Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Rep ; 13(1): 14308, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37652940

ABSTRACT

Human milk contains over 200 distinct oligosaccharides, which are critical to shaping the developing neonatal gut microbiome. To investigate whether a complex mixture of human milk oligosaccharides (HMOs) would similarly modulate the adult gut microbiome, HMO-Concentrate derived from pooled donor breast milk was administered orally to 32 healthy adults for 7 days followed by 21 days of monitoring. Fecal samples were collected for 16S rRNA gene sequencing, shotgun metagenomics, and metabolomics analyses. HMO-Concentrate induced dose-dependent Bifidobacterium expansion, reduced microbial diversity, and altered microbial gene content. Following HMO cessation, a microbial succession occurred with diverse taxonomic changes-including Bacteroides expansion-that persisted through day 28. This was associated with altered microbial gene content, shifts in serum metabolite levels, and increased circulating TGFß and IL-10. Incubation of cultured adult microbiota with HMO-Concentrate induced dose-dependent compositional shifts that were not recapitulated by individual HMOs or defined mixtures of the 10 most abundant HMOs in HMO-Concentrate at their measured concentrations. These findings support that pooled donor HMOs can exert direct effects on adult gut microbiota and that complex mixtures including low abundance HMOs present in donor milk may be required for maximum effect.Registration: ClinicalTrials.gov NCT05516225.


Subject(s)
Gastrointestinal Microbiome , Milk, Human , Oligosaccharides , Adult , Female , Humans , Infant, Newborn , Milk, Human/chemistry , Oligosaccharides/pharmacology , RNA, Ribosomal, 16S/genetics
2.
Paediatr Respir Rev ; 48: 24-29, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37268507

ABSTRACT

The past decades have seen markedly improved survival of increasingly immature preterm infants, yet major health complications persist. This is particularly true for bronchopulmonary dysplasia (BPD), the chronic lung disease of prematurity, which has become the most common sequelae of prematurity and a significant predictor of respiratory morbidity throughout childhood as well as adult life, neurodevelopmental disability, cardiovascular disease, and even death. The need for novel approaches to reduce BPD and related complications of prematurity has never been more critical. Thus, despite major advances in the use of antenatal steroids, surfactant therapy, and improvements in respiratory support, there is a persistent need for developing therapeutic strategies that more specifically reflect our growing understanding of BPD in the post-surfactant age, or the "new BPD." In contrast with the severe lung injury leading to marked fibroproliferative disease from the past, the "new BPD" is primarily characterized by an arrest of lung development as related to more extreme prematurity. This distinction and the continued high incidence of BPD and related sequelae suggest the need to identify therapies that target critical mechanisms that support lung growth and maturation in conjunction with treatments to improve respiratory outcomes across the lifespan. As the prevention of BPD and its severity remains a primary goal, we highlight the concept from preclinical and early clinical observations that insulin-like growth factor 1 (IGF-1) can potentially support the natural sequence of lung growth as a replacement therapy after preterm birth. Data supporting this hypothesis are robust and include observations that low IGF-1 levels persist after extremely preterm birth in human infants and strong preclinical data from experimental models of BPD highlight the therapeutic benefit of IGF-1 in reducing disease. Importantly, phase 2a clinical data in extremely premature infants where replacement of IGF-1 with a human recombinant human IGF-1 complexed with its main IGF-1 binding protein 3, significantly reduced the most severe form of BPD, which is strongly associated with multiple morbidities that have lifelong consequences. As physiologic replacement therapy of surfactant heralded the success of reducing acute respiratory distress syndrome in preterm infants, the paradigm has the potential to become the platform for discovering the next generation of therapies like IGF-1, which becomes deficient after extremely premature birth where endogenous production by the infant is not sufficient to maintain the physiologic levels adequate to support normal organ development and maturation.


Subject(s)
Bronchopulmonary Dysplasia , Premature Birth , Pulmonary Surfactants , Respiratory System Agents , Infant , Adult , Infant, Newborn , Female , Humans , Pregnancy , Child , Infant, Premature , Insulin-Like Peptides , Insulin-Like Growth Factor I/therapeutic use , Lung , Bronchopulmonary Dysplasia/therapy , Pulmonary Surfactants/therapeutic use , Respiratory System Agents/therapeutic use , Surface-Active Agents/therapeutic use
3.
Am J Perinatol ; 39(S 01): S14-S17, 2022 12.
Article in English | MEDLINE | ID: mdl-36318942

ABSTRACT

Bronchopulmonary dysplasia (BPD) and poor neurodevelopmental outcome after preterm birth are closely associated. However, mechanistic links are uncertain. We are exploring the hypothesis that decreased circulating insulin-like growth factor (IGF)-1 after preterm birth due to the abrupt end of supply by the placenta impairs growth during critical windows of development in most organs, including the lung and brain. Throughout gestation, the fetus uses glycolysis as its main source of energy. Metabolism is mainly stopped at pyruvate, which serves as a "metabolic crossroad", allowing for the production of amino acids and other "building blocks" for new cells. Metabolic pathways are differentially regulated in the nucleus and the cytoplasm. The ratio between pyruvate dehydrogenase (PDH) and pyruvate dehydrogenase kinase (PDK) determines the biochemical activity which irreversibly metabolizes pyruvate to acetyl-co-A. Metabolites in the nucleus modulate epigenetic remodeling, an essential mechanism of normal growth and maturation during development. IGF-1 has been shown to contribute significantly to the development of virtually all organs, especially related to the regulation of microvascular growth, based on extensive studies of the brain, retina, lung, and intestine. With a preterm birth, the abrupt withdrawal of the placental supply of IGF-1 and its local production directly affects metabolism and microvascular development, which may contribute to a high risk for organ maldevelopment and injury after birth. We speculate that reduced bioavailability of IGF-1 is a possible link between lung and brain development disruption and increases susceptibility for major pulmonary and neurocognitive morbidities in preterm babies. KEY POINTS: · Metabolic changes inherent to prterm birth may cause epigenetic changes which cause "dysmaturational" development.. · IGF-1 may be the potential link between BPD and brain development.. · The ratio between pyruvate dehydrogenase and pyruvate dehydrogenase kinase determines the biochemical activity..


Subject(s)
Bronchopulmonary Dysplasia , Epigenesis, Genetic , Neurodevelopmental Disorders , Premature Birth , Female , Humans , Infant, Newborn , Pregnancy , Bronchopulmonary Dysplasia/complications , Insulin-Like Growth Factor I , Placenta/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/genetics
4.
J Med Econ ; 24(1): 1290-1298, 2021.
Article in English | MEDLINE | ID: mdl-34709122

ABSTRACT

BACKGROUND: Extremely preterm (EP) infants have high rates of respiratory morbidity and correspondingly high healthcare resource utilization. OBJECTIVES: Data from the PHARMO Perinatal Research Network were analyzed to quantify the burden of EP birth in the Netherlands. METHODS: A retrospective analysis included infants <28 weeks gestational age with a birth record in the Perinatal Registry (1999-2015) and data in the PHARMO Database Network. Outcomes of interest included select comorbidities, hospital readmissions, and costs of hospitalization and medication up to 1- and 2-years corrected age. Outcomes were stratified by birth period (1999-2005, 2000-2009, 2010-2015) and by diagnosis of bronchopulmonary dysplasia (BPD) and chronic lung disease (CLD). RESULTS: The cohort included 168 EP infants (37 born 1999-2005, 51 born 2006-2009, 80 born 2010-2015). Median (Q1-Q3) birth weights decreased by birth period from 970 (840-1,035) g in 1999-2005 to 853 (695-983) g in 2010-2015. Overall, BPD and CLD were reported during the birth hospitalization in 40% and 29% of infants, respectively; rates of BPD increased and rates of CLD decreased by birth period. Eighty-four percent of EP infants had an additional comorbidity. Mean (standard deviation) costs of birth hospitalization were €110,600 (€73,000) for 1999-2005, €119,350 (€60,650) for 2006-2009, and €138,800 (€130,100) for 2010-2015. Birth hospitalization and total costs for up to 1- and 2-years corrected age were higher for infants with BPD and/or CLD than for those without either complication. CONCLUSION: Healthcare resource utilization and costs for EP infants, especially for those with respiratory morbidities, increased between 1999 and 2015. Future cost-effectiveness analyses are essential to determine the economic impact of this change and underscore the need for new therapeutic interventions to decrease clinical sequelae in this vulnerable population.


Subject(s)
Bronchopulmonary Dysplasia , Premature Birth , Bronchopulmonary Dysplasia/epidemiology , Delivery of Health Care , Female , Gestational Age , Humans , Infant , Infant, Extremely Premature , Infant, Newborn , Morbidity , Netherlands/epidemiology , Pregnancy , Premature Birth/epidemiology , Retrospective Studies
5.
Pediatr Rev ; 40(10): 517-527, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31575803

ABSTRACT

Necrotizing enterocolitis (NEC) has been recognized for well over 5 decades yet remains the most common life-threatening surgical emergency in the newborn. The incidence of NEC has decreased steadily in preterm and very-low-birthweight infants over several decades and is typically uncommon in term newborns and infants with a birthweight greater than 2,500 g. Evidence accumulating during the past decade, however, suggests that practitioners should consider NEC in this broader subset of term infants with chromosomal and congenital anomalies complicated by heart or gastrointestinal defects when signs and symptoms of feeding intolerance, abdominal illness, or sepsis are present. The short- and long-term consequences of NEC are devastating in all infants, and although early disease recognition and treatment are essential, promoting human milk feeding as a primary modality in prevention is critical. This article highlights our current understanding of the pathophysiology, the clinical presentation, the risk factors for NEC in term infants compared with premature infants, and the treatment of NEC and discusses strategies in the prevention of NEC. Finally, we review the long-term consequences of NEC and the importance of primary care practitioners in the long-term care of infants after hospitalization for NEC.


Subject(s)
Enterocolitis, Necrotizing/diagnosis , Enterocolitis, Necrotizing/therapy , Infant Care/methods , Infant, Premature, Diseases/diagnosis , Infant, Premature, Diseases/therapy , Infant, Premature , Diagnosis, Differential , Enterocolitis, Necrotizing/etiology , Enterocolitis, Necrotizing/physiopathology , Humans , Infant, Newborn , Infant, Premature, Diseases/etiology , Infant, Premature, Diseases/physiopathology , Risk Factors , Treatment Outcome
6.
Breastfeed Med ; 13(6): 408-411, 2018.
Article in English | MEDLINE | ID: mdl-29877722

ABSTRACT

OBJECTIVE: Articles previously published by Sullivan et al. and Cristofalo et al. were reanalyzed using the proportion of cow milk-based nutrition received to determine whether that affected clinical outcomes during hospitalization for infants birth weight 500-1250 g. Abrams et al. showed in the same cohort incidences of necrotizing enterocolitis (NEC), NEC requiring surgery and sepsis increased proportionally to the amount of dietary cow milk. METHODS: The data from the two studies conducted under essentially the same protocol were combined yielding a cohort of 260 infants receiving a diet ranging from 0% to 100% cow milk. Data analysis utilized negative binomial regression which mitigates differences between subjects in terms of their time on study by incorporating that number into the statistical model. The percent of cow milk-based nutrition was the only predictor investigated. RESULTS: For all outcomes the larger the amount of cow's milk in the diet the greater the number of days of that intervention required. A trend toward statistical significance was seen for ventilator days; however, only parenteral nutrition (PN) days and days to full feeds achieved statistical significance. CONCLUSIONS: Incorporation of any cow milk-based nutrition into the diet of extremely premature infants correlates with more days on PN and a longer time to achieve full feeds. There was a nonstatistically significant trend toward increased ventilator days. These represent additional clinical consequences of the use of any cow milk-based protein in feeding EP infants.


Subject(s)
Breast Feeding , Enterocolitis, Necrotizing/prevention & control , Milk, Human , Parenteral Nutrition/adverse effects , Respiration, Artificial/statistics & numerical data , Animals , Birth Weight , Enteral Nutrition/adverse effects , Enterocolitis, Necrotizing/therapy , Female , Gestational Age , Humans , Infant Formula , Infant Nutritional Physiological Phenomena , Infant, Extremely Premature , Infant, Newborn , Infant, Very Low Birth Weight , Male , Milk , Oxygen Inhalation Therapy
7.
Pediatr Ann ; 45(11): e394-e398, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27841922

ABSTRACT

Incessant crying is one of the most common caregiver complaints during emergency department (ED) visits in the first few months of the child's life. Although the majority of cases are attributed to normal infant behavior, the differential diagnosis remains broad. Moreover, the potential for the negative impact of incessant crying on the mental well-being of caregivers as well as the infants necessitates that complaints be taken seriously and that "red flags" for underlying organic causes be ruled out and caregiver anxiety quelled. In addition, the apparent triviality of incessant crying in the face of the life-threatening illnesses or injuries that confront practitioners in the ED necessitates a high level of due diligence in the evaluation of these infants and their families. Ensuring the availability of family support is essential in the discharge planning. Families should also perceive the empathy of the physician and feel reassured about their safe discharge home. Although it is a challenge to examine an incessantly crying infant in all care settings, the failure to recognize the small percentage of infants that present with incessant crying as a manifestation of an underlying organic illness may have grave consequences. [Pediatr Ann. 2016;45(11):e394-e398.].


Subject(s)
Clinical Decision-Making/methods , Crying , Emergency Service, Hospital , Triage/methods , Diagnosis, Differential , Humans , Infant , Infant Behavior
8.
J Pediatr ; 175: 68-73.e3, 2016 08.
Article in English | MEDLINE | ID: mdl-27260839

ABSTRACT

OBJECTIVE: To evaluate the safety and explore the efficacy of recombinant human lactoferrin (talactoferrin [TLf]) to reduce infection. STUDY DESIGN: We conducted a randomized, double blind, placebo-controlled trial in infants with birth weight of 750-1500 g. Infants received enteral TLf (n = 60) or placebo (n = 60) on days 1 through 28 of life; the TLf dose was 150 mg/kg every 12 hours. Primary outcomes were bacteremia, pneumonia, urinary tract infection, meningitis, and necrotizing enterocolitis (NEC). Secondary outcomes were sepsis syndrome and suspected NEC. We recorded clinical, laboratory, and radiologic findings, along with diseases and adverse events, in a database used for statistical analyses. RESULTS: Demographic data were similar in the 2 groups of infants. We attributed no enteral or organ-specific adverse events to TLf. There were 2 deaths in the TLf group (1 each due to posterior fossa hemorrhage and postdischarge sudden infant death), and 1 death in the placebo group, due to NEC. The rate of hospital-acquired infections was 50% lower in the TLf group compared with the placebo group (P < .04), including fewer blood or line infections, urinary tract infections, and pneumonia. Fourteen infants in the TLf group weighing <1 kg at birth had no gram-negative infections, compared with only 3 of 14 such infants in the placebo group. Noninfectious outcomes were not statistically significantly different between the 2 groups, and there were no between-group differences in growth or neurodevelopment over a 1-year posthospitalization period. CONCLUSION: We found no clinical or laboratory toxicity and a trend toward less infectious morbidity in the infants treated with TLf. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00854633.


Subject(s)
Cross Infection/prevention & control , Gram-Negative Bacterial Infections/prevention & control , Gram-Positive Bacterial Infections/prevention & control , Infant, Premature, Diseases/prevention & control , Lactoferrin/therapeutic use , Protective Agents/therapeutic use , Administration, Oral , Bacteremia/prevention & control , Double-Blind Method , Enterocolitis, Necrotizing/prevention & control , Female , Follow-Up Studies , Humans , Infant, Newborn , Infant, Premature , Male , Meningitis/prevention & control , Pneumonia/prevention & control , Sepsis/prevention & control , Treatment Outcome , Urinary Tract Infections/prevention & control
9.
J Pediatr ; 173 Suppl: S37-42, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27234409

ABSTRACT

UNLABELLED: The purpose of this study is to evaluate the effects of enteral lactoferrin on the fecal microbiome and contrast those influences with the neonatal intensive care unit (NICU) environment. We theorized that lactoferrin and the NICU habitat shape the fecal microbial composition of very preterm infants. Although functions attributed to lactoferrin include intestinal immune system development and emergence of a healthy gut microbiota, evidence is limited. Twenty-one very low birth weight (VLBW <1500 g) infants received twice-daily talactoferrin (TLf, a drug designation for recombinant human lactoferrin) or its excipient by gastric gavage from day 1-28 of life. Twenty-four-hour fecal samples were collected on day 21 of life and compared with fecal operational taxonomy units (OTUs) in treated and control infants in 2 NICUs. Workflow included fecal DNA isolation, generation of amplicons for the V1-V3 region of bacterial 16S ribosomal RNA, and sequencing of a gel-purified multiplex amplicon library using a Roche 454 GS FLX Titanium (Roche, Branford, Connecticut) platform and protocols. Fecal OTUs per infant were higher in NICU 1 vs NICU 2 (P < .001), consistent with fewer antibiotic days (P < .02) and a shorter duration of parenteral nutrition (P < .007) in NICU 1. Proteobacteria and Firmicutes were the major phyla in infants treated with TLf and placebo. Among Enterobacteriaceae, TLf prophylaxis reduced Enterobacter and Klebsiella, but increased Citrobacter in feces of VLBW infants. Citrobacter caused no neonatal infections in the study population. OTUs for Clostridiaceae increased in NICU 1 among infants treated with TLf. Importantly, OTUs of staphylococci were barely detectable in both NICUs among infants fed TLf. Fewer hospital-acquired infections occurred in infants treated with TLf vs controls, although the reduction was seen mostly in coagulase-negative staphylococci-related bloodstream and central line infections (P = .06). TLf modified the fecal microbiome in VLBW infants, but care practices in the NICU habitat also contributed. Future research must establish whether elimination vs enrichment of gut-related microbiota reduces clinically significant hospital-acquired infections and promotes a healthy commensal microflora in the intestines of VLBW infants. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00854633.


Subject(s)
Anti-Infective Agents/pharmacology , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Infant, Premature , Infant, Very Low Birth Weight , Intensive Care, Neonatal/methods , Lactoferrin/pharmacology , Administration, Oral , Anti-Infective Agents/therapeutic use , Cross Infection/prevention & control , Double-Blind Method , Female , Follow-Up Studies , Humans , Infant Nutritional Physiological Phenomena , Infant, Newborn , Infant, Premature, Diseases/prevention & control , Intensive Care Units, Neonatal , Lactoferrin/therapeutic use , Male , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Treatment Outcome
10.
Pediatr Ann ; 44(5): e97-102, 2015 May.
Article in English | MEDLINE | ID: mdl-25996200

ABSTRACT

Herpes simplex virus (HSV) infection in the newborn carries a high mortality rate and can result in lifelong neurologic impairment. The severity of HSV infection in the newborn has always dictated conservative management when prodromal symptoms or active genital lesions (or those suggestive of genital herpes) are present during labor and delivery. The risk of intrapartum infection, however, is related to the presence or absence of maternal immunity (neutralizing antibody) to HSV. The most significant risk of transmission is in first-episode primary infections with active lesions at delivery. Recent recommendations from the American Academy of Pediatrics Committees on Infectious Diseases and the Fetus and Newborn use rapid serologic and virologic screening in the management of asymptomatic infants born to mothers with active genital herpes. The revised guidelines highlight infants at greatest risk for HSV disease but do not apply to asymptomatic infants born to mothers with a history of HSV but no genital lesions at delivery. The current guidelines also stipulate that maternal serologic screening and molecular assays for HSV in newborn blood and cerebrospinal fluid must be available and reported in a timely fashion.


Subject(s)
Herpes Genitalis/diagnosis , Infectious Disease Transmission, Vertical/prevention & control , Neonatal Screening/methods , Pathology, Molecular/methods , Simplexvirus/isolation & purification , Female , Herpes Genitalis/prevention & control , Humans , Infant, Newborn , Pregnancy , Pregnancy Complications, Infectious , Risk
11.
Case Rep Pediatr ; 2015: 584735, 2015.
Article in English | MEDLINE | ID: mdl-25866693

ABSTRACT

Congenital hypothyroidism (CH) is the most common endocrine disorder affecting the newborn. Universal newborn screening (NBS) has virtually eliminated the static encephalopathy and devastating neurodevelopmental syndrome known as cretinism. This report describes the presentation of an infant referred by the primary pediatrician to our hospital at 12 days of age for confirmatory testing after the NBS was consistent with CH. The infant had hypoglycemia secondary to lethargy and poor feeding and required transfer to the neonatal intensive care unit for worsening abdominal distension despite normalization of serum thyroid function tests following hormone replacement. In particular, the recalcitrant ileus and secondary bowel obstruction resulted in an additional diagnostic workup and lengthened hospital day. Our report highlights the acute gastrointestinal consequences of hypothyroidism despite evidence of effective treatment. We believe that the preclinical detection and immediate therapy for CH have lessened the prevalence of this presentation in general practice, and hence practitioners are less likely to be familiar with its natural history and management.

12.
Pediatr Res ; 77(1-2): 127-35, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25303278

ABSTRACT

The conceptual framework for a gut-brain axis has existed for decades. The Human Microbiome Project is responsible for establishing intestinal dysbiosis as a mediator of inflammatory bowel disease, obesity, and neurodevelopmental disorders in adults. Recent advances in metagenomics implicate gut microbiota and diet as key modulators of the bidirectional signaling pathways between the gut and brain that underlie neurodevelopmental and psychiatric disorders in adults. Evidence linking intestinal dysbiosis to neurodevelopmental disease outcomes in preterm infants is emerging. Recent clinical studies show that intestinal dysbiosis precedes late-onset neonatal sepsis and necrotizing enterocolitis in intensive care nurseries. Moreover, strong epidemiologic evidence links late-onset neonatal sepsis and necrotizing enterocolitis in long-term psychomotor disabilities of very-low-birth-weight infants. The notion of the gut-brain axis thereby supports that intestinal microbiota can indirectly harm the brain of preterm infants. In this review, we highlight the anatomy and physiology of the gut-brain axis and describe transmission of stress signals caused by immune-microbial dysfunction in the gut. These messengers initiate neurologic disease in preterm infants. Understanding neural and humoral signaling through the gut-brain axis will offer insight into therapeutic and dietary approaches that may improve the outcomes of very-low-birth-weight infants.


Subject(s)
Brain/physiology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiology , Infant, Newborn, Diseases/microbiology , Microbiota/physiology , Models, Biological , Signal Transduction/physiology , Dysbiosis/microbiology , Enterocolitis, Necrotizing/microbiology , Humans , Infant, Newborn , Infant, Very Low Birth Weight , Sepsis/microbiology
13.
Curr Opin Pediatr ; 26(2): 146-50, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24503532

ABSTRACT

PURPOSE OF REVIEW: There is an intense interest among neonatal caregivers as to whether lactoferrin given enterally may reduce the incidence of necrotizing enterocolitis in preterm infants. This review presents scientific and clinical evidence that lactoferrin alleviates or prevents this life-threatening disease. RECENT FINDINGS: Preclinical studies in neonatal rats showed that lactoferrin given orally before enteral infection with pathogenic Escherichia coli reduced bacteremia and mortality. A multicentered clinical trial found that very low-birth weight preterm infants given bovine lactoferrin had a significant reduction in late-onset sepsis; there was also a trend towards a diminished incidence of necrotizing enterocolitis. Although multicentered trials of lactoferrin use in preterm infants are near completion, regulatory burdens required to bring lactoferrin to the bedside may limit its availability. SUMMARY: Extremely preterm infants should receive colostrum, a natural lactoferrin concentrate, immediately after birth and, ideally, continue on breast milk throughout the hospital stay. This practice appears well tolerated, but additional experience will tell us whether this practice reduces the prevalence of necrotizing enterocolitis.


Subject(s)
Colostrum , Enterocolitis, Necrotizing/prevention & control , Gastrointestinal Tract/immunology , Inflammation/immunology , Lactoferrin/therapeutic use , Sepsis/prevention & control , Animals , Enteral Nutrition , Enterocolitis, Necrotizing/immunology , Humans , Immunity, Innate , Incidence , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/prevention & control , Infant, Very Low Birth Weight , Lactoferrin/administration & dosage , Lactoferrin/immunology , Randomized Controlled Trials as Topic , Sepsis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...