1
|
Franco S, Abdelhemid A, Fordjour L, Kohlhoff S, Hammerschlag MR. Stenotrophomonas maltophilia Associated Factors and Outcomes in a Neonatal Intensive Care Unit: A Retrospective Matched Case-control Study. Pediatr Infect Dis J 2025; 44:69-73. [PMID: 39705406 DOI: 10.1097/inf.0000000000004530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
BACKGROUND Stenotrophomonas maltophilia is a multi-drug-resistant, hospital-acquired Gram-negative bacillus associated with significant morbidity and mortality. The objective of this study is to identify risk factors and outcomes associated with S. maltophilia isolation in a high-risk neonatal population. METHODS This was a retrospective matched case-control study. Cases were matched 1:2 for years of neonatal intensive care unit admission, completed weeks' gestational age and birth weight in 250-gram incremental categories. RESULTS A total of 15 cases and 35 controls were included in the analyses. Risk factors for S. maltophilia isolation included days of antibiotics (24 vs. 18, P = 0.036), days of broad-spectrum antibiotics (19 vs. 12 days, P = 0.027), days of meropenem (9 vs. 6 days, P = 0.018) and any meropenem exposure (100% vs. 22%, P = 0.005). Other risk factors were any corticosteroid exposure (66.7% vs. 20%, P = 0.001), days of total parenteral nutrition (55 vs. 31 days, P = 0.017) and days of invasive mechanical ventilation (28 vs. 7, P = 0.015). S. maltophilia isolation was associated with increased length of neonatal intensive care unit stay (134 vs. 69 days, P < 0.001) and mortality (33.3% vs. 0%, P = 0.001). CONCLUSIONS Antibiotic stewardship efforts should be made to decrease the risk of S. maltophilia isolation and associated mortality. Carbapenem over-use should be specifically addressed with institutional policies and unit-based guidelines. Additional neonatal studies are needed to confirm these findings and explore other possible risk factors.
Collapse
Affiliation(s)
- Susannah Franco
- From the Department of Pharmacy, University Hospital at Downstate, Brooklyn, New York
- Division of Pediatric Infectious Diseases, Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, New York
| | - Ashraf Abdelhemid
- Division of Pediatric Infectious Diseases, Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, New York
| | - Lawrence Fordjour
- Division of Neonatology, Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, New York
| | - Stephan Kohlhoff
- Division of Pediatric Infectious Diseases, Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, New York
| | - Margaret R Hammerschlag
- Division of Pediatric Infectious Diseases, Department of Pediatrics, State University of New York Downstate Health Sciences University, Brooklyn, New York
| |
Collapse
|
2
|
Rueda MS, Soghier L, Campos J, Bahar B, Bost JE, Gai J, Hamdy RF. Blood volume collected for cultures in infants with suspected neonatal sepsis. J Perinatol 2024; 44:1800-1804. [PMID: 39341980 DOI: 10.1038/s41372-024-02120-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/17/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVES To evaluate blood culture sample volumes, identify factors linked to insufficient samples, and compare volumes among neonates treated for culture-negative-sepsis, sepsis-rule-outs, and bloodstream infections (BSI). METHODS Observational cohort of blood cultures collected during NICU stay. Association of age, weight, gender, source, and collection time with lower-than-recommended volumes was determined by logistic regression. Blood culture inocula of patients with culture-negative-sepsis, sepsis rule-out, and BSI were compared using ANOVA. RESULTS 742 blood cultures were obtained from 292 neonates. Median inoculum was 1 mL (IQR:0.6-1.4), and 259 bottles (35%) had inocula <0.9 mL. Night shift sample collection was associated with lower-than-recommended volumes (p = 0.006). No difference in sample volumes was observed between culture-negative-sepsis, sepsis-rule-outs, and BSI (p = 0.5). CONCLUSIONS Median NICU blood culture volumes align with recommendations. Night shift collections correlate with lower volumes. Sample volumes don't differ in patients with culture-negative-sepsis, BSI, and sepsis-rule-out, and should not be a justification for longer duration of antibiotics.
Collapse
Affiliation(s)
- Maria S Rueda
- Division of Pediatric Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Lamia Soghier
- Division of Neonatology, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Joseph Campos
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Division of Laboratory Medicine, Children's National Hospital, Washington, DC, USA
- Department of Pathology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Department of Microbiology/Immunology/Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Burak Bahar
- Division of Laboratory Medicine, Children's National Hospital, Washington, DC, USA
| | - James E Bost
- Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Jiaxiang Gai
- Division of Biostatistics and Study Methodology, Children's National Hospital, Washington, DC, USA
| | - Rana F Hamdy
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Division of Infectious Diseases, Children's National Hospital, Washington, DC, USA
| |
Collapse
|
3
|
Raturi A, Chandran S. Neonatal Sepsis: Aetiology, Pathophysiology, Diagnostic Advances and Management Strategies. Clin Med Insights Pediatr 2024; 18:11795565241281337. [PMID: 39371316 PMCID: PMC11452898 DOI: 10.1177/11795565241281337] [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: 12/25/2023] [Accepted: 08/21/2024] [Indexed: 10/08/2024] Open
Abstract
Neonatal sepsis, a bloodstream infection in the first 28 days of life, is a leading cause of morbidity and mortality among infants in both developing and developed countries. Additionally, sepsis is distinguished in neonates by unique pathophysiological and presentational factors relating to its development in immature neonatal immune systems. This review focuses on the current understanding of the mechanics and implications of neonatal sepsis, providing a comprehensive overview of the epidemiology, aetiology, pathophysiology, major risk factors, signs and symptoms and recent consensus on the diagnosis and management of both early-onset and late-onset neonatal sepsis. It also includes a discussion on novel biomarkers and upcoming treatment strategies for the condition as well as the potential of COVID-19 infection to progress to sepsis in infants.
Collapse
Affiliation(s)
- Adi Raturi
- University of Glasgow School of Medicine, Glasgow, UK
| | - Suresh Chandran
- Department of Neonatology, KK Women’s and Children’s Hospital, Singapore, Singapore
- Duke NUS Medical School, Singapore, Singapore
- Lee Kong Chian School of Medicine, Singapore, Singapore
- Yong Loo Lin School of Medicine, Singapore, Singapore
| |
Collapse
|
4
|
Alosaimi HM, Alshammari MK, Fetyani MM, Allehidan MS, Almalki TJ, Hussain KH, Hussain HH, Althobaiti MD, Alharbi AS, Alharthi AA, Al-Shammari AA, Al Jamea ZA, Alamro RA, Najmi A. Point prevalence survey of antibiotics use among hospitalised neonates and children in Saudi Arabia: findings and implications. J Pharm Policy Pract 2024; 17:2371411. [PMID: 39011353 PMCID: PMC11249164 DOI: 10.1080/20523211.2024.2371411] [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: 04/17/2024] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
Background Neonates and children are more susceptible to a variety of infections, leading to frequent antibiotic prescriptions. However, the inappropriate use of antibiotics leads to antibiotic resistance and higher mortality rates. Therefore, this study aimed to determine the prevalence of antibiotic use, and current antibiotic prescribing practices among neonates and children admitted in the selected hospitals of Saudi Arabia. Methods A cross-sectional study was conducted from September to November 2023 to assess the prevalence of antibiotic use, and the current antibiotic prescribing practices across six hospitals of Saudi Arabia. Results The study included 499 children and neonates, with 94.6% receiving antibiotic prescriptions. The most frequently prescribed antibiotic class was third-generation cephalosporin (31.5%), with ceftriaxone being the most commonly prescribed antibiotic (15%). The majority of patients were prescribed one antibiotic (81.4%), and the intravenous route (96.4%) was the primary route for administration. The majority of patients were prescribed antibiotics empirically (69.7%), and community-acquired infections (64.2%) were the most common type of infection for antibiotic prescription. Similarly, sepsis (39.2%) was the most common indication for antibiotics, and the majority of prescribed antibiotics (61.7%) belonged to the 'Watch' category as per WHO AWaRe classification. Conclusion Our study revealed excessive antibiotic consumption in neonates and children, therefore quality improvement programmes including antimicrobial stewardship programmes are urgently needed to address ongoing issues.
Collapse
Affiliation(s)
- Hind M Alosaimi
- Department of Pharmacy Services Administration, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh, Saudi Arabia
| | - Mohammed K Alshammari
- Department of Clinical Pharmacy, King Fahad Medical City, Kingdom of Saudi Arabia, Riyadh, Saudi Arabia
| | - Mohammad M Fetyani
- Department of Pharmacy Services, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh, Saudi Arabia
| | - Maha S Allehidan
- Department of Pharmacy Services, NICU/Pediatric Clinical Pharmacist, Alyamam Hospital, Second Health Cluster, Riyadh, Saudi Arabia
| | - Tahani J Almalki
- Pharmaceutical Care Administration, Armed Forces Hospital Southern Region, Khamis Mushait, Saudi Arabia
| | - Khansa H Hussain
- Department of Cardiac Science, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Haifaa H Hussain
- Department of Nursing, Advanced Practice Nurse, Pediatric Nurse Practitioner, King Faisal Specialist Hospital and Research Center, Kingdom of Saudi Arabia, Riyadh, Saudi Arabia
| | - Mohammed D Althobaiti
- Department of Medical Surgical Nursing, King Faisal Specialist Hospital & Research Center, Kingdom of Saudi Arabia, Riyadh, Saudi Arabia
| | - Abrar S Alharbi
- Department of Pharmaceutical Services, Maternity and Children's Hospital, West Zone, Kingdom of Saudi Arabia, Mecca, Saudi Arabia
| | | | - Amosha A Al-Shammari
- Department of Pharmacy, Maternity and Children Hospital in Rafah, Kingdom of Saudi Arabia, Rafha, Saudi Arabia
| | - Zainab A Al Jamea
- Department of Pharmaceutical Care, King Fahd Hospital of University, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Rayed A Alamro
- Department of Pharmacy Services, Dr. Sulaiman Al Habib Medical Group Alrayyan Hospital, Riyadh, Saudi Arabia
| | - Ali Najmi
- Pharmaceutical Care Administration, Armed Forces Hospital Southern Region, Khamis Mushait, Saudi Arabia
| |
Collapse
|
5
|
Sharma V, Grover R, Priyadarshi M, Chaurasia S, Bhat NK, Basu S, Singh P. Point-of-Care Serum Amyloid A as a Diagnostic Marker for Neonatal Sepsis. Indian J Pediatr 2024; 91:571-577. [PMID: 37368220 DOI: 10.1007/s12098-023-04677-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 03/17/2023] [Indexed: 06/28/2023]
Abstract
OBJECTIVES To evaluate diagnostic accuracy of point-of-care Serum Amyloid A (POC-SAA) and its comparison with procalcitonin for diagnosis of neonatal sepsis. METHODS The present diagnostic accuracy study consecutively recruited neonates with suspected sepsis. Blood samples for sepsis screen, culture, high sensitivity C-reactive protein (CRP) (hs-CRP, as a part of sepsis screen), procalcitonin and POC-SAA were collected before starting antibiotics. The optimum cut-off level of biomarkers (POC-SAA and procalcitonin) was determined by receiver-operating-characteristics curve (ROC) analysis. Sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV) of POC-SAA and procalcitonin were derived for 'clinical sepsis (neonates with suspected sepsis and either positive sepsis screen and/or blood culture)' and 'culture positive sepsis' (neonates with suspected sepsis and positive blood culture). RESULTS Seventy-four neonates with mean±SD gestational age of 32.8±3.7 wk were evaluated for suspected sepsis, of which the proportion of 'clinical sepsis' and 'culture positive sepsis' was 37.8% had 16.2%, respectively. At a cut-off of 25.4 mg/L, POC-SAA had sensitivity, specificity, PPV and NPV of 53.6%, 80.4%, 62.5% and 74.0%, respectively for diagnosis of clinical sepsis. The sensitivity, specificity, PPV and NPV of POC-SAA for detection of culture positive sepsis were 83.3%, 61.3%, 29.4% and 95.0%, respectively at a cut-off of 10.3 mg/L. There was no significant difference in the diagnostic accuracy of biomarkers for detection of culture positive sepsis (area under the curve, AUC of POC-SAA vs. procalcitonin vs. hs-CRP: 0.72 vs. 0.85 vs. 0.85; p = 0.21). CONCLUSIONS POC-SAA is comparable to procalcitonin and hs-CRP for diagnosis of neonatal sepsis.
Collapse
Affiliation(s)
- Vishakha Sharma
- Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, 249203, India
| | - Rajat Grover
- Department of Neonatology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, 249203, India
| | - Mayank Priyadarshi
- Department of Neonatology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, 249203, India
| | - Suman Chaurasia
- Department of Neonatology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, 249203, India
| | - Nowneet Kumar Bhat
- Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, 249203, India
| | - Sriparna Basu
- Department of Neonatology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, 249203, India
| | - Poonam Singh
- Department of Neonatology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, 249203, India.
| |
Collapse
|
6
|
Singh NK, Will L, Al-Mulaabed S, Ruoss L, Li N, de La Cruz D, Gurka M, Neu J. Antibiotics Use and Its Effects on the Establishment of Feeding Tolerance in Preterm Neonates. Am J Perinatol 2024; 41:e2248-e2253. [PMID: 37308133 DOI: 10.1055/a-2108-1960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
OBJECTIVE Antibiotics are one of the most widely used medications in today's neonatal intensive care units. Indiscriminate antibiotic usage persists in preterm newborns who are symptomatic due to factors linked to prematurity rather than being septic. Previous studies in older infants suggest that prior antibiotic administration is associated with possible dysmotility and microbial dysbiosis in the intestinal tract. We hypothesize that early antibiotic administration impacts high-risk preterm infants' tolerance to enteral feeding advancement. STUDY DESIGN As part of the Routine Early Antibiotic Use in SymptOmatic Preterm Neonates study, symptomatic preterm newborns without maternal infection risk factors were randomized to receive or not receive antibiotics, with C1 receiving antibiotics and C2 not. Of the 55 newborns that underwent pragmatic randomization, 28 preterm neonates in group C1 received antibiotics. RESULTS The premature neonates in the randomized groups who received antibiotics and those who did not showed no differences in sustained feeding tolerance. CONCLUSION Our investigation of the risk of feeding issues in babies who get antibiotics early in life revealed no differences between neonates who received antibiotics and those who did not when the randomized controlled trial data alone was reviewed. Given the sample sizes, it is uncertain if the preceding analysis is powerful enough to detect differences (a significant percentage of neonates who were randomly assigned to NOT get antibiotics subsequently received early treatment due to changing clinical conditions). This affirms the requirement for a meticulously designed prospective randomized study. KEY POINTS · Defining feeding tolerance for the first time in neonates.. · Patients from the REASON trial were evaluated.. · Preterm neonates were the focus of this study..
Collapse
Affiliation(s)
- Neel K Singh
- Department of Pediatrics, Division of Neonatology, Shands Children's Hospital, University of Florida, Gainesville, Florida
| | - Lester Will
- Department of Pediatrics, Division of Neonatology, Shands Children's Hospital, University of Florida, Gainesville, Florida
| | - Sharef Al-Mulaabed
- Department of Pediatrics, Presbyterian Medical Group, Albuquerque, New Mexico
| | - Lauren Ruoss
- Department of Pediatrics, Division of Neonatology, Shands Children's Hospital, University of Florida, Gainesville, Florida
| | - Nan Li
- Department of Pediatrics, Division of Neonatology, Shands Children's Hospital, University of Florida, Gainesville, Florida
| | - Diomel de La Cruz
- Department of Pediatrics, Division of Neonatology, Shands Children's Hospital, University of Florida, Gainesville, Florida
| | - Matthew Gurka
- Pediatrics Research Hub (PoRCH), Department of Pediatrics in the College of Medicine, University of Florida, Gainesville, Florida
| | - Josef Neu
- Department of Pediatrics, Division of Neonatology, Shands Children's Hospital, University of Florida, Gainesville, Florida
| |
Collapse
|
7
|
Stevens J, Culberson E, Kinder J, Ramiriqui A, Gray J, Bonfield M, Shao TY, Al Gharabieh F, Peterson L, Steinmeyer S, Zacharias W, Pryhuber G, Paul O, Sengupta S, Alenghat T, Way SS, Deshmukh H. Microbiota-derived inosine programs protective CD8 + T cell responses against influenza in newborns. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588427. [PMID: 38645130 PMCID: PMC11030415 DOI: 10.1101/2024.04.09.588427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The immunological defects causing susceptibility to severe viral respiratory infections due to early-life dysbiosis remain ill-defined. Here, we show that influenza virus susceptibility in dysbiotic infant mice is caused by CD8+ T cell hyporesponsiveness and diminished persistence as tissue-resident memory cells. We describe a previously unknown role for nuclear factor interleukin 3 (NFIL3) in repression of memory differentiation of CD8+ T cells in dysbiotic mice involving epigenetic regulation of T cell factor 1 (TCF 1) expression. Pulmonary CD8+ T cells from dysbiotic human infants share these transcriptional signatures and functional phenotypes. Mechanistically, intestinal inosine was reduced in dysbiotic human infants and newborn mice, and inosine replacement reversed epigenetic dysregulation of Tcf7 and increased memory differentiation and responsiveness of pulmonary CD8+ T cells. Our data unveils new developmental layers controlling immune cell activation and identifies microbial metabolites that may be used therapeutically in the future to protect at-risk newborns.
Collapse
Affiliation(s)
- Joseph Stevens
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
- Medical Scientist Training Program, University of Cincinnati College of Medicine
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center
| | - Erica Culberson
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
- Medical Scientist Training Program, University of Cincinnati College of Medicine
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center
| | - Jeremy Kinder
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center
| | - Alicia Ramiriqui
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
| | - Jerilyn Gray
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
| | - Madeline Bonfield
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center
| | - Tzu-Yu Shao
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center
| | - Faris Al Gharabieh
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
| | - Laura Peterson
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
| | - Shelby Steinmeyer
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
| | - William Zacharias
- Department of Pediatrics, University of Cincinnati College of Medicine
- Medical Scientist Training Program, University of Cincinnati College of Medicine
| | - Gloria Pryhuber
- Department of Pediatrics, University of Rochester, School of Medicine
| | - Oindrila Paul
- Division of Neonatology, Children’s Hospital of Philadelphia; Perelman School of Medicine, University of Pennsylvania
| | - Shaon Sengupta
- Division of Neonatology, Children’s Hospital of Philadelphia; Perelman School of Medicine, University of Pennsylvania
| | - Theresa Alenghat
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center
| | - Sing Sing Way
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Infectious Disease, Cincinnati Children’s Hospital Medical Center
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center
| | - Hitesh Deshmukh
- Department of Pediatrics, University of Cincinnati College of Medicine
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center
| |
Collapse
|
8
|
Nuzum ND, Deady C, Kittel-Schneider S, Cryan JF, O'Mahony SM, Clarke G. More than just a number: the gut microbiota and brain function across the extremes of life. Gut Microbes 2024; 16:2418988. [PMID: 39567371 PMCID: PMC11583591 DOI: 10.1080/19490976.2024.2418988] [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: 05/28/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024] Open
Abstract
Understanding the interrelationship between the gut microbiota and host physiology, although still in its relative infancy, has taken important steps forward over the past decade. In the context of brain disorders including those characterized by neurodevelopmental and neurodegenerative changes there have been important advances. However, initially research involved correlational analyses, had limited translational scope, and lacked functional assessments. Thus, largescale longitudinal clinical investigations that assess causation and underlying mechanisms via in depth analysis methods are needed. In neurodegeneration research, strong causal evidence now links the gut microbiome to Alzheimer's (AD), and Parkinson's Disease (PD), as supported by human-to-animal transplantation studies. Longitudinal interventions are being conducted in AD, PD, amyotrophic lateral sclerosis, Huntington's disease, and multiple sclerosis. Neurodevelopmental research has also seen a boon in microbiome-related clinical research including in autism, Attention-deficit/hyperactivity disorder, and schizophrenia, which is confirming prior animal model work regarding the key time-windows in the gut microbiome important for infant cognition. While recent research advances represent important progress, fundamental knowledge gaps and obstacles remain. Knowing how and why the gut microbiome changes at the extremes of life will develop our mechanistic understanding and help build the evidence base as we strive toward counteracting microbial missteps with precision therapeutic interventions.
Collapse
Affiliation(s)
- Nathan D Nuzum
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Sarah Kittel-Schneider
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Siobhain M O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| |
Collapse
|
9
|
Baharin NS, Duan M, Loe MWC, Goh GL, Thoon KC, Rajadurai VS, Yeo KT. Burden of antibiotic resistance in infections among very-low-birthweight infants in Singapore. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2023; 52:561-569. [PMID: 38920145 DOI: 10.47102/annals-acadmedsg.2023190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Introduction Recent reports have described the increasing predominance of Gram-negative organisms among invasive bacterial infections affecting preterm infants. This changing pattern of infections is concerning due to the spread of antibiotic resistance among Gram-negatives. Method We conducted a single-centre, retrospective cohort study involving very-low-birthweight (VLBW) (<1500 grams) infants born <32 weeks gestation, with culture-proven infections (blood, urine, cerebrospinal fluid [CSF]) in the neonatal intensive care unit from 1 January 2005 to 31 October 2017. Results A total of 278 out of 2431 (11.4%) VLBW infants born <32 weeks gestation developed 334 infections, i.e. 52 (15.6%) early-onset infections (EOIs) and 282 (84.4%) late-onset infections (LOIs). The overall incidence decreased from 247 to 68 infections per 1000 infants over the study period, corresponding to reductions in LOI (211 to 62 infections per 1000 infants). A total of 378 bacteria were isolated, i.e. Gram-negatives accounted for 70.9% (45 of 59 [76.3%] EOI; 223 of 319 [69.9%] LOI). Specific resistant organisms were noted, i.e. Methicillin-resistant Staphylococcus aureus (8 of 21 S. aureus infections [38.1%]); Cephalosporin-resistant Klebsiella (18 of 62 isolates [29.0%]) and multidrug-resistant [MDR] Acinetobacter (10 of 27 isolates [37.0%]). MDR organisms accounted for 85 of 195 (43.6%) Gram-negative infections from the bloodstream and CSF. Based on laboratory susceptibility testing, only 63.5% and 49.3% of infecting bacteria isolated in blood were susceptible to empiric antibiotic regimens used for suspected EOI and LOI, respectively. Conclusion Gram-negative bacteria are the predominant causative organisms for EOI and LOI and are frequently MDR. Understanding the pattern of antimicrobial resistance is important in providing appropriate empiric coverage for neonatal infections.
Collapse
MESH Headings
- Humans
- Singapore/epidemiology
- Infant, Newborn
- Retrospective Studies
- Infant, Very Low Birth Weight
- Female
- Male
- Intensive Care Units, Neonatal/statistics & numerical data
- Anti-Bacterial Agents/therapeutic use
- Anti-Bacterial Agents/pharmacology
- Gram-Negative Bacterial Infections/epidemiology
- Gram-Negative Bacterial Infections/drug therapy
- Gram-Negative Bacterial Infections/microbiology
- Incidence
- Gram-Negative Bacteria/drug effects
- Gram-Negative Bacteria/isolation & purification
- Infant, Premature
- Drug Resistance, Bacterial
- Infant, Premature, Diseases/epidemiology
- Infant, Premature, Diseases/microbiology
- Infant, Premature, Diseases/drug therapy
- Methicillin-Resistant Staphylococcus aureus/isolation & purification
- Methicillin-Resistant Staphylococcus aureus/drug effects
- Microbial Sensitivity Tests
Collapse
Affiliation(s)
| | - Menghao Duan
- Department of Neonatology, KK Women's & Children's Hospital, Singapore
| | - Marcus Wing Choy Loe
- Department of Neonatology, KK Women's & Children's Hospital, Singapore
- Duke-NUS Medical School, Singapore
| | - Guan Lin Goh
- Department of Neonatology, KK Women's & Children's Hospital, Singapore
| | - Koh Cheng Thoon
- Duke-NUS Medical School, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Infectious Disease Service, KK Women's & Children's Hospital, Singapore
| | - Victor Samuel Rajadurai
- Department of Neonatology, KK Women's & Children's Hospital, Singapore
- Duke-NUS Medical School, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Kee Thai Yeo
- Department of Neonatology, KK Women's & Children's Hospital, Singapore
- Duke-NUS Medical School, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
10
|
Graspeuntner S, Lupatsii M, Dashdorj L, Rody A, Rupp J, Bossung V, Härtel C. First-Day-of-Life Rectal Swabs Fail To Represent Meconial Microbiota Composition and Underestimate the Presence of Antibiotic Resistance Genes. Microbiol Spectr 2023; 11:e0525422. [PMID: 37097170 PMCID: PMC10269712 DOI: 10.1128/spectrum.05254-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/10/2023] [Indexed: 04/26/2023] Open
Abstract
The human gut microbiome plays a vital role in health and disease. In particular, the first days of life provide a unique window of opportunity for development and establishment of microbial community. Currently, stool samples are known to be the most widely used sampling approach for studying the gut microbiome. However, complicated sample acquisition at certain time points, challenges in transportation, and patient discomfort underline the need for development of alternative sampling approaches. One of the alternatives is rectal swabs, shown to be a reliable proxy for gut microbiome analysis when obtained from adults. Here, we compare the usability of rectal swabs and meconium paired samples collected from infants on the first days of life. Our results indicate that the two sampling approaches display significantly distinct patterns in microbial composition and alpha and beta diversity as well as detection of resistance genes. Moreover, the dissimilarity between the two collection methods was greater than the interindividual variation. Therefore, we conclude that rectal swabs are not a reliable proxy compared to stool samples for gut microbiome analysis when collected on the first days of a newborn's life. IMPORTANCE Currently, there are numerous suggestions on how to ease the notoriously complex and error-prone methodological setups to study the gut microbiota of newborns during the first days of life. Especially, meconium samples are regularly failing to yield meaningful data output and therefore have been suggested to be replaced by rectal swabs as done in adults as well. We find this development toward a simplified method to be producing dramatically erroneous results, skewing data interpretation away from the real aspects to be considered for neonatal health during the first days of life. We have put together our knowledge on this critical aspect with careful consideration and identified the failure of rectal swabs to be a replacement for sampling of meconium in term-born newborns.
Collapse
Affiliation(s)
- S. Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - M. Lupatsii
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - L. Dashdorj
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - A. Rody
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - J. Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - V. Bossung
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Lübeck, Germany
- Department of Pediatrics, University Hospital of Würzburg, Würzburg, Germany
| | - C. Härtel
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
- Department of Obstetrics, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Abstract
Antibiotic therapy remains a cornerstone of treatment of both medical and surgical presentations of necrotizing enterocolitis (NEC). However, guidelines regarding the administration of antibiotics for the treatment of NEC are lacking and practices vary amongst clinicians. Although the pathogenesis of NEC is unknown, there is consensus that the infant gastrointestinal microbiome contributes to the disease. The presumed connection between dysbiosis and NEC has prompted some to study whether early prophylactic enteral antibiotics can prevent NEC. Yet others have taken an opposing approach, studying whether perinatal antibiotic exposure increases the risk of NEC by inducing a state of dysbiosis. This narrative review summarizes what is known about antibiotics and their association with the infant microbiome and NEC, current antibiotic prescribing practices for infants with medical and surgical NEC, as well as potential strategies to further optimize the use of antibiotics in this population of infants.
Collapse
Affiliation(s)
- Elizabeth Pace
- University of Pittsburgh Department of Surgery, United States
| | - Toby D Yanowitz
- University of Pittsburgh Department of Pediatrics, Division of Neonatology, United States
| | - Paul Waltz
- University of Pittsburgh Department of Surgery, Division of Pediatric General and Thoracic Surgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, United States
| | - Michael J Morowitz
- University of Pittsburgh Department of Surgery, Division of Pediatric General and Thoracic Surgery, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, United States.
| |
Collapse
|
12
|
Sourour W, Sanchez V, Sourour M, Burdine J, Lien ER, Nguyen D, Jain SK. The Association between Prolonged Antibiotic Use in Culture Negative Infants and Length of Hospital Stay and Total Hospital Costs. Am J Perinatol 2023; 40:525-531. [PMID: 33975363 DOI: 10.1055/s-0041-1729560] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE This study aimed to determine if prolonged antibiotic use at birth in neonates with a negative blood culture increases the total cost of hospital stay. STUDY DESIGN This was a retrospective study performed at a 60-bed level IV neonatal intensive care unit. Neonates born <30 weeks of gestation or <1,500 g between 2016 and 2018 who received antibiotics were included. A multivariate linear regression analysis was conducted to determine if clinical factors contributed to increased hospital cost or length of stay. RESULTS In total, 190 patients met inclusion criteria with 94 infants in the prolonged antibiotic group and 96 in the control group. Prolonged antibiotic use was associated with an increase length of hospital stay of approximately 31.87 days, resulting in a $69,946 increase in total cost of hospitalization. CONCLUSION Prolonged antibiotics in neonates with negative blood culture were associated with significantly longer hospital length of stay and increased total cost of hospitalization. KEY POINTS · Prolonged antibiotic use at birth is associated with prolonged hospital stay.. · Prolonged antibiotic use at birth is associated with increased cost of hospitalization.. · Prolonged antibiotic use at birth is associated with increased days on total parenteral nutrition.. · Prolonged antibiotic use at birth is associated with increased subsequent courses of antibiotics..
Collapse
Affiliation(s)
- Wesam Sourour
- Division of Pediatrics, University of Texas Medical Branch, Galveston, Texas
| | - Valeria Sanchez
- University of Texas Medical Branch School of Medicine, Galveston, Texas
| | - Michel Sourour
- Division of Neurosurgery, McMaster University, Hamilton, Ontario, Canada
| | - Jordan Burdine
- Department of Pharmacy, University of Texas Medical Branch, Galveston, Texas
| | | | - Diana Nguyen
- Division of Pediatric Infectious Diseases, University of Texas Medical Branch, Galveston, Texas
| | - Sunil K Jain
- Division of Neonatology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
13
|
Neonatal prophylactic antibiotics after preterm birth affect plasma proteome and immune development in pigs. Pediatr Res 2023:10.1038/s41390-023-02492-7. [PMID: 36804504 DOI: 10.1038/s41390-023-02492-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/12/2022] [Accepted: 01/15/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND Most preterm infants receive antibiotics to prevent serious infections shortly after birth. However, prolonged antibiotic treatment predisposes to gut dysbiosis and late-onset sepsis. Using preterm pigs as model, we hypothesized that neonatal prophylactic antibiotics impair systemic immune development beyond the days of antibiotic treatment. METHODS Preterm pigs (90% gestation) were fed formula for 9 days, treated with sterile water (CON) or enteral antibiotics from day 1 to 4. On days 5 and 9, blood was collected for haematology, in vitro LPS stimulation, and plasma proteomics. RESULTS Antibiotic treatment altered the abundance of 21 and 47 plasma proteins on days 5 and 9, representing 6.6% and 14.8% of the total annotated proteins, respectively. Most antibiotics-induced proteome changes related to complement cascade, neutrophil degranulation, and acute phase responses. Neutrophil and lymphocyte counts were higher in antibiotics-treated pigs on day 5 but did not change from days 5-9, in contrast to increasing cell counts in CON. The antibiotics treatment suppressed TNF-alpha and IL-10 responses to in vitro LPS challenge on day 5, 7 and 9. CONCLUSION Few days of antibiotics treatment following preterm birth alter the plasma proteome and inhibit systemic immune development, even beyond the days of treatment. IMPACT 1. Neonatal prophylactic antibiotics alter the plasma proteome and suppress systemic immune development in preterm pigs 2. The effects of prophylactic antibiotics last beyond the days of treatment. 3. Neonatal antibiotics treatment for compromised human newborns may predispose to longer-term risks of impaired immunity and infections.
Collapse
|
14
|
Incidence of Early and Late Onset Neonatal Sepsis in Suriname: A National Tertiary Hospital Birth-cohort Study. Pediatr Infect Dis J 2022; 41:1007-1011. [PMID: 36102696 DOI: 10.1097/inf.0000000000003683] [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: 01/09/2023]
Abstract
BACKGROUND Early onset neonatal sepsis (EONS) and late onset neonatal sepsis (LONS) are important causes of neonatal mortality and morbidity. A pressing need for reliable and detailed data of low- and middle-income countries exists. This study aimed to describe the incidence and outcome of neonatal sepsis in the only tertiary hospital of Suriname, a middle-income country in South America. METHODS Infants born at the Academic Hospital of Paramaribo from May 2017 through December 2018 were prospectively included at birth. Perinatal data, duration of antibiotic treatment, blood culture results and mortality data were gathered. Neonatal sepsis was defined as positive blood culture with a pathogenic microorganism within the first 28 days of life. RESULTS Of the 2190 infants included, 483 (22%) were admitted to neonatal (intensive) care. The incidence of EONS was 2.1 (95% CI: 0.9-5) per 1000 live births, with no deaths. Antibiotics for suspected EONS were administrated to 189 (8.6%) infants, of whom 155 (82%) were born prematurely. The incidence of LONS cases was 145 (95% CI: 114-176) per 1000 admissions. Gramnegative bacteria accounted for 70% (48 out of 70) of causative organisms. Seventeen deaths were directly caused by sepsis (35 per 1000 admissions). CONCLUSIONS Findings from this tertiary center birth cohort study in a middle-income setting indicate EONS incidence and outcomes comparable to high-income settings, whereas LONS is a more prevalent and significant challenge with a predominance of gram-negative bacteria, and high mortality.
Collapse
|
15
|
Madany AM, Hughes HK, Ashwood P. Prenatal Maternal Antibiotics Treatment Alters the Gut Microbiota and Immune Function of Post-Weaned Prepubescent Offspring. Int J Mol Sci 2022; 23:12879. [PMID: 36361666 PMCID: PMC9655507 DOI: 10.3390/ijms232112879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/07/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
This study aimed to investigate the immediate and continual perturbation to the gut microbiota of offspring in the weeks post-weaning and how these may be modulated by treating pregnant C57BL/6J dams with antibiotics (ABX). We used a broad-spectrum antibiotic cocktail consisting of ampicillin 1 mg/mL, neomycin 1 mg/mL, and vancomycin 0.5 mg/mL, or vancomycin 0.5 mg/mL alone, administered ad-lib orally to dams via drinking water during gestation and stopped after delivery. We analyzed the gut microbiota of offspring, cytokine profiles in circulation, and the brain to determine if there was evidence of a gut-immune-brain connection. Computationally predicted metabolic pathways were calculated from 16s rRNA sequencing data. ABX treatment can negatively affect the gut microbiota, including reduced diversity, altered metabolic activity, and immune function. We show that the maternal ABX-treatment continues to alter the offspring's gut microbiota diversity, composition, and metabolic pathways after weaning, with the most significant differences evident in 5-week-olds as opposed to 4-week-olds. Lower levels of chemokines and inflammatory cytokines, such as interleukin (IL)-1α and IL-2, are also seen in the periphery and brains of offspring, respectively. In conclusion, this study shows maternal antibiotic administration alters gut microbiome profiles in offspring, which undergoes a continuous transformation, from week to week, at an early age after weaning.
Collapse
Affiliation(s)
- Abdullah M. Madany
- Department of Psychiatry and Behavioral Sciences, University of California at Davis, 2230 Stockton Blvd., Sacramento, CA 95817, USA
- The M.I.N.D. Institute, University of California at Davis, 2825 50th Street, Sacramento, CA 95817, USA
| | - Heather K. Hughes
- The M.I.N.D. Institute, University of California at Davis, 2825 50th Street, Sacramento, CA 95817, USA
- Department of Medical Microbiology and Immunology, University of California at Davis, 3146 One Shields Avenue, Davis, CA 95616, USA
| | - Paul Ashwood
- The M.I.N.D. Institute, University of California at Davis, 2825 50th Street, Sacramento, CA 95817, USA
- Department of Medical Microbiology and Immunology, University of California at Davis, 3146 One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|
16
|
Huang D, Li H, Lin Y, Lin J, Li C, Kuang Y, Zhou W, Huang B, Wang P. Antibiotic-induced depletion of Clostridium species increases the risk of secondary fungal infections in preterm infants. Front Cell Infect Microbiol 2022; 12:981823. [PMID: 36118040 PMCID: PMC9473543 DOI: 10.3389/fcimb.2022.981823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/12/2022] [Indexed: 11/28/2022] Open
Abstract
Preterm infants or those with low birth weight are highly susceptible to invasive fungal disease (IFD) and other microbial or viral infection due to immaturity of their immune system. Antibiotics are routinely administered in these vulnerable infants in treatment of sepsis and other infectious diseases, which might cause perturbation of gut microbiome and hence development of IFD. In this study, we compared clinical characteristics of fungal infection after antibiotic treatment in preterm infants. As determined by 16S rRNA sequencing, compared with non-IFD patients with or without antibiotics treatment, Clostridium species in the intestinal tracts of patients with IFD were almost completely eliminated, and Enterococcus were increased. We established a rat model of IFD by intraperitoneal inoculation of C. albicans in rats pretreated with meropenem and vancomycin. After pretreatment with antibiotics, the intestinal microbiomes of rats infected with C. albicans were disordered, as characterized by an increase of proinflammatory conditional pathogens and a sharp decrease of Clostridium species and Bacteroides. Immunofluorescence analysis showed that C. albicans-infected rats pretreated with antibiotics were deficient in IgA and IL10, while the number of Pro-inflammatory CD11c+ macrophages was increased. In conclusion, excessive use of antibiotics promoted the imbalance of intestinal microbiome, especially sharp decreases of short-chain fatty acids (SCFA)-producing Clostridium species, which exacerbated the symptoms of IFD, potentially through decreased mucosal immunomodulatory molecules. Our results suggest that inappropriate use of broad-spectrum antibiotics may promote the colonization of invasive fungi. The results of this study provide new insights into the prevention of IFD in preterm infants.
Collapse
Affiliation(s)
- Dabin Huang
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huixian Li
- Department of Data Center, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Yuying Lin
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jinting Lin
- Department of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Chengxi Li
- Department of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Yashu Kuang
- Division of Birth Cohort Study, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wei Zhou
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Wei Zhou, ; Bing Huang, ; Ping Wang,
| | - Bing Huang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Wei Zhou, ; Bing Huang, ; Ping Wang,
| | - Ping Wang
- Department of Neonatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Wei Zhou, ; Bing Huang, ; Ping Wang,
| |
Collapse
|
17
|
Lingel I, Wilburn AN, Hargis J, McAlees JW, Laumonnier Y, Chougnet CA, Deshmukh H, König P, Lewkowich IP, Schmudde I. Prenatal antibiotics exposure does not influence experimental allergic asthma in mice. Front Immunol 2022; 13:937577. [PMID: 36032166 PMCID: PMC9399857 DOI: 10.3389/fimmu.2022.937577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Changes in microbiome (dysbiosis) contribute to severity of allergic asthma. Preexisting epidemiological studies in humans correlate perinatal dysbiosis with increased long-term asthma severity. However, these studies cannot discriminate between prenatal and postnatal effects of dysbiosis and suffer from a high variability of dysbiotic causes ranging from antibiotic treatment, delivery by caesarian section to early-life breastfeeding practices. Given that maternal antibiotic exposure in mice increases the risk of newborn bacterial pneumonia in offspring, we hypothesized that prenatal maternal antibiotic-induced dysbiosis induces long-term immunological effects in the offspring that also increase long-term asthma severity. Therefore, dams were exposed to antibiotics (gentamycin, ampicillin, vancomycin) from embryonic day 15 until birth. Six weeks later, asthma was induced in the offspring by repeated applications of house dust mite extract. Airway function, cytokine production, pulmonary cell composition and distribution were assessed. Our study revealed that prenatally induced dysbiosis in mice led to an increase in pulmonary Th17+ non-conventional T cells with limited functional effect on airway resistance, pro-asthmatic Th2/Th17 cytokine production, pulmonary localization and cell-cell contacts. These data indicate that dysbiosis-related immune-modulation with long-term effects on asthma development occurs to a lesser extent prenatally and will allow to focus future studies on more decisive postnatal timeframes.
Collapse
Affiliation(s)
- Imke Lingel
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Lübeck, Germany
| | - Adrienne N. Wilburn
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, University of Cincinnati, Cincinnati, OH, United States
| | - Julie Hargis
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Jaclyn W. McAlees
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Yves Laumonnier
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Lübeck, Germany
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Hitesh Deshmukh
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Peter König
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Lübeck, Germany
| | - Ian P. Lewkowich
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Inken Schmudde
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Lübeck, Germany
- *Correspondence: Inken Schmudde,
| |
Collapse
|
18
|
Morowitz MJ, Katheria AC, Polin RA, Pace E, Huang DT, Chang CCH, Yabes JG. The NICU Antibiotics and Outcomes (NANO) trial: a randomized multicenter clinical trial assessing empiric antibiotics and clinical outcomes in newborn preterm infants. Trials 2022; 23:428. [PMID: 35606829 PMCID: PMC9125935 DOI: 10.1186/s13063-022-06352-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 04/25/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Early-onset sepsis is an important cause of neonatal morbidity and mortality in the preterm population. Infants perceived to be at increased risk for early-onset sepsis are often treated empirically with broad-spectrum antibiotics while awaiting confirmatory blood cultures, despite an overall incidence of early-onset sepsis of 2-3% among extremely-low-birthweight (ELBW) infants. Recent observational studies associate perinatal antibiotic use with an increased incidence of necrotizing enterocolitis, late-onset sepsis, and mortality among ELBW infants. Given currently available data and variability in clinical practice, we designed a prospective multi-institutional randomized controlled trial to determine the safety of early antibiotic use in ELBW infants. METHODS The NICU Antibiotics and Outcomes (NANO) trial is a multicenter, double-blinded, randomized controlled trial. A sample of 802 ELBW preterm infants will undergo web-based stratified block randomization to receive empiric antibiotics (EA; ampicillin and gentamicin) or placebo during routine evaluation for early-onset sepsis. Participating sites will use preexisting institutional protocols for antibiotic dosage and duration. Infants born at participating sites with a gestational age of 29 weeks or less are eligible for enrollment. Exclusion criteria include maternal intrauterine infection, hemodynamic or respiratory instability, delivery by caesarean section for maternal indications without labor or prolonged rupture of membranes, and prior administration of antibiotics. The primary outcome is the composite incidence of necrotizing enterocolitis, late-onset sepsis, or death during participants' index hospitalization. Maternal and infant samples will be collected longitudinally and assessed for differences in microbiome composition and diversity. DISCUSSION The NANO trial is designed to compare the rate of adverse outcomes of EA use at birth versus placebo in ELBW preterm infants. If EA at birth worsens clinical outcomes, then the results of the trial may help providers decrease antibiotic utilization in the NICU and subsequently decrease the incidence of complications associated with early antibiotic use in ELBW infants. If we instead find that EA improve outcomes, then the trial will validate a longstanding clinical practice that has not previously been supported by high-quality data. Future studies will assess long-term clinical and microbial outcomes in infants who received empiric antibiotics following delivery. TRIAL REGISTRATION Trial registration data: June 25, 2019 NCT03997266 .
Collapse
Affiliation(s)
- Michael J. Morowitz
- Division of Pediatric General and Thoracic Surgery, University of Pittsburgh School of Medicine, Children’s Hospital of Pittsburgh of UPMC, Rangos Research Center 6th Floor, 4401 Penn Avenue, Pittsburgh, PA 15224 USA
| | - Anup C. Katheria
- Division of Pediatrics, Sharp Mary Birch Hospital for Women & Newborns, San Diego, CA 92123 USA
| | - Richard A. Polin
- Department of Pediatrics, Columbia University, New York, NY 10032 USA
| | - Elizabeth Pace
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - David T. Huang
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Chung-Chou H. Chang
- Department of General Internal Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Johathan G. Yabes
- Department of General Internal Medicine, University of Pittsburgh, Pittsburgh, USA
| |
Collapse
|
19
|
Kahn DJ, Perkins BS, Barrette CE, Godin R. Reducing Antibiotic Use in a Level III and Two Level II Neonatal Intensive Care Units Targeting Prescribing Practices for Both Early and Late-onset Sepsis: A Quality Improvement Project. Pediatr Qual Saf 2022; 7:e555. [PMID: 35720871 PMCID: PMC9197376 DOI: 10.1097/pq9.0000000000000555] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/17/2022] [Indexed: 11/26/2022] Open
Abstract
Introduction Variation in antibiotic (ATB) use exists between neonatal intensive care units (NICUs) without demonstrated benefit to outcomes tested. Studies show that early-onset sepsis occurs in up to 2% of NICU patients, yet antibiotics (ABX) were started in over 50% of neonates admitted to our NICUs. An internal audit identified variations in prescribing practices and excessive use of ABX. As a result, we introduced ATB stewardship to our NICUs in 2015 to reduce unnecessary usage of these medications. Methods We used standard quality improvement methodology utilizing multiple iterative plan-do-study-act cycles during a 6-year project to test various interventions aimed at using ABX wisely. Specifically, our goals were to reduce ABX on admission (AA), percent of patients who continued on ABX beyond 72 hours of life (AC), and ATB utilization rate in our 3 NICUs by 28% for each metric. Interventions implemented included the development of an ATB stewardship program consisting of a multidisciplinary team that met regularly, creation of tools and guidelines for evaluations of sepsis and ATB use, universal use of the neonatal early-onset sepsis calculator for all newborns 34 weeks and older gestational age, education regarding noninitiation of ABX for maternal indications in clinically well newborns, and discontinuation within 48 hours for asymptomatic newborns with negative blood cultures. Results AA, AC, and ATB utilization rate decreased by 34.1%, 45.3%, and 34.9%, respectively, in our 3 NICUs. Conclusions By introducing ATB stewardship in our NICUs, we exceeded our predetermined goal of significantly reducing ATB usage.
Collapse
Affiliation(s)
- Doron J. Kahn
- From the Division of Neonatology, Joe DiMaggio Children’s Hospital, Hollywood, FL
- Mednax Services, Inc., Sunrise, FL
| | - Beckett S. Perkins
- From the Division of Neonatology, Joe DiMaggio Children’s Hospital, Hollywood, FL
- Mednax Services, Inc., Sunrise, FL
| | - Claire E. Barrette
- From the Division of Neonatology, Joe DiMaggio Children’s Hospital, Hollywood, FL
- Mednax Services, Inc., Sunrise, FL
| | - Robert Godin
- Department of Pharmacy, Joe DiMaggio Children’s Hospital, Hollywood, FL
| |
Collapse
|
20
|
Navarro-Garcia de Llano JP, Ponce-Ayala A, Ceja-Espinosa A, Vera-Márquez CD, Mendizabal-Guerra R. Wound Dehiscence After Occipital Encephalocele Surgical Repair in a Neonate: Management Alternative. Cureus 2022; 14:e23685. [PMID: 35505708 PMCID: PMC9055975 DOI: 10.7759/cureus.23685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2022] [Indexed: 11/25/2022] Open
Abstract
Encephaloceles are congenital malformations of the neural tube, mostly located in the occipital region in the Western world. Its presence is related to many complications, among which cognitive impairment and death are the most important. The diagnosis is usually made in the prenatal period, but sometimes due to poor control, this is not feasible. Surgery is required as early as possible to prevent further damage. Sometimes we can face complications related to the procedure, such as wound dehiscence, which has been the aim of this work. Many different types of treatments have been proposed for this complication: nevertheless, they result in invasive management. We present the case of a neonate's wound dehiscence, managed with potable water washes and a correct sterile technique, shown to be safe, reduce the in-patient costs, as well as improve the patient’s and their family's quality of life (QoL).
Collapse
|
21
|
Untargeted 1H-NMR Urine Metabolomic Analysis of Preterm Infants with Neonatal Sepsis. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12041932] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
One of the most critical medical conditions occurring after preterm birth is neonatal sepsis, a systemic infection with high rates of morbidity and mortality, chiefly amongst neonates hospitalized in Neonatal Intensive Care Units (NICU). Neonatal sepsis is categorized as early-onset sepsis (EOS) and late-onset sepsis (LOS) regarding the time of the disease onset. The accurate early diagnosis or prognosis have hurdles to overcome, since there are not specific clinical signs or laboratory tests. Herein, a need for biomarkers presents, with the goals of aiding accurate medical treatment, reducing the clinical severity of symptoms and the hospitalization time. Through nuclear magnetic resonance (NMR) based metabolomics, we aim to investigate the urine metabolomic profile of septic neonates and reveal those metabolites which could be indicative for an initial discrimination between the diseased and the healthy ones. Multivariate and univariate statistical analysis between NMR spectroscopic data of urine samples from neonates that developed EOS, LOS, and a healthy control group revealed a discriminate metabolic profile of septic newborns. Gluconate, myo-inositol, betaine, taurine, lactose, glucose, creatinine and hippurate were the metabolites highlighted as significant in most comparisons.
Collapse
|
22
|
Ting JY, Autmizguine J, Dunn MS, Choudhury J, Blackburn J, Gupta-Bhatnagar S, Assen K, Emberley J, Khan S, Leung J, Lin GJ, Lu-Cleary D, Morin F, Richter LL, Viel-Thériault I, Roberts A, Lee KS, Skarsgard ED, Robinson J, Shah PS. Practice Summary of Antimicrobial Therapy for Commonly Encountered Conditions in the Neonatal Intensive Care Unit: A Canadian Perspective. Front Pediatr 2022; 10:894005. [PMID: 35874568 PMCID: PMC9304938 DOI: 10.3389/fped.2022.894005] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/13/2022] [Indexed: 11/29/2022] Open
Abstract
Neonates are highly susceptible to infections owing to their immature cellular and humoral immune functions, as well the need for invasive devices. There is a wide practice variation in the choice and duration of antimicrobial treatment, even for relatively common conditions in the NICU, attributed to the lack of evidence-based guidelines. Early decisive treatment with broad-spectrum antimicrobials is the preferred clinical choice for treating sick infants with possible bacterial infection. Prolonged antimicrobial exposure among infants without clear indications has been associated with adverse neonatal outcomes and increased drug resistance. Herein, we review and summarize the best practices from the existing literature regarding antimicrobial use in commonly encountered conditions in neonates.
Collapse
Affiliation(s)
- Joseph Y Ting
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Julie Autmizguine
- Division of Infectious Diseases, Department of Pediatrics, Université de Montreal, Montreal, QC, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Michael S Dunn
- Division of Neonatology, Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Julie Choudhury
- Department of Pharmacy, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Julie Blackburn
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montreal, Montreal, QC, Canada
| | - Shikha Gupta-Bhatnagar
- Division of Neonatology, Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| | - Katrin Assen
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Julie Emberley
- Division of Neonatology, Department of Pediatrics, University of Manitoba, Winnipeg, MB, Canada
| | - Sarah Khan
- Department of Microbiology, McMaster University, Hamilton, ON, Canada
| | - Jessica Leung
- Department of Pediatrics, University of Massachusetts, Worcester, MA, United States
| | - Grace J Lin
- School of Medicine, Queen's University, Kingston, ON, Canada
| | | | - Frances Morin
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Lindsay L Richter
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Isabelle Viel-Thériault
- Division of Infectious Diseases, Department of Pediatrics, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Ashley Roberts
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Kyong-Soon Lee
- Division of Neonatology, Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Erik D Skarsgard
- Division of Pediatric Surgery, Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Joan Robinson
- Division of Infectious Diseases, Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Prakesh S Shah
- Division of Neonatology, Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
23
|
Kilpatrick R, Scarrow E, Hornik C, Greenberg RG. Neonatal invasive candidiasis: updates on clinical management and prevention. THE LANCET CHILD & ADOLESCENT HEALTH 2021; 6:60-70. [PMID: 34672994 DOI: 10.1016/s2352-4642(21)00272-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/30/2021] [Accepted: 08/13/2021] [Indexed: 01/15/2023]
Abstract
Neonatal invasive candidiasis is an important cause of morbidity and mortality in preterm infants. The incidence of invasive candidiasis in this population has been declining in high-income settings, largely due to preventive measures, although there are still considerable variations in incidence between health-care centres. Surveillance data and large, multicentre studies in lower-income settings are not available, although preventive measures in these settings have been shown to decrease the incidence of neonatal invasive candidiasis. Understanding risk factors and pathogenesis are key to the prevention of invasive candidiasis. The difficulty of a definitive diagnosis of invasive candidiasis and the high risk for death or substantial neurodevelopmental impairment, even with appropriate treatment, further increase the need for effective preventive measures. In this Review, we examine the pathogenesis, clinical presentation, and diagnosis of invasive candidiasis. We highlight commonly used and emerging preventive and prophylactic measures, including standardised central line care, antibiotic stewardship, antifungal prophylaxis, and probiotics. Finally, we provide updates on empirical treatment, clinical management in confirmed cases of invasive candidiasis, and antifungal pharmacotherapy.
Collapse
Affiliation(s)
- Ryan Kilpatrick
- Department of Pediatrics, Duke Clinical Research Institute, Durham, NC, USA
| | - Evelyn Scarrow
- Department of Pediatrics, Duke Clinical Research Institute, Durham, NC, USA
| | - Chi Hornik
- Department of Pediatrics, Duke Clinical Research Institute, Durham, NC, USA
| | - Rachel G Greenberg
- Department of Pediatrics, Duke Clinical Research Institute, Durham, NC, USA.
| |
Collapse
|
24
|
Tran VL, Greenberg J, Nuibe A. Evaluating the Incidence of Sepsis Post-Central Catheter Removal When Using Prophylactic Vancomycin in the Neonatal Intensive Care Unit. J Pediatr Pharmacol Ther 2021; 26:728-733. [PMID: 34588937 DOI: 10.5863/1551-6776-26.7.728] [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: 10/15/2020] [Accepted: 12/14/2020] [Indexed: 11/11/2022]
Abstract
OBJECTIVE With no consensus, the practice of using prophylactic antibiotics prior to central venous catheter (CVC) removal in NICU patients remains controversial. The objective of this study was to compare the incidence of sepsis post-CVC removal in those who received a dose of vancomycin prophylactically with those who did not. METHODS This single-center, retrospective chart review included NICU patients who had CVCs removed. Patients were excluded if they had a confirmed or suspected infection at the time of CVC removal or if the indwelling CVC was removed prior to 30 days from insertion. Primary outcome was the occurrence of a sepsis evaluation within 72 hours from CVC removal. Secondary outcomes included the development of acute kidney injury, source and identification of positive cultures, time to onset of suspected or confirmed sepsis, and the appropriate administration of intravenous vancomycin. RESULTS Eighty-two CVC removals received prophylactic vancomycin (P-VAN), and 22 CVCs did not receive prophylactic vancomycin (NP-VAN) prior to CVC removal. There were no significant differences in patient demographics between groups and median duration of indwelling CVC. Two clinical sepsis evaluations occurred in the P-VAN group compared with none in the NP-VAN group. Of all the P-VAN CVC removals, 45 (55%) received vancomycin appropriately. There were no statistical differences in all evaluated secondary outcomes. CONCLUSIONS Vancomycin administered prophylactically prior to CVC removal did not reduce the number of subsequent clinical sepsis evaluations or infections in NICU patients.
Collapse
|
25
|
Begnaud CM, Lemoine J, Broussard L, Rholdon R, Doshi H. A Quality Improvement Project to Reduce Antibiotic Utilization and Ancillary Laboratory Tests in the Appraisal of Early-Onset Sepsis in the NICU. J Pediatr Nurs 2021; 60:215-222. [PMID: 34273817 DOI: 10.1016/j.pedn.2021.06.012] [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: 03/29/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Diagnosis and treatment of early-onset sepsis (EOS) of the newborn remains a controversial issue among providers due to the non-infectious symptomology which exists in the newborn period. METHODS Pre/post interventional quality improvement project in a level III NICU to reduce antibiotic utilization and ancillary laboratory tests with the introduction of an evidence-based guideline for the evaluation of EOS in the NICU. RESULTS Primary outcome measures include mean number of empiric antibiotic treatment days and utilization rate (AUR), number of laboratory tests ordered, and incidence of unwarranted antibiotic therapy beyond the 48-h rule out period. Mean empiric antibiotic treatment days decreased from 2.94 to 1.58 days and overall antibiotic use decreased from 73.7% to 57.1%. Likewise, the mean AUR decreased from 212.5 to 147.6 days of therapy per 1000 patient days. There was an 86% decline in the number of ancillary tests and unwarranted antibiotic use beyond 48- h was reduced by 74%. DISCUSSION Guidelines for EOS of the newborn should include a thorough baseline evaluation of the drivers of antibiotic use to create an evidence-based foundation. Reducing unnecessary antibiotic use and EOS evaluations in a safe and effective manner have the potential to lower consumer and healthcare expenditures while improving the long-term health of the newborn in the NICU. CONCLUSIONS These findings emphasize the importance of implementing an evidence-based protocol for antibiotic stewardship in the NICU. With further research there is the potential to improve the healthcare of newborns while reducing expenditures in a safe, effective evaluation of EOS in the newborn population.
Collapse
Affiliation(s)
- C Martin Begnaud
- Our Lady of Lourdes Women's and Children's Hospital and Pediatrix Medical Group, LA, United States of America; University of Louisiana at Lafayette, College of Nursing and Allied Health Professions, LA, United States of America.
| | - Jennifer Lemoine
- University of Louisiana at Lafayette, College of Nursing and Allied Health Professions, LA, United States of America
| | - Lisa Broussard
- University of Louisiana at Lafayette, College of Nursing and Allied Health Professions, LA, United States of America
| | - Roger Rholdon
- University of Louisiana at Lafayette, College of Nursing and Allied Health Professions, LA, United States of America
| | - Harshit Doshi
- Golisano Childrens Hospital of Southwest FL, FL, United States of America
| |
Collapse
|
26
|
Lack of utility of tracheal aspirates in the management of suspected pneumonia in intubated neonates. Infect Control Hosp Epidemiol 2021; 41:660-665. [PMID: 32209148 DOI: 10.1017/ice.2020.57] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES To evaluate the utility of tracheal aspirates in suspected pneumonia in intubated neonates and to measure the burden of antibiotic use associated with a positive tracheal aspirate culture. DESIGN Retrospective cohort study between January 2016 and December 2017. SETTING A level IV neonatal intensive care unit (NICU). PATIENTS Intubated patients with a tracheal aspirate culture. METHODS Data on temporally associated clinical measures of illness, laboratory and radiographic testing, and clinical demographic information were analyzed. RESULTS Positive tracheal aspirate cultures were associated with lower birth weight and a normal immature to total neutrophil ratio (I/T ratio). Positive tracheal aspirates were not significantly associated with clinical, laboratory, or radiographic markers used in clinical practice to screen for infection. Despite the lack of positive clinical associations, a positive tracheal aspirate culture was associated with increased risk of prolonged antibiotic exposure. CONCLUSION These findings suggest that positive tracheal aspirates do not always represent clinical infection and may result in unnecessary antibiotic exposure.
Collapse
|
27
|
Korang SK, Safi S, Nava C, Gordon A, Gupta M, Greisen G, Lausten-Thomsen U, Jakobsen JC. Antibiotic regimens for early-onset neonatal sepsis. Cochrane Database Syst Rev 2021; 5:CD013837. [PMID: 33998666 PMCID: PMC8127574 DOI: 10.1002/14651858.cd013837.pub2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
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. Possibly 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. The last Cochrane Review was updated in 2004. Given the clinical importance, an updated systematic review assessing the effects of different antibiotic regimens for early-onset neonatal sepsis is needed. OBJECTIVES To assess the beneficial and harmful effects of different antibiotic regimens for early-onset neonatal sepsis. SEARCH METHODS We searched the following electronic databases: CENTRAL (2020, Issue 8); Ovid MEDLINE; Embase Ovid; CINAHL; LILACS; Science Citation Index EXPANDED and Conference Proceedings Citation Index - Science on 12 March 2021. We 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 early-onset neonatal sepsis. We included participants from birth to 72 hours of life at randomisation. 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 (865 participants). All trials were at high risk of bias. The certainty of the evidence according to GRADE was very low. The included trials assessed five different comparisons of antibiotics. We did not conduct any meta-analyses due to lack of relevant data. Of the five included trials one trial compared ampicillin plus gentamicin with benzylpenicillin plus gentamicin; one trial compared piperacillin plus tazobactam with amikacin; one trial compared ticarcillin plus clavulanic acid with piperacillin plus gentamicin; one trial compared piperacillin with ampicillin plus amikacin; and one trial compared ceftazidime with benzylpenicillin plus gentamicin. 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. Large RCTs assessing different antibiotic regimens in early-onset neonatal sepsis with low risk 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
| | - Adrienne Gordon
- Neonatology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Munish Gupta
- Neonatology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, 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
- 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
|
28
|
Simon A, Zemlin M, Geipel M, Gärtner B, Armann J, Meyer S. [Infection prevention in neonatal intensive care units]. DER GYNAKOLOGE 2021; 54:428-434. [PMID: 33967301 PMCID: PMC8094126 DOI: 10.1007/s00129-021-04804-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 04/09/2021] [Indexed: 12/24/2022]
Abstract
In diesem Beitrag werden einige Besonderheiten der Infektionsprävention bei intensivmedizinisch behandelten Früh und Neugeborenen dargestellt. Ergänzend finden sich Hinweise zum krankenhaushygienischen Management der SARS-CoV-2(„severe acute respiratory syndrome coronavirus 2“)-Pandemie und zur Antibiotic Stewardship in der neonatologischen Intensivmedizin.
Collapse
Affiliation(s)
- Arne Simon
- Pädiatrische Onkologie und Hämatologie, Universitätsklinikum des Saarlandes, Kirrberger Str. 9, Geb. 9, 66421 Homburg/Saar, Deutschland
| | - Michael Zemlin
- Allgemeine Pädiatrie und Neonatologie, Universitätsklinikum des Saarlandes, Homburg/Saar, Deutschland
| | - Martina Geipel
- Allgemeine Pädiatrie und Neonatologie, Universitätsklinikum des Saarlandes, Homburg/Saar, Deutschland
| | - Barbara Gärtner
- Institut für Medizinische Mikrobiologie und Hygiene (IMMH), Universität und Universitätsklinikums des Saarlandes, Homburg/Saar, Deutschland
| | - Jakob Armann
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Deutschland
| | - Sascha Meyer
- Allgemeine Pädiatrie und Neonatologie, Universitätsklinikum des Saarlandes, Homburg/Saar, Deutschland
| |
Collapse
|
29
|
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
|
30
|
Pyle AK, Cantey JB, Brown LS, Heyne RJ, Wozniak PS, Heyne E, Holcombe A, Brammer EM, Lair CS, Sánchez PJ. Antibiotic exposure and growth patterns in preterm, very low birth weight infants. Matern Health Neonatol Perinatol 2021; 7:7. [PMID: 33514436 PMCID: PMC7846997 DOI: 10.1186/s40748-021-00126-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Antibiotic exposure in term infants has been associated with later obesity. Premature, very-low-birth-weight (birth weight ≤ 1500 g) infants in the neonatal intensive care unit frequently are exposed to antibiotics. Our hypothesis was that in preterm infants, there is a positive linear and dose-dependent relationship between antibiotic exposure and growth from birth through 12 months' corrected age. METHODS Retrospective analysis of prospectively collected data of all antibiotic use among inborn, preterm (≤32 weeks' gestation), very-low-birth-weight infants admitted to the neonatal intensive care unit at Parkland Memorial Hospital and followed in the Low Birth Weight Clinic at Children's Medical Center, Dallas, TX. Antibiotic use was quantified by days of therapy which was compared with weight and length parameters at birth, 36 weeks' postmenstrual age, and 2, 4, 6, and 12 months' corrected age. The change in weight and length z-scores from birth to all subsequent age points was calculated. Stepwise multivariate regression analysis was performed to determine predictors of weight, length, and weight-for-length delta z-scores from birth to each subsequent age point. RESULTS During the 18-month study, 161 infants received a median of 11 (IQR, 5.5-19.5) antibiotic days of therapy which was not associated with weight or length delta z-scores from birth through 12 months' corrected age. CONCLUSION Association of prolonged antibiotic use and neonatal morbidities and mortality may override the potential association with increased weight gain in the NICU and beyond.
Collapse
Affiliation(s)
- Alaina K Pyle
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, Division of Neonatology, Connecticut Children's Medical Center, Hartford, CT, USA
| | - Joseph B Cantey
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, Divisions of Neonatology and Pediatric Infectious Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - L Steven Brown
- Health System Research, Parkland Health and Hospital System, Dallas, TX, USA
| | - Roy J Heyne
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Phillip S Wozniak
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pediatrics, Divisions of Neonatology and Pediatric Infectious Diseases, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| | | | | | | | - Cheryl S Lair
- Nutrition Services, Parkland Health and Hospital System, Dallas, TX, USA
| | - Pablo J Sánchez
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Pediatrics, Divisions of Neonatology and Pediatric Infectious Diseases, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
31
|
Russell JT, Lauren Ruoss J, de la Cruz D, Li N, Bazacliu C, Patton L, McKinley KL, Garrett TJ, Polin RA, Triplett EW, Neu J. Antibiotics and the developing intestinal microbiome, metabolome and inflammatory environment in a randomized trial of preterm infants. Sci Rep 2021; 11:1943. [PMID: 33479274 PMCID: PMC7820285 DOI: 10.1038/s41598-021-80982-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/01/2021] [Indexed: 01/29/2023] Open
Abstract
Antibiotic use in neonates can have detrimental effects on the developing gut microbiome, increasing the risk of morbidity. A majority of preterm neonates receive antibiotics after birth without clear evidence to guide this practice. Here microbiome, metabolomic, and immune marker results from the routine early antibiotic use in symptomatic preterm Neonates (REASON) study are presented. The REASON study is the first trial to randomize symptomatic preterm neonates to receive or not receive antibiotics in the first 48 h after birth. Using 16S rRNA sequencing of stool samples collected longitudinally for 91 neonates, the effect of such antibiotic use on microbiome diversity is assessed. The results illustrate that type of nutrition shapes the early infant gut microbiome. By integrating data for the gut microbiome, stool metabolites, stool immune markers, and inferred metabolic pathways, an association was discovered between Veillonella and the neurotransmitter gamma-aminobutyric acid (GABA). These results suggest early antibiotic use may impact the gut-brain axis with the potential for consequences in early life development, a finding that needs to be validated in a larger cohort.Trial Registration This project is registered at clinicaltrials.gov under the name "Antibiotic 'Dysbiosis' in Preterm Infants" with trial number NCT02784821.
Collapse
Affiliation(s)
- Jordan T Russell
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| | - J Lauren Ruoss
- Division of Neonatology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Diomel de la Cruz
- Division of Neonatology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Nan Li
- Division of Neonatology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Catalina Bazacliu
- Division of Neonatology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Laura Patton
- Division of Neonatology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Kelley Lobean McKinley
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Richard A Polin
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Eric W Triplett
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA.
| | - Josef Neu
- Division of Neonatology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
32
|
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
|
33
|
Korang SK, Safi S, Gupta M, Gordon A, Greisen G, Lausten-Thomsen U, Jakobsen JC. Antibiotic regimens for early-onset neonatal sepsis. Hippokratia 2021. [DOI: 10.1002/14651858.cd013837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
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
|
34
|
Berezin E, Manjabosco ABL, Michelin L, Gallacci C. Antimicrobial use in a neonatal intensive care unit during a 4-year period. J Clin Neonatol 2021. [DOI: 10.4103/jcn.jcn_143_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
35
|
Al-Haqan A, Boswihi SS, Pathan S, Udo EE. Antimicrobial resistance and virulence determinants in coagulase-negative staphylococci isolated mainly from preterm neonates. PLoS One 2020; 15:e0236713. [PMID: 32750089 PMCID: PMC7402503 DOI: 10.1371/journal.pone.0236713] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 07/13/2020] [Indexed: 12/22/2022] Open
Abstract
Coagulase-negative staphylococci (CoNS) are the most common isolates from blood culture in neonates resulting in high mortality and morbidity. This study investigated CoNS obtained from blood cultures of neonates for antibiotic resistance and virulence factors, and possible association with inflammatory response (C-reactive protein). A total of 93 CoNS isolates were collected from 76 blood cultures of neonates at the Maternity hospital in Kuwait in a six-month period and investigated for susceptibility to antibiotics, carriage of staphylococcal cassette chromosome mec (SCCmec), and virulence-associated genes. The 93 CoNS isolates consisted of S. epidermidis (76; 81.7%), S. capitis (12; 12.9%), S. hominis (2; 2.1%), S. warneri (2; 2.1%) and S. haemolyticus (1; 1.0%). Eighty-six (92.4%) of the isolates were resistant to cefoxitin (MR-CoNS) while 49 (52.7%) expressed multi-antibiotic resistance. The methicillin-resistant isolates (MR-CoNS) carried SCCmec III, SCCmec IVa and four combinations of SCCmec types including SCCmec types I+IVa (one S. warneri and 25 S. epidermidis isolates), types I+III (one S. epidermidis isolate), types III+IVa (six S. epidermidis isolates) and types I+III+IVa (one S. epidermidis isolate). The most common virulence-related genes were icaC, seb, arc detected in 69.7%, 60.5%, 40.8% of the isolates respectively. Two isolates were positive for tst1. No association between C-reactive protein and antibiotic resistance or virulence factors was established. This study revealed that S. epidermidis carrying different SCCmec genetic elements, was the dominant CoNS species isolated from neonatal blood cultures with 90.3% and 36.6% of the isolates positive for genes for biofilm and ACME production respectively.
Collapse
Affiliation(s)
- Aishah Al-Haqan
- Microbiology Laboratory, Maternity Hospital, Kuwait city, Kuwait
| | - Samar S. Boswihi
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait city, Kuwait
| | - Seema Pathan
- Microbiology Laboratory, Maternity Hospital, Kuwait city, Kuwait
| | - Edet E. Udo
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait city, Kuwait
- * E-mail:
| |
Collapse
|
36
|
Sola A, Mir R, Lemus L, Fariña D, Ortiz J, Golombek S. Suspected Neonatal Sepsis: Tenth Clinical Consensus of the Ibero-American Society of Neonatology (SIBEN). Neoreviews 2020; 21:e505-e534. [PMID: 32737171 DOI: 10.1542/neo.21-8-e505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Suspected neonatal sepsis is one of the most common diagnoses made in newborns (NBs), but very few NBs actually have sepsis. There is no international consensus to clearly define suspected neonatal sepsis, but each time that this suspected diagnosis is assumed, blood samples are taken, venous accesses are used to administer antibiotics, and the mother-child pair is separated, with prolonged hospital stays. X-rays, urine samples, and a lumbar puncture are sometimes taken. This is of concern, as generally <10% and no more than 25%-30% of the NBs in whom sepsis is suspected have proven neonatal sepsis. It seems easy to start antibiotics with suspicion of sepsis, but stopping them is difficult, although there is little or no support to maintain them. Unfortunately, the abuse of antibiotics in inpatient and outpatient NBs is foolish. Its negative impact on neonatal health and the economy is a public health problem of epidemiological and even epidemic proportions. This manuscript is a shortened version of the 10th Clinical Consensus of the Ibero-American Society of Neonatology (SIBEN) on suspected neonatal sepsis at the end of 2018, updated with publications from its completion to February 2020. This manuscript describes useful strategies for everyday neonatal practice when neonatal sepsis is suspected, along with important aspects about the indisputable value of clinical evaluation of the NB and about obtaining and interpreting blood cultures, urine cultures, and other cultures. Likewise, the low value of laboratory tests in suspected neonatal sepsis is demonstrated with evidence and clinical recommendations are made on the appropriate use of antibiotics.
Collapse
Affiliation(s)
- Augusto Sola
- Medical Director, Ibero-American Society of Neonatology, Wellington, FL
| | - Ramón Mir
- Neonatology Department Chief in Hospital de Clìnicas Universidad Nacional de Asunciòn, Paraguay
| | - Lourdes Lemus
- Departamento de Neonatología, Hospital de Pediatría UMAE, Instituto Mexicano del Seguro Social, Guadalajara Jalisco, México
| | - Diana Fariña
- Director of the Neonatal Intensive Care Unit, Hospital de Pediatría, Buenos Aires, Argentina
| | - Javier Ortiz
- Ángeles del Pedregal Hospital, Mexico City, Mexico
| | - Sergio Golombek
- Joseph M. Sanzari Children's Hospital at Hackensack University Medical Center, Hackensack, NJ
| | | |
Collapse
|
37
|
Raju V, Pournami F, Nandakumar A, Prabhakar J, Nair PMC, Jain N. Improving Microbe Detection and Optimizing Antibiotic Use in Neonatal Sepsis With Multiplex Polymerase Chain Reaction. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2020. [DOI: 10.1097/ipc.0000000000000836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
38
|
Simeonova M, Piszczek J, Hoi S, Harder C, Pelligra G. Evaluation of compliance with the 2017 Canadian Paediatric Society Position Statement for the management of newborns at risk for early-onset sepsis: A retrospective cohort study. Paediatr Child Health 2020; 26:e152-e157. [PMID: 33936345 DOI: 10.1093/pch/pxaa042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/26/2020] [Indexed: 11/14/2022] Open
Abstract
Introduction Due to the nonspecific clinical presentation, clinicians often empirically treat newborns at risk of early-onset sepsis (EOS). Recently, the Canadian Paediatric Society (CPS) published updated recommendations that promote a more judicious approach to EOS management. Objective To examine the compliance with the CPS statement at a tertiary perinatal site and characterize the types of deviations. Methods A retrospective chart review was conducted for all term and late pre-term newborns at risk for sepsis, between January 1 and June 30, 2018. The prevalence of newborns with EOS risk factors was measured during the first month. Management strategies for eligible newborns during the 6-month period were compared to the CPS recommendations to establish the rate of noncompliance. The type of noncompliance, readmission rate, and rate of culture-positive EOS were examined. Results In the first month, 29% (66 of 228) of newborns had EOS risk factors. Among the 100 newborns born in the 6-month period for whom the CPS recommendations apply, 47 (47%) received noncompliant management. Of those, 51% (N=24) had inappropriately initiated investigations, 17% (N=8) had inappropriate antibiotics, and 32% (N=15) had both. The rate of readmission for a septic workup was 1.6% (N= 2). None had culture-positive sepsis while admitted. Conclusion A large proportion of term and late preterm newborns (29%) had EOS risk factors, but none had culture-confirmed EOS. The rate of noncompliance with the CPS recommendations was high (47%), mainly due to overzealous management. Future initiatives should aim at increasing compliance, particularly in newborns at lower EOS risk.
Collapse
Affiliation(s)
- Marina Simeonova
- Department of Pharmacy, Victoria General Hospital, Victoria, British Columbia.,Department of Pharmacy, Royal Jubilee Hospital, Victoria, British Columbia.,Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia
| | - Jolanta Piszczek
- Department of Pharmacy, Royal Jubilee Hospital, Victoria, British Columbia
| | - Sannifer Hoi
- Department of Pharmacy, Victoria General Hospital, Victoria, British Columbia.,Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia
| | - Curtis Harder
- Department of Pharmacy, Victoria General Hospital, Victoria, British Columbia.,Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, British Columbia
| | - Gustavo Pelligra
- Department of Maternity Care and Pediatrics, Victoria General Hospital, Victoria, British Columbia.,Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia
| |
Collapse
|
39
|
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: 15] [Impact Index Per Article: 2.5] [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
|
40
|
Raffaeli G, Pokorna P, Allegaert K, Mosca F, Cavallaro G, Wildschut ED, Tibboel D. Drug Disposition and Pharmacotherapy in Neonatal ECMO: From Fragmented Data to Integrated Knowledge. Front Pediatr 2019; 7:360. [PMID: 31552205 PMCID: PMC6733981 DOI: 10.3389/fped.2019.00360] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/16/2019] [Indexed: 12/27/2022] Open
Abstract
Extracorporeal membrane oxygenation (ECMO) is a lifesaving support technology for potentially reversible neonatal cardiac and/or respiratory failure. As the survival and the overall outcome of patients rely on the treatment and reversal of the underlying disease, effective and preferentially evidence-based pharmacotherapy is crucial to target recovery. Currently limited data exist to support the clinicians in their every-day intensive care prescribing practice with the contemporary ECMO technology. Indeed, drug dosing to optimize pharmacotherapy during neonatal ECMO is a major challenge. The impact of the maturational changes of the organ function on both pharmacokinetics (PK) and pharmacodynamics (PD) has been widely established over the last decades. Next to the developmental pharmacology, additional non-maturational factors have been recognized as key-determinants of PK/PD variability. The dynamically changing state of critical illness during the ECMO course impairs the achievement of optimal drug exposure, as a result of single or multi-organ failure, capillary leak, altered protein binding, and sometimes a hyperdynamic state, with a variable effect on both the volume of distribution (Vd) and the clearance (Cl) of drugs. Extracorporeal membrane oxygenation introduces further PK/PD perturbation due to drug sequestration and hemodilution, thus increasing the Vd and clearance (sequestration). Drug disposition depends on the characteristics of the compounds (hydrophilic vs. lipophilic, protein binding), patients (age, comorbidities, surgery, co-medications, genetic variations), and circuits (roller vs. centrifugal-based systems; silicone vs. hollow-fiber oxygenators; renal replacement therapy). Based on the potential combination of the above-mentioned drug PK/PD determinants, an integrated approach in clinical drug prescription is pivotal to limit the risks of over- and under-dosing. The understanding of the dose-exposure-response relationship in critically-ill neonates on ECMO will enable the optimization of dosing strategies to ensure safety and efficacy for the individual patient. Next to in vitro and clinical PK data collection, physiologically-based pharmacokinetic modeling (PBPK) are emerging as alternative approaches to provide bedside dosing guidance. This article provides an overview of the available evidence in the field of neonatal pharmacology during ECMO. We will identify the main determinants of altered PK and PD, elaborate on evidence-based recommendations on pharmacotherapy and highlight areas for further research.
Collapse
Affiliation(s)
- Genny Raffaeli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Pavla Pokorna
- Department of Pediatrics—ICU, General University Hospital, 1st Faculty of Medicine Charles University, Prague, Czechia
- Department of Pharmacology, General University Hospital, 1st Faculty of Medicine Charles University, Prague, Czechia
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Karel Allegaert
- Division of Neonatology, Department of Pediatrics, Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Fabio Mosca
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Giacomo Cavallaro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Enno D. Wildschut
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| |
Collapse
|
41
|
Audit of Antibiotic Prescribing Practices for Neonatal Sepsis and Measurement of Outcome in New Born Unit at Kenyatta National Hospital. Int J Pediatr 2019; 2019:7930238. [PMID: 31182965 PMCID: PMC6512059 DOI: 10.1155/2019/7930238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/01/2019] [Indexed: 01/30/2023] Open
Abstract
Background Neonatal sepsis is a leading cause of morbidity and mortality globally. A high index of suspicion is required since features of sepsis are nonspecific. Auditing of antibiotic use is necessary to reduce misuse and minimise development of antibiotic resistance. Objectives To assess the antibiotic prescribing practices in NBU at KNH against recommended Kenyan guidelines for neonatal sepsis. In addition, outcome within 7 days was described. Methods This was a prospective audit of 320 neonates over a 2-month period at NBU of KNH. Data were collected using a structured questionnaire, stored in MS-EXCEL, and analysed using STATA. Results Documentation of perinatal risk factors and clinical features at admission and at the time of change of antibiotics was very poor. The rate of investigations to confirm infection was very low. Blood cultures were done only in 13 (4%) neonates on admission, while complete blood count and C reactive protein were done in 224 (70%) and 198 (62%), respectively. Appropriate antibiotics as per the Kenyan guidelines were prescribed in 313 (97.8%) of neonates on admission. However, these were not stopped at 48-72 hours for the 148 (53.62%) who had improved. Overall mortality was high in neonates at 80 (25%). Majority (55%) died within 48 hours. Mortality was high among preterm neonates; 70 (43.8%) died out of 160. Conclusion Overall documentation and investigations to confirm infection was poor. The continuation of antibiotics was inappropriate. Overall mortality was high especially in the first 48 hours of admission. To improve documentation, availability of a checklist on admission is recommended.
Collapse
|
42
|
Prävention von Gefäßkatheter-assoziierten Infektionen bei Früh- und Neugeborenen. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2019; 61:608-626. [PMID: 29671025 DOI: 10.1007/s00103-018-2718-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
43
|
Gkentzi D, Kortsalioudaki C, Cailes BC, Zaoutis T, Kopsidas J, Tsolia M, Spyridis N, Siahanidou S, Sarafidis K, Heath PT, Dimitriou G. Epidemiology of infections and antimicrobial use in Greek Neonatal Units. Arch Dis Child Fetal Neonatal Ed 2019; 104:F293-F297. [PMID: 29954881 DOI: 10.1136/archdischild-2018-315024] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/07/2018] [Accepted: 06/09/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To describe the epidemiology of neonatal infections and of antimicrobial use in Greek Neonatal Units (NNUs) in order to develop national, evidence-based guidelines on empiric antimicrobial use for neonatal sepsis in Greece. DESIGN Retrospective analysis of prospectively collected infection surveillance data from 2012 to 2015, together with a Point Prevalence Survey (PPS) on antimicrobial use and the collection of data on local empiric antimicrobial policies. SETTING 16 NNUs in Greece participating in the neonIN infection surveillance network PATIENTS: Newborns in participating NNUs who had a positive blood, cerebrospinal fluid or urine culture and were treated with at least 5 days of antibiotics. RESULTS 459 episodes were recorded in 418 infants. The overall incidence of infection was 50/1000 NNU-admissions. The majority of episodes were late-onset sepsis (LOS) (413, 90%). Coagulase-negative Staphylococci (80%) were the most common Gram-positive organisms causing LOS and Klebsiella spp (39%) the most common Gram-negative. Nearly half (45%) of the Klebsiella spp were resistant to at least one aminoglycoside. The PPS revealed that 196 of 484 (40%) neonates were on antimicrobials. The survey revealed wide variation in empiric antimicrobial policies for LOS. CONCLUSIONS This is the largest collection of data on the epidemiology of neonatal infections in Greece and on neonatal antimicrobial use. It provides the background for the development of national evidence-based guidelines. Continuous surveillance, the introduction of antimicrobial stewardship interventions and evidence-based guidelines are urgently required.
Collapse
Affiliation(s)
- Despoina Gkentzi
- Department of Paediatrics, Patras Medical School, University Hospital of Patras, Patras, Greece
| | | | | | - Theoklis Zaoutis
- The Stavros Niarchos Foundation-Collaborative Centre for Clinical Epidemiology and Outcomes Research, School of Medicine, University of Athens, Athens, Greece
| | - John Kopsidas
- The Stavros Niarchos Foundation-Collaborative Centre for Clinical Epidemiology and Outcomes Research, School of Medicine, University of Athens, Athens, Greece
| | - Maria Tsolia
- Paediatric Infectious Diseases Unit, Aglaia Kyriakou Children's Hospital, Athens, Greece
| | - Nikos Spyridis
- Paediatric Infectious Diseases Unit, Aglaia Kyriakou Children's Hospital, Athens, Greece
| | - Soultana Siahanidou
- Neonatal Unit, First Department of Pediatrics, Athens University Medical School, Athens, Greece
| | - Kosmas Sarafidis
- 1st Department of Neonatology and Neonatal Intensive Care Unit, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paul T Heath
- Paediatric Infectious Diseases Research Group, St George's University of London, London, UK
| | - Gabriel Dimitriou
- Department of Paediatrics, Patras Medical School, University Hospital of Patras, Patras, Greece
| | | |
Collapse
|
44
|
Gong CL, Dasgupta-Tsinikas S, Zangwill KM, Bolaris M, Hay JW. Early onset sepsis calculator-based management of newborns exposed to maternal intrapartum fever: a cost benefit analysis. J Perinatol 2019; 39:571-580. [PMID: 30692615 DOI: 10.1038/s41372-019-0316-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/05/2018] [Accepted: 12/27/2018] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To determine potential net monetary benefit of an early onset sepsis calculator-based approach for management of neonates exposed to maternal intrapartum fever, compared to existing guidelines. STUDY DESIGN We performed a cost-benefit analysis comparing two management approaches for newborns >34 weeks gestational age exposed to maternal intrapartum fever. Probabilities of sepsis and meningitis, consequences of infection and antibiotic use, direct medical costs, and indirect costs for long-term disability and mortality were considered. RESULTS A calculator-based approach resulted in a net monetary benefit of $3998 per infant with a 60% likelihood of net benefit in probabilistic sensitivity analysis. Our model predicted a 67% decrease in antibiotic use in the calculator arm. The absolute difference for all adverse clinical outcomes between approaches was ≤0.6%. CONCLUSIONS Compared to existing guidelines, a calculator-based approach for newborns exposed to maternal intrapartum fever yields a robust net monetary benefit, largely by preventing unnecessary antibiotic treatment.
Collapse
Affiliation(s)
- Cynthia L Gong
- University of Southern California Leonard D. Schaeffer Center for Health Policy & Economics, Los Angeles, CA, USA. .,Children's Hospital of Los Angeles, Fetal & Neonatal Institute, Los Angeles, CA, USA.
| | - Shom Dasgupta-Tsinikas
- Division of Pediatric Infectious Diseases, Harbor-UCLA Medical Center, Los Angeles, CA, USA
| | - Kenneth M Zangwill
- Division of Pediatric Infectious Diseases, Harbor-UCLA Medical Center, Los Angeles, CA, USA.,Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Los Angeles, CA, USA
| | - Michael Bolaris
- Division of Pediatric Infectious Diseases, Harbor-UCLA Medical Center, Los Angeles, CA, USA.,Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Los Angeles, CA, USA
| | - Joel W Hay
- University of Southern California Leonard D. Schaeffer Center for Health Policy & Economics, Los Angeles, CA, USA
| |
Collapse
|
45
|
Magréault S, Leroux S, Touati J, Storme T, Jacqz-Aigrain E. UPLC/MS/MS assay for the simultaneous determination of seven antibiotics in human serum-Application to pediatric studies. J Pharm Biomed Anal 2019; 174:256-262. [PMID: 31181488 DOI: 10.1016/j.jpba.2019.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 02/28/2019] [Accepted: 03/02/2019] [Indexed: 10/27/2022]
Abstract
A rapid and highly sensitive ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) assay was developed for quantification of 7 antibiotics in low sample volumes (50 μL): amoxicillin, azithromycin, cefotaxime, ciprofloxacin, meropenem, metronidazole and piperacillin, for both routine monitoring and pharmacokinetic studies. After protein precipitation by acetonitrile, the antibiotics were separated on an Acquity UPLC HSS T3 column (run time, 4 min). The mobile phase consisted of a mixture of (A) ammonium acetate (pH 2.4; 5 mM) and (B) acetonitrile acidified with 0.1% formic acid, delivered at 500 μl/min in a gradient elution mode. Total time run was 2.75 min. Ions were detected in the turbo-ion-spray-positive and multiple-reaction-monitoring modes. The assay was accurate and reproductible for the quantification of the seven antibiotics in serum samples over large concentration ranges.
Collapse
Affiliation(s)
- Sophie Magréault
- Department of Pediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, APHP, Paris, France
| | - Stephanie Leroux
- Department of Pediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, APHP, Paris, France
| | - Jeremie Touati
- Department of Pediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, APHP, Paris, France
| | - Thomas Storme
- Department of Pharmacy, Hôpital Robert Debré, APHP, Paris, France
| | - Evelyne Jacqz-Aigrain
- Department of Pediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, APHP, Paris, France; Clinical Investigation Center CIC1426, INSERM, Paris, France; University Paris 7 - Diderot - Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
46
|
Abstract
BACKGROUND Neonatal sepsis is a leading cause of child morbidity and mortality, especially in premature and low birth weight infants. Prompt antibiotic therapy is warranted, but its inappropriate use leads to bacterial resistance and adverse outcomes. Our objective is to describe the antibiotic use for late-onset sepsis in Peruvian premature infants. METHODS This study is a prospective study as a secondary analysis of a clinical trial in 3 neonatal care units in Peru. We included infants in the first 72 hours of life, with birth weight (BW) <2000 g. We described the antibiotic use as length of therapy (LOT) per 1000 patient days (PD) and antibiotic courses. RESULTS We included 408 neonates, with 12,204 PD of follow-up; 253 infants (62%) had a BW ≤1500 g. Total antibiotic use for late-onset sepsis was 2395 LOT (196 LOT/1000 PD). Two-hundred and seventy-one patients (66.4%) did not receive antibiotics for late-onset sepsis during their hospitalization. In total, 204 antibiotic courses were administered; 92 infants (22.5%) received 1 course, and 45 (11.0%) received 2-5 antibiotic courses. Mean duration of antibiotic course was 10.8 days (standard deviation: ±7.3). We found a significant association between a lower BW and increased antibiotic use per day (P < 0.001). The most commonly used antibiotics were vancomycin (143 LOT/1000 PD), carbapenems (115 LOT/1000 PD), aminoglycosides (72 LOT/1000 PD) and ampicillin (41 LOT/1000 PD). CONCLUSIONS Premature infants receive antibiotics for longer than recommended periods of time. Antibiotic overuse is greater in neonates with lower BW. Vancomycin is the most used antibiotic. There is an urgent need to develop antimicrobial stewardship programs in our setting.
Collapse
|
47
|
Gkentzi D, Dimitriou G. Antimicrobial Stewardship in the Neonatal Intensive Care Unit: An Update. Curr Pediatr Rev 2019; 15:47-52. [PMID: 30657041 PMCID: PMC6696821 DOI: 10.2174/1573396315666190118101953] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 12/19/2022]
Abstract
Neonates represent a vulnerable population for infections and neonatal sepsis is a major cause of mortality and morbidity worldwide. Therefore, antimicrobials are the most commonly prescribed drugs in the Neonatal Intensive Care Unit Setting but unfortunately are quite often used inappropriately with various short and long-term effects. The rational use of antimicrobials is of paramount importance in this population and structured antimicrobial stewardship interventions should be in place. These interventions are slightly different from those used in adults and older children due to the particularities of the neonatal medicine. The aim of this review is to provide an update in the field and identify areas for further consideration and future research.
Collapse
Affiliation(s)
- Despoina Gkentzi
- Department of Paediatrics, Patras Medical School, Patras, Greece
| | | |
Collapse
|
48
|
Ykema JMA, D'Haens EJ, Havenith M, van Eyck J, van Lingen RA, Hemels MAC. Pilot study demonstrates that placental histology can provide an additional tool for diagnosing early-onset neonatal sepsis. Acta Paediatr 2018; 107:2086-2091. [PMID: 29786145 DOI: 10.1111/apa.14410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/20/2018] [Accepted: 05/16/2018] [Indexed: 12/29/2022]
Abstract
AIM We explored whether placental histology could help to diagnose early-onset neonatal sepsis (EONS), guide clinical decision-making 48 hours after birth and reduce antibiotic use. METHODS This study comprised 109 infants born at less than 32 weeks of gestation, who were admitted to the neonatal intensive care unit of Isala, Zwolle, The Netherlands, between January 2013 and December 2013. EONS was defined as clinical symptoms plus raised serial C-reactive protein (CRP) >10 mg/L and a positive (proven EONS) or a negative (suspected EONS) blood culture. Placentas were studied for a histological inflammatory response and scored according to Redline's criteria. RESULTS A histological inflammatory response was seen in 15/88 (17%) placentas and this occurred significantly more often in infants with a high suspicion of EONS (p < 0.05). No histological inflammatory response was seen if maternal risk factors for EONS were absent, despite a raised CRP level. Based on placental histology, the duration of antibiotic therapy was reduced from more than five days to 48 hours in 20/27 infants (74%). CONCLUSION Histological examination of the placenta helped to diagnose EONS and guide clinical decision-making 48 hours after birth and led to a clinically relevant reduction in antibiotic use.
Collapse
Affiliation(s)
- J M A Ykema
- Princess Amalia Children's Centre; Department of Neonatology; Isala Women and Children's Hospital Zwolle; AB Zwolle The Netherlands
| | - E J D'Haens
- Princess Amalia Children's Centre; Department of Neonatology; Isala Women and Children's Hospital Zwolle; AB Zwolle The Netherlands
| | - M Havenith
- Department of Pathology; Isala Women and Children's Hospital Zwolle; AB Zwolle The Netherlands
| | - J van Eyck
- Department of Obstetrics and Gynaecology; Isala Woman and Children's Hospital Zwolle; AB Zwolle The Netherlands
| | - R A van Lingen
- Princess Amalia Children's Centre; Department of Neonatology; Isala Women and Children's Hospital Zwolle; AB Zwolle The Netherlands
| | - M A C Hemels
- Princess Amalia Children's Centre; Department of Neonatology; Isala Women and Children's Hospital Zwolle; AB Zwolle The Netherlands
| |
Collapse
|
49
|
Verstraete EH, Mahieu L, d'Haese J, De Coen K, Boelens J, Vogelaers D, Blot S. Blood culture indications in critically ill neonates: a multicenter prospective cohort study. Eur J Pediatr 2018; 177:1565-1572. [PMID: 30051146 DOI: 10.1007/s00431-018-3203-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/29/2018] [Accepted: 07/01/2018] [Indexed: 12/29/2022]
Abstract
Due to potential lethality of healthcare-associated sepsis (HAS), a low threshold for blood culturing and antimicrobial therapy (ABT) initiation is accepted. We assessed variability in the trigger for blood culturing between three neonatal intensive care units. A multicenter prospective cohort study was conducted. In newborns with suspicion of HAS, 10 predefined clinical signs, nosocomial sepsis (NOSEP) score, C-reactive protein, ABT initiation, and risk factors were registered at time of culturing. Outcome was lab-confirmed HAS, defined according to the NeoKISS-criteria. Two hundred ninety-nine suspected HAS episodes were considered in 212 infants, of which 118 had birth-weight ≤ 1500 g; proportion of lab-confirmed HAS per suspected episode was 30/192 (center 1), 28/60 (center 2), and 8/47 (center 3) (p < 0.001). Median C-reactive protein and number of clinical signs at time of culturing differed between centers 1, 2, and 3 (respectively 11 vs. 5 vs. 3 mg/L, p = 0.001; 1 sign [IQR 0-2, center 1] vs. 3 signs [IQR 2-4, centers 2 and 3], p < 0.001). Median NOSEP score at time of culturing was 5 (IQR 3-8, center 1), 5 (IQR 3-9, center 2), and 8 (IQR 5-11, center 3) (p = 0.016). Difference in ABT initiation was noticed (82 vs. 93 vs. 74%, p = 0.05). CONCLUSION Center heterogeneity in sampling practice is substantial. Optimizing sampling practice can be recommended. What is Known: • Blood culture test is a common diagnostic procedure in critically-ill newborns. • A low threshold for sampling and antimicrobial therapy initiation is accepted. What is New: • Variability in blood culture practice was assessed between 3 neonatal intensive care units by the registration of sampling frequencies, clinical indications, and antimicrobial therapy initiation.
Collapse
Affiliation(s)
| | - Ludo Mahieu
- Department of Neonatal Medicine, Antwerp University Hospital, Antwerp, Belgium
- Department of Pediatrics, University of Antwerp, Antwerp, Belgium
| | - James d'Haese
- Department of Neonatal Medicine, General Hospital of Saint-Jan Bruges, Bruges, Belgium
| | - Kris De Coen
- Department of Neonatal Medicine, Ghent University Hospital, Ghent, Belgium
| | - Jerina Boelens
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Dirk Vogelaers
- Department of Internal Medicine, Ghent University, Ghent, Belgium
- Department of General Internal Medicine, Infectious Diseases and Psychosomatic Disorders, Ghent University Hospital, Ghent, Belgium
| | - Stijn Blot
- Department of Internal Medicine, Ghent University, Ghent, Belgium
- Burns, Trauma and Critical Care Research Centre, The University of Queensland, Brisbane, Australia
| |
Collapse
|
50
|
Fajardo C, Alshaikh B, Harabor A. Prolonged use of antibiotics after birth is associated with increased morbidity in preterm infants with negative cultures. J Matern Fetal Neonatal Med 2018; 32:4060-4066. [PMID: 29792103 DOI: 10.1080/14767058.2018.1481042] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background: Most preterm infants are exposed to a variable duration of antibiotic therapy after birth despite negative cultures. Data is emerging about the risks of prolonged antibiotics. We sought to assess the association between length of initial antibiotic course and neonatal outcomes in a cohort from a single large perinatal center.Methods: Retrospective cohort study of prospectively collected data on all infants with a birth weight of less than 1250 g hospitalized in our NICU in a 4 year window and who had negative blood and CSF cultures in the first 2 days of life. The primary outcome is a composite of necrotizing enterocolitis (NEC), late onset sepsis (LOS) and death evaluated using multivariable regression analysis.Results: A total of 620 infants less than 1250 g with negative cultures were eligible for study over a 4 year period. The 238 infants with more than 5 days initial antibiotic use were significantly smaller and of lower gestational age than the 382 infants who received up to 5 days of antibiotics. Their mothers had more clinical chorioamnionitis, less maternal hypertension and greater perinatal use of antibiotics. On multivariate analysis, infants who received empiric antibiotics for longer than 5 days had higher rates of neonatal morbidities after adjusting for gestational age, SNAP II, small-for-gestational age status, gender, maternal hypertension, prenatal steroid treatment, clinical chorioamnionitis, intrapartum antibiotic treatment, and multiple births. Composite outcome OR: 1.83 (1.15 to 2.92), LOS OR: 2.02 (1.20 to 3.39), bronchopulmonary dysplasia OR: 1.58 (1.04 to 2.29). Mortality and NEC were not significantly different.Conclusion: More than 5 days of antibiotic treatment in very preterm infants with negative cultures was associated with increased morbidity in our population, and that included BPD. It is of note that patterns of increased morbidity and/or mortality differ between studies. Prospective trials of clinical protocols for starting and stopping antibiotics in the very preterm infants are required.
Collapse
Affiliation(s)
- Carlos Fajardo
- Department of Pediatrics, University of Calgary, Peter Lougheed Centre, Calgary, AB, Canada
| | - Belal Alshaikh
- Department of Pediatrics, University of Calgary, Section of Neonatology, South Health Campus, Calgary, AB, Canada
| | - Andrei Harabor
- Regina General Hospital, Division of Neonatology, Saskatchewan Health Authority, Pediatrics, Regina, SK, Canada
| |
Collapse
|