: 10.1002/14651858.cd002972.pub3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Artificial formula can be manipulated to contain higher amounts of macro-nutrients than maternal breast milk but breast milk confers important immuno-nutritional advantages for preterm or low birth weight (LBW) infants. OBJECTIVES To determine the effect of feeding preterm or LBW infants with formula compared with maternal breast milk on growth and developmental outcomes. SEARCH METHODS We used the standard strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL 2018, Issue 9), and Ovid MEDLINE, Ovid Embase, Ovid Maternity & Infant Care Database, and CINAHL to October 2018. We searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles. SELECTION CRITERIA Randomised or quasi-randomised controlled trials that compared feeding preterm or low birth weight infants with formula versus maternal breast milk. DATA COLLECTION AND ANALYSIS Two review authors planned independently to assess trial eligibility and risk of bias, and extract data. We planned to analyse treatment effects as described in the individual trials and report risk ratios and risk differences for dichotomous data, and mean differences for continuous data, with 95% confidence intervals. We planned to use a fixed-effect model in meta-analyses and to explore potential causes of heterogeneity in subgroup analyses. We planned to use the GRADE approach to assess the certainty of evidence. MAIN RESULTS We did not identify any eligible trials. AUTHORS' CONCLUSIONS There are no trials of formula versus maternal breast milk for feeding preterm or low birth weight infants. Such trials are unlikely to be conducted because of the difficulty of allocating an alternative form of nutrition to an infant whose mother wishes to feed with her own breast milk. Maternal breast milk remains the default choice of enteral nutrition because observational studies, and meta-analyses of trials comparing feeding with formula versus donor breast milk, suggest that feeding with breast milk has major immuno-nutritional advantages for preterm or low birth weight infants.
Collapse
|
36
|
Abstract
Probiotic administration to preterm infants is not universal despite randomised trial data from >10,000 infants, significant observational data and multiple meta-analyses. Advocates point to reductions in necrotising enterocolitis and sepsis, 'sceptics' hold concerns over data quality/interpretation or risks. Issues revolve around different products, primary outcomes, uncertain dosing strategies and individual large 'negative' trials alongside probiotic associated sepsis and quality control concerns. We review concerns and how to move probiotic use forward. Surprisingly little is known about parental perspectives, vital to inform next steps. How to share information and decisions around probiotic use now, and how this impacts on future available strategies is discussed. We address placebo controlled trials and propose alternate designs, including head to head studies, using 'routine' data collection systems, opt out consents and 'learning technologies' embedded in health care systems. We also raise the importance of underpinning mechanistic work to inform future trials.
Collapse
|
37
|
Walsh V, Brown JVE, Askie LM, Embleton ND, McGuire W. Nutrient-enriched formula versus standard formula for preterm infants. Cochrane Database Syst Rev 2019; 7:CD004204. [PMID: 31314903 PMCID: PMC6636703 DOI: 10.1002/14651858.cd004204.pub3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Preterm infants may accumulate nutrient deficits leading to extrauterine growth restriction. Feeding preterm infants with nutrient-enriched rather than standard formula might increase nutrient accretion and growth rates and might improve neurodevelopmental outcomes. OBJECTIVES To compare the effects of feeding with nutrient-enriched formula versus standard formula on growth and development of preterm infants. SEARCH METHODS We used the Cochrane Neonatal standard search strategy. This included electronic searches of the Cochrane Central Register of Controlled Trials (CENTRAL; 2018, Issue 11), MEDLINE, Embase, and the Cumulative Index to Nursing and Allied Health Literature (until November 2018), as well as conference proceedings, previous reviews, and clinical trials databases. SELECTION CRITERIA Randomised and quasi-randomised controlled trials that compared feeding preterm infants with nutrient-enriched formula (protein and energy plus minerals, vitamins, or other nutrients) versus standard formula. DATA COLLECTION AND ANALYSIS We extracted data using the Cochrane Neonatal standard methods. Two review authors separately evaluated trial quality and extracted and synthesised data using risk ratios (RRs), risk differences, and mean differences (MDs). We assessed certainty of evidence at the outcome level using Grading of Recommendations Assessment, Development and Evaluation (GRADE) methods. MAIN RESULTS We identified seven trials in which a total of 590 preterm infants participated. Most participants were clinically stable preterm infants of birth weight less than 1850 g. Few participants were extremely preterm, extremely low birth weight, or growth restricted at birth. Trials were conducted more than 30 years ago, were formula industry funded, and were small with methodological weaknesses (including lack of masking) that might bias effect estimates. Meta-analyses of in-hospital growth parameters were limited by statistical heterogeneity. There is no evidence of an effect on time to regain birth weight (MD -1.48 days, 95% confidence interval (CI) -4.73 to 1.77) and low-certainty evidence suggests that feeding with nutrient-enriched formula increases in-hospital rates of weight gain (MD 2.43 g/kg/d, 95% CI 1.60 to 3.26) and head circumference growth (MD 1.04 mm/week, 95% CI 0.18 to 1.89). Meta-analysis did not show an effect on the average rate of length gain (MD 0.22 mm/week, 95% CI -0.70 to 1.13). Fewer data are available for growth and developmental outcomes assessed beyond infancy, and these do not show consistent effects of nutrient-enriched formula feeding. Data from two trials did not show an effect on Bayley Mental Development Index scores at 18 months post term (MD 2.87, 95% CI -1.38 to 7.12; moderate-certainty evidence). Infants who received nutrient-enriched formula had higher Bayley Psychomotor Development Index scores at 18 months post term (MD 6.56. 95% CI 2.87 to 10.26; low-certainty evidence), but no evidence suggested an effect on cerebral palsy (typical RR 0.79, 95% CI 0.30 to 2.07; 2 studies, 377 infants). Available data did not indicate any other benefits or harms and provided low-certainty evidence about the effect of nutrient-enriched formula feeding on the risk of necrotising enterocolitis in preterm infants (typical RR 0.72, 95% CI 0.41 to 1.25; 3 studies, 489 infants). AUTHORS' CONCLUSIONS Available trial data show that feeding preterm infants nutrient-enriched (compared with standard) formulas has only modest effects on growth rates during their initial hospital admission. No evidence suggests effects on long-term growth or development. The GRADE assessment indicates that the certainty of this evidence is low, and that these findings should be interpreted and applied with caution. Further randomised trials would be needed to resolve this uncertainty.
Collapse
Affiliation(s)
- Verena Walsh
- University of YorkCentre for Reviews and DisseminationYorkUKY010 5DD
| | | | - Lisa M Askie
- University of SydneyNHMRC Clinical Trials CentreLocked Bag 77CamperdownNSWAustralia2050
| | - Nicholas D Embleton
- Newcastle Hospitals NHS Foundation Trust and University of NewcastleNewcastle Neonatal ServiceRichardson RoadNewcastle upon TyneUKNE1 4LP
| | - William McGuire
- University of YorkCentre for Reviews and DisseminationYorkUKY010 5DD
| | | |
Collapse
|
38
|
Kumbhare SV, Patangia DVV, Patil RH, Shouche YS, Patil NP. Factors influencing the gut microbiome in children: from infancy to childhood. J Biosci 2019; 44:49. [PMID: 31180062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The human microbiota plays a crucial role in educating the immune system and influencing host health right since birth. Various maternal factors along with the vertical microbial transfer from the mother, as well as the horizontal environmental transmission and internal factors relating to the infant, play a crucial role in modulating the gut microbiota. The early life microflora is highly unstable and undergoes dynamic changes during the first few years, converging towards a more stabilized adult microbiota by co-evolving with the host by the age of 3-4 years. Microbiota studies have underlined the role of dysbiosis in developing several metabolic disorders like obesity, diabetes and immune-related disorders like asthma, to name a few. Thus, understanding early life microbial composition and various factors affecting the microbial community will provide a platform for developing strategies/techniques to maintain host health by restoring gut microbial flora. This review focuses on the factors that affect the microbial composition of the foetus in utero, during birth, infancy through childhood.
Collapse
Affiliation(s)
- Shreyas V Kumbhare
- National Centre for Cell Science, Savitribai Phule University of Pune Campus, Pune, Maharashtra 411007, India
| | | | | | | | | |
Collapse
|
39
|
Griffiths J, Jenkins P, Vargova M, Bowler U, Juszczak E, King A, Linsell L, Murray D, Partlett C, Patel M, Berrington J, Embleton N, Dorling J, Heath PT, McGuire W, Oddie S. Enteral lactoferrin to prevent infection for very preterm infants: the ELFIN RCT. Health Technol Assess 2019; 22:1-60. [PMID: 30574860 DOI: 10.3310/hta22740] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Infections acquired in hospital are an important cause of morbidity and mortality in very preterm infants. Several small trials have suggested that supplementing the enteral diet of very preterm infants with lactoferrin, an antimicrobial protein processed from cow's milk, prevents infections and associated complications. OBJECTIVE To determine whether or not enteral supplementation with bovine lactoferrin (The Tatua Cooperative Dairy Company Ltd, Morrinsville, New Zealand) reduces the risk of late-onset infection (acquired > 72 hours after birth) and other morbidity and mortality in very preterm infants. DESIGN Randomised, placebo-controlled, parallel-group trial. Randomisation was via a web-based portal and used an algorithm that minimised for recruitment site, weeks of gestation, sex and single versus multiple births. SETTING UK neonatal units between May 2014 and September 2017. PARTICIPANTS Infants born at < 32 weeks' gestation and aged < 72 hours at trial enrolment. INTERVENTIONS Eligible infants were allocated individually (1 : 1 ratio) to receive enteral bovine lactoferrin (150 mg/kg/day; maximum 300 mg/day) or sucrose (British Sugar, Peterborough, UK) placebo (same dose) once daily from trial entry until a postmenstrual age of 34 weeks. Parents, caregivers and outcome assessors were unaware of group assignment. OUTCOMES Primary outcome - microbiologically confirmed or clinically suspected late-onset infection. Secondary outcomes - microbiologically confirmed infection; all-cause mortality; severe necrotising enterocolitis (NEC); retinopathy of prematurity (ROP); bronchopulmonary dysplasia (BPD); a composite of infection, NEC, ROP, BPD and mortality; days of receipt of antimicrobials until 34 weeks' postmenstrual age; length of stay in hospital; and length of stay in intensive care, high-dependency and special-care settings. RESULTS Of 2203 enrolled infants, primary outcome data were available for 2182 infants (99%). In the intervention group, 316 out of 1093 (28.9%) infants acquired a late-onset infection versus 334 out of 1089 (30.7%) infants in the control group [adjusted risk ratio (RR) 0.95, 95% confidence interval (CI) 0.86 to 1.04]. There were no significant differences in any secondary outcomes: microbiologically confirmed infection (RR 1.05, 99% CI 0.87 to 1.26), mortality (RR 1.05, 99% CI 0.66 to 1.68), NEC (RR 1.13, 99% CI 0.68 to 1.89), ROP (RR 0.89, 99% CI 0.62 to 1.28), BPD (RR 1.01, 99% CI 0.90 to 1.13), or a composite of infection, NEC, ROP, BPD and mortality (RR 1.01, 99% CI 0.94 to 1.08). There were no differences in the number of days of receipt of antimicrobials, length of stay in hospital, or length of stay in intensive care, high-dependency or special-care settings. There were 16 reports of serious adverse events for infants in the lactoferrin group and 10 for infants in the sucrose group. CONCLUSIONS Enteral supplementation with bovine lactoferrin does not reduce the incidence of infection, mortality or other morbidity in very preterm infants. FUTURE WORK Increase the precision of the estimates of effect on rarer secondary outcomes by combining the data in a meta-analysis with data from other trials. A mechanistic study is being conducted in a subgroup of trial participants to explore whether or not lactoferrin supplementation affects the intestinal microbiome and metabolite profile of very preterm infants. TRIAL REGISTRATION Current Controlled Trials ISRCTN88261002. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 22, No. 74. See the NIHR Journals Library website for further project information. This trial was also sponsored by the University of Oxford, Oxford, UK. The funder provided advice and support and monitored study progress but did not have a role in study design or data collection, analysis and interpretation.
Collapse
Affiliation(s)
- James Griffiths
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Paula Jenkins
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Monika Vargova
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Ursula Bowler
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Edmund Juszczak
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Andrew King
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Louise Linsell
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - David Murray
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | | | | | - Janet Berrington
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Nicholas Embleton
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | | | - Paul T Heath
- St George's, University of London and St George's University Hospitals NHS Trust, London, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sam Oddie
- Bradford Institute for Health Research, Bradford, UK
| |
Collapse
|
40
|
Walsh V, Brown JVE, Askie LM, Embleton ND, McGuire W. Nutrient-enriched formula versus standard formula milk for preterm infants. Hippokratia 2019. [DOI: 10.1002/14651858.cd004204.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Verena Walsh
- University of York; Centre for Reviews and Dissemination; York UK Y010 5DD
| | | | - Lisa M Askie
- University of Sydney; NHMRC Clinical Trials Centre; Locked Bag 77 Camperdown NSW Australia 2050
| | - Nicholas D Embleton
- Newcastle Hospitals NHS Foundation Trust and University of Newcastle; Newcastle Neonatal Service; Richardson Road Newcastle upon Tyne UK NE1 4LP
| | - William McGuire
- University of York; Centre for Reviews and Dissemination; York UK Y010 5DD
| |
Collapse
|
41
|
|
42
|
Enteral lactoferrin supplementation for very preterm infants: a randomised placebo-controlled trial. Lancet 2019; 393:423-433. [PMID: 30635141 PMCID: PMC6356450 DOI: 10.1016/s0140-6736(18)32221-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 08/31/2018] [Accepted: 09/05/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Infections acquired in hospital are an important cause of morbidity and mortality in very preterm infants. Several small trials have suggested that supplementing the enteral diet of very preterm infants with lactoferrin, an antimicrobial protein processed from cow's milk, prevents infections and associated complications. The aim of this large randomised controlled trial was to collect data to enhance the validity and applicability of the evidence from previous trials to inform practice. METHODS In this randomised placebo-controlled trial, we recruited very preterm infants born before 32 weeks' gestation in 37 UK hospitals and younger than 72 h at randomisation. Exclusion criteria were presence of a severe congenital anomaly, anticipated enteral fasting for longer than 14 days, or no realistic prospect of survival. Eligible infants were randomly assigned (1:1) to receive either enteral bovine lactoferrin (150 mg/kg per day; maximum 300 mg/day; lactoferrin group) or sucrose (same dose; control group) once daily until 34 weeks' postmenstrual age. Web-based randomisation minimised for recruitment site, gestation (completed weeks), sex, and single versus multifetal pregnancy. Parents, caregivers, and outcome assessors were unaware of group assignment. The primary outcome was microbiologically confirmed or clinically suspected late-onset infection (occurring >72 h after birth), which was assessed in all participants for whom primary outcome data was available by calculating the relative risk ratio with 95% CI between the two groups. The trial is registered with the International Standard Randomised Controlled Trial Number 88261002. FINDINGS We recruited 2203 participants between May 7, 2014, and Sept 28, 2017, of whom 1099 were assigned to the lactoferrin group and 1104 to the control group. Four infants had consent withdrawn or unconfirmed, leaving 1098 infants in the lactoferrin group and 1101 in the sucrose group. Primary outcome data for 2182 infants (1093 [99·5%] of 1098 in the lactoferrin group and 1089 [99·0] of 1101 in the control group) were available for inclusion in the modified intention-to-treat analyses. 316 (29%) of 1093 infants in the intervention group acquired a late-onset infection versus 334 (31%) of 1089 in the control group. The risk ratio adjusted for minimisation factors was 0·95 (95% CI 0·86-1·04; p=0·233). During the trial there were 16 serious adverse events for infants in the lactoferrin group and 10 for infants in the control group. Two events in the lactoferrin group (one case of blood in stool and one death after intestinal perforation) were assessed as being possibly related to the trial intervention. INTERPRETATION Enteral supplementation with bovine lactoferrin does not reduce the risk of late-onset infection in very preterm infants. These data do not support its routine use to prevent late-onset infection and associated morbidity or mortality in very preterm infants. FUNDING UK National Institute for Health Research Health Technology Assessment programme (10/57/49).
Collapse
|
43
|
Walsh V, McGuire W. Immunonutrition for Preterm Infants. Neonatology 2019; 115:398-405. [PMID: 30974431 DOI: 10.1159/000497332] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 11/19/2022]
Abstract
Care and outcomes for very preterm infants continue to improve, but important causes of mortality and acute and long-term morbidity associated with prolonged hospitalisation remain. Necrotising enterocolitis (NEC) and late-onset infection have emerged as the major causes of death beyond the early neonatal period and of neurodisability in very preterm infants. Although the pathogenesis of these conditions is incompletely understood, it appears to be related to the content and mode of delivery of the enteral diet, particularly the impact of immunonutrients from human breast milk on the microbial and metabolic balance within the immature intestine. Evidence exists to support investment in measures to help mothers to express breast milk as the primary source of nutrition for their very preterm infants. In the absence of maternal milk, pasteurised donor breast milk provides protection against NEC, but its nutritive adequacy is not clear and its cost-effectiveness is uncertain. Supplementation with individual immunonutrients, including immunoglobulins and lactoferrin, has not been shown to be effective in preventing NEC or infection in randomised controlled trials. The evidence base for prebiotics and probiotics is stronger, but concerns exist about the choice, safety and availability of formulations. Other strategies - including avoidance of drugs such as gastric acid suppressants that compromise innate immunity, as well as evidence-based progressive feeding strategies that reduce exposure to invasive interventions - are emerging as key components of care packages to reduce the burden of NEC, infection and associated growth and developmental faltering for very preterm infants.
Collapse
Affiliation(s)
- Verena Walsh
- Centre for Reviews and Dissemination, University of York, York, United Kingdom
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, United Kingdom,
| |
Collapse
|
44
|
Neu J, Pammi M. Necrotizing enterocolitis: The intestinal microbiome, metabolome and inflammatory mediators. Semin Fetal Neonatal Med 2018; 23:400-405. [PMID: 30172660 DOI: 10.1016/j.siny.2018.08.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Necrotizing enterocolitis (NEC) is a disease of preterm infants and associated with significant mortality and morbidity. Although the pathogenesis of NEC is not clear, microbial dysbiosis, with a bloom of the phylum Proteobacteria, has been reported. Antibiotics and the use of H2 blockers, which affect the gut microbiome, are associated with increased incidence of NEC. In association with dysbiosis, inflammatory processes are upregulated with increased Toll-like receptor signaling, leading to translocation of nuclear factor kappa-β, a transcription factor that induces transcription of various pro-inflammatory cytokines and chemokines. Microbial metabolites, short chain fatty acids including acetate and butyrate, may modulate immunity, inflammation, intestinal integrity and regulate transcription by epigenetic mechanisms. Evaluation of microbiome and metabolome may provide biomarkers for early diagnosis of NEC and microbial therapeutic approaches to correct microbial dysbiosis.
Collapse
Affiliation(s)
- Josef Neu
- Section of Neonatology, Department of Pediatrics, University of Florida, Gainesville, FL, USA.
| | - Mohan Pammi
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
45
|
Laing B, Barnett MPG, Marlow G, Nasef NA, Ferguson LR. An update on the role of gut microbiota in chronic inflammatory diseases, and potential therapeutic targets. Expert Rev Gastroenterol Hepatol 2018; 12:969-983. [PMID: 30052094 DOI: 10.1080/17474124.2018.1505497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The human microbiome plays a critical role in human health, having metabolic, protective, and trophic functions, depending upon its' exact composition. This composition is affected by a number of factors, including the genetic background of the individual, early life factors (including method of birth, length of breastfeeding) and nature of the diet and other environmental exposures (including cigarette smoking) and general life habits. It plays a key role in the control of inflammation, and in turn, its' composition is significantly influenced by inflammation. Areas covered: We consider metabolic, protective, and trophic functions of the microbiome and influences through the lifespan from post-partum effects, to diet later in life in healthy older adults, the effects of aging on both its' composition, and influence on health and potential therapeutic targets that may have anti-inflammatory effects. Expert commentary: The future will see the growth of more effective therapies targeting the microbiome particularly with respect to the use of specific nutrients and diets personalized to the individual.
Collapse
Affiliation(s)
- Bobbi Laing
- a Discipline of Nutrition and Dietetics, Faculty of Medical Health Sciences , The University of Auckland , Auckland , New Zealand.,b School of Nursing, Faculty of Medical and Health Sciences , The University of Auckland , Auckland , New Zealand
| | - Matthew P G Barnett
- c Food Nutrition & Health Team, Food & Bio-Based Products Group , AgResearch Limited , Palmerston North , New Zealand.,d Liggins Institute , The High-Value Nutrition National Science Challenge , Auckland , New Zealand.,e Riddet Institute , Massey University , Palmerston North , New Zealand
| | - Gareth Marlow
- f Institute of Medical Genetics , Cardiff University , Cardiff , Wales , UK
| | - Noha Ahmed Nasef
- e Riddet Institute , Massey University , Palmerston North , New Zealand.,g College of Health, Massey Institute of Food Science and Technology , Palmerston North , New Zealand
| | - Lynnette R Ferguson
- a Discipline of Nutrition and Dietetics, Faculty of Medical Health Sciences , The University of Auckland , Auckland , New Zealand.,h Auckland Cancer Research Society, Faculty of Medical and Health Sciences, Grafton Campus , The University of Auckland , Auckland , New Zealand
| |
Collapse
|
46
|
Effects of lactoferrin on neonatal pathogens and Bifidobacterium breve in human breast milk. PLoS One 2018; 13:e0201819. [PMID: 30133470 PMCID: PMC6104981 DOI: 10.1371/journal.pone.0201819] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/22/2018] [Indexed: 01/22/2023] Open
Abstract
Supplementation with probiotics in preterm infants reduces necrotizing enterocolitis and sepsis. Bovine lactoferrin is a promising supplement that may further reduce disease burden, but its effects on probiotic bacteria in human breast milk has not been evaluated. We aimed to characterise the antimicrobial activity of bovine and human lactoferrin in human breast milk against probiotics and typical neonatal sepsis pathogens. Lactoferrin levels were determined by enzyme linked immunosorbent assay in fresh and pasteurised human breast milk. The neonatal pathogens Staphylococcus epidermidis and Escherichia coli, and the probiotic Bifidobacterium breve strain M-16V were cultured in human breast milk or infant formula in the presence or absence of clinically relevant doses of bovine or human lactoferrin. Standard microbiological methods were used to determine the effects of lactoferrin on bacterial growth. Unpasteurised human breast milk contained significantly higher lactoferrin levels and resulted in superior inhibition of pathogenic bacterial growth compared to infant formula and pasteurised human breast milk. Human lactoferrin was significantly more effective at inhibiting bacterial growth, when compared to bovine lactoferrin. Supplementation with human lactoferrin or high dose bovine lactoferrin inhibited growth of the probiotic strain B. breve M-16V in pasteurised human breast milk. Although unpasteurised human breast milk and human lactoferrin had the greatest antimicrobial activity against all bacterial species tested, higher doses of bovine lactoferrin also showed activity against B. breve and. S. epidermidis. This study suggests that simultaneous administration of lactoferrins and probiotics may affect colonisation with probiotic bacteria, warranting further investigations.
Collapse
|
47
|
Berkhout DJC, Niemarkt HJ, de Boer NKH, Benninga MA, de Meij TGJ. The potential of gut microbiota and fecal volatile organic compounds analysis as early diagnostic biomarker for necrotizing enterocolitis and sepsis in preterm infants. Expert Rev Gastroenterol Hepatol 2018; 12:457-470. [PMID: 29488419 DOI: 10.1080/17474124.2018.1446826] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although the exact pathophysiological mechanisms of both necrotizing enterocolitis (NEC) and late-onset sepsis (LOS) in preterm infants are yet to be elucidated, evidence is emerging that the gut microbiota plays a key role in their pathophysiology. Areas covered: In this review, initial microbial colonization and factors influencing microbiota composition are discussed. For both NEC and LOS, an overview of studies investigating preclinical alterations in gut microbiota composition and fecal volatile organic compounds (VOCs) is provided. Fecal VOCs are considered to reflect not only gut microbiota composition, but also their metabolic activity and concurrent interaction with the host. Expert review: Heterogeneity in study protocols and applied analytical techniques hampers reliable comparison between outcomes of different microbiota studies, limiting the ability to draw firm conclusions. This dilemma is illustrated by the finding that study results often cannot be reproduced, or even contradict each other. A NEC- and sepsis specific microbial or metabolic signature has not yet been discovered. Identification of 'disease-specific' VOCs and microbiota composition may increase understanding on pathophysiological mechanisms and may allow for development of an accurate screening tool, opening avenues towards timely identification and initiation of targeted treatment for preterm infants at increased risk for NEC and sepsis.
Collapse
Affiliation(s)
- Daniel Johannes Cornelis Berkhout
- a Department of Pediatric Gastroenterology , Emma Children's Hospital/Academic Medical Center , Amsterdam , the Netherlands.,b Department of Pediatric Gastroenterology , VU University Medical Center , Amsterdam , the Netherlands
| | | | - Nanne Klaas Hendrik de Boer
- d Department of Gastroenterology and Hepatology , VU University Medical Center , Amsterdam , the Netherlands
| | - Marc Alexander Benninga
- a Department of Pediatric Gastroenterology , Emma Children's Hospital/Academic Medical Center , Amsterdam , the Netherlands
| | | |
Collapse
|
48
|
Paul SP, Kirkham EN, Hawton KA, Mannix PA. Feeding growth restricted premature neonates: a challenging perspective. Sudan J Paediatr 2018; 18:5-14. [PMID: 30799892 DOI: 10.24911/sjp.106-1519511375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Nutrition in the postnatal period is essential to achieve optimal growth and maintain biochemical normality. Feeding growth-restricted premature neonates remains a big challenge for pediatricians and neonatologists. The choice of milk is one of the biggest challenges. Breast milk is recommended, although feeding with preterm formulas can ensure a more consistent delivery of optimal levels of nutrients. The timing of introduction of feeds and the rate of advancement of those feeds in preterm infants are both topics of significant controversy. Early feeding is advantageous because it improves the functional adaptation of the gastrointestinal tract and reduces the duration of total parenteral nutrition. A faster rate of advancement will also reduce the duration of need for parenteral nutrition. Despite this, enteral feeding is often delayed and is often slowly increased in high-risk infants because of a possible increased risk of necrotizing enterocolitis (NEC). Growth-restricted neonates are at increased risk of developing NEC due to a combination of antenatal and postnatal disturbances in gut perfusion. If enteral feeding is introduced earlier and advanced more quickly, this may lead to increased risk of NEC, but slower feeds extend the duration of parenteral nutrition and its risks and may have adverse consequences for survival, growth, and development. Premature infants pose a significant nutritional challenge. Overall, we would suggest the preferential use of human breast milk, early minimal enteral feeds, and standardized feeding protocols with cautious advancements of feeds to facilitate gastrointestinal adaptation and reduce the risk of NEC, however further research is needed.
Collapse
|
49
|
Stewart CJ, Embleton ND, Marrs ECL, Smith DP, Fofanova T, Nelson A, Skeath T, Perry JD, Petrosino JF, Berrington JE, Cummings SP. Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls. MICROBIOME 2017; 5:75. [PMID: 28701177 PMCID: PMC5508794 DOI: 10.1186/s40168-017-0295-1] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/29/2017] [Indexed: 05/28/2023]
Abstract
BACKGROUND Late onset sepsis (LOS) in preterm infants is associated with considerable morbidity and mortality. While studies have implicated gut bacteria in the aetiology of the disease, functional analysis and mechanistic insights are generally lacking. We performed temporal bacterial (n = 613) and metabolomic (n = 63) profiling on extensively sampled stool from 7 infants with LOS and 28 matched healthy (no LOS or NEC) controls. RESULTS The bacteria isolated in diagnostic blood culture usually corresponded to the dominant bacterial genera in the gut microbiome. Longitudinal changes were monitored based on preterm gut community types (PGCTs), where control infants had an increased number of PGCTs compared to LOS infants (P = 0.011). PGCT 6, characterised by Bifidobacteria dominance, was only present in control infants. Metabolite profiles differed between LOS and control infants at diagnosis and 7 days later, but not 7 days prior to diagnosis. Bifidobacteria was positively correlated with control metabolites, including raffinose, sucrose, and acetic acid. CONCLUSIONS Using multi-omic analysis, we show that the gut microbiome is involved in the pathogenesis of LOS. While the causative agent of LOS varies, it is usually abundant in the gut. Bifidobacteria dominance was associated with control infants, and the presence of this organism may directly protect, or act as a marker for protection, against gut epithelial translocation. While the metabolomic data is preliminary, the findings support that gut development and protection in preterm infants is associated with increased in prebiotic oligosaccharides (e.g. raffinose) and the growth of beneficial bacteria (e.g. Bifidobacterium).
Collapse
Affiliation(s)
- Christopher J Stewart
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, 77030, USA.
| | - Nicholas D Embleton
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, NE1 4LP.77030, UK
| | - Emma C L Marrs
- Department of Microbiology, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - Daniel P Smith
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Tatiana Fofanova
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Andrew Nelson
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, NE1 8ST, UK
| | - Tom Skeath
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, NE1 4LP.77030, UK
| | - John D Perry
- Department of Microbiology, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - Joseph F Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Janet E Berrington
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, NE1 4LP.77030, UK
| | - Stephen P Cummings
- School of Science and Engineering, Teesside University, Middlesbrough, TS1 3BX, UK
| |
Collapse
|
50
|
Walsh V, Brown JVE, Askie LM, Embleton ND, McGuire W. Nutrient-enriched formula versus standard formula milk for preterm infants. Hippokratia 2003. [DOI: 10.1002/14651858.cd004204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Verena Walsh
- University of York; Centre for Reviews and Dissemination; York UK Y010 5DD
| | | | - Lisa M Askie
- University of Sydney; NHMRC Clinical Trials Centre; Locked Bag 77 Camperdown NSW Australia 2050
| | - Nicholas D Embleton
- Newcastle Hospitals NHS Foundation Trust and University of Newcastle; Newcastle Neonatal Service; Richardson Road Newcastle upon Tyne UK NE1 4LP
| | - William McGuire
- University of York; Centre for Reviews and Dissemination; York UK Y010 5DD
| |
Collapse
|