151
|
Greenberg RG, Chowdhury D, Hansen NI, Smith PB, Stoll BJ, Sánchez PJ, Das A, Puopolo KM, Mukhopadhyay S, Higgins RD, Cotten CM. Prolonged duration of early antibiotic therapy in extremely premature infants. Pediatr Res 2019; 85:994-1000. [PMID: 30737489 PMCID: PMC6531328 DOI: 10.1038/s41390-019-0300-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Prolonged early antibiotics in extremely premature infants may have negative effects. We aimed to assess prevalence and outcomes of provision of prolonged early antibiotics to extremely premature infants in the absence of culture-confirmed infection or NEC. METHODS Cohort study of infants from 13 centers born without a major birth defect from 2008-2014 who were 401-1000 grams birth weight, 22-28 weeks gestation, and survived ≥5 days without culture-confirmed infection, NEC, or spontaneous intestinal perforation. We determined the proportion of infants who received prolonged early antibiotics, defined as ≥5 days of antibiotic therapy started at ≤72 h of age, by center and over time. Associations between prolonged early antibiotics and adverse outcomes were assessed using multivariable logistic regression. RESULTS A total of 5730 infants were included. The proportion of infants receiving prolonged early antibiotics varied from 30-69% among centers and declined from 49% in 2008 to 35% in 2014. Prolonged early antibiotics was not significantly associated with death (adjusted odds ratio 1.17 [95% CI: 0.99-1.40], p = 0.07) and was not associated with NEC. CONCLUSIONS The proportion of extremely premature infants receiving prolonged early antibiotics decreased, but significant center variation persists. Prolonged early antibiotics were not significantly associated with increased odds of death or NEC.
Collapse
Affiliation(s)
- Rachel G Greenberg
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA.
| | - Dhuly Chowdhury
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC, Durham, USA
| | - Nellie I Hansen
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC, Durham, USA
| | - P Brian Smith
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Barbara J Stoll
- Department of Pediatrics, University of Texas Medical School at Houston, Houston, TX, USA
| | - Pablo J Sánchez
- Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA
| | - Abhik Das
- Social, Statistical and Environmental Sciences Unit, RTI International, Rockville, MD, USA
| | - Karen M Puopolo
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sagori Mukhopadhyay
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rosemary D Higgins
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - C Michael Cotten
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
152
|
O'Mara K, Gattoline SJ, Campbell CT. Female low dose tip syringes-increased complexity of use may compromise dosing accuracy in paediatric patients. J Clin Pharm Ther 2019; 44:463-470. [PMID: 30763471 DOI: 10.1111/jcpt.12810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/28/2018] [Accepted: 01/10/2019] [Indexed: 11/29/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE The International Organization for Standardization (ISO) created enteral device specifications to reduce tubing misconnections. The Global Enteral Device Supplier Association (GEDSA) supports a female design: standard and low dose tip (LDT). Concerns include increased complexity of use with adapters, dosing accuracy and workflow. No peer-reviewed studies have evaluated dosing accuracy of the complete female system with adapters. The objective of this study was to compare dosing accuracy of the female design to legacy syringes. METHODS An in vitro study was conducted at the University of Florida College of Pharmacy pharmaceutics laboratory. Assessments were completed for syringe size, dispense methods and volumes, and adapters when applicable. A gravimetric scale and specific gravity were used to calculate administration volumes. The primary outcome was frequency administration volume exceeded 10% expected amount. RESULTS AND DISCUSSION A total of 576 tests were performed. The LDT resulted in significantly higher rates of unacceptable dosing variance compared to legacy (21.2% vs 7.4%, P = 0.003). Variance exceeding 10% occurred more frequently with LDT 0.5 and 1 mL syringes, medication cup dispensing (liquid or tablet) and inappropriate LDT adapter use. Unapproved adapter processes compared to FDA-approved processes held a higher likelihood of unacceptable dosing variance (28% vs 7.4%, P < 0.001). FDA-approved use of adapters with prefilled syringes compared to bedside administration may result in higher rates of dosing inaccuracy (18.8% vs 5.6%, P = 0.06). WHAT IS NEW AND CONCLUSIONS This study raises clinical concerns of dosing inaccuracies with the LDT syringes, particularly with 0.5 and 1 mL sizes. The use of adapters significantly increases the opportunity for inaccurate dosing.
Collapse
Affiliation(s)
- Keliana O'Mara
- Department of Pharmacy, UF Health Shands Children's Hospital, Newberry, Florida
| | | | - Christopher T Campbell
- Department of Pharmacy, UF Health Shands Children's Hospital, Newberry, Florida
- University of Florida College of Pharmacy, Gainesville, Florida
| |
Collapse
|
153
|
Abstract
Antimicrobial medications are the most commonly used medications in the neonatal intensive care unit. Antibiotics are used for infection prophylaxis, empiric treatment, and definitive treatment of confirmed infection. The choice of medication should be informed by the epidemiology and microbiology of infection in specific clinical scenarios and by the clinical condition of the infant. Understanding evolving pathogen susceptibility to antimicrobials and key pharmacotherapy determinants in neonates can inform optimal antibiotic use.
Collapse
Affiliation(s)
- Sagori Mukhopadhyay
- Section on Newborn Medicine, Pennsylvania Hospital, Philadelphia, PA, USA; Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; CHOP Newborn Care, Pennsylvania Hospital, 800 Spruce Street, Philadelphia, PA 19107, USA
| | - Kelly C Wade
- Section on Newborn Medicine, Pennsylvania Hospital, Philadelphia, PA, USA; Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; CHOP Newborn Care, Pennsylvania Hospital, 800 Spruce Street, Philadelphia, PA 19107, USA
| | - Karen M Puopolo
- Section on Newborn Medicine, Pennsylvania Hospital, Philadelphia, PA, USA; Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; CHOP Newborn Care, Pennsylvania Hospital, 800 Spruce Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
154
|
Allegaert K, van den Anker J. Neonates are not just little children and need more finesse in dosing of antibiotics. Acta Clin Belg 2019; 74:157-163. [PMID: 29745792 DOI: 10.1080/17843286.2018.1473094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
OBJECTIVES Neonates are not just little children. They need more finesse in decisions on when to treat, which antibiotics to use and how to dose these antibiotics. METHODS Representative compounds of three major classes of antibiotics (beta-lactams, aminoglycosides, glycopeptides) are discussed in a narrative review to illustrate the recent progress in the knowledge on PK and its covariates (how to dose). RESULTS This knowledge can subsequently be converted to targeted exposure dosing regimens. This is because it is reasonable to postulate that pharmacodynamics (PD) of antibiotics are similar in neonates to that in other populations if a similar concentration-time profile and targeted exposure are attained. However, this approach has its limitations, since the clinical response may be different in neonates because of maturational differences in innate immunity or toxicity. These dosing regimens should at least be validated. CONCLUSION Relevant information on the PK of antibiotics and its covariates have been generated, but the next steps are to validate the dosing regimens suggested, and consider more sophisticated dosing regimens. This approach should subsequently pave the way to conduct comparative studies to assess the efficacy and safety of these commonly used drugs in neonates.
Collapse
Affiliation(s)
- Karel Allegaert
- Intensive Care and Department of Pediatric Surgery, Erasmus MC – Sophia Children’s Hospital, Rotterdam, The Netherlands
- Department of Neonatology, Erasmus MC – Sophia Children’s Hospital, Rotterdam, The Netherlands
- Department of Development and Regeneration, KU, Leuven, Belgium
| | - John van den Anker
- Intensive Care and Department of Pediatric Surgery, Erasmus MC – Sophia Children’s Hospital, Rotterdam, The Netherlands
- Paediatric Pharmacology and Pharmacometrics, University of Basel Children’s Hospital, Basel, Switzerland
- Division of Clinical Pharmacology, Children’s National Health System, Washington, DC, USA
| |
Collapse
|
155
|
Gerhart JG, Watt KM, Edginton A, Wade KC, Salerno SN, Benjamin DK, Smith PB, Hornik CP, Cohen-Wolkowiez M, Duara S, Ross A, Shattuck K, Stewart DL, Neu N, Gonzalez D. Physiologically-Based Pharmacokinetic Modeling of Fluconazole Using Plasma and Cerebrospinal Fluid Samples From Preterm and Term Infants. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:500-510. [PMID: 31087536 PMCID: PMC6656941 DOI: 10.1002/psp4.12414] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 12/16/2022]
Abstract
Fluconazole is used to treat hematogenous Candida meningoencephalitis in preterm and term infants. To characterize plasma and central nervous system exposure, an adult fluconazole physiologically‐based pharmacokinetic (PBPK) model was scaled to infants, accounting for age dependencies in glomerular filtration and metabolism. The model was optimized using 760 plasma samples from 166 infants (median postmenstrual age (range) 28 weeks (24–50)) and 27 cerebrospinal fluid (CSF) samples from 22 infants (postmenstrual age 28 weeks (24–33)). Simulations evaluated achievement of the surrogate efficacy target of area under the unbound concentration‐time curve ≥ 400 mg • hour/L over the dosing interval in plasma and CSF using dosing guidelines. Average fold error of predicted concentrations was 0.73 and 1.14 for plasma and CSF, respectively. Target attainment in plasma and CSF was reached faster after incorporating a loading dose of 25 mg/kg. PBPK modeling can be useful in exploring CNS kinetics of drugs in children.
Collapse
Affiliation(s)
- Jacqueline G Gerhart
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kevin M Watt
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Andrea Edginton
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| | - Kelly C Wade
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sara N Salerno
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Daniel K Benjamin
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - P Brian Smith
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Christoph P Hornik
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Michael Cohen-Wolkowiez
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Shahnaz Duara
- Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Ashley Ross
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Karen Shattuck
- Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Dan L Stewart
- Department of Pediatrics, University of Louisville, Louisville, Kentucky, USA
| | - Natalie Neu
- Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
156
|
Frymoyer A, Stockmann C, Hersh AL, Goswami S, Keizer RJ. Individualized Empiric Vancomycin Dosing in Neonates Using a Model-Based Approach. J Pediatric Infect Dis Soc 2019; 8:97-104. [PMID: 29294072 DOI: 10.1093/jpids/pix109] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 12/11/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND Vancomycin dosing in neonates is challenging because of the large variation in pharmacokinetics. Existing empiric dosing recommendations use table-based formats, within which a neonate is categorized on the basis of underlying characteristics. The ability to individualize dosing is limited because of the small number of "dose categories," and achieving narrow exposure targets is difficult. Our objective was to evaluate a model-based dosing approach (which we designated Neo-Vanco) designed to individualize empiric vancomycin dosing in neonates. METHODS Neo-Vanco was developed on the basis of a published, externally validated population pharmacokinetic model. Using a simulation-based methodology, individualized empiric doses that maximize the probability of attaining a 24-hour area under the curve/minimum inhibitory concentration ratio (AUC24/MIC) of >400 while minimizing troughs >20 mg/L are calculated. To evaluate the Neo-Vanco strategy, retrospective data from neonates treated with vancomycin at 2 healthcare systems were used, and empiric dose recommendations from the following 4 sources were examined: Neo-Vanco, Neofax, Red Book, and Lexicomp. Predicted AUC24 and troughs were calculated and compared. RESULTS Overall, 492 neonates were evaluated (median postmenstrual age, 36 weeks [5th-95th percentiles (90% range), 25-47 weeks]; median weight, 2.4 kg [90% range, 0.6-4.8 kg]). The percentage of neonates predicted to achieve an AUC24/MIC of >400 was 94% with Neo-Vanco, 18% with Neofax, 23% with Red Book, and 55% with Lexicomp (all P < .0001 vs Neo-Vanco). Predicted troughs of >20 mg/L were infrequent and similar across the dosing approaches (Neo-Vanco, 2.8%; Neofax, 1.0% [P = .03]; Red Book, 2.6% [P = .99]; and Lexicomp, 4.1% [P = .27]. CONCLUSION A model-based dosing approach that individualizes empiric vancomycin dosing was predicted to improve achievement of target exposure levels in neonates. Prospective clinical evaluation is warranted.
Collapse
Affiliation(s)
- Adam Frymoyer
- Department of Pediatrics, Stanford University, Palo Alto, California
| | - Chris Stockmann
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City
| | - Adam L Hersh
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City
| | | | | |
Collapse
|
157
|
Hornik CP, Atz AM, Bendel C, Chan F, Downes K, Grundmeier R, Fogel B, Gipson D, Laughon M, Miller M, Smith M, Livingston C, Kluchar C, Heath A, Jarrett C, McKerlie B, Patel H, Hunter C. Creation of a Multicenter Pediatric Inpatient Data Repository Derived from Electronic Health Records. Appl Clin Inform 2019; 10:307-315. [PMID: 31067576 DOI: 10.1055/s-0039-1688477] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Integration of electronic health records (EHRs) data across sites and access to that data remain limited. OBJECTIVE We developed an EHR-based pediatric inpatient repository using nine U.S. centers from the National Institute of Child Health and Human Development Pediatric Trials Network. METHODS A data model encompassing 147 mandatory and 99 optional elements was developed to provide an EHR data extract of all inpatient encounters from patients <17 years of age discharged between January 6, 2013 and June 30, 2017. Sites received instructions on extractions, transformation, testing, and transmission to the coordinating center. RESULTS We generated 177 staging reports to process all nine sites' 147 mandatory and 99 optional data elements to the repository. Based on 520 prespecified criteria, all sites achieved 0% errors and <2% warnings. The repository includes 386,159 inpatient encounters from 264,709 children to support study design and conduct of future trials in children. CONCLUSION Our EHR-based data repository of pediatric inpatient encounters utilized a customized data model heavily influenced by the PCORnet format, site-based data mapping, a comprehensive set of data testing rules, and an iterative process of data submission. The common data model, site-based extraction, and technical expertise were key to our success. Data from this repository will be used in support of Pediatric Trials Network studies and the labeling of drugs and devices for children.
Collapse
Affiliation(s)
- Christoph P Hornik
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Andrew M Atz
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Catherine Bendel
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| | - Francis Chan
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, California, United States
| | - Kevin Downes
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Robert Grundmeier
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Ben Fogel
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Debbie Gipson
- Department of Pediatrics and Communicable Disease, University of Michigan, Ann Arbor, Michigan, United States
| | - Matthew Laughon
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Michael Miller
- Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, United States
| | - Michael Smith
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, Kentucky, United States.,Division of Pediatric Infectious Diseases, Duke University School of Medicine, Durham North Carolina, United States
| | - Chad Livingston
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Cindy Kluchar
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Anne Heath
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Chanda Jarrett
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Brian McKerlie
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Hetalkumar Patel
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | - Christina Hunter
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, United States
| | | |
Collapse
|
158
|
Mukhopadhyay S, Sengupta S, Puopolo KM. Challenges and opportunities for antibiotic stewardship among preterm infants. Arch Dis Child Fetal Neonatal Ed 2019; 104:F327-F332. [PMID: 30425110 PMCID: PMC6491257 DOI: 10.1136/archdischild-2018-315412] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/13/2018] [Accepted: 10/18/2018] [Indexed: 12/20/2022]
Abstract
Antibiotic stewardship programmes aim to optimise antimicrobial use to prevent the emergence of resistance species and protect patients from the side effects of unnecessary medication. The high incidence of systemic infection and associated mortality from these infections leads neonatal providers to frequently initiate antibiotic therapy and make empiric antibiotic courses one of the main contributors of antibiotic use in the neonatal units. Yet, premature infants are also at risk for acute life-threatening complications associated with antibiotic use such as necrotising enterocolitis and for long-term morbidities such as asthma. In this review, we discuss specific aspects of antibiotic use in the very low birthweight preterm infants, with a focus on empiric use, that provide opportunities for stewardship practice. We discuss strategies to risk-stratify antibiotic initiation for the risk of early-onset sepsis, optimise empiric therapy duration and antibiotic choice in late-onset sepsis, and standardise decisions for stopping empiric therapy. Lastly, review the evolving role of biomarkers in antibiotic stewardship.
Collapse
Affiliation(s)
- Sagori Mukhopadhyay
- Division of Neonatology, Children’s Hospital of Philadelphia,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shaon Sengupta
- Division of Neonatology, Children’s Hospital of Philadelphia,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Karen M. Puopolo
- Division of Neonatology, Children’s Hospital of Philadelphia,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
159
|
Taylor G, Jackson W, Hornik CP, Koss A, Mantena S, Homsley K, Gattis B, Kudumu-Clavell M, Clark R, Smith PB, Laughon MM. Surfactant Administration in Preterm Infants: Drug Development Opportunities. J Pediatr 2019; 208:163-168. [PMID: 30580975 PMCID: PMC6486873 DOI: 10.1016/j.jpeds.2018.11.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/26/2018] [Accepted: 11/26/2018] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To evaluate how frequently surfactant is used off-label in preterm infants. STUDY DESIGN We conducted a retrospective cohort analysis of prospectively collected administrative data for 2005-2015 from 348 neonatal intensive care units in the US. We quantified off-label administration of poractant alfa, calfactant, or beractant in inborn infants born at <37 weeks of gestational age (GA). Off-label surfactant administration was defined according to the Food and Drug Administration (FDA) label. RESULTS Of a total of 110 822 preterm infants who received surfactant, 68 226 (62%) received the surfactant off-label. The majority of infants who received surfactant off-label had a higher birth weight than those who received surfactant on-label (40 716 [37%]), had an older GA than those who received surfactant on-label (35 191 [32%]), or were treated with intubation and surfactant administration followed by immediate extubation (INSURE) (32 310 [29%]). Poractant alfa was administered via INSURE more frequently than beractant or calfactant (16 688 [38%], 7137 [20%], and 8485 [27%], respectively). An increasing number of infants received surfactant via INSURE from 2005 to 2015 (from 1697 [19%] to 3368 [36%]). CONCLUSIONS The majority of surfactant given to preterm infants is administered off-label. The uptrend in administration via INSURE coincides with increased supporting evidence. The gap between FDA labeling and current clinic practice exemplifies an opportunity for label expansion, which may require additional prospective or retrospective safety and/or effectiveness data for infants of older GA and higher birth weight.
Collapse
Affiliation(s)
- Genevieve Taylor
- Department of Pediatrics, The University of North Carolina, Chapel Hill, NC.
| | - Wesley Jackson
- Department of Pediatrics, The University of North Carolina, Chapel Hill, NC
| | - Christoph P Hornik
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Alec Koss
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC
| | - Sreekar Mantena
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC
| | - Kenya Homsley
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC
| | - Blair Gattis
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC
| | | | - Reese Clark
- Pediatrix-Obstetrix Center for Research and Education, Sunrise, FL
| | - P Brian Smith
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Matthew M Laughon
- Department of Pediatrics, The University of North Carolina, Chapel Hill, NC
| |
Collapse
|
160
|
Chen CY, Essien MD, Johnson AJ, Lee GT, Chou FS. Use of mean platelet volume in the assessment of intrauterine infection in newborns with combined thrombocytopenia and leukopenia at birth. J Matern Fetal Neonatal Med 2019; 34:346-352. [PMID: 30983434 DOI: 10.1080/14767058.2019.1608174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Objective: Intrauterine stress can be reflected on hematological disturbance at birth. Thrombocytopenia and leukopenia may be a result of exposure to maternal hypertensive disorders but may also indicate fetal inflammatory response to intrauterine infection, prompting empiric antibiotics use during the initial assessment after birth. Emerging data suggest long-term adverse health outcomes associated with antibiotics exposure early in life. In this report, we sought to assess the use of mean platelet volume (MPV) at birth in predicting fetal inflammatory response in newborns with combined thrombocytopenia and leukopenia.Materials and methods: This is a retrospective study from a single academic medical center. Data were collected prospectively on all newborns with thrombocytopenia and leukopenia within the first 24 h of life. The primary outcome was a composite of fetal tachycardia, premature preterm rupture of membrane with preterm labor, and histological evidence of chorioamnionitis/funisitis/villitis on placental pathology reports evaluated using a multiple logistical regression analysis.Results: The prevalence of combined thrombocytopenia and leukopenia was 5.8% (99 out of 1693 newborns) during the study period. The prevalence was highly associated with gestational age (R2 = 0.873). Twenty-four (32.4%) had abnormal MPV values at birth, defined as MPV > 9 or < 7 fL. Newborns with abnormal MPV had lower platelet counts and were more likely to have I:T ratio ≥0.2. In a univariate analysis, abnormal MPV (OR: 6.205, 95% CI: 1.923-20.022, p = .002), I:T ratio ≥0.2 (OR: 8.462, 95% CI: 1.396-51.281, p = .02), and platelet counts (OR: 98.4, 95% CI: 96.9%-99.9%, p = .035) were each significantly associated with a positive composite outcome. In a multivariate analysis, only abnormal MPV remained significantly associated with an increased likelihood of having a positive composite outcome, with an OR of 3.922 (95% CI: 1.094-14.06, p = .036).Conclusions: MPV may be a more reliable marker than I:T ratio ≥0.2 for fetal inflammatory response in newborns with combined thrombocytopenia and leukopenia during the initial assessment of intrauterine infection. Future prospective studies are required to confirm findings from this report.
Collapse
Affiliation(s)
- Chu-Yen Chen
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, MI, USA
| | | | - Amy J Johnson
- School of Medicine, University of Kansas Medical Center, Kansas City, MI, USA
| | - Gene T Lee
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, MI, USA
| | - Fu-Sheng Chou
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, MI, USA.,Department of Pediatrics, University of Missouri - Kansas City, Kansas City, MI, USA.,Division of Neonatology, Children's Mercy Kansas City, Kansas City, MI, USA
| |
Collapse
|
161
|
Fjalstad JW, Esaiassen E, Juvet LK, van den Anker JN, Klingenberg C. Antibiotic therapy in neonates and impact on gut microbiota and antibiotic resistance development: a systematic review. J Antimicrob Chemother 2019; 73:569-580. [PMID: 29182785 DOI: 10.1093/jac/dkx426] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 10/17/2017] [Indexed: 12/15/2022] Open
Abstract
Objectives To systematically review the impact of antibiotic therapy in the neonatal period on changes in the gut microbiota and/or antibiotic resistance development. Methods Data sources were PubMed, Embase, Medline and the Cochrane Database, supplemented by manual searches of reference lists. Randomized controlled trials (RCTs) and observational studies were included if they provided data on different categories of antibiotic treatment (yes versus no, long versus short duration and/or broad- versus narrow-spectrum regimens) and subsequent changes in the gut microbiota and/or antibiotic resistance development. We evaluated risk of bias using the Cochrane Handbook, adapted to include observational studies. When appropriate, we used the vote-counting method to perform semi-quantitative meta-analyses. We applied the Grades of Recommendation, Assessment, Development and Evaluation approach to rate the quality of evidence (QoE). Study protocol registration: PROSPERO CRD42015026743. Results We included 48 studies, comprising 3 RCTs and 45 observational studies. Prolonged antibiotic treatment was associated with reduced gut microbial diversity in all three studies investigating this outcome (very low QoE). Antibiotic treatment was associated with reduced colonization rates of protective commensal anaerobic bacteria in four of five studies (very low QoE). However, all three categories of antibiotic treatment were associated with an increased risk of antibiotic resistance development, in particular MDR in Gram-negative bacteria, and we graded the QoE for these outcomes as moderate. Conclusions We are moderately confident that antibiotic treatment leads to antibiotic resistance development in neonates and it may also induce potentially disease-promoting gut microbiota alterations. Our findings emphasize the need to reduce unnecessary antibiotic treatment in neonates.
Collapse
Affiliation(s)
- Jon Widding Fjalstad
- Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway.,Paediatric Research Group, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Eirin Esaiassen
- Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway.,Paediatric Research Group, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Lene Kristine Juvet
- Norwegian Institute of Public Health, PO Box 4404, Nydalen, N-0403 Oslo, Norway.,University College of Southeast Norway, Notodden, Norway
| | - John N van den Anker
- Division of Paediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland.,Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA
| | - Claus Klingenberg
- Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway.,Paediatric Research Group, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
162
|
Shah L, Hossain J, Xie S, Zaritsky J. Poor adherence to early childhood blood pressure measurement guidelines in a large pediatric healthcare system. Pediatr Nephrol 2019; 34:697-701. [PMID: 30406366 DOI: 10.1007/s00467-018-4132-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/12/2018] [Accepted: 10/29/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Children who were born prematurely, those with a very low birthweight, or who have survived the neonatal intensive care unit (NICU) are at risk for the development of hypertension and chronic kidney disease (CKD), and thus require blood pressure screening less than 3 years of age, per American Academy of Pediatrics (AAP) 2004 and 2017 guidelines. METHODS We reviewed the practice patterns of a large pediatric health care system and assessed adherence to the AAP clinical practice guidelines on blood pressure measurements in children less than 3 years of age for hypertension and CKD with the following risk factors: prematurity, very low birthweight, and a neonatal intensive care setting encounter. This retrospective chart review included a total of 9965 patients with a median gestational age of 34 weeks. RESULTS Overall, 38% of patients had at least one blood pressure measured less than 3 years of age. Primary care accounted for 41% of all outpatient encounters and 4% of all blood pressure measurements. Surgical specialties (i.e., ophthalmology, otolaryngology, and orthopedics) accounted for many non-primary care visits and were less likely than medical specialties (i.e., cardiology and nephrology) to obtain a blood pressure measurement (p < 0.0001). CONCLUSIONS This study of a large healthcare system's practice revealed a lack of basic screening for hypertension in a population known to be at risk for hypertension and CKD.
Collapse
Affiliation(s)
- Lokesh Shah
- Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| | - Jobayer Hossain
- Alfred I. duPont Hospital for Children, 1600 Rockland Rd, Wilmington, DE, 19803, USA
| | - Shirlly Xie
- Drexel University, 3141 Chestnut Street, Philadelphia, PA, 19104, USA
| | - Joshua Zaritsky
- Alfred I. duPont Hospital for Children, 1600 Rockland Rd, Wilmington, DE, 19803, USA
| |
Collapse
|
163
|
Abstract
Multicenter groups have reported reductions in the incidence of necrotizing enterocolitis (NEC) among preterm infants over the past 2 decades. These large-scale prevalence studies have coincided with reports from multicenter consortia and single centers of modifications in practice using quality-improvement techniques aimed at either reducing NEC risk specifically or reducing risk of mortality and multiple morbidities associated with extreme prematurity. The modifications in practice have been based on mechanistic studies, epidemiologic association data, and clinical trials. Recent reports from centers modifying practice to reduce NEC are reviewed and select modified/modifiable practices discussed.
Collapse
Affiliation(s)
- C Michael Cotten
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Duke University School of Medicine, Box 2739 DUMC, Durham, NC 27710, USA.
| |
Collapse
|
164
|
Allegaert K, Simons S, Van Den Anker J. Research on medication use in the neonatal intensive care unit. Expert Rev Clin Pharmacol 2019; 12:343-353. [PMID: 30741041 DOI: 10.1080/17512433.2019.1580569] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Research on medication use aims at assessing how much of current pharmacotherapy is rational. In neonates, this is hampered by extensive off-label drug use and limited knowledge. Areas covered: We report on medication use research and have conducted a systematic review of observational studies on medication use to provide an updated overview on characteristics, objectives, methods, and patterns in hospitalized neonates. Moreover, a review on aspects of medication use for opioids, anti-epileptics, gastric acid-related disorders and respiratory stimulants with emphasis on trends and impact of interventions is presented, illustrating how research on medication use can contribute to improved neonatal pharmacotherapy and more focused research. Medication use reports describe patterns and provide signals on irrational use, benchmarking, or can guide research priorities. Moreover, this may generate information on how neonatal health topics and their pharmacotherapy are handled over time or across regions. Expert opinion: Research on medicine utilization is relevant, since it will inform us on aspects like trends, variability, or about the impact and pattern of implementation of guidelines in neonates. Further progress necessitates to merge datasets on medication use with clinical characteristics, and perinatal drug use remains an area in need of additional research.
Collapse
Affiliation(s)
- Karel Allegaert
- a Department of Pediatrics, Division of Neonatology , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands.,b Department of Development and Regeneration , KU Leuven , Leuven , Belgium
| | - Sinno Simons
- a Department of Pediatrics, Division of Neonatology , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - John Van Den Anker
- c Division of Clinical Pharmacology, Department of Pediatrics , Children's National Health System , Washington , DC , USA.,d Division of Paediatric Pharmacology and Pharmacometrics , University of Basel Children's Hospital , Basel , Switzerland.,e Intensive Care and Department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , The Netherlands
| |
Collapse
|
165
|
Hendrikx D, Smits A, Lavanga M, De Wel O, Thewissen L, Jansen K, Caicedo A, Van Huffel S, Naulaers G. Measurement of Neurovascular Coupling in Neonates. Front Physiol 2019; 10:65. [PMID: 30833901 PMCID: PMC6387909 DOI: 10.3389/fphys.2019.00065] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 01/21/2019] [Indexed: 01/01/2023] Open
Abstract
Neurovascular coupling refers to the mechanism that links the transient neural activity to the subsequent change in cerebral blood flow, which is regulated by both chemical signals and mechanical effects. Recent studies suggest that neurovascular coupling in neonates and preterm born infants is different compared to adults. The hemodynamic response after a stimulus is later and less pronounced and the stimulus might even result in a negative (hypoxic) signal. In addition, studies both in animals and neonates confirm the presence of a short hypoxic period after a stimulus in preterm infants. In clinical practice, different methodologies exist to study neurovascular coupling. The combination of functional magnetic resonance imaging or functional near-infrared spectroscopy (brain hemodynamics) with EEG (brain function) is most commonly used in neonates. Especially near-infrared spectroscopy is of interest, since it is a non-invasive method that can be integrated easily in clinical care and is able to provide results concerning longer periods of time. Therefore, near-infrared spectroscopy can be used to develop a continuous non-invasive measurement system, that could be used to study neonates in different clinical settings, or neonates with different pathologies. The main challenge for the development of a continuous marker for neurovascular coupling is how the coupling between the signals can be described. In practice, a wide range of signal interaction measures exist. Moreover, biomedical signals often operate on different time scales. In a more general setting, other variables also have to be taken into account, such as oxygen saturation, carbon dioxide and blood pressure in order to describe neurovascular coupling in a concise manner. Recently, new mathematical techniques were developed to give an answer to these questions. This review discusses these recent developments.
Collapse
Affiliation(s)
- Dries Hendrikx
- Department of Electrical Engineering, KU Leuven, Leuven, Belgium
- imec, Leuven, Belgium
| | - Anne Smits
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Mario Lavanga
- Department of Electrical Engineering, KU Leuven, Leuven, Belgium
- imec, Leuven, Belgium
| | - Ofelie De Wel
- Department of Electrical Engineering, KU Leuven, Leuven, Belgium
- imec, Leuven, Belgium
| | - Liesbeth Thewissen
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Katrien Jansen
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
- Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Alexander Caicedo
- Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Sabine Van Huffel
- Department of Electrical Engineering, KU Leuven, Leuven, Belgium
- imec, Leuven, Belgium
| | - Gunnar Naulaers
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
166
|
Rational Use of Antibiotics in Neonates: Still in Search of Tailored Tools. Healthcare (Basel) 2019; 7:healthcare7010028. [PMID: 30781454 PMCID: PMC6473895 DOI: 10.3390/healthcare7010028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 01/31/2019] [Accepted: 02/14/2019] [Indexed: 12/13/2022] Open
Abstract
Rational medicine use in neonates implies the prescription and administration of age-appropriate drug formulations, selecting the most efficacious and safe dose, all based on accurate information on the drug and its indications in neonates. This review illustrates that important uncertainties still exist concerning the different aspects (when, what, how) of rational antibiotic use in neonates. Decisions when to prescribe antibiotics are still not based on robust decision tools. Choices (what) on empiric antibiotic regimens should depend on the anticipated pathogens, and the available information on the efficacy and safety of these drugs. Major progress has been made on how (beta-lactam antibiotics, aminoglycosides, vancomycin, route and duration) to dose. Progress to improve rational antibiotic use necessitates further understanding of neonatal pharmacology (short- and long-term safety, pharmacokinetics, duration and route) and the use of tailored tools and smarter practices (biomarkers, screening for colonization, and advanced therapeutic drug monitoring techniques). Implementation strategies should not only facilitate access to knowledge and guidelines, but should also consider the most effective strategies (‘skills’) and psychosocial aspects involved in the prescription process: we should be aware that both the decision not to prescribe as well as the decision to prescribe antibiotics is associated with risks and benefits.
Collapse
|
167
|
Sucasas Alonso A, Avila-Alvarez A, Combarro Eiriz M, Martínez Roca C, Yáñez Gómez P, Codias López A, Fernández Trisac JL, Pértega Díaz S. [Use of off-label drugs in neonatal intensive care]. An Pediatr (Barc) 2019; 91:237-243. [PMID: 30772271 DOI: 10.1016/j.anpedi.2018.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/10/2018] [Accepted: 12/29/2018] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To evaluate the prevalence of non-approved prescriptions (off-label and unlicensed) in a Neonatal Intensive Care Unit (NICU), and to describe factors of the neonate associated with its use. MATERIALS AND METHODS Observational prospective study in a level III NICU during a 6-month period. Every prescription was analysed using the summary of product characteristics as a reference. A sequential algorithm was used to create a classification of prescriptions based on current status: approved, unlicensed, off-label (by age, route of administration, dosage, or indication). RESULTS The study included 84 patients and 564 prescriptions. A total of 127 (22.5%) prescriptions were considered off-label, and 45 (8%) were considered unlicensed. More than half (59.5%) of the patients received at least one of these drugs, and this increases to 100% among very preterm neonates and surgical patients (P<.001). A positive linear correlation was found between duration of NICU stay and the number of off-label prescriptions (correlation coefficient 0.6; P<.001). CONCLUSIONS Non-licensed drugs are frequently prescribed in NICU, especially in the most vulnerable patients. Our results show the need to move forward on clinical research in order to homogenise the existing data about neonatology drugs, with the aim of making an efficient and safe prescription.
Collapse
Affiliation(s)
- Andrea Sucasas Alonso
- Unidad de Neonatología, Servicio de Pediatría, Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, A Coruña, España
| | - Alejandro Avila-Alvarez
- Unidad de Neonatología, Servicio de Pediatría, Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, A Coruña, España.
| | - Marina Combarro Eiriz
- Unidad de Neonatología, Servicio de Pediatría, Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, A Coruña, España
| | - Cristina Martínez Roca
- Servicio de Farmacia, Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, A Coruña, España
| | - Pedro Yáñez Gómez
- Servicio de Farmacia, Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, A Coruña, España
| | - Alejandra Codias López
- Unidad de Neonatología, Servicio de Pediatría, Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, A Coruña, España
| | - Jose Luis Fernández Trisac
- Unidad de Neonatología, Servicio de Pediatría, Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, A Coruña, España
| | - Sonia Pértega Díaz
- Unidad de Epidemiología Clínica y Bioestadística, Complexo Hospitalario Universitario A Coruña (CHUAC)-Instituto de Investigación Biomédica A Coruña (INIBIC) Sergas, A Coruña, España
| |
Collapse
|
168
|
Hoste L, George I. Ranitidine-induced Thrombocytopenia in a Neonate - A Case Report and Review of Literature. J Pediatr Pharmacol Ther 2019; 24:66-71. [PMID: 30837818 DOI: 10.5863/1551-6776-24.1.66] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Thrombocytopenia (platelet count <150 × 109/L) regularly occurs in newborns but is especially observed in critically ill neonates. We describe the case of a small for gestational age (SGA) neonate, who showed an unexpected, severe thrombocytopenia (8 × 109/L) at day 5 of life. The thrombocytopenia recovered completely after cessation of ranitidine (0.5 mg/kg/6 hr), which was started in a context of feeding difficulties. Other causes of neonatal thrombocytopenia were ruled out. Besides a brief report on a cimetidine-induced thrombocytopenia over 25 years ago, no other neonatal or pediatric cases of H2 antagonist-induced thrombocytopenia have been reported to date, although being widely used in routine care. Moreover, several adult cases have been published. In general, neonatal thrombocytopenia, although one of the most frequent hematological conditions in newborns, is only rarely attributed to an adverse drug reaction. Clinicians should be aware of the risks for adverse reactions, especially in routinely used drugs and in critically ill patients.
Collapse
|
169
|
Ziesenitz VC, Fox E, Zocchi M, Samiee-Zafarghandy S, van den Anker JN, Mazer-Amirshahi M. Prescription Drug Shortages: Impact on Neonatal Intensive Care. Neonatology 2019; 115:108-115. [PMID: 30384374 DOI: 10.1159/000493119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/22/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND Prescription drug shortages have increased significantly during the past two decades and also impact drugs used in critical care and pediatrics. OBJECTIVES To analyze drug shortages affecting medications used in neonatal intensive care units (NICUs). METHODS Drug shortage data for the top 100 NICU drugs were retrieved from the University of Utah Drug Information Service from 2001 to 2016. Data were analyzed focusing on drug class, formulation, reason for shortage, and shortage duration. RESULTS Seventy-four of the top 100 NICU drugs were impacted by 227 shortages (10.3% of total shortages). Twenty-eight (12.3%) shortages were unresolved as of December 2016. Resolved shortages had a median duration of 8.8 months (interquartile range 3.6-21.3), and generic drugs were involved in 175 (87.9%). An alternative agent was available for 171 (85.8%) drugs but 120 (70.2%) of alternatives were also affected by shortages. Parenteral drugs were involved in 172 (86.4%) shortages, with longer durations than nonparenteral drugs (9.9 vs. 6.4 months, p = 0.022). The most common shortage reason was manufacturing problems (32.2%). CONCLUSIONS Drug shortages affected many agents used in NICUs, which can have quality and safety implications for patient care, especially in extremely low birth weight infants. Neonatologists must be aware of current shortages and implement mitigation strategies to optimize patient care.
Collapse
Affiliation(s)
- Victoria C Ziesenitz
- Division of Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland, .,Department of Pediatric Cardiology, University Children's Hospital, Heidelberg, Germany,
| | - Erin Fox
- Drug Information Service, University of Utah Health, Salt Lake City, Utah, USA.,College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Mark Zocchi
- Center for Healthcare Innovation and Policy Research, George Washington University, Washington, District of Columbia, USA
| | | | - Johannes N van den Anker
- Division of Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland.,Division of Clinical Pharmacology, Children's National Health System, Washington, District of Columbia, USA
| | - Maryann Mazer-Amirshahi
- Department of Emergency Medicine, MedStar Washington Hospital Center, Washington, District of Columbia, USA.,Georgetown University School of Medicine, Washington, District of Columbia, USA
| |
Collapse
|
170
|
Cantey JB, Pyle AK, Wozniak PS, Hynan LS, Sánchez PJ. Early Antibiotic Exposure and Adverse Outcomes in Preterm, Very Low Birth Weight Infants. J Pediatr 2018; 203:62-67. [PMID: 30172430 DOI: 10.1016/j.jpeds.2018.07.036] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/28/2018] [Accepted: 07/11/2018] [Indexed: 01/19/2023]
Abstract
OBJECTIVES To determine whether antibiotic use in the first 14 postnatal days in preterm, very low birth weight (birth weight of ≤1500 g) infants is associated with risk after 14 days of age for late-onset sepsis, necrotizing enterocolitis (NEC), or death after controlling for severity of illness using the Clinical Risk Index in Babies II score, and determine whether duration of antibiotic exposure was associated with risk of adverse outcomes. STUDY DESIGN This retrospective cohort study included very low birth weight infants born at ≤326/7 weeks of gestation admitted to the neonatal intensive care unit from September 2010 to June 2014. Infants were excluded if they had major congenital anomalies or culture-proven sepsis, NEC, or death during the first 14 days of life. Antibiotic exposure was recorded as days of therapy and length of therapy in days. RESULTS Of 374 infants, 70 (19%) had late-onset sepsis, NEC, or death after 14 days of age. The median number of antibiotic days of therapy and length of therapy were 5.5 and 3.0, respectively. In multivariate analysis after controlling for severity of illness, each antibiotic day of therapy was associated with a 1.24 times increased risk of sepsis, NEC, or death (OR, 1.24; 95% CI, 1.17-1.31). Risk was similar when length of therapy was used (OR, 1.47; 95% CI, 1.32-1.64). CONCLUSIONS After controlling for severity of illness, each day of antibiotic therapy provided to preterm, very low birth weight infants in the first 2 weeks of age is associated with an increased risk of late-onset sepsis, NEC, or death.
Collapse
Affiliation(s)
- Joseph B Cantey
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX; Division of Neonatal-Perinatal Medicine and Pediatric Infectious Diseases, University of Texas Southwestern Medical Center, Dallas, TX; University of Texas Health Science Center San Antonio, San Antonio, TX.
| | - Alaina K Pyle
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX; Division of Neonatal-Perinatal Medicine and Pediatric Infectious Diseases, University of Texas Southwestern Medical Center, Dallas, TX; Division of Neonatology, Yale School of Medicine, New Haven, CT
| | - Phillip S Wozniak
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX; Division of Neonatology and Pediatric Infectious Diseases, Center for Perinatal Research, Nationwide Children's Hospital-The Ohio State University College of Medicine, Columbus, OH
| | - Linda S Hynan
- Clinical Sciences and Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Pablo J Sánchez
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX; Division of Neonatal-Perinatal Medicine and Pediatric Infectious Diseases, University of Texas Southwestern Medical Center, Dallas, TX; Division of Neonatology and Pediatric Infectious Diseases, Center for Perinatal Research, Nationwide Children's Hospital-The Ohio State University College of Medicine, Columbus, OH
| |
Collapse
|
171
|
Puia-Dumitrescu M, Bretzius OM, Brown N, Fitz-Henley JA, Ssengonzi R, Wechsler CS, Gray KD, Benjamin DK, Smith PB, Clark RH, Gonzalez D, Hornik CP. Evaluation of Gentamicin Exposure in the Neonatal Intensive Care Unit and Hearing Function at Discharge. J Pediatr 2018; 203:131-136. [PMID: 30244991 PMCID: PMC6361629 DOI: 10.1016/j.jpeds.2018.07.101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/25/2018] [Accepted: 07/30/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To characterize the association between gentamicin dosing, duration of treatment, and ototoxicity in hospitalized infants. STUDY DESIGN This retrospective cohort study conducted at 330 neonatal intensive care units (2002-2014) included inborn infants exposed to gentamicin with available hearing screen results, and excluded infants with incomplete dosing data and major congenital anomalies. Our primary outcome was the final hearing screen result performed during hospitalization: abnormal (failed or referred for further testing in one or both ears) or normal (bilateral passed). The 4 measures of gentamicin exposure were highest daily dose, average daily dose, cumulative dose, and cumulative duration of exposure. We fitted separate multivariable logistic regression models adjusted for demographics, comorbidities, and other clinical events. RESULTS A total of 84 808 infants met inclusion/exclusion criteria; median (25th, 75th percentile) gestational age and birth weight were 35 weeks (33, 38) and 2480 g (1890, 3184), respectively. Failed hearing screens occurred in 3238 (3.8%) infants; failed screens were more likely in infants of lower gestational age and birth weight, who had longer hospital lengths of stay, higher rates of morbidities, and were small for gestational age. Median highest daily dose, average daily dose, and cumulative dose were 4.0 mg/kg/day (3.0, 4.0), 3.8 mg/kg/day (3.0, 4.0), and 12.1 mg/kg (9.1, 20.5), respectively. Median cumulative duration of exposure was 3 days (3, 6). In adjusted analysis, gentamicin dose and duration of therapy were not associated with hearing screen failure. CONCLUSIONS Gentamicin dosing and duration of treatment were not associated with increased odds of failed hearing screen at the time of discharge from initial neonatal intensive care unit stay.
Collapse
Affiliation(s)
- Mihai Puia-Dumitrescu
- Department of Pediatrics, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC
| | | | - Nia Brown
- Duke Clinical Research Institute, Durham, NC
| | | | | | | | - Keyaria D Gray
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | | | - P Brian Smith
- Department of Pediatrics, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC
| | - Reese H Clark
- Pediatrix-Obstetrix Center for Research and Education, Sunrise, FL
| | - Daniel Gonzalez
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC
| | - Christoph P Hornik
- Department of Pediatrics, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC.
| |
Collapse
|
172
|
Jackson W, Taylor G, Selewski D, Smith PB, Tolleson-Rinehart S, Laughon MM. Association between furosemide in premature infants and sensorineural hearing loss and nephrocalcinosis: a systematic review. Matern Health Neonatol Perinatol 2018; 4:23. [PMID: 30473868 PMCID: PMC6240934 DOI: 10.1186/s40748-018-0092-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/23/2018] [Indexed: 11/10/2022] Open
Abstract
Furosemide is a potent loop diuretic commonly and variably used by neonatologists to improve oxygenation and lung compliance in premature infants. There are several safety concerns with use of furosemide in premature infants, specifically the risk of sensorineural hearing loss (SNHL), and nephrocalcinosis/nephrolithiasis (NC/NL). We conducted a systematic review of all trials and observational studies examining the association between these outcomes with exposure to furosemide in premature infants. We searched MEDLINE, EMBASE, CINAHL, and clinicaltrials.gov. We included studies reporting either SNHL or NC/NL in premature infants (< 37 weeks completed gestational age) who received at least one dose of enteral or intravenous furosemide. Thirty-two studies met full inclusion criteria for the review, including 12 studies examining SNHL and 20 studies examining NC/NL. Only one randomized controlled trial was identified in this review. We found no evidence that furosemide exposure increases the risk of SNHL or NC/NL in premature infants, with varying quality of studies and found the strength of evidence for both outcomes to be low. The most common limitation in these studies was the lack of control for confounding factors. The evidence for the risk of SNHL and NC/NL in premature infants exposed to furosemide is low. Further randomized controlled trials of furosemide in premature infants are urgently needed to adequately assess the risk of SNHL and NC/NL, provide evidence for improved FDA labeling, and promote safer prescribing practices.
Collapse
Affiliation(s)
- Wesley Jackson
- 1Division of Pediatrics, University of North Carolina at Chapel Hill, UNC Hospitals 101 Manning Dr. 4th Floor, Chapel Hill, NC CB 7596 USA
| | - Genevieve Taylor
- 1Division of Pediatrics, University of North Carolina at Chapel Hill, UNC Hospitals 101 Manning Dr. 4th Floor, Chapel Hill, NC CB 7596 USA
| | - David Selewski
- 2Division of Nephrology, Department of Pediatrics and Communicable Diseases, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, MI USA
| | - P Brian Smith
- 3Duke Department of Pediatrics, Duke University Medical Center, Durham, NC USA
| | - Sue Tolleson-Rinehart
- 1Division of Pediatrics, University of North Carolina at Chapel Hill, UNC Hospitals 101 Manning Dr. 4th Floor, Chapel Hill, NC CB 7596 USA.,4Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Matthew M Laughon
- 1Division of Pediatrics, University of North Carolina at Chapel Hill, UNC Hospitals 101 Manning Dr. 4th Floor, Chapel Hill, NC CB 7596 USA
| |
Collapse
|
173
|
Abstract
Time of day is a critical factor for most biological functions, but concepts from the field of chronobiology have yet to be fully translated to clinical practice. Circadian rhythms, generated internally and synchronised to the external environment, promote function and support survival in almost every living species. Fetal circadian rhythms can be observed in utero from 30weeks gestation, coupled to the maternal rhythm, but synchronise to the external environment only after birth. Important cues for synchronisation include the light/dark cycle, the timing of feeding, and exposure to melatonin in breast milk. Disruption to these cues may occur during admission to the neonatal intensive care unit. This can impair the development of circadian rhythms, and influence survival and function in the neonatal period, with a potential to impact health and well-being throughout adult life. Here we outline the rationale and evidence to support a chronobiological approach to neonatal care.
Collapse
Affiliation(s)
- Helen McKenna
- Critical Care Unit, Royal Free Hospital, Pond Street, London NW3 2QG, UK; Division of Surgery and Interventional Science, University College London, UK.
| | - Irwin Karl Marcel Reiss
- Division of Neonatology, Department of Paediatrics, Erasmus Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
174
|
McCarthy KN, Hawke A, Dempsey EM. Antimicrobial stewardship in the neonatal unit reduces antibiotic exposure. Acta Paediatr 2018; 107:1716-1721. [PMID: 29603353 DOI: 10.1111/apa.14337] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/09/2018] [Accepted: 03/21/2018] [Indexed: 11/30/2022]
Abstract
AIM Antimicrobial stewardship plays an important role in ensuring that the appropriate drug, dose, route and duration are employed to provide adequate treatment while minimising the risks of unnecessary antibiotic use. Surveillance of antibiotic use with prescriber feedback is recommended as a high-impact stewardship intervention. The aim of this study was to reduce unnecessary antimicrobial use in a neonatal unit. METHODS A prospective audit was performed to assess compliance with antimicrobial guidelines. Following this, educational interventions were applied, electronic prescribing was introduced to the neonatal unit, and re-audit was performed. The primary outcome was a reduction in days of therapy (DOT). RESULTS There were 312 neonatal admissions. There was a significant overall reduction in the primary outcome of DOT/1000 patient days from 572 to 417 DOT. This represents a 27% reduction in total antibiotic use. Prolonged antibiotic treatment courses >36 hours in negative sepsis evaluations were reduced from 82 DOT to 7.5 DOT. Similarly, treatment courses greater than five days for culture-negative sepsis were reduced from 46.5 DOT to 7 DOT. CONCLUSION Monitoring antibiotic prescribing data can provide useful insights into the trends of antibiotic use and also inform clinicians of potential areas where antibiotic use may be safely reduced.
Collapse
Affiliation(s)
- KN McCarthy
- Department of Paediatrics and Child Health; Neonatal Intensive Care Unit; Cork Ireland
| | - A Hawke
- Department of Paediatrics and Child Health; Neonatal Intensive Care Unit; Cork Ireland
| | - EM Dempsey
- Department of Paediatrics and Child Health; Neonatal Intensive Care Unit; Cork Ireland
- INFANT, Irish Centre for Fetal and Neonatal Translational Research; University College Cork; Cork Ireland
| |
Collapse
|
175
|
McPherson C, Liviskie C, Zeller B, Nelson MP, Newland JG. Antimicrobial Stewardship in Neonates: Challenges and Opportunities. Neonatal Netw 2018; 37:116-123. [PMID: 29615159 DOI: 10.1891/0730-0832.37.2.116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Neonatal infections result in significant morbidity and mortality. Antibiotics are vital for the treatment of infections but disrupt the neonatal microbiome, put the infant at risk for an adverse drug reaction, and may lead to the development of antibiotic resistance. Immediately after birth, clinicians must determine which infants require empiric antibiotics. Online risk stratification tools may provide a superior approach to decision trees. In infants who require empiric therapy for early-onset sepsis, ampicillin and an aminoglycoside with dosing based on recent pharmacokinetic studies represents the most appropriate first-line agents; third-generation cephalosporins should be reserved for patients with a high likelihood of Gram-negative meningitis. An antistaphylococcal penicillin and gentamicin should be utilized for suspected late-onset sepsis. Vancomycin and other broad-spectrum agents are reserved for patients with a history of resistant organisms. Antibiotic duration should be guided by understanding the clinical indications and obtaining the necessary cultures appropriately (i.e., adequate volume blood cultures). In the absence of a positive culture, antibiotic duration should often be limited. Individual institutions should leverage a multidisciplinary, interprofessional team to identify opportunities for antimicrobial stewardship. A collaborative, transparent system is required to change unit culture and generate a sustained impact on antibiotic utilization with optimal patient outcomes.
Collapse
|
176
|
Costa HTMDL, Costa TX, Martins RR, Oliveira AG. Use of off-label and unlicensed medicines in neonatal intensive care. PLoS One 2018; 13:e0204427. [PMID: 30252920 PMCID: PMC6155521 DOI: 10.1371/journal.pone.0204427] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 09/09/2018] [Indexed: 01/18/2023] Open
Abstract
Purpose To evaluate the use of off-label and unlicensed medicines in a neonatal intensive care unit (NICU) of a teaching maternity hospital specialized in high risk pregnancy. Methods A prospective cohort study was conducted between August 2015 and July 2016. All newborns admitted to the NICU who had at least one medication prescribed and a hospital stay longer than 24 hours were included. The classification of off-label and unlicensed drugs for the neonatal population was done according to the information of Food and Drug Administration. Results A total of 17421 medication items were analyzed in 3935 prescriptions of 220 newborns. The proportion of newborns exposed to off-label drugs was 96.4%, and to unlicensed medicines was 66.8%. About one-half (49.3%) of the medication items were off-label and 24.6% were unlicensed. The main reason for off-label and unlicensed classification was, respectively, frequency of administration and the administration of adaptations of pharmaceutical forms. Conclusions Although there are actions to encourage the development of pharmacological studies with neonates, this study observed a high rate of prescription and exposure of newborns to off-label and unlicensed drugs in NICUs and pointed out areas of neonatal therapy that require scientific investment.
Collapse
Affiliation(s)
- Haline Tereza Matias de Lima Costa
- Integrated Multiprofessional Health Residency Program—Neonatal Intensive Care Unit, Pharmacy Department, Health Sciences Centre, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
- * E-mail:
| | - Tatiana Xavier Costa
- School Maternity Januário Cicco, Health Sciences Centre, Universidade Federal do Rio Grande Norte, Natal, RN, Brazil
| | - Rand Randall Martins
- Pharmacy Department, Health Sciences Centre, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | - Antônio Gouveia Oliveira
- Pharmacy Department, Health Sciences Centre, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
177
|
Allegaert K. Rational Use of Medicines in Neonates: Current Observations, Areas for Research and Perspectives. Healthcare (Basel) 2018; 6:healthcare6030115. [PMID: 30223533 PMCID: PMC6165407 DOI: 10.3390/healthcare6030115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 12/16/2022] Open
Abstract
A focused reflection on rational medicines use in neonates is valuable and relevant, because indicators to assess rational medicines use are difficult to apply to neonates. Polypharmacy and exposure to antibiotics are common, while dosing regimens or clinical guidelines are only rarely supported by robust evidence in neonates. This is at least in part due to the extensive variability in pharmacokinetics and subsequent effects of medicines in neonates. Medicines utilization research informs us on trends, on between unit variability and on the impact of guideline implementation. We illustrate these aspects using data on drugs for gastroesophageal reflux, analgesics or anti-epileptic drugs. Areas for additional research are drug-related exposure during breastfeeding (exposure prediction) and how to assess safety (tools to assess seriousness, causality, and severity tailored to neonates) since both efficacy and safety determine rational drug use. To further improve rational medicines use, we need more data and tools to assess efficacy and safety in neonates. Moreover, we should facilitate access to such data, and explore strategies for effective implementation. This is because prescription practices are not only rational decisions, but also have psychosocial aspects that may guide clinicians to irrational practices, in part influenced by the psychosocial characteristics of this population.
Collapse
Affiliation(s)
- Karel Allegaert
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Doctor Molenwaterplein 40, 3015 GD Rotterdam, The Netherlands.
- Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
178
|
Godamudunage MP, Grech AM, Scott EE. Comparison of Antifungal Azole Interactions with Adult Cytochrome P450 3A4 versus Neonatal Cytochrome P450 3A7. Drug Metab Dispos 2018; 46:1329-1337. [PMID: 29991575 PMCID: PMC6081698 DOI: 10.1124/dmd.118.082032] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/09/2018] [Indexed: 12/31/2022] Open
Abstract
Adult drug metabolism is dominated by cytochrome P450 3A4 (CYP3A4), which is often inhibited by antifungal azole drugs, resulting in potential alterations in drug metabolism and adverse drug/drug interactions. In the fetal and neonatal stages of life, the 87% identical cytochrome P450 3A7 (CYP3A7) is expressed but not CYP3A4. Azole antifungals developed for adults are also used in neonates, assuming they interact similarly with both enzymes, but systematic information is lacking. Herein a method was developed for generating recombinant purified CYP3A7. Thirteen different azoles were then evaluated for binding and inhibition of purified human CYP3A4 versus CYP3A7. All imidazole-containing azoles bound both enzymes via coordination to the heme iron and inhibited both with IC50 values ranging from 180 nM for clotrimazole to the millimolar range for imidazole itself. Across this wide range of potencies, CYP3A4 was consistently inhibited more strongly than CYP3A7, with clotrimazole being the least selective (1.5-fold) inhibitor and econazole the most selective (12-fold). Observations for 1,2,4-triazole-containing azoles were more varied. Most bound to CYP3A4 via coordination to the heme iron, but several also demonstrated evidence of a distinct binding mode at low concentrations. However, only posaconazole inhibited CYP3A4. Of the triazoles, only posaconazole inhibited CYP3A7, again less potently than CYP3A4. Spectral evidence for binding was weak or nonexistent for all triazoles. Overall, although the details of binding interactions do vary, the same azole compounds inhibit both enzymes, albeit with weaker interactions with CYP3A7 compared with CYP3A4.
Collapse
Affiliation(s)
- Malika P Godamudunage
- Departments of Medicinal Chemistry (M.P.G., A.M.G., E.E.S.) and Pharmacology (E.E.S.), University of Michigan, Ann Arbor, Michigan
| | - Anne M Grech
- Departments of Medicinal Chemistry (M.P.G., A.M.G., E.E.S.) and Pharmacology (E.E.S.), University of Michigan, Ann Arbor, Michigan
| | - Emily E Scott
- Departments of Medicinal Chemistry (M.P.G., A.M.G., E.E.S.) and Pharmacology (E.E.S.), University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
179
|
Proteomics of human liver membrane transporters: a focus on fetuses and newborn infants. Eur J Pharm Sci 2018; 124:217-227. [PMID: 30171984 DOI: 10.1016/j.ejps.2018.08.042] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Hepatic membrane transporters are involved in the transport of many endogenous and exogenous compounds, including drugs. We aimed to study the relation of age with absolute transporter protein expression in a cohort of 62 mainly fetus and newborn samples. METHODS Protein expressions of BCRP, BSEP, GLUT1, MCT1, MDR1, MRP1, MRP2, MRP3, NTCP, OCT1, OATP1B1, OATP1B3, OATP2B1 and ATP1A1 were quantified with LC-MS/MS in isolated crude membrane fractions of snap-frozen post-mortem fetal and pediatric, and surgical adult liver samples. mRNA expression was quantified using RNA sequencing, and genetic variants with TaqMan assays. We explored relationships between protein expression and age (gestational age [GA], postnatal age [PNA], and postmenstrual age); between protein and mRNA expression; and between protein expression and genotype. RESULTS We analyzed 36 fetal (median GA 23.4 weeks [range 15.3-41.3]), 12 premature newborn (GA 30.2 weeks [24.9-36.7], PNA 1.0 weeks [0.14-11.4]), 10 term newborn (GA 40.0 weeks [39.7-41.3], PNA 3.9 weeks [0.3-18.1]), 4 pediatric (PNA 4.1 years [1.1-7.4]) and 8 adult liver samples. A relationship with age was found for BCRP, BSEP, GLUT1, MDR1, MRP1, MRP2, MRP3, NTCP, OATP1B1 and OCT1, with the strongest relationship for postmenstrual age. For most transporters mRNA and protein expression were not correlated. No genotype-protein expression relationship was detected. DISCUSSION AND CONCLUSION Various developmental patterns of protein expression of hepatic transporters emerged in fetuses and newborns up to four months of age. Postmenstrual age was the most robust factor predicting transporter expression in this cohort. Our data fill an important gap in current pediatric transporter ontogeny knowledge.
Collapse
|
180
|
Thompson EJ, Greenberg RG, Kumar K, Laughon M, Smith PB, Clark RH, Crowell A, Shaw L, Harrison L, Scales G, Bell N, Hornik CP. Association between Furosemide Exposure and Patent Ductus Arteriosus in Hospitalized Infants of Very Low Birth Weight. J Pediatr 2018; 199:231-236. [PMID: 29752171 PMCID: PMC6063759 DOI: 10.1016/j.jpeds.2018.03.067] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/26/2018] [Accepted: 03/27/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To evaluate the association between furosemide exposure and patent ductus arteriosus (PDA) in a large, contemporary cohort of hospitalized infants with very low birth weight (VLBW). STUDY DESIGN Using the Pediatrix Medical Group Clinical Data Warehouse, we identified all inborn infants of VLBW <37 weeks of gestation discharged from the neonatal intensive care unit after the first postnatal week from 2011 to 2015. We defined PDA as any medical (ibuprofen or indomethacin) or surgical PDA therapy. We collected data up to the day of PDA treatment or postnatal day 18 for infants not diagnosed with PDA. We performed multivariable logistic regression to evaluate the association between PDA and exposure to furosemide. RESULTS We included 43 576 infants from 337 neonatal intensive care units, of whom 6675 (15%) underwent PDA treatment. Infants with PDA were more premature and more often exposed to mechanical ventilation and inotropes. Furosemide was prescribed to 4055 (9%) infants. On multivariable regression, exposure to furosemide was associated with decreased odds of PDA treatment (OR 0.72; 95% CI 0.65-0.79). Increasing percentage of days with furosemide exposure was not associated with PDA treatment (OR 1.01; 95% CI 0.97-1.06). CONCLUSIONS Furosemide exposure was not associated with increased odds of PDA treatment in hospitalized infants of VLBW. Further studies are needed to characterize the efficacy and safety of furosemide in premature infants.
Collapse
Affiliation(s)
| | - Rachel G Greenberg
- Duke Clinical Research Institute & Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Karan Kumar
- Duke Clinical Research Institute & Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Matthew Laughon
- Department of Pediatrics & Division of Neonatology, University of North Carolina, Chapel Hill, NC
| | - P Brian Smith
- Duke Clinical Research Institute & Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Reese H Clark
- The Pediatrix-Obstetrix Center for Research, Education, and Quality, Sunrise, FL
| | | | - Layla Shaw
- Duke Star Program, Duke Clinical Research Institute, Durham, NC
| | - Louis Harrison
- Duke Star Program, Duke Clinical Research Institute, Durham, NC
| | | | - Nicole Bell
- Duke Star Program, Duke Clinical Research Institute, Durham, NC
| | - Christoph P Hornik
- Duke Clinical Research Institute & Department of Pediatrics, Duke University School of Medicine, Durham, NC.
| |
Collapse
|
181
|
Does duration of caffeine therapy in preterm infants born ≤1250 g at birth influence neurodevelopmental (ND) outcomes at 3 years of age? J Perinatol 2018; 38:889-899. [PMID: 29740190 DOI: 10.1038/s41372-018-0106-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/28/2018] [Accepted: 03/06/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To evaluate the effect of duration of caffeine use on long-term neurodevelopmental (ND) outcomes at 3 years corrected age (CA) in preterm infants with birthweights (BW) ≤ 1250 g. DESIGN/METHODS All surviving infants with BW ≤ 1250 g admitted to the Foothills Medical Center neonatal intensive care unit (NICU) from January 2002 to December 2009 who received the first dose of caffeine in the first week of life and were followed up at three years CA were included in the study. Demographics and follow-up outcomes were compared based on early cessation of caffeine ≤ 14 days (ECC), intermediate cessation of caffeine 15-30 days (ICC), and late cessation of caffeine >30 days (LCC). The primary outcome of ND impairment was present if a child had any one of the following: cerebral palsy, cognitive delay, visual impairment, or hearing impairment or deafness. Univariate and logistic regression analyses were performed. RESULTS Of the 508 eligible infants, 448 (88%) were seen at 3 years CA at follow-up. ECC (n = 139), ICC (n = 122) and LCC (n = 187) groups had a median (range) BW of 979 (560-1250), 1010 (530-1250), and 980 (520-1250) g (p = 0.524) and median (range) gestational age (GA) of 27 (23-33), 28 (24-33), and 27 (24-32) weeks, respectively (p = 0.034). In logistic regression models adjusting for GA, maternal smoking, and each neonatal risk factor separately (IVH, NEC, sepsis, blood transfusions, BPD, postnatal dexamethasone, SNAP-II, and ventilator days), none of the models showed a statistically significant association between caffeine duration and ND impairment. CONCLUSION The duration of caffeine use in premature infants in the NICU does not impact on long-term ND outcomes at 3 years CA.
Collapse
|
182
|
Soo JY, Wiese MD, Berry MJ, McMillen IC, Morrison JL. Intrauterine growth restriction may reduce hepatic drug metabolism in the early neonatal period. Pharmacol Res 2018; 134:68-78. [PMID: 29890254 DOI: 10.1016/j.phrs.2018.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/23/2018] [Accepted: 06/04/2018] [Indexed: 11/26/2022]
Abstract
The effects of intrauterine growth restriction (IUGR) extend well into postnatal life. IUGR is associated with an increased risk of adverse health outcomes, which often leads to greater medication usage. Many medications require hepatic metabolism for activation or clearance, but hepatic function may be altered in IUGR fetuses. Using a sheep model of IUGR, we determined the impact of IUGR on hepatic drug metabolism and drug transporter expression, both important mediators of fetal drug exposure, in late gestation and in neonatal life. In the late gestation fetus, IUGR decreased the gene expression of uptake drug transporter OATPC and increased P-glycoprotein protein expression in the liver, but there was no change in the activity of the drug metabolising enzymes CYP3A4 or CYP2D6. In contrast, at 3 weeks of age, CYP3A4 activity was reduced in the livers of lambs born with low birth weight (LBW), indicating that LBW results in changes to drug metabolising capacity in neonatal life. Together, these results suggest that IUGR may reduce hepatic drug metabolism in fetal and neonatal life through different mechanisms.
Collapse
Affiliation(s)
- Jia Yin Soo
- Early Origins of Adult Health Research Group, Adelaide, SA, 5001, Australia; School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, 5001, Australia
| | - Michael D Wiese
- School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, 5001, Australia
| | - Mary J Berry
- Centre for Translational Physiology, Wellington, New Zealand; Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | | | - Janna L Morrison
- Early Origins of Adult Health Research Group, Adelaide, SA, 5001, Australia; School of Pharmacy & Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, 5001, Australia.
| |
Collapse
|
183
|
Pakvasa MA, Saroha V, Patel RM. Optimizing Caffeine Use and Risk of Bronchopulmonary Dysplasia in Preterm Infants: A Systematic Review, Meta-analysis, and Application of Grading of Recommendations Assessment, Development, and Evaluation Methodology. Clin Perinatol 2018; 45:273-291. [PMID: 29747888 DOI: 10.1016/j.clp.2018.01.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Caffeine reduces the risk of bronchopulmonary dysplasia (BPD). Optimizing caffeine use could increase therapeutic benefit. We performed a systematic-review and random-effects meta-analysis of studies comparing different timing of initiation and dose of caffeine on the risk of BPD. Earlier initiation, compared to later, was associated with a decreased risk of BPD (5 observational studies; n = 63,049, adjusted OR 0.69; 95% CI 0.64-0.75, GRADE: low quality). High-dose caffeine, compared to standard-dose, was associated with a decreased risk of BPD (3 randomized trials, n = 432, OR 0.65; 95% CI 0.43-0.97; GRADE: low quality). Higher quality evidence is needed to guide optimal caffeine use.
Collapse
Affiliation(s)
- Mitali Atul Pakvasa
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Drive Northeast, 3rd Floor, Atlanta, GA 30322, USA
| | - Vivek Saroha
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Drive Northeast, 3rd Floor, Atlanta, GA 30322, USA
| | - Ravi Mangal Patel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Drive Northeast, 3rd Floor, Atlanta, GA 30322, USA.
| |
Collapse
|
184
|
Cantey JB, Anderson KR, Kalagiri RR, Mallett LH. Morbidity and mortality of coagulase-negative staphylococcal sepsis in very-low-birth-weight infants. World J Pediatr 2018. [PMID: 29536341 DOI: 10.1007/s12519-018-0145-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Coagulase-negative staphylococci (CoNS) are the most common cause of late-onset sepsis in the neonatal intensive care unit (NICU) and usually require vancomycin treatment. Our objective was to determine whether CoNS are associated with neonatal morbidity and mortality. METHODS This was a retrospective cohort study of very-low-birth-weight (VLBW, ≤ 1500 g) infants from 1989 to 2015. Exclusion criteria were major congenital anomaly or death within 72 h. CoNS was considered a pathogen if recovered from ≥ 2 cultures, or 1 culture if treated for ≥ 5 days and signs of sepsis were present. Logistic regression was used to examine factors associated with morbidity and mortality. RESULTS Of 2242 VLBW infants, 285 (12.7%) had late-onset sepsis. CoNS (125, 44%), Staphylococcus aureus (52, 18%), and Escherichia coli (36, 13%) were the most commonly recovered organisms. In multivariate analysis, CoNS sepsis was not associated with mortality [OR 0.6 (95% CI 0.2-2.6)), but sepsis with other organisms was [OR 4.5 (95% CI 2.6-8.0)]. CoNS sepsis was associated with longer hospitalization but not risk for bronchopulmonary dysplasia, intraventricular hemorrhage, or retinopathy of prematurity. CONCLUSION CoNS sepsis was not associated with mortality or morbidities other than length of stay. These findings support vancomycin-reduction strategies in the NICU.
Collapse
Affiliation(s)
- Joseph B Cantey
- Department of Pediatrics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| | | | - Ram R Kalagiri
- Texas A&M Health Science Center, Baylor/Scott & White Health, Temple, TX, USA
| | - Lea H Mallett
- Texas A&M Health Science Center, Baylor/Scott & White Health, Temple, TX, USA
| |
Collapse
|
185
|
Flannery DD, Ross RK, Mukhopadhyay S, Tribble AC, Puopolo KM, Gerber JS. Temporal Trends and Center Variation in Early Antibiotic Use Among Premature Infants. JAMA Netw Open 2018; 1:e180164. [PMID: 30646054 PMCID: PMC6324528 DOI: 10.1001/jamanetworkopen.2018.0164] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
IMPORTANCE Premature infants are frequently administered empirical antibiotic therapy at birth. Early and prolonged antibiotic exposures among infants without culture-confirmed infection have been associated with increased risk of adverse outcomes. OBJECTIVE To examine early antibiotic use among premature infants over time and across hospitals in the United States. DESIGN, SETTING, AND PARTICIPANTS This retrospective cohort study used a comprehensive administrative database of inpatient encounters from 297 academic and community hospitals across the United States to examine data concerning very low-birth-weight (VLBW) infants (<1500 g), including extremely low-birth-weight (ELBW) infants (<1000 g), who were admitted to the neonatal intensive care unit and survived for at least 1 day. Data collection took place in November 2015 and analysis took place from February 2016 to November 2016. EXPOSURES Antibiotic initiation within the first 3 days of age and subsequent antibiotic administration for more than 5 days. MAIN OUTCOMES AND MEASURES Temporal trends in early antibiotic initiation and duration from 2009 to 2015, and center variation in early antibiotic use from 2014 to 2015. RESULTS We identified 40 364 VLBW infants (20 447 female [50.7%]) who survived for at least 1 day, including 12 947 ELBW infants, from 297 centers. The majority of premature infants had early antibiotic initiation (31 715 VLBW infants [78.6%] and 11 264 ELBW infants [87.0%]), and no differences were observed over time in temporal trend analyses (P = .12 for VLBW and P = .52 for ELBW). The annual risk difference in the proportion of VLBW infants administered early antibiotic therapy ranged from -0.75% (95% CI, -1.61% to 0.11%) to -0.87% (95% CI, -2.04% to 0.30%); in ELBW infants the annual risk difference ranged from -0.34% (95% CI, -1.28% to 0.61%) to -0.38% (95% CI, -1.61% to 0.85%). There was a small but significant decrease over time in the rate of prolonged antibiotic duration for VLBW infants (P = .02), but not for ELBW infants (P = .22). The annual risk difference in the proportion of VLBW infants with prolonged antibiotic duration ranged from -0.94% (95% CI, -1.65% to -0.23%) to -1.08% (95% CI, -2.00% to -0.16%); in ELBW infants the annual risk difference ranged from -0.72% (95% CI, -1.83% to 0.39%) to -0.75% (95% CI, -1.96% to 0.46%). We also observed variation in early antibiotic exposures across centers. Sixty-nine of 113 centers (61.1%) started antibiotic therapy for more than 75% of VLBW infants, and 56 of 66 centers (84.8%) started antibiotic therapy for more than 75% of ELBW infants. The proportion of VLBW and ELBW infants administered prolonged antibiotics ranged from 0% to 80.4% and 0% to 92.0% across centers, respectively. CONCLUSIONS AND RELEVANCE Most premature infants in this study received empirical early antibiotic therapy with little change over a recent 7-year period. The variability in exposure rates across centers, however, suggests that neonatal antimicrobial stewardship efforts are warranted to optimize antibiotic use for VLBW and ELBW infants.
Collapse
Affiliation(s)
- Dustin D Flannery
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia
| | - Rachael K Ross
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Pediatric Infectious Diseases, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia
| | - Sagori Mukhopadhyay
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia
| | - Alison C Tribble
- Division of Pediatric Infectious Diseases, C. S. Mott Children's Hospital, University of Michigan, Ann Arbor
| | - Karen M Puopolo
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia
| | - Jeffrey S Gerber
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Pediatric Infectious Diseases, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia
| |
Collapse
|
186
|
Flint RB, van Beek F, Andriessen P, Zimmermann LJ, Liem KD, Reiss IKM, de Groot R, Tibboel D, Burger DM, Simons SHP. Large differences in neonatal drug use between NICUs are common practice: time for consensus? Br J Clin Pharmacol 2018; 84:1313-1323. [PMID: 29624207 PMCID: PMC5980600 DOI: 10.1111/bcp.13563] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 01/24/2018] [Accepted: 02/11/2018] [Indexed: 01/27/2023] Open
Abstract
Aims Evidence for drug use in newborns is sparse, which may cause large differences in drug prescriptions. We aimed to investigate the differences between neonatal intensive care units (NICUs) in the Netherlands in currently prescribed drugs. Methods This multicentre study included neonates admitted during 12 months to four different NICUs. Drugs were classified in accordance with the Anatomical Therapeutic Chemical (ATC) classification system and assessed for on/off‐label status in relation to neonatal age. The treatment protocols for four common indications for drug use were compared: pain, intubation, convulsions and hypotension. Results A total of 1491 neonates (GA range 23+6–42+2 weeks) were included with a total of 32 182 patient days, 181 different drugs and 10 895 prescriptions of which 23% was off‐label in relation to neonatal age. Overall, anti‐infective drugs were most frequently used with a total of 3161 prescriptions, of which 4% was off‐label in relation to neonatal age. Nervous system drugs included 2500 prescriptions of which 31% was off‐label in relation to neonatal age. Nervous system drugs, blood and blood forming organs, and cardiovascular drugs showed the largest differences between NICUs with ranges of 919–2278, 554–1465, and 238–952 total prescriptions per 1000 patients per ATC class, respectively. Conclusions We showed that drug use varies widely in neonatal clinical practice. The drug classes with the highest proportion of off‐label drugs in relation to neonatal age showed the largest differences between NICUs, i.e. cardiovascular and nervous system drugs. Drug research in neonates should receive high priority to guarantee safe and appropriate medicines and optimal treatment.
Collapse
Affiliation(s)
- Robert B Flint
- Department of Pediatrics, Division of Neonatology, Erasmus University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Pharmacy and Radboud Institute of Health Sciences (RIHS), Radboudumc, Nijmegen, The Netherlands.,Department of Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Floor van Beek
- Department of Pediatrics, Division of Neonatology, Erasmus University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Peter Andriessen
- Department of Pediatrics, Division of Neonatology, Máxima Medical Centre, Veldhoven, The Netherlands
| | - Luc J Zimmermann
- Department of Pediatrics, Maastricht University Medical Center, School of Oncology and Developmental Biology, School of Mental Health and Neuroscience, Maastricht, The Netherlands
| | - Kian D Liem
- Department of Pediatrics, Division of Neonatology, Radboudumc, Nijmegen, Nijmegen, The Netherlands
| | - Irwin K M Reiss
- Department of Pediatrics, Division of Neonatology, Erasmus University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Ronald de Groot
- Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Department of Pediatrics, Erasmus University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - David M Burger
- Department of Pharmacy and Radboud Institute of Health Sciences (RIHS), Radboudumc, Nijmegen, The Netherlands
| | - Sinno H P Simons
- Department of Pediatrics, Division of Neonatology, Erasmus University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands
| | | |
Collapse
|
187
|
Reducing antibiotic utilization rate in preterm infants: a quality improvement initiative. J Perinatol 2018; 38:421-429. [PMID: 29396511 DOI: 10.1038/s41372-018-0041-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 12/10/2017] [Accepted: 12/21/2017] [Indexed: 02/03/2023]
Abstract
BACKGROUND Judicious use of antibiotic therapy in preterm infants is necessary as prolonged and unwarranted use of antibiotics have been associated with adverse short-term and long-term outcomes. LOCAL PROBLEM Our baseline data review revealed overuse and unnecessary prolonged antibiotic exposure among preterm infants despite a low suspicion for sepsis. METHODS AND INTERVENTIONS The baseline overall AUR was calculated retrospectively from our pharmacy database for a period of 4 months prior to the quality improvement (QI) initiative (pre-QI phase). The principal QI intervention included the development and implementation of guidance algorithms for evaluation and management of suspected sepsis incorporating key QI measures, such as an emphasis on early discontinuation of antibiotics by 36 h if blood culture remained negative and the introduction of multiplex polymerase chain reaction assay for early identification of causative organisms. This QI initiative was implemented through multiple Plan-Do-Study-Act cycles, starting in February 2016 (QI phase), with an objective to achieve a 10% reduction in the baseline overall AUR by December 2016, in preterm infants with gestational ages between 250/7 and 336/7 weeks. Data for the QI phase of the study were collected prospectively. RESULT The overall AUR (outcome measure) decreased from 154.8 to 138.4 days of treatment per 1000 hospital days (10.6% decrease, p < 0.05) over the 11-month period. However, the overall rate of adherence to guidance algorithm (process measure) remained below the target goal of 90%. CONCLUSION This multiphase QI initiative was able to reduce the overall AUR at our NICU. The beneficial impact of this decrease in AUR in preterm infants remains to be determined.
Collapse
|
188
|
|
189
|
Esaiassen E, Hjerde E, Cavanagh JP, Pedersen T, Andresen JH, Rettedal SI, Støen R, Nakstad B, Willassen NP, Klingenberg C. Effects of Probiotic Supplementation on the Gut Microbiota and Antibiotic Resistome Development in Preterm Infants. Front Pediatr 2018; 6:347. [PMID: 30505830 PMCID: PMC6250747 DOI: 10.3389/fped.2018.00347] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/26/2018] [Indexed: 12/15/2022] Open
Abstract
Objectives: In 2014 probiotic supplementation (Lactobacillus acidophilus and Bifidobacterium longum subspecies infantis; InfloranⓇ) was introduced as standard of care to prevent necrotizing enterocolitis (NEC) in extremely preterm infants in Norway. We aimed to evaluate the influence of probiotics and antibiotic therapy on the developing gut microbiota and antibiotic resistome in extremely preterm infants, and to compare with very preterm infants and term infants not given probiotics. Study design: A prospective, observational multicenter study in six tertiary-care neonatal units. We enrolled 76 infants; 31 probiotic-supplemented extremely preterm infants <28 weeks gestation, 35 very preterm infants 28-31 weeks gestation not given probiotics and 10 healthy full-term control infants. Taxonomic composition and collection of antibiotic resistance genes (resistome) in fecal samples, collected at 7 and 28 days and 4 months age, were analyzed using shotgun-metagenome sequencing. Results: Median (IQR) birth weight was 835 (680-945) g and 1,290 (1,150-1,445) g in preterm infants exposed and not exposed to probiotics, respectively. Two extremely preterm infants receiving probiotic developed NEC requiring surgery. At 7 days of age we found higher median relative abundance of Bifidobacterium in probiotic supplemented infants (64.7%) compared to non-supplemented preterm infants (0.0%) and term control infants (43.9%). Lactobacillus was only detected in small amounts in all groups, but the relative abundance increased up to 4 months. Extremely preterm infants receiving probiotics had also much higher antibiotic exposure, still overall microbial diversity and resistome was not different than in more mature infants at 4 weeks and 4 months. Conclusion: Probiotic supplementation may induce colonization resistance and alleviate harmful effects of antibiotics on the gut microbiota and antibiotic resistome. Clinical Trial Registration: Clinicaltrials.gov: NCT02197468. https://clinicaltrials.gov/ct2/show/NCT02197468.
Collapse
Affiliation(s)
- Eirin Esaiassen
- Paediatric Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| | - Erik Hjerde
- Department of Chemistry, Norstruct, UiT The Arctic University of Norway, Tromsø, Norway
| | - Jorunn Pauline Cavanagh
- Paediatric Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| | - Tanja Pedersen
- Department of Paediatrics, Haukeland University Hospital, Bergen, Norway
| | - Jannicke H Andresen
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway
| | - Siren I Rettedal
- Department of Paediatrics, Stavanger University Hospital, Stavanger, Norway
| | - Ragnhild Støen
- Department of Paediatrics, St. Olavs University Hospital, Trondheim, Norway.,Department of Laboratory Medicine, Children's and Women's Health, University of Science and Technology, Trondheim, Norway
| | - Britt Nakstad
- Department of Paediatric and Adolescents Medicine, Akershus University Hospital, Nordbyhagen, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nils P Willassen
- Department of Chemistry, Norstruct, UiT The Arctic University of Norway, Tromsø, Norway
| | - Claus Klingenberg
- Paediatric Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
190
|
Abstract
Invasive fungal infections are a significant cause of morbidity and mortality in infants and children. Early diagnosis is critical, and treatment with the appropriate drug and dose should be initiated promptly. Although an increasing number of studies have examined dosing of antifungals in this population, pediatric safety and efficacy data are lacking.
Collapse
Affiliation(s)
- Mihai Puia-Dumitrescu
- Department of Pediatrics, Division of Neonatal Medicine, Duke University Medical Center, 2424 Erwin Road, Suite 504, Durham, NC 27705, USA; Department of Pediatrics, Duke Clinical Research Institute, P.O. Box 17969, Durham, NC 27715, USA
| | - P Brian Smith
- Department of Pediatrics, Division of Neonatal Medicine, Duke University Medical Center, 2424 Erwin Road, Suite 504, Durham, NC 27705, USA; Department of Pediatrics, Duke Clinical Research Institute, P.O. Box 17969, Durham, NC 27715, USA.
| |
Collapse
|
191
|
Hwang MF, Beechinor RJ, Wade KC, Benjamin DK, Smith PB, Hornik CP, Capparelli EV, Duara S, Kennedy KA, Cohen-Wolkowiez M, Gonzalez D. External Evaluation of Two Fluconazole Infant Population Pharmacokinetic Models. Antimicrob Agents Chemother 2017; 61:e01352-17. [PMID: 28893774 PMCID: PMC5700313 DOI: 10.1128/aac.01352-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 09/04/2017] [Indexed: 11/20/2022] Open
Abstract
Fluconazole is an antifungal agent used for the treatment of invasive candidiasis, a leading cause of morbidity and mortality in premature infants. Population pharmacokinetic (PK) models of fluconazole in infants have been previously published by Wade et al. (Antimicrob Agents Chemother 52:4043-4049, 2008, https://doi.org/10.1128/AAC.00569-08) and Momper et al. (Antimicrob Agents Chemother 60:5539-5545, 2016, https://doi.org/10.1128/AAC.00963-16). Here we report the results of the first external evaluation of the predictive performance of both models. We used patient-level data from both studies to externally evaluate both PK models. The predictive performance of each model was evaluated using the model prediction error (PE), mean prediction error (MPE), mean absolute prediction error (MAPE), prediction-corrected visual predictive check (pcVPC), and normalized prediction distribution errors (NPDE). The values of the parameters of each model were reestimated using both the external and merged data sets. When evaluated with the external data set, the model proposed by Wade et al. showed lower median PE, MPE, and MAPE (0.429 μg/ml, 41.9%, and 57.6%, respectively) than the model proposed by Momper et al. (2.45 μg/ml, 188%, and 195%, respectively). The values of the majority of reestimated parameters were within 20% of their respective original parameter values for all model evaluations. Our analysis determined that though both models are robust, the model proposed by Wade et al. had greater accuracy and precision than the model proposed by Momper et al., likely because it was derived from a patient population with a wider age range. This study highlights the importance of the external evaluation of infant population PK models.
Collapse
Affiliation(s)
- Michael F Hwang
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ryan J Beechinor
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kelly C Wade
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Daniel K Benjamin
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - P Brian Smith
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Christoph P Hornik
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Edmund V Capparelli
- University of California, San Diego, Department of Pediatrics and Skaggs School of Pharmacy, La Jolla, California, USA
| | - Shahnaz Duara
- University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kathleen A Kennedy
- University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Michael Cohen-Wolkowiez
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
- Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
192
|
Abstract
The spread of antibiotic resistance due to the use and misuse of antibiotics around the world is now a major health crisis. Neonates are exposed to antibiotics both before and after birth, often empirically because of risk factors for infection, or for non-specific signs which may or may not indicate sepsis. There is increasing evidence that, apart from antibiotic resistance, the use of antibiotics in pregnancy and in the neonatal period alters the microbiome in the fetus and neonate with an increased risk of immediate and long-term adverse effects. Antibiotic stewardship is a co-ordinated program that promotes the appropriate use of antibiotics, improves patient outcomes, reduces microbial resistance, and decreases the spread of infections caused by multidrug-resistant organisms. This review addresses some of the controversies in antibiotic use in the perinatal period, examines opportunities for reduction of unnecessary antibiotic exposure in neonates, and provides a framework for antibiotic stewardship in neonatal care.
Collapse
Affiliation(s)
- Jayashree Ramasethu
- Division of Neonatal Perinatal Medicine, MedStar Georgetown University Hospital, Washington DC, USA.
| | - Tetsuya Kawakita
- Department of Obstetrics and Gynecology, MedStar Washington Hospital Center, Washington DC, USA
| |
Collapse
|
193
|
Rivera-Chaparro ND, Cohen-Wolkowiez M, Greenberg RG. Dosing antibiotics in neonates: review of the pharmacokinetic data. Future Microbiol 2017; 12:1001-1016. [PMID: 28758800 PMCID: PMC5627030 DOI: 10.2217/fmb-2017-0058] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 05/23/2017] [Indexed: 12/20/2022] Open
Abstract
Antibiotics are often used in neonates despite the absence of relevant dosing information in drug labels. For neonatal dosing, clinicians must extrapolate data from studies for adults and older children, who have strikingly different physiologies. As a result, dosing extrapolation can lead to increased toxicity or efficacy failures in neonates. Driven by these differences and recent legislation mandating the study of drugs in children and neonates, an increasing number of pharmacokinetic studies of antibiotics are being performed in neonates. These studies have led to new dosing recommendations with particular consideration for neonate body size and maturation. Herein, we highlight the available pharmacokinetic data for commonly used systemic antibiotics in neonates.
Collapse
Affiliation(s)
- Nazario D Rivera-Chaparro
- Duke Clinical Research Institute, 2400 Pratt Street, Durham, NC 27705, USA
- Department of Pediatrics, Duke University, Durham, NC 27710, USA
| | - Michael Cohen-Wolkowiez
- Duke Clinical Research Institute, 2400 Pratt Street, Durham, NC 27705, USA
- Department of Pediatrics, Duke University, Durham, NC 27710, USA
| | - Rachel G Greenberg
- Duke Clinical Research Institute, 2400 Pratt Street, Durham, NC 27705, USA
- Department of Pediatrics, Duke University, Durham, NC 27710, USA
| |
Collapse
|
194
|
Salerno S, Hornik CP, Cohen-Wolkowiez M, Smith PB, Ku LC, Kelly MS, Clark R, Gonzalez D. Use of Population Pharmacokinetics and Electronic Health Records to Assess Piperacillin-Tazobactam Safety in Infants. Pediatr Infect Dis J 2017; 36:855-859. [PMID: 28410277 PMCID: PMC5555808 DOI: 10.1097/inf.0000000000001610] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Piperacillin, in combination with tazobactam, is frequently used in infants for treating nosocomial infections, although safety data in this population are limited. Electronic health record (EHR) data can be used to evaluate drug safety in infants, but measures of drug exposure are lacking. METHODS To relate simulated piperacillin exposure with adverse events (AEs) in infants using EHR data, we identified infants discharged from 333 neonatal intensive care units managed by the Pediatrix Medical Group between 1997 and 2012. Using a previously published population pharmacokinetic model in the target population, we simulated piperacillin steady state area under the concentration versus time curve from zero to τ (AUCss,0-τ) and steady state maximal drug concentration (Cmaxss). Next, we used multivariable logistic regression to evaluate the association between simulated AUCss,0-τ and Cmaxss with clinical AEs (seizure and rash) and laboratory AEs controlling for gestational age. The odds ratios (95% confidence intervals) comparing the third versus the first tertiles for AUCss,0-τ and Cmaxss were reported. RESULTS We identified 746 infants with a median (interquartile range) gestational age of 30 weeks (26-33) and postnatal age of 11 days (6-25). The median (interquartile range) piperacillin dose was 225 mg/kg/d (176-300). No significant associations were found between simulated piperacillin exposure (AUCss,0-τ and Cmaxss) and clinical and laboratory AEs. CONCLUSIONS We found no associations between predicted piperacillin exposures and the occurrence of AEs. This study confirms the feasibility of using population pharmacokinetics and EHR to relate drug exposure with safety.
Collapse
Affiliation(s)
- Sara Salerno
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christoph P. Hornik
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, USA
| | - Michael Cohen-Wolkowiez
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, USA
| | - P. Brian Smith
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC, USA
| | - Lawrence C. Ku
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Matthew S. Kelly
- Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Reese Clark
- Pediatrix Medical Group, Inc., Sunrise, FL, USA
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
195
|
Greenberg RG, Wu H, Laughon M, Capparelli E, Rowe S, Zimmerman KO, Smith PB, Cohen-Wolkowiez M. Population Pharmacokinetics of Dexmedetomidine in Infants. J Clin Pharmacol 2017; 57:1174-1182. [PMID: 28444697 PMCID: PMC5561462 DOI: 10.1002/jcph.904] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/02/2017] [Indexed: 12/31/2022]
Abstract
Despite limited pharmacokinetic (PK) data, dexmedetomidine is increasingly being used off-label for sedation in infants. We aimed to characterize the developmental PK changes of dexmedetomidine during infancy. In this open-label, single-center PK study of dexmedetomidine in infants receiving dexmedetomidine per clinical care, ≤10 blood samples per infant were collected. A set of structural PK models and residual error models were explored using nonlinear mixed-effects modeling in NONMEM. Covariates including postmenstrual age (PMA), serum creatinine, and recent history of cardiac surgery requiring cardiopulmonary bypass were investigated for their influence on PK parameters. Univariable generalized estimating equation models were used to evaluate the association of hypotension with dexmedetomidine concentrations. A total of 89 PK samples were collected from 20 infants with a median PMA of 44 weeks (range, 33-61). The median maximum dexmedetomidine infusion dose during the study period was 1.8 μg/(kg·h) (0.5-2.5), and 16/20 infants had a maximum dose >1 μg/(kg·h). A 1-compartment model best described the data. Younger PMA was a significant predictor of lower clearance. Infants with a history of cardiac surgery had ∼40% lower clearance compared to those without a history of cardiac surgery. For infants with PMA of 33 to 61 weeks and body weight of 2 to 6 kg, the estimated clearance and volume of distribution were 0.87 to 2.65 L/(kg·h) and 1.5 L/kg, respectively. No significant associations were found between dexmedetomidine concentrations and hypotension. Infants with younger PMA and recent cardiac surgery may require relatively lower doses of dexmedetomidine to achieve exposure similar to older patients and those without cardiac surgery.
Collapse
Affiliation(s)
- Rachel G. Greenberg
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Huali Wu
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Matthew Laughon
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Edmund Capparelli
- Department of Pediatrics and Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California, San Diego, CA, USA
| | - Stevie Rowe
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Kanecia O. Zimmerman
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - P. Brian Smith
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Michael Cohen-Wolkowiez
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
196
|
Peeples ES. An evaluation of hydrocortisone dosing for neonatal refractory hypotension. J Perinatol 2017; 37:943-946. [PMID: 28518133 DOI: 10.1038/jp.2017.68] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/28/2017] [Accepted: 04/12/2017] [Indexed: 11/09/2022]
Abstract
OBJECTIVE The objective of the study was to compare blood pressure, vasoactive medication requirements and adverse outcomes after administration of high- versus low-dose hydrocortisone (HC) in preterm infants. STUDY DESIGN This is a retrospective analysis of 106 infants ⩽28 weeks gestational age with hypotension requiring vasoactive infusions and high-dose (4 mg kg-1 per day, n=50), low-dose (1 to 3 mg kg-1 per day, n=20) or no HC (n=36) from 2011 to 2015. Groups were compared by two-tailed t-test or χ2, and correlation estimated by multivariable logistic regression. RESULTS There were no differences in measured efficacy between the low- and high-dose groups. Infants with pre-treatment cortisol >15 mcg dl-1 who received HC therapy showed less improvement in vasoactive burden, increased hyperglycemia (P=0.015) and increased death independent of HC dose (odds ratio 26.3, 3.5 to 198.3, P=0.002). CONCLUSION These results support using the lowest effective HC dose in preterm infants. In addition, HC therapy should likely be avoided in infants who are not cortisol deficient.
Collapse
Affiliation(s)
- E S Peeples
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
197
|
Buckley LA, Salunke S, Thompson K, Baer G, Fegley D, Turner MA. Challenges and strategies to facilitate formulation development of pediatric drug products: Safety qualification of excipients. Int J Pharm 2017; 536:563-569. [PMID: 28729174 DOI: 10.1016/j.ijpharm.2017.07.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/10/2017] [Accepted: 07/12/2017] [Indexed: 11/30/2022]
Abstract
A public workshop entitled "Challenges and strategies to facilitate formulation development of pediatric drug products" focused on current status and gaps as well as recommendations for risk-based strategies to support the development of pediatric age-appropriate drug products. Representatives from industry, academia, and regulatory agencies discussed the issues within plenary, panel, and case-study breakout sessions. By enabling practical and meaningful discussion between scientists representing the diversity of involved disciplines (formulators, nonclinical scientists, clinicians, and regulators) and geographies (eg, US, EU), the Excipients Safety workshop session was successful in providing specific and key recommendations for defining paths forward. Leveraging orthogonal sources of data (eg. food industry, agro science), collaborative data sharing, and increased awareness of the existing sources such as the Safety and Toxicity of Excipients for Paediatrics (STEP) database will be important to address the gap in excipients knowledge needed for risk assessment. The importance of defining risk-based approaches to safety assessments for excipients vital to pediatric formulations was emphasized, as was the need for meaningful stakeholder (eg, patient, caregiver) engagement.
Collapse
Affiliation(s)
- Lorrene A Buckley
- Toxicology and Drug Disposition, Eli Lilly & Co., Inc, Indianapolis, IN 46285, USA.
| | - Smita Salunke
- European Paediatric Formulations Initiative (EuPFI), Dept of Pharmaceutics and Centre for Paediatric Pharmacy Research, UCL School of Pharmacy, London, UK
| | - Karen Thompson
- Oral Formulation Science & Technology, MRL, West Point, PA 19486, USA
| | - Gerri Baer
- US Food and Drug Administration, OC/OSMP/OPT, Silver Spring, MD 20993, USA
| | - Darren Fegley
- US Food and Drug Administration, CDER/OND/ODEI/DPP, Silver Spring, MD 20993, USA
| | - Mark A Turner
- Institution of Translational Medicine, University of Liverpool, Liverpool, L8 7SS, UK
| |
Collapse
|
198
|
Abstract
Hospitals are challenged to implement measures to improve health outcomes, decrease costly interventions, and increase patient satisfaction. By following a nurse-driven protocol, our institution has successfully met these three challenges in our treatment of newborns diagnosed with neonatal hypoglycemia (NH). Based on results of a randomized clinical trial, a multidisciplinary team trialed glucose gel as a standard treatment for NH. During the first year, admission rates to the NICU for NH decreased by 73 percent. Exclusive breastfeeding rates for this population increased to 49 percent and 40 additional families remained together on the mother baby unit. This practice change is improving health outcomes, decreasing expensive interventions, and increasing satisfaction among the population of infants at risk for NH.
Collapse
|
199
|
Ravisankar S, Kuehn D, Clark RH, Greenberg RG, Smith PB, Hornik CP. Antihypertensive drug exposure in premature infants from 1997 to 2013. Cardiol Young 2017; 27:905-911. [PMID: 27748228 PMCID: PMC5393975 DOI: 10.1017/s1047951116001591] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Systemic hypertension is increasingly recognised in premature infants. There is limited evidence regarding treatment, and most published treatment recommendations are based solely on expert opinions. METHODS We identified all infants born ⩽32 weeks of gestation and ⩽1500 g birth weight discharged from one of 348 neonatal ICUs managed by the Pediatrix Medical Group between 1997 and 2013. We defined antihypertensive drugs as vasodilators, angiotensin-converting enzyme inhibitors, β receptor blockers, calcium channel blockers, and central α2 receptor agonists. We compared characteristics between infants who were treated with at least one antihypertensive drug during their initial hospitalisation and infants who were not prescribed antihypertensive drugs using Wilcoxon's ranked sum test or Pearson's χ2-test. RESULTS We identified 2504/119,360 (2.1%) infants who required at least one antihypertensive drug. The median postnatal age of first exposure was 48 days (25th, 75th percentile 15, 86), and the median length of therapy was 6 days (1, 16). Hydralazine was the most commonly prescribed antihypertensive with 1280/2504 (51.1%) treated infants exposed to the drug. More than two antihypertensive drugs were administered in 582/2504 (23.2%) infants, and 199/2097 (9.5%) of the treated infants were discharged home on antihypertensive therapy. Infants who received antihypertensive drugs were of lower gestational age (p<0.001) and birth weight (p<0.001) compared with infants not prescribed antihypertensive drugs. CONCLUSIONS Our study is the largest to describe current antihypertensive drug exposure in a cohort of exclusively premature infants born ⩽32 weeks of gestation. We found wide variations in practice for treating hypertension in premature infants.
Collapse
Affiliation(s)
- Srikanth Ravisankar
- Department of Pediatrics, East Carolina University, Greenville, North Carolina, USA
| | - Devon Kuehn
- Department of Pediatrics, East Carolina University, Greenville, North Carolina, USA
| | - Reese H. Clark
- Pediatrix Medical Group, Greenville, Sunrise, Florida, USA
| | | | - P. Brian Smith
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | | |
Collapse
|
200
|
Avant D, Baer G, Moore J, Zheng P, Sorbello A, Ariagno R, Yao L, Burckart GJ, Wang J. Neonatal Safety Information Reported to the FDA During Drug Development Studies. Ther Innov Regul Sci 2017; 2017:1-9. [PMID: 28804696 DOI: 10.1177/2168479017716713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Relatively few neonatal drug development studies have been conducted, but an increase is expected with the enactment of the Food and Drug Administration Safety and Innovation Act (FDASIA). Understanding the safety of drugs studied in neonates is complicated by the unique nature of the population and the level of illness. The objective of this study was to examine neonatal safety data submitted to the FDA in studies pursuant to the Best Pharmaceuticals for Children Act (BPCA) and the Pediatric Research Equity Act (PREA) between 1998 and 2015. METHODS FDA databases were searched for BPCA and/or PREA studies that enrolled neonates. Studies that enrolled a minimum of 3 neonates were analyzed for the presence and content of neonatal safety data. RESULTS The analysis identified 40 drugs that were studied in 3 or more neonates. Of the 40 drugs, 36 drugs received a pediatric labeling change as a result of studies between 1998 and 2015, that included information from studies including neonates. Fourteen drugs were approved for use in neonates. Clinical trials for 20 of the drugs reported serious adverse events (SAEs) in neonates. The SAEs primarily involved cardiovascular events such as bradycardia and/or hypotension or laboratory abnormalities such as anemia, neutropenia, and electrolyte disturbances. Deaths were reported during studies of 9 drugs. CONCLUSIONS Our analysis revealed that SAEs were reported in studies involving 20 of the 40 drugs evaluated in neonates, with deaths identified in 9 of those studies. Patients enrolled in studies were often critically ill, which complicated determination of whether an adverse event was drug-related. We conclude that the traditional means for collecting safety information in drug development trials needs to be adjusted for neonates and will require the collaboration of regulators, industry, and the clinical and research communities to establish appropriate definitions and reporting strategies for the neonatal population.
Collapse
Affiliation(s)
- Debbie Avant
- Office of Pediatric Therapeutics, Office of the Commissioner, US Food and Drug Administration, Silver Spring, MD, USA
| | - Gerri Baer
- Office of Pediatric Therapeutics, Office of the Commissioner, US Food and Drug Administration, Silver Spring, MD, USA
| | - Jason Moore
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD, USA.,Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | - Panli Zheng
- University of North Carolina School of Pharmacy, Chapel Hill, NC, USA
| | - Alfred Sorbello
- Office of Translational Sciences, US Food and Drug Administration, Silver Spring, MD, USA
| | - Ron Ariagno
- Office of Pediatric Therapeutics, Office of the Commissioner, US Food and Drug Administration, Silver Spring, MD, USA.,Stanford University School of Medicine, Stanford, CA, USA
| | - Lynne Yao
- Division of Pediatric and Maternal Health, Office of New Drugs, CDER, US Food and Drug Administration, Silver Spring, MD, USA
| | - Gilbert J Burckart
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research (CDER), US Food and Drug Administration, Silver Spring, MD, USA
| | - Jian Wang
- Office of Drug Evaluation IV, CDER, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|