1
|
Keshet A, Hochwald O, Lavon A, Borenstein-Levin L, Shoer S, Godneva A, Glantz-Gashai Y, Cohen-Dolev N, Timstut F, Lotan-Pompan M, Solt I, Weinberger A, Segal E, Shilo S. Development of antibiotic resistome in premature infants. Cell Rep 2025; 44:115515. [PMID: 40198224 DOI: 10.1016/j.celrep.2025.115515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/17/2024] [Accepted: 03/14/2025] [Indexed: 04/10/2025] Open
Abstract
Preterm birth is a major concern in neonatal care, significantly impacting infant survival and long-term health. The gut microbiome, essential for infant development, often becomes imbalanced in preterm infants, making it crucial to understand the effects of antibiotics on its development. Our study analyzed weekly, 6-month, and 1-year stool samples from 100 preterm infants, correlating clinical data on antibiotic use and feeding patterns. Comparing infants who received no antibiotics with those given empirical post-birth treatment, we observed notable alterations in the gut microbiome's composition and an increase in antibiotic resistance gene abundance early in life. Although these effects diminished over time, their long-term clinical impacts remain unclear. Human milk feeding was associated with beneficial microbiota like Actinobacteriota and reduced antibiotic resistance genes, underscoring its protective role. This highlights the importance of judicious antibiotic use and promoting human milk to foster a healthy gut microbiome in preterm infants.
Collapse
Affiliation(s)
- Ayya Keshet
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ori Hochwald
- Neonatal Intensive Care Unit, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel; Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amit Lavon
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Liron Borenstein-Levin
- Neonatal Intensive Care Unit, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel; Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Saar Shoer
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Anastasia Godneva
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yitav Glantz-Gashai
- Neonatal Intensive Care Unit, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Noa Cohen-Dolev
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Fanny Timstut
- Neonatal Intensive Care Unit, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Maya Lotan-Pompan
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Solt
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Department of Obstetrics and Gynecology, Rambam Health Care Campus, Haifa, Israel
| | - Adina Weinberger
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Smadar Shilo
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel; Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel; The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Faculty of Medical and Health Sciences, Tel Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
2
|
Tas M, Onal E, Tapisiz A, Bas S, Hirfanoglu IM, Tezer H, Turkyilmaz C, Ergenekon E, Koc E. Evaluating the effectiveness of a vancomycin stewardship programme in a level IV neonatal intensive care unit. J Hosp Infect 2025; 158:11-18. [PMID: 39922500 DOI: 10.1016/j.jhin.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/27/2024] [Accepted: 01/26/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Despite recommendations to limit the use of vancomycin for known resistant infections, it remains one of the most commonly prescribed antibiotics in neonatal intensive care units (NICUs). One of the most effective approaches to reducing unnecessary antibiotic exposure is through the implementation of antibiotic stewardship programmes (ASPs). AIM The objective of this study was to evaluate the effectiveness of ASPs in reducing the use of vancomycin in neonates hospitalized in our NICU. METHODS This study was a quasi-experimental single-centre study for a quality improvement (QI) initiative. Interventions were implemented to limit the use of vancomycin, including education of the neonatal intensive care team, standardization of vancomycin therapy, and prospective audit and feedback. The pre-intervention period was compared with the post-intervention period. FINDINGS The initiation of vancomycin decreased from 166 times in the pre-intervention period to 71 times after stewardship implementations, representing a 57.2% reduction. Total vancomycin days of therapy per 1000 patient days gradually declined from 113 to 45 (60.2%) (P<0.001) during the study period. There was an increase in the Gram-positive growth in the culture of patients who were started on vancomycin (P=0.04). The number of patients receiving two or more courses of vancomycin treatment decreased by 85.7% (P=0.03). CONCLUSIONS This study has demonstrated that implementing effective multi-disciplinary strategies can significantly reduce vancomycin exposure in the NICU. The application of ASP practices and management in the NICU is essential and achievable, without any increase in the duration of hospitalization or mortality rates.
Collapse
Affiliation(s)
- M Tas
- Gazi University Medical Faculty, Department of Pediatrics, Neonatology Unit, Ankara, Turkey.
| | - E Onal
- Gazi University Medical Faculty, Department of Pediatrics, Neonatology Unit, Ankara, Turkey
| | - A Tapisiz
- Gazi University Medical Faculty, Department of Pediatrics, Pediatric Infectious Diseases, Ankara, Turkey
| | - S Bas
- Gazi University Medical Faculty, Department of Infection Committee, Ankara, Turkey
| | - I M Hirfanoglu
- Gazi University Medical Faculty, Department of Pediatrics, Neonatology Unit, Ankara, Turkey
| | - H Tezer
- Gazi University Medical Faculty, Department of Pediatrics, Pediatric Infectious Diseases, Ankara, Turkey
| | - C Turkyilmaz
- Gazi University Medical Faculty, Department of Pediatrics, Neonatology Unit, Ankara, Turkey
| | - E Ergenekon
- Gazi University Medical Faculty, Department of Pediatrics, Neonatology Unit, Ankara, Turkey
| | - E Koc
- Gazi University Medical Faculty, Department of Pediatrics, Neonatology Unit, Ankara, Turkey
| |
Collapse
|
3
|
Fillistorf L, Carra G, Matusiak R, Dimopoulou V, Despraz J, Meylan S, Giannoni E. Absence of association between early antibiotic exposure and short-term adverse outcomes in very preterm infants: a single-center retrospective study. Front Pediatr 2025; 13:1563979. [PMID: 40166661 PMCID: PMC11955704 DOI: 10.3389/fped.2025.1563979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Background Antibiotics save lives but also carry significant risks, including increased antimicrobial resistance, higher healthcare costs, and disruption of the microbiome. However, the association between antibiotic exposure and short-term adverse outcomes remains uncertain. Our study aimed to evaluate whether early unnecessary antibiotic exposure in the first 7 days of life of very preterm infants is linked to short-term adverse outcomes. Methods This retrospective study included infants born below 32 weeks of gestation and hospitalized at the University Hospital of Lausanne between January 1, 2007 and December 31, 2022. Antibiotic exposure was quantified during the first seven postnatal days by the median number of days of antibiotics. Multilinear regressions and mixed effect models analyzed the association between the number of days of antibiotics and death, late-onset sepsis, necrotizing enterocolitis, severe bronchopulmonary dysplasia, severe retinopathy of prematurity and cystic periventricular leukomalacia. The primary outcome was a composite of at least one of the listed adverse outcomes, while the secondary outcomes consisted of each adverse outcome individually. Adjusted odds ratio (aOR) and p-value were calculated. Results We included 1,398 preterm infants. The median gestational age was 29 weeks (IQR: 27-30) and the median birthweight was 1,144 grams (895-1,420). The median number of days of antibiotics declined by 53%, from 4 days in 2007 to 1.9 days in 2022 (p < 0.0001). The number of days of antibiotics was not associated with the composite outcome [aOR: 0.97 (0.82-1.17), p = 0.80, adjusted p = 0.80] or any of the following adverse outcomes: mortality [aOR: 1.10 (0.78-1.55), p = 0.58, adjusted p = 0.69], late-onset sepsis [aOR: 0.74 (0.59-0.93), p = 0.01, adjusted p = 0.07], necrotizing enterocolitis [aOR: 1.22 (0.86-1.74), p = 0.26, adjusted p = 0.65], severe bronchopulmonary dysplasia [aOR: 1.12 (0.88-1.42), p = 0.36, adjusted p = 0.65], severe retinopathy of prematurity [aOR: 1.34 (0.65-2.78), p = 0.43, adjusted p = 0.65], and cystic periventricular leukomalacia [aOR: 1.02 (0.69-1.99), p = 0.91, adjusted p = 0.91]. Conclusion We found no association between early antibiotic exposure and short-term adverse outcomes.
Collapse
Affiliation(s)
- Laura Fillistorf
- Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Giorgia Carra
- Biomedical Data Science Center, Department of Innovation and Clinical Research, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Raphaël Matusiak
- Biomedical Data Science Center, Department of Innovation and Clinical Research, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Swiss Data Science Center, Swiss Federal Institute of Technology in Lausanne, Lausanne, Switzerland
| | - Varvara Dimopoulou
- Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jérémie Despraz
- Biomedical Data Science Center, Department of Innovation and Clinical Research, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sylvain Meylan
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eric Giannoni
- Clinic of Neonatology, Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
4
|
Coggins SA, Greenberg RG. Pharmacokinetic and Pharmacodynamic Approaches to Optimize Antibiotic Use in Neonates. Clin Perinatol 2025; 52:67-86. [PMID: 39892955 DOI: 10.1016/j.clp.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Newborn infants (particularly those born preterm) are frequently exposed to empiric antibiotics at birth, and antibiotics are among the most commonly prescribed medications in neonatal intensive care units. Challenges in optimizing neonatal antibiotic dosing include: technical and ethical barriers to neonatal pharmacoanalytic study design and sampling, difficulty in extrapolating adult and pediatric data due to unique neonatal physiology, and a lack of validated pharmacodynamic targets specific to neonatal populations. In this review, we summarize basic concepts in pharmacokinetics (PK) and pharmacodynamics (PD), describe pharmacometric strategies utilized in contemporary PK/PD analyses, and review the evolution of PK/PD data guiding neonatal dosing among 3 commonly used antibiotics.
Collapse
Affiliation(s)
- Sarah A Coggins
- Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Neonatology (2 Main NW), Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - Rachel G Greenberg
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA; Duke Clinical Research Institute, 300 West Morgan Street Suite 800, Durham, NC 27701, USA
| |
Collapse
|
5
|
Massaro AN, Fletcher EP, Madabushi R, Baer G, Green D. A Pediatric Research Imperative: Addressing Neonates in Drug Development Through Understanding Neonatal Clinical Pharmacology. J Pediatr Pharmacol Ther 2025; 30:8-16. [PMID: 39935560 PMCID: PMC11809530 DOI: 10.5863/1551-6776-30.1.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/10/2024] [Indexed: 02/13/2025]
Abstract
Drug development in the neonatal population remains an unmet need in health care. While incentives and legislative mandates have had some impact on increasing drug development in pediatric patients, the advancement of neonatal therapeutics faces unique challenges. This review summarizes relevant regulatory history, clinical, pharmacological and ethical considerations that characterize the landscape of drug development in neonates. Research priorities and future directions for advancing safe and effective medicines for the vulnerable neonatal population are discussed.
Collapse
Affiliation(s)
- An N. Massaro
- Office of Pediatric Therapeutics, Office of the Chief Medical Officer, Office of the Commissioner, US FDA (DG, ANM), Silver Spring, MD
| | - Elimika Pfuma Fletcher
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US FDA (EPF, RM), Silver Spring, MD
| | - Rajanikanth Madabushi
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US FDA (EPF, RM), Silver Spring, MD
| | - Gerri Baer
- Division of Hepatology and Nutrition, Office of New Drugs, Center for Drug Evaluation and Research, US FDA (GB), Silver Spring, MD
| | - Dionna Green
- Office of Pediatric Therapeutics, Office of the Chief Medical Officer, Office of the Commissioner, US FDA (DG, ANM), Silver Spring, MD
| |
Collapse
|
6
|
Lloyd LG, van Weissenbruch MM, Bekker A, Ferreyra C, Gleeson B, Dramowski A. Theoretical impact of a bedside decision-making tool on antibiotic use for suspected neonatal healthcare-associated infection: an observational study. BMC Pediatr 2025; 25:52. [PMID: 39838378 PMCID: PMC11749325 DOI: 10.1186/s12887-024-05323-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Healthcare-associated infections (HAI) are a leading contributor to morbidity and mortality in hospitalised neonates. Diagnosing neonatal HAI is challenging owing to non-specific symptoms and lack of definitive diagnostic markers, contributing to high rates of inappropriate antibiotic use. This study evaluated the theoretical impact of implementing a bedside tool for decision-making on antibiotic length of therapy (LOT). METHODS This prospective observational physician-blinded study consecutively enrolled patients with suspected HAI events at a large South African neonatal unit from September 2022 to September 2023. The antibiotic decision-making tool included an infection prediction score (NeoHoP), and a point-of-care C-reactive protein test (CRP) performed at HAI diagnosis and 24 h later. The theoretical impact of the tool on antibiotic LOT was calculated. RESULTS We recruited 180 neonates with 214 episodes of suspected HAI, of which 22 (10.3%) were proven HAI, 56 (26.2%) were presumed HAI and 136 (63.6%) had HAI ruled out. The median observed antibiotic LOT was three days (9 days for proven HAI, 7 days for presumed HAI, and 3 days for no HAI). The antibiotic decision-making tool would theoretically reduce overall antibiotic LOT by 2 days (p < 0.001), particularly in neonates where HAI was subsequently excluded. CONCLUSION We developed an antibiotic decision-making tool to support the clinical evaluation of suspected neonatal HAI and demonstrated a significant potential impact on reducing antibiotic LOT. Given increasing antibiotic resistance rates globally, this tool should be further evaluated to minimise unnecessary antibiotic use in hospitalised neonates.
Collapse
Affiliation(s)
- Lizel Georgi Lloyd
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| | | | - Adrie Bekker
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | | | | | - Angela Dramowski
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
7
|
Lawrence SM, Wynn JL, Gordon SM. Neonatal bacteremia and sepsis. REMINGTON AND KLEIN'S INFECTIOUS DISEASES OF THE FETUS AND NEWBORN INFANT 2025:183-232.e25. [DOI: 10.1016/b978-0-323-79525-8.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
8
|
Nishihara Y, MacBrayne CE, Prinzi A, Pearce K, Melara D, Weikel BW, Zenge J, Grover T, Parker SK. Time to positivity of blood cultures in a level IV NICU varies based on organism category and population subgroups: is a 36-hour rule out safe? J Perinatol 2024; 44:1807-1815. [PMID: 38902516 DOI: 10.1038/s41372-024-02031-0] [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: 12/19/2023] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Time to positivity (TTP) of blood cultures and organism characteristics may be different in a Level IV NICU population. METHODS Retrospective study of 309 Level IV NICU positive blood cultures between January 2012 to December 2018 describing TTP and organism characteristics. RESULTS Median TTP [IQR] was 21.1 [14.3, 25.2] hours, with 91.2% positive at 36 h, and 96.1% positive at 48 h. Gram negative definite pathogens had the shortest TTP (13.0 [11.4, 15.4] hours) compared to gram positive definite pathogens (16.3 [13.0, 22.4] hours). TTP for treated gram positive commensal organisms (22.3 [20.1, 30.4] hours) and those considered contaminants (23.6 [21.4, 26.0] hours), was significantly longer than both gram positive and negative definite pathogens. CONCLUSION When antimicrobials are initiated due to concern for bacteremia and blood cultures have not identified a causative pathogen at 36 h, antimicrobials may be safely discontinued in the majority of Level IV NICU patients.
Collapse
Affiliation(s)
- Yo Nishihara
- Department of Neonatology, Children's Hospital Colorado and School of Medicine, University of Colorado, Aurora, CO, USA.
| | | | - Andrea Prinzi
- Graduate School of Medicine, University of Colorado, Aurora, CO, USA
| | - Kelly Pearce
- Department of Epidemiology, Children's Hospital Colorado, Aurora, CO, USA
| | - Diane Melara
- Department of Neonatology, Children's Hospital Colorado and School of Medicine, University of Colorado, Aurora, CO, USA
| | - Blair W Weikel
- Department of Neonatology, Children's Hospital Colorado and School of Medicine, University of Colorado, Aurora, CO, USA
| | - Jeanne Zenge
- Department of Neonatology, Children's Hospital Colorado and School of Medicine, University of Colorado, Aurora, CO, USA
| | - Theresa Grover
- Department of Neonatology, Children's Hospital Colorado and School of Medicine, University of Colorado, Aurora, CO, USA
| | - Sarah K Parker
- Department of Pediatric Infectious Diseases, Children's Hospital Colorado and School of Medicine, University of Colorado, Aurora, CO, USA
| |
Collapse
|
9
|
Thänert R, Schwartz DJ, Keen EC, Hall-Moore C, Wang B, Shaikh N, Ning J, Rouggly-Nickless LC, Thänert A, Ferreiro A, Fishbein SRS, Sullivan JE, Radmacher P, Escobedo M, Warner BB, Tarr PI, Dantas G. Clinical sequelae of gut microbiome development and disruption in hospitalized preterm infants. Cell Host Microbe 2024; 32:1822-1837.e5. [PMID: 39197454 PMCID: PMC11466706 DOI: 10.1016/j.chom.2024.07.027] [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: 05/10/2024] [Revised: 06/24/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Aberrant preterm infant gut microbiota assembly predisposes to early-life disorders and persistent health problems. Here, we characterize gut microbiome dynamics over the first 3 months of life in 236 preterm infants hospitalized in three neonatal intensive care units using shotgun metagenomics of 2,512 stools and metatranscriptomics of 1,381 stools. Strain tracking, taxonomic and functional profiling, and comprehensive clinical metadata identify Enterobacteriaceae, enterococci, and staphylococci as primarily exploiting available niches to populate the gut microbiome. Clostridioides difficile lineages persist between individuals in single centers, and Staphylococcus epidermidis lineages persist within and, unexpectedly, between centers. Collectively, antibiotic and non-antibiotic medications influence gut microbiome composition to greater extents than maternal or baseline variables. Finally, we identify a persistent low-diversity gut microbiome in neonates who develop necrotizing enterocolitis after day of life 40. Overall, we comprehensively describe gut microbiome dynamics in response to medical interventions in preterm, hospitalized neonates.
Collapse
Affiliation(s)
- Robert Thänert
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Drew J Schwartz
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Women's Infectious Diseases Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric C Keen
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla Hall-Moore
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bin Wang
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie Ning
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Anna Thänert
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aura Ferreiro
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Skye R S Fishbein
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Janice E Sullivan
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY 40202, USA
| | - Paula Radmacher
- Department of Pediatrics, University of Louisville School of Medicine, Norton Children's Hospital, Louisville, KY 40202, USA
| | - Marilyn Escobedo
- Department of Pediatrics, University of Oklahoma, Oklahoma City, OK 73104, USA
| | - Barbara B Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
10
|
Viana Oliveira CR, Almeida Alves I, de Oliveira-Filho AD, Boa-Sorte N. Profile of antimicrobial use and potential use of the defined daily dose in neonatology. Sci Rep 2024; 14:22979. [PMID: 39362955 PMCID: PMC11450155 DOI: 10.1038/s41598-024-74559-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024] Open
Abstract
One of the measures for monitoring microbial resistance is the calculation of the defined daily dose of antimicrobial agents. For this calculation, the weight of an adult of 70 kg is used as a standard, so that application in neonatology is not possible. The aim of this study is to describe the use profile and calculate the defined daily dose (DDD) of antimicrobials in a neonatal intensive care unit (NICU) of a public hospital in the interior of Bahia, Brazil. From March 2020 to December 2021, the medical records of 712 newborns admitted to a NICU between September 2018 and June 2020 were analyzed. A total of 410 newborns diagnosed with neonatal sepsis were included. The most used antimicrobials per patient were gentamicin (408/410; 99.5%), ampicillin (407; 99.3%), amikacin (29; 7.1%) and oxacillin (21; 5.1%), with a mean (SD) treatment duration of 9.8 (3.9) days. The most commonly used combination of antimicrobials was ampicillin with gentamicin, which was used in 406 patients (99.0%). The values for neonatal DDDs were on average 26 times lower than those for adult DDDs. The neonatal DDDs were similar to those observed in other studies. Ampicilin and cefepime were the antimicrobials for which the greatest differences were observed in neonatal DDDs compared with adult DDDs, which differed mainly between maintenance doses, reflecting the lack of international standards in neonatology. Standardization of DDDs as a surveillance measure has the potential to clarify the pattern of antimicrobial use in neonatal patients worldwide and, in particular, to prevent indiscriminate use and bacterial resistance.
Collapse
Affiliation(s)
- Cinara Rejane Viana Oliveira
- Graduate Program in Pharmaceutical Sciences, Federal University of Sergipe, Avenue Marechal Rondon, Jd. Rosa Elze, São Cristóvão, 49100-000, Sergipe, Brazil.
- Laboratory of Teaching and Research in Social Pharmacy (LEPFS), Department of Pharmacy, Federal University of Sergipe, Avenue Marechal Rondon, Jd. Rosa Elze, 49100-000, São Cristóvão, Sergipe, Brazil.
| | - Izabel Almeida Alves
- Department of Life Sciences (DCV), State University of Bahia (UNEB), Campus I, 2555 Silveira Martins Street, Cabula, Salvador, 41150-000, Bahia, Brazil
| | - Alfredo Dias de Oliveira-Filho
- Laboratory of Teaching and Research in Social Pharmacy (LEPFS), Department of Pharmacy, Federal University of Sergipe, Avenue Marechal Rondon, Jd. Rosa Elze, 49100-000, São Cristóvão, Sergipe, Brazil
| | - Ney Boa-Sorte
- Department of Life Sciences (DCV), State University of Bahia (UNEB), Campus I, 2555 Silveira Martins Street, Cabula, Salvador, 41150-000, Bahia, Brazil
| |
Collapse
|
11
|
Barca HC, Ferber J, Richards M, Strickland M, Fernandez AJ, Li DK, Darrow LA. Antimicrobial exposure during infancy in a longitudinal California cohort. Pediatr Res 2024; 96:805-813. [PMID: 38570559 DOI: 10.1038/s41390-024-03171-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/05/2024] [Accepted: 03/10/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND To describe temporal and sociodemographic patterns of antimicrobial exposure during the first year of life in a large US cohort. METHODS Singleton infants born 1998-2014 enrolled in Kaiser Permanente Northern California integrated health system (n = 345,550) were followed longitudinally via comprehensive electronic health records, capturing all systemic antimicrobial inpatient administrations and outpatient dispensings. Antimicrobial exposure was summarized by maternal and infant characteristics, birth year, inpatient/outpatient status, age in months, and drug class. RESULTS Overall, 44% of infants in this cohort received at least one dose of antimicrobials during infancy. Decreases over time were driven by reduced outpatient dispensings specifically in later infancy, primarily for penicillins. Among infants receiving any antimicrobials the median number of exposure-days was 16. Inpatient dispensings peaked in the first 30 days of life and outpatient dispensings peaked at 10-11 months. Birth characteristics (i.e., NICU admission, gestational age) were strong independent predictors of antimicrobial exposure between 0- < 3 months; sociodemographic factors were modest predictors of exposure for 3-12 months. CONCLUSION Predictors of antimicrobial exposure in early and late infancy are distinct with early infancy exposures highly correlated to birth characteristics. The cumulative proportion of infants exposed has decreased due to fewer late infancy outpatient dispensings. IMPACT Comprehensive antimicrobial exposure histories and the maternal and infant characteristics predicting exposure have not been well described in US populations. This analysis provides estimates of cumulative antimicrobial exposures by sociodemographic factors, delivery characteristics, month of life, inpatient/outpatient status, and antibiotic class among one of the largest US HMOs. Predictors of early infancy antimicrobial exposures differ from those in late infancy, with early exposures strongly correlated to birth characteristics and late infancy exposures modestly related to sociodemographic factors. Antimicrobial exposure among infants decreased over the time period primarily due to reduced outpatient dispensings in later infancy.
Collapse
Affiliation(s)
- Hannah C Barca
- School of Public Health, University of Nevada, Reno, Reno, NV, USA.
| | | | - Megan Richards
- School of Public Health, University of Nevada, Reno, Reno, NV, USA
| | | | | | - De-Kun Li
- Kaiser Permanente Division of Research, Oakland, CA, USA
| | - Lyndsey A Darrow
- School of Public Health, University of Nevada, Reno, Reno, NV, USA
| |
Collapse
|
12
|
Kontou A, Agakidou E, Chatziioannidis I, Chotas W, Thomaidou E, Sarafidis K. Antibiotics, Analgesic Sedatives, and Antiseizure Medications Frequently Used in Critically Ill Neonates: A Narrative Review. CHILDREN (BASEL, SWITZERLAND) 2024; 11:871. [PMID: 39062320 PMCID: PMC11275925 DOI: 10.3390/children11070871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024]
Abstract
Antibiotic, analgesic sedative, and antiseizure medications are among the most commonly used medications in preterm/sick neonates, who are at high risk of nosocomial infections, central nervous system complications, and are exposed to numerous painful/stressful procedures. These severe and potentially life-threatening complications may have serious short- and long-term consequences and should be prevented and/or promptly treated. The reported variability in the medications used in neonates indicates the lack of adequate neonatal studies regarding their effectiveness and safety. Important obstacles contributing to inadequate studies in preterm/sick infants include difficulties in obtaining parental consent, physicians' unwillingness to recruit preterm infants, the off-label use of many medications in neonates, and other scientific and ethical concerns. This review is an update on the use of antimicrobials (antifungals), analgesics (sedatives), and antiseizure medications in neonates, focusing on current evidence or knowledge gaps regarding their pharmacokinetics, indications, safety, dosage, and evidence-based guidelines for their optimal use in neonates. We also address the effects of early antibiotic use on the intestinal microbiome and its association with long-term immune-related diseases, obesity, and neurodevelopment (ND). Recommendations for empirical treatment and the emergence of pathogen resistance to antimicrobials and antifungals are also presented. Finally, future perspectives on the prevention, modification, or reversal of antibiotic resistance are discussed.
Collapse
Affiliation(s)
- Angeliki Kontou
- Department of Neonatology and Neonatal Intensive Care, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Ippokrateion General Hospital, 54642 Thessaloniki, Greece; (E.A.); (I.C.); (K.S.)
| | - Eleni Agakidou
- Department of Neonatology and Neonatal Intensive Care, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Ippokrateion General Hospital, 54642 Thessaloniki, Greece; (E.A.); (I.C.); (K.S.)
| | - Ilias Chatziioannidis
- Department of Neonatology and Neonatal Intensive Care, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Ippokrateion General Hospital, 54642 Thessaloniki, Greece; (E.A.); (I.C.); (K.S.)
| | - William Chotas
- Department of Neonatology, University of Vermont, Burlington, VT 05405, USA
| | - Evanthia Thomaidou
- Department of Anesthesia and Intensive Care, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, AHEPA University General Hospital of Thessaloniki, 54621 Thessaloniki, Greece;
| | - Kosmas Sarafidis
- Department of Neonatology and Neonatal Intensive Care, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Ippokrateion General Hospital, 54642 Thessaloniki, Greece; (E.A.); (I.C.); (K.S.)
| |
Collapse
|
13
|
Chirico V, Lacquaniti A, Tripodi F, Conti G, Marseglia L, Monardo P, Gitto E, Chimenz R. Acute Kidney Injury in Neonatal Intensive Care Unit: Epidemiology, Diagnosis and Risk Factors. J Clin Med 2024; 13:3446. [PMID: 38929977 PMCID: PMC11205241 DOI: 10.3390/jcm13123446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Acute kidney injury (AKI) is associated with long-term consequences and poor outcomes in the neonatal intensive care unit. Its precocious diagnosis represents one of the hardest challenges in clinical practice due to the lack of sensitive and specific biomarkers. Currently, neonatal AKI is defined with urinary markers and serum creatinine (sCr), with limitations in early detection and individual treatment. Biomarkers and risk factor scores were studied to predict neonatal AKI, to early identify the stage of injury and not the damage and to anticipate late increases in sCr levels, which occurred when the renal function already began to decline. Sepsis is the leading cause of AKI, and sepsis-related AKI is one of the main causes of high mortality. Moreover, preterm neonates, as well as patients with post-neonatal asphyxia or after cardiac surgery, are at a high risk for AKI. Critical patients are frequently exposed to nephrotoxic medications, representing a potentially preventable cause of AKI. This review highlights the definition of neonatal AKI, its diagnosis and new biomarkers available in clinical practice and in the near future. We analyze the risk factors involving patients with AKI, their outcomes and the risk for the transition from acute damage to chronic kidney disease.
Collapse
Affiliation(s)
- Valeria Chirico
- Pediatric Nephrology and Dialysis Unit, University Hospital “G. Martino”, 98124 Messina, Italy (F.T.)
| | - Antonio Lacquaniti
- Nephrology and Dialysis Unit, Papardo Hospital, 98158 Messina, Italy (P.M.)
| | - Filippo Tripodi
- Pediatric Nephrology and Dialysis Unit, University Hospital “G. Martino”, 98124 Messina, Italy (F.T.)
| | - Giovanni Conti
- Pediatric Nephrology and Dialysis Unit, University Hospital “G. Martino”, 98124 Messina, Italy (F.T.)
| | - Lucia Marseglia
- Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98124 Messina, Italy; (L.M.)
| | - Paolo Monardo
- Nephrology and Dialysis Unit, Papardo Hospital, 98158 Messina, Italy (P.M.)
| | - Eloisa Gitto
- Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98124 Messina, Italy; (L.M.)
| | - Roberto Chimenz
- Pediatric Nephrology and Dialysis Unit, University Hospital “G. Martino”, 98124 Messina, Italy (F.T.)
| |
Collapse
|
14
|
Flannery DD, Coggins SA, Medoro AK. Antibiotic Stewardship in the Neonatal Intensive Care Unit. J Intensive Care Med 2024:8850666241258386. [PMID: 38835250 DOI: 10.1177/08850666241258386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Antibiotic stewardship is a multidisciplinary, evidence-based approach to optimize antibiotic use and mitigate development of antibiotic resistance. Neonates have high rates of antibiotic exposure, particularly those born preterm and admitted to the NICU, and mounting evidence describes the adverse consequences of such exposures in the absence of infection. Here, we review the general principles of antibiotic stewardship and how they can be applied in NICUs. The unique characteristics of NICUs and patients cared for in this setting, which warrant unique implementation strategies and special considerations are discussed. We summarize current antibiotic use metrics for assessment of responses to stewardship interventions and changes over time, and review evidence-based infection prevention practices in the NICU. Current recommendations for empiric antibiotic use in the NICU and the utility of infection biomarkers are summarized. Lastly, given the growing global threat of increasing antibiotic resistance, specific threats in the NICU are highlighted.
Collapse
Affiliation(s)
- Dustin D Flannery
- Division of Neonatology, Department of Pediatrics, University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sarah A Coggins
- Division of Neonatology, Department of Pediatrics, University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alexandra K Medoro
- Division of Neonatology, Department of Pediatrics, University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Infectious Diseases, Department of Pediatrics, University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
15
|
Arribas C, Decembrino N, Raffaeli G, Amodeo I, González-Caballero JL, Riaza M, Ortiz-Movilla R, Massenzi L, Gizzi C, Araimo G, Cattarelli D, Aversa S, Martinelli S, Frezza S, Orfeo L, Mosca F, Cavallaro G, Garrido F. Ototoxic and nephrotoxic drugs in neonatal intensive care units: results of a Spanish and Italian survey. Eur J Pediatr 2024; 183:2625-2636. [PMID: 38492032 DOI: 10.1007/s00431-024-05467-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 03/18/2024]
Abstract
Neonates face heightened susceptibility to drug toxicity, often exposed to off-label medications with dosages extrapolated from adult or pediatric studies. Premature infants in Neonatal Intensive Care Units (NICUs) are particularly at risk due to underdeveloped pharmacokinetics and exposure to multiple drugs. The study aimed to survey commonly used medications with a higher risk of ototoxicity and nephrotoxicity in Spanish and Italian neonatal units. A prospective cross-sectional study was conducted in Italian and Spanish neonatal units using a web-based survey with 43 questions. A modified Delphi method involved experts refining the survey through online consensus. Ethical approval was obtained, and responses were collected from January to July 2023. The survey covered various aspects, including drug-related ototoxic and nephrotoxic management, hearing screening, and therapeutic drug monitoring. Responses from 131 participants (35.9% from Spain and 64.1% from Italy) revealed awareness of drug toxicity risks. Varied practices were observed in hearing screening protocols, and a high prevalence of ototoxic and nephrotoxic drug use, including aminoglycosides (100%), vancomycin (70.2%), loop diuretics (63.4%), and ibuprofen (62.6%). Discrepancies existed in guideline availability and adherence, with differences between Italy and Spain in therapeutic drug monitoring practices. CONCLUSIONS The study underscores the need for clinical guidelines and uniform practices in managing ototoxic and nephrotoxic drugs in neonatal units. Awareness is high, but inconsistencies in practices indicate a necessity for standardization, including the implementation of therapeutic drug monitoring and the involvement of clinical pharmacologists. Addressing these issues is crucial for optimizing neonatal care in Southern Europe. WHAT IS KNOWN • Neonates in intensive care face a high risk of nephrotoxicity and ototoxicity from drugs like aminoglycosides, vancomycin, loop diuretics, and ibuprofen. • Therapeutic drug monitoring is key for managing these risks, optimizing dosing for efficacy and minimizing side effects. WHAT IS NEW • NICUs in Spain and Italy show high drug toxicity awareness but differ in ototoxic/nephrotoxic drug management. • Urgent need for standard guidelines and practices to address nephrotoxic risks from aminoglycosides, vancomycin, loop diuretics, and ibuprofen.
Collapse
Affiliation(s)
- Cristina Arribas
- Neonatal Intensive Care Unit, Clínica Universidad de Navarra, 28027, Madrid, Spain
| | - Nunzia Decembrino
- Neonatal Intensive Care Unit, AOU Policlinico G. Rodolico San Marco, 95123, Catania, Italy
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy.
| | - Ilaria Amodeo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | | | - Mónica Riaza
- Department of Pediatrics, Clínica Universidad de Navarra, 31008, Pamplona, Spain
| | - Roberto Ortiz-Movilla
- Neonatal Intensive Care Unit, Hospital Universitario Puerta de Hierro-Majadahonda, 28222, Madrid, Spain
| | - Luca Massenzi
- Neonatal Intensive Care Unit, Ospedale Regionale Di Bolzano, 39100, Bolzano, Italy
| | - Camilla Gizzi
- Division of Pediatrics and Neonatology, Sandro Pertini Hospital, 00157, Rome, Italy
| | - Gabriella Araimo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Donatella Cattarelli
- Division of Pediatrics and Neonatology, ASST del Garda, 25015, Desenzano del Garda (BS), Italy
| | - Salvatore Aversa
- Neonatal Intensive Care Unit, Children's Hospital, ASST Spedali Civili, 25123, Brescia, Italy
| | - Stefano Martinelli
- Neonatal Intensive Care Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162, Milan, Italy
| | - Simonetta Frezza
- Division of Neonatology, Area of Child Health, Department of Woman, Child Health and Public Health, Fondazione IRCCS Policlinico Universitario Agostino Gemelli, 00168, Rome, Italy
| | - Luigi Orfeo
- Neonatal Intensive Care Unit, Isola Tiberina Hospital Gemelli Isola, 00186, Rome, Italy
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, 20122, Milan, Italy
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Felipe Garrido
- Neonatal Intensive Care Unit, Clínica Universidad de Navarra, 28027, Madrid, Spain
| |
Collapse
|
16
|
Kuo CM, Su PC, Yang ST, Chung HW, Chen HL. Furosemide and Ductus Arteriosus Closure in Very-Low-Birth-Weight Preterm Infants: A Comprehensive Retrospective Study. CHILDREN (BASEL, SWITZERLAND) 2024; 11:610. [PMID: 38790605 PMCID: PMC11119670 DOI: 10.3390/children11050610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/23/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024]
Abstract
Ductus arteriosus closure may be delayed in preterm infants, and prostaglandin, a vasodilator, can affect ductal patency. Furosemide can increase renal prostaglandin synthesis, so its net effect on patent ductus arteriosus (PDA) is uncertain. Our goal is to explore the relationship between furosemide and spontaneous ductal closure in very-low-birth-weight preterm infants. Our treatment for PDA involves fluid restriction initially and furosemide administration for hemodynamically significant PDA until closure is confirmed by the echocardiogram. We enrolled 105 infants from 1 January 2019 to 30 June 2022 and evaluated the impact of furosemide on ductal closure, including exposure duration and cumulative dose. There is no correlation between furosemide exposure and spontaneous ductal closure (p = 0.384). Furosemide exposure does not delay the postmenstrual age at which spontaneous ductal closure occurs (p = 0.558). The time for spontaneous ductal closure is positively associated with furosemide prescription days (coefficient value = 0.547, p = 0.026) and negatively with gestational age (coefficient value = -0.384, p = 0.062). The prescription of furosemide does not impact the probability or time duration of ductus arteriosus spontaneous closure. The cumulative dose of furosemide has minimal impact on ductal closure. The correlation between furosemide exposure duration and ductal patency duration is likely due to our treatment protocol, with gestational age being a significant factor.
Collapse
Affiliation(s)
- Chi-Mei Kuo
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan; (C.-M.K.); (P.-C.S.); (S.-T.Y.); (H.-W.C.)
| | - Pin-Chun Su
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan; (C.-M.K.); (P.-C.S.); (S.-T.Y.); (H.-W.C.)
| | - Shu-Ting Yang
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan; (C.-M.K.); (P.-C.S.); (S.-T.Y.); (H.-W.C.)
| | - Hao-Wei Chung
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan; (C.-M.K.); (P.-C.S.); (S.-T.Y.); (H.-W.C.)
| | - Hsiu-Lin Chen
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan; (C.-M.K.); (P.-C.S.); (S.-T.Y.); (H.-W.C.)
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung City 807378, Taiwan
| |
Collapse
|
17
|
Sobel AL, Melamed J, Haas D, LeBlanc G, Cirone A, Constantinides MG. Antibiotic use in early life subsequently impairs MAIT cell-mediated immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593643. [PMID: 38798453 PMCID: PMC11118404 DOI: 10.1101/2024.05.10.593643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are predominantly located in barrier tissues where they rapidly respond to pathogens and commensals by recognizing microbial derivatives of riboflavin synthesis. Early-life exposure to these metabolites imprints the abundance of MAIT cells within tissues, so we hypothesized that antibiotic use during this period may abrogate their development. We identified antibiotics that deplete riboflavin-synthesizing commensals and revealed an early period of susceptibility during which antibiotic administration impaired MAIT cell development. The reduction in MAIT cell abundance rendered mice more susceptible to pneumonia, while MAIT cell-deficient mice were unaffected by early-life antibiotics. Concomitant administration of a riboflavin-synthesizing commensal during antibiotic treatment was sufficient to restore MAIT cell development and immunity. Our work demonstrates that transient depletion of riboflavin-synthesizing commensals in early life can adversely affect responses to subsequent infections.
Collapse
|
18
|
Leiting C, Kerns E, Euteneuer JC, McCulloh RJ, Peeples ES. Inhaled Corticosteroid Exposure in Hospitalized Infants with Bronchopulmonary Dysplasia. Am J Perinatol 2024; 41:e85-e93. [PMID: 35523409 PMCID: PMC9637235 DOI: 10.1055/a-1845-2669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
OBJECTIVE The objective of this paper was to determine inhaled corticosteroid (IC) use in infants with bronchopulmonary dysplasia (BPD), define the interhospital variation of IC administration to infants with BPD, and compare clinical, demographic, and hospital factors associated with IC use. STUDY DESIGN Using the Pediatric Health Information System database, a retrospective multicenter cohort of 4,551 infants born at <32 weeks of gestation with developing BPD was studied. The clinical, demographic, and hospital characteristics of infants exposed and not exposed to ICs were compared. RESULTS IC use varied markedly between hospitals, ranging from 0 to 66% of infants with BPD exposed to ICs. Increased annual BPD census was not associated with IC use. In total, 25% (1,144 out of 4,551) of patients with BPD and 43% (536 out of 1,244) of those with severe BPD received ICs. Increased IC exposure was associated with lower birth weight and gestational age, days on respiratory support, need for positive pressure ventilation at 36-week postmenstrual age, need for tracheostomy, and increased use of systemic steroids, bronchodilators, and diuretics. CONCLUSION IC exposure is common in infants with BPD, with substantial interhospital variability. IC use was associated with more severe disease. Hospital experience did not account for the wide variability in IC use by the hospital. Further research into the effects of ICs use is urgently needed to help guide their use in this vulnerable population. KEY POINTS · The risks and benefits of IC use in infants with BPD are incompletely understood.. · IC use is common in infants with BPD (25%) and severe BPD (43%) varies widely by hospital (0-66% of patients with BPD received an IC).. · Hospital experience did not account for the wide interhospital variation in IC use..
Collapse
Affiliation(s)
| | - Ellen Kerns
- Children’s Hospital & Medical Center, Omaha, NE
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE
| | - Joshua C. Euteneuer
- Children’s Hospital & Medical Center, Omaha, NE
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE
| | - Russell J. McCulloh
- Children’s Hospital & Medical Center, Omaha, NE
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE
| | - Eric S. Peeples
- Children’s Hospital & Medical Center, Omaha, NE
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
19
|
Aithal N, Kandasamy Y. The Babyccino: The Role of Caffeine in the Prevention of Acute Kidney Injury in Neonates-A Literature Review. Healthcare (Basel) 2024; 12:529. [PMID: 38470639 PMCID: PMC10931184 DOI: 10.3390/healthcare12050529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Acute kidney injury (AKI) in neonates is associated with increased morbidity and mortality. Theophylline (a methylxanthine) has been shown to prevent neonatal AKI but is seldom used due to its unfavorable profile. Caffeine, another methylxanthine, is utilized ubiquitously to treat apnea of prematurity, but there are no randomized trials evaluating its efficacy in preventing neonatal AKI. This literature review aims to summarize the existing research pertaining to the relationship between caffeine and neonatal AKI. The review was conducted using Pubmed, Embase, Google Scholar, and Cochrane. Inclusion criteria incorporated empirical studies, being published in English, and being available electronically. All eight studies identified were included. Seven studies found caffeine-exposed premature neonates had lower rates of AKI than caffeine-unexposed neonates. Four found reduced AKI severity with caffeine exposure. One study included term neonates and did not find a difference in the AKI rate between caffeine-exposed and non-exposed babies. Limitations include exclusively observational studies, short study periods, heterogenous definitions of prematurity, and a lack of assessment of dose-effect relationships. In conclusion, premature neonates exposed to caffeine appear to have lower rates and potentially less severe AKI. Further research is needed before caffeine can be considered for use in the primary prevention of neonatal AKI.
Collapse
Affiliation(s)
- Nimisha Aithal
- Department of Pediatrics, Townsville University Hospital, Townsville, QLD 4811, Australia
| | - Yogavijayan Kandasamy
- Department of Neonatology, Townsville University Hospital, Townsville, QLD 4811, Australia
- College of Medicine and Dentistry, James Cook University, Townsville, QLD 4814, Australia
| |
Collapse
|
20
|
Dumont O, Iacono D, Jacob A, Aggarwal A, Hagmann SHF. Experience With a Vancomycin-sparing Empiric Antibiotic Guideline for Late-onset Sepsis in a Level-4 Neonatal Intensive Care Unit. Pediatr Infect Dis J 2024; 43:e49-e51. [PMID: 38108378 DOI: 10.1097/inf.0000000000004172] [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: 12/19/2023]
Abstract
A vancomycin-sparing guideline for suspected late-onset sepsis helped reduce vancomycin usage in our level-4 neonatal intensive care unit. Significant reduction in overall vancomycin use, with its likely unit-wide beneficial downstream effects, may need to be measured against the rare case of methicillin-resistant Staphylococcus aureus infection and delayed effective therapy.
Collapse
Affiliation(s)
- Olivia Dumont
- From the Department of Pharmacy
- Steven and Alexandra Cohen Children's Medical Center of New York/Northwell Health, New Hyde Park, New York
| | - Denise Iacono
- From the Department of Pharmacy
- Steven and Alexandra Cohen Children's Medical Center of New York/Northwell Health, New Hyde Park, New York
| | - Alby Jacob
- From the Department of Pharmacy
- Steven and Alexandra Cohen Children's Medical Center of New York/Northwell Health, New Hyde Park, New York
| | - Alpna Aggarwal
- Steven and Alexandra Cohen Children's Medical Center of New York/Northwell Health, New Hyde Park, New York
- Division of Neonatal Medicine
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, New York
| | - Stefan H F Hagmann
- Steven and Alexandra Cohen Children's Medical Center of New York/Northwell Health, New Hyde Park, New York
- Division of Pediatric Infectious Diseases
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Hempstead, New York
| |
Collapse
|
21
|
do Vale MS, Marques PF, Cavalcante MCV, Brito MN, dos Santos AM, Salgado-Filho N, Duarte JLMB. Renal deficit and associated factors in children born with low birth weight. J Bras Nefrol 2024; 46:62-69. [PMID: 37015048 PMCID: PMC10962415 DOI: 10.1590/2175-8239-jbn-2022-0154en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/13/2023] [Indexed: 04/06/2023] Open
Abstract
INTRODUCTION Kidney problems may be due to low birth weight alone or may occur in association with other conditions. The objective this study was to evaluate the association between maternal and birth characteristics, anthropometric measurements, and kidney function deficit in low birth weight infants. METHODS Cross-sectional study with children who were born weighing < 2500 grams and were under outpatient follow-up. Maternal factors investigated were prenatal care and presence of hypertension, diabetes, and infection during pregnancy. The children's variables were sex, gestational age, birth weight, Apgar score, use of nephrotoxic medications, age, body weight at the time of evaluation, height, and serum creatinine and cystatin C dosages. The glomerular filtration rate (GFR) was estimated with the combined Zapittelli equation. Multivariate logistic regression model was used for identification of associated factors, with renal function deficit (GFR < 60 mL/min/1.73 m2) as the dependent variable. RESULTS Of the 154 children evaluated, 34.42% had kidney function deficit. Most of them had a gestational age > 32 weeks (56.6%), a mean birth weight of 1439.7 grams, and mean estimated GFR of 46.9 ± 9.3 mL/min/1.73 m2. There was a significant association of GFR < 60 mL/min/1.73 m2 with children's current weight and use of nephrotoxic drugs. DISCUSSION Children born with low birth weight had a high prevalence of kidney function deficit and current normal weight was a protective factor while the use of nephrotoxic drugs during perinatal period increased the chance of kidney deficit. These findings reinforce the need to evaluate the kidney function in these children, especially those who use nephrotoxic drugs.
Collapse
Affiliation(s)
| | | | | | - Mateus Noleto Brito
- Universidade Federal do Maranhão, Departamento de Medicina I, São
Luís, MA, Brazil
| | | | | | - José Luiz M. B. Duarte
- Universidade do Estado do Rio de Janeiro, Departamento de Pediatria,
Rio de Janeiro, RJ, Brazil
| |
Collapse
|
22
|
Yan B, Li Y, Sun M, Meng Y, Li X. Variables related to bronchopulmonary dysplasia severity: a Six-Year retrospective study. J Matern Fetal Neonatal Med 2023; 36:2248335. [PMID: 37580063 DOI: 10.1080/14767058.2023.2248335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 06/29/2023] [Accepted: 08/10/2023] [Indexed: 08/16/2023]
Abstract
OBJECTIVES This was a retrospective observational study conducted in a tertiary neonatal intensive care unit, in order to investigate factors which influenced the severity of bronchopulmonary dysplasia under NICHD new classification. METHODS Six years of clinical data with different grades of bronchopulmonary dysplasia patients were collected and analyzed, bivariate ordinal logistic regression model and multivariable ordinal logistic regression model were used with sensitivity analyses. RESULTS We identified seven variables were associated with the severity of BPD via a bivariate ordinal logistic regression model, including the level of referral hospital (OR 0.273;95% CI 0.117, 0.636), method of caffeine administration (OR 00.418;95% CI 0.177, 0.991), more than two occurrences of reintubation (OR 4.925;95% CI 1.878, 12.915), CPAP reapplication (OR 2.255;95% CI 1.059, 4.802), presence of positive sputum cultures (OR 2.574;95% CI 1.200, 5.519), the cumulative duration of invasive ventilation (OR 1.047;95% CI 1.017, 1.078), and postmenstrual age at the discontinuation of oxygen supplementation (OR 1.190;95% CI 1.027, 1.38). These seven variables were further analyzed via all multivariable ordinal logistic regression models, and we found that tertiary hospital birth and early administration of caffeine could reduce the severity of BPD by approximately 70% (OR 0.263;95% CI 0.090, 0.770) and 60% (OR 0.371;95% CI 0.138, 0.995), respectively. In contrast, multiple reintubations were related to higher BPD severity with an OR of 3.358 (95% CI 1.002, 11.252). CONCLUSION Improving perinatal care in level II hospitals, standardized caffeine administration, and optimized extubation strategy could potentially decrease the severity of BPD.
Collapse
Affiliation(s)
- Beibei Yan
- Department of Neonatology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
- Department of Neonatology, Jinan Children's Hospital, Jinan, Shandong, P.R. China
| | - Yunxia Li
- Department of Neonatology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
- Department of Neonatology, Jinan Children's Hospital, Jinan, Shandong, P.R. China
| | - Mingying Sun
- Department of Neonatology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
- Department of Neonatology, Jinan Children's Hospital, Jinan, Shandong, P.R. China
| | - Yan Meng
- Department of Neonatology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
- Department of Neonatology, Jinan Children's Hospital, Jinan, Shandong, P.R. China
| | - Xiaoying Li
- Department of Neonatology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, P.R. China
- Department of Neonatology, Jinan Children's Hospital, Jinan, Shandong, P.R. China
| |
Collapse
|
23
|
Villanueva-García D, Ghezzi M, Mora-Medina P, Hernández-Ávalos I, Olmos-Hernández A, Casas-Alvarado A, Lezama-García K, Domínguez-Oliva A, Rodríguez-González D, Marcet-Rius M. Caffeine Administration in Piglets with Low Birthweight and Low Vitality Scores, and Its Effect on Physiological Blood Profile, Acid-Base Balance, Gas Exchange, and Infrared Thermal Response. Animals (Basel) 2023; 13:3491. [PMID: 38003109 PMCID: PMC10668766 DOI: 10.3390/ani13223491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/18/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Intrapartum asphyxia, fetal hypoxia, and their consequences (e.g., acidosis, hypercapnia, hypoglycemia, and hypothermia) are the main factors related to physio-metabolic imbalances that increase neonatal mortality in piglets, particularly in piglets with low birthweight and low vitality scores. This study aimed to evaluate the effect of three different doses of caffeine (10, 20, and 30 mg/kg) administered orally to 480 newborn piglets with low birthweight and low vitality scores. Blood gas parameters (pH, pO2, pCO2, and HCO3-), physio-metabolic profile (Ca++, glucose, and lactate), and the thermal response assessed through infrared thermography in four thermal windows (ocular, auricular, snout, and hindlimb) and rectal temperature were evaluated during the first 24 h of life. Doses of 30 mg/kg resulted in significant differences at 24 h for all evaluated parameters, suggesting that caffeine administration improved the cardiorespiratory function and metabolic activity of piglets by reducing acidosis, restoring glycemia, and increasing surface and rectal temperature. In conclusion, caffeine at 30 mg/kg could be suggested as an appropriate dose to use in piglets with low birthweight and low vitality scores. Future research might need to study the presentation of adverse effects due to higher caffeine concentrations.
Collapse
Affiliation(s)
- Dina Villanueva-García
- Division of Neonatology, National Institute of Health, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
| | - Marcelo Ghezzi
- Animal Welfare Area, Faculty of Veterinary Sciences (FCV), Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), University Campus, Tandil 7000, Argentina
| | - Patricia Mora-Medina
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Mexico
| | - Ismael Hernández-Ávalos
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Mexico
| | - Adriana Olmos-Hernández
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Alejandro Casas-Alvarado
- Neurophysiology, Behavior and Animal Welfare Assesment, DPAA, Xochimilco Campus, Universidad Autónoma Metropolitana, México City 04960, Mexico (K.L.-G.)
| | - Karina Lezama-García
- Neurophysiology, Behavior and Animal Welfare Assesment, DPAA, Xochimilco Campus, Universidad Autónoma Metropolitana, México City 04960, Mexico (K.L.-G.)
| | - Adriana Domínguez-Oliva
- Neurophysiology, Behavior and Animal Welfare Assesment, DPAA, Xochimilco Campus, Universidad Autónoma Metropolitana, México City 04960, Mexico (K.L.-G.)
| | - Daniela Rodríguez-González
- Neurophysiology, Behavior and Animal Welfare Assesment, DPAA, Xochimilco Campus, Universidad Autónoma Metropolitana, México City 04960, Mexico (K.L.-G.)
| | - Miriam Marcet-Rius
- Department of Animal Behaviour and Welfare, Research Institute in Semiochemistry and Applied Ethology (IRSEA), 84400 Apt, France
| |
Collapse
|
24
|
Mathevula H, Schellack N, Orubu S, Godman B, Matlala M. Off-Label and Unlicenced Medicine Use among Hospitalised Children in South Africa: Practice and Policy Implications. PHARMACY 2023; 11:174. [PMID: 37987384 PMCID: PMC10661306 DOI: 10.3390/pharmacy11060174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/25/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Information regarding off-label and unlicensed medicine use among South African children is limited. This is a concern as the prescribing of off-label and unlicensed medicines can lead to issues of effectiveness and safety as well as raise liability issues in the event of adverse events. This potentially exposes physicians to legal penalties. Consequently, we sought to determine the prevalence of off-label and unlicensed medicine use among paediatric patients in South Africa to provide future direction. METHODS This study retrospectively examined the use of medicine in a point-prevalence survey study (PPS) involving paediatric patients aged (0-2 years) admitted to selected public hospitals in Gauteng Province, South Africa. Data were collected per hospital over two days between February 2022 and July 2022. Demographics, duration of treatment, diagnosis, and medicines prescribed were collected from patient medical records using a mobile application. Prescribed medicines were reviewed against the medicine formularies and other databases to assess their appropriateness. RESULTS From three academic hospitals, 184 patient records were reviewed. A total of 592 medicines were dispensed, of which 379 (64.0%) were licensed and 213 (36.0%) were used off-label/unlicensed for paediatric patients 0-2 years of age. The most prevalent off-label and unlicensed medicines were multivitamins (n = 32, 15.0%) and ampicillin injections (n = 15, 7.0%). CONCLUSION The frequency of unlicensed and off-label medicine prescribing shown in this study is consistent with the literature and can be considered high. This practice can pose a risk because it adversely affects patients if not properly regulated. Attention is needed to ensure future high-quality, safe, and effective use of medicines.
Collapse
Affiliation(s)
- Hlayiseka Mathevula
- School of Pharmacy, Sefako Makgatho Health Sciences University, Molotlegi Street, Pretoria 0204, South Africa; (B.G.); (M.M.)
| | - Natalie Schellack
- Department of Pharmacology, University of Pretoria, Pretoria 0084, South Africa;
| | - Samuel Orubu
- Pharmacy Department, Niger Delta University, Yenagoa P.O. BOX 72, Nigeria;
- Global Strategy Lab, York University, Toronto, ON 4700, Canada
| | - Brian Godman
- School of Pharmacy, Sefako Makgatho Health Sciences University, Molotlegi Street, Pretoria 0204, South Africa; (B.G.); (M.M.)
- Department of Pharmacoepidemiology, Strathclyde Institute of Pharmacy and Biomedical Science (SIPBS), University of Strathclyde, Glasgow G4 0RE, UK
| | - Moliehi Matlala
- School of Pharmacy, Sefako Makgatho Health Sciences University, Molotlegi Street, Pretoria 0204, South Africa; (B.G.); (M.M.)
| |
Collapse
|
25
|
Pichichero ME. Variability of vaccine responsiveness in early life. Cell Immunol 2023; 393-394:104777. [PMID: 37866234 DOI: 10.1016/j.cellimm.2023.104777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/18/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
Vaccinations in early life elicit variable antibody and cellular immune responses, sometimes leaving fully vaccinated children unprotected against life-threatening infectious diseases. Specific immune cell populations and immune networks may have a critical period of development and calibration in a window of opportunity occurring during the first 100 days of early life. Among the early life determinants of vaccine responses, this review will focus on modifiable factors involving development of the infant microbiota and metabolome: antibiotic exposure, breast versus formula feeding, and Caesarian section versus vaginal delivery of newborns. How microbiota may serve as natural adjuvants for vaccine responses and how microbiota-derived metabolites influence vaccine responses are also reviewed. Early life poor vaccine responsiveness can be linked to increased infection susceptibility because both phenotypes share similar immunity dysregulation profiles. An early life pre-vaccination endotype, when interventions have the highest potential for success, should be sought that predicts vaccine response trajectories.
Collapse
Affiliation(s)
- Michael E Pichichero
- Center for Infectious Diseases and Immunology, Research Institute, Rochester General Hospital, 1425 Portland Ave, Rochester, NY 14621, USA.
| |
Collapse
|
26
|
Schill EM, Joyce EL, Floyd AN, Udayan S, Rusconi B, Gaddipati S, Barrios BE, John V, Kaye ME, Kulkarni DH, Pauta JT, McDonald KG, Newberry RD. Vancomycin-induced gut microbial dysbiosis alters enteric neuron-macrophage interactions during a critical period of postnatal development. Front Immunol 2023; 14:1268909. [PMID: 37901245 PMCID: PMC10602895 DOI: 10.3389/fimmu.2023.1268909] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/20/2023] [Indexed: 10/31/2023] Open
Abstract
Vancomycin is a broad-spectrum antibiotic widely used in cases of suspected sepsis in premature neonates. While appropriate and potentially lifesaving in this setting, early-life antibiotic exposure alters the developing microbiome and is associated with an increased risk of deadly complications, including late-onset sepsis (LOS) and necrotizing enterocolitis (NEC). Recent studies show that neonatal vancomycin treatment disrupts postnatal enteric nervous system (ENS) development in mouse pups, which is in part dependent upon neuroimmune interactions. This suggests that early-life antibiotic exposure could disrupt these interactions in the neonatal gut. Notably, a subset of tissue-resident intestinal macrophages, muscularis macrophages, has been identified as important contributors to the development of postnatal ENS. We hypothesized that vancomycin-induced neonatal dysbiosis impacts postnatal ENS development through its effects on macrophages. Using a mouse model, we found that exposure to vancomycin in the first 10 days of life, but not in adult mice, resulted in an expansion of pro-inflammatory colonic macrophages by increasing the recruitment of bone-marrow-derived macrophages. Single-cell RNA sequencing of neonatal colonic macrophages revealed that early-life vancomycin exposure was associated with an increase in immature and inflammatory macrophages, consistent with an influx of circulating monocytes differentiating into macrophages. Lineage tracing confirmed that vancomycin significantly increased the non-yolk-sac-derived macrophage population. Consistent with these results, early-life vancomycin exposure did not expand the colonic macrophage population nor decrease enteric neuron density in CCR2-deficient mice. Collectively, these findings demonstrate that early-life vancomycin exposure alters macrophage number and phenotypes in distinct ways compared with vancomycin exposure in adult mice and results in altered ENS development.
Collapse
Affiliation(s)
- Ellen Merrick Schill
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Elisabeth L. Joyce
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Alexandria N. Floyd
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Sreeram Udayan
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Brigida Rusconi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Shreya Gaddipati
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Bibiana E. Barrios
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Vini John
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Mitchell E. Kaye
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Devesha H. Kulkarni
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Jocelyn T. Pauta
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Keely G. McDonald
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Rodney D. Newberry
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
27
|
Roilides E, Bradley JS, Lonchar J, Huntington JA, Wickremasingha P, Su FH, Bruno CJ, Johnson MG. Subgroup analysis of phase 2 study of ceftolozane/tazobactam in neonates and young infants with pyelonephritis. Microbiol Spectr 2023; 11:e0180023. [PMID: 37698430 PMCID: PMC10581202 DOI: 10.1128/spectrum.01800-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/13/2023] [Indexed: 09/13/2023] Open
Abstract
Ceftolozane/tazobactam is approved for the treatment of patients from birth to <18 y old with complicated urinary tract infections (cUTI). This post hoc analysis evaluated the safety, efficacy, and pharmacokinetics (PK) of ceftolozane/tazobactam compared with meropenem in neonates and young infants. NCT03230838 was a phase 2, randomized, active comparator-controlled, double-blind study of patients from birth to <18 y of age with cUTI, including pyelonephritis, given ceftolozane/tazobactam or meropenem in a 3:1 ratio. This subset analysis included only neonates and young infants < 3 mo of age. The microbiologic modified intent-to-treat population (mMITT) included 20 patients (ceftolozane/tazobactam, n = 14; meropenem, n = 6). All patients had pyelonephritis at baseline; two patients in each treatment group had bacteremia (overall 4/20, 20%). Escherichia coli was the most common baseline pathogen (overall 16/20, 80%). Safety and efficacy results were similar between treatment groups and consistent with the overall pediatric population. There were no serious drug-related adverse events (AEs), no discontinuations due to AEs, and no AEs leading to death in either treatment group. For the ceftolozane/tazobactam and meropenem treatment groups, clinical cure rates in the mMITT population were 92.9% and 100%, respectively. The population PK analysis of neonates and young infants demonstrated similar ceftolozane and tazobactam exposures to those of adults, achieving pharmacodynamic targets associated with clinical and microbiologic cure. Ceftolozane/tazobactam has a favorable safety profile and achieves high clinical cure and microbiologic eradication rates in neonates and young infants < 3 mo of age with cUTI and pyelonephritis. IMPORTANCE Extrapolation of antibacterial agent pharmacokinetics from adults to newborns and young infants may not be appropriate; similarly, the clinical manifestations of infectious diseases and outcomes following antibacterial treatment may not be similar. Ceftolozane/tazobactam is an antibacterial drug combination active against Pseudomonas aeruginosa and other multidrug-resistant gram-negative bacteria. A clinical study led to the approval for ceftolozane/tazobactam in patients from birth to 18 y of age who have complicated urinary tract infections, including those with serious kidney infections. Based on data collected during that clinical study, we compared newborns and young infants who were treated with ceftolozane/tazobactam (14 patients) and those who were treated with meropenem (6 patients). We found that ceftolozane/tazobactam treatment of newborns and young infants up to 3 mo of age who have complicated urinary tract infections demonstrated a favorable safety profile and high clinical cure and microbiologic eradication rates, similar to meropenem.
Collapse
Affiliation(s)
- Emmanuel Roilides
- Third Department of Pediatrics, Infectious Diseases Unit, School of Medicine, Aristotle University and Hippokration General Hospital, Thessaloniki, Greece
| | - John S. Bradley
- Department of Pediatrics, University of California, San Diego School of Medicine and Rady Children’s Hospital of San Diego, San Diego, California, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Gallaway KA, Cann K, Oetting K, Rothenberger M, Raibulet A, Slaven JE, Suhrie K, Tillman EM. The Potential Impact of Preemptive Pharmacogenetic Genotyping in the Neonatal Intensive Care Unit. J Pediatr 2023; 259:113489. [PMID: 37201679 DOI: 10.1016/j.jpeds.2023.113489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/26/2023] [Accepted: 05/10/2023] [Indexed: 05/20/2023]
Abstract
OBJECTIVE To evaluate the use of drugs with pharmacogenomic (PGx) guidelines from the Clinical Pharmacogenetics Implementation Consortium in early childhood. STUDY DESIGN A retrospective observational study of patients admitted to the neonatal intensive care (NICU) between 2005 and 2018 with at least 1 subsequent hospitalization at or after 5 years of age was performed to determine PGx drug exposure. Data regarding hospitalizations, drug exposures, gestational age, birth weight, and congenital anomalies and/or a primary genetic diagnosis were collected. Incidence of PGx drug and drug class exposures was determined and patient specific factors predictive of exposure were investigated. RESULTS During the study, 19 195 patients received NICU care and 4196 (22%) met study inclusion; 67% received 1-2, 28% 3-4, and 5% 5 or more PGx-drugs in early childhood. Preterm gestation, low birth weight (<2500 g), and the presence of any congenital anomalies and/or a primary genetic diagnosis were statistically significant predictors of Clinical Pharmacogenetics Implementation Consortium drug exposures (P < .01, P < .01, P < .01, respectively). CONCLUSIONS Preemptive PGx testing in patients in the NICU could have a significant impact on medical management during the NICU stay and throughout early childhood.
Collapse
Affiliation(s)
- Katherine A Gallaway
- Division of Pediatric Critical Care, Indiana University School of Medicine, Indianapolis, IN
| | - Kayla Cann
- Purdue University College of Pharmacy, Purdue University, West Lafayette, IN
| | - Katherine Oetting
- Purdue University College of Pharmacy, Purdue University, West Lafayette, IN
| | - Mary Rothenberger
- Purdue University College of Pharmacy, Purdue University, West Lafayette, IN
| | - Andra Raibulet
- College of Pharmacy and Health Sciences, Butler University, Indianapolis, IN
| | - James E Slaven
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN
| | - Kristen Suhrie
- Division of Neonatology, Department of Pediatrics, and Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Emma M Tillman
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN.
| |
Collapse
|
29
|
Steflik HJ, Charlton JR, Briley M, Selewski DT, Gist KM, Hanna MH, Askenazi D, Griffin R. Neonatal nephrotoxic medication exposure and early acute kidney injury: results from the AWAKEN study. J Perinatol 2023; 43:1029-1037. [PMID: 37100984 PMCID: PMC10725773 DOI: 10.1038/s41372-023-01684-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND We aimed to describe nephrotoxic medication exposure and investigate associations between exposure and acute kidney injury (AKI) in the neonatal intensive care unit during the first postnatal week. DESIGN/METHODS Secondary analysis of the AWAKEN cohort. We evaluated nephrotoxic medication exposure during the first postnatal week and associations with AKI using time-varying Cox proportional hazard regressions models. Nephrotoxic medication exposure categories were defined as: no nephrotoxic medication, nephrotoxic medications excluding aminoglycosides, aminoglycoside alone, and aminoglycoside and another nephrotoxic medication. RESULTS Of 2162 neonates, 1616 (74.7%) received ≥1 nephrotoxic medication. Aminoglycoside receipt was most common (72%). AKI developed in 211(9.8%) neonates and was associated with a nephrotoxic medication exposure (p < 0.01). Nephrotoxic medication exposures including a nephrotoxic medication excluding aminoglycoside (aHR 3.14, 95% CI 1.31-7.55) and aminoglycoside and another nephrotoxic medication (aHR 4.79, 95% CI 2.19-10.50) were independently associated with AKI and severe AKI (stage 2/3), respectively. CONCLUSIONS Nephrotoxic medication exposure in critically ill infants is common during the first postnatal week. Specific nephrotoxic medication exposure, principally aminoglycosides with another nephrotoxic medication, are independently associated with early AKI.
Collapse
Affiliation(s)
- Heidi J Steflik
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA.
| | | | - Meagan Briley
- Department of Pediatrics, Vanderbilt University, Nashville, TN, USA
| | - David T Selewski
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Katja M Gist
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - David Askenazi
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Russell Griffin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
30
|
Suhrie K, Tillman EM. Pharmacogenetic informed care in early childhood: options for improving access and health equity. Pharmacogenomics 2023; 24:579-582. [PMID: 37466126 DOI: 10.2217/pgs-2023-0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Affiliation(s)
- Kristen Suhrie
- Department of Pediatrics, Division of Neonatology, & Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Emma M Tillman
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
31
|
Sánchez PJ, Prusakov P, de Alba Romero C, Zamora-Flores E, Reyes Escamilla MC, White NO, Miller RR, Moraille R, Theile AR, Magers JK. Short-course empiric antibiotic therapy for possible early-onset sepsis in the NICU. J Perinatol 2023; 43:741-745. [PMID: 36813903 DOI: 10.1038/s41372-023-01634-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 02/05/2023] [Accepted: 02/10/2023] [Indexed: 02/24/2023]
Abstract
OBJECTIVE On 2/2019, the Neonatal Antimicrobial Stewardship Program at Nationwide Children's Hospital recommended reducing empirical antibiotic therapy for early-onset sepsis (EOS) from 48 to 24 hours with a TIME-OUT. We describe our experience with this guideline and assess its safety. METHODS Retrospective review of newborns evaluated for possible EOS at 6 NICUs from 12/2018-7/2019. Safety endpoints were re-initiation of antibiotics within 7 days after discontinuation of the initial course, positive bacterial blood or cerebrospinal fluid culture in the 7 days after antibiotic discontinuation, and overall and sepsis-related mortality. RESULT Among 414 newborns evaluated for EOS, 196 (47%) received a 24 hour rule-out sepsis antibiotic course while 218 (53%) were managed with a 48 hour course. The 24-hour rule-out group were less likely to have antibiotics re-initiated and did not differ in the other predefined safety endpoints. CONCLUSION Antibiotic therapy for suspected EOS may be discontinued safely within 24 hours.
Collapse
Affiliation(s)
- Pablo J Sánchez
- Divisions of Neonatology and Pediatric Infectious Diseases, Department of Pediatrics, Nationwide Children's Hospital, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Pavel Prusakov
- Department of Pharmacy, Nationwide Children's Hospital, Columbus, OH, USA.
| | - Concepción de Alba Romero
- Division of Neonatology, Department of Pediatrics, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Neonatology, Hospital 12 de Octubre, Madrid, Spain
| | - Elena Zamora-Flores
- Division of Neonatology, Department of Pediatrics, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Neonatology, Gregorio Marañon University Hospital, Madrid, Spain
| | - María Camila Reyes Escamilla
- Division of Neonatology, Department of Pediatrics, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Neonatology, Universidad del Valle, Cali, Colombia
| | | | | | | | | | | |
Collapse
|
32
|
Mota-Rojas D, Villanueva-García D, Hernández-Ávalos I, Casas-Alvarado A, Domínguez-Oliva A, Lezama-García K, Miranda-Cortés A, Martínez-Burnes J. Cardiorespiratory and Neuroprotective Effects of Caffeine in Neonate Animal Models. Animals (Basel) 2023; 13:1769. [PMID: 37889643 PMCID: PMC10252037 DOI: 10.3390/ani13111769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 07/15/2023] Open
Abstract
Caffeine is widely used to improve neonatal health in animals with low vitality. Due to its pharmacokinetics and pharmacodynamics, caffeine stimulates the cardiorespiratory system by antagonism of adenosine receptors and alteration in Ca+2 ion channel activity. Moreover, the availability of intracellular Ca+2 also has positive inotropic effects by increasing heart contractibility and by having a possible positive effect on neonate vitality. Nonetheless, since neonatal enzymatic and tissular systems are immature at birth, there is a controversy about whether caffeine is an effective therapy for newborns. This review aims to analyze the basic concepts of caffeine in neonatal animal models (rat and mouse pups, goat kids, lambs, and piglets), and it will discuss the neuroprotective effect and its physiological actions in reducing apnea in newborns.
Collapse
Affiliation(s)
- Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Dina Villanueva-García
- Division of Neonatology, National Institute of Health, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
| | - Ismael Hernández-Ávalos
- Clinical Pharmacology and Veterinary Anesthesia, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán 54714, Mexico; (I.H.-Á.)
| | - Alejandro Casas-Alvarado
- Neurophysiology, Behavior and Animal Welfare Assessment, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Adriana Domínguez-Oliva
- Neurophysiology, Behavior and Animal Welfare Assessment, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Karina Lezama-García
- Neurophysiology, Behavior and Animal Welfare Assessment, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Agatha Miranda-Cortés
- Clinical Pharmacology and Veterinary Anesthesia, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán 54714, Mexico; (I.H.-Á.)
| | - Julio Martínez-Burnes
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Tamaulipas, Victoria City 87000, Mexico
| |
Collapse
|
33
|
Vale MSD, Marques PF, Cavalcante MCV, Brito MN, Santos AMD, Salgado-Filho N, Duarte JLMB. Déficit renal e fatores associados em crianças nascidas com baixo peso. J Bras Nefrol 2023. [DOI: 10.1590/2175-8239-jbn-2022-0154pt] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
RESUMO Introdução: Problemas renais podem ser devido apenas ao baixo peso ao nascer ou podem ocorrer em associação com outras condições. O objetivo deste estudo foi avaliar a associação entre características maternas e de nascimento, medidas antropométricas e déficit da função renal em bebês de baixo peso ao nascer. Métodos: Estudo transversal com crianças que nasceram com peso < 2500 gramas e estavam sob acompanhamento ambulatorial. Os fatores maternos investigados foram cuidados pré-natal e presença de hipertensão, diabetes e infecção durante a gravidez. As variáveis das crianças foram sexo, idade gestacional, peso ao nascer, índice Apgar, uso de medicamentos nefrotóxicos, idade, peso corporal no momento da avaliação, altura e dosagens séricas de creatinina e cistatina C. A taxa de filtração glomerular (TFG) foi estimada com a equação combinada de Zapittelli. Utilizou-se um modelo de regressão logística multivariada para identificação de fatores associados, com déficit da função renal (TFG < 60 mL/min/1,73 m2) como variável dependente. Resultados: Das 154 crianças avaliadas, 34,42% apresentaram déficit da função renal. A maioria tinha idade gestacional > 32 semanas (56,6%), peso médio ao nascer de 1439,7 gramas, e TFG média estimada de 46,9 ± 9,3 mL/min/1,73 m2. Houve uma associação significativa da TFG < 60 mL/min/1,73 m2 com o peso atual das crianças e o uso de medicamentos nefrotóxicos. Discussão: Crianças nascidas com baixo peso apresentaram alta prevalência de déficit da função renal e o peso atual normal foi um fator de proteção, enquanto o uso de medicamentos nefrotóxicos durante o período perinatal aumentou a chance de déficit renal. Estes achados reforçam a necessidade de avaliar a função renal destas crianças, especialmente aquelas que usam medicamentos nefrotóxicos.
Collapse
|
34
|
Villanueva-Bueno C, Montecatine-Alonso E, Jiménez-Parrilla F, González-López M, Manrique-Rodríguez S, Moreno-Ramos F, Cañete-Ramírez C, Dolz E, García-Robles A, Caro-Teller JM, Moral-Pumarega MT, Bergon-Sendin E, Gómez-Trevecedo Calvo MT, Gallego-Fernández C, del Vayo-Benito CÁ, Mejías-Trueba M, Gil-Navarro MV. Antimicrobial Defined Daily Dose in Neonatal Population: Validation in the Clinical Practice. Antibiotics (Basel) 2023; 12:antibiotics12030602. [PMID: 36978469 PMCID: PMC10044623 DOI: 10.3390/antibiotics12030602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/21/2023] [Accepted: 03/09/2023] [Indexed: 03/19/2023] Open
Abstract
Background: Currently, there is no validated method for estimating antimicrobial consumption in the neonatal population, as it exists for adults using Defined Daily Doses (DDD). In neonatology, although there are different methods, each one with advantages and disadvantages, there is no unified criterion for use. The aim of this study is to validate the neonatal DDD designed as a new standardised form of antimicrobial consumption over this population. Methods: The validation of the neonatal DDD, Phase II of the research project, was carried out through a descriptive observational study. Periodic cut-offs were performed to collect antimicrobial prescriptions of neonates admitted to the neonatology and intensive care units of nine Spanish hospitals. The data collected included demographic variables (gestational age, postnatal age, weight and sex), antimicrobial dose, frequency and route of administration. The selection of the optimal DDD value takes into account power value, magnitude obtained from the differences in the DDD, statistical significance obtained by the Wilcoxon test and degree of agreement in the stipulated doses. Results: Set of 904 prescriptions were collected and finally 860 were analysed based on the established criteria. The antimicrobials were mostly prescribed in the intensive care unit (63.1%). 32 different antimicrobials were collected, and intravenous administration was the most commonly used route. Neonatal DDD were defined for 11 different antimicrobials. A potency > 80% was obtained in 7 antibiotics. The 57.1% of the selected DDD correspond to phase I and 21.4% from phase II. Conclusion: DDD validation has been achieved for the majority of intravenously administered antimicrobials used in clinical practice in the neonatal population. This will make it possible to have an indicator that will be used globally to estimate the consumption of antimicrobials in this population, thus confirming its usefulness and applicability.
Collapse
Affiliation(s)
- Cristina Villanueva-Bueno
- Department of Pharmacy, Hospital Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañon, 28007 Madrid, Spain
- Correspondence:
| | | | | | - María González-López
- Department of Neonatology, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Silvia Manrique-Rodríguez
- Department of Pharmacy, Hospital Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañon, 28007 Madrid, Spain
| | | | - Carme Cañete-Ramírez
- Department of Pharmacy, Hospital Universitario Vall d’Hebron, 08035 Barcelona, Spain
| | - Elisenda Dolz
- Department of Pharmacy, Complejo Hospitalario Universitario Insular-Materno Infantil, 35016 Las Palmas de Gran Canarias, Spain
| | - Ana García-Robles
- Division of Pharmacy, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - José Manuel Caro-Teller
- Department of Pharmacy, Hospital Universitario 12 de Octubre, Research Institute 12 de Octubre i+12, 28041 Madrid, Spain
| | | | - Elena Bergon-Sendin
- Department of Neonatology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | | | | | | | - Marta Mejías-Trueba
- Department of Pharmacy, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain (M.M.-T.)
- Department of Infectious Diseases, Microbiology and Parasitology, Institute of Biomedicine of Seville, University Hospital Virgen del Rocío, Spanish National Research Council, University of Seville, 41013 Seville, Spain
| | - María Victoria Gil-Navarro
- Department of Pharmacy, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain (M.M.-T.)
- Institute of Biomedicine of Seville, Virgen del Rocío University Hospital, CSIC, University of Seville, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, 28029 Madrid, Spain
| | | |
Collapse
|
35
|
Developmental Pharmacokinetics of Antibiotics Used in Neonatal ICU: Focus on Preterm Infants. Biomedicines 2023; 11:biomedicines11030940. [PMID: 36979919 PMCID: PMC10046592 DOI: 10.3390/biomedicines11030940] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/22/2023] Open
Abstract
Neonatal Infections are among the most common reasons for admission to the intensive care unit. Neonatal sepsis (NS) significantly contributes to mortality rates. Empiric antibiotic therapy of NS recommended by current international guidelines includes benzylpenicillin, ampicillin/amoxicillin, and aminoglycosides (gentamicin). The rise of antibacterial resistance precipitates the growth of the use of antibiotics of the Watch (second, third, and fourth generations of cephalosporines, carbapenems, macrolides, glycopeptides, rifamycins, fluoroquinolones) and Reserve groups (fifth generation of cephalosporines, oxazolidinones, lipoglycopeptides, fosfomycin), which are associated with a less clinical experience and higher risks of toxic reactions. A proper dosing regimen is essential for effective and safe antibiotic therapy, but its choice in neonates is complicated with high variability in the maturation of organ systems affecting drug absorption, distribution, metabolism, and excretion. Changes in antibiotic pharmacokinetic parameters result in altered efficacy and safety. Population pharmacokinetics can help to prognosis outcomes of antibiotic therapy, but it should be considered that the neonatal population is heterogeneous, and this heterogeneity is mainly determined by gestational and postnatal age. Preterm neonates are common in clinical practice, and due to the different physiology compared to the full terms, constitute a specific neonatal subpopulation. The objective of this review is to summarize the evidence about the developmental changes (specific for preterm and full-term infants, separately) of pharmacokinetic parameters of antibiotics used in neonatal intensive care units.
Collapse
|
36
|
Good PI, Li L, Hurst HA, Serrano Herrera I, Xu K, Rao M, Bateman DA, Al-Awqati Q, D’Agati VD, Costantini F, Lin F. Low nephron endowment increases susceptibility to renal stress and chronic kidney disease. JCI Insight 2023; 8:e161316. [PMID: 36626229 PMCID: PMC9977438 DOI: 10.1172/jci.insight.161316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Preterm birth results in low nephron endowment and increased risk of acute kidney injury (AKI) and chronic kidney disease (CKD). To understand the pathogenesis of AKI and CKD in preterm humans, we generated potentially novel mouse models with a 30%-70% reduction in nephron number by inhibiting or deleting Ret tyrosine kinase in the developing ureteric bud. These mice developed glomerular and tubular hypertrophy, followed by the transition to CKD, recapitulating the renal pathological changes seen in humans born preterm. We injected neonatal mice with gentamicin, a ubiquitous nephrotoxic exposure in preterm infants, and detected more severe proximal tubular injury in mice with low nephron number compared with controls with normal nephron number. Mice with low nephron number had reduced proliferative repair with more rapid development of CKD. Furthermore, mice had more profound inflammation with highly elevated levels of MCP-1 and CXCL10, produced in part by damaged proximal tubules. Our study directly links low nephron endowment with postnatal renal hypertrophy, which in this model is maladaptive and results in CKD. Underdeveloped kidneys are more susceptible to gentamicin-induced AKI, suggesting that AKI in the setting of low nephron number is more severe and further increases the risk of CKD in this vulnerable population.
Collapse
Affiliation(s)
| | - Ling Li
- Department of Pediatrics and
| | | | | | - Katherine Xu
- Department of Internal Medicine, Columbia University Vagelos College of Physicians and Surgeons New York, New York, USA
| | - Meenakshi Rao
- Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston Massachusetts, USA
| | | | - Qais Al-Awqati
- Department of Internal Medicine, Columbia University Vagelos College of Physicians and Surgeons New York, New York, USA
| | - Vivette D. D’Agati
- Department of Pathology and Cellular Biology at Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Frank Costantini
- Department of Genetics and Development at Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | | |
Collapse
|
37
|
Baltogianni M, Giapros V, Kosmeri C. Antibiotic Resistance and Biofilm Infections in the NICUs and Methods to Combat It. Antibiotics (Basel) 2023; 12:antibiotics12020352. [PMID: 36830264 PMCID: PMC9951928 DOI: 10.3390/antibiotics12020352] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Neonatal sepsis is an important cause of neonatal morbidity and mortality. A significant proportion of bacteria causing neonatal sepsis is resistant to multiple antibiotics, not only to the usual empirical first-line regimens, but also to second- and third-line antibiotics in many neonatal intensive care units (NICUs). NICUs have unique antimicrobial stewardship goals. Apart from antimicrobial resistance, NICUs have to deal with another problem, namely biofilm infections, since neonates often have central and peripheral lines, tracheal tubes and other foreign bodies for a prolonged duration. The aim of this review is to describe traditional and novel ways to fight antibiotic-resistant bacteria and biofilm infections in NICUs. The topics discussed will include prevention and control of the spread of infection in NICUs, as well as the wise use of antimicrobial therapy and ways to fight biofilm infections.
Collapse
Affiliation(s)
- Maria Baltogianni
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45500 Ioannina, Greece
| | - Vasileios Giapros
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45500 Ioannina, Greece
- Correspondence: ; Tel.: +30-26-5100-7546
| | - Chrysoula Kosmeri
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece
| |
Collapse
|
38
|
Kaufmann M, Eckart F, Rüdiger M, Birdir C, Mense L. [Telemedical Support of Feto-Neonatal Care in One Region - Part I: Demand Analysis Using the Example of East Saxony]. Z Geburtshilfe Neonatol 2023; 227:17-23. [PMID: 36702136 DOI: 10.1055/a-1977-9030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Excellence in feto-neonatal care forms the basis for health in adulthood and requires a collaboration of stakeholders in the health care system. As in other regions, demographic changes such as rural depopulation pose a risk to feto-neonatal care in Eastern Saxony. Areas in need of regional, perinatal collaboration have been identified: (I) multi-professional counselling of families with a suspected fetal disease, (II) immediately available expertise of a neonatologist during neonatal resuscitation, (III) evidence-based neonatal antibiotic therapy, (IV) backtransfer of extremely preterm infants or sick neonates, and (V) adequate psychosocial support of families with extremely preterm infants or sick neonates. Telemedicine enables regional partners to communicate efficiently and gives an audiovisual impression of the patient. The Saxony Center for feto/neonatal Health (SCFNH) collaborates with regional partners to establish a feto-neonatal telemedicine network "Sichere Geburt". The network will be scientifically evaluated and might be of help as a model for other regions with structural challenges.
Collapse
Affiliation(s)
- Maxi Kaufmann
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Fachbereich Neonatologie & Pädiatrische Intensivmedizin, Medizinische Fakultät, TU Dresden, Dresden, Germany
- Zentrum für Feto/Neonatale Gesundheit, Medizinische Fakultät, TU Dresden, Dresden, Germany
| | - Falk Eckart
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Fachbereich Neonatologie & Pädiatrische Intensivmedizin, Medizinische Fakultät, TU Dresden, Dresden, Germany
- Zentrum für Feto/Neonatale Gesundheit, Medizinische Fakultät, TU Dresden, Dresden, Germany
| | - Mario Rüdiger
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Fachbereich Neonatologie & Pädiatrische Intensivmedizin, Medizinische Fakultät, TU Dresden, Dresden, Germany
- Zentrum für Feto/Neonatale Gesundheit, Medizinische Fakultät, TU Dresden, Dresden, Germany
| | - Cahit Birdir
- Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Medizinische Fakultät, TU Dresden, Dresden, Germany
- Zentrum für Feto/Neonatale Gesundheit, Medizinische Fakultät, TU Dresden, Dresden, Germany
| | - Lars Mense
- Klinik und Poliklinik für Kinder- und Jugendmedizin, Fachbereich Neonatologie & Pädiatrische Intensivmedizin, Medizinische Fakultät, TU Dresden, Dresden, Germany
- Zentrum für Feto/Neonatale Gesundheit, Medizinische Fakultät, TU Dresden, Dresden, Germany
| |
Collapse
|
39
|
Maalouf FI, Saad T, Zakhour R, Yunis K. Successful establishment and five-year sustainability of a neonatal-specific antimicrobial stewardship program in a low middle-income country. Front Pharmacol 2023; 13:1076392. [PMID: 36686698 PMCID: PMC9848584 DOI: 10.3389/fphar.2022.1076392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/15/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction: Antibiotic use varies substantially among neonatal intensive care units (NICUs) without any appreciable impact on outcomes. An increased use of antimicrobials has been reported in low-middle income countries. This raises the concern for potential overuse of antibiotics in a fragile patient population, thus increasing the rates of multidrug resistant organisms and affecting the developing microbiome. The presence of a neonatal-specific antimicrobial stewardship program can aid with the judicious use of antibiotics in the neonatal population and thus decrease the overuse of such medications. Methods: In this quality improvement project, we established and implemented a neonatal-specific antimicrobial stewardship program with the aim of reducing antimicrobial use in the neonatal intensive care units within a year of starting. Several interventions using a multidisciplinary approach included implementing standard algorithms, direct audit and feedback, and automated hard stops. Results: These series of interventions led to a 35% decrease in antimicrobial usage in the first 3 months and further decrease was seen with a median of 63% decline for a total of 5 years after project implementation. The use of the most commonly prescribed antibiotics, ampicillin and gentamicin, decreased by 63% and 79%, respectively. There was no evidence that this change in practice affected or jeopardized patient outcomes. Additionally, it showed sustainability and resilience despite the many challenges such as COVID-19 pandemic, political and financial unrest, and healthcare sector collapse. Discussion: This model-based and multidisciplinary low-cost approach can lead to marked improvement affecting neonatal outcomes and can be replicated in other similar centers.
Collapse
Affiliation(s)
- Faouzi I. Maalouf
- Division of Neonatology, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Therese Saad
- Department of Pharmacy, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ramia Zakhour
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Khalid Yunis
- Division of Neonatology, Department of Pediatrics and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
40
|
Salamat-Miller N, Turner MA, Bandekar A, Dixit N, Jochim E, Mangum B, McPherson C, Tenjarla S, Singh S, Hwang YS, Barton N. Assessment of compatibility of rhIGF-1/rhIGFBP-3 with neonatal intravenous medications. World J Pediatr 2023; 19:58-67. [PMID: 36344872 PMCID: PMC9832074 DOI: 10.1007/s12519-022-00610-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/21/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Recombinant human (rh)IGF-1/IGFBP-3 protein complex, administered as a continuous intravenous infusion in preterm infants, is being studied for the prevention of complications of prematurity. METHODS We conducted in vitro studies to evaluate the physical and chemical compatibility of rhIGF-1/IGFBP-3 with medications routinely administered to preterm neonates. In vitro mixing of rhIGF-1/IGFBP-3 drug product with small-molecule test medications plus corresponding controls was performed. Physical compatibility was defined as no color change, precipitation, turbidity, gas evolution, no clinically relevant change in pH/osmolality or loss in medication content. Chemical compatibility of small molecules was assessed using liquid chromatography (e.g., reverse-phase HPLC and ion chromatography), with incompatibility defined as loss of concentration of ≥ 10%. A risk evaluation was conducted for each medication based on in vitro compatibility data and potential for chemical modification. RESULTS In vitro physical compatibility was established for 11/19 medications: caffeine citrate, fentanyl, fluconazole, gentamicin, insulin, intravenous fat emulsion, midazolam, morphine sulfate, custom-mixed parenteral nutrition solution (with/without electrolytes), parenteral nutrition solution + intravenous fat emulsion, and vancomycin (dosed from a 5 mg/mL solution), but not for 8/19 medications: amikacin, ampicillin, dopamine, dobutamine, furosemide, meropenem, norepinephrine, and penicillin G, largely owing to changes in pH after mixing. Small-molecule compatibility was unaffected post-mixing, with no loss of small-molecule content. For physically compatible medications, risk analyses confirmed low probability and severity of a risk event. CONCLUSION Co-administration of rhIGF-1/rhIGFBP-3 drug product with various medications was assessed by in vitro studies using case-by-case risk analyses to determine the suitability of the products for co-administration.
Collapse
Affiliation(s)
| | - Mark A. Turner
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | - Nitin Dixit
- Takeda, 200 Shire Way, Lexington, MA 02421 USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Feng K, He Y, Liu W, Zhang X, Song P, Hua Z. Evaluation of antibiotic stewardship among near-term and term infants admitted to a neonatal unit. Eur J Pediatr 2023; 182:245-254. [PMID: 36289096 DOI: 10.1007/s00431-022-04668-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/30/2022] [Accepted: 10/15/2022] [Indexed: 01/12/2023]
Abstract
UNLABELLED To evaluate the safety and effectiveness of evidence-based antibiotic stewardship in a neonatal unit in China. The study period consisted of two phases, one retrospective (the baseline period, January to December 2018, and the transition period, January 2019 to August 2020) and one prospective intervention period (September 2020 to August 2021). During the prospective period, evidence-based antibiotic stewardship was applied to neonates with suspected infections, pneumonia, and culture-negative sepsis. The antibiotic stewardship included the observation form of neonatal infections, antibiotic therapy of no more than 48 h for suspected infections, and 5 days for pneumonia and culture-negative sepsis. The change in antibiotic use measured by days of therapy per 1000 patient-days between the baseline and intervention period was analyzed. Safety outcomes included reinitiation of antibiotics within 14 days, length of stay, occurrence of late-onset sepsis and necrotizing enterocolitis (Bell stage ≥ II), multidrug-resistant organism infections, and mortality. A total of 7705 neonates were enrolled during the baseline (n = 4804) and the intervention periods (n = 2901). The total antibiotic usage during the baseline period was 771 days of therapy per 1000 patient-days, while that was 525 days of therapy per 1000 patient-days during the intervention period, indicating a 32% decrease in antibiotic consumption. No significant difference in safety outcomes was observed between the baseline and intervention period (P > 0.05), whereas the length of stay was longer during the intervention period (P < 0.001). CONCLUSION The evidence-based antibiotic stewardship can safely and effectively reduce antibiotic use and shorten the duration of therapy in the neonatal unit. WHAT IS KNOWN • Overuse of antibiotics has been associated with adverse events in neonates, including necrotizing enterocolitis, multidrug-resistant organism infections, and death. • More clinical effectiveness evidence is needed to support antibiotic stewardship of neonates in China. WHAT IS NEW • Using prospective audit, targeted stewardship interventions, this study shows that a 32% reduction in overall antibiotic consumption was achieved safely. • Implementation of evidence-based neonatal antibiotic stewardship, including the observation form of neonatal infections, antibiotic therapy of no more than 48 h for suspected infections, and 5 days for pneumonia and culture-negative sepsis, is safe and effective among newborns in a developing country.
Collapse
Affiliation(s)
- Kun Feng
- Department of Neonatology, Children's Hospital of Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Er Rd, Yuzhong District, Chongqing, 400014, China
| | - Yunyan He
- Department of Neonatology, Children's Hospital of Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Er Rd, Yuzhong District, Chongqing, 400014, China
| | - Weiqin Liu
- Department of Neonatology, Children's Hospital of Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Er Rd, Yuzhong District, Chongqing, 400014, China
| | - Xinyin Zhang
- Department of Neonatology, Children's Hospital of Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Er Rd, Yuzhong District, Chongqing, 400014, China
| | - Ping Song
- Children's Medical Big Data Intelligent Application, Chongqing University Engineering Research Center, Chongqing, 400014, China
| | - Ziyu Hua
- Department of Neonatology, Children's Hospital of Chongqing Medical University/Ministry of Education Key Laboratory of Child Development and Disorders/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Er Rd, Yuzhong District, Chongqing, 400014, China.
| |
Collapse
|
42
|
Dai HR, Guo HL, Hu YH, Xu J, Ding XS, Cheng R, Chen F. Precision caffeine therapy for apnea of prematurity and circadian rhythms: New possibilities open up. Front Pharmacol 2022; 13:1053210. [PMID: 36532766 PMCID: PMC9753576 DOI: 10.3389/fphar.2022.1053210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/18/2022] [Indexed: 09/10/2024] Open
Abstract
Caffeine is the globally consumed psychoactive substance and the drug of choice for the treatment of apnea of prematurity (AOP), but its therapeutic effects are highly variable among preterm infants. Many of the molecular underpinnings of the marked individual response have remained elusive yet. Interestingly, the significant association between Clock gene polymorphisms and the response to caffeine therapy offers an opportunity to advance our understanding of potential mechanistic pathways. In this review, we delineate the functions and mechanisms of human circadian rhythms. An up-to-date advance of the formation and ontogeny of human circadian rhythms during the perinatal period are concisely discussed. Specially, we summarize and discuss the characteristics of circadian rhythms in preterm infants. Second, we discuss the role of caffeine consumption on the circadian rhythms in animal models and human, especially in neonates and preterm infants. Finally, we postulate how circadian-based therapeutic initiatives could open new possibilities to promote precision caffeine therapy for the AOP management in preterm infants.
Collapse
Affiliation(s)
- Hao-Ran Dai
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hong-Li Guo
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Ya-Hui Hu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Xu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan-Sheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rui Cheng
- Neonatal Intensive Care Unit, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
43
|
A whole blood microsampling furosemide assay: development, validation and use in a pediatric pharmacokinetic study. Bioanalysis 2022; 14:1025-1038. [PMID: 36165919 PMCID: PMC9540403 DOI: 10.4155/bio-2022-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Furosemide is a commonly used diuretic for the treatment of edema. The pharmacokinetics of furosemide in neonates as they mature remains poorly understood. Microsampling assays facilitate research in pediatric populations. Results: We developed and validated a liquid chromatography-tandem mass spectrometry method for the quantitation of furosemide in human whole blood with volumetric absorptive microsampling (VAMS) devices (10 μl). Furosemide was stable in human whole blood VAMS under the study's assay conditions. This work established stability for furosemide for 161 days when stored as dried microsamples at -78°C. Conclusion: This method is being applied for the quantitation of furosemide in whole blood VAMS in an ongoing prospective pediatric clinical study. Representative clinical data are reported.
Collapse
|
44
|
Guitor AK, Yousuf EI, Raphenya AR, Hutton EK, Morrison KM, McArthur AG, Wright GD, Stearns JC. Capturing the antibiotic resistome of preterm infants reveals new benefits of probiotic supplementation. MICROBIOME 2022; 10:136. [PMID: 36008821 PMCID: PMC9414150 DOI: 10.1186/s40168-022-01327-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/14/2022] [Indexed: 05/28/2023]
Abstract
BACKGROUND Probiotic use in preterm infants can mitigate the impact of antibiotic exposure and reduce rates of certain illnesses; however, the benefit on the gut resistome, the collection of antibiotic resistance genes, requires further investigation. We hypothesized that probiotic supplementation of early preterm infants (born < 32-week gestation) while in hospital reduces the prevalence of antibiotic resistance genes associated with pathogenic bacteria in the gut. We used a targeted capture approach to compare the resistome from stool samples collected at the term corrected age of 40 weeks for two groups of preterm infants (those that routinely received a multi-strain probiotic during hospitalization and those that did not) with samples from full-term infants at 10 days of age to identify if preterm birth or probiotic supplementation impacted the resistome. We also compared the two groups of preterm infants up to 5 months of age to identify persistent antibiotic resistance genes. RESULTS At the term corrected age, or 10 days of age for the full-term infants, we found over 80 antibiotic resistance genes in the preterm infants that did not receive probiotics that were not identified in either the full-term or probiotic-supplemented preterm infants. More genes associated with antibiotic inactivation mechanisms were identified in preterm infants unexposed to probiotics at this collection time-point compared to the other infants. We further linked these genes to mobile genetic elements and Enterobacteriaceae, which were also abundant in their gut microbiomes. Various genes associated with aminoglycoside and beta-lactam resistance, commonly found in pathogenic bacteria, were retained for up to 5 months in the preterm infants that did not receive probiotics. CONCLUSIONS This pilot survey of preterm infants shows that probiotics administered after preterm birth during hospitalization reduced the diversity and prevented persistence of antibiotic resistance genes in the gut microbiome. The benefits of probiotic use on the microbiome and the resistome should be further explored in larger groups of infants. Due to its high sensitivity and lower sequencing cost, our targeted capture approach can facilitate these surveys to further address the implications of resistance genes persisting into infancy without the need for large-scale metagenomic sequencing. Video Abstract.
Collapse
Affiliation(s)
- Allison K Guitor
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Canada
| | - Efrah I Yousuf
- Department of Pediatrics, McMaster University, Hamilton, Canada
| | - Amogelang R Raphenya
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Canada
| | - Eileen K Hutton
- Department of Obstetrics & Gynecology, McMaster University, Hamilton, Canada
- The Baby & Mi and the Baby & Pre-Mi Cohort Studies, Hamilton, Canada
| | - Katherine M Morrison
- Department of Pediatrics, McMaster University, Hamilton, Canada
- The Baby & Mi and the Baby & Pre-Mi Cohort Studies, Hamilton, Canada
| | - Andrew G McArthur
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Canada
| | - Gerard D Wright
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Canada
| | - Jennifer C Stearns
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada.
- The Baby & Mi and the Baby & Pre-Mi Cohort Studies, Hamilton, Canada.
- Department of Medicine, McMaster University, Hamilton, Canada.
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada.
| |
Collapse
|
45
|
Armann J, Rüdiger M, Berner R, Mense L. Restrictive prescription of antibiotics in preterm infants with premature rupture of membranes. BMC Pediatr 2022; 22:408. [PMID: 35820876 PMCID: PMC9275236 DOI: 10.1186/s12887-022-03476-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In preterm infants with premature rupture of membranes, antibiotic treatment is frequently started but rates of early onset sepsis are lower. In line with national guidelines, a stratified approach in the decision to start antibiotic treatment using maternal history, clinical impression and biomarkers has been implemented in our level III neonatal center and its results are evaluated. METHODS Retrospective cohort study of all preterm newborns with rupture of membranes at least 1 h prior to delivery admitted to our tertiary neonatal intensive care unit. Data on antibiotic exposure, mortality and major neonatal complications were extracted from the electronic patient charts to evaluate the effects and safety of our stratified approach. RESULTS Four hundred fifty-six infants met the inclusion criteria. 120 (26%) received primary antibiotics whereas 336 (74%) did not. Of those receiving primary antibiotics, 13 (11%) had a blood culture positive sepsis, 46 (38%) met the criteria of clinical sepsis and in 61 (51%) sepsis was ruled out and antibiotics were stopped after 48-96 h. All infants with blood culture positive sepsis were identified and treated within the first 24 h of life using this approach. None of the 336 infants who were not started on antibiotics primarily needed antibiotic therapy within the first 5 days of life. There were no deaths or major neonatal complications in the group that did not receive empiric antibiotics. CONCLUSIONS Our stratified approach for preterm infants with premature rupture of membranes allows a safe reduction of antibiotic exposure even in this high risk population. As a result, only 25% of high risk preterm newborns are treated with antibiotics of which more than half receive less than 5 days of treatment. To treat one infant with blood culture positive sepsis, only 9 infants receive empiric antibiotics.
Collapse
Affiliation(s)
- Jakob Armann
- Department of Pediatrics, University Hospital Carl Gustav Carus Dresden, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Mario Rüdiger
- Department of Pediatrics, University Hospital Carl Gustav Carus Dresden, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
- Department of Pediatrics, Division of Neonatology & Pediatric Intensive Care Medicine, University Hospital Carl Gustav Carus Dresden, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Reinhard Berner
- Department of Pediatrics, University Hospital Carl Gustav Carus Dresden, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Lars Mense
- Department of Pediatrics, University Hospital Carl Gustav Carus Dresden, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
- Department of Pediatrics, Division of Neonatology & Pediatric Intensive Care Medicine, University Hospital Carl Gustav Carus Dresden, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| |
Collapse
|
46
|
Kabir M, Padilha EC, Shah P, Huang R, Sakamuru S, Gonzalez E, Ye L, Hu X, Henderson MJ, Xia M, Xu X. Identification of Selective CYP3A7 and CYP3A4 Substrates and Inhibitors Using a High-Throughput Screening Platform. Front Pharmacol 2022; 13:899536. [PMID: 35847040 PMCID: PMC9283723 DOI: 10.3389/fphar.2022.899536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/27/2022] [Indexed: 11/26/2022] Open
Abstract
Cytochrome P450 (CYP) 3A7 is one of the major xenobiotic metabolizing enzymes in human embryonic, fetal, and newborn liver. CYP3A7 expression has also been observed in a subset of the adult population, including pregnant women, as well as in various cancer patients. The characterization of CYP3A7 is not as extensive as other CYPs, and health authorities have yet to provide guidance towards DDI assessment. To identify potential CYP3A7-specific molecules, we used a P450-Glo CYP3A7 enzyme assay to screen a library of ∼5,000 compounds, including FDA-approved drugs and drug-like molecules, and compared these screening data with that from a P450-Glo CYP3A4 assay. Additionally, a subset of 1,000 randomly selected compounds were tested in a metabolic stability assay. By combining the data from the qHTS P450-Glo and metabolic stability assays, we identified several chemical features important for CYP3A7 selectivity. Halometasone was chosen for further evaluation as a potential CYP3A7-selective inhibitor using molecular docking. From the metabolic stability assay, we identified twenty-two CYP3A7-selective substrates over CYP3A4 in supersome setting. Our data shows that CYP3A7 has ligand promiscuity, much like CYP3A4. Furthermore, we have established a large, high-quality dataset that can be used in predictive modeling for future drug metabolism and interaction studies.
Collapse
Affiliation(s)
- Md Kabir
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
- Department of Pharmacology, The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Elias C. Padilha
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Pranav Shah
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Ruili Huang
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Srilatha Sakamuru
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Eric Gonzalez
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
- Novartis Institutes for BioMedical Research, Cambridge, MA, United States
| | - Lin Ye
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Xin Hu
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Mark J. Henderson
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Menghang Xia
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
- *Correspondence: Menghang Xia, ; Xin Xu,
| | - Xin Xu
- Division of Pre-Clinical Innovation, National Center for Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
- *Correspondence: Menghang Xia, ; Xin Xu,
| |
Collapse
|
47
|
Le J, Greenberg RG, Yoo Y, Clark RH, Benjamin DK, Zimmerman KO, Cohen-Wolkowiez M, Wade KC. Ampicillin dosing in premature infants for early-onset sepsis: exposure-driven efficacy, safety, and stewardship. J Perinatol 2022; 42:959-964. [PMID: 35210541 PMCID: PMC9262754 DOI: 10.1038/s41372-022-01344-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/06/2022] [Accepted: 02/03/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Define optimal ampicillin dosing for empiric early-onset sepsis (EOS) therapy in preterm neonates. STUDY DESIGN We simulated ampicillin concentrations in newborns (birthweight < 1500 g; gestational age 22-27 weeks), summarizing three 48 h regimens: high 100 mg/kg Q8hr, medium 100 mg/kg Q12hr, and standard 50 mg/kg Q12hr. Concentration data were analyzed for concentration above minimum inhibitory concentration (MIC), below neurotoxicity threshold (Cmax ≤ 140 mcg/mL), and duration limited to 48 h. RESULTS Among 34,689 newborns, all dosing regimens provided concentrations above MIC through 48 h, but Cmax exceeded the neurotoxicity threshold. With the 4-dose standard regimen, >90% maintained concentrations >MIC beyond 48 h. With the 2-dose regimen, newborns maintained the mean concentration >MIC within the 48 h culture window and below neurotoxicity level. Infants 22-24 weeks' gestation had higher drug concentrations and more prolonged exposure duration than 25-27 weeks' gestation. CONCLUSIONS For EOS in preterm infants, two ampicillin doses (50 mg/kg) provided optimal bactericidal exposures, while minimizing potential toxicity.
Collapse
Affiliation(s)
- Jennifer Le
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, CA, USA
| | - Rachel G Greenberg
- Department of Pediatrics, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - YoungJun Yoo
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, CA, USA
| | | | - Daniel K Benjamin
- Department of Pediatrics, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Kanecia O Zimmerman
- Department of Pediatrics, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Michael Cohen-Wolkowiez
- Department of Pediatrics, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Kelly C Wade
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Graus JM, Herbozo C, Hernandez R, Pantoja AF, Zegarra J. Managing antibiotics wisely in a neonatal intensive care unit in a low resource setting. J Perinatol 2022; 42:965-970. [PMID: 35459905 PMCID: PMC9023725 DOI: 10.1038/s41372-022-01388-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 03/17/2022] [Accepted: 03/31/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND AND OBJECTIVES Unnecessary early antibiotic exposure is deleterious, it may induce the selection of multi-drug-resistant organisms. The objective of this project was to decrease antibiotic exposure of newborns admitted to the neonatal intensive care unit at Hospital Cayetano Heredia, a level 3 unit in Lima, Peru. METHODS Quality improvement project in which we implemented an antibiotic stewardship program for early onset sepsis in the neonatal intensive care unit. Primary outcome measure was antibiotic usage rate, total number of days infants were exposed to antibacterial agents divided by 1000 patient-days. RESULTS Antibiotic usage rate declined from 291/1000 patient-days to 82/1000 patient-days during the last months of 2020, representing a total decrease of 65.1%. CONCLUSIONS Antibiotic stewardship for early-onset sepsis implemented in a perinatal center like ours is effective, appears to be safe and results in a sustained and significant decrease in the use of antibiotics for early-onset sepsis.
Collapse
Affiliation(s)
- Juan M Graus
- Universidad Peruana Cayetano Heredia, School of Medicine, Lima, Peru
- Hospital Cayetano Heredia, Lima, Peru
| | - Cecilia Herbozo
- Universidad Peruana Cayetano Heredia, School of Medicine, Lima, Peru.
- Hospital Cayetano Heredia, Lima, Peru.
| | - Roger Hernandez
- Universidad Peruana Cayetano Heredia, School of Medicine, Lima, Peru
- Hospital Cayetano Heredia, Lima, Peru
| | - Alfonso Francisco Pantoja
- Universidad Peruana Cayetano Heredia, School of Medicine, Lima, Peru
- St Joseph Hospital, Denver, CO, United States
| | - Jaime Zegarra
- Universidad Peruana Cayetano Heredia, School of Medicine, Lima, Peru
- Hospital Cayetano Heredia, Lima, Peru
| |
Collapse
|
49
|
Gilfillan M, Bhandari V. Moving bronchopulmonary dysplasia research from the bedside to the bench. Am J Physiol Lung Cell Mol Physiol 2022; 322:L804-L821. [PMID: 35437999 DOI: 10.1152/ajplung.00452.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/22/2022] Open
Abstract
Although advances in the respiratory management of extremely preterm infants have led to improvements in survival, this progress has not yet extended to a reduction in the incidence of bronchopulmonary dysplasia (BPD). BPD is a complex multifactorial condition that primarily occurs due to disturbances in the regulation of normal pulmonary airspace and vascular development. Preterm birth and exposure to invasive mechanical ventilation also compromises large airway development, leading to significant morbidity and mortality. Although both predisposing and protective genetic and environmental factors have been frequently described in the clinical literature, these findings have had limited impact on the development of effective therapeutic strategies. This gap is likely because the molecular pathways that underlie these observations are yet not fully understood, limiting the ability of researchers to identify novel treatments that can preserve normal lung development and/or enhance cellular repair mechanisms. In this review article, we will outline various well-established clinical observations while identifying key knowledge gaps that need to be filled with carefully designed preclinical experiments. We will address these issues by discussing controversial topics in the pathophysiology, the pathology, and the treatment of BPD, including an evaluation of existing animal models that have been used to answer important questions.
Collapse
Affiliation(s)
- Margaret Gilfillan
- Division of Neonatology, St. Christopher's Hospital for Children/Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Vineet Bhandari
- Division of Neonatology, The Children's Regional Hospital at Cooper/Cooper Medical School of Rowan University, Camden, New Jersey
| |
Collapse
|
50
|
Chu X, Prasad B, Neuhoff S, Yoshida K, Leeder JS, Mukherjee D, Taskar K, Varma MVS, Zhang X, Yang X, Galetin A. Clinical Implications of Altered Drug Transporter Abundance/Function and PBPK Modeling in Specific Populations: An ITC Perspective. Clin Pharmacol Ther 2022; 112:501-526. [PMID: 35561140 DOI: 10.1002/cpt.2643] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022]
Abstract
The role of membrane transporters on pharmacokinetics (PKs), drug-drug interactions (DDIs), pharmacodynamics (PDs), and toxicity of drugs has been broadly recognized. However, our knowledge of modulation of transporter expression and/or function in the diseased patient population or specific populations, such as pediatrics or pregnancy, is still emerging. This white paper highlights recent advances in studying the changes in transporter expression and activity in various diseases (i.e., renal and hepatic impairment and cancer) and some specific populations (i.e., pediatrics and pregnancy) with the focus on clinical implications. Proposed alterations in transporter abundance and/or activity in diseased and specific populations are based on (i) quantitative transporter proteomic data and relative abundance in specific populations vs. healthy adults, (ii) clinical PKs, and emerging transporter biomarker and/or pharmacogenomic data, and (iii) physiologically-based pharmacokinetic modeling and simulation. The potential for altered PK, PD, and toxicity in these populations needs to be considered for drugs and their active metabolites in which transporter-mediated uptake/efflux is a major contributor to their absorption, distribution, and elimination pathways and/or associated DDI risk. In addition to best practices, this white paper discusses current challenges and knowledge gaps to study and quantitatively predict the effects of modulation in transporter activity in these populations, together with the perspectives from the International Transporter Consortium (ITC) on future directions.
Collapse
Affiliation(s)
- Xiaoyan Chu
- Department of ADME and Discovery Toxicology, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | | | - Kenta Yoshida
- Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, California, USA
| | - James Steven Leeder
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Dwaipayan Mukherjee
- Clinical Pharmacology & Pharmacometrics, Research & Development, AbbVie, Inc., North Chicago, Illinois, USA
| | | | - Manthena V S Varma
- Pharmacokinetics, Dynamics and Metabolism, Medicine Design, Worldwide R&D, Pfizer Inc, Groton, Connecticut, USA
| | - Xinyuan Zhang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Xinning Yang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|