1
|
Hetzroni A, Yosef PG, Hedar M, Ishay RTB, Sharon N, Rubinstein U. Length of hospital stay not affected by empirical treatment with ceftriaxone versus cefuroxime for bacteraemia. Acta Paediatr 2025. [PMID: 39754407 DOI: 10.1111/apa.17572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 12/10/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
AIM The preferred antibiotic treatment for bacteraemia in infants continues to be debated. We examined the duration of hospital stays as a surrogate for the effectiveness of initial treatment with ceftriaxone versus cefuroxime. METHODS This was a retrospective review of the medical records of all infants aged 3-36 months, admitted with suspected occult bacteraemia to the paediatric department at Laniado Hospital, Israel, between 2016 and 2022. The effect of antibiotic treatment, namely ceftriaxone versus cefuroxime, on hospital stays was determined, in both the total study population and population subgroups. RESULTS We identified 217 patients (59.0% male) with a median age of 13 months and 12.4% had positive blood cultures. Approximately three-quarters (75.6%) received cefuroxime as their initial treatment for bacteraemia and the other quarter (24.4%) received ceftriaxone. The median length of hospital stay was 3.0 (interquartile range 3.0-4.0), with no statistically significant difference between the two drugs. However, we did notice a statistically significant shorter median length of hospital stay among fully vaccinated infants treated with cefuroxime rather than ceftriaxone (p = 0.055). CONCLUSION The length of hospital stay among infants diagnosed with bacteraemia was not affected by whether they initially received ceftriaxone or cefuroxime. Further studies in larger populations are needed.
Collapse
Affiliation(s)
| | | | - Muna Hedar
- Pediatric Department, Laniado Hospital, Netanya, Israel
| | | | | | | |
Collapse
|
2
|
Li Q, Guan Y, Xia C, Wu L, Zhang H, Wang Y. Physiologically-Based Pharmacokinetic Modeling and Dosing Optimization of Cefotaxime in Preterm and Term Neonates. J Pharm Sci 2024; 113:2605-2615. [PMID: 38460573 DOI: 10.1016/j.xphs.2024.03.002] [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: 11/02/2023] [Revised: 03/02/2024] [Accepted: 03/02/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND Cefotaxime is commonly used in treating bacterial infections in neonates. To characterize the pharmacokinetic process in neonates and evaluate different recommended dosing schedules of cefotaxime, a physiologically-based pharmacokinetic (PBPK) model of cefotaxime was established in adults and scaled to neonates. METHODS A whole-body PBPK model was built in PK-SIM® software. Three elimination pathways are composed of enzymatic metabolism in the liver, passive filtration through glomerulus, and active tubular secretion mediated by renal transporters. The ontogeny information was applied to account for age-related changes in cefotaxime pharmacokinetics. The established models were verified with realistic clinical data in adults and pediatric populations. Simulations in neonates were conducted and 100 % of the dosing interval where the unbound concentration in plasma was above the minimum inhibitory concentration (fT>MIC) was selected as the target index for dosing regimen evaluation. RESULTS The developed PBPK models successfully described the pharmacokinetic process of cefotaxime in adults and were scaled to the pediatric population. Good verification results were achieved in both adults' and neonates' PBPK models, indicating a good predictive performance. The optimal dosage regimen of cefotaxime was proposed according to the postnatal age (PNA) and gestational age (GA) of neonates. For preterm neonates (GA < 36 weeks), dosages of 25 mg/kg every 8 h in PNA 0-6 days and 25 mg/kg every 6 h in PNA 7-28 days were suggested. For term neonates (GA ≥ 36 weeks), dosages of 33 mg/kg every 8 h in PNA 0-6 days and 33 mg/kg every 6 h in PNA 7-28 days were recommended. CONCLUSIONS Our study may provide useful experience in practicing PBPK model-informed precision dosing in the pediatric population.
Collapse
Affiliation(s)
- Qiaoxi Li
- Department of pharmacy, the first people's hospital of Foshan, Foshan, China
| | - Yanping Guan
- Institute of clinical pharmacology, school of pharmaceutical sciences, Sun Yat-sen University, Guangzhou, China
| | - Chen Xia
- Department of pharmacy, the first people's hospital of Foshan, Foshan, China
| | - Lili Wu
- Department of pharmacy, the first people's hospital of Foshan, Foshan, China
| | - Hongyu Zhang
- Department of pharmacy, the first people's hospital of Foshan, Foshan, China
| | - Yan Wang
- Department of pharmacy, the first people's hospital of Foshan, Foshan, China.
| |
Collapse
|
3
|
Knowledge gaps in late-onset neonatal sepsis in preterm neonates: a roadmap for future research. Pediatr Res 2022; 91:368-379. [PMID: 34497356 DOI: 10.1038/s41390-021-01721-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022]
Abstract
Late-onset neonatal sepsis (LONS) remains an important threat to the health of preterm neonates in the neonatal intensive care unit. Strategies to optimize care for preterm neonates with LONS are likely to improve survival and long-term neurocognitive outcomes. However, many important questions on how to improve the prevention, early detection, and therapy for LONS in preterm neonates remain unanswered. This review identifies important knowledge gaps in the management of LONS and describe possible methods and technologies that can be used to resolve these knowledge gaps. The availability of computational medicine and hypothesis-free-omics approaches give way to building bedside feedback tools to guide clinicians in personalized management of LONS. Despite advances in technology, implementation in clinical practice is largely lacking although such tools would help clinicians to optimize many aspects of the management of LONS. We outline which steps are needed to get possible research findings implemented on the neonatal intensive care unit and provide a roadmap for future research initiatives. IMPACT: This review identifies knowledge gaps in prevention, early detection, antibiotic, and additional therapy of late-onset neonatal sepsis in preterm neonates and provides a roadmap for future research efforts. Research opportunities are addressed, which could provide the means to fill knowledge gaps and the steps that need to be made before possible clinical use. Methods to personalize medicine and technologies feasible for bedside clinical use are described.
Collapse
|
4
|
Schroepf S, Burau D, Muench HG, Derendorf H, Zeitlinger M, Genzel-Boroviczény O, Adam D, Kloft C. Microdialysis sampling to monitor target-site vancomycin concentrations in septic infants: a feasible way to close the knowledge gap. Int J Antimicrob Agents 2021; 58:106405. [PMID: 34289402 DOI: 10.1016/j.ijantimicag.2021.106405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/24/2021] [Accepted: 07/03/2021] [Indexed: 10/20/2022]
Abstract
This work is dedicated to the memory of Hartmut Derendorf (1953-2020), a pioneer of modern pharmacokinetics and valued mentor of this project. OBJECTIVES Septic infants/neonates need effective antibiotic exposure, but dosing recommendations are challenging as the pharmacokinetics in this age are highly variable. For vancomycin, which is used as a standard treatment, comprehensive pharmacokinetic knowledge especially at the infection site is lacking. Hence, an exploratory clinical study was conducted to assess the feasibility and safety of microdialysis sampling for vancomycin monitoring at the target site. METHODS Nine infants/neonates with therapeutic indications for vancomycin treatment were administered 15 mg/kg as 1-hour infusions every 8-24 hours. Microdialysis catheters were implanted in the subcutaneous interstitial space fluid of the lateral thigh. Samples were collected every 30 minutes over 24 hours, followed by retrodialysis for catheter calibration. Prior in vitro investigations have evaluated impact factors on relative recovery and retrodialysis. RESULTS In vitro investigations showed the applicability of microdialysis for vancomycin monitoring. Microdialysis sampling was well tolerated in all infants/neonates (23-255 days) without major bleeding or other adverse events. Pharmacokinetic profiles were obtained and showed plausible vancomycin concentration-time courses. CONCLUSIONS Microdialysis as a minimally invasive technique for continuous longer-term sampling is feasible and safe in infants/neonates. Interstitial space fluid profiles were plausible and showed substantial interpatient variation. Hence, a larger microdialysis trial is warranted to further characterise the pharmacokinetics and variability of vancomycin at the target site and ultimately improve vancomycin dosing in these vulnerable patients.
Collapse
Affiliation(s)
- Sebastian Schroepf
- Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Muenchen, Germany.
| | - Daniela Burau
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany
| | - Hans-Georg Muench
- Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University Munich, Muenchen, Germany
| | - Hartmut Derendorf
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Wien, Austria
| | | | | | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany
| |
Collapse
|
5
|
Korang SK, Safi S, Nava C, Greisen G, Gupta M, Lausten-Thomsen U, Jakobsen JC. Antibiotic regimens for late-onset neonatal sepsis. Cochrane Database Syst Rev 2021; 5:CD013836. [PMID: 33998665 PMCID: PMC8127057 DOI: 10.1002/14651858.cd013836.pub2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Neonatal sepsis is a major cause of morbidity and mortality. It is the third leading cause of neonatal mortality globally constituting 13% of overall neonatal mortality. Despite the high burden of neonatal sepsis, high-quality evidence in diagnosis and treatment is scarce. Due to the diagnostic challenges of sepsis and the relative immunosuppression of the newborn, many neonates receive antibiotics for suspected sepsis. Antibiotics have become the most used therapeutics in neonatal intensive care units, and observational studies in high-income countries suggest that 83% to 94% of newborns treated with antibiotics for suspected sepsis have negative blood cultures. The last Cochrane Review was updated in 2005. There is a need for an updated systematic review assessing the effects of different antibiotic regimens for late-onset neonatal sepsis. OBJECTIVES To assess the beneficial and harmful effects of different antibiotic regimens for late-onset neonatal sepsis. SEARCH METHODS We searched the following electronic databases: CENTRAL (2021, Issue 3); Ovid MEDLINE; Embase Ovid; CINAHL; LILACS; Science Citation Index EXPANDED and Conference Proceedings Citation Index - Science on 12 March 2021. We also searched clinical trials databases and the reference lists of retrieved articles for randomised controlled trials (RCTs) and quasi-RCTs. SELECTION CRITERIA We included RCTs comparing different antibiotic regimens for late-onset neonatal sepsis. We included participants older than 72 hours of life at randomisation, suspected or diagnosed with neonatal sepsis, meningitis, osteomyelitis, endocarditis, or necrotising enterocolitis. We excluded trials that assessed treatment of fungal infections. DATA COLLECTION AND ANALYSIS Three review authors independently assessed studies for inclusion, extracted data, and assessed risk of bias. We used the GRADE approach to assess the certainty of evidence. Our primary outcome was all-cause mortality, and our secondary outcomes were: serious adverse events, respiratory support, circulatory support, nephrotoxicity, neurological developmental impairment, necrotising enterocolitis, and ototoxicity. Our primary time point of interest was at maximum follow-up. MAIN RESULTS We included five RCTs (580 participants). All trials were at high risk of bias, and had very low-certainty evidence. The five included trials assessed five different comparisons of antibiotics. We did not conduct a meta-analysis due to lack of relevant data. Of the five included trials one trial compared cefazolin plus amikacin with vancomycin plus amikacin; one trial compared ticarcillin plus clavulanic acid with flucloxacillin plus gentamicin; one trial compared cloxacillin plus amikacin with cefotaxime plus gentamicin; one trial compared meropenem with standard care (ampicillin plus gentamicin or cefotaxime plus gentamicin); and one trial compared vancomycin plus gentamicin with vancomycin plus aztreonam. None of the five comparisons found any evidence of a difference when assessing all-cause mortality, serious adverse events, circulatory support, nephrotoxicity, neurological developmental impairment, or necrotising enterocolitis; however, none of the trials were near an information size that could contribute significantly to the evidence of the comparative benefits and risks of any particular antibiotic regimen. None of the trials assessed respiratory support or ototoxicity. The benefits and harms of different antibiotic regimens remain unclear due to the lack of well-powered trials and the high risk of systematic errors. AUTHORS' CONCLUSIONS Current evidence is insufficient to support any antibiotic regimen being superior to another. RCTs assessing different antibiotic regimens in late-onset neonatal sepsis with low risks of bias are warranted.
Collapse
Affiliation(s)
- Steven Kwasi Korang
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sanam Safi
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Chiara Nava
- Neonatal Intensive Care Unit, Ospedale "A. Manzoni", Lecco, Italy
| | - Gorm Greisen
- Department of Neonatology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Munish Gupta
- Neonatology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Ulrik Lausten-Thomsen
- Pediatric and Neonatal Intensive Care Unit, Paris South University Hospitals Le Kremlin-Bicêtre, Paris, France
| | - Janus C Jakobsen
- Cochrane Hepato-Biliary Group, Copenhagen Trial Unit, Centre for Clinical Intervention Research, The Capital Region of Denmark, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
6
|
D'Agate S, Musuamba FT, Jacqz-Aigrain E, Della Pasqua O. Simplified Dosing Regimens for Gentamicin in Neonatal Sepsis. Front Pharmacol 2021; 12:624662. [PMID: 33762945 PMCID: PMC7982486 DOI: 10.3389/fphar.2021.624662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/04/2021] [Indexed: 11/30/2022] Open
Abstract
Background: The effectiveness of antibiotics for the treatment of severe bacterial infections in newborns in resource-limited settings has been determined by empirical evidence. However, such an approach does not warrant optimal exposure to antibiotic agents, which are known to show different disposition characteristics in this population. Here we evaluate the rationale for a simplified regimen of gentamicin taking into account the effect of body size and organ maturation on pharmacokinetics. The analysis is supported by efficacy data from a series of clinical trials in this population. Methods: A previously published pharmacokinetic model was used to simulate gentamicin concentration vs. time profiles in a virtual cohort of neonates. Model predictive performance was assessed by supplementary external validation procedures using therapeutic drug monitoring data collected in neonates and young infants with or without sepsis. Subsequently, clinical trial simulations were performed to characterize the exposure to intra-muscular gentamicin after a q.d. regimen. The selection of a simplified regimen was based on peak and trough drug levels during the course of treatment. Results: In contrast to current World Health Organization guidelines, which recommend gentamicin doses between 5 and 7.5 mg/kg, our analysis shows that gentamicin can be used as a fixed dose regimen according to three weight-bands: 10 mg for patients with body weight <2.5 kg, 16 mg for patients with body weight between 2.5 and 4 kg, and 30 mg for those with body weight >4 kg. Conclusion: The choice of the dose of an antibiotic must be supported by a strong scientific rationale, taking into account the differences in drug disposition in the target patient population. Our analysis reveals that a simplified regimen is feasible and could be used in resource-limited settings for the treatment of sepsis in neonates and young infants with sepsis aged 0–59 days.
Collapse
Affiliation(s)
- S D'Agate
- Clinical Pharmacology and Therapeutics Group, University College London, London, United Kingdom
| | - F Tshinanu Musuamba
- Clinical Pharmacology and Therapeutics Group, University College London, London, United Kingdom
| | - E Jacqz-Aigrain
- Department of Paediatric Pharmacology and Pharmacogenetics, Centre Hospitalier Universitaire, Hôpital Robert Debré, Paris, France
| | - O Della Pasqua
- Clinical Pharmacology and Therapeutics Group, University College London, London, United Kingdom
| |
Collapse
|
7
|
Abstract
BACKGROUND Early-onset sepsis, occurring within 72 hours of birth, and late-onset sepsis, occurring after this time period, present serious risks for neonates. While culture-based screening and intrapartum antibiotics have decreased the number of early-onset cases, sepsis remains a top cause of neonatal morbidity and mortality in the United States. PURPOSE To provide a review of neonatal sepsis by identifying its associated risk factors and most common causative pathogens, reviewing features of the term and preterm neonatal immune systems that increase vulnerability to infection, describing previous and the most current management recommendations, and discussing relevant implications for the neonatal nurse and novice neonatal nurse practitioner. METHODS/SEARCH STRATEGY An integrative review of literature was conducted using key words in CINAHL, Google Scholar, and PubMed. FINDINGS/RESULTS Group B streptococcus and Escherichia coli are the most common pathogens in early-onset sepsis, while Coagulase-negative staphylococci comprise the majority of cases in late-onset. The neonatal immune system is vulnerable due to characteristics including decreased cellular activity, underdeveloped complement systems, preferential anti-inflammatory responses, and insufficient pathogenic memory. Blood cultures remain the criterion standard of diagnosis, with several other adjunct tests under investigation for clinical use. The recent development of the sepsis calculator has been a useful tool in the management of early-onset cases. IMPLICATIONS FOR PRACTICE It is vital to understand the mechanisms behind the neonate's elevated risk for infection and to implement evidence-based management. IMPLICATIONS FOR RESEARCH Research needs exist for diagnostic methods that deliver timely and sensitive results. A tool similar to the sepsis calculator does not exist for preterm infants or late-onset sepsis, groups for which antibiotic stewardship is not as well practiced.Video Abstract available athttps://journals.lww.com/advancesinneonatalcare/Pages/videogallery.aspx?autoPlay=false&videoId=40.
Collapse
|
8
|
Korang SK, Safi S, Gupta M, Greisen G, Lausten-Thomsen U, Jakobsen JC. Antibiotic regimens for late-onset neonatal sepsis. Hippokratia 2021. [DOI: 10.1002/14651858.cd013836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Steven Kwasi Korang
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812; Rigshospitalet, Copenhagen University Hospital; Copenhagen Denmark
- Pediatric Department; Holbaek Sygehus; Holbaek Denmark
| | - Sanam Safi
- Copenhagen Trial Unit, Centre for Clinical Intervention Research; Department 7812, Rigshospitalet, Copenhagen University Hospital; Copenhagen Denmark
| | - Munish Gupta
- Neonatology; Beth Israel Deaconess Medical Center; Boston USA
| | - Gorm Greisen
- Department of Neonatology; Rigshospitalet, Copenhagen University Hospital; Copenhagen Denmark
| | - Ulrik Lausten-Thomsen
- Pediatric and Neonatal Intensive Care Unit; Paris South University Hospitals Le Kremlin-Bicêtre; Paris France
| | - Janus C Jakobsen
- Cochrane Hepato-Biliary Group; Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Rigshospitalet; Copenhagen Denmark
- Department of Cardiology; Holbaek Hospital; Holbaek Denmark
- Department of Regional Health Research, the Faculty of Health Sciences; University of Southern Denmark; Holbaek Denmark
| |
Collapse
|
9
|
Effects of inflammation on the developing respiratory system: Focus on hypoglossal (XII) neuron morphology, brainstem neurochemistry, and control of breathing. Respir Physiol Neurobiol 2020; 275:103389. [PMID: 31958568 DOI: 10.1016/j.resp.2020.103389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 01/03/2020] [Accepted: 01/16/2020] [Indexed: 12/20/2022]
Abstract
Breathing is fundamental to life and any adverse change in respiratory function can endanger the health of an organism or even be fatal. Perinatal inflammation is known to adversely affect breathing in preterm babies, but lung infection/inflammation impacts all stages of life from birth to death. Little is known about the role of inflammation in respiratory control, neuronal morphology, or neural function during development. Animal models of inflammation can provide understanding and insight into respiratory development and how inflammatory processes alter developmental phenotype in addition to providing insight into new treatment modalities. In this review, we focus on recent work concerning the development of neurons, models of perinatal inflammation with an emphasis on two common LPS-based models, inflammation and its impact on development, and current and potential treatments for inflammation within the respiratory control circuitry of the mammalian brainstem. We have also discussed models of inflammation in adults and have specifically focused on hypoglossal motoneurons (XII) and neurons of the nucleus tractus solitarii (nTS) as these nuclei have been studied more extensively than other brainstem nuclei participating in breathing and airway control. Understanding the impact of inflammation on the developmental aspects of respiratory control and breathing pattern is critical to addressing problems of cardiorespiratory dysregulation in disease and this overview points out many gaps in our current knowledge.
Collapse
|
10
|
Korang SK, Safi S, Gluud C, Lausten-Thomsen U, Jakobsen JC. Antibiotic regimens for neonatal sepsis - a protocol for a systematic review with meta-analysis. Syst Rev 2019; 8:306. [PMID: 31805993 PMCID: PMC6896287 DOI: 10.1186/s13643-019-1207-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/22/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Sepsis is a major cause of morbidity and mortality among neonates and infants. Antibiotics are a central part of the first line treatment for sepsis in neonatal intensive care units worldwide. However, the evidence on the clinical effects of the commonly used antibiotic regimens for sepsis in neonates remains scarce. This systematic review aims to assess the efficacy and harms of antibiotic regimens for neonatal sepsis. METHODS Electronic searches will be conducted in MEDLINE, Embase, The Cochrane Library, CINAHL, ZETOC and clinical trial registries (clinicaltrials.gov and ISRCTN). We will include randomised controlled trials of different antibiotic regimens for sepsis of neonates and infants. Eligible interventions will be any antibiotic regimen. Two reviewers will independently screen, select, and extract data. The methodological quality of individual studies will be appraised following Cochrane methodology. Primary outcomes will be 'all-cause mortality' and 'serious adverse events'. Secondary outcomes will be 'need for respiratory support', 'need for circulatory support', 'neurodevelopmental impairment', ototoxicity, nephrotoxicity and necrotizing enterocolitis. We plan to perform a meta-analysis with trial sequential analysis. DISCUSSION This is the study protocol for a systematic review on the effects of different antibiotic regimens for neonatal sepsis. The results of this systematic review intent to adequately inform stakeholders or health care professionals in the field of neonatal sepsis, and to aid appropriate development of treatment guidelines. SYSTEMATIC REVIEW REGISTRATION PROSPERO reference number: CRD42019134300.
Collapse
Affiliation(s)
- Steven Kwasi Korang
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Sanam Safi
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christian Gluud
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ulrik Lausten-Thomsen
- Department of Neonatology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Janus C Jakobsen
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Regional Health Research, The Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Cardiology, Holbæk Hospital, Holbæk, Denmark
| |
Collapse
|
11
|
Extracorporeal membrane oxygenation and bloodstream infection in congenital diaphragmatic hernia. J Perinatol 2019; 39:1384-1391. [PMID: 31383944 DOI: 10.1038/s41372-019-0435-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/22/2019] [Accepted: 06/03/2019] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To characterize the risk of bloodstream (BSI) and urinary tract infection (UTI) and describe antibiotic use in infants with congenital diaphragmatic hernia (CDH) requiring extracorporeal membrane oxygenation (ECMO). STUDY DESIGN The Children's Hospitals Neonatal Database was queried for infants with CDH and ECMO treatment from 2010 to 2016. The outcomes included BSI, UTI, and antimicrobial medication. Member institutions completed a survey on infection practices. RESULT Eighteen of the 338 patients identified (5.3%) had ≥1 BSI during their ECMO course. The likelihood of BSI increased with time: 1.2/1000 ECMO days; 0.6% (2/315) in the first week and rising to 14.6/1000; 8.6% (5/58) after 21 days (p = 0.002). More than 95% of patients received antibiotics each week on ECMO. CONCLUSIONS Confirmed BSI is rare in infants with CDH treated with ECMO in the first week, but increases with the duration of ECMO. Use of antibiotics was extensive and did not correspond to infection frequency.
Collapse
|
12
|
Penetration of Cefotaxime into Cerebrospinal Fluid in Neonates and Young Infants. Antimicrob Agents Chemother 2018; 62:AAC.02448-17. [PMID: 29437625 DOI: 10.1128/aac.02448-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/31/2018] [Indexed: 01/11/2023] Open
Abstract
Cefotaxime is the first-line treatment for meningitis in neonates and young infants. However, limited data on cefotaxime cerebrospinal fluid (CSF) concentrations in neonates and young infants were available. The aim of the present study was to evaluate the penetration of cefotaxime into CSF in neonates and young infants. Blood and CSF samples were collected from neonates and young infants treated with cefotaxime using an opportunistic pharmacokinetic sampling strategy, and concentrations were quantified by high-performance liquid chromatography-tandem mass spectrometry. The analysis was performed using NONMEM and R software. Thirty neonates and young infants (postmenstrual age range, 25.4 to 47.4 weeks) were included. A total of 67 plasma samples and 30 CSF samples were available for analysis. Cefotaxime plasma and CSF concentrations ranged from 2.30 to 175.42 mg/liter and from 0.39 to 25.38 mg/liter, respectively. The median ratio of the CSF concentration to the plasma concentration was 0.28 (range, 0.06 to 0.76). Monte Carlo simulation demonstrated that 88.4% and 63.9% of hypothetical neonates treated with 50 mg/kg of body weight three times a day (TID) would reach the pharmacodynamic target (the percentage of the dosing interval that the free antimicrobial drug concentration remains above the MIC, 70%) using the standard EUCAST MIC susceptibility breakpoints of 2 mg/liter and 4 mg/liter, respectively. The penetration of cefotaxime into the CSF of neonates and young infants was evaluated using an opportunistic sampling approach. A dosage regimen of 50 mg/kg TID could cover the most causative pathogens with MICs of <2 mg/liter. Individual dosage adaptation was required for more resistant bacterial strains, such as Staphylococcus aureus.
Collapse
|
13
|
Li W, Luo S, Zhu Y, Wen Y, Shu M, Wan C. C-reactive protein concentrations can help to determine which febrile infants under three months should receive blood cultures during influenza seasons. Acta Paediatr 2017; 106:2017-2024. [PMID: 28799220 DOI: 10.1111/apa.14022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/03/2017] [Accepted: 08/08/2017] [Indexed: 11/26/2022]
Abstract
AIM We explored whether C-reactive protein (CRP) concentrations could indicate which infants with fever without source (FWS) should receive undergo blood culture tests during influenza seasons. METHODS This retrospective study focused on patients under three months of age with FWS who had received blood culture tests at the West China Second University Hospital Paediatric Emergency Department during the influenza seasons from June 2013 to January 2015. The statistical analysis comprised specificity, sensitivity, multilevel likelihood ratios (LRs), receiver operating characteristic analysis and a multivariate logistic regression model. RESULTS We enrolled 592 febrile patients and 7.1% had bacteraemia, with levels falling with increasing age. According to the receiver operating characteristic analysis, the optimum threshold of CRP was 30.5 mg/L, and when the CRP level was higher than 30.5 mg/L, the positive LR of bacteraemia was 2.32. In patients aged 29-90 days, when the CRP level was higher than 5 mg/L, the negative LR of bacteraemia was 0.38. In the neonatal group, a CRP level of ≥30.5 mg/L had a positive LR of bacteraemia of 3.55. CONCLUSION We found that CRP concentrations could indicate which febrile children under three months of age should undergo blood culture tests during influenza seasons.
Collapse
Affiliation(s)
- Weiran Li
- Department of Paediatrics; West China Second Hospital; Sichuan University; Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University); Ministry of Education; Chengdu China
| | - Shuanghong Luo
- Department of Paediatrics; West China Second Hospital; Sichuan University; Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University); Ministry of Education; Chengdu China
| | - Yu Zhu
- Department of Paediatrics; West China Second Hospital; Sichuan University; Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University); Ministry of Education; Chengdu China
| | - Yang Wen
- Department of Paediatrics; West China Second Hospital; Sichuan University; Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University); Ministry of Education; Chengdu China
| | - Min Shu
- Department of Paediatrics; West China Second Hospital; Sichuan University; Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University); Ministry of Education; Chengdu China
| | - Chaomin Wan
- Department of Paediatrics; West China Second Hospital; Sichuan University; Chengdu China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University); Ministry of Education; Chengdu China
| |
Collapse
|
14
|
Job KM, Olson J, Stockmann C, Constance JE, Enioutina EY, Rower JE, Linakis MW, Balch AH, Yu T, Liu X, Thorell EA, Sherwin CMT. Pharmacodynamic studies of voriconazole: informing the clinical management of invasive fungal infections. Expert Rev Anti Infect Ther 2017; 14:731-46. [PMID: 27355512 DOI: 10.1080/14787210.2016.1207526] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Voriconazole is a broad-spectrum antifungal agent commonly used to treat invasive fungal infections (IFI), including aspergillosis, candidiasis, Scedosporium infection, and Fusarium infection. IFI often occur in immunocompromised patients, leading to increased morbidity and mortality. AREAS COVERED The objective of this review is to summarize the pharmacodynamic properties of voriconazole and to provide considerations for potential optimal dosing strategies. Studies have demonstrated superior clinical response when an AUC/MIC >25 or Cmin/MIC >1 is attained in adult patients, correlating to a trough concentration range as narrow as 2-4.5 mg/L; however, these targets are poorly established in the pediatric population. Topics in this discussion include voriconazole use in multiple age groups, predisposing patient factors for IFI, and considerations for clinicians managing IFI. Expert commentary: The relationship between voriconazole dosing and exposure is not well defined due to the large inter- and intra-subject variability. Development of comprehensive decision support tools for individualizing dosing, particularly in children who require higher dosing, will help to increase the probability of achieving therapeutic efficacy and decrease sub-therapeutic dosing and adverse events.
Collapse
Affiliation(s)
- Kathleen M Job
- a Division of Clinical Pharmacology , University of Utah , Salt Lake City , UT , USA
| | - Jared Olson
- b Pharmacy, Primary Children's Hospital, Intermountain Healthcare , University of Utah , Salt Lake City , UT , USA
| | - Chris Stockmann
- c Division of Pediatric Infectious Diseases, Department of Pediatrics , University of Utah , Salt Lake City , UT , USA
| | - Jonathan E Constance
- a Division of Clinical Pharmacology , University of Utah , Salt Lake City , UT , USA
| | - Elena Y Enioutina
- a Division of Clinical Pharmacology , University of Utah , Salt Lake City , UT , USA.,d Division of Microbiology and Immunology, Department of Pathology , University of Utah , Salt Lake City , UT , USA
| | - Joseph E Rower
- a Division of Clinical Pharmacology , University of Utah , Salt Lake City , UT , USA
| | - Matthew W Linakis
- a Division of Clinical Pharmacology , University of Utah , Salt Lake City , UT , USA
| | - Alfred H Balch
- a Division of Clinical Pharmacology , University of Utah , Salt Lake City , UT , USA
| | - Tian Yu
- a Division of Clinical Pharmacology , University of Utah , Salt Lake City , UT , USA
| | - Xiaoxi Liu
- a Division of Clinical Pharmacology , University of Utah , Salt Lake City , UT , USA
| | - Emily A Thorell
- c Division of Pediatric Infectious Diseases, Department of Pediatrics , University of Utah , Salt Lake City , UT , USA
| | - Catherine M T Sherwin
- a Division of Clinical Pharmacology , University of Utah , Salt Lake City , UT , USA.,e Department of Pharmacology and Toxicology, College of Pharmacy , University of Utah , Salt Lake City , UT , USA
| |
Collapse
|
15
|
A Population and Developmental Pharmacokinetic Analysis To Evaluate and Optimize Cefotaxime Dosing Regimen in Neonates and Young Infants. Antimicrob Agents Chemother 2016; 60:6626-6634. [PMID: 27572399 DOI: 10.1128/aac.01045-16] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 08/14/2016] [Indexed: 01/05/2023] Open
Abstract
Cefotaxime is one of the most frequently prescribed antibiotics for the treatment of Gram-negative bacterial sepsis in neonates. However, the dosing regimens routinely used in clinical practice vary considerably. The objective of the present study was to conduct a population pharmacokinetic study of cefotaxime in neonates and young infants in order to evaluate and optimize the dosing regimen. An opportunistic sampling strategy combined with population pharmacokinetic analysis using NONMEM software was performed. Cefotaxime concentrations were measured by high-performance liquid chromatography-tandem mass spectrometry. Developmental pharmacokinetics-pharmacodynamics, the microbiological pathogens, and safety aspects were taken into account to optimize the dose. The pharmacokinetic data from 100 neonates (gestational age [GA] range, 23 to 42 weeks) were modeled with an allometric two-compartment model with first-order elimination. The median values for clearance and the volume of distribution at steady state were 0.12 liter/h/kg of body weight and 0.64 liter/kg, respectively. The covariate analysis showed that current weight, GA, and postnatal age (PNA) had significant impacts on cefotaxime pharmacokinetics. Monte Carlo simulations demonstrated that the current dose recommendations underdosed older newborns. A model-based dosing regimen of 50 mg/kg twice a day to four times a day, according to GA and PNA, was established. The associated risk of overdose for the proposed dosing regimen was 0.01%. We determined the population pharmacokinetics of cefotaxime and established a model-based dosing regimen to optimize treatment for neonates and young infants.
Collapse
|
16
|
Evaluation of Vancomycin Use in Late-Onset Neonatal Sepsis Using the Area Under the Concentration-Time Curve to the Minimum Inhibitory Concentration ≥400 Target. Ther Drug Monit 2016; 37:756-65. [PMID: 26562817 DOI: 10.1097/ftd.0000000000000216] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AIM To develop a vancomycin population pharmacokinetic model and assess the probability of attaining a pharmacodynamic target associated with clinical and microbiological success, a ratio of the 24-hour area under the concentration-time curve to the minimum inhibitory concentration (MIC) ≥ 400, in a 5-year clinical cohort of preterm and term neonatal patients with late-onset staphylococcal sepsis. METHODS Therapeutic drug monitoring data were obtained from septic neonates with ≥1 vancomycin concentration(s) from January 2006 to September 2011. Only neonates with a postnatal age of >72 hours and a positive microbiological culture were included. Population pharmacokinetic model was developed using nonlinear mixed effects modeling (NONMEM 7.2). Eleven demographic characteristics were evaluated as covariates. Probabilities of achieving the pharmacodynamic target were evaluated. RESULTS A 1-compartment model with first-order elimination was constructed from 528 vancomycin concentrations collected from 152 preterm and term neonates. Body weight, creatinine clearance (CL), and postmenstrual age were identified as significant covariates. Estimated vancomycin CL and volume of distribution for typical neonates were 0.068 ± 0.03 L·h·kg and 0.62 ± 0.13 L/kg, respectively. Coagulase-negative staphylococci (85.5%) and Staphylococcus aureus (14.5%) were the common pathogenic organisms. The distribution of vancomycin MIC breakpoints was composed of approximately 70% MIC breakpoint of ≤2 mcg/mL. Approximately 54% of neonates, with a median serum creatinine concentration of 0.44 mg/dL, achieved the target ratio of 24-hour area under the concentration-time curve to the MIC ≥ 400 with a median daily dose of 30 (interquartile range, 21-42) mg/kg. CONCLUSIONS Body weight, creatinine CL, and postmenstrual age significantly influenced vancomycin CL. The current vancomycin doses are acceptable at MICs ≤1 mcg/mL because they are likely to achieve the pharmacodynamic target in the majority of neonatal patients, although higher doses may be considered for more resistant staphylococcal infections.
Collapse
|
17
|
Antimicrobial Agent Dosing in Infants. Clin Ther 2016; 38:1948-60. [DOI: 10.1016/j.clinthera.2016.06.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 11/19/2022]
|
18
|
|
19
|
Huang FK, Chen HL, Yang PH, Lin HC. Bird's Eye View of a Neonatologist: Clinical Approach to Emergency Neonatal Infection. Pediatr Neonatol 2016; 57:167-73. [PMID: 26701838 DOI: 10.1016/j.pedneo.2015.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 04/21/2015] [Accepted: 06/01/2015] [Indexed: 10/22/2022] Open
Abstract
Though the incidence of neonatal infection in term and near-term infants is relatively low, incidence of infection in preterm very low birth weight infants is as high as 20-30% and may result in neurodevelopmental impairment or mortality. Pediatricians should be familiar with recognition and emergency management of life-threatening neonatal infections, such as congenital pneumonia, early onset sepsis, late onset sepsis, bacterial and fungal meningitis, disseminated neonatal herpes simplex virus (HSV), and HSV meningoencephalitis. For the pediatrician, it is logical to approach the management of these infections by time of onset, i.e., early versus late onset of infection. Perinatal risk factors and simple laboratory tests, such as total white blood-cell count, immature/total ratio, and C-reactive protein are helpful in guiding the decision of antibiotics therapy. Successful management of these critical infections depends upon early diagnosis and timely administration of adequate antibiotics. Empiric antibiotic therapy must cover the most likely pathogens according to the risk factors of each individual neonate, and therapy duration is dependent upon culture results, clinical course, and the microorganism. Future research may focus on developing a practical neonatal sepsis score system based on risk factors and common biomarkers, which are readily available at bedside to make early accurate decisions and achieve better outcomes.
Collapse
Affiliation(s)
- Fu-Kuei Huang
- Department of Pediatrics, China Medical University Children Hospital, Taichung, Taiwan
| | - Hsiu-Lin Chen
- Department of Pediatrics of Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Peng-Hong Yang
- Department of Neonatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hung-Chih Lin
- Department of Pediatrics, China Medical University Children Hospital, Taichung, Taiwan; School of Chinese Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
20
|
Pai S, Enoch DA, Aliyu SH. Bacteremia in children: epidemiology, clinical diagnosis and antibiotic treatment. Expert Rev Anti Infect Ther 2015; 13:1073-88. [PMID: 26143645 DOI: 10.1586/14787210.2015.1063418] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The diagnosis of bacteremia in children is important and it can be clinically challenging to recognize the signs and symptoms. The reported rates of bacteremia are higher in young children but with the increasing vaccine coverage, there has been a decrease in bacteremia due to the three vaccine preventable bacteria (Streptococcus pneumoniae, Haemophilus influenzae group b and Neisseria meningitidis). Notably, there have been increases in healthcare-associated bacteremias with a rise in Staphylococcus aureus and Gram negative bacteremias. This review provides a brief overview of the clinical diagnosis of bacteremia in children, focusing on the epidemiology, clinical characteristics, risk factors, antibiotic treatment, outcomes and preventative measures to reduce the incidence of bacteremia and improve morbidity and mortality.
Collapse
Affiliation(s)
- Sumita Pai
- Public Health England, Clinical Microbiology and Public Health Laboratory, Cambridge, UK
| | | | | |
Collapse
|
21
|
Abstract
Neonatal bacterial meningitis is uncommon but devastating. Morbidity among survivors remains high. The types and distribution of pathogens are related to gestational age, postnatal age, and geographic region. Confirming the diagnosis is difficult. Clinical signs are often subtle, lumbar punctures are frequently deferred, and cerebrospinal fluid (CSF) cultures can be compromised by prior antibiotic exposure. Infants with bacterial meningitis can have negative blood cultures and normal CSF parameters. Promising tests such as the polymerase chain reaction require further study. Prompt treatment with antibiotics is essential. Clinical trials investigating a vaccine for preventing neonatal Group B Streptococcus infections are ongoing.
Collapse
Affiliation(s)
- Lawrence C. Ku
- Duke Clinical Research Institute, Box 17969, Durham, NC, 27715; ; phone: 919-668-1592; fax: 919-668-7058 (corresponding author)
| | - Kim A. Boggess
- University of North Carolina School of Medicine, Dept. of Ob/Gyn CB 7570, Chapel Hill, NC 27599-7570; ; phone: 919-966-1601; fax: 919-966-6377
| | - Michael Cohen-Wolkowiez
- Department of Pediatrics, Duke Clinical Research Institute, Box 17969, Durham, NC 27715, USA.
| |
Collapse
|
22
|
Kraus-Haas M, Mielke M, Simon A. [Update on outbreaks reported from neonatal intensive care units (2010-203): Staphylococcus aureus]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2015; 58:323-38. [PMID: 25566845 DOI: 10.1007/s00103-014-2115-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In terms of the unique risk profile of the patients and the morbidity associated with S. aureus infections in this vulnerable patient population, the literature on outbreaks of S. aureus (including MRSA) in neonatal intensive care units (NICUs) needs to be analyzed separately from reports derived from other intensive care units. With the objective of updating important information for those involved in outbreak management and fostering preventive efforts, this article summarizes the results of a systematic literature analysis, referring to an earlier publication by Gastmeier et al. It focuses on NICU outbreaks caused by S. aureus (including MRSA) and on controlling them.
Collapse
|
23
|
Manage neonatal sepsis with prompt initiation of empirical antibacterial therapy, followed by targeted therapy. DRUGS & THERAPY PERSPECTIVES 2014. [DOI: 10.1007/s40267-014-0137-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
24
|
Roberts JK, Stockmann C, Constance JE, Stiers J, Spigarelli MG, Ward RM, Sherwin CMT. Pharmacokinetics and Pharmacodynamics of Antibacterials, Antifungals, and Antivirals Used Most Frequently in Neonates and Infants. Clin Pharmacokinet 2014; 53:581-610. [DOI: 10.1007/s40262-014-0147-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|