1
|
Stokes MA, Kamel NA, Festa MS, Sandaradura I, Stocker SL. Scoping Review of Paediatric Population Pharmacokinetic Models of Morphine. Clin Pharmacokinet 2025:10.1007/s40262-025-01477-5. [PMID: 40310579 DOI: 10.1007/s40262-025-01477-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2025] [Indexed: 05/02/2025]
Abstract
OBJECTIVE AND PURPOSE This scoping review aimed to summarise all available population pharmacokinetic models of morphine and its metabolites (morphine-3-glucoronide [M3G], morphine-6-glucoronide [M6G]) in children and describe how morphine exposure varies across paediatric age groups and settings. Identifying the factors that contribute to pharmacokinetic variability may improve our understanding of a patient's pharmacodynamic response to morphine. METHODS We searched Embase and MEDLINE databases from inception to 8 March 2024 for paediatric population pharmacokinetic models of morphine and its metabolites. Two reviewers independently screened abstracts and full texts and extracted the data. The review was conducted according to Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. RESULTS In total, 21 paediatric population pharmacokinetic models of morphine were identified; 12 studies also included morphine metabolites (M3G and/or M6G). Neonates and young children (< 6 years) were the most studied age groups (18/21; 86%), whereas older children (> 6 years) and adolescents (> 10 years) were included in only 6 of the 21 (29%) models. Morphine pharmacokinetics were most commonly described with two-compartment (52%) and one-compartment (38%) structure with first-order elimination. Several model covariates were identified: bodyweight, post-natal age for neonates, body temperature, therapeutic cooling, duration of mechanical ventilation, and genetic variation in drug transporters that mediate the uptake of morphine (e.g. OCT1). CONCLUSION Several population pharmacokinetic models of morphine and its metabolites in paediatrics have been published across diverse patient groups. Bodyweight and age-related covariates emerged as the most common factors affecting clearance and distribution; other covariates, including mechanical ventilation, therapeutic cooling, and genetic variation, also impacted morphine pharmacokinetics. Further research should focus on validating the predictive accuracy of paediatric morphine models in different patient populations and the combined effect of covariates, such as those related to critical illness and genetic variation, on morphine pharmacokinetics.
Collapse
Affiliation(s)
- Michael A Stokes
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, A15 Pharmacy and Bank Building, Science Road, Camperdown, NSW, 2006, Australia
- Kids Critical Care Research, Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Noha A Kamel
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, A15 Pharmacy and Bank Building, Science Road, Camperdown, NSW, 2006, Australia
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Marino S Festa
- Kids Critical Care Research, Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Indy Sandaradura
- Centre for Infectious Diseases and Microbiology, Westmead Hospital, Sydney, NSW, Australia
- Faculty of Medicine and Health, Westmead Clinical School, The University of Sydney, Sydney, NSW, Australia
- Institute of Clinical Pathology and Medical Research, New South Wales Health Pathology, Westmead Hospital, Sydney, NSW, Australia
| | - Sophie L Stocker
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, A15 Pharmacy and Bank Building, Science Road, Camperdown, NSW, 2006, Australia.
- Department of Clinical Pharmacology and Toxicology, St. Vincent's Hospital Sydney, Sydney, NSW, 2010, Australia.
- St. Vincent's Clinical Campus, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia.
- Sydney Musculoskeletal Health, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
2
|
Abdelwahab AH, Abdullah LO, Mohammed N, Alikunju M, Abdul Rouf PV, Thomas B, Sree B, Elkassem WM. The effect of morphine in the development of neonatal cholestasis in extremely preterm neonates. J Neonatal Perinatal Med 2025:19345798251336731. [PMID: 40250844 DOI: 10.1177/19345798251336731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2025]
Abstract
BackgroundMorphine is commonly used in ventilated neonates as an analgesic with many side effects like respiratory depression and hypotension. Only a few studies have investigated morphine's influence on biliary tract dynamics. Therefore, we planned this study to find out the relationship between morphine and liver function in preterm neonates.MethodsThis retrospective study was conducted at the Women's Wellness and Research Centre by reviewing the case records of neonates admitted between 01st January 2015 and 31st December 2020. Preterm infants on mechanical ventilation, between gestation ages (GA) ≥24 and ≤37 weeks, were included in the study. Participants were divided into two groups according to morphine use, and liver function test values were compared, including direct (DB), total bilirubin (TB), alanine transaminase (ALT), and aspartate transaminase (AST).ResultsThe study comprised 133 preterm newborns; out of them, 73 received morphine (Group A) and 60 did not receive it (Group B). The mean GA was 29.90 ± 5.18 weeks, and birth weight was 1250 ± 418 grams. At admission, DB levels were higher in group B than in group A (54.71 ± 50.0 vs 36.77±48.53, p = .038). However, the peak and discharge values were higher in group A than in group B (p = .001 and p = .196, respectively). The peak AST, DB, and TB values were higher in group A.ConclusionThe present study showed the impact of morphine on liver function in preterm neonates and the possibility of morphine hepatotoxicity. Clinicians should use morphine cautiously in preterm neonates to avoid the risk of developing cholestasis and hepatotoxicity.
Collapse
Affiliation(s)
- A H Abdelwahab
- Neonatology Department, Hamad Medical Corporation, NICU\Women's Wellness and Research Center, Doha, Qatar
| | - L O Abdullah
- Neonatology Department, Hamad Medical Corporation, NICU\Women's Wellness and Research Center, Doha, Qatar
| | - N Mohammed
- Neonatology Department, Hamad Medical Corporation, NICU\Women's Wellness and Research Center, Doha, Qatar
| | - Mrp Alikunju
- Pharmacy Department, Hamad Medical Corporation, Doha, Qatar
| | - P V Abdul Rouf
- Pharmacy Department, Hamad Medical Corporation, Doha, Qatar
| | - B Thomas
- Pharmacy Department, Hamad Medical Corporation, Doha, Qatar
| | - B Sree
- Pharmacy Department, Hamad Medical Corporation, Doha, Qatar
| | - W M Elkassem
- Pharmacy Department, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
3
|
Cai L, Zhai J, Ji B, Han F, Niu T, Wang L, Wang J. Intranasal diamorphine population pharmacokinetics modeling and simulation in pediatric breakthrough pain. CPT Pharmacometrics Syst Pharmacol 2025; 14:435-447. [PMID: 39949065 PMCID: PMC11919270 DOI: 10.1002/psp4.13186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 05/01/2024] [Accepted: 05/24/2024] [Indexed: 03/20/2025] Open
Abstract
Intranasal diamorphine (IND), approved for managing breakthrough pain in the UK, has been identified as an acceptable alternative offering effective, expedient, and less traumatic analgesia for children. However, the current dose regimen in pediatric populations relies on clinical expertise while the pharmacokinetics properties are poorly understood. This study aimed to develop diamorphine population pharmacokinetics (pop-PK) models and simulate the IND dosing in virtual pediatric subjects. An integrated four-compartment pop-PK model with first-order absorption and elimination provided an appropriate fit and characterized publicly available 385 concentration measurements of diamorphine, 6-monoacetylmorphine, and morphine collected from adults. Body weight allometry and renal function maturation (age) were incorporated into the final model, serving as two covariates. The estimated IND relative bioavailability was around 52% compared with intramuscularly injected diamorphine. Using this final model, the morphine plasma concentrations, as the active metabolite for pain relief, were simulated in virtual subjects. The utility of model extrapolation was supported by external verification with acceptable average fold errors of 1.06 ± 0.30 and 0.83 ± 0.07 for morphine maximum concentration and exposures. Meanwhile, the simulated morphine concentration-time profiles could recover the PK profiles observed in children after a single dose of IND. The model-based dosing simulations were therefore assessed in four children age groups to match the therapeutic window of morphine concentrations in steady state (10-20 μg/L). Our study demonstrates that the dose regimen of 0.3 mg/kg loading dose plus 0.1 mg/kg hourly maintenance dose is generally appropriate for multiple pediatric populations with breakthrough pain, in the view of PK.
Collapse
Affiliation(s)
- Lianjin Cai
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Jingchen Zhai
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Beihong Ji
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Fengyang Han
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Taoyu Niu
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Luxuan Wang
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Junmei Wang
- Department of Pharmaceutical Sciences, School of PharmacyUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
4
|
Gastine S, Morse JD, Leung MT, Wong ICK, Howard RF, Harrop E, Liossi C, Standing JF, Jassal SS, Hain RD, Skene S, Oulton K, Law SL, Quek WT, Anderson BJ. Diamorphine pharmacokinetics and conversion factor estimates for intranasal diamorphine in paediatric breakthrough pain:systematic review. BMJ Support Palliat Care 2024; 13:e485-e493. [PMID: 35184039 DOI: 10.1136/bmjspcare-2021-003461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/26/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND Intranasal diamorphine is a potential treatment for breakthrough pain but few paediatric data are available to assist dose estimation. AIM To determine an intranasal diamorphine dose in children through an understanding of pharmacokinetics. DESIGN A systematic review of the literature was undertaken to seek diamorphine pharmacokinetic parameters in neonates, children and adults. Parenteral and enteral diamorphine bioavailability were reviewed with respect to formation of the major metabolite, morphine. Clinical data quantifying equianalgesic effects of diamorphine and morphine were reviewed. REVIEW SOURCES PubMed (1960-2020); EMBASE (1980-2020); IPA (1973-2020) and original human research studies that reported diacetylmorphine and metabolite after any dose or route of administration. RESULTS The systematic review identified 19 studies: 16 in adults and 1 in children and 2 neonatal reports. Details of study participants were extracted. Age ranged from premature neonates to 67 years and weight 1.4-88 kg. Intranasal diamorphine bioavailability was predicted as 50%. The equianalgesic intravenous conversion ratio of morphine:diamorphine was 2:1. There was heterogeneity between pharmacokinetic parameter estimates attributed to routes of administration, lack of size standardisation, methodology and pharmacokinetic analysis. Estimates of the pharmacokinetic parameters clearance and volume of distribution were reduced in neonates. There were insufficient paediatric data to characterise clearance or volume maturation of either diamorphine or its metabolites. CONCLUSIONS We estimate equianalgesic ratios of intravenous morphine:diamorphine 2:1, intravenous morphine:intranasal diamorphine 1:1 and oral morphine:intranasal diamorphine of 1:3. These ratios are based on adult literature, but are reasonable for deciding on an initial dose of 0.1 mg/kg in children 4-13 years.
Collapse
Affiliation(s)
- Silke Gastine
- Great Ormond St Institute of Child Health, University College London, London, UK
| | - James D Morse
- Department of Pharmacology & Clinical Pharmacology, The University of Auckland Faculty of Medical and Health Sciences, Auckland, New Zealand
| | - Miriam Ty Leung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, Hong Kong
| | - Ian Chi Kei Wong
- Research Department of Practice and Policy, University College London School of Pharmacy, London, UK
| | - Richard F Howard
- Department of Anaesthesia and Pain Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | | | - Christina Liossi
- School of Psychology, University of Southampton, Southampton, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Joseph F Standing
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- Department of Immunity and Inflammation, University College London, London, UK
| | - Satbir Singh Jassal
- Palliative Care, Rainbows Hospice for Children and Young Adults, Loughborough, UK
| | - Richard D Hain
- All-Wales Managed Clinical Network in Paediatric Palliative Medicine, Cardiff and Vale University Health Board, Cardiff, UK
| | - Simon Skene
- Faculty of Arts and Human Sciences, Surrey Clinical Trials Unit, University of Surrey, Guildford, UK
| | - Kate Oulton
- Centre for Outcomes and Experience Research in Children's Health, Illness and Disability, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Siew L Law
- Research Department of Practice and Policy, University College London School of Pharmacy, London, UK
| | - Wan T Quek
- Research Department of Practice and Policy, University College London School of Pharmacy, London, UK
| | - Brian J Anderson
- Department of Anaesthesiology, The University of Auckland Faculty of Medical and Health Sciences, Auckland, New Zealand
| |
Collapse
|
5
|
Kinoshita M, Borges do Nascimento IJ, Styrmisdóttir L, Bruschettini M. Systemic opioid regimens for postoperative pain in neonates. Cochrane Database Syst Rev 2023; 4:CD015016. [PMID: 37018131 PMCID: PMC10075508 DOI: 10.1002/14651858.cd015016.pub3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
BACKGROUND Postoperative pain clinical management in neonates has always been a challenging medical issue. Worldwide, several systemic opioid regimens are available for pediatricians, neonatologists, and general practitioners to control pain in neonates undergoing surgical procedures. However, the most effective and safe regimen is still unknown in the current body of literature. OBJECTIVES To determine the effects of different regimens of systemic opioid analgesics in neonates submitted to surgery on all-cause mortality, pain, and significant neurodevelopmental disability. Potentially assessed regimens might include: different doses of the same opioid, different routes of administration of the same opioid, continuous infusion versus bolus administration, or 'as needed' administration versus 'as scheduled' administration. SEARCH METHODS Searches were conducted in June 2022 using the following databases: Cochrane Central Register of Controlled Trials [CENTRAL], PubMed, and CINAHL. Trial registration records were identified via CENTRAL and an independent search of the ISRCTN registry. SELECTION CRITERIA We included randomized controlled trials (RCTs), quasi-randomized, cluster-randomized, and cross-over controlled trials evaluating systemic opioid regimens' effects on postoperative pain in neonates (pre-term or full-term). We considered suitable for inclusion: I) studies evaluating different doses of the same opioid; 2) studies evaluating different routes of administration of the same opioid; 3) studies evaluating the effectiveness of continuous infusion versus bolus infusion; and 4) studies establishing an assessment of an 'as needed' administration versus 'as scheduled' administration. DATA COLLECTION AND ANALYSIS According to Cochrane methods, two investigators independently screened retrieved records, extracted data, and appraised the risk of bias. We stratified meta-analysis by the type of intervention: studies evaluating the use of opioids for postoperative pain in neonates through continuous infusion versus bolus infusion and studies assessing the 'as needed' administration versus 'as scheduled' administration. We used the fixed-effect model with risk ratio (RR) for dichotomous data and mean difference (MD), standardized mean difference (SMD), median, and interquartile range (IQR) for continuous data. Finally, we used the GRADEpro approach for primary outcomes to evaluate the quality of the evidence across included studies. MAIN RESULTS In this review, we included seven randomized controlled clinical trials (504 infants) from 1996 to 2020. We identified no studies comparing different doses of the same opioid, or different routes. The administration of continuous opioid infusion versus bolus administration of opioids was evaluated in six studies, while one study compared 'as needed' versus 'as scheduled' administration of morphine given by parents or nurses. Overall, the effectiveness of continuous infusion of opioids over bolus infusion as measured by the visual analog scale (MD 0.00, 95% confidence interval (CI) -0.23 to 0.23; 133 participants, 2 studies; I² = 0); or using the COMFORT scale (MD -0.07, 95% CI -0.89 to 0.75; 133 participants, 2 studies; I² = 0), remains unclear due to study designs' limitations, such as the unclear risk of attrition, reporting bias, and imprecision among reported results (very low certainty of the evidence). None of the included studies reported data on other clinically important outcomes such as all-cause mortality rate during hospitalization, major neurodevelopmental disability, the incidence of severe retinopathy of prematurity or intraventricular hemorrhage, and cognitive- and educational-related outcomes. AUTHORS' CONCLUSIONS: Limited evidence is available on continuous infusion compared to intermittent boluses of systemic opioids. We are uncertain whether continuous opioid infusion reduces pain compared with intermittent opioid boluses; none of the studies reported the other primary outcomes of this review, i.e. all-cause mortality during initial hospitalization, significant neurodevelopmental disability, or cognitive and educational outcomes among children older than five years old. Only one small study reported on morphine infusion with parent- or nurse-controlled analgesia.
Collapse
Affiliation(s)
- Mari Kinoshita
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
- Fetal Medicine Research Center, University of Barcelona, Barcelona, Spain
| | - Israel Junior Borges do Nascimento
- School of Medicine and University Hospital, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Matteo Bruschettini
- Paediatrics, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Department of Research and Education, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
6
|
Kinoshita M, Borges do Nascimento IJ, Styrmisdóttir L, Bruschettini M. Systemic opioid regimens for postoperative pain in neonates. Cochrane Database Syst Rev 2023; 1:CD015016. [PMID: 36645224 PMCID: PMC9841767 DOI: 10.1002/14651858.cd015016.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Postoperative pain clinical management in neonates has always been a challenging medical issue. Worldwide, several systemic opioid regimens are available for pediatricians, neonatologists, and general practitioners to control pain in neonates undergoing surgical procedures. However, the most effective and safe regimen is still unknown in the current body of literature. OBJECTIVES To determine the effects of different regimens of systemic opioid analgesics in neonates submitted to surgery on all-cause mortality, pain, and significant neurodevelopmental disability. Potentially assessed regimens might include: different doses of the same opioid, different routes of administration of the same opioid, continuous infusion versus bolus administration, or 'as needed' administration versus 'as scheduled' administration. SEARCH METHODS Searches were conducted in June 2022 using the following databases: Cochrane Central Register of Controlled Trials [CENTRAL], PubMed, and CINAHL. Trial registration records were identified via CENTRAL and an independent search of the ISRCTN registry. SELECTION CRITERIA We included randomized controlled trials (RCTs), quasi-randomized, cluster-randomized, and cross-over controlled trials evaluating systemic opioid regimens' effects on postoperative pain in neonates (pre-term or full-term). We considered suitable for inclusion: I) studies evaluating different doses of the same opioid; 2) studies evaluating different routes of administration of the same opioid; 3) studies evaluating the effectiveness of continuous infusion versus bolus infusion; and 4) studies establishing an assessment of an 'as needed' administration versus 'as scheduled' administration. DATA COLLECTION AND ANALYSIS According to Cochrane methods, two investigators independently screened retrieved records, extracted data, and appraised the risk of bias. We stratified meta-analysis by the type of intervention: studies evaluating the use of opioids for postoperative pain in neonates through continuous infusion versus bolus infusion and studies assessing the 'as needed' administration versus 'as scheduled' administration. We used the fixed-effect model with risk ratio (RR) for dichotomous data and mean difference (MD), standardized mean difference (SMD), median, and interquartile range (IQR) for continuous data. Finally, we used the GRADEpro approach for primary outcomes to evaluate the quality of the evidence across included studies. MAIN RESULTS In this review, we included seven randomized controlled clinical trials (504 infants) from 1996 to 2020. We identified no studies comparing different doses of the same opioid, or different routes. The administration of continuous opioid infusion versus bolus administration of opioids was evaluated in six studies, while one study compared 'as needed' versus 'as scheduled' administration of morphine given by parents or nurses. Overall, the effectiveness of continuous infusion of opioids over bolus infusion as measured by the visual analog scale (MD 0.00, 95% confidence interval (CI) -0.23 to 0.23; 133 participants, 2 studies; I² = 0); or using the COMFORT scale (MD -0.07, 95% CI -0.89 to 0.75; 133 participants, 2 studies; I² = 0), remains unclear due to study designs' limitations, such as the unclear risk of attrition, reporting bias, and imprecision among reported results (very low certainty of the evidence). None of the included studies reported data on other clinically important outcomes such as all-cause mortality rate during hospitalization, major neurodevelopmental disability, the incidence of severe retinopathy of prematurity or intraventricular hemorrhage, and cognitive- and educational-related outcomes. AUTHORS' CONCLUSIONS: Limited evidence is available on continuous infusion compared to intermittent boluses of systemic opioids. We are uncertain whether continuous opioid infusion reduces pain compared with intermittent opioid boluses; none of the studies reported the other primary outcomes of this review, i.e. all-cause mortality during initial hospitalization, significant neurodevelopmental disability, or cognitive and educational outcomes among children older than five years old. Only one small study reported on morphine infusion with parent- or nurse-controlled analgesia.
Collapse
Affiliation(s)
- Mari Kinoshita
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
- Fetal Medicine Research Center, University of Barcelona, Barcelona, Spain
| | - Israel Junior Borges do Nascimento
- School of Medicine and University Hospital, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
7
|
Morse JD, Anderson BJ, Gastine S, Wong ICK, Standing JF. Pharmacokinetic modeling and simulation to understand diamorphine dose-response in neonates, children, and adolescents. Paediatr Anaesth 2022; 32:716-726. [PMID: 35212432 DOI: 10.1111/pan.14425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/19/2022] [Accepted: 02/21/2022] [Indexed: 11/28/2022]
Abstract
Pharmacokinetic-pharmacodynamic modeling and simulation can facilitate understanding and prediction of exposure-response relationships in children with acute or chronic pain. The pharmacokinetics of diamorphine (diacetylmorphine, heroin), a strong opioid, remain poorly quantified in children and dose is often guided by clinical acumen. This tutorial demonstrates how a model to describe intranasal and intravenous diamorphine pharmacokinetics can be fashioned from a model for diamorphine disposition in adults and a model describing morphine disposition in children. Allometric scaling and maturation models were applied to clearances and volumes to account for differences in size and age between children and adults. The utility of modeling and simulation to gain insight into the analgesic exposure-response relationship is demonstrated. The model explains reported observations, can be used for interrogation, interpolated to determine equianalgesia and inform future clinical studies. Simulation was used to illustrate how diamorphine is rapidly metabolized to morphine via its active metabolite 6-monoacetylmorphine, which mediates an early dopaminergic response accountable for early euphoria. Morphine formation is then responsible for the slower, prolonged analgesic response. Time-concentration profiles of diamorphine and its metabolites reflected disposition changes with age and were used to describe intravenous and intranasal dosing regimens. These indicated that morphine exposure in children after intranasal diamorphine 0.1 mg.kg-1 was similar to that after intranasal diamorphine 5 mg in adults. A target concentration of morphine 30 μg.L-1 can be achieved by a diamorphine intravenous infusion in neonates 14 μg.kg-1 .h-1 , in a 5-year-old child 42 μg.kg-1 .h-1 and in an 15 year-old-adolescent 33 μg.kg-1 .h-1 .
Collapse
Affiliation(s)
- James D Morse
- Department of Pharmacology & Clinical Pharmacology, Auckland University, Auckland, New Zealand
| | - Brian J Anderson
- Department of Anaesthesiology, University of Auckland, Auckland, New Zealand
| | - Silke Gastine
- Infection, Immunity, and Inflammation, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Ian C K Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Joseph F Standing
- Infection, Immunity, and Inflammation, Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
8
|
Postoperative breakthrough pain in paediatric cardiac surgery not reduced by increased morphine concentrations. Pediatr Res 2021; 90:1201-1206. [PMID: 33603216 DOI: 10.1038/s41390-021-01383-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/13/2020] [Accepted: 12/22/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Morphine is commonly used for postoperative analgesia in children. Here we studied the pharmacodynamics of morphine in children after cardiac surgery receiving protocolized morphine. METHODS Data on morphine rescue requirements guided by validated pain scores in children (n = 35, 3-36 months) after cardiac surgery receiving morphine as loading dose (100 μg kg-1) with continuous infusion (40 μg kg-1 h-1) from a previous study on morphine pharmacokinetics were analysed using repeated time-to-event (RTTE) modelling. RESULTS During the postoperative period (38 h (IQR 23-46)), 130 morphine rescue events (4 (IQR 1-5) per patient) mainly occurred in the first 24 h (107/130) at a median morphine concentration of 29.5 ng ml-1 (range 7-180 ng ml-1). In the RTTE model, the hazard of rescue morphine decreased over time (half-life 18 h; P < 0.001), while the hazard for rescue morphine (21.9% at 29.5 ng ml-1) increased at higher morphine concentrations (P < 0.001). CONCLUSIONS In this study on protocolized morphine analgesia in children, rescue morphine was required at a wide range of morphine concentrations and further increase of the morphine concentration did not lead to a decrease in hazard. Future studies should focus on a multimodal approach using other opioids or other analgesics to treat breakthrough pain in children. IMPACT In children receiving continuous morphine infusion, administration of rescue morphine is an indicator for insufficient effect or an event. Morphine rescue events were identified at a wide range of morphine concentrations upon a standardized pain protocol consisting of continuous morphine infusion and morphine as rescue boluses. The expected number of rescue morphine events was found to increase at higher morphine concentrations. Instead of exploring more aggressive morphine dosing, future research should focus on a multimodal approach to treat breakthrough pain in children.
Collapse
|
9
|
Model-Informed Bayesian Estimation Improves the Prediction of Morphine Exposure in Neonates and Infants. Ther Drug Monit 2021; 42:778-786. [PMID: 32427759 DOI: 10.1097/ftd.0000000000000763] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Pain control in infants is an important clinical concern, with potential long-term adverse neurodevelopmental effects. Intravenous morphine is routinely administered for postoperative pain management; however, its dose-concentration-response relationship in neonates and infants has not been well characterized. Although the current literature provides dosing guidelines for the average infant, it fails to control for the large unexplained variability in morphine clearance and response in individual patients. Bayesian estimation can be used to control for some of this variability. The authors aimed to evaluate morphine pharmacokinetics (PKs) and exposure in critically ill neonates and infants receiving standard-of-care morphine therapy and compare a population-based approach to the model-informed Bayesian techniques. METHODS The PKs and exposure of morphine and its active metabolites were evaluated in a prospective opportunistic PK study using 221 discarded blood samples from 57 critically ill neonates and infants in the neonatal intensive care unit. Thereafter, a population-based PK model was compared with a Bayesian adaptive control strategy to predict an individual's PK profile and morphine exposure over time. RESULTS Among the critically ill neonates and infants, morphine clearance showed substantial variability with a 40-fold range (ie, 2.2 to 87.1, mean 23.7 L/h/70 kg). Compared with the observed morphine concentrations, the population-model based predictions had an R of 0.13, whereas the model-based Bayesian predictions had an R of 0.61. CONCLUSIONS Model-informed Bayesian estimation is a better predictor of morphine exposure than PK models alone in critically ill neonates and infants. A large variability was also identified in morphine clearance. A further study is warranted to elucidate the predictive covariates and precision dosing strategies that use morphine concentration and pain scores as feedbacks.
Collapse
|
10
|
Chapron BD, Chapron A, Leeder JS. Recent advances in the ontogeny of drug disposition. Br J Clin Pharmacol 2021; 88:4267-4284. [PMID: 33733546 DOI: 10.1111/bcp.14821] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 02/12/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Developmental changes that occur throughout childhood have long been known to impact drug disposition. However, pharmacokinetic studies in the paediatric population have historically been limited due to ethical concerns arising from incorporating children into clinical trials. As such, much of the early work in the field of developmental pharmacology was reliant on difficult-to-interpret in vitro and in vivo animal studies. Over the last 2 decades, our understanding of the mechanistic processes underlying age-related changes in drug disposition has advanced considerably. Progress has largely been driven by technological advances in mass spectrometry-based methods for quantifying proteins implicated in drug disposition, and in silico tools that leverage these data to predict age-related changes in pharmacokinetics. This review summarizes our current understanding of the impact of childhood development on drug disposition, particularly focusing on research of the past 20 years, but also highlighting select examples of earlier foundational research. Equally important to the studies reviewed herein are the areas that we cannot currently describe due to the lack of research evidence; these gaps provide a map of drug disposition pathways for which developmental trends still need to be characterized.
Collapse
Affiliation(s)
- Brian D Chapron
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA
| | - Alenka Chapron
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA
| | - J Steven Leeder
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, USA.,Schools of Medicine and Pharmacy, University of Missouri-Kansas City, MO, USA
| |
Collapse
|
11
|
Abstract
Neuraxial (spinal and epidural) anesthesia has become commonplace in the care of neonates undergoing surgical procedures. These techniques afford many benefits, and, when properly performed, are extremely safe. This article reviews the benefits, risks, and applications of neuraxial anesthesia in neonates.
Collapse
|
12
|
Tang F, Bada H, Ng CM, Leggas M. Validation of a HPLC/MS method for simultaneous quantification of clonidine, morphine and its metabolites in human plasma. Biomed Chromatogr 2019; 33:e4527. [PMID: 30830964 DOI: 10.1002/bmc.4527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/20/2019] [Accepted: 02/26/2019] [Indexed: 11/08/2022]
Abstract
A high-performance liquid chromatography-tandem mass spectrometry method was developed and validated for the simultaneous quantification of morphine, morphine's major metabolites morphine-3-glucuronide and morphine-6-glucuronide, and clonidine, to support the pharmacokinetic analysis of an ongoing double-blinded randomized clinical trial that compares the use of morphine and clonidine in infants diagnosed with neonatal abstinence syndrome. Plasma samples were processed by solid-phase extraction and separated on an Inertsil ODS-3 (4 μm) column using an 0.1% formic acid in water-0.1% formic acid in methanol gradient. Detection of the analytes was conducted in the positive multiple reaction monitoring mode. The range of quantitation was 1-1000 ng/mL for morphine, morphine-3-glucuronide and morphine-6-glucuronide, and 0.25-100 ng/mL for clonidine. Intra-day and inter-day accuracy and precision were ≤15% for all analytes across the quantitation range. Extraction recovery rates were ≥94% for morphine, ≥90% for M3G, ≥87% for M6G and ≥ 79% for clonidine. Matrix effect ranged from 85-94% for clonidine to 101-106% for M3G. The method fulfilled all predetermined acceptance criteria and required only 100 μL of starting plasma volume. Furthermore, it was successfully applied to 30 clinical trial plasma samples.
Collapse
Affiliation(s)
- Fei Tang
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA.,Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Henrietta Bada
- Department of Pediatrics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Chee M Ng
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Markos Leggas
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA.,Center for Pharmaceutical Research and Innovation, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
13
|
Rodieux F, Piguet V, Desmeules J, Samer CF. Safety Issues of Pharmacological Acute Pain Treatment in Children. Clin Pharmacol Ther 2019; 105:1130-1138. [DOI: 10.1002/cpt.1358] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/16/2018] [Indexed: 02/01/2023]
Affiliation(s)
- Frédérique Rodieux
- Division of Clinical Pharmacology and Toxicology; Department of Anesthesiology, Pharmacology, Intensive Care, and Emergency Medicine; Geneva University Hospitals; Geneva Switzerland
- Multidisciplinary Pain Center; Geneva University Hospitals; Geneva Switzerland
| | - Valérie Piguet
- Division of Clinical Pharmacology and Toxicology; Department of Anesthesiology, Pharmacology, Intensive Care, and Emergency Medicine; Geneva University Hospitals; Geneva Switzerland
- Multidisciplinary Pain Center; Geneva University Hospitals; Geneva Switzerland
| | - Jules Desmeules
- Division of Clinical Pharmacology and Toxicology; Department of Anesthesiology, Pharmacology, Intensive Care, and Emergency Medicine; Geneva University Hospitals; Geneva Switzerland
- Multidisciplinary Pain Center; Geneva University Hospitals; Geneva Switzerland
- Faculty of Medicine; University of Geneva; Geneva Switzerland
- Faculty of Sciences; School of Pharmacy; University of Geneva and Lausanne; Lausanne Switzerland
| | - Caroline F. Samer
- Division of Clinical Pharmacology and Toxicology; Department of Anesthesiology, Pharmacology, Intensive Care, and Emergency Medicine; Geneva University Hospitals; Geneva Switzerland
- Multidisciplinary Pain Center; Geneva University Hospitals; Geneva Switzerland
- Faculty of Medicine; University of Geneva; Geneva Switzerland
| |
Collapse
|
14
|
Favié LMA, Groenendaal F, van den Broek MPH, Rademaker CMA, de Haan TR, van Straaten HLM, Dijk PH, van Heijst A, Dudink J, Dijkman KP, Rijken M, Zonnenberg IA, Cools F, Zecic A, van der Lee JH, Nuytemans DHGM, van Bel F, Egberts TCG, Huitema ADR. Pharmacokinetics of morphine in encephalopathic neonates treated with therapeutic hypothermia. PLoS One 2019; 14:e0211910. [PMID: 30763356 PMCID: PMC6375702 DOI: 10.1371/journal.pone.0211910] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Morphine is a commonly used drug in encephalopathic neonates treated with therapeutic hypothermia after perinatal asphyxia. Pharmacokinetics and optimal dosing of morphine in this population are largely unknown. The objective of this study was to describe pharmacokinetics of morphine and its metabolites morphine-3-glucuronide and morphine-6-glucuronide in encephalopathic neonates treated with therapeutic hypothermia and to develop pharmacokinetics based dosing guidelines for this population. STUDY DESIGN Term and near-term encephalopathic neonates treated with therapeutic hypothermia and receiving morphine were included in two multicenter cohort studies between 2008-2010 (SHIVER) and 2010-2014 (PharmaCool). Data were collected during hypothermia and rewarming, including blood samples for quantification of morphine and its metabolites. Parental informed consent was obtained for all participants. RESULTS 244 patients (GA mean (sd) 39.8 (1.6) weeks, BW mean (sd) 3,428 (613) g, male 61.5%) were included. Morphine clearance was reduced under hypothermia (33.5°C) by 6.89%/°C (95% CI 5.37%/°C- 8.41%/°C, p<0.001) and metabolite clearance by 4.91%/°C (95% CI 3.53%/°C- 6.22%/°C, p<0.001) compared to normothermia (36.5°C). Simulations showed that a loading dose of 50 μg/kg followed by continuous infusion of 5 μg/kg/h resulted in morphine plasma concentrations in the desired range (between 10 and 40 μg/L) during hypothermia. CONCLUSIONS Clearance of morphine and its metabolites in neonates is affected by therapeutic hypothermia. The regimen suggested by the simulations will be sufficient in the majority of patients. However, due to the large interpatient variability a higher dose might be necessary in individual patients to achieve the desired effect. TRIAL REGISTRATION www.trialregister.nl NTR2529.
Collapse
Affiliation(s)
- Laurent M. A. Favié
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Floris Groenendaal
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marcel P. H. van den Broek
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - Carin M. A. Rademaker
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Timo R. de Haan
- Department of Neonatology, Emma Children’s Hospital, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Peter H. Dijk
- Department of Neonatology, Groningen University Medical Centre, Groningen, the Netherlands
| | - Arno van Heijst
- Department of Neonatology, Radboud university medical center-Amalia Children’s Hospital, Nijmegen, the Netherlands
| | - Jeroen Dudink
- Department of Pediatrics, Division of Neonatology, Erasmus Medical Centre-Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Koen P. Dijkman
- Department of Neonatology, Máxima Medical Center Veldhoven, Veldhoven, the Netherlands
| | - Monique Rijken
- Department of Neonatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Inge A. Zonnenberg
- Department of Neonatology, VU University Medical Center, Amsterdam, the Netherlands
| | - Filip Cools
- Department of Neonatology, UZ Brussel—Vrije Universiteit Brussel, Brussels, Belgium
| | - Alexandra Zecic
- Department of Neonatology, University Hospital Gent, Gent, Belgium
| | - Johanna H. van der Lee
- Paediatric Clinical Research Office, Emma Children’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Frank van Bel
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Toine C. G. Egberts
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Alwin D. R. Huitema
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | |
Collapse
|
15
|
Hannam JA, Anderson BJ. Contribution of Morphine and Morphine-6-Glucuronide to Respiratory Depression in a Child. Anaesth Intensive Care 2019; 40:867-70. [DOI: 10.1177/0310057x1204000516] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- J. A. Hannam
- Department of Anaesthesiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - B. J. Anderson
- Department of Anaesthesiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
16
|
Anderson BJ, Lerman J, Coté CJ. Pharmacokinetics and Pharmacology of Drugs Used in Children. A PRACTICE OF ANESTHESIA FOR INFANTS AND CHILDREN 2019:100-176.e45. [DOI: 10.1016/b978-0-323-42974-0.00007-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
17
|
Rodieux F, Gotta V, Pfister M, van den Anker JN. Causes and Consequences of Variability in Drug Transporter Activity in Pediatric Drug Therapy. J Clin Pharmacol 2017; 56 Suppl 7:S173-92. [PMID: 27385174 DOI: 10.1002/jcph.721] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/26/2016] [Accepted: 02/11/2016] [Indexed: 01/06/2023]
Abstract
Drug transporters play a key role in mediating the uptake of endo- and exogenous substances into cells as well as their efflux. Therefore, variability in drug transporter activity can influence pharmaco- and toxicokinetics and be a determinant of drug safety and efficacy. In children, particularly in neonates and young infants, the contribution of tissue-specific drug transporters to drug absorption, distribution, and excretion may differ from that in adults. In this review 5 major factors and their interdependence that may influence drug transporter activity in children are discussed: developmental differences, genetic polymorphisms, pediatric comorbidities, interacting comedication, and environmental factors. Even if data are sparse, altered drug transporter activity due to those factors have been associated with clinically relevant differences in drug disposition, efficacy, and safety in pediatric patients. Single nucleotide polymorphisms in drug transporter-encoding genes were the most studied source of drug transporter variability in children. However, in the age group where drug transporter activity has been reported to differ from that in adults, namely neonates and young infants, hardly any studies have been performed. Longitudinal studies in this young population are required to investigate the age- and disease-dependent genotype-phenotype relationships and relevance of drug transporter drug-drug interactions. Physiologically based pharmacokinetic modeling approaches can integrate drug- and patient-specific parameters, including drug transporter ontogeny, and may further improve in silico predictions of pediatric-specific pharmacokinetics.
Collapse
Affiliation(s)
- Frédérique Rodieux
- Pediatric Pharmacology, University of Basel Children's Hospital (UKBB), Basel, Switzerland
| | - Verena Gotta
- Pediatric Pharmacology, University of Basel Children's Hospital (UKBB), Basel, Switzerland
| | - Marc Pfister
- Pediatric Pharmacology, University of Basel Children's Hospital (UKBB), Basel, Switzerland.,Quantitative Solutions/Certara, Menlo Park, CA, USA
| | - Johannes N van den Anker
- Pediatric Pharmacology, University of Basel Children's Hospital (UKBB), Basel, Switzerland.,Division of Pediatric Clinical Pharmacology, Children's National Health System, Washington, DC, USA.,Intensive Care and Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
18
|
Predicting postoperative morphine consumption in children. Anaesth Crit Care Pain Med 2017; 36:179-184. [DOI: 10.1016/j.accpm.2016.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 08/23/2016] [Accepted: 08/30/2016] [Indexed: 11/21/2022]
|
19
|
Abstract
Pain management in the neonatal ICU remains challenging for many clinicians and in many complex care circumstances. The authors review general pain management principles and address the use of pain scales, non-pharmacologic management, and various agents that may be useful in general neonatal practice, procedurally, or at the end of life. Chronic pain and neonatal abstinence are also noted.
Collapse
Affiliation(s)
- Brian S Carter
- Department of Pediatrics, Division of Neonatology, University of Missouri at Kansas City School of Medicine, 2401 Gillham Rd, Kansas City, MO 64108; Children׳s Mercy Bioethics Center, Kansas City, MO.
| | - Jessica Brunkhorst
- Department of Pediatrics, Division of Neonatology, University of Missouri at Kansas City School of Medicine, 2401 Gillham Rd, Kansas City, MO 64108
| |
Collapse
|
20
|
Martin LD, Jimenez N, Lynn AM. A review of perioperative anesthesia and analgesia for infants: updates and trends to watch. F1000Res 2017; 6:120. [PMID: 28232869 PMCID: PMC5302152 DOI: 10.12688/f1000research.10272.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2017] [Indexed: 01/13/2023] Open
Abstract
This review focuses on pharmacokinetics and pharmacodynamics of opioid and non-opioid analgesics in neonates and infants. The unique physiology of this population differs from that of adults and impacts drug handling. Morphine and remifentanil are described as examples of older versus recently developed opiates to compare and contrast pharmacokinetics and pharmacodynamics in infants. Exploration of genetics affecting both pharmacokinetics and pharmacodynamics of opiates is an area of active research, as is the investigation of a new class of mu-opiate-binding agents which seem selective for analgesic pathways while having less activity in pathways linked to side effects. The kinetics of acetaminophen and of ketorolac as examples of parenteral non-steroidal analgesics in infants are also discussed. The growth in regional anesthesia for peri-operative analgesia in infants can fill an important role minimizing intra-operative anesthetic exposure to opioids and transitioning to post-operative care. Use of multi-modal techniques is recommended to decrease undesirable opiate-related side effects in this vulnerable population.
Collapse
Affiliation(s)
- Lizabeth D Martin
- University of Washington School of Medicine, Department of Anesthesiology & Pain Medicine, Seattle Children’s Hospital, Seattle, WA, USA
| | - Nathalia Jimenez
- University of Washington School of Medicine, Department of Anesthesiology & Pain Medicine, Seattle Children’s Hospital, Seattle, WA, USA
| | - Anne M Lynn
- University of Washington School of Medicine, Department of Anesthesiology & Pain Medicine, Seattle Children’s Hospital, Seattle, WA, USA
| |
Collapse
|
21
|
Knøsgaard KR, Foster DJR, Kreilgaard M, Sverrisdóttir E, Upton RN, van den Anker JN. Pharmacokinetic models of morphine and its metabolites in neonates:: Systematic comparisons of models from the literature, and development of a new meta-model. Eur J Pharm Sci 2016; 92:117-30. [PMID: 27373670 DOI: 10.1016/j.ejps.2016.06.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 11/19/2022]
Abstract
Morphine is commonly used for pain management in preterm neonates. The aims of this study were to compare published models of neonatal pharmacokinetics of morphine and its metabolites with a new dataset, and to combine the characteristics of the best predictive models to design a meta-model for morphine and its metabolites in preterm neonates. Moreover, the concentration-analgesia relationship for morphine in this clinical setting was also investigated. A population of 30 preterm neonates (gestational age: 23-32weeks) received a loading dose of morphine (50-100μg/kg), followed by a continuous infusion (5-10μg/kg/h) until analgesia was no longer required. Pain was assessed using the Premature Infant Pain Profile. Five published population models were compared using numerical and graphical tests of goodness-of-fit and predictive performance. Population modelling was conducted using NONMEM® and the $PRIOR subroutine to describe the time-course of plasma concentrations of morphine, morphine-3-glucuronide, and morphine-6-glucuronide, and the concentration-analgesia relationship for morphine. No published model adequately described morphine concentrations in this new dataset. Previously published population pharmacokinetic models of morphine, morphine-3-glucuronide, and morphine-6-glucuronide were combined into a meta-model. The meta-model provided an adequate description of the time-course of morphine and the concentrations of its metabolites in preterm neonates. Allometric weight scaling was applied to all clearance and volume terms. Maturation of morphine clearance was described as a function of postmenstrual age, while maturation of metabolite elimination was described as a function of postnatal age. A clear relationship between morphine concentrations and pain score was not established.
Collapse
Affiliation(s)
- Katrine Rørbæk Knøsgaard
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - David John Richard Foster
- Australian Centre for Pharmacometrics and Sansom Institute, School of Pharmaceutical and Medical Sciences, City East Campus, North Terrace, University of South Australia, Adelaide, SA 5000, Australia
| | - Mads Kreilgaard
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Eva Sverrisdóttir
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Richard Neil Upton
- Australian Centre for Pharmacometrics and Sansom Institute, School of Pharmaceutical and Medical Sciences, City East Campus, North Terrace, University of South Australia, Adelaide, SA 5000, Australia.
| | - Johannes N van den Anker
- Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA; Division of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, Switzerland; Intensive Care and Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
22
|
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
|
23
|
Elkomy MH, Drover DR, Glotzbach KL, Galinkin JL, Frymoyer A, Su F, Hammer GB. Pharmacokinetics of Morphine and Its Metabolites in Infants and Young Children After Congenital Heart Surgery. AAPS JOURNAL 2015; 18:124-33. [PMID: 26349564 DOI: 10.1208/s12248-015-9826-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 08/26/2015] [Indexed: 11/30/2022]
Abstract
The objective of this study was to characterize morphine glucuronidation in infants and children following cardiac surgery for possible treatment individualization in this population. Twenty children aged 3 days to 6 years, admitted to the cardiovascular intensive care unit after congenital heart surgery, received an intravenous (IV) loading dose of morphine (0.15 mg/kg) followed by subsequent intermittent IV bolus doses based on a validated pain scale. Plasma samples were collected over 6 h after the loading dose and randomly after follow-up doses to measure morphine and its major metabolite concentrations. A population pharmacokinetic model was developed with the non-linear mixed effects software NONMEM. Parent disposition was adequately described by a linear two-compartment model. Effect of growth (size and maturation) on morphine parameters was accounted for by allometric body weight-based models. An intermediate compartment with Emax model best characterized glucuronide concentrations. Glomerular filtration rate was identified as a significant predictor of glucuronide formation time delay and maximum concentrations. Clearance of morphine in children with congenital heart disease is comparable to that reported in children without cardiac abnormalities of similar age. Children 1-6 months of age need higher morphine doses per kilogram to achieve an area under concentration-time curve comparable to that in older children. Pediatric patients with renal failure receiving morphine therapy are at increased risk of developing opioid toxicity due to accumulation of morphine metabolites.
Collapse
Affiliation(s)
- Mohammed H Elkomy
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California, 94305-5117, USA.,Department of Pharmaceutics and Industrial Pharmacy, Beni Suef University, Beni Suef, Egypt
| | - David R Drover
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California, 94305-5117, USA.
| | - Kristi L Glotzbach
- Division of Pediatric Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Jeffery L Galinkin
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Adam Frymoyer
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Felice Su
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Gregory B Hammer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California, 94305-5117, USA.,Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
24
|
Abou Elella R, Adalaty H, Koay YN, Mokrusova P, Theresa M, Male B, Francis B, Jarrab C, Al Wadai A. The efficacy of the COMFORT score and pain management protocol in ventilated pediatric patients following cardiac surgery. Int J Pediatr Adolesc Med 2015; 2:123-127. [PMID: 31528681 PMCID: PMC6738520 DOI: 10.1016/j.ijpam.2015.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVES An optimal scoring system for pain assessment in pediatric intensive care is necessary to determine the efficacy of analgesics. We assess the COMFORT scale in postoperative ventilated children and study the effect of pain and sedation protocols on their outcomes. PATIENTS AND METHODS We included postoperative ventilated patients. The unit-based pain management protocol was used. The assessment of the COMFORT and FLACC scales was performed by 2-nurses at 2-h intervals on the day of surgery and at 4-h intervals during the first 2-postoperative days or until the patient was ex-tubated. The patients' outcomes were compared with age-matched and RACHS score matched patients prior to the application of the pain protocol. RESULTS One-hundred-ten prospective patients were included. The mean age and weight was 24 months and 9.8 ± 8.4 kg, respectively. There was a weak, statistically significant correlation between the COMFORT and FLACC scales, with a range of (r = 0.01-0.7). The COMFORT scale demonstrated good internal consistency, with a Cronbach's alpha of 0.75. The mean ventilation days were 1.3 ± 3, with a mean ICU and hospital stay of 5 ± 5 and 10 ± 9 days, respectively. The 110 patients were compared to 50 retrospective matching patients. The prospective group demonstrated statistically less ventilation days, ICU stay time and hospital stay time, with P-values of 0.0004, 0.001 and 0.0003, respectively. CONCLUSION The COMFORT scale is a valuable and reliable pain assessment tool for use in postoperative ventilated pediatric patients. The implementation of a pain and sedation protocol decreased the incidence of withdrawal and the duration of mechanical ventilation as well as ICU and hospital stays.
Collapse
Affiliation(s)
- Raja Abou Elella
- Heart Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Altamimi MI, Choonara I, Sammons H. Inter-individual variation in morphine clearance in children. Eur J Clin Pharmacol 2015; 71:649-655. [PMID: 25845657 PMCID: PMC4430598 DOI: 10.1007/s00228-015-1843-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 03/24/2015] [Indexed: 11/29/2022]
Abstract
Objectives The aim of the study was to determine the extent of inter-individual variation in clearance of intravenous morphine in children and to establish which factors are responsible for this variation. Methods A systematic literature review was performed to identify papers describing the clearance of morphine in children. The following databases were searched: Medline, Embase, International Pharmaceutical Abstracts, CINAHL, and Cochrane library. From the papers, the range in plasma clearance and the coefficient of variation (CV) in plasma clearance were determined. Results Twenty-eight studies were identified. After quality assessment, 20 studies were included. Only 10 studies gave clearance values for individual patients. The majority of the studies were in critically ill patients. Inter-individual variability of morphine clearance was observed in all age groups, but greatest in critically ill neonates (both preterm and term) and infants. In critically ill patients, the CV was 16–9 7 % in preterm neonates, 24–87 % in term neonates, 35 and 134 % in infants, 39 and 55 % in children, and 74 % in adolescents. The CV was 37 and 44 % respectively in non-critically ill neonates and infants. The mean clearance was higher in children (32 and 52 ml min-1 kg-1) than in neonates (2 to 16 ml min-1 kg-1). Conclusions Large inter-individual variation was seen in morphine clearance values in critically ill neonates and infants. Electronic supplementary material The online version of this article (doi:10.1007/s00228-015-1843-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mohammed I Altamimi
- Academic Division of Child Health, University of Nottingham, Derbyshire Children's Hospital, Derby, DE22 3DT, UK.
| | - Imti Choonara
- Academic Division of Child Health, University of Nottingham, Derbyshire Children's Hospital, Derby, DE22 3DT, UK
| | - Helen Sammons
- Academic Division of Child Health, University of Nottingham, Derbyshire Children's Hospital, Derby, DE22 3DT, UK
| |
Collapse
|
26
|
Schnabel A, Thyssen NM, Goeters C, Zheng H, Zahn PK, Van Aken H, Pogatzki-Zahn EM. Age- and Procedure-Specific Differences of Epidural Analgesia in Children—A Database Analysis. PAIN MEDICINE 2015; 16:544-53. [DOI: 10.1111/pme.12633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Abstract
As a standard of care for preterm/term newborns effective pain management may improve their clinical and neurodevelopmental outcomes. Neonatal pain is assessed using context-specific, validated, and objective pain methods, despite the limitations of currently available tools. Therapeutic approaches reducing invasive procedures and using pharmacologic, behavioral, or environmental measures are used to manage neonatal pain. Nonpharmacologic approaches like kangaroo care, facilitated tucking, non-nutritive sucking, sucrose, and others can be used for procedural pain or adjunctive therapy. Local/topical anesthetics, opioids, NSAIDs/acetaminophen and other sedative/anesthetic agents can be incorporated into NICU protocols for managing moderate/severe pain or distress in all newborns.
Collapse
|
28
|
|
29
|
Brasher C, Gafsous B, Dugue S, Thiollier A, Kinderf J, Nivoche Y, Grace R, Dahmani S. Postoperative pain management in children and infants: an update. Paediatr Drugs 2014; 16:129-40. [PMID: 24407716 DOI: 10.1007/s40272-013-0062-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Many factors contribute to suboptimal pain management in children. Current evidence suggests that severe pain in children has significant long-lasting effects, even more so than in adults. In particular, recent evidence suggests a lack of optimal postoperative pain management in children, especially following ambulatory surgery. This review provides simple guidelines for the management of postoperative pain in children. It discusses the long-term effects of severe pain and how to evaluate pain in both healthy and neurologically impaired children, including neonates. Currently available treatment options are discussed with reference to the efficacy and side effects of opioid and non-opioid and regional analgesic techniques. The impact of preoperative anxiety on postoperative pain, and the efficacy of some nonpharmacological techniques such as hypnosis or distraction, are also discussed. Finally, basic organizational strategies are described, aiming to promote safer and more efficient postoperative pain management in children.
Collapse
Affiliation(s)
- Christopher Brasher
- Department of Anesthesiology, Intensive Care, Robert Debré Hospital, 48 Bd Sérurier, 75019, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Czarnecki ML, Hainsworth K, Simpson PM, Arca MJ, Uhing MR, Varadarajan J, Weisman SJ. Is there an alternative to continuous opioid infusion for neonatal pain control? A preliminary report of parent/nurse-controlled analgesia in the neonatal intensive care unit. Paediatr Anaesth 2014; 24:377-85. [PMID: 24417623 PMCID: PMC4331187 DOI: 10.1111/pan.12332] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND Continuous opioid infusion (COI) remains the mainstay of analgesic therapy in the neonatal intensive care unit (NICU). Parent/nurse-controlled analgesia (PNCA) has been accepted as safe and effective for pediatric patients, but few reports include use in neonates. This study sought to compare outcomes of PNCA and COI in postsurgical neonates and young infants. METHODS Twenty infants treated with morphine PNCA were retrospectively compared with 13 infants treated with fentanyl COI in a Midwestern pediatric hospital in the United States. Outcome measures included opioid consumption, pain scores, frequency of adverse events, and subsequent methadone use. RESULTS The PNCA group (median 6.4 μg · kg(-1) · h(-1) morphine equivalents, range 0.0-31.4) received significantly less opioid (P < 0.001) than the COI group (median 40.0 μg · kg(-1) · h(-1) morphine equivalents; range 20.0-153.3), across postoperative days 0-3. Average daily pain scores (based on 0-10 scale) were low for both groups, but median scores differed nonetheless (0.8 PNCA vs 0.3 COI, P < 0.05). There was no significant difference in the frequency of adverse events or methadone use. CONCLUSION Results suggest PNCA may be a feasible and effective alternative to COI for pain management in postsurgical infants in the NICU. Results also suggest PNCA may provide more individualized care for this vulnerable population and in doing so, may potentially reduce opioid consumption; however, more studies are needed.
Collapse
Affiliation(s)
- Michelle L. Czarnecki
- Jane B. Pettit Pain Management Center, Children’s Hospital of Wisconsin, Milwaukee, USA
| | - Keri Hainsworth
- Jane B. Pettit Pain Management Center, Children’s Hospital of Wisconsin, Milwaukee, USA,Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, USA
| | - Pippa M. Simpson
- Department of Quantitative Health Sciences, Medical College of Wisconsin, Milwaukee, USA,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, USA
| | - Marjorie J. Arca
- Department of Surgery, Medical College of Wisconsin, Milwaukee, USA,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, USA
| | - Michael R. Uhing
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, USA,Department of neonatology, Medical College of Wisconsin, Milwaukee, USA
| | - Jaya Varadarajan
- Jane B. Pettit Pain Management Center, Children’s Hospital of Wisconsin, Milwaukee, USA,Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, USA
| | - Steven J. Weisman
- Jane B. Pettit Pain Management Center, Children’s Hospital of Wisconsin, Milwaukee, USA,Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, USA,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, USA
| |
Collapse
|
31
|
Maitra S, Baidya DK, Khanna P, Ray BR, Panda SS, Bajpai M. Acute perioperative pain in neonates: An evidence-based review of neurophysiology and management. ACTA ACUST UNITED AC 2014; 52:30-7. [DOI: 10.1016/j.aat.2014.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 02/10/2014] [Indexed: 10/25/2022]
|
32
|
Anderson BJ, van den Anker J. Why is there no morphine concentration-response curve for acute pain? Paediatr Anaesth 2014; 24:233-8. [PMID: 24467568 DOI: 10.1111/pan.12361] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Brian J Anderson
- Department of Anaesthesiology, University of Auckland, Auckland, New Zealand.
| | | |
Collapse
|
33
|
Krekels EHJ, Tibboel D, de Wildt SN, Ceelie I, Dahan A, van Dijk M, Danhof M, Knibbe CAJ. Evidence-Based Morphine Dosing for Postoperative Neonates and Infants. Clin Pharmacokinet 2014; 53:553-63. [DOI: 10.1007/s40262-014-0135-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
34
|
West N, Nilforushan V, Stinson J, Ansermino JM, Lauder G. Critical incidents related to opioid infusions in children: a five-year review and analysis. Can J Anaesth 2014; 61:312-21. [PMID: 24442987 DOI: 10.1007/s12630-013-0097-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 12/11/2013] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Opioids have a narrow therapeutic index and have the potential to cause significant harm. Developmental and pharmacogenetic factors put children, and especially infants, at increased risk of complications. We performed a retrospective root cause analysis to identify the factors associated with critical incidents in children receiving opioid infusions in a tertiary care children's hospital. METHODS Following institutional ethical approval, we identified potential critical incidents during 2004 to 2009 from patient safety and pharmacy data. Patients' medical charts were reviewed and a timeline of events that occurred before, during, and following each incident was generated. A safety assessment code score was assigned to each incident according to its severity and probability of recurrence, and incidents with a score ≥ 8 were selected for root cause analysis. Root causes were identified and classified, formal causal statements were written, and action plans were recommended. RESULTS One hundred and sixty-six medical charts were reviewed, and 58 of these included one (45/58) or more (13/58) relevant critical incidents. The resulting harms were of minor to moderate severity. Fourteen incidents were submitted for detailed analysis, from which 31 root causes were identified. The most frequent and significant root causes involved defects in pre-printed order sheets, lack of a nursing guidelines for infusions (rate, adjustment, weaning), and inadequate guidelines for monitoring and recording pain, vital signs, and arousal scores. DISCUSSION The root causes of a range of critical incidents have been identified, and these have been used to generate recommendations for improving both patient safety and quality of analgesia for children receiving opioid infusions for acute pain management.
Collapse
Affiliation(s)
- Nicholas West
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | |
Collapse
|
35
|
Kwok CHT, Devonshire IM, Bennett AJ, Hathway GJ. Postnatal maturation of endogenous opioid systems within the periaqueductal grey and spinal dorsal horn of the rat. Pain 2013; 155:168-178. [PMID: 24076162 PMCID: PMC3894430 DOI: 10.1016/j.pain.2013.09.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 09/03/2013] [Accepted: 09/20/2013] [Indexed: 12/31/2022]
Abstract
Significant opioid-dependent changes occur during the fourth postnatal week in supraspinal sites (rostroventral medulla [RVM], periaqueductal grey [PAG]) that are involved in the descending control of spinal excitability via the dorsal horn (DH). Here we report developmentally regulated changes in the opioidergic signalling within the PAG and DH, which further increase our understanding of pain processing during early life. Microinjection of the μ-opioid receptor (MOR) agonist DAMGO (30 ng) into the PAG of Sprague-Dawley rats increased spinal excitability and lowered mechanical threshold to noxious stimuli in postnatal day (P)21 rats, but had inhibitory effects in adults and lacked efficacy in P10 pups. A tonic opioidergic tone within the PAG was revealed in adult rats by intra-PAG microinjection of CTOP (120 ng, MOR antagonist), which lowered mechanical thresholds and increased spinal reflex excitability. Spinal adminstration of DAMGO inhibited spinal excitability in all ages, yet the magnitude of this was greater in younger animals than in adults. The expression of MOR and related peptides were also investigated using TaqMan real-time polymerase chain reaction and immunohistochemistry. We found that pro-opiomelanocortin peaked at P21 in the ventral PAG, and MOR increased significantly in the DH as the animals aged. Enkephalin mRNA transcripts preceded the increase in enkephalin immunoreactive fibres in the superficial dorsal horn from P21 onwards. These results illustrate that profound differences in the endogenous opioidergic signalling system occur throughout postnatal development.
Collapse
Affiliation(s)
- Charlie H T Kwok
- Laboratory of Developmental Nociception, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK FRAME Laboratory, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | | | | | | |
Collapse
|
36
|
Abstract
The use of genetic information to guide medication decisions holds great promise to improve therapeutic outcomes through increased efficacy and reduced adverse events. As in many areas of medicine, pediatric research and clinical implementation in pharmacogenetics lag behind corresponding adult discovery and clinical applications. In adults, genotype-guided clinical decision support for medications such as clopidogrel, warfarin and simvastatin are in use in some medical centers. However, research conducted in pediatric populations demonstrates that the models and practices developed in adults may be inaccurate in children, and some applications lack any pediatric research to guide clinical decisions. To account for additional factors introduced by developmental considerations in pediatric populations and provide pediatric patients with maximal benefit from genotype-guided therapy, the field will need to develop and employ creative solutions. In this article, we detail some concerns about research and clinical implementation of pharmacogenetics in pediatrics, and present potential mechanisms for addressing them.
Collapse
Affiliation(s)
- Sara L Van Driest
- Department of Pediatrics, Vanderbilt University, 2200 Children's Way, Nashville, TN 37232, USA ; The Monroe Carell Jr Children's Hospital at Vanderbilt, 8232 DOT, 2200 Children's Way, Nashville, TN 37232, USA
| | - Tracy L McGregor
- Department of Pediatrics, Vanderbilt University, 2200 Children's Way, Nashville, TN 37232, USA ; The Monroe Carell Jr Children's Hospital at Vanderbilt, 8232 DOT, 2200 Children's Way, Nashville, TN 37232, USA ; The Center for Human Genetics Research, 519 Light Hall, 2215 Garland Avenue, Nashville, TN 37232-0700, USA
| |
Collapse
|
37
|
Abstract
The use of genetic information to guide medication decisions holds great promise to improve therapeutic outcomes through increased efficacy and reduced adverse events. As in many areas of medicine, pediatric research and clinical implementation in pharmacogenetics lag behind corresponding adult discovery and clinical applications. In adults, genotype-guided clinical decision support for medications such as clopidogrel, warfarin and simvastatin are in use in some medical centers. However, research conducted in pediatric populations demonstrates that the models and practices developed in adults may be inaccurate in children, and some applications lack any pediatric research to guide clinical decisions. To account for additional factors introduced by developmental considerations in pediatric populations and provide pediatric patients with maximal benefit from genotype-guided therapy, the field will need to develop and employ creative solutions. In this article, we detail some concerns about research and clinical implementation of pharmacogenetics in pediatrics, and present potential mechanisms for addressing them.
Collapse
Affiliation(s)
- Sara L Van Driest
- Department of Pediatrics, Vanderbilt University, 2200 Children’s Way, Nashville, TN 37232, USA
- The Monroe Carell Jr Children’s Hospital at Vanderbilt, 8232 DOT, 2200 Children’s Way, Nashville, TN 37232, USA
| | - Tracy L McGregor
- Department of Pediatrics, Vanderbilt University, 2200 Children’s Way, Nashville, TN 37232, USA
- The Monroe Carell Jr Children’s Hospital at Vanderbilt, 8232 DOT, 2200 Children’s Way, Nashville, TN 37232, USA
- The Center for Human Genetics Research, 519 Light Hall, 2215 Garland Avenue, Nashville, TN 37232-0700, USA
| |
Collapse
|
38
|
Abstract
The provision of care to the newborn or young infant at the end of life is primarily motivated by concern and compassion. When examining the evidence base for most interventions, it is lacking - but this is not unique to this aspect of neonatal care. Nevertheless, a redirection of care from cure-oriented and life-extending measures to comfort and limitations of life-sustaining technologic interventions requires the neonatologist to apply practical knowledge skillfully and with prudence. Clinicians can acknowledge that patient needs require managing their end-of-life symptoms now; neither these patients nor their families should have to wait for research to catch up to their current needs.
Collapse
Affiliation(s)
- Brian S Carter
- University of Missouri-Kansas City, Bioethics Center & Section of Neonatology, Children's Mercy Hospital, Kansas City, MO, USA.
| | | |
Collapse
|
39
|
Abstract
AIM We performed a retrospective audit of intravenous morphine infusion administered to children in an effort to characterize the relationship between dose and age. METHODS A retrospective audit of morphine infusions was reviewed for a 24-months period and included all children who received continuous intravenous nurse-controlled morphine infusions and patient-controlled analgesia; a population undergoing acute and elective surgical procedures, as well as medical and oncological treatments. The relationship between age and infusion rate was investigated using nonlinear mixed effects models. RESULTS There were 886 children whose data were acceptable for review. Morphine dose increased with age from 9.97 (CV 28%) μg · kg(-1) per h in neonates. The Hill equation with an exponential of 1.5 best described these changes. Morphine rate reached 90% of its mean final rate of 22.5 (CV 167%) μg · kg(-1) per h, observed in teenagers, at approximately 5 years of age. There was considerable uncertainty of this age-morphine rate profile, and the maturation half-life of this profile was 20 months of age (CV 632%). An increase in dosing variability was observed with increasing age. CONCLUSIONS Morphine infusions at steady-state did not mirror clearance maturation in children nursed in our hospital. We suggest that initial infusion rates in children are started at 10 μg · kg(-1) per h in neonates, 15 μg · kg(-1) per h in toddlers and 25 μg · kg(-1) per h in children above the age of 5 years. The large variability associated with infusion rates means that subsequent infusion rates will depend on feedback from pain scores, adjuvant medications and adverse effects.
Collapse
Affiliation(s)
- Jonathan Taylor
- Department of Anaesthesia, Starship Children's Hospital, Auckland, New Zealand
| | | | | |
Collapse
|
40
|
de la Brière F, Wodey E. Analgésie systémique à domicile après chirurgie pédiatrique ambulatoire. ACTA ACUST UNITED AC 2013. [DOI: 10.1016/j.annfar.2012.10.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
41
|
Hunfeld JAM, Passchier J. Participation in medical research; a systematic review of the understanding and experience of children and adolescents. PATIENT EDUCATION AND COUNSELING 2012; 87:268-276. [PMID: 22018733 DOI: 10.1016/j.pec.2011.09.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2011] [Revised: 09/16/2011] [Accepted: 09/18/2011] [Indexed: 05/31/2023]
Abstract
OBJECTIVE To systematically review the state of knowledge regarding the understanding and experience of psychological burden and risk of children and adolescents who participate in medical research. Further to provide recommendations to maximize understanding and minimize the burden and risk. METHODS Studies were selected that focus on children and adolescents as source of information via PubMed and PsycINFO; search terms were medical research or trial or experiment, child or adolescent, non-therapeutic, therapeutic, psychology, consent or understanding, risk, burden or stress. RESULTS Of 413 abstracts identified, ten studies on understanding and eight on the experience of burden and risk met our selection criteria. Overall, understanding of the study purpose was reasonable, but low for the procedure. Understanding increased with age and appeared to be overestimated by the young participants. Research on experience of burden was scarce, but identified invasive procedures and procedures related to sexual development as the most burdensome. Only one study assessed psychological risks. CONCLUSION Systematic evidence should be collected on the various aspects of medical procedures related to the experienced burden and risk. This will be useful in informing children and adolescents, parents and medical ethical committees.
Collapse
|
42
|
Abstract
BACKGROUND Morphine is widely used throughout the human life span. Several pharmacokinetic models have been proposed to predict how morphine clearance changes with weight and age. This study uses a large external data set to evaluate the ability of pharmacokinetic models to predict morphine doses. METHODS A data set of morphine clearance estimates was created from published reports in premature neonates, full-term neonates, infants, children, and adults. This external data set was used to evaluate published models for morphine clearance as well as other models proposed for use in neonates and infants. Morphine clearance predictions were used to predict morphine dose rates to achieve similar target concentrations in all age groups. RESULTS An allometric ¾ power model using weight combined with a sigmoid maturation model using postmenstrual age successfully predicted the morphine dose rate (within 25% of target) in all age groups except infants [predicted dose 30% under target (95% CI, 7-46%)]. Other published models based on empirical allometric scaling all made unacceptable predictions (>100% of target) in at least one age group. CONCLUSIONS Clearance based on empirical allometric scaling predicted unacceptable doses. Theory-based allometric scaling combined with a maturation function has been confirmed by external evaluation to provide a sound basis for describing clearance and predicting morphine doses in humans of all ages.
Collapse
Affiliation(s)
- Nick H G Holford
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand.
| | | | | |
Collapse
|
43
|
Ottenhoff MJ, Dammers R, Kompanje EJO, Tibboel D, de Jong THR. Discomfort and pain in newborns with myelomeningocele: a prospective evaluation. Pediatrics 2012; 129:e741-7. [PMID: 22371456 DOI: 10.1542/peds.2011-1645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE In a worldwide debate on deliberately terminating the lives of newborns, proponents point at newborns with very severe forms of myelomeningocele (MMC) and their assumed suffering, claiming there are no effective means of alleviating their distress. Nevertheless, the degree of discomfort and pain in these newborns has never been assessed in a structured manner. METHODS In a prospective cohort study, 28 consecutive newborns with MMC were included over a 5-year period and were followed up throughout their hospital stay for initial treatment. We created 2 disease severity groups on the basis of the Lorber criteria. The primary outcomes were discomfort and pain, assessed by simultaneously scoring 2 validated scales: the visual analog scale for pain and the Comfort Behavioral Scale for discomfort. These scores were coupled to a validated and evidence-based analgesia algorithm. RESULTS Overall, discomfort related to pain was measured in 3.3% of the scores. This percentage differed little between the preoperative and postoperative periods and did not significantly differ between newborns with less severe MMC and severe MMC (3.9% vs 2.8%; P = .3). The mean dosage of paracetamol was 35 mg/kg per day (95% confidence interval: 32-39); the mean dosage of morphine was 0.9 μg/kg per hour (95% confidence interval: 0.6 -1.2). CONCLUSION Over the length of their hospital stays for initial treatment, all newborns with MMC presented with low levels of discomfort and pain independent of disease severity and time frame.
Collapse
Affiliation(s)
- Myrthe J Ottenhoff
- Department of Neurosurgery, Erasmus MC, Sophia Children’s Hospital, Rotterdam, Netherlands
| | | | | | | | | |
Collapse
|
44
|
Berde CB, Walco GA, Krane EJ, Anand KJS, Aranda JV, Craig KD, Dampier CD, Finkel JC, Grabois M, Johnston C, Lantos J, Lebel A, Maxwell LG, McGrath P, Oberlander TF, Schanberg LE, Stevens B, Taddio A, von Baeyer CL, Yaster M, Zempsky WT. Pediatric analgesic clinical trial designs, measures, and extrapolation: report of an FDA scientific workshop. Pediatrics 2012; 129:354-64. [PMID: 22250028 PMCID: PMC9923552 DOI: 10.1542/peds.2010-3591] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Analgesic trials pose unique scientific, ethical, and practical challenges in pediatrics. Participants in a scientific workshop sponsored by the US Food and Drug Administration developed consensus on aspects of pediatric analgesic clinical trial design. The standard parallel-placebo analgesic trial design commonly used for adults has ethical and practical difficulties in pediatrics, due to the likelihood of subjects experiencing pain for extended periods of time. Immediate-rescue designs using opioid-sparing, rather than pain scores, as a primary outcome measure have been successfully used in pediatric analgesic efficacy trials. These designs maintain some of the scientific benefits of blinding, with some ethical and practical advantages over traditional designs. Preferred outcome measures were recommended for each age group. Acute pain trials are feasible for children undergoing surgery. Pharmacodynamic responses to opioids, local anesthetics, acetaminophen, and nonsteroidal antiinflammatory drugs appear substantially mature by age 2 years. There is currently no clear evidence for analgesic efficacy of acetaminophen or nonsteroidal antiinflammatory drugs in neonates or infants younger than 3 months of age. Small sample designs, including cross-over trials and N of 1 trials, for particular pediatric chronic pain conditions and for studies of pain and irritability in pediatric palliative care should be considered. Pediatric analgesic trials can be improved by using innovative study designs and outcome measures specific for children. Multicenter consortia will help to facilitate adequately powered pediatric analgesic trials.
Collapse
Affiliation(s)
- Charles B. Berde
- Division of Pain Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Children's Hospital, Boston, Boston, Massachusetts;,Harvard Medical School, Boston, Massachusetts;,Address correspondence to Charles Berde, MD, PhD, Division of Pain Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Children’s Hospital, Boston, 333 Longwood Ave, 5th floor, Boston, MA 02115. E-mail:
| | - Gary A. Walco
- Department of Anesthesiology and Pain Medicine, Seattle Children's Hospital, Seattle, Washington;,University of Washington School of Medicine, Seattle, Washington
| | - Elliot J. Krane
- Stanford University School of Medicine, Stanford, California;,Lucile Packard Children's Hospital, Stanford, California
| | - K. J. S. Anand
- Division of Pediatric Critical Care Medicine, Le Bonheur Children's Hospital, Memphis, Tennessee;,University of Tennessee Health Science Center, Memphis, Tennessee
| | - Jacob V. Aranda
- The Children's Hospital of Brooklyn, State University of New York, New York, New York;,Pediatric Pharmacology Research Unit Network, Children's Hospital of Michigan, Detroit, Michigan
| | - Kenneth D. Craig
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carlton D. Dampier
- Emory University School of Medicine, Atlanta, Georgia;,Atlanta Clinical Translational Science Institute, Atlanta, Georgia
| | - Julia C. Finkel
- Department of Anesthesiology George Washington University, Washington, District of Columbia;,Division of Anesthesiology and Pain Medicine, Children's National Medical Center, Washington, District of Columbia
| | - Martin Grabois
- Baylor College of Medicine, Houston, Texas;,University of Texas Health Science Center-Houston, Houston, Texas
| | | | - John Lantos
- Children's Mercy Bioethics Center, Children's Mercy Hospital, Kansas City, Missouri;,University of Missouri–Kansas City, Kansas City, Missouri
| | - Alyssa Lebel
- Division of Pain Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Children's Hospital, Boston, Boston, Massachusetts;,Harvard Medical School, Boston, Massachusetts
| | - Lynne G. Maxwell
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania;,Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Patrick McGrath
- IWK Health Centre, Halifax, Nova Scotia, Canada;,Dalhousie University, Halifax, Nova Scotia, Canada
| | - Timothy F. Oberlander
- Division of Developmental Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada;,BC Children's Hospital, Vancouver, British Columbia, Canada
| | | | - Bonnie Stevens
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Anna Taddio
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Carl L. von Baeyer
- Department of Psychology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Myron Yaster
- Division of Pediatric Anesthesiology, Department of Anesthesiology and Critical Care Medicine, Children's Medical and Surgical Center, The Johns Hopkins Hospital, Baltimore, Maryland; and
| | - William T. Zempsky
- Division of Pain and Palliative Medicine, Connecticut Children's Medical Center, University of Connecticut School of Medicine, Hartford, Connecticut
| |
Collapse
|
45
|
Abstract
Pain in neonates is now well established. Studies of the developmental neurobiology of pain have revealed that pain processing in the immature is very different from that in the mature nervous system. Neonates undergo considerable maturation of peripheral, spinal and supraspinal afferent pain transmission over the early postnatal period but are able to respond to tissue injury with specific behaviour and with autonomic, hormonal and metabolic signs of stress and distress. Opioid analgesia is now widely used in neonates. There is evidence that morphine requirements may be low in the youngest patients. Sensory threshold testing in rat pups has shown that the analgesic potency of systemic morphine mechanical stimulation is significantly greater in the neonate and declines with postnatal age. The changing morphine sensitivity in the postnatal period may be part of a general reorganisation in the structure and function of primary afferent synapses, neurotransmitter/receptor expression and function and excitatory and inhibitory modulation from higher brain centres. Importantly opioid receptor expression undergoes significant developmental regulation - mu opioid receptors, observed to be exuberantly expressed in the neonatal rat, have been found to be functional. These findings have important implications for the human neonate as they provide a possible explanation for the differences in morphine requirements observed in the youngest patients. The study of the underlying mechanisms of pain and analgesia in development has enabled important changes in clinical practice. However, pain in the newborn remains poorly understood and continued research and intensive study in this area is essential for further effective analgesic intervention and the discovery of new targets for therapy.
Collapse
Affiliation(s)
- R Nandi
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | |
Collapse
|
46
|
Ceelie I, Wildt S, Jong M, Ista E, Tibboel D, Dijk M. Protocolized post-operative pain management in infants; do we stick to it? Eur J Pain 2011; 16:760-6. [DOI: 10.1002/j.1532-2149.2011.00056.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2011] [Indexed: 11/08/2022]
Affiliation(s)
- I. Ceelie
- Department of Pediatric Surgery & Intensive Care; Erasmus MC - Sophia Children's Hospital; Rotterdam; The Netherlands
| | - S.N. Wildt
- Department of Pediatric Surgery & Intensive Care; Erasmus MC - Sophia Children's Hospital; Rotterdam; The Netherlands
| | - M. Jong
- Department of Pediatric Surgery & Intensive Care; Erasmus MC - Sophia Children's Hospital; Rotterdam; The Netherlands
| | - E. Ista
- Department of Pediatric Surgery & Intensive Care; Erasmus MC - Sophia Children's Hospital; Rotterdam; The Netherlands
| | - D. Tibboel
- Department of Pediatric Surgery & Intensive Care; Erasmus MC - Sophia Children's Hospital; Rotterdam; The Netherlands
| | - M. Dijk
- Department of Pediatric Surgery & Intensive Care; Erasmus MC - Sophia Children's Hospital; Rotterdam; The Netherlands
| |
Collapse
|
47
|
Monitto CL, Kost-Byerly S, White E, Lee CKK, Rudek MA, Thompson C, Yaster M. The optimal dose of prophylactic intravenous naloxone in ameliorating opioid-induced side effects in children receiving intravenous patient-controlled analgesia morphine for moderate to severe pain: a dose finding study. Anesth Analg 2011; 113:834-42. [PMID: 21890885 DOI: 10.1213/ane.0b013e31822c9a44] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Opioid-induced side effects, such as pruritus, nausea, and vomiting are common and may be more debilitating than pain itself. A continuous low-dose naloxone infusion (0.25 μg/kg/h) ameliorates some of these side effects in many but not all patients without adversely affecting analgesia. We sought to determine the optimal dose of naloxone required to minimize opioid-induced side effects and to measure plasma morphine and naloxone levels in a dose escalation study. METHODS Fifty-nine pediatric patients (24 male/35 female; average age 14.2 ± 2.2 years) experiencing moderate to severe postoperative pain were started on IV patient-controlled analgesia morphine (basal infusion 20 μg/kg/h, demand dose 20 μg/kg, 5 doses/h) and a low-dose naloxone infusion (initial cohort: 0.05 μg/kg/h; subsequent cohorts: 0.10, 0.15, 0.25, 0.40, 0.65, 1, and 1.65 μg/kg/h). If 2 patients developed intolerable nausea, vomiting, or pruritus, the naloxone infusion was increased for subsequent patients. Dose/treatment success occurred when 10 patients had minimal side effects at a naloxone dose. Blood samples were obtained for measurement of plasma morphine and naloxone levels after initiation of the naloxone infusion, processed, stored, and measured by tandem mass spectrometry with electrospray positive ionization. RESULTS The minimum naloxone dose at which patients were successfully treated with a <10% side effect/failure rate was 1 μg/kg/h; cohort size varied between 4 and 11 patients. Naloxone was more effective in preventing pruritus than nausea and vomiting. Concomitant use of supplemental medicines to treat opioid-induced side effects was required at all naloxone infusion rates. Plasma naloxone levels were below the level of assay quantification (0.1 ng/mL) for infusion rates ≤0.15 μg/kg/h. At rates >0.25 μg/kg/h, plasma levels increased linearly with increasing infusion rate. In each dose cohort, patients who failed therapy had comparable or higher plasma naloxone levels than those levels measured in patients who did not fail treatment. Plasma morphine levels ranged between 3.52 and 172 ng/mL, and >90% of levels ranged between 10.2 and 61.6 ng/mL. Plasma morphine levels were comparable between patients who failed therapy and those patients who achieved symptom control. CONCLUSIONS Naloxone infusion rates ≥1 μg/kg/h significantly reduced, but did not eliminate, the incidence of opioid-induced side effects in postoperative pediatric patients receiving IV patient-controlled analgesia morphine. Patients who failed therapy generally had plasma naloxone and morphine levels that were comparable to those who had good symptom relief suggesting that success or failure to ameliorate opioid-induced side effects was unrelated to plasma levels.
Collapse
Affiliation(s)
- Constance L Monitto
- Department of Anesthesiology and Critical Care Medicine, Division of Pediatric Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, 600 North Wolfe St., Blalock 904, Baltimore, MD 21287, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
A comparison of pain measures in newborn infants after cardiac surgery. Pain 2011; 152:1758-1765. [DOI: 10.1016/j.pain.2011.03.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 02/26/2011] [Accepted: 03/15/2011] [Indexed: 11/23/2022]
|
49
|
Baxter AL, Watcha MF, Baxter WV, Leong T, Wyatt MM. Development and validation of a pictorial nausea rating scale for children. Pediatrics 2011; 127:e1542-9. [PMID: 21624874 DOI: 10.1542/peds.2010-1410] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE The lack of a widely used, validated measure limits pediatric nausea management. The goal of this study was to create and validate a pictorial scale with regular incremental levels between scores depicting increasing nausea intensity. METHODS A pictorial nausea scale of 0 to 10 with 6 faces (the Baxter Retching Faces [BARF] scale) was developed in 3 stages. The BARF scale was validated in emergency department patients with vomiting and in healthy patients undergoing day surgery procedures. Patients were presented with visual analog scales for nausea and pain, the pictorial Faces Pain Scale-Revised, and the BARF scale. Patients receiving opioid analgesics or antiemetic agents had their pain and nausea assessed before and 30 minutes after therapy. Spearman's ρ correlation coefficients were calculated. A Wilcoxon matched-pair rank test compared pain and nausea scores before and after antiemetic therapy. RESULTS Thirty oncology patients and 15 nurses participated in the development of the scale, and 127 patients (52, emergency department; 75, day surgery) ages 7 to 18 years participated in the validation. The Spearman ρ correlation coefficient of the first paired BARF and visual analog scale for nausea scores was 0.93. Visual analog scales for nausea and BARF scores were significantly higher in patients requiring antiemetic agents (P = .0001) and decreased significantly after treatment (P = .0002), while posttreatment VAS (P = .20) and FPSR scores (P =.47) for patients receiving only antiemetic agents did not [corrected]. CONCLUSIONS We describe the development of a pictorial scale with beginning evidence of construct validity for a self-report assessment of the severity of pediatric nausea. The scale had convergent and discriminant validity, along with an ability to detect change after treatment.
Collapse
Affiliation(s)
- Amy L Baxter
- Medical College of Georgia, Pediatric Emergency Medicine Associates, Atlanta, Georgia, USA.
| | | | | | | | | |
Collapse
|
50
|
Improved practices for safe administration of intravenous bolus morphine in a pediatric setting. Pain Manag Nurs 2011; 12:146-53. [PMID: 21893303 DOI: 10.1016/j.pmn.2011.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 02/22/2011] [Accepted: 03/07/2011] [Indexed: 11/20/2022]
Abstract
Postoperative pain control is a clinical imperative, for which morphine is a preferred opioid. However, interpatient variability and drug accumulation with repeated doses, as well as medication errors, may result in respiratory arrest with this medication. Early detection of respiratory depression is essential for safe use of morphine, following both initial and repeated doses. A multidisciplinary team contributed to development of an intravenous (IV) bolus morphine monitoring guideline that reflects current knowledge of morphine pharmacokinetics. Monitoring over a 22-week period in a postsurgical unit was then assessed via record review. A total of 270 postsurgical patients received a first dose of IV bolus morphine, with 784 subsequent doses also administered. Complete monitoring (heart rate, respiratory rate, blood pressure, sedation score, oxygen saturation, and pain score) after the morphine bolus was documented at baseline and 10 and 20 minutes for 34%, 30%, and 23%, respectively, of the patients; partial monitoring (respiratory rate and oxygen saturation) was documented for an additional 22%, 15%, and 9% of patients; 43% of subsequent morphine doses were followed with complete monitoring, and an additional 30% with at least partial monitoring. Adherence to the monitoring procedure fluctuated over the study period with no consistent upward or downward trend. A small number of children exhibited a reduced respiratory rate potentially indicating respiratory depression, but no child required antidote or respiratory support. Despite suboptimal guideline adherence, potential signs of respiratory depression were detected that might otherwise have gone unnoticed. This validates the improved guideline and suggests that some incidents may have remained undetected. Front-line staff must be involved to optimize change, champion the initiative, and promote patient safety.
Collapse
|