1
|
Hailu WB, Yimer G, Deyno S, Bacha LT. Time to response and predictors of seizure response to phenobarbitone therapy among neonates admitted with hypoxic-ischemic encephalopathy at Nekemte Comprehensive Specialized Hospital, Ethiopia. J Clin Neurosci 2025; 134:111104. [PMID: 39914182 DOI: 10.1016/j.jocn.2025.111104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 03/12/2025]
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy is a brain injury that occurs in newborns when there is not enough blood flow to the brain. Recent studies have raised concerns about how well phenobarbitone works for treating seizures in newborns, as it may not effectively control seizures with the initial loading and repeated doses. OBJECTIVE This study evaluated the time to respond and predictors of seizure response to phenobarbitone therapy among neonates admitted with hypoxic-ischemic encephalopathy. METHODS AND MATERIALS A retrospective cohort study was conducted at Nekemte Comprehensive Specialized Hospital, using randomly selected medical records of 284 neonates who were treated between January 2020 and December 31, 2023. The study included neonates diagnosed with perinatal asphyxia stage II and III hypoxic-ischemic encephalopathy, who were treated with nasogastric phenobarbitone. The treatment included an initial loading dose of 20 mg/kg and two repeated doses of 10 mg/kg. Survival analysis was conducted. Predictor variables with a p-value ≤ 0.25 in bivariate Cox regression were included in the multivariable Cox regression analysis. Adjusted Hazard Ratios with 95 % confidence intervals were computed, and a p-value < 0.05 was considered statistically significant. RESULTS Out of the 284 neonates, 210 (73.9 %) responded to the phenobarbitone treatment. The incidence rate of response was 27.73 per 1000 person-hours of observation, with a median time to response of 29 h (IQR 26.5-32 h). Low birth weight (AHR = 0.59; 95 %CI 0.58, 0.98), subtle seizure type (AHR: 2.35; 95 % CI 1.09, 5.08), severe hypothermia (AHR = 0.23; 95 % CI 0.052, 0.26), and seizure frequency of twice or more (AHR = 0.436, 95 % CI 0.31, 0.61) were identified as predictors of seizure response. CONCLUSION AND RECOMMENDATIONS The overall incidence rate of response was low. Having a history of twice or more frequency of seizure insult, severe hypothermia and low birth weight decreased the response rate while subtle types of seizure increased the likelihood of response to phenobarbitone therapy. Electroencephalogram-confirmed seizure treatment and combined management with therapeutic hypothermia for high-risk newborns need to be started for better response and reduced response time. Further controlled studies utilizing both clinical and neuroimaging for definitive outcome measurement are recommended.
Collapse
Affiliation(s)
- Wase Benti Hailu
- Department of Public Health Institute of Health Sciences Wollega University Ethiopia.
| | - Getnet Yimer
- Associate Professor of Pharmacology at College of Medicine and Health Science Addis Ababa University Ethiopia
| | - Serawit Deyno
- Postdoctoral Fellow at CDT Africa Addis Ababa University Ethiopia
| | - Lensa Tamiru Bacha
- Paediatrician at Nekemte Comprehensive Specialized Hospital Oromia Regional State Nekemte Ethiopia
| |
Collapse
|
2
|
ELMeneza S, Agaba N, Fawaz RAES, Abd Elgawad SS. Review of Precision Medicine and Diagnosis of Neonatal Illness. Diagnostics (Basel) 2025; 15:478. [PMID: 40002629 PMCID: PMC11854428 DOI: 10.3390/diagnostics15040478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Precision medicine is a state-of-the-art medicine tactic that tailors information about people's genes, environment, and lifestyle to aid the prevention, diagnosis, and treatment of various diseases to provide an overview of the currently available knowledge and applicability of precision medicine in the diagnosis of different cases admitted to the NICU, such as encephalopathies, respiratory distress syndrome of prematurity, hemodynamic instability, acute kidney injury, sepsis, and hyperbilirubinemia. Methods: The authors searched databases, such as PubMed and PubMed Central, for the terms neonatal "precision medicine", "personalized medicine", "genomics", and "metabolomics", all related to precision medicine in the diagnosis of neonatal illness. The related studies were collected. Results: The review highlights the diagnostic approach that serves to implement precision medicine in the NICU and provide precision diagnosis, monitoring, and treatment. Conclusions: In this review, we projected several diagnostic approaches that provide precision identification of health problems among sick neonates with complex illnesses in the NICU; some are noninvasive and available in ordinary healthcare settings, while others are invasive or not feasible or still in ongoing research as machine learning algorithms. Future studies are needed for the wide implementation of artificial intelligence tools in the diagnosis of neonatal illnesses.
Collapse
Affiliation(s)
- Safaa ELMeneza
- Pediatrics Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo 11651, Egypt; (N.A.); (R.A.E.S.F.); (S.S.A.E.)
| | | | | | | |
Collapse
|
3
|
Bersani I, Lapergola G, Patacchiola R, D'Adamo E, Stuppia L, de Laurenzi V, Damiani V, Cataldo I, Clemente K, Primavera A, Salomone R, Barbante E, Campi F, Savarese I, Ronci S, Dotta A, Braguglia A, Longo D, Gavilanes DAW, Gazzolo F, Serpero L, Strozzi MC, Maconi A, Cassinari M, Libener R, Gazzolo D. Elevated S100B urine levels predict seizures in infants complicated by perinatal asphyxia and undergoing therapeutic hypothermia. Clin Chem Lab Med 2024; 62:1109-1117. [PMID: 38290722 DOI: 10.1515/cclm-2023-1471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024]
Abstract
OBJECTIVES Seizures (SZ) are one of the main complications occurring in infants undergoing therapeutic hypothermia (TH) due to perinatal asphyxia (PA) and hypoxic ischemic encephalopathy (HIE). Phenobarbital (PB) is the first-line therapeutic strategy, although data on its potential side-effects need elucidation. We investigated whether: i) PB administration in PA-HIE TH-treated infants affects S100B urine levels, and ii) S100B could be a reliable early predictor of SZ. METHODS We performed a prospective case-control study in 88 PA-HIE TH infants, complicated (n=44) or not (n=44) by SZ requiring PB treatment. S100B urine levels were measured at 11 predetermined monitoring time-points from first void up to 96-h from birth. Standard-of-care monitoring parameters were also recorded. RESULTS S100B significantly increased in the first 24-h independently from HIE severity in the cases who later developed SZ and requested PB treatment. ROC curve analysis showed that S100B, as SZ predictor, at a cut-off of 2.78 μg/L achieved a sensitivity/specificity of 63 and 84 %, positive/negative predictive values of 83 and 64 %. CONCLUSIONS The present results offer additional support to the usefulness of S100B as a trustable diagnostic tool in the clinical daily monitoring of therapeutic and pharmacological procedures in infants complicated by PA-HIE.
Collapse
Affiliation(s)
- Iliana Bersani
- Department of Neonatology, Neonatal Intensive Care Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Giuseppe Lapergola
- Neonatal Intensive Care Unit, G. d'Annunzio University of Chieti, Chieti, Italy
| | - Roberta Patacchiola
- Neonatal Intensive Care Unit, G. d'Annunzio University of Chieti, Chieti, Italy
| | - Ebe D'Adamo
- Neonatal Intensive Care Unit, G. d'Annunzio University of Chieti, Chieti, Italy
| | - Liborio Stuppia
- Center for Advances Studies and Technology (CAST), G. d'Annunzio University, Chieti, Italy
| | - Vincenzo de Laurenzi
- Center for Advances Studies and Technology (CAST), G. d'Annunzio University, Chieti, Italy
| | - Verena Damiani
- Center for Advances Studies and Technology (CAST), G. d'Annunzio University, Chieti, Italy
| | - Ivana Cataldo
- Laboratory of Clinical Pathology, SS Annunziata Hospital, Chieti, Italy
| | - Katia Clemente
- Laboratory of Clinical Pathology, SS Annunziata Hospital, Chieti, Italy
| | - Adele Primavera
- Neonatal Intensive Care Unit, G. d'Annunzio University of Chieti, Chieti, Italy
| | - Rita Salomone
- Neonatal Intensive Care Unit, G. d'Annunzio University of Chieti, Chieti, Italy
| | - Elisabetta Barbante
- Neonatal Intensive Care Unit, G. d'Annunzio University of Chieti, Chieti, Italy
| | - Francesca Campi
- Department of Neonatology, Neonatal Intensive Care Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Immacolata Savarese
- Department of Neonatology, Neonatal Intensive Care Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Sara Ronci
- Department of Neonatology, Neonatal Intensive Care Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Andrea Dotta
- Department of Neonatology, Neonatal Intensive Care Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Annabella Braguglia
- Department of Neonatology, Neonatal Intensive Care Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Daniela Longo
- Department of Neonatology, Neonatal Intensive Care Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Danilo A W Gavilanes
- Department of Pediatrics and Neonatology, Maastricht University, Maastricht, The Netherlands
| | | | - Laura Serpero
- Department of Maternal, Fetal and Neonatal Medicine, SS Antonio, Biagio and C. Arrigo Hospital, Alessandria, Italy
| | - Maria Chiara Strozzi
- Department of Pediatrics and Neonatology, Ospedale Cardinal Massaia, Asti, Italy
| | - Antonio Maconi
- Social Security Administration Development and Promotion of Scientific Research Unit, SS Antonio, Biagio and C. Arrigo Hospital, Alessandria, Italy
| | - Maurizio Cassinari
- Department of Clinical Biochemistry, Transfusion and Regeneration Medicine Alessandria Hospital, Alessandria, Italy
| | - Roberta Libener
- Department of Clinical Biochemistry, Transfusion and Regeneration Medicine Alessandria Hospital, Alessandria, Italy
| | - Diego Gazzolo
- Neonatal Intensive Care Unit, G. d'Annunzio University of Chieti, Chieti, Italy
| |
Collapse
|
4
|
Pressler R, Boylan G, Dempsey E, Klotz KA, Krauwinkel W, Will E, Morita D, Floricel F, Elshoff J, van den Anker J. Pharmacokinetics and safety of brivaracetam in neonates with repeated electroencephalographic seizures: A multicenter, open-label, single-arm study. Epilepsia Open 2024; 9:522-533. [PMID: 38049197 PMCID: PMC10984296 DOI: 10.1002/epi4.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023] Open
Abstract
OBJECTIVE To evaluate the pharmacokinetics (PK), safety, and tolerability of brivaracetam (BRV) in neonates with repeated electroencephalographic seizures not controlled with previous antiseizure medications (ASMs). METHODS Phase 2/3, multicenter, open-label, single-arm study (N01349/NCT03325439) in neonates with repeated electroencephalographic seizures (lasting ≥10 s) confirmed by video-electroencephalography, and inadequate seizure control with at least one ASM. A screening period (up to 36 h) was followed by a 48-h evaluation period during which patients received 0.5 mg/kg BRV twice daily (b.i.d) intravenously (IV). Patients who benefitted from BRV (investigator's opinion) could continue 0.5 mg/kg b.i.d (IV or oral solution) in an extension period. Outcomes included plasma concentrations of BRV following the first dose (primary), and incidence of treatment-emergent adverse events (TEAEs). RESULTS Six patients (median [range] postnatal age: 1.5 [1.0, 6.0] days) received ≥1 dose of BRV. All six patients completed the evaluation period; two entered and completed the extension period. Overall (evaluation and extension periods), three patients received one dose of 0.5 mg/kg BRV and three received more than one dose. The median (range) duration of exposure to BRV (IV and oral solution) was 1.5 (1.0, 29.0) days (n = 6). At 0.5-1, 2-4, and 8-12 h following IV BRV administration, the GeoMean (GeoCV) plasma concentrations of BRV were 0.53 mg/L (15.40% [n = 5]), 0.50 mg/L (28.20% [n = 6]), and 0.34 mg/L (13.20% [n = 5]), respectively. Individual and population BRV PK profiles were estimated, and individual PK parameters were calculated using Bayesian feedback. The observed concentrations were consistent with the predicted PK. Three patients experienced four TEAEs, none of which were considered related to BRV. SIGNIFICANCE BRV plasma concentrations in neonates were consistent with data in older children receiving BRV oral solution, and with data from adults receiving a nominal IV dose of 25 mg b.i.d. BRV was well tolerated, with no drug-related TEAEs reported. PLAIN LANGUAGE SUMMARY Few drugs are available to treat seizures in newborn babies. Brivaracetam is approved to treat focal-onset seizures in children and adults in Europe (patients 2 years of age and older) and the United States (patients 1 month of age or older). In this study, six newborns with repeated seizures were treated with intravenous brivaracetam. The study doctors took samples of blood from the newborns and measured the levels of brivaracetam. The concentrations of brivaracetam in the newborns' blood plasma were consistent with data from studies in older children and in adults. No brivaracetam-related medical problems were reported.
Collapse
Affiliation(s)
- Ronit Pressler
- Department of Clinical Neurophysiology, Great Ormond Street Hospital and Clinical NeuroscienceUCL‐GOS Institute of Child HealthLondonUK
| | - Geraldine Boylan
- INFANT Research Centre and Department of Paediatrics and Child HealthCorkIreland
| | - Eugene Dempsey
- INFANT Research Centre and Department of Paediatrics and Child HealthCorkIreland
| | - Kerstin Alexandra Klotz
- Department of Neuropediatrics and Muscle Disorders, Medical CenterUniversity of FreiburgFreiburgGermany
| | | | | | | | | | | | | |
Collapse
|
5
|
Bättig L, Dünner C, Cserpan D, Rüegger A, Hagmann C, Schmitt B, Pisani F, Ramantani G. Levetiracetam versus Phenobarbital for Neonatal Seizures: A Retrospective Cohort Study. Pediatr Neurol 2023; 138:62-70. [PMID: 36401982 DOI: 10.1016/j.pediatrneurol.2022.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/22/2022] [Accepted: 10/15/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Although phenobarbital (PB) is commonly used as a first-line antiseizure medication (ASM) for neonatal seizures, in 2015 we chose to replace it with levetiracetam (LEV), a third-generation ASM. Here, we compared the safety and efficacy of LEV and PB as first-line ASM, considering the years before and after modifying our treatment protocol. METHODS We conducted a retrospective cohort study of 108 neonates with electroencephalography (EEG)-confirmed seizures treated with first-line LEV or PB in 2012 to 2020. RESULTS First-line ASM was LEV in 33 (31%) and PB in 75 (69%) neonates. The etiology included acute symptomatic seizures in 69% of cases (30% hypoxic-ischemic encephalopathy, 32% structural vascular, 6% infectious, otherwise metabolic) and neonatal epilepsy in 22% (5% structural due to brain malformation, 17% genetic). Forty-two of 108 (39%) neonates reached seizure freedom following first-line therapy. Treatment response did not vary by first-line ASM among all neonates, those with acute symptomatic seizures, or those with neonatal-onset epilepsy. Treatment response was lowest for neonates with a higher seizure frequency, particularly for those with status epilepticus versus rare seizures (P < 0.001), irrespective of gestational age, etiology, or EEG findings. Adverse events were noted in 22 neonates treated with PB and in only one treated with LEV (P < 0.001). CONCLUSIONS Our study suggests a potential noninferiority and a more acceptable safety profile for LEV, which may thus be a reasonable option as first-line ASM for neonatal seizures in place of PB. Treatment should be initiated as early as possible since higher seizure frequencies predispose to less favorable responses.
Collapse
Affiliation(s)
- Linda Bättig
- Department of Neuropediatrics, University Children's Hospital Zurich, Switzerland
| | - Corinne Dünner
- Department of Neuropediatrics, University Children's Hospital Zurich, Switzerland
| | - Dorottya Cserpan
- Department of Neuropediatrics, University Children's Hospital Zurich, Switzerland
| | - Andrea Rüegger
- Department of Neuropediatrics, University Children's Hospital Zurich, Switzerland
| | - Cornelia Hagmann
- Department of Neonatology and Pediatric Intensive Care, University Children's Hospital Zurich, Switzerland; Children's Research Center, University Children's Hospital Zurich, Switzerland; University of Zurich, Switzerland
| | - Bernhard Schmitt
- Department of Neuropediatrics, University Children's Hospital Zurich, Switzerland
| | - Francesco Pisani
- Child Neuropsychiatry Unit, Medicine and Surgery Department, University of Parma, Italy
| | - Georgia Ramantani
- Department of Neuropediatrics, University Children's Hospital Zurich, Switzerland; Children's Research Center, University Children's Hospital Zurich, Switzerland; University of Zurich, Switzerland.
| |
Collapse
|
6
|
Bäcke P, Bruschettini M, Sibrecht G, Thernström Blomqvist Y, Olsson E. Pharmacological interventions for pain and sedation management in newborn infants undergoing therapeutic hypothermia. Cochrane Database Syst Rev 2022; 11:CD015023. [PMID: 36354070 PMCID: PMC9647594 DOI: 10.1002/14651858.cd015023.pub2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Newborn infants affected by hypoxic-ischemic encephalopathy (HIE) undergo therapeutic hypothermia. As this treatment seems to be associated with pain, and intensive and invasive care is needed, pharmacological interventions are often used. Moreover, painful procedures in the newborn period can affect pain responses later in life, impair brain development, and possibly have a long-term negative impact on neurodevelopment and quality of life. OBJECTIVES To determine the effects of pharmacological interventions for pain and sedation management in newborn infants undergoing therapeutic hypothermia. Primary outcomes were analgesia and sedation, and all-cause mortality to discharge. SEARCH METHODS We searched CENTRAL, PubMed, CINAHL (Cumulative Index to Nursing and Allied Health Literature), and the trial register ISRCTN in August 2021. We also checked the reference lists of relevant articles to identify additional studies. SELECTION CRITERIA We included randomized controlled trials (RCT), quasi-RCTs and cluster-randomized trials comparing drugs used for the management of pain or sedation, or both, during therapeutic hypothermia: any opioids (e.g. morphine, fentanyl), alpha-2 agonists (e.g. clonidine, dexmedetomidine), N-Methyl-D-aspartate (NMDA) receptor antagonist (e.g. ketamine), other analgesics (e.g. paracetamol), and sedatives (e.g. benzodiazepines such as midazolam) versus another drug, placebo, no intervention, or non-pharmacological interventions. Primary outcomes were analgesia and sedation, and all-cause mortality to discharge. DATA COLLECTION AND ANALYSIS Two review authors independently assessed studies identified by the search strategy for inclusion. We planned to use the GRADE approach to assess the certainty of evidence. We planned to assess the methodological quality of included trials using Cochrane Effective Practice and Organisation of Care Group (EPOC) criteria (assessing randomization, blinding, loss to follow-up, and handling of outcome data). We planned to evaluate treatment effects using a fixed-effect model with risk ratio (RR) for categorical data and mean, standard deviation (SD), and mean difference (MD) for continuous data. MAIN RESULTS: We did not find any completed studies for inclusion. Amongst the four excluded studies, topiramate and atropine were used in two and one trial, respectively; one study used dexmedetomidine and was initially reported in 2019 to be a randomized trial. However, it was an observational study (correction in 2021). We identified one ongoing study comparing dexmedetomidine to morphine. AUTHORS' CONCLUSIONS We found no studies that met our inclusion criteria and hence there is no evidence to recommend or refute the use of pharmacological interventions for pain and sedation management in newborn infants undergoing therapeutic hypothermia.
Collapse
Affiliation(s)
- Pyrola Bäcke
- Neonatal Intensive Care Unit, University Hospital, Uppsala, Sweden
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Lund University, Skåne University Hospital, Lund, Sweden
| | - Greta Sibrecht
- Newborns' Infectious Diseases Department, Poznan University of Medical Sciences, Poznan, Poland
| | - Ylva Thernström Blomqvist
- Neonatal Intensive Care Unit, University Hospital, Uppsala, Sweden
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Emma Olsson
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Faculty of Medicine and Health, School of Health Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
7
|
Palaparthy V, Kumar M, Rebekah G, Thomas N. Comparing the effect of different loading doses of phenobarbitone on serum phenobarbitone levels in babies with neonatal seizures and effect of therapeutic hypothermia on phenobarbitone levels. J Trop Pediatr 2022; 68:6712682. [PMID: 36150145 DOI: 10.1093/tropej/fmac078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
BACKGROUND With current recommendation for phenobarbitone dosing, we have noted that babies are extremely sedated with elevated serum phenobarbitone levels. We postulate that asphyxiated neonates with hypoxic liver injury have impaired drug metabolism and renal injury affects drug elimination, thus elevating serum drug levels. Therapeutic hypothermia (TH) could further affect the drug levels. OBJECTIVE To determine the serum levels of the phenobarbitone in babies receiving different loading doses of phenobarbitone for neonatal seizures and to study the effect of asphyxia and TH on drug levels. DESIGN Prospective observational cohort study. MATERIAL AND METHODS Term neonates with seizures of any cause were given phenobarbitone up to a maximum loading of 40 mg/kg followed by maintenance dose of 5 mg/kg/day. Serum phenobarbitone levels were assessed after 4 h of the initial loading dose and subsequently at 24, 48 and 72 h from the time after maximum loading dose. Babies were divided into three groups Group 1 (HIE + TH-hypoxic ischemic encephalopathy undergoing TH), Group 2 (HIE - TH-hypoxic ischemic encephalopathy without TH) and Group 3 (non-HIE group). RESULTS A total of 47 babies completed the study. Twenty-three (49%) received 20 mg/kg, 14 (30%) received 30 mg/kg and 10 (21%) received 40 mg per kg of phenobarbitone as loading dose. HIE was the major cause of seizures 28 (59%) followed by hypoglycemia 7 (14%), cerebral malformations 4 (8%), inborn errors of metabolism 2 (4%) and hypocalcemia 1 (2%) while the cause of seizures was not known in 6 (13%). Median (IQR) Phenobarbitone levels at 72 h in babies who received 20 mg/kg loading dose of phenobarbitone was 46.72 (44.02-50.49) mcg/ml in HIE + TH group, 40.53 (28.66-65.09) mcg/ml in HIE - TH group and 49 (37-65) mcg/ml in non-HIE group. After a loading dose of 30 mg/kg, phenobarbitone level was 63.76 (59.5-65.94) mcg/ml in HIE + TH group, 42.5 (34.75-48.75) mcg/ml in HIE - TH group and 42.07 (40-49.05) mcg/ml in non-HIE group. After 40 mg/kg loading dose, it was 62.3 (60.2-64.9) mcg/ml in HIE + TH group, 57.0 (49.8-60.2) mcg/ml in HIE - TH group and 48.15 (40.8-50.97) mcg/ml in non-HIE group. In babies who received >20 mg/kg loading dose, 100% of HIE + TH, 80% of HIE - TH and 60% of non-HIE had supratherapeutic levels of phenobarbitone. CONCLUSION At higher loading doses of 30 and 40 mg/kg, steady state concentration of serum phenobarbitone is higher in babies with hypoxic ischemic encephalopathy who underwent TH than in babies with non-HIE causes of seizures. Loading dose beyond 20 mg/kg should be used with close monitoring of serum drug level.
Collapse
Affiliation(s)
- Vinod Palaparthy
- Department of Neonatology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Manish Kumar
- Department of Neonatology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Grace Rebekah
- Department of Neonatology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Niranjan Thomas
- Department of Neonatology, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
8
|
Keene JC, Wainwright M, Morgan LA, Mietzsch U, Musa N, Bozarth XL, Natarajan N. Retrospective Evaluation of First-line Levetiracetam use for Neonatal Seizures after Congenital Heart Defect repair with or without Extracorporeal Membrane Oxygenation. J Pediatr Pharmacol Ther 2022; 27:254-262. [PMID: 35350164 PMCID: PMC8939278 DOI: 10.5863/1551-6776-27.3.254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/17/2021] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Levetiracetam (LEV) efficacy for neonatal seizures is debated. We evaluated LEV as a first line anti-seizure medicine (ASM) in neonates following neonatal congenital heart defect (CHD) repair who did not require extracorporeal membrane oxygenation (ECMO) vs neonates who required ECMO. METHODS A single center retrospective review of neonates with CHD from 2015 to 2020 was conducted. Neonates were included if seizures were present on continuous EEG after CHD repair either on or off ECMO, and they received LEV as a first line ASM. Primary outcomes were seizure resolution with LEV, adverse events and response to subsequent ASM. RESULTS Eighteen total neonates were evaluated, 10 with seizures post-CHD repair who did not require ECMO and 8 who required ECMO. In the non-ECMO cohort, nine of ten were successfully treated with LEV monotherapy with no adverse events. In comparison, the eight neonates who required ECMO had a higher initial seizure burden (1.6% vs 17%, p=0.003), were more likely to have injury on neuroimaging (12.5 vs 75%, p= 0.04), and all neonates required multiple ASMs. Seizure burden did not decrease with LEV, but significantly decreased with phenobarbital and fosphenytoin (14.4% and 10.5%, p = 0.024). CONCLUSIONS Neonates with CHD and seizures on and off ECMO demonstrated divergent seizure characteristics including seizure burden and response to LEV. LEV may reduce neonatal seizure burden after uncomplicated CHD repair. However, in neonates requiring ECMO, multiple ASMs were required. A prospective evaluation of ASM efficacy and safety in this high-risk population is urgently needed.
Collapse
Affiliation(s)
- Jennifer C. Keene
- University of Washington, Department of Neurology, Division of Child Neurology (JK, MW, LM, XB, NN), Seattle, WA
| | - Mark Wainwright
- University of Washington, Department of Neurology, Division of Child Neurology (JK, MW, LM, XB, NN), Seattle, WA
| | - Lindsey A. Morgan
- University of Washington, Department of Neurology, Division of Child Neurology (JK, MW, LM, XB, NN), Seattle, WA
| | | | - Ndidi Musa
- Division of Pediatric Critical Care Medicine (NM), Seattle, WA
| | - Xiuhua L. Bozarth
- University of Washington, Department of Neurology, Division of Child Neurology (JK, MW, LM, XB, NN), Seattle, WA
| | - Niranjana Natarajan
- University of Washington, Department of Neurology, Division of Child Neurology (JK, MW, LM, XB, NN), Seattle, WA
| |
Collapse
|
9
|
Jenkins DD, Moss HG, Brown TR, Yazdani M, Thayyil S, Montaldo P, Vento M, Kuligowski J, Wagner C, Hollis BW, Wiest DB. NAC and Vitamin D Improve CNS and Plasma Oxidative Stress in Neonatal HIE and Are Associated with Favorable Long-Term Outcomes. Antioxidants (Basel) 2021; 10:1344. [PMID: 34572976 PMCID: PMC8466838 DOI: 10.3390/antiox10091344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/11/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
N-acetylcysteine (NAC) and vitamin D provide effective neuroprotection in animal models of severe or inflammation-sensitized hypoxic ischemic encephalopathy (HIE). To translate these FDA-approved drugs to HIE neonates, we conducted an early phase, open-label trial of 10 days of NAC (25, 40 mg/kg q12h) + 1,25(OH)2D (calcitriol 0.05 mg/kg q12h, 0.03 mg/kg q24h), (NVD), for pharmacokinetic (PK) estimates during therapeutic hypothermia and normothermia. We paired PK samples with pharmacodynamic (PD) targets of plasma isoprostanoids, CNS glutathione (GSH) and total creatine (tCr) by serial MRS in basal ganglia (BG) before and after NVD infusion at five days. Infants had moderate (n = 14) or severe HIE (n = 16), funisitis (32%), and vitamin D deficiency (75%). NVD resulted in rapid, dose-responsive increases in CNS GSH and tCr that correlated positively with plasma [NAC], inversely with plasma isofurans, and was greater in infants with lower baseline [GSH] and [tCr], suggesting increases in these PD markers were titrated by neural demand. Hypothermia and normothermia altered NAC PK estimates. NVD was well tolerated. Excluding genetic syndromes (2), prolonged ECMO (2), lost-to-follow-up (1) and SIDS death (1), 24 NVD treated HIE infants have no evidence of cerebral palsy, autism or cognitive delay at 24-48 months. These data confirm that low, safe doses of NVD in HIE neonates decreased oxidative stress in plasma and CNS, improved CNS energetics, and are associated with favorable developmental outcomes at two to four years.
Collapse
Affiliation(s)
- Dorothea D Jenkins
- Division of Neonatology, Department of Pediatrics, Medical University of South Carolina, 10 McClennan Banks Drive, Charleston, SC 29425, USA; (C.W.); (B.W.H.)
| | - Hunter G Moss
- Center for Biomedical Imaging, Department of Radiology, Medical University of South Carolina, Charleston, SC 29425, USA; (H.G.M.); (T.R.B.); (M.Y.)
| | - Truman R Brown
- Center for Biomedical Imaging, Department of Radiology, Medical University of South Carolina, Charleston, SC 29425, USA; (H.G.M.); (T.R.B.); (M.Y.)
| | - Milad Yazdani
- Center for Biomedical Imaging, Department of Radiology, Medical University of South Carolina, Charleston, SC 29425, USA; (H.G.M.); (T.R.B.); (M.Y.)
| | - Sudhin Thayyil
- Centre for Perinatal Neuroscience, Imperial College London, London W12 0HS, UK; (S.T.); (P.M.)
| | - Paolo Montaldo
- Centre for Perinatal Neuroscience, Imperial College London, London W12 0HS, UK; (S.T.); (P.M.)
| | - Maximo Vento
- Neonatal Research Group, Health Research Institute Hospital La Fe, 46026 Valencia, Spain; (M.V.); (J.K.)
| | - Julia Kuligowski
- Neonatal Research Group, Health Research Institute Hospital La Fe, 46026 Valencia, Spain; (M.V.); (J.K.)
| | - Carol Wagner
- Division of Neonatology, Department of Pediatrics, Medical University of South Carolina, 10 McClennan Banks Drive, Charleston, SC 29425, USA; (C.W.); (B.W.H.)
| | - Bruce W Hollis
- Division of Neonatology, Department of Pediatrics, Medical University of South Carolina, 10 McClennan Banks Drive, Charleston, SC 29425, USA; (C.W.); (B.W.H.)
| | - Donald B Wiest
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
10
|
Bäcke P, Bruschettini M, Sibrecht G, Thernström Blomqvist Y, Olsson E. Pharmacological interventions for pain and sedation management in newborn infants undergoing therapeutic hypothermia. Hippokratia 2021. [DOI: 10.1002/14651858.cd015023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Pyrola Bäcke
- Neonatal Intensive Care Unit; University Hospital; Uppsala Sweden
- Department of Women's and Children's Health; Uppsala University; Uppsala Sweden
| | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics; Lund University, Skåne University Hospital; Lund Sweden
- Cochrane Sweden; Lund University, Skåne University Hospital; Lund Sweden
| | - Greta Sibrecht
- Newborns' Infectious Diseases Department; Poznan University of Medical Sciences; Poznan Poland
| | - Ylva Thernström Blomqvist
- Neonatal Intensive Care Unit; University Hospital; Uppsala Sweden
- Department of Women's and Children's Health; Uppsala University; Uppsala Sweden
| | - Emma Olsson
- Department of Pediatrics, Faculty of Medicine and Health; Örebro University; Örebro Sweden
- Faculty of Medicine and Health, School of Health Sciences; Örebro University; Örebro Sweden
| |
Collapse
|
11
|
Ryu S, Jung WJ, Jiao Z, Chae JW, Yun HY. External evaluation of the predictive performance of seven population pharmacokinetic models for phenobarbital in neonates. Br J Clin Pharmacol 2021; 87:3878-3889. [PMID: 33638184 DOI: 10.1111/bcp.14803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 02/09/2021] [Accepted: 02/13/2021] [Indexed: 02/06/2023] Open
Abstract
AIM Several studies have reported population pharmacokinetic models for phenobarbital (PB), but the predictive performance of these models has not been well documented. This study aims to do external evaluation of the predictive performance in published pharmacokinetic models. METHODS Therapeutic drug monitoring data collected in neonates and young infants treated with PB for seizure control was used for external evaluation. A literature review was conducted through PubMed to identify population pharmacokinetic models. Prediction- and simulation-based diagnostics, and Bayesian forecasting were performed for external evaluation. The incorporation of allometric scaling for body size and maturation factors into the published models was also tested for prediction improvement. RESULTS A total of 79 serum concentrations from 28 subjects were included in the external dataset. Seven population pharmacokinetic studies of PB were identified as relevant in the literature search and included for our evaluation. The model by Voller et al showed the best performance concerning prediction-based evaluation. In simulation-based analyses, the normalized prediction distribution error of two models (those of Shellhaas et al and Marsot et al) obeyed a normal distribution. Bayesian forecasting with more than one observation improved predictive capability. Incorporation of both allometric size scaling and maturation function generally enhanced the predictive performance, with improvement as observed in the model of Vucicevic et al. CONCLUSIONS: The predictive performance of published pharmacokinetic models of PB was diverse. Bayesian forecasting and incorporation of both size and maturation factors could improve the predictability of the models for neonates.
Collapse
Affiliation(s)
- Sunae Ryu
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea.,National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju, Republic of Korea
| | - Woo Jin Jung
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jung-Woo Chae
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Hwi-Yeol Yun
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
12
|
Šíma M, Michaličková D, Slanař O. What is the Best Predictor of Phenobarbital Pharmacokinetics to Use for Initial Dosing in Neonates? Pharmaceutics 2021; 13:301. [PMID: 33668911 PMCID: PMC7996486 DOI: 10.3390/pharmaceutics13030301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 11/17/2022] Open
Abstract
Phenobarbital is a first-line treatment of various seizure types in newborns. Dosage individualization maximizing the proportion of patients with drug levels in therapeutic range or sufficient treatment response is still challenging. The aim of this review was to summarize the available evidence on phenobarbital pharmacokinetics in neonates and to identify its possible covariates suitable for individualization of initial drug dosing. Several covariates have been considered: body weight and height, body surface area, gestational and postnatal age, laboratory parameters of renal and hepatic functions, asphyxia, therapeutic hypothermia, extracorporeal membrane oxygenation (ECMO), drug interactions, and genetic polymorphisms. The most frequently studied and well-founded covariate for the estimation of phenobarbital dosing is actual body weight. Loading dose of 15-20 mg/kg followed by a maintenance dose of 3-5 mg/kg/day seems to be accurate. However, the evidence for the other covariates with respect to dosing individualization is not sufficient. Doses at the lower limit of suggested range should be preferred in patients with severe asphyxia, while the upper limit of the range should be targeted in neonates receiving ECMO support.
Collapse
Affiliation(s)
- Martin Šíma
- Department of Pharmacology, First Faculty of Medicine, Charles University, 110 00 Prague, Czech Republic; (D.M.); (O.S.)
- General University Hospital in Prague, 128 00 Prague, Czech Republic
| | - Danica Michaličková
- Department of Pharmacology, First Faculty of Medicine, Charles University, 110 00 Prague, Czech Republic; (D.M.); (O.S.)
- General University Hospital in Prague, 128 00 Prague, Czech Republic
| | - Ondřej Slanař
- Department of Pharmacology, First Faculty of Medicine, Charles University, 110 00 Prague, Czech Republic; (D.M.); (O.S.)
- General University Hospital in Prague, 128 00 Prague, Czech Republic
| |
Collapse
|
13
|
van Hoogdalem MW, McPhail BT, Hahn D, Wexelblatt SL, Akinbi HT, Vinks AA, Mizuno T. Pharmacotherapy of neonatal opioid withdrawal syndrome: a review of pharmacokinetics and pharmacodynamics. Expert Opin Drug Metab Toxicol 2020; 17:87-103. [PMID: 33049155 DOI: 10.1080/17425255.2021.1837112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Neonatal opioid withdrawal syndrome (NOWS) often arises in infants born to mothers who used opioids during pregnancy. Morphine, methadone, and buprenorphine are the most common first-line treatments, whereas clonidine and phenobarbital are generally reserved for adjunctive therapy. These drugs exhibit substantial pharmacokinetic (PK) and pharmacodynamic (PD) variability. Current pharmacological treatments for NOWS are based on institutional protocols and largely rely on empirical treatment of patient symptoms. AREAS COVERED This article reviews the PK/PD of NOWS pharmacotherapies with a focus on the implication of physiological development and maturation. Body size-standardized clearance is consistently low in neonates, except for methadone. This can be ascribed to underdeveloped metabolic and elimination pathways. The effects of pharmacogenetics have been clarified especially for morphine. The PK/PD relationship of medications used in the treatment of NOWS is generally understudied. EXPERT OPINION Providing an appropriate opioid dose in neonates is challenging. Advancements in quantitative pharmacology and PK/PD modeling approaches facilitate identification of key factors driving PK/PD variability and characterization of exposure-response relationships. PK/PD model-informed simulations have been widely employed to define age-appropriate pediatric dosing regimens. The model-informed approach holds promise to aid more rational use of medications in the treatment of NOWS.
Collapse
Affiliation(s)
- Matthijs W van Hoogdalem
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,James L. Winkle College of Pharmacy, University of Cincinnati , Cincinnati, OH, USA
| | - Brooks T McPhail
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,School of Medicine Greenville, University of South Carolina , Greenville, SC, USA
| | - David Hahn
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA
| | - Scott L Wexelblatt
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati , Cincinnati, OH, USA.,Center for Addiction Research, College of Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Henry T Akinbi
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati , Cincinnati, OH, USA.,Center for Addiction Research, College of Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center , Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati , Cincinnati, OH, USA.,Center for Addiction Research, College of Medicine, University of Cincinnati , Cincinnati, OH, USA
| |
Collapse
|
14
|
Pokorná P, Michaličková D, Völler S, Hronová K, Tibboel D, Slanař O, Krekels EH. Severity parameters for asphyxia or hypoxic-ischemic encephalopathy do not explain inter-individual variability in the pharmacokinetics of phenobarbital in newborns treated with therapeutic hypothermia. Minerva Pediatr (Torino) 2020; 74:107-115. [PMID: 33107271 DOI: 10.23736/s2724-5276.20.05740-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The current study uses a population modeling approach to evaluate and quantify the impact of severity of asphyxia and hypoxic-ischemic encephalopathy (HIE) on the pharmacokinetics of phenobarbital in asphyxiated newborns treated with therapeutic hypothermia. METHODS Included newborns received phenobarbital (the TOBY trial protocol). 120 plasma samples were available from 50 newborns, median (IQR) weight 3.3 (2.8-3.5) kg and gestational age 39 (39-40) weeks. NONMEM® version 7.2 was used for the data analysis. Age, body weight, sex, concomitant medications, kidney and liver function markers, as well as severity parameters of asphyxia and HIE were tested as potential covariates of pharmacokinetics of phenobarbital. Severe asphyxia was defined as pH of arterial umbilical cord blood ≤7.1 and Apgar 5 ≤5, and severe HIE was defined as time to normalization of amplitude-integrated electroencephalography (aEEG) >24 h. RESULTS Weight was found to be the only statistically significant covariate for the volume of distribution. At weight of 1 kg volume of distribution was 0.91 L and for every additional kg it increased in 0.91 L. Clearance was 0.00563 L/h. No covariates were statistically significant for the clearance of phenobarbital. CONCLUSIONS Phenobarbital dose adjustments are not indicated in the studied population, irrespective of the severity of asphyxia or HIE.
Collapse
Affiliation(s)
- Pavla Pokorná
- Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital.,st Faculty of Medicine, Charles University, Prague, Czech Republic.,Institute of Pharmacology, General University Hospital.,st Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Swantje Völler
- st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Karolina Hronová
- st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Dick Tibboel
- Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital.,Institute of Pharmacology, General University Hospital
| | - Ondřej Slanař
- st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Elke H Krekels
- st Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
15
|
De Rose DU, Cairoli S, Dionisi M, Santisi A, Massenzi L, Goffredo BM, Dionisi-Vici C, Dotta A, Auriti C. Therapeutic Drug Monitoring Is a Feasible Tool to Personalize Drug Administration in Neonates Using New Techniques: An Overview on the Pharmacokinetics and Pharmacodynamics in Neonatal Age. Int J Mol Sci 2020; 21:E5898. [PMID: 32824472 PMCID: PMC7460644 DOI: 10.3390/ijms21165898] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
Therapeutic drug monitoring (TDM) should be adopted in all neonatal intensive care units (NICUs), where the most preterm and fragile babies are hospitalized and treated with many drugs, considering that organs and metabolic pathways undergo deep and progressive maturation processes after birth. Different developmental changes are involved in interindividual variability in response to drugs. A crucial point of TDM is the choice of the bioanalytical method and of the sample to use. TDM in neonates is primarily used for antibiotics, antifungals, and antiepileptic drugs in clinical practice. TDM appears to be particularly promising in specific populations: neonates who undergo therapeutic hypothermia or extracorporeal life support, preterm infants, infants who need a tailored dose of anticancer drugs. This review provides an overview of the latest advances in this field, showing options for a personalized therapy in newborns and infants.
Collapse
Affiliation(s)
- Domenico Umberto De Rose
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, “Bambino Gesù” Children’s Hospital IRCCS, 00165 Rome, Italy; (D.U.D.R.); (A.S.); (A.D.)
| | - Sara Cairoli
- Laboratory of Metabolic Biochemistry Unit, Department of Specialist Pediatrics, “Bambino Gesù” Children’s Hospital IRCCS, 00165 Rome, Italy; (S.C.); (M.D.); (B.M.G.); (C.D.-V.)
| | - Marco Dionisi
- Laboratory of Metabolic Biochemistry Unit, Department of Specialist Pediatrics, “Bambino Gesù” Children’s Hospital IRCCS, 00165 Rome, Italy; (S.C.); (M.D.); (B.M.G.); (C.D.-V.)
| | - Alessandra Santisi
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, “Bambino Gesù” Children’s Hospital IRCCS, 00165 Rome, Italy; (D.U.D.R.); (A.S.); (A.D.)
| | - Luca Massenzi
- Neonatal Intensive Care Unit and Neonatal Pathology, Fatebenefratelli Hospital, 00186 Rome, Italy;
| | - Bianca Maria Goffredo
- Laboratory of Metabolic Biochemistry Unit, Department of Specialist Pediatrics, “Bambino Gesù” Children’s Hospital IRCCS, 00165 Rome, Italy; (S.C.); (M.D.); (B.M.G.); (C.D.-V.)
| | - Carlo Dionisi-Vici
- Laboratory of Metabolic Biochemistry Unit, Department of Specialist Pediatrics, “Bambino Gesù” Children’s Hospital IRCCS, 00165 Rome, Italy; (S.C.); (M.D.); (B.M.G.); (C.D.-V.)
| | - Andrea Dotta
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, “Bambino Gesù” Children’s Hospital IRCCS, 00165 Rome, Italy; (D.U.D.R.); (A.S.); (A.D.)
| | - Cinzia Auriti
- Neonatal Intensive Care Unit, Department of Medical and Surgical Neonatology, “Bambino Gesù” Children’s Hospital IRCCS, 00165 Rome, Italy; (D.U.D.R.); (A.S.); (A.D.)
| |
Collapse
|
16
|
Abstract
OBJECTIVES To develop a population pharmacokinetic model for IV phenobarbital in neonates following cardiac surgery and perform simulations to identify optimal dosing regimens. DESIGN Retrospective single-center pharmacokinetic study. SETTING Cardiac ICU at Children's Hospital of Philadelphia. PATIENTS Consecutive neonates who received greater than or equal to one dose of IV phenobarbital and had greater than or equal to one phenobarbital concentration drawn per standard of care from June 15, 2012, to October 15, 2018. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS A population pharmacokinetic model was developed using nonlinear mixed-effects modeling. Simulations were performed using the final model variables. Optimal phenobarbital loading doses were determined based on attainment of peak and maintenance concentrations between 20 and 40 mg/L. A total of 37 neonates contributed 159 pharmacokinetic samples. The median (range) weight, postmenstrual age, and postnatal age were 3.2 kg (1.3-3.8), 39 2/7 weeks (28 2/7 to 42 6/7), and 5 days (0-26 d), respectively. Twelve patients (32%) were on extracorporeal membrane oxygenation. An one-compartment model best described the data. The final population pharmacokinetic model included (1) weight and postnatal age for clearance and (2) weight, extracorporeal membrane oxygenation, and albumin for volume of distribution. In neonates not on extracorporeal membrane oxygenation, loading doses of 30 and 20 mg/kg reached goal concentration with albumin values less than or equal to 3 and 3.5 mg/dL, respectively. Loading doses of 30 mg/kg reached goal concentration on extracorporeal membrane oxygenation regardless of albumin values. Maintenance doses of 4-5 mg/kg/d reached goal concentration in all neonates. CONCLUSIONS In neonates following cardiac surgery, phenobarbital clearance increased with postnatal age. Volume of distribution increased with extracorporeal membrane oxygenation and lower albumin values. Loading doses of 30 mg/kg on extracorporeal membrane oxygenation and 20-30 mg/kg without extracorporeal membrane oxygenation were needed to reach goal concentration based on simulations.
Collapse
|
17
|
Lutz IC, Allegaert K, de Hoon JN, Marynissen H. Pharmacokinetics during therapeutic hypothermia for neonatal hypoxic ischaemic encephalopathy: a literature review. BMJ Paediatr Open 2020; 4:e000685. [PMID: 32577535 PMCID: PMC7299043 DOI: 10.1136/bmjpo-2020-000685] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Neonatal hypoxic ischaemic encephalopathy due to perinatal asphyxia, can result in severe neurodevelopmental disability or mortality. Hypothermia is at present the only proven neuroprotective intervention. During hypothermia, the neonate may need a variety of drugs with their specific pharmacokinetic profile. The aim of this paper is to determine the effect that hypothermia for neonates suffering from hypoxic ischaemic encephalopathy has on the pharmacokinetics and to what extent dosing regimens need adjustments. METHOD A systematic search was performed on PubMed, Embase and Cochrane Library of literature (2000-2020) using a combination of the following search terms: therapeutic hypothermia, neonate, hypoxic ischemic encephalopathy and pharmacokinetics. Titles and abstracts were screened, and inclusion/exclusion criteria were applied. Finally, relevant full texts were read, and secondary inclusion was applied on the identified articles. RESULTS A total of 380 articles were retrieved, and 34 articles included after application of inclusion/exclusion criteria and duplicate removal, two additional papers were included as suggested by the reviewers. Twelve out of 36 studies on 15 compounds demonstrated a significant decrease in clearance, be it that the extent differs between routes of elimination and compounds, most pronounced for renal elimination (phenobarbital no difference, midazolam metabolite -21%, lidocaine -24%; morphine -21% to -47%, gentamicin -25% to -35%, amikacin -40%) during hypothermia. The data as retrieved in literature were subsequent compared with the dosing regimen as stated in the Dutch paediatric formulary. CONCLUSION Depending on the drug-specific disposition characteristics, therapeutic hypothermia in neonates with hypoxic ischaemic encephalopathy affects pharmacokinetics.
Collapse
Affiliation(s)
| | - Karel Allegaert
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Intensive Care and Pediatric Surgery, Erasmus MC Sophia, Rotterdam, The Netherlands
| | - Jan N de Hoon
- Department of Pharmaceutical and Pharmacological Sciences, Center for Clinical Pharmacology, KU Leuven, Leuven, Belgium
| | - Heleen Marynissen
- Department of Pharmaceutical and Pharmacological Sciences, Center for Clinical Pharmacology, KU Leuven, Leuven, Belgium
| |
Collapse
|
18
|
Thibault C, Naim MY, Abend NS, Licht DJ, Gaynor JW, Xiao R, Massey SL. A retrospective comparison of phenobarbital and levetiracetam for the treatment of seizures following cardiac surgery in neonates. Epilepsia 2020; 61:627-635. [PMID: 32162678 DOI: 10.1111/epi.16469] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/07/2020] [Accepted: 02/13/2020] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To compare the safety and efficacy of phenobarbital and levetiracetam in a cohort of neonates with seizures following cardiac surgery. METHODS We performed a retrospective single-center study of consecutive neonates with electrographically confirmed seizures managed with antiseizure medication after cardiac surgery from June 15, 2012 to December 31, 2018. We compared the safety and efficacy of phenobarbital and levetiracetam as first-line therapy. RESULTS First-line therapy was phenobarbital in 31 neonates and levetiracetam in 22 neonates. Phenobarbital was associated with more adverse events (P = .006). Eight neonates (14%) experienced an adverse event related to phenobarbital use, including seven with hypotension and one with respiratory depression. No adverse events were reported with levetiracetam use. The cessation of electrographic seizures was similar in both groups, including 18 neonates (58%) with seizure cessation after phenobarbital and 12 neonates (55%) with seizure cessation after levetiracetam (P = 1.0). The combined cessation rates of phenobarbital and levetiracetam when used as first- or second-line therapy were 58% and 47%, respectively (P = .47). SIGNIFICANCE Phenobarbital was associated with more adverse events than levetiracetam, and the two drugs were equally but incompletely effective in treating electrographically confirmed seizures in neonates following cardiac surgery. Given its more acceptable safety profile and potential noninferiority, levetiracetam may be a reasonable option for first-line therapy for treatment of seizures in this population. Further prospective studies are needed to confirm these results.
Collapse
Affiliation(s)
- Céline Thibault
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Maryam Y Naim
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Nicholas S Abend
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel J Licht
- Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | - J William Gaynor
- Division of Cardiothoracic Surgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Rui Xiao
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Shavonne L Massey
- Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
19
|
Raffaeli G, Pokorna P, Allegaert K, Mosca F, Cavallaro G, Wildschut ED, Tibboel D. Drug Disposition and Pharmacotherapy in Neonatal ECMO: From Fragmented Data to Integrated Knowledge. Front Pediatr 2019; 7:360. [PMID: 31552205 PMCID: PMC6733981 DOI: 10.3389/fped.2019.00360] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/16/2019] [Indexed: 12/27/2022] Open
Abstract
Extracorporeal membrane oxygenation (ECMO) is a lifesaving support technology for potentially reversible neonatal cardiac and/or respiratory failure. As the survival and the overall outcome of patients rely on the treatment and reversal of the underlying disease, effective and preferentially evidence-based pharmacotherapy is crucial to target recovery. Currently limited data exist to support the clinicians in their every-day intensive care prescribing practice with the contemporary ECMO technology. Indeed, drug dosing to optimize pharmacotherapy during neonatal ECMO is a major challenge. The impact of the maturational changes of the organ function on both pharmacokinetics (PK) and pharmacodynamics (PD) has been widely established over the last decades. Next to the developmental pharmacology, additional non-maturational factors have been recognized as key-determinants of PK/PD variability. The dynamically changing state of critical illness during the ECMO course impairs the achievement of optimal drug exposure, as a result of single or multi-organ failure, capillary leak, altered protein binding, and sometimes a hyperdynamic state, with a variable effect on both the volume of distribution (Vd) and the clearance (Cl) of drugs. Extracorporeal membrane oxygenation introduces further PK/PD perturbation due to drug sequestration and hemodilution, thus increasing the Vd and clearance (sequestration). Drug disposition depends on the characteristics of the compounds (hydrophilic vs. lipophilic, protein binding), patients (age, comorbidities, surgery, co-medications, genetic variations), and circuits (roller vs. centrifugal-based systems; silicone vs. hollow-fiber oxygenators; renal replacement therapy). Based on the potential combination of the above-mentioned drug PK/PD determinants, an integrated approach in clinical drug prescription is pivotal to limit the risks of over- and under-dosing. The understanding of the dose-exposure-response relationship in critically-ill neonates on ECMO will enable the optimization of dosing strategies to ensure safety and efficacy for the individual patient. Next to in vitro and clinical PK data collection, physiologically-based pharmacokinetic modeling (PBPK) are emerging as alternative approaches to provide bedside dosing guidance. This article provides an overview of the available evidence in the field of neonatal pharmacology during ECMO. We will identify the main determinants of altered PK and PD, elaborate on evidence-based recommendations on pharmacotherapy and highlight areas for further research.
Collapse
Affiliation(s)
- Genny Raffaeli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Pavla Pokorna
- Department of Pediatrics—ICU, General University Hospital, 1st Faculty of Medicine Charles University, Prague, Czechia
- Department of Pharmacology, General University Hospital, 1st Faculty of Medicine Charles University, Prague, Czechia
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Karel Allegaert
- Division of Neonatology, Department of Pediatrics, Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Fabio Mosca
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Giacomo Cavallaro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, NICU, Milan, Italy
| | - Enno D. Wildschut
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, Netherlands
| |
Collapse
|
20
|
Application of Size and Maturation Functions to Population Pharmacokinetic Modeling of Pediatric Patients. Pharmaceutics 2019; 11:pharmaceutics11060259. [PMID: 31163633 PMCID: PMC6630378 DOI: 10.3390/pharmaceutics11060259] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/12/2019] [Accepted: 05/19/2019] [Indexed: 12/11/2022] Open
Abstract
Traditionally, dosage for pediatric patients has been optimized using simple weight-scaled methods, but these methods do not always meet the requirements of children. To overcome this discrepancy, population pharmacokinetic (PK) modeling of size and maturation functions has been proposed. The main objective of the present study was to evaluate a new modeling method for pediatric patients using clinical data from three different clinical studies. To develop the PK models, a nonlinear mixed effect modeling method was employed, and to explore PK differences in pediatric patients, size with allometric and maturation with Michaelis–Menten type functions were evaluated. Goodness of fit plots, visual predictive check and bootstrap were used for model evaluation. Single application of size scaling to PK parameters was statistically significant for the over one year old group. On the other hand, simultaneous use of size and maturation functions was statistically significant for infants younger than one year old. In conclusion, population PK modeling for pediatric patients was successfully performed using clinical data. Size and maturation functions were applied according to established criteria, and single use of size function was applicable for over one year ages, while size and maturation functions were more effective for PK analysis of neonates and infants.
Collapse
|
21
|
Falsaperla R, Mauceri L, Pavone P, Barbagallo M, Vitaliti G, Ruggieri M, Pisani F, Corsello G. Short-Term Neurodevelopmental Outcome in Term Neonates Treated with Phenobarbital versus Levetiracetam: A Single-Center Experience. Behav Neurol 2019; 2019:3683548. [PMID: 31281546 PMCID: PMC6589264 DOI: 10.1155/2019/3683548] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/08/2019] [Accepted: 04/14/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Phenobarbital (PB) has been traditionally used as the first-line treatment for neonatal seizures. More recently, levetiracetam (LEV) has been increasingly used as a promising newer antiepileptic medication for treatment of seizures in neonates. OBJECTIVES The aim of our study was to compare the effect of PB vs. LEV on short-term neurodevelopmental outcome in infants treated for neonatal seizures. METHOD This randomized, one-blind prospective study was conducted on term neonates admitted to the Neonatal Intensive Care Unit of S. Bambino Hospital, University Hospital "Policlinico-Vittorio Emanuele," Catania, Italy, from February 2016 to February 2018. Thirty term neonates with seizures were randomized to receive PB or LEV; the Hammersmith Neonatal Neurological Examination (HNNE) was used at baseline (T0) and again one month after the initial treatment (T1). RESULTS We found a significantly positive HNNE score for the developmental outcomes, specifically tone and posture, in neonates treated with LEV. There was no significant improvement in the HNNE score at T1 in the neonates treated with PB. CONCLUSION This study suggests a positive effect of levetiracetam on tone and posture in term newborns treated for neonatal seizures. If future randomized-controlled studies also show better efficacy of LEV in the treatment of neonatal seizures, LEV might potentially be considered as the first-line anticonvulsant in this age group.
Collapse
Affiliation(s)
- Raffaele Falsaperla
- Neonatal Intensive Care Unit, Santo Bambino Hospital, University Hospital “Policlinico-Vittorio Emanuele”, Via Tindaro 2, 95124 Catania, Italy
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico-Vittorio Emanuele”, Via Plebiscito 628, 95124 Catania, Italy
| | - Laura Mauceri
- Neonatal Intensive Care Unit, Santo Bambino Hospital, University Hospital “Policlinico-Vittorio Emanuele”, Via Tindaro 2, 95124 Catania, Italy
| | - Piero Pavone
- Department of Pediatrics, AOU Policlinico-Vittorio Emanuele, University of Catania, Via S. Sofia 78, 95123 Catania, Italy
| | - Massimo Barbagallo
- Department of Pediatrics, Azienda Ospedaliera di Rilievo Nazionale e di Alta Specializzazione Garibaldi, Catania, Italy
| | - Giovanna Vitaliti
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico-Vittorio Emanuele”, Via Plebiscito 628, 95124 Catania, Italy
| | - Martino Ruggieri
- Unit of Rare Diseases of the Nervous System in Childhood, Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, University of Catania, 95124 Catania, Italy
| | - Francesco Pisani
- Child Neuropsychiatry Unit, Medicine & Surgery Department, Neuroscience Division, University of Parma, Parma, Italy
| | - Giovanni Corsello
- Department of Maternal and Child Health, University of Palermo, Palermo, Italy
| |
Collapse
|
22
|
Verrotti A, Iapadre G, Di Donato G, Di Francesco L, Zagaroli L, Matricardi S, Belcastro V, Iezzi ML. Pharmacokinetic considerations for anti-epileptic drugs in children. Expert Opin Drug Metab Toxicol 2019; 15:199-211. [PMID: 30689454 DOI: 10.1080/17425255.2019.1575361] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 01/24/2019] [Indexed: 10/27/2022]
Abstract
Epilepsy is a chronic and debilitating neurological disease, with a peak of incidence in the first years of life. Today, the vast armamentarium of antiepileptic drugs (AEDs) available make even more challenging to select the most appropriate AED and establish the most effective dosing regimen. In fact, AEDs pharmacokinetics is under the influence of important age-related factors which cannot be ignored. Areas covered: Physiological changes occurring during development age (different body composition, immature metabolic patterns, reduced renal activity) can significantly modify the pharmacokinetic profile of AEDs (adsorption, volume of distribution, half-life, clearance), leading to an altered treatment response. We reviewed the main pharmacokinetic characteristics of AEDs used in children, focusing on age-related factors which are of relevance when treating this patient population. Expert opinion: To deal with this pharmacokinetic variability, physicians have at their disposal two tools: 1) therapeutic drug concentration monitoring, which may help to set the optimal therapeutic regimen for each patient and to monitor eventual fluctuation, and 2) the use of extended-release drug formulations, when available. In the next future, the development of 'ad-hoc' electronic dashboard systems will represent relevant decision-support tools making the AED therapy even more individualized and precise, especially in children.
Collapse
Affiliation(s)
- Alberto Verrotti
- a Department of Pediatrics , University of L'Aquila , L'Aquila , Italy
| | - Giulia Iapadre
- a Department of Pediatrics , University of L'Aquila , L'Aquila , Italy
| | - Giulia Di Donato
- a Department of Pediatrics , University of L'Aquila , L'Aquila , Italy
| | | | - Luca Zagaroli
- a Department of Pediatrics , University of L'Aquila , L'Aquila , Italy
| | - Sara Matricardi
- b Child Neurology and Psychiatry Unit , Children's Hospital G. Salesi , Ancona , Italy
| | | | - Maria Laura Iezzi
- a Department of Pediatrics , University of L'Aquila , L'Aquila , Italy
| |
Collapse
|
23
|
Boylan GB, Kharoshankaya L, Mathieson SR. Diagnosis of seizures and encephalopathy using conventional EEG and amplitude integrated EEG. HANDBOOK OF CLINICAL NEUROLOGY 2019; 162:363-400. [PMID: 31324321 DOI: 10.1016/b978-0-444-64029-1.00018-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Seizures are more common in the neonatal period than at any other time of life, partly due to the relative hyperexcitability of the neonatal brain. Brain monitoring of sick neonates in the NICU using either conventional electroencephalography or amplitude integrated EEG is essential to accurately detect seizures. Treatment of seizures is important, as evidence increasingly indicates that seizures damage the brain in addition to that caused by the underlying etiology. Prompt treatment has been shown to reduce seizure burden with the potential to ameliorate seizure-mediated damage. Neonatal encephalopathy most commonly caused by a hypoxia-ischemia results in an alteration of mental status and problems such as seizures, hypotonia, apnea, and feeding difficulties. Confirmation of encephalopathy with EEG monitoring can act as an important adjunct to other investigations and the clinical examination, particularly when considering treatment strategies such as therapeutic hypothermia. Brain monitoring also provides useful early prognostic indicators to clinicians. Recent use of machine learning in algorithms to continuously monitor the neonatal EEG, detect seizures, and grade encephalopathy offers the exciting prospect of real-time decision support in the NICU in the very near future.
Collapse
Affiliation(s)
- Geraldine B Boylan
- Department of Paediatrics and Child Health, Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland.
| | - Liudmila Kharoshankaya
- Department of Paediatrics and Child Health, Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland
| | - Sean R Mathieson
- Department of Paediatrics and Child Health, Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland
| |
Collapse
|
24
|
Iapadre G, Balagura G, Zagaroli L, Striano P, Verrotti A. Pharmacokinetics and Drug Interaction of Antiepileptic Drugs in Children and Adolescents. Paediatr Drugs 2018; 20:429-453. [PMID: 30003498 DOI: 10.1007/s40272-018-0302-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Selecting the most appropriate antiepileptic drug (AED) or combination of drugs for each patient and identifying the most suitable therapeutic regimen for their needs is increasingly challenging, especially among pediatric populations. In fact, the pharmacokinetics of several drugs vary widely in children with epilepsy because of age-related factors, which can influence the absorption, distribution, metabolism, and elimination of the pharmacological agent. In addition, individual factors, such as seizure type, associated comorbidities, individual pharmacokinetics, and potential drug interactions, may contribute to large fluctuations in serum drug concentrations and, therefore, clinical response. Therapeutic drug concentration monitoring (TDM) is an essential tool to deal with this complexity, enabling the definition of individual therapeutic concentrations and adaptive control of dosing to minimize drug interactions and prevent loss of efficacy or toxicity. Moreover, pharmacokinetic/pharmacodynamic modelling integrated with dashboard systems have recently been tested in antiepileptic therapy, although more clinical trials are required to support their use in clinical practice. We review the mechanism of action, pharmacokinetics, drug-drug interactions, and safety/tolerability profiles of the main AEDs currently used in children and adolescents, paying particular regard to issues of relevance when treating this patient population. Indications for TDM are provided for each AED as useful support to the clinical management of pediatric patients with epilepsy by optimizing pharmacological therapy.
Collapse
Affiliation(s)
- Giulia Iapadre
- Department of Pediatrics, University of L'Aquila, Via Vetoio, 1. Coppito, L'Aquila, Italy
| | - Ganna Balagura
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Opthalmology, Genetics, Maternal and Child Health, University of Genoa, "G. Gaslini" Institute, Genoa, Italy
| | - Luca Zagaroli
- Department of Pediatrics, University of L'Aquila, Via Vetoio, 1. Coppito, L'Aquila, Italy
| | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Opthalmology, Genetics, Maternal and Child Health, University of Genoa, "G. Gaslini" Institute, Genoa, Italy
| | - Alberto Verrotti
- Department of Pediatrics, University of L'Aquila, Via Vetoio, 1. Coppito, L'Aquila, Italy.
| |
Collapse
|
25
|
Pokorná P, Posch L, Šíma M, Klement P, Slanar O, van den Anker J, Tibboel D, Allegaert K. Severity of asphyxia is a covariate of phenobarbital clearance in newborns undergoing hypothermia. J Matern Fetal Neonatal Med 2018; 32:2302-2309. [PMID: 29357720 DOI: 10.1080/14767058.2018.1432039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
AIM Phenobarbital (PB) pharmacokinetics (PK) in asphyxiated newborns show large variability, not only explained by hypothermia (HT). We evaluated potential relevant covariates of PK of PB in newborns treated with or without HT for hypoxic-ischemic encephalopathy (HIE). METHODS Clearance (CL), distribution volume (Vd) and elimination half-life (t1/2) were calculated using one-compartment analysis. Covariates were clinical characteristics (weight, gestational age, hepatic, renal, and circulatory status), comedication and HIE severity [time to reach normal aEEG pattern (TnormaEEG), dichotomous, within 24 h] and asphyxia severity [severe aspyhxia = pH ≤7.1 + Apgar score ≤5 (5 min), dichotomous]. Student's t-test, two-way ANOVA, correlation and Pearson's chi-square test were used. RESULTS Forty newborns were included [14 non-HT; 26 HT with TnormaEEG <24 h in 14/26 (group1-HT) and TnormaEEG ≥24 h in 12/26 (group2-HT)]. Severe asphyxia was present in 26/40 [5/14 non-HT, 11/14 and 10/12 in both HT groups]. PB-CL, Vd and t1/2 were similar between the non-HT and HT group. However, within the HT group, PB-CL was significantly different between group1-HT and group2-HT (p = .043). ANOVA showed that HT (p = .034) and severity of asphyxia (p = .038) reduced PB-CL (-50%). CONCLUSION The interaction of severity of asphyxia and HT is associated with a clinical relevant reduced PB-CL, suggesting the potential relevance of disease characteristics beyond HT itself.
Collapse
Affiliation(s)
- Pavla Pokorná
- a Department of Pediatrics, First Faculty of Medicine , Charles University in Prague and General University Hospital , Prague , Czech Republic.,b Institute of Pharmacology, First Faculty of Medicine , Charles University in Prague and General University Hospital in Prague , Prague , Czech Republic.,c Intensive Care and Department of Pediatric Surgery , Erasmus MC, Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Lenka Posch
- a Department of Pediatrics, First Faculty of Medicine , Charles University in Prague and General University Hospital , Prague , Czech Republic
| | - Martin Šíma
- b Institute of Pharmacology, First Faculty of Medicine , Charles University in Prague and General University Hospital in Prague , Prague , Czech Republic
| | - Petr Klement
- a Department of Pediatrics, First Faculty of Medicine , Charles University in Prague and General University Hospital , Prague , Czech Republic
| | - Ondrej Slanar
- b Institute of Pharmacology, First Faculty of Medicine , Charles University in Prague and General University Hospital in Prague , Prague , Czech Republic
| | - John van den Anker
- c Intensive Care and Department of Pediatric Surgery , Erasmus MC, Sophia Children's Hospital , Rotterdam , The Netherlands.,d Departments of Pediatrics, Pharmacology and Physiology , George Washington University School of Medicine and Health Sciences , Washington , DC , USA.,e Division of Clinical Pharmacology , Children's National Health System , Washington , DC , USA.,f Intensive Care, Erasmus MC, Sophia Children's Hospital , Rotterdam , The Netherlands.,g Division of Paediatric Pharmacology and Pharmacometrics , University of Basel Children's Hospital , Basel , Switzerland
| | - Dick Tibboel
- a Department of Pediatrics, First Faculty of Medicine , Charles University in Prague and General University Hospital , Prague , Czech Republic.,c Intensive Care and Department of Pediatric Surgery , Erasmus MC, Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Karel Allegaert
- c Intensive Care and Department of Pediatric Surgery , Erasmus MC, Sophia Children's Hospital , Rotterdam , The Netherlands.,h Department of Development and Regeneration , KU Leuven , Leuven , Belgium
| |
Collapse
|
26
|
Yozawitz E, Stacey A, Pressler RM. Pharmacotherapy for Seizures in Neonates with Hypoxic Ischemic Encephalopathy. Paediatr Drugs 2017; 19:553-567. [PMID: 28770451 DOI: 10.1007/s40272-017-0250-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Seizures are common in neonates with moderate and severe hypoxic ischemic encephalopathy (HIE) and are associated with worse outcomes, independent of HIE severity. In contrast to adults and older children, no new drugs have been licensed for treatment of neonatal seizures over the last 50 years, because of a lack of controlled clinical trials. Hence, many antiseizure medications licensed in older children and adults are used off-label for neonatal seizure, which is associated with potential risks of adverse effects during a period when the brain is particularly vulnerable. Phenobarbital is worldwide the first-line drug and is considered standard of care, although there is a limited evidence base for its efficacy. Second-line agents include phenytoin, benzodiazepines, levetiracetam, and lidocaine. These drugs are discussed in more detail along with two emerging drugs (bumetanide and topiramate). More safety, pharmacokinetic, and efficacy data are needed from well-designed clinical trials to develop safe and effective antiseizure regimes for the treatment of neonatal seizures in HIE.
Collapse
Affiliation(s)
- Elissa Yozawitz
- Department of Neurology and Pediatrics, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Arthur Stacey
- UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Ronit M Pressler
- Department of Clinical Neurophysiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, Great Ormond Street, London, WC1N 3JH, UK. .,Clinical Neurosciences, UCL- Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
27
|
Šíma M, Pokorná P, Hartinger J, Slanař O. Estimation of initial phenobarbital dosing in term neonates with moderate-to-severe hypoxic ischaemic encephalopathy following perinatal asphyxia. J Clin Pharm Ther 2017; 43:196-201. [PMID: 28940525 DOI: 10.1111/jcpt.12632] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 09/07/2017] [Indexed: 11/26/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Phenobarbital is the first-line treatment of seizures in asphyxiated neonates; however, due to the high pharmacokinetic variability in this population, there is no consensus on the optimal dosage regimen. This study was conducted to identify variables that affect phenobarbital fate during routine clinical care and then to evaluate the dosage schedule that could be applied in term asphyxiated neonates with respect to achieving the target therapeutic range. METHODS Phenobarbital pharmacokinetics was calculated based on serum concentrations measurements using one-compartmental model. Body weight, body surface area, gestational age, creatinine clearance, total bilirubin, alanine aminotransferase, aspartate aminotransferase, international normalized ratio, Apgar scores, umbilical cord arterial pH and base excess were explored as covariates in linear regression models. Based on this analysis, phenobarbital loading and maintenance dose regimen were projected. RESULTS AND DISCUSSION In the whole study population (N = 36), phenobarbital volume of distribution, clearance and half-life median (interquartile range) values were 0.49 (0.38-0.59) L/kg, 0.0045 (0.0034-0.0055) L/h/kg and 75.1 (60.2-103.3) hours, respectively. The drug volume of distribution was associated with body weight, length and body surface area, whereas clearance was not in relationship with any explored features. Weight-normalized loading dose of 15 mg/kg and weight-normalized daily maintenance dose of 3 mg/kg proved to be optimal in our study population to reach phenobarbital therapeutic range. WHAT IS NEW AND CONCLUSIONS This study presents basis for phenobarbital initial dosing in term asphyxiated neonates during first week of life. Phenobarbital weight-normalized loading dose of 15 mg/kg lead to simulated target peak concentrations in 72% of neonates, weight-normalized maintenance dose of 3 mg/kg lead to steady state within therapeutic window in the same proportion of patients.
Collapse
Affiliation(s)
- M Šíma
- Department of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - P Pokorná
- Department of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.,Department of Pediatrics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.,Intensive Care and Department of Pediatric Surgery, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - J Hartinger
- Department of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - O Slanař
- Department of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
28
|
Baud O, Auvin S, Saliba E, Biran V. [Therapeutic management of neonatal arterial cerebral infarction and neuroprotection perspectives]. Arch Pediatr 2017; 24:9S46-9S50. [PMID: 28867038 DOI: 10.1016/s0929-693x(17)30331-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neonatal seizure related to stroke is a common diagnostic feature. Their treatment, although widely debated even today must be initiated in case of status epilepticus, clinical seizures of more than 5 minutes duration or short (> 30 secondes) and repeated clinical seizures (2 or more per hour). The treatment of neonatal seizures is a challenge that remains only partially solved. It should take into account the etiology of seizures, type of brain lesions and clinical/electrical response to treatment after the first line treatment. It is based on using a single anti-epileptic at its maximum dosage, and if needed, on the association with another anti-epileptic drug with a different mechanism of action. Phenobarbital remains the most commonly used drug for initial treatment of neonatal seizures and for which the most clinical experience has been accumulated. The lack of randomized controlled trials makes difficult recommendations about the optimal duration of treatment, but most experts agree that once arrested seizures, the duration of treatment should be as short as possible because of its potential risk on the developing brain. Novel neuroprotective strategies for reducing impact of neonatal stroke or promoting brain repair remain for the moment the concept stage, pre-clinical or parcel clinical data.
Collapse
Affiliation(s)
- O Baud
- AP-HP, néonatalogie, Hôpital universitaire Robert-Debré, 48, boulevard Sérurier, Paris, 7509 France.
| | - S Auvin
- AP-Hp neurologie et maladies métaboliques, Hôpital universitaire Robert-Debré, 48, boulevard Sérurier, Paris, 75019 France
| | - E Saliba
- Centre hospitalier régional universitaire, service de réanimation - néonatalogie, Hôpital d'enfants de Clocheville, 49, boulevard Béranger, Tours, 37044 France
| | - V Biran
- AP-HP, néonatalogie, Hôpital universitaire Robert-Debré, 48, boulevard Sérurier, Paris, 7509 France
| |
Collapse
|
29
|
Sung IK. Therapeutic Hypothermia for Hypoxic-Ischemic Encephalopathy in Newborn Infants. NEONATAL MEDICINE 2017. [DOI: 10.5385/nm.2017.24.4.145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- In Kyung Sung
- Department of Pediatrics, Collge of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
30
|
Anderson KB, Poloyac SM, Kochanek PM, Empey PE. Effect of Hypothermia and Targeted Temperature Management on Drug Disposition and Response Following Cardiac Arrest: A Comprehensive Review of Preclinical and Clinical Investigations. Ther Hypothermia Temp Manag 2016; 6:169-179. [PMID: 27622966 DOI: 10.1089/ther.2016.0003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Targeted temperature management (TTM) has been shown to reduce mortality and improve neurological outcomes in out-of-hospital cardiac arrest (CA) patients and in neonates with hypoxic-ischemic encephalopathy (HIE). TTM has also been associated with adverse drug events in the critically ill patient due to its effect on drug pharmacokinetics (PKs) and pharmacodynamics (PDs). We aim to evaluate the current literature on the effect of TTM on drug PKs and PDs following CA. MEDLINE/PubMed databases were searched for publications, which include the MeSH terms hypothermia, drug metabolism, drug transport, P450, critical care, cardiac arrest, hypoxic-ischemic encephalopathy, pharmacokinetics, and pharmacodynamics between July 2006 and October 2015. Twenty-three studies were included in this review. The studies demonstrate that hypothermia impacts PK parameters and increases concentrations of cytochrome-P450-metabolized drugs in the cooling and rewarming phase. Furthermore, the current data demonstrate a combined effect of CA and hypothermia on drug PK. Importantly, these effects can last greater than 4-5 days post-treatment. Limited evidence suggests hypothermia-mediated changes in the Phase II metabolism and the Phase III transport of drugs. Hypothermia also has been shown to potentially decrease the effect of specific drugs at the receptor level. Therapeutic hypothermia, as commonly deployed/applied during TTM, alters PK, and elevates concentrations of several commonly used medications. Hypothermia-mediated effects are an important factor when dosing and monitoring patients undergoing TTM treatment.
Collapse
Affiliation(s)
- Kacey B Anderson
- 1 Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Samuel M Poloyac
- 1 Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Patrick M Kochanek
- 2 Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Philip E Empey
- 3 Department of Pharmacy and Therapeutics, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
31
|
Mathieson SR, Livingstone V, Low E, Pressler R, Rennie JM, Boylan GB. Phenobarbital reduces EEG amplitude and propagation of neonatal seizures but does not alter performance of automated seizure detection. Clin Neurophysiol 2016; 127:3343-50. [PMID: 27514722 PMCID: PMC5034854 DOI: 10.1016/j.clinph.2016.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/20/2016] [Accepted: 07/16/2016] [Indexed: 11/28/2022]
Abstract
Phenobarbital reduces both amplitude and propagation of neonatal seizures. These changes may help to explain electroclinical uncoupling. The performance of our seizure detection algorithm was unaffected.
Objective Phenobarbital increases electroclinical uncoupling and our preliminary observations suggest it may also affect electrographic seizure morphology. This may alter the performance of a novel seizure detection algorithm (SDA) developed by our group. The objectives of this study were to compare the morphology of seizures before and after phenobarbital administration in neonates and to determine the effect of any changes on automated seizure detection rates. Methods The EEGs of 18 term neonates with seizures both pre- and post-phenobarbital (524 seizures) administration were studied. Ten features of seizures were manually quantified and summary measures for each neonate were statistically compared between pre- and post-phenobarbital seizures. SDA seizure detection rates were also compared. Results Post-phenobarbital seizures showed significantly lower amplitude (p < 0.001) and involved fewer EEG channels at the peak of seizure (p < 0.05). No other features or SDA detection rates showed a statistical difference. Conclusion These findings show that phenobarbital reduces both the amplitude and propagation of seizures which may help to explain electroclinical uncoupling of seizures. The seizure detection rate of the algorithm was unaffected by these changes. Significance The results suggest that users should not need to adjust the SDA sensitivity threshold after phenobarbital administration.
Collapse
Affiliation(s)
- Sean R Mathieson
- Academic Research Department of Neonatology, Institute for Women's Health, University College London, London, United Kingdom; Neonatal Brain Research Group, Irish Centre for Fetal and Neonatal Translational Research and Department of Paediatrics and Child Health, University College Cork, Ireland.
| | - Vicki Livingstone
- Neonatal Brain Research Group, Irish Centre for Fetal and Neonatal Translational Research and Department of Paediatrics and Child Health, University College Cork, Ireland
| | - Evonne Low
- Neonatal Brain Research Group, Irish Centre for Fetal and Neonatal Translational Research and Department of Paediatrics and Child Health, University College Cork, Ireland
| | - Ronit Pressler
- Department of Clinical Neurophysiology, Great Ormond Street Hospital, London, United Kingdom
| | - Janet M Rennie
- Academic Research Department of Neonatology, Institute for Women's Health, University College London, London, United Kingdom; Neonatal Brain Research Group, Irish Centre for Fetal and Neonatal Translational Research and Department of Paediatrics and Child Health, University College Cork, Ireland
| | - Geraldine B Boylan
- Neonatal Brain Research Group, Irish Centre for Fetal and Neonatal Translational Research and Department of Paediatrics and Child Health, University College Cork, Ireland
| |
Collapse
|
32
|
Frymoyer A, Bonifacio SL, Drover DR, Su F, Wustoff CJ, Van Meurs KP. Decreased Morphine Clearance in Neonates With Hypoxic Ischemic Encephalopathy Receiving Hypothermia. J Clin Pharmacol 2016; 57:64-76. [PMID: 27225747 DOI: 10.1002/jcph.775] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/18/2016] [Accepted: 05/19/2016] [Indexed: 12/19/2022]
Abstract
Morphine is commonly used in neonates with hypothermic ischemic encephalopathy (HIE) during therapeutic hypothermia to provide comfort and analgesia. However, pharmacokinetic data to support morphine dosing in this vulnerable population are lacking. A prospective, 2-center clinical pharmacokinetic study of morphine was conducted in 20 neonates (birthweight, 1.82-5.3 kg) with HIE receiving hypothermia. Morphine dosing was per standard of care at each center. Morphine and glucuronide metabolites (morphine-3-glucuronide and morphine-6-gluronide) were measured via a validated dried blood spot liquid chromatography-tandem mass spectrometry assay. From the available concentration data (n = 106 for morphine; n = 106 for each metabolite), a population pharmacokinetic model was developed using nonlinear mixed-effects modeling. The clearance of morphine and glucuronide metabolites was best predicted by birthweight allometrically scaled using an exponent of 1.23. In addition, the clearance of each glucuronide metabolite was influenced by serum creatinine. No other significant predictors of clearance or volume of distribution were found. For a 3.5-kg neonate, morphine clearance was 0.77 L/h (CV, 48%), and the steady-state volume of distribution was 8.0 L (CV, 49%). Compared with previous studies in full-term newborns without HIE, morphine clearance was markedly lower. Dosing strategies customized for this vulnerable population will be needed. Applying the final population pharmacokinetic model, repeated Monte Carlo simulations (n = 1000 per simulation) were performed to evaluate various morphine dosing strategies that optimized achievement of morphine concentrations between 10 and 40 ng/mL. An optimized morphine loading dose of 50 μg/kg followed by a continuous infusion of 5 μg/kg/h was predicted across birthweights.
Collapse
Affiliation(s)
- Adam Frymoyer
- Department of Pediatrics, Stanford University, Palo Alto, CA, USA
| | | | - David R Drover
- Department of Anesthesiology, Perioperative and Pain Medicine, Palo Alto, CA, USA
| | - Felice Su
- Department of Pediatrics, Stanford University, Palo Alto, CA, USA
| | | | | |
Collapse
|
33
|
|
34
|
ŠÍMA M, POKORNÁ P, HRONOVÁ K, SLANAŘ O. Effect of Co-Medication on the Pharmacokinetic Parameters of Phenobarbital in Asphyxiated Newborns. Physiol Res 2015; 64:S513-9. [DOI: 10.33549/physiolres.933213] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Phenobarbital is an anticonvulsive drug widely used in newborns with hypoxic-ischemic encephalopathy. The objective of our study was to describe possible effect of frequently co-administered medications (dopamine, dobutamine, norepinephrine, furosemide, phenytoin, and analgesics) on the phenobarbital pharmacokinetics in full term newborns with hypoxic-ischemic encephalopathy. Phenobarbital pharmacokinetic parameters (standardized intravenous loading dose was 10-20 mg/kg, maintenance dose 2-6 mg/kg/day) were computed using non-compartmental analysis. Co-medication was evaluated throughout the whole treatment period up to 5 days. Volume of distribution, clearance, and half-life median values (95 % CI) for phenobarbital in the whole study population (n=37) were 0.48 (0.41-0.56) l/kg, 0.0034 (0.0028-0.0040) l/h/kg, and 93.7 (88.1-99.2) h, respectively. Phenobarbital pharmacokinetic parameters were not significantly affected by vasoactive drugs (dopamine, dobutamine, and norepinephrine), furosemide, phenytoin, or analgesics. Furthermore, no dose-dependent alteration of phenobarbital pharmacokinetic parameters was noted for vasoactive medication at doses equivalent to cumulative vasoactive-inotropic score (area under the curve in a plot of vasoactive-inotropic score against time) 143.2-8473.6, furosemide at cumulative doses of 0.2-42.9 mg/kg, or phenytoin at cumulative doses of 10.3-46.2 mg/kg. Phenobarbital pharmacokinetics was not affected by investigated co-administered drugs used in newborns with hypoxic-ischemic encephalopathy in real clinical settings.
Collapse
Affiliation(s)
- M. ŠÍMA
- Department of Pharmacology, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | | | | | | |
Collapse
|
35
|
Donovan MD, Boylan GB, Murray DM, Cryan JF, Griffin BT. Treating disorders of the neonatal central nervous system: pharmacokinetic and pharmacodynamic considerations with a focus on antiepileptics. Br J Clin Pharmacol 2015; 81:62-77. [PMID: 26302437 DOI: 10.1111/bcp.12753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 08/05/2015] [Accepted: 08/13/2015] [Indexed: 12/31/2022] Open
Abstract
A major consideration in the treatment of neonatal disorders is that the selected drug, dose and dosage frequency is safe, effective and appropriate for the intended patient population. Thus, a thorough knowledge of the pharmacokinetics and pharmacodynamics of the chosen drug within the patient population is essential. In paediatric and neonatal populations two additional challenges can often complicate drug treatment - the inherently greater physiological variability, and a lack of robust clinical evidence of therapeutic range. There has traditionally been an overreliance in paediatric medicine on extrapolating doses from adult values by adjusting for bodyweight or body surface area, but many other sources of variability exist which complicate the choice of dose in neonates. The lack of reliable drug dosage data in neonates has been highlighted by regulatory authorities, as only ~50% of the most commonly used paediatric medicines have been examined in a paediatric population. Moreover, there is a paucity of information on the pharmacokinetic parameters which affect drug concentrations in different body tissues, and pharmacodynamic responses to drugs in the neonate. Thus, in the present review, we draw attention to the main pharmacokinetic factors that influence the unbound brain concentration of neuroactive drugs. Moreover, the pharmacodynamic differences between neonates and adults that affect the activity of centrally-acting therapeutic agents are briefly examined, with a particular emphasis on antiepileptic drugs.
Collapse
Affiliation(s)
- Maria D Donovan
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Geraldine B Boylan
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,Irish Centre for Fetal and Neonatal Translational Research, University College Cork and Cork University Maternity Hospital, Cork, Ireland
| | - Deirdre M Murray
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Brendan T Griffin
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| |
Collapse
|
36
|
Lynch NE, Stevenson NJ, Livingstone V, Mathieson S, Murphy BP, Rennie JM, Boylan GB. The temporal characteristics of seizures in neonatal hypoxic ischemic encephalopathy treated with hypothermia. Seizure 2015; 33:60-5. [PMID: 26571073 DOI: 10.1016/j.seizure.2015.10.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 12/25/2022] Open
Abstract
PURPOSE The characteristics of electrographic seizures in newborns with hypoxic-ischaemic encephalopathy (HIE) treated with therapeutic hypothermia (TH) are poorly described. This retrospective, observational study provides reference data on the characteristics of seizures and their evolution over time in newborns with HIE receiving whole-body TH. METHOD The cohort under analysis included 23 infants with HIE and seizures defined by multi-channel EEG recordings. Clinical presentation, details of TH and antiepileptic drugs used were recorded. Time from first to last-recorded electrographic seizure (seizure period) was calculated. Temporal characteristics of seizures - total burden, duration, number, burden in minutes per hour, distribution of burden over time (temporal evolution), time from seizure onset to maximum seizure burden (Tmsb), T1, and time from Tmsb to seizure offset, T2 - were analysed. RESULTS The median age at electrographic seizure onset was 13.1h (IQR: 11.4 to 22.0). Tmsb was reached at a median age of 19.4 hours (IQR: 12.2 to 29.7). Median seizure period was 16.5h (IQR: 7.0 to 49.7), median number of seizures per hour was 1.9 (IQR: 1.0 to 3.3). The seizure burden was 4.0 min/h (IQR: 2.0 to 7.0). There was no consistent pattern in the temporal evolution of seizures in neonates treated with TH. The skewness was neither positive nor negative (p-value=0.15), there was no difference between the duration of T1 and T2 (p-value=0.09) and no difference in the seizure burden between T1 and T2 (p=0.09). There was an association between Tmsb and Phenobarbital (PB) administration (r=0.76, p-value<0.001). CONCLUSION There is no consistent temporal evolution of seizure burden in neonates treated with TH. Seizures are diffuse, and their characteristics are variable.
Collapse
Affiliation(s)
- Niamh E Lynch
- Department of Paediatrics and Child Health, University College Cork; Neonatal Brain Research Group, Irish Centre for Fetal and Neonatal Translational Research, University College Cork
| | - Nathan J Stevenson
- Neonatal Brain Research Group, Irish Centre for Fetal and Neonatal Translational Research, University College Cork
| | - Vicki Livingstone
- Neonatal Brain Research Group, Irish Centre for Fetal and Neonatal Translational Research, University College Cork
| | - Sean Mathieson
- Elizabeth Garrett Anderson Institute for Women's Health, University College London Hospitals, London
| | - Brendan P Murphy
- Department of Paediatrics and Child Health, University College Cork; Neonatal Brain Research Group, Irish Centre for Fetal and Neonatal Translational Research, University College Cork
| | - Janet M Rennie
- Elizabeth Garrett Anderson Institute for Women's Health, University College London Hospitals, London
| | - Geraldine B Boylan
- Department of Paediatrics and Child Health, University College Cork; Neonatal Brain Research Group, Irish Centre for Fetal and Neonatal Translational Research, University College Cork.
| |
Collapse
|
37
|
Julian S, Burnham CAD, Sellenriek P, Shannon WD, Hamvas A, Tarr PI, Warner BB. Impact of neonatal intensive care bed configuration on rates of late-onset bacterial sepsis and methicillin-resistant Staphylococcus aureus colonization. Infect Control Hosp Epidemiol 2015; 36:1173-82. [PMID: 26108888 PMCID: PMC5089903 DOI: 10.1017/ice.2015.144] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Infections cause morbidity and mortality in neonatal intensive care units (NICUs). The association between nursery design and nosocomial infections is unclear. OBJECTIVE To determine whether rates of colonization by methicillin-resistant Staphylococcus aureus (MRSA), late-onset sepsis, and mortality are reduced in single-patient rooms. DESIGN Retrospective cohort study. SETTING NICU in a tertiary referral center. METHODS Our NICU is organized into single-patient and open-unit rooms. Clinical data sets including bed location and microbiology results were examined over 29 months. Differences in outcomes between bed configurations were determined by χ2 and Cox regression. PATIENTS All NICU patients. RESULTS Among 1,823 patients representing 55,166 patient-days, single-patient and open-unit models had similar incidences of MRSA colonization and MRSA colonization-free survival times. Average daily census was associated with MRSA colonization rates only in single-patient rooms (hazard ratio, 1.31; P=.039), whereas hand hygiene compliance on room entry and exit was associated with lower colonization rates independent of bed configuration (hazard ratios, 0.834 and 0.719 per 1% higher compliance, respectively). Late-onset sepsis rates were similar in single-patient and open-unit models as were sepsis-free survival and the combined outcome of sepsis or death. After controlling for demographic, clinical, and unit-based variables, multivariate Cox regression demonstrated that bed configuration had no effect on MRSA colonization, late-onset sepsis, or mortality. CONCLUSIONS MRSA colonization rate was impacted by hand hygiene compliance, regardless of room configuration, whereas average daily census affected only infants in single-patient rooms. Single-patient rooms did not reduce the rates of MRSA colonization, late-onset sepsis, or death.
Collapse
Affiliation(s)
- Samuel Julian
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Carey-Ann D. Burnham
- Departments of Pathology & Immunology and Pediatrics, Washington University School of Medicine, St. Louis, MO
| | | | - William D. Shannon
- Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Aaron Hamvas
- Department of Pediatrics, Northwestern Feinberg School of Medicine, Chicago, IL
| | - Phillip I. Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Barbara B. Warner
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
38
|
Wietstock SO, Bonifacio SL, McCulloch CE, Kuzniewicz MW, Glass HC. Neonatal Neurocritical Care Service Is Associated With Decreased Administration of Seizure Medication. J Child Neurol 2015; 30:1135-41. [PMID: 25380602 PMCID: PMC4424192 DOI: 10.1177/0883073814553799] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 08/25/2014] [Indexed: 11/16/2022]
Abstract
This cohort study examines medication use in term neonates with hypoxic-ischemic encephalopathy and seizures before and after implementation of a Neonatal Neurocritical Care Service (N = 108), which included increased seizure monitoring. Nearly all neonates received phenobarbital (96% pre- vs 95% post-Neonatal Neurocritical Care Service) and total loading dose did not vary among groups (33 [95% confidence interval 29-37] vs 30 [26-34] mg/kg). After adjustment for seizure burden, neonates managed during the Neonatal Neurocritical Care Service era, on average, received 30 mg/kg less cumulative phenobarbital (95% confidence interval 15-46 mg/kg) and were on maintenance 5 fewer days (95% confidence interval 3-8 days) than those who were treated prior to implementation of the service. In spite of the enhanced ability to detect seizures because of improved monitoring and increased vigilance by bedside practitioners, implementation of the Neonatal Neurocritical Care Service was associated with decreased use of potentially harmful phenobarbital treatment among neonates with hypoxic-ischemic encephalopathy.
Collapse
Affiliation(s)
- Sharon O Wietstock
- Departments of Pediatrics and Neurology, University of California, San Francisco, CA, USA
| | - Sonia L Bonifacio
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Charles E McCulloch
- Department of Epidemiology & Biostatistics, University of California, San Francisco, CA, USA
| | - Michael W Kuzniewicz
- Department of Pediatrics, University of California, San Francisco, CA, USA Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Hannah C Glass
- Department of Pediatrics, University of California, San Francisco, CA, USA Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
39
|
Therapeutic drug monitoring of carbamazepine and its metabolite in children from dried blood spots using liquid chromatography and tandem mass spectrometry. J Pharm Biomed Anal 2015; 109:164-70. [DOI: 10.1016/j.jpba.2015.02.045] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/20/2015] [Accepted: 02/22/2015] [Indexed: 11/23/2022]
|
40
|
Shetty J. Neonatal seizures in hypoxic-ischaemic encephalopathy--risks and benefits of anticonvulsant therapy. Dev Med Child Neurol 2015; 57 Suppl 3:40-3. [PMID: 25800491 DOI: 10.1111/dmcn.12724] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/17/2014] [Indexed: 12/14/2022]
Abstract
The risk of seizures is at its highest during the neonatal period, and the most common cause of neonatal seizures is hypoxic-ischaemic encephalopathy (HIE). This enhanced vulnerability is caused by an imbalance in the expression of receptors for excitatory and inhibitory neurotransmission, which is age dependent. There has been progress in detecting the electrophysiological abnormalities associated with seizures using amplitude-integrated electroencephalography (aEEG). Data from animal studies indicate a variety of risk factors for seizures, but there are limited clinical data looking at the long-term neurodevelopmental consequences of seizures alone. Neonatal seizures are also associated with increased risk of further epileptic seizures; however, it is less clear whether or not this results from an underlying pathology, and whether or not seizures confer additional risk. Phenobarbital and phenytoin are still the first-line antiepileptic drugs (AEDs) used to treat neonatal seizures, although they are effective in only one-third of affected infants. Furthermore, based on findings from animal studies, there are concerns regarding the risks associated with using these AEDs. Clinicians face a difficult challenge because, although seizures can be easily identified using aEEG, treatment options are limited, and there are uncertainties regarding treatment outcomes. There is a need to obtain long-term follow-up data, comparing groups of infants treated with or without current therapies. If these analyses indicate a definite benefit of treating neonatal seizures, then novel therapeutic approaches should be developed.
Collapse
Affiliation(s)
- Jayakara Shetty
- Department of Paediatric Neurosciences, Royal Hospital for Sick Children, Edinburgh, UK
| |
Collapse
|
41
|
Seizures and hypothermia: importance of electroencephalographic monitoring and considerations for treatment. Semin Fetal Neonatal Med 2015; 20:103-8. [PMID: 25683598 DOI: 10.1016/j.siny.2015.01.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hypoxic-ischemic encephalopathy is a common cause of seizures in neonates. Despite the introduction of therapeutic hypothermia, seizure rates are similar to those reported in the pre-therapeutic hypothermia era. However, the seizure profile has been altered resulting in a lower overall seizure burden, shorter individual seizure durations, and seizures that are harder to detect. Electroencephalographic (EEG) monitoring is the gold standard for detecting all seizures in neonates and this is even more critical in neonates who are cooled, as they are often sedated, making seizures more difficult to detect. Several studies have shown that the majority of seizures in neonates undergoing therapeutic hypothermia remain subclinical, thus requiring EEG monitoring for diagnosis. Amplitude-integrated EEG monitoring is useful but shorter duration seizures are more likely to be missed. Evidence is emerging about the pharmacokinetic profile of routinely used antiepileptic drugs during therapeutic hypothermia and some modifications have been suggested, particularly for lidocaine use.
Collapse
|
42
|
Sarkar S, Barks J. Management of neonatal morbidities during hypothermia treatment. Semin Fetal Neonatal Med 2015; 20:97-102. [PMID: 25701292 DOI: 10.1016/j.siny.2015.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Although the primary goal of therapeutic hypothermia is to improve the neurodevelopmental outcome in asphyxiated infants, optimal management of the full range of multi-organ system complications typically presented by such infants during cooling treatment is necessary for improvement of the overall outcome. For this reason, adequate knowledge of how cooling affects all organ systems of asphyxiated infants with multi-organ hypoxic-ischemic injury is essential. Adequate diagnostic resources, readily available subspecialty consultant services and trained multidisciplinary staff to monitor and manage multi-organ system complications in asphyxiated infants during therapeutic cooling must be ensured during implementation of a cooling program. As therapeutic hypothermia is being used more widely, centers should consider participation in national or international benchmarking of outcomes and short-term adverse events during cooling to facilitate continuous quality improvement efforts.
Collapse
Affiliation(s)
- Subrata Sarkar
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, The University of Michigan, C.S. Mott Children's Hospital, Ann Arbor, MI, USA
| | - John Barks
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, The University of Michigan, C.S. Mott Children's Hospital, Ann Arbor, MI, USA.
| |
Collapse
|
43
|
Abstract
Neonatal seizures are the most important indicators of underlying brain injury. Seizures in a neonate are different from seizures in older children in many aspects including clinical presentation and etiology. The neonatal brain is immature and tends to have a decreased seizure threshold. Neonatal seizures are classified, based on their presentation as, clinical seizures, electroclinical seizures and electroencephalographic seizures; based on the pathophysiology as epileptic and nonepileptic seizures; and also on the basis of the etiology. Hypoxic ischemic encephalopathy is the leading cause of neonatal seizures, followed by intracranial hemorrhage, metabolic causes such as hypoglycemia and hypocalcemia, intracranial infections and strokes. Neonatal epilepsy syndromes are rare. Electroencephalography (EEG) is the gold standard for diagnosis. Amplitude integrated EEG (aEEG) is also used for continuous monitoring. The approach to management consists of initial stabilization of the neonate followed by treatment of potentially correctable injurious processes such as hypocalcemia, hypoglycemia and electrolyte disturbances, etiology specific therapies and antiepileptic drug (AED) therapy. Phenobarbital remains the first line AED therapy. Pharmacokinetic data on newer drugs is limited. Prognosis depends on the etiology, seizure type, neurological examination at discharge and EEG. Long term neurodevelopmental follow up is essential for babies with neonatal seizures.
Collapse
Affiliation(s)
- Sujata Kanhere
- Division of Pediatric Neurology, Department of Pediatrics & Neonatology, K.J. Somaiya Medical College, Hospital & Research Centre, Mumbai, Maharashtra, 400022, India,
| |
Collapse
|
44
|
Gutherz SB, Kulick CV, Soper C, Kondratyev A, Gale K, Forcelli PA. Brief postnatal exposure to phenobarbital impairs passive avoidance learning and sensorimotor gating in rats. Epilepsy Behav 2014; 37:265-9. [PMID: 25112558 PMCID: PMC4170015 DOI: 10.1016/j.yebeh.2014.07.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 06/10/2014] [Accepted: 07/03/2014] [Indexed: 10/24/2022]
Abstract
Phenobarbital is the most commonly utilized drug for the treatment of neonatal seizures. However, mounting preclinical evidence suggests that even brief exposure to phenobarbital in the neonatal period can induce neuronal apoptosis, alterations in synaptic development, and long-lasting changes in behavioral functions. In the present report, we treated neonatal rat pups with phenobarbital and evaluated behavior in adulthood. Pups were treated initially with a loading dose (80 mg/kg) on postnatal day (P)7 and with a lower dose (40 mg/kg) on P8 and P9. We examined sensorimotor gating (prepulse inhibition), passive avoidance, and conditioned place preference for cocaine when the animals reached adulthood. Consistent with our previous reports, we found that three days of neonatal exposure to phenobarbital significantly impaired prepulse inhibition compared with vehicle-exposed control animals. Using a step-though passive avoidance paradigm, we found that animals exposed to phenobarbital as neonates and tested as adults showed significant deficits in passive avoidance retention compared with matched controls, indicating impairment in associative memory and/or recall. Finally, we examined place preference conditioning in response to cocaine. Phenobarbital exposure did not alter the normal conditioned place preference associated with cocaine exposure. Our findings expand the profile of behavioral toxicity induced by phenobarbital.
Collapse
Affiliation(s)
- Samuel B. Gutherz
- Department of Pharmacology and Physiology, Georgetown University, School of Medicine, Washington, DC 20007
| | - Catherine V. Kulick
- Department of Pharmacology and Physiology, Georgetown University, School of Medicine, Washington, DC 20007
| | - Colin Soper
- Department of Pharmacology and Physiology, Georgetown University, School of Medicine, Washington, DC 20007
| | - Alexei Kondratyev
- Department of Pharmacology and Physiology, Georgetown University, School of Medicine, Washington, DC 20007,Interdisciplinary Program in Neuroscience, Georgetown University, School of Medicine, Washington, DC 20007
| | - Karen Gale
- Department of Pharmacology and Physiology, Georgetown University, School of Medicine, Washington, DC 20007,Interdisciplinary Program in Neuroscience, Georgetown University, School of Medicine, Washington, DC 20007
| | - Patrick A. Forcelli
- Department of Pharmacology and Physiology, Georgetown University, School of Medicine, Washington, DC 20007,Interdisciplinary Program in Neuroscience, Georgetown University, School of Medicine, Washington, DC 20007
| |
Collapse
|
45
|
Shankaran S. Current status of hypothermia for hypoxemic ischemia of the newborn. Indian J Pediatr 2014; 81:578-84. [PMID: 24820235 DOI: 10.1007/s12098-014-1468-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/21/2014] [Indexed: 11/27/2022]
Abstract
This article reviews the pathophysiology of hypoxic-ischemic brain injury, and the impact of hypothermia as neuroprotection in the clinical setting. The results of trials performed in well resourced and mid and low resourced countries is presented. Infant and childhood outcome following hypothermia is provided. Biomarkers of outcome that are clinical, electrophysiological and imaging which will be helpful to clinicians are noted. Management of infants with encephalopathy, including safety of hypothermia is reviewed. The article concludes with knowledge gaps in neuroprotection and the future of hypothermia therapy.
Collapse
Affiliation(s)
- Seetha Shankaran
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, 48201, USA,
| |
Collapse
|
46
|
Bijleveld Y, de Haan TR, Toersche J, Jorjani S, van der Lee J, Groenendaal F, Dijk P, van Heijst A, Gavilanes AWD, de Jonge R, Dijkman KP, van Straaten H, Rijken M, Zonnenberg I, Cools F, Nuytemans D, Mathôt R. A simple quantitative method analysing amikacin, gentamicin, and vancomycin levels in human newborn plasma using ion-pair liquid chromatography/tandem mass spectrometry and its applicability to a clinical study. J Chromatogr B Analyt Technol Biomed Life Sci 2014; 951-952:110-118. [PMID: 24548921 DOI: 10.1016/j.jchromb.2014.01.035] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 01/16/2014] [Accepted: 01/22/2014] [Indexed: 01/11/2023]
Abstract
Neuroprotective controlled therapeutic hypothermia is the standard of care for newborns suffering perinatal asphyxia. Antibiotic drugs, such as amikacin, gentamicin, and vancomycin are frequently administered during controlled hypothermia, which possibly alters their pharmacokinetic (PK) and pharmacodynamic (PD) profiles. In order to examine this effect an LC-MS/MS method for the simultaneous quantification of amikacin, the major gentamicin components (gentamicin C, C1a and C2), and vancomycin in plasma was developed. In 25μL plasma proteins were precipitated with trichloroacetic acid (TCA) and detection of the components was achieved using ion-pair reversed phase chromatography coupled with electrospray ionization tandem mass spectrometry. The chromatographic runtime was 7.5min per sample. Calibration standards were prepared over a range of 0.3-50mgL(-1) for amikacin and gentamicin and 1.0-100mgL(-1) for vancomycin. At LLOQ accuracy was between 103 and 120% and imprecision was less than 19%. For concentrations above LLOQ accuracy ranged from 98% to 102% and imprecision was less than 6%. Process efficiency, ionization efficiency, and recovery were acceptable. Samples and stock solutions were stable during the time periods and at the different temperatures examined. The applicability of the method was shown by analysing plasma samples from 3 neonatal patients. The developed method allows accurate and precise simultaneous quantification of amikacin, gentamicin, and vancomycin in a small volume (25μL) of plasma.
Collapse
Affiliation(s)
- Yuma Bijleveld
- Department of Pharmacy, Academic Medical Centre, Amsterdam, The Netherlands.
| | - Timo R de Haan
- Department of Neonatology, Emma Children's Hospital, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan Toersche
- Department of Pharmacy, Academic Medical Centre, Amsterdam, The Netherlands
| | - Sona Jorjani
- Department of Pharmacy, Academic Medical Centre, Amsterdam, The Netherlands
| | - Johanna van der Lee
- Department of Paediatric Clinical Epidemiology, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Floris Groenendaal
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter Dijk
- Department of Neonatology, University Medical Center Groningen, Groningen, The Netherlands
| | - Arno van Heijst
- Department of Neonatology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Antonio W D Gavilanes
- Department of Neonatology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Rogier de Jonge
- Department of Neonatology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Koen P Dijkman
- Department of Neonatology, Maxima Medical Center Veldhoven, Veldhoven, The Netherlands
| | | | - Monique Rijken
- Department of Neonatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Inge Zonnenberg
- Department of Neonatology, VU University Medical Center, Amsterdam, The Netherlands
| | - Filip Cools
- Department of Neonatology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Ron Mathôt
- Department of Pharmacy, Academic Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Himebauch AS, Zuppa A. Methods for pharmacokinetic analysis in young children. Expert Opin Drug Metab Toxicol 2014; 10:497-509. [DOI: 10.1517/17425255.2014.885502] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
48
|
Simultaneous LC–MS/MS quantification of P-glycoprotein and cytochrome P450 probe substrates and their metabolites in DBS and plasma. Bioanalysis 2014; 6:151-64. [DOI: 10.4155/bio.13.289] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: An LC–MS/MS method has been developed for the simultaneous quantification of P-glycoprotein (P-gp) and cytochrome P450 (CYP) probe substrates and their Phase I metabolites in DBS and plasma. P-gp (fexofenadine) and CYP-specific substrates (caffeine for CYP1A2, bupropion for CYP2B6, flurbiprofen for CYP2C9, omeprazole for CYP2C19, dextromethorphan for CYP2D6 and midazolam for CYP3A4) and their metabolites were extracted from DBS (10 µl) using methanol. Analytes were separated on a reversed-phase LC column followed by SRM detection within a 6 min run time. Results: The method was fully validated over the expected clinical concentration range for all substances tested, in both DBS and plasma. The method has been successfully applied to a PK study where healthy male volunteers received a low dose cocktail of the here described P-gp and CYP probes. Good correlation was observed between capillary DBS and venous plasma drug concentrations. Conclusion: Due to its low-invasiveness, simple sample collection and minimal sample preparation, DBS represents a suitable method to simultaneously monitor in vivo activities of P-gp and CYP.
Collapse
|
49
|
Primum non nocere: are seizure medications safe in neonates? Epilepsy Curr 2013; 13:179-81. [PMID: 24009483 DOI: 10.5698/1535-7597-13.4.179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
50
|
Srinivasakumar P, Zempel J, Wallendorf M, Lawrence R, Inder T, Mathur A. Therapeutic hypothermia in neonatal hypoxic ischemic encephalopathy: electrographic seizures and magnetic resonance imaging evidence of injury. J Pediatr 2013; 163:465-70. [PMID: 23452588 DOI: 10.1016/j.jpeds.2013.01.041] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 12/20/2012] [Accepted: 01/22/2013] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To evaluate the electrographic seizure burden in neonates with hypoxic ischemic encephalopathy (HIE) treated with or without therapeutic hypothermia and stratified results by severity of HIE and severity of injury as assessed by magnetic resonance imaging (MRI). STUDY DESIGN Between 2007 and 2011, video-electroencephalography (EEG) monitoring was initiated in neonates with moderate to severe HIE. Seizure burden (in seconds) was calculated, and brain MRI scans were quantitatively scored. Data were analyzed by ANOVA, the Student t test, and the χ(2) test. RESULTS Sixty-nine neonates with moderate or severe HIE were prospectively enrolled, including 51 who received therapeutic hypothermia and 18 who did not. The mean duration of video-EEG monitoring was longer in the therapeutic hypothermia group (72 ± 34 hours vs 48 ± 34 hours; P = .01). The therapeutic hypothermia group had a lower electrographic seizure burden (log units) after controlling for injury, as assessed by MRI (2.9 ± 0.6 vs 6.2 ± 0.9; P = .003). A reduction in seizure burden was seen in neonates with moderate HIE (P = .0001), but not in those with severe HIE (P = .80). Among neonates with injury assessed by MRI, seizure burden was lower in those with mild (P = .0004) and moderate (P = .02) injury, but not in those with severe injury (P = .90). CONCLUSION Therapeutic hypothermia was associated with reduced electrographic seizure burden in neonatal HIE. This effect was detected on video-EEG in infants with moderate HIE, but not in those with severe HIE. When stratified by injury as assessed by MRI, therapeutic hypothermia was associated with a reduced seizure burden in infants with mild and moderate injury, but not in those with severe injury.
Collapse
|