1
|
Fong J, Lewis J, Lam M, Kesavan K. Developmental Outcomes after Opioid Exposure in the Fetus and Neonate. Neoreviews 2024; 25:e325-e337. [PMID: 38821910 DOI: 10.1542/neo.25-6-e325] [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: 10/25/2023] [Revised: 11/05/2023] [Accepted: 12/06/2023] [Indexed: 06/02/2024]
Abstract
The overall prevalence of opiate use has been increasing, currently affecting approximately 0.6% of the global population and resulting in a significant proportion of infants being born with prenatal opioid exposure. Animal and human models of prenatal opioid exposure demonstrate detrimental effects on brain anatomy as well as neurodevelopment. Less is known about the neurologic sequelae of postnatal opioid exposure in hospitalized infants. In this review, we summarize our current understanding of the impact of prenatal and postnatal opioid exposure on the brain and on neurodevelopment outcomes. We also identify resources and management strategies that may help mitigate neurodevelopmental delays and deficits associated with opioid exposure in this vulnerable population.
Collapse
Affiliation(s)
- Jeanette Fong
- Department of Pediatrics, University of California, Los Angeles, CA
| | - Juanita Lewis
- Department of Pediatrics, Olive View UCLA Medical Center, Sylmar, CA
| | - Melanie Lam
- Department of Pediatrics, University of California, Los Angeles, CA
| | | |
Collapse
|
2
|
Selvanathan T, Zaki P, McLean MA, Au-Young SH, Chau CMY, Chau V, Synnes AR, Ly LG, Kelly E, Grunau RE, Miller SP. Early-life exposure to analgesia and 18-month neurodevelopmental outcomes in very preterm infants. Pediatr Res 2023:10.1038/s41390-023-02536-y. [PMID: 36859445 DOI: 10.1038/s41390-023-02536-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/28/2023] [Accepted: 02/06/2023] [Indexed: 03/03/2023]
Abstract
BACKGROUND We assessed variability of analgesic use across three tertiary neonatal intensive care units (NICUs) accounting for early-life pain, quantified as number of invasive procedures. We also determined whether analgesia exposure modifies associations between early-life pain and neurodevelopment. METHODS Multicenter prospective study of 276 very preterm infants (born <24-32 weeks' gestational age [GA]). Detailed data of number of invasive procedures and duration of analgesia exposure were collected in initial weeks after birth. Eighteen-month neurodevelopmental assessments were completed in 215 children with Bayley Scales for Infant Development-Third edition. RESULTS Multivariable linear regressions revealed significant differences in morphine use across sites, for a given exposure to early-life pain (interaction p < 0.001). Associations between early-life pain and motor scores differed by duration of morphine exposure (interaction p = 0.01); greater early-life pain was associated with poorer motor scores in infants with no or long (>7 days) exposure, but not short exposure (≤7 days). CONCLUSIONS Striking cross-site differences in morphine exposure in very preterm infants are observed even when accounting for early-life pain. Negative associations between greater early-life pain and adverse motor outcomes were attenuated in infants with short morphine exposure. These findings emphasize the need for further studies of optimal analgesic approaches in preterm infants. IMPACT In very preterm neonates, both early-life exposure to pain and analgesia are associated with adverse neurodevelopment and altered brain maturation, with no clear guidelines for neonatal pain management in this population. We found significant cross-site variability in morphine use across three tertiary neonatal intensive care units in Canada. Morphine use modified associations between early-life pain and motor outcomes. In infants with no or long durations of morphine exposure, greater early-life pain was associated with lower motor scores, this relationship was attenuated in those with short morphine exposure. Further trials of optimal treatment approaches with morphine in preterm infants are warranted.
Collapse
Affiliation(s)
- Thiviya Selvanathan
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Pearl Zaki
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Mia A McLean
- Department of Pediatrics, University of British Columbia, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Stephanie H Au-Young
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Cecil M Y Chau
- Department of Pediatrics, University of British Columbia, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Vann Chau
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Anne R Synnes
- Department of Pediatrics, University of British Columbia, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Linh G Ly
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Edmond Kelly
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada.,Department of Pediatrics, Mount Sinai Hospital, Toronto, ON, Canada
| | - Ruth E Grunau
- Department of Pediatrics, University of British Columbia, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,BC Women's Hospital, Vancouver, BC, Canada
| | - Steven P Miller
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada. .,Department of Pediatrics, University of British Columbia, BC Children's Hospital Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
3
|
Zhang M, Alamaw E, Jampachaisri K, Huss M, Pacharinsak C. Effectiveness of two extended-release buprenorphine formulations during postoperative period in neonatal rats. PLoS One 2022; 17:e0276327. [PMID: 36251720 PMCID: PMC9576048 DOI: 10.1371/journal.pone.0276327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
Information on the effectiveness of a new long-lasting buprenorphine formulation, extended-release buprenorphine, in the neonatal rat is very limited. This study compares whether a high dose of extended-release buprenorphine (XR-Hi) attenuates thermal hypersensitivity for a longer period than a low dose of extended-release buprenorphine (XR-Lo) in a neonatal rat incisional pain model. Two experiments were performed. Experiment one: Male and female postnatal day-5 rat pups (n = 38) were randomly assigned to 1 of 4 treatment groups and received a subcutaneous administration of one of the following: 1) 0.9%NaCl (Saline), 0.1 mL; 2) sustained release buprenorphine (Bup-SR), 1 mg/kg; 3) XR-Lo, 0.65 mg/kg; and 4) XR-Hi, 1.3 mg/kg. Pups were anesthetized with sevoflurane in 100% O2 and a 5 mm long skin incision was made over the left lateral thigh and underlying muscle dissected. The skin was closed with surgical tissue glue. Thermal hypersensitivity testing (using a laser diode) and clinical observations were conducted 1 hour (h) prior to surgery and subsequently after 1, 4, 8, 24, 48, 72 h of treatment. Experiment two: The plasma buprenorphine concentration level was evaluated at 1, 4, 8, 24, 48, 72 h on five-day-old rat pups. Plasma buprenorphine concentration for all treatment groups remained above the clinically effective concentration of 1 ng/mL for at least 4 h in the Bup-SR group, 8 h in XR-Lo and 24 h in XR-Hi group with no abnormal clinical observations. This study demonstrates that XR-Hi did not attenuate postoperative thermal hypersensitivity for a longer period than XR-Lo in 5-day-old rats; XR-Hi attenuated postoperative thermal hypersensitivity for up to 4 h while Bup-SR and XR-Lo for at least 8 h in this model.
Collapse
Affiliation(s)
- Mingyun Zhang
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| | - Eden Alamaw
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | | | - Monika Huss
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Cholawat Pacharinsak
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
4
|
Campbell-Yeo M, Eriksson M, Benoit B. Assessment and Management of Pain in Preterm Infants: A Practice Update. CHILDREN (BASEL, SWITZERLAND) 2022; 9:244. [PMID: 35204964 PMCID: PMC8869922 DOI: 10.3390/children9020244] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/20/2022] [Accepted: 02/02/2022] [Indexed: 12/11/2022]
Abstract
Infants born preterm are at a high risk for repeated pain exposure in early life. Despite valid tools to assess pain in non-verbal infants and effective interventions to reduce pain associated with medical procedures required as part of their care, many infants receive little to no pain-relieving interventions. Moreover, parents remain significantly underutilized in provision of pain-relieving interventions, despite the known benefit of their involvement. This narrative review provides an overview of the consequences of early exposure to untreated pain in preterm infants, recommendations for a standardized approach to pain assessment in preterm infants, effectiveness of non-pharmacologic and pharmacologic pain-relieving interventions, and suggestions for greater active engagement of parents in the pain care for their preterm infant.
Collapse
Affiliation(s)
- Marsha Campbell-Yeo
- School of Nursing, Faculty of Health, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Department of Pediatrics, Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
- IWK Health, Halifax, NS B3K 6R8, Canada
| | - Mats Eriksson
- School of Health Sciences, Faculty of Medicine and Health, Örebro University, SE-701 82 Örebro, Sweden;
| | - Britney Benoit
- Rankin School of Nursing, St. Francis Xavier University, Antigonish, NS B2G 2N5, Canada;
| |
Collapse
|
5
|
Pre- and Postnatal Maturation are Important for Fentanyl Exposure in Preterm and Term Newborns: A Pooled Population Pharmacokinetic Study. Clin Pharmacokinet 2021; 61:401-412. [PMID: 34773609 PMCID: PMC8891207 DOI: 10.1007/s40262-021-01076-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2021] [Indexed: 10/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Fentanyl is an opioid commonly used to prevent and treat severe pain in neonates; however, its use is off label and mostly based on bodyweight. Given the limited pharmacokinetic information across the entire neonatal age range, we characterized the pharmacokinetics of fentanyl across preterm and term neonates to individualize dosing. METHODS We pooled data from two previous studies on 164 newborns with a median gestational age of 29.0 weeks (range 23.9-42.3), birthweight of 1055 g (range 390-4245), and postnatal age (PNA) of 1 day (range 0-68). In total, 673 plasma samples upon bolus dosing (69 patients; median dose 2.1 μg/kg, median 2 boluses per patient) or continuous infusions (95 patients; median dose 1.1 μg/kg/h for 30 h) with and without boluses were used for population pharmacokinetic modeling in NONMEM® 7.4. RESULTS Clearance in neonates with birthweight of 2000 and 3000 g was 2.8- and 5.0-fold the clearance in a neonate with birthweight of 1000 g, respectively. Fentanyl clearance at PNA of 7, 14, and 21 days was 2.7-fold, 3.8-fold, and 4.6-fold the clearance at 1 day, respectively. Bodyweight-based dosing resulted in large differences in fentanyl concentrations. Depending on PNA and birthweight, fentanyl concentrations increased slowly after the start of therapy for both intermittent boluses and continuous infusion and reached a maximum concentration at 12-48 h. CONCLUSIONS As both prenatal and postnatal maturation are important for fentanyl exposure, we propose a birthweight- and PNA-based dosage regimen. To provide rapid analgesia in the first 24 h of treatment, additional loading doses need to be considered.
Collapse
|
6
|
Verscheijden LFM, Litjens CHC, Koenderink JB, Mathijssen RHJ, Verbeek MM, de Wildt SN, Russel FGM. Physiologically based pharmacokinetic/pharmacodynamic model for the prediction of morphine brain disposition and analgesia in adults and children. PLoS Comput Biol 2021; 17:e1008786. [PMID: 33661919 PMCID: PMC7963108 DOI: 10.1371/journal.pcbi.1008786] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 03/16/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022] Open
Abstract
Morphine is a widely used opioid analgesic, which shows large differences in clinical response in children, even when aiming for equivalent plasma drug concentrations. Age-dependent brain disposition of morphine could contribute to this variability, as developmental increase in blood-brain barrier (BBB) P-glycoprotein (Pgp) expression has been reported. In addition, age-related pharmacodynamics might also explain the variability in effect. To assess the influence of these processes on morphine effectiveness, a multi-compartment brain physiologically based pharmacokinetic/pharmacodynamic (PB-PK/PD) model was developed in R (Version 3.6.2). Active Pgp-mediated morphine transport was measured in MDCKII-Pgp cells grown on transwell filters and translated by an in vitro-in vivo extrapolation approach, which included developmental Pgp expression. Passive BBB permeability of morphine and its active metabolite morphine-6-glucuronide (M6G) and their pharmacodynamic parameters were derived from experiments reported in literature. Model simulations after single dose morphine were compared with measured and published concentrations of morphine and M6G in plasma, brain extracellular fluid (ECF) and cerebrospinal fluid (CSF), as well as published drug responses in children (1 day– 16 years) and adults. Visual predictive checks indicated acceptable overlays between simulated and measured morphine and M6G concentration-time profiles and prediction errors were between 1 and -1. Incorporation of active Pgp-mediated BBB transport into the PB-PK/PD model resulted in a 1.3-fold reduced brain exposure in adults, indicating only a modest contribution on brain disposition. Analgesic effect-time profiles could be described reasonably well for older children and adults, but were largely underpredicted for neonates. In summary, an age-appropriate morphine PB-PK/PD model was developed for the prediction of brain pharmacokinetics and analgesic effects. In the neonatal population, pharmacodynamic characteristics, but not brain drug disposition, appear to be altered compared to adults and older children, which may explain the reported differences in analgesic effect. Developmental processes in children can affect pharmacokinetics: “what the body does to the drug” as well as pharmacodynamics: “what the drug does to the body”. A typical example is morphine, of which the analgesic response is variable and particularly neonates suffer more often from respiratory depression, even when receiving doses corrected for differences in elimination. One way to mathematically incorporate developmental processes is by employing physiologically based pharmacokinetic/pharmacodynamic (PB-PK/PD) models, where physiological differences between individuals are incorporated. In this study, we developed a morphine PB-PK/PD model to predict brain drug disposition as well as analgesic response in adults and children, as both processes could potentially contribute to developmental variability in the effect of morphine. We found that age-related variation in BBB expression of the main morphine efflux transporter P-glycoprotein was not responsible for differences in brain exposure. In contrast, pharmacodynamic modelling suggested an increased sensitivity to morphine in neonates.
Collapse
Affiliation(s)
- Laurens F. M. Verscheijden
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Carlijn H. C. Litjens
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Jan B. Koenderink
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Ron H. J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Marcel M. Verbeek
- Departments of Neurology and Laboratory Medicine, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, The Netherlands
| | - Saskia N. de Wildt
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- Intensive Care and Department of Paediatric Surgery, Erasmus MC-Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Frans G. M. Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
7
|
Watson E, Khandelwal A, Freijer J, van den Anker J, Lefeber C, Eerdekens M. Population pharmacokinetic modeling to facilitate dose selection of tapentadol in the pediatric population. J Pain Res 2019; 12:2835-2850. [PMID: 31686902 PMCID: PMC6800464 DOI: 10.2147/jpr.s208454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 07/26/2019] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE The main aim of this analysis was to characterize the pharmacokinetics (PK) of the strong analgesic tapentadol in 2-year-old to <18-year-old patients with acute pain and to inform the optimal dosing strategy for a confirmatory efficacy trial in this patient population. METHODS The analysis dataset included tapentadol concentrations obtained from 92 pediatric patients receiving a single tapentadol oral solution (OS) dose of 1.0 mg/kg bodyweight in two single-dose PK clinical trials. Population PK analysis was performed using nonlinear mixed effects modeling. Simulations were performed to identify tapentadol OS doses in pediatric subjects (2 to <18 years) that would produce exposures similar to those in adults receiving safe and efficacious doses of tapentadol IR (50-100 mg every 4 hrs). RESULTS Tapentadol PK in children aged from 2 to <18 years was best described by a one-compartment model. Mean population apparent clearance and apparent volume of distribution for a typical subject weighing 45 kg were 170 L/h and 685 L, respectively. Clearance, expressed in bodyweight units as L/h/kg, decreased with increasing age whereas total clearance (L/h) increased with increasing age. Model-based simulations suggested that a tapentadol OS dose of 1.25 mg/kg to children and adolescents aged 2 to <18 years would result in efficacious tapentadol exposures similar to those in adults receiving tapentadol immediate release 50-100 mg every 4 hrs. The proposed tapentadol OS dose was subsequently applied in a confirmatory efficacy trial in 2 to <18-year-old patients suffering from acute postsurgical pain. CONCLUSION This analysis provides an example of a model-based approach for a dose recommendation to be used in an efficacy trial in the pediatric population. Uniform dosing based on bodyweight was proposed for the treatment of acute pain in children aged from 2 to <18 years.
Collapse
Affiliation(s)
| | | | | | - John van den Anker
- Division of Paediatric Pharmacology and Pharmacometrics, University of Basel Children’s Hospital, Basel, Switzerland
- Division of Clinical Pharmacology, Children’s National Medical Center, Washington, DC, USA
| | | | | |
Collapse
|
8
|
Eerdekens M, Beuter C, Lefeber C, van den Anker J. The challenge of developing pain medications for children: therapeutic needs and future perspectives. J Pain Res 2019; 12:1649-1664. [PMID: 31213880 PMCID: PMC6536714 DOI: 10.2147/jpr.s195788] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/27/2019] [Indexed: 12/11/2022] Open
Abstract
It is broadly accepted that children of all age groups including (preterm) neonates and young infants can perceive pain and that there is an absolute need to treat their pain safely and effectively. The approved treatment options for children, particularly (preterm) neonates and young infants, are very limited with only a few medications specifically labelled for this population. This article presents the challenges of developing pain medications for children. A short overview gives information on pain in children, including pain perception, prevalence of pain and the long-term consequences of leaving pain untreated in this vulnerable population. Current pain management practices are briefly discussed. The challenges of conducting pediatric clinical trials in general and trials involving analgesic medications in particular within the regulatory framework available to develop these medications for children are presented. Emphasis is given to the operational hurdles faced in conducting a pediatric clinical trial program. Some suggestions to overcome these hurdles are provided based on our experience during the pediatric trial program for the strong analgesic tapentadol used for the treatment of moderate to severe acute pain.
Collapse
Affiliation(s)
| | | | | | - John van den Anker
- Division of Paediatric Pharmacology and Pharmacometrics, University of Basel Children’s Hospital, Basel, Switzerland
- Division of Clinical Pharmacology, Children’s National Medical Center, Washington, DC, USA
| |
Collapse
|
9
|
Towards personalized medicine in preterm newborns: Morphine analgesia predicted by genotype. EBioMedicine 2019; 40:41-42. [PMID: 30738831 PMCID: PMC6413580 DOI: 10.1016/j.ebiom.2019.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/03/2019] [Indexed: 11/30/2022] Open
|
10
|
Oliveira C, Scarabelot VL, Vercelino R, Silveira NP, Adachi LN, Regner GG, Silva LS, Macedo IC, Souza A, Caumo W, Torres IL. Morphine exposure and maternal deprivation during the early postnatal period alter neuromotor development and nerve growth factor levels. Int J Dev Neurosci 2017; 63:8-15. [DOI: 10.1016/j.ijdevneu.2017.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 09/02/2017] [Accepted: 09/03/2017] [Indexed: 12/17/2022] Open
Affiliation(s)
- Carla Oliveira
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduaçăo em Medicina: Ciências MédicasFaculdade de MedicinaUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Vanessa L. Scarabelot
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Rafael Vercelino
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Centro Universitário FADERGSPorto AlegreRSBrazil
- Health and Wellness School Laureate International Universities
| | - Natalia P. Silveira
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Lauren N.S. Adachi
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduaçăo em Medicina: Ciências MédicasFaculdade de MedicinaUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Gabriela G. Regner
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Lisiane S. Silva
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Isabel Cristina Macedo
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Universidade Federal do PampaAvenida Antônio Trilha, 184797300‐000São GabrielRSBrazil
| | - Andressa Souza
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Wolnei Caumo
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduaçăo em Medicina: Ciências MédicasFaculdade de MedicinaUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
| | - Iraci L.S. Torres
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré‐ClínicasDepartamento de FarmacologiaInstituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
- Programa de Pós‐Graduaçăo em Medicina: Ciências MédicasFaculdade de MedicinaUniversidade Federal do Rio Grande do SulPorto AlegreBrazil
- Unidade de Experimentação Animal e Grupo de Pesquisa e Pós‐Graduação, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do SulPorto AlegreBrazil
| |
Collapse
|
11
|
Allegaert K, Simons SHP, Tibboel D, Krekels EH, Knibbe CA, van den Anker JN. Non-maturational covariates for dynamic systems pharmacology models in neonates, infants, and children: Filling the gaps beyond developmental pharmacology. Eur J Pharm Sci 2017; 109S:S27-S31. [PMID: 28506866 DOI: 10.1016/j.ejps.2017.05.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 02/07/2023]
Abstract
Pharmacokinetics and -dynamics show important changes throughout childhood. Studies on the different maturational processes that influence developmental pharmacology have been used to create population PK/PD models that can yield individualized pediatric drug dosages. These models were subsequently translated to semi-physiologically or physiology-based PK (PBPK) models that support predictions in pediatric patient cohorts and other special populations. Although these translational efforts are crucial, these models should be further improved towards individual patient predictions by including knowledge on non-maturational covariates. These efforts are needed to ultimately get to systems pharmacology models for children. These models take developmental changes relating to the pediatric dynamical system into account but also other aspects that may be of importance such as abnormal body composition, pharmacogenetics, critical illness and inflammatory status.
Collapse
Affiliation(s)
- Karel Allegaert
- Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Development and Regeneration KU Leuven, Leuven, Belgium
| | - Sinno H P Simons
- Department of Pediatrics, Division of Neonatology, Erasmus MC Sophia Children's Hospital, Rotterdam, the Netherlands.
| | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Elke H Krekels
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, the Netherlands
| | - Catherijne A Knibbe
- Division of Pharmacology, Leiden Academic Center for Drug Research, Leiden, the Netherlands; Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, the Netherlands
| | - John N van den Anker
- Intensive Care and Department of Pediatric Surgery, Erasmus MC Sophia Children's Hospital, Rotterdam, the Netherlands; Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA; Division of Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| |
Collapse
|
12
|
Panahi Y, Saboory E, Rassouli A, Sadeghi‐Hashjin G, Roshan‐Milani S, Derafshpour L, Rasmi Y. The effect of selective opioid receptor agonists and antagonists on epileptiform activity in morphine‐dependent infant mice hippocampal slices. Int J Dev Neurosci 2017; 60:56-62. [DOI: 10.1016/j.ijdevneu.2017.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 04/01/2017] [Accepted: 04/24/2017] [Indexed: 01/13/2023] Open
Affiliation(s)
- Yousef Panahi
- Department of Pharmacology, Faculty of Veterinary MedicineUniversity of TehranTehranIran
| | - Ehsan Saboory
- Neurophysiology Research Center, Urmia University of Medical SciencesUrmiaIran
| | - Ali Rassouli
- Department of Pharmacology, Faculty of Veterinary MedicineUniversity of TehranTehranIran
| | | | - Shiva Roshan‐Milani
- Department of Physiology, Faculty of MedicineUrmia University of Medical SciencesUrmiaIran
| | - Leila Derafshpour
- Neurophysiology Research Center, Urmia University of Medical SciencesUrmiaIran
| | - Yousef Rasmi
- Cellular and Molecular Research Center, Urmia University of Medical SciencesUrmiaIran
| |
Collapse
|
13
|
Baarslag MA, Allegaert K, Van Den Anker JN, Knibbe CAJ, Van Dijk M, Simons SHP, Tibboel D. Paracetamol and morphine for infant and neonatal pain; still a long way to go? Expert Rev Clin Pharmacol 2016; 10:111-126. [PMID: 27785937 DOI: 10.1080/17512433.2017.1254040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Pharmacologic pain management in newborns and infants is often based on limited scientific data. To close the knowledge gap, drug-related research in this population is increasingly supported by the authorities, but remains very challenging. This review summarizes the challenges of analgesic studies in newborns and infants on morphine and paracetamol (acetaminophen). Areas covered: Aspects such as the definition and multimodal character of pain are reflected to newborn infants. Specific problems addressed include defining pharmacodynamic endpoints, performing clinical trials in this population and assessing developmental changes in both pharmacokinetics and pharmacodynamics. Expert commentary: Neonatal and infant pain management research faces two major challenges: lack of clear biomarkers and very heterogeneous pharmacokinetics and pharmacodynamics of analgesics. There is a clear call for integral research addressing the multimodality of pain in this population and further developing population pharmacokinetic models towards physiology-based models.
Collapse
Affiliation(s)
- Manuel A Baarslag
- a Intensive Care and department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands
| | - Karel Allegaert
- a Intensive Care and department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands.,b Department of development and regeneration , KU Leuven , Leuven , Belgium
| | - John N Van Den Anker
- a Intensive Care and department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands.,c Division of Clinical Pharmacology , Children's National Health System , Washington , DC , USA.,d Division of Pediatric Pharmacology and Pharmacometrics , University of Basel Children's Hospital , Basel , Switzerland
| | - Catherijne A J Knibbe
- e Department of Clinical Pharmacy , St. Antonius Hospital , Nieuwegein , The Netherlands.,f Division of Pharmacology, Leiden Academic Center for Drug Research , Leiden University , Leiden , the Netherlands
| | - Monique Van Dijk
- a Intensive Care and department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands.,g Department of Pediatrics, division of Neonatology , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands
| | - Sinno H P Simons
- g Department of Pediatrics, division of Neonatology , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands
| | - Dick Tibboel
- a Intensive Care and department of Pediatric Surgery , Erasmus MC-Sophia Children's Hospital , Rotterdam , the Netherlands
| |
Collapse
|
14
|
Mermet-Joret N, Chatila N, Pereira B, Monconduit L, Dallel R, Antri M. Lamina specific postnatal development of PKCγ interneurons within the rat medullary dorsal horn. Dev Neurobiol 2016; 77:102-119. [PMID: 27346325 DOI: 10.1002/dneu.22414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/21/2016] [Accepted: 06/22/2016] [Indexed: 01/17/2023]
Abstract
Protein kinase C gamma (PKCγ) interneurons, located in the superficial spinal (SDH) and medullary dorsal horns (MDH), have been shown to play a critical role in cutaneous mechanical hypersensitivity. However, a thorough characterization of their development in the MDH is lacking. Here, it is shown that the number of PKCγ-ir interneurons changes from postnatal day 3 (P3) to P60 (adult) and such developmental changes differ according to laminae. PKCγ-ir interneurons are already present at P3-5 in laminae I, IIo, and III. In lamina III, they then decrease from P11-P15 to P60. Interestingly, PKCγ-ir interneurons appear only at P6 in lamina IIi, and they conversely increase to reach adult levels at P11-15. Analysis of neurogenesis using bromodeoxyuridine (BrdU) does not detect any PKCγ-BrdU double-labeling in lamina IIi. Quantification of the neuronal marker, NeuN, reveals a sharp neuronal decline (∼50%) within all superficial MDH laminae during early development (P3-15), suggesting that developmental changes in PKCγ-ir interneurons are independent from those of other neurons. Finally, neonatal capsaicin treatment, which produces a permanent loss of most unmyelinated afferent fibers, has no effect on the development of PKCγ-ir interneurons. Together, the results show that: (i) the expression of PKCγ-ir interneurons in MDH is developmentally regulated with a critical period at P11-P15, (ii) PKCγ-ir interneurons are developmentally heterogeneous, (iii) lamina IIi PKCγ-ir interneurons appear less vulnerable to cell death, and (iv) postnatal maturation of PKCγ-ir interneurons is due to neither neurogenesis, nor neuronal migration, and is independent of C-fiber development. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 102-119, 2017.
Collapse
Affiliation(s)
- Noemie Mermet-Joret
- Neuro-Dol, Clermont Université, Université D'Auvergne, BP 10448, F-63000, Clermont-Ferrand & Inserm U1107, Clermont-Ferrand, F-63100, France
| | - Nadwa Chatila
- Neuro-Dol, Clermont Université, Université D'Auvergne, BP 10448, F-63000, Clermont-Ferrand & Inserm U1107, Clermont-Ferrand, F-63100, France
| | - Bruno Pereira
- Biostatistics Unit (DRCI), CHU Clermont-Ferrand, Clermont-Ferrand, F-63100, France
| | - Lénaic Monconduit
- Neuro-Dol, Clermont Université, Université D'Auvergne, BP 10448, F-63000, Clermont-Ferrand & Inserm U1107, Clermont-Ferrand, F-63100, France
| | - Radhouane Dallel
- Neuro-Dol, Clermont Université, Université D'Auvergne, BP 10448, F-63000, Clermont-Ferrand & Inserm U1107, Clermont-Ferrand, F-63100, France.,Service D'Odontologie, CHU Clermont-Ferrand, Clermont-Ferrand, F-63000, France
| | - Myriam Antri
- Neuro-Dol, Clermont Université, Université D'Auvergne, BP 10448, F-63000, Clermont-Ferrand & Inserm U1107, Clermont-Ferrand, F-63100, France
| |
Collapse
|
15
|
Craig MM, Bajic D. Long-term behavioral effects in a rat model of prolonged postnatal morphine exposure. Behav Neurosci 2015; 129:643-55. [PMID: 26214209 PMCID: PMC4586394 DOI: 10.1037/bne0000081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Prolonged morphine treatment in neonatal pediatric populations is associated with a high incidence of opioid tolerance and dependence. Despite the clinical relevance of this problem, our knowledge of long-term consequences is sparse. The main objective of this study was to investigate whether prolonged morphine administration in a neonatal rat is associated with long-term behavioral changes in adulthood. Newborn animals received either morphine (10 mg/kg) or equal volume of saline subcutaneously twice daily for the first 2 weeks of life. Morphine-treated animals underwent 10 days of morphine weaning to reduce the potential for observable physical signs of withdrawal. Animals were subjected to nonstressful testing (locomotor activity recording and a novel-object recognition test) at a young age (Postnatal Days [PDs] 27-31) or later in adulthood (PDs 55-56), as well as stressful testing (calibrated forceps test, hot plate test, and forced swim test) only in adulthood. Analysis revealed that prolonged neonatal morphine exposure resulted in decreased thermal but not mechanical threshold. Importantly, no differences were found for total locomotor activity (proxy of drug reward/reinforcement behavior), individual forced swim test behaviors (proxy of affective processing), or novel-object recognition test. Performance on the novel-object recognition test was compromised in the morphine-treated group at the young age, but the effect disappeared in adulthood. These novel results provide insight into the long-term consequences of opioid treatment during an early developmental period and suggest long-term neuroplastic differences in sensory processing related to thermal stimuli.
Collapse
Affiliation(s)
- Michael M. Craig
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA, USA
| | - Dusica Bajic
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children’s Hospital, 300 Longwood Ave., Boston, MA, USA
- Department of Anaesthesia, Harvard Medical School, 25 Shattuck St., Boston, MA, USA
| |
Collapse
|
16
|
Interaction of prenatal stress and morphine alters prolactin and seizure in rat pups. Physiol Behav 2015; 149:181-6. [DOI: 10.1016/j.physbeh.2015.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 06/01/2015] [Accepted: 06/03/2015] [Indexed: 11/17/2022]
|
17
|
Schunck RVA, Torres IL, Laste G, de Souza A, Macedo IC, Valle MTC, Salomón JL, Moreira S, Kuo J, Arbo MD, Dallegrave E, Leal MB. Protracted alcohol abstinence induces analgesia in rats: Possible relationships with BDNF and interleukin-10. Pharmacol Biochem Behav 2015; 135:64-9. [DOI: 10.1016/j.pbb.2015.05.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 05/05/2015] [Accepted: 05/11/2015] [Indexed: 11/24/2022]
|
18
|
D'Amato FR. Evaluation of social and nonsocial behaviors mediated by opioids in mouse pups. Methods Mol Biol 2015; 1230:313-322. [PMID: 25293338 DOI: 10.1007/978-1-4939-1708-2_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The experimental approach to carry out a behavioral study involving opioids in mouse pups needs equipments and procedures different from those used for adult animals. Pups are immature at birth and only slowly acquire all the potentialities that characterize adult con-specifics. The standard and abnormal development of behavioral systems and their neural correlates can be followed during the first postnatal weeks, using appropriate methodologies that exploit characteristic pups' capabilities. Behavioral tests designed for pups to evaluate the activity and involvement of the opioid system, according to the well-known role of the system in adult animals, are described in this chapter.
Collapse
Affiliation(s)
- Francesca R D'Amato
- Cell Biology and Neurobiology Institute, CNR/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, Rome, 00143, Italy,
| |
Collapse
|
19
|
Therapeutic touch is not therapeutic for procedural pain in very preterm neonates: a randomized trial. Clin J Pain 2014; 29:824-9. [PMID: 23817594 DOI: 10.1097/ajp.0b013e3182757650] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Preterm neonates below 30 weeks' gestational age undergo numerous painful procedures. Many management approaches are not appropriate for this population. Therapeutic Touch, an alternative approach based on the theory of energy medicine, has been shown to promote physiological stability in preterm neonates and reduce pain in some adult studies. The objective was to determine whether Therapeutic Touch is efficacious in decreasing pain in preterm neonates. METHODS Infants < 30 weeks' gestational age participated in a randomized control trial in 2 level III neonatal intensive care units. All evaluations, analyses, and heel lance procedure were conducted with only the therapist knowing the group assignment. Immediately before and after the heel lance procedure, the therapist performed nontactile Therapeutic Touch (n = 27) with infant behind curtains, leaving the curtained area for the heel lance, performed by another. In the sham condition (n = 28), the therapist stood by the incubator with hands by her side. The Premature Infant Pain Profile was used for pain response and time for heart rate to return to baseline for recovery. Heart rate variability and stress response were secondary outcomes. RESULTS There were no group differences in any of the outcomes. Mean Premature Infant Pain Profile scores across 2 minutes of heel lance procedure in 30-second blocks ranged from 7.92 to 8.98 in the Therapeutic Touch group and 7.64 to 8.46 in the sham group. INTERPRETATION Therapeutic Touch given immediately before and after heel lance has no comforting effect in preterm neonates. Other effective strategies involving actual touch should be considered.
Collapse
|
20
|
Campbell-Yeo M, Johnston C, Benoit B, Latimer M, Vincer M, Walker CD, Streiner D, Inglis D, Caddell K. Trial of repeated analgesia with Kangaroo Mother Care (TRAKC Trial). BMC Pediatr 2013; 13:182. [PMID: 24284002 PMCID: PMC3828622 DOI: 10.1186/1471-2431-13-182] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 10/09/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Skin-to-skin contact (SSC) between mother and infant, commonly referred to as Kangaroo Mother Care (KMC), is recommended as an intervention for procedural pain. Evidence demonstrates its consistent efficacy in reducing pain for a single painful procedure. The purpose of this study is to examine the sustained efficacy of KMC, provided during all routine painful procedures for the duration of Neonatal Intensive Care Unit (NICU) hospitalization, in diminishing behavioral pain response in preterm neonates. The efficacy of KMC alone will be compared to standard care of 24% oral sucrose, as well as the combination of KMC and 24% oral sucrose. METHODS/DESIGN Infants admitted to the NICU who are less than 36 6/7 weeks gestational age (according to early ultrasound), that are stable enough to be held in KMC, will be considered eligible (N = 258). Using a single-blinded randomized parallel group design, participants will be assigned to one of three possible interventions: 1) KMC, 2) combined KMC and sucrose, and 3) sucrose alone, when they undergo any routine painful procedure (heel lance, venipuncture, intravenous, oro/nasogastric insertion). The primary outcome is infant's pain intensity, which will be assessed using the Premature Infant Pain Profile (PIPP). The secondary outcome will be maturity of neurobehavioral functioning, as measured by the Neurobehavioral Assessment of the Preterm Infant (NAPI). Gestational age, cumulative exposure to KMC provided during non-pain contexts, and maternal cortisol levels will be considered in the analysis. Clinical feasibility will be accounted for from nurse and maternal questionnaires. DISCUSSION This will be the first study to examine the repeated use of KMC for managing procedural pain in preterm neonates. It is also the first to compare KMC to sucrose, or the interventions in combination, across time. Based on the theoretical framework of the brain opioid theory of attachment, it is expected that KMC will be a preferred standard of care. However, current pain management guidelines are based on minimal data on repeated use of either intervention. Therefore, regardless of the outcomes of this study, results will have important implications for guidelines and practices related to management of procedural pain in preterm infants. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01561547.
Collapse
|
21
|
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
|
22
|
Saboory E, Gholami M, Zare S, Roshan-Milani S. The long-term effects of neonatal morphine administration on the pentylenetetrazol seizure model in rats: the role of hippocampal cholinergic receptors in adulthood. Dev Psychobiol 2013; 56:498-509. [PMID: 23775703 DOI: 10.1002/dev.21117] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 03/04/2013] [Indexed: 11/07/2022]
Abstract
Early life exposure to opiates may affect neuropathological conditions, such as epilepsy, during adulthood. We investigated whether neonatal morphine exposure affects pentylenetetrazol (PTZ)-induced seizures in adulthood. Male rats were subcutaneously injected with morphine or saline on postnatal days 8-14. During adulthood, each rat was assigned to 1 of the following 10 sub-groups: saline, nicotine (0.1, 0.5, or 1 μg), atropine (0.25 or 1 μg), oxotremorine M (0.1 or 1 μg), or mecamylamine (2 or 8 μg). An intrahippocampal infusion of the indicated compound was administered 30 min before seizure induction (80 mg/kg PTZ). Compared with the saline/oxotremorine (1 μg), saline/saline, and morphine/saline groups, the morphine/oxotremorine (1 μg) group showed a significantly increased latency to the first epileptic behavior. The duration of tonic-clonic seizures was significantly lower in the morphine/oxotremorine (1 μg) group compared to the saline/saline and morphine/saline groups. The severity of seizure was significantly decreased in the morphine/atropine (1 μg) group than in the saline/atropine (1 μg). Seizure severity was also decreased in the morphine/mecamylamine (2 μg) group than in the saline/mecamylamine (2 μg) group. Latency for death was significantly lower in the morphine/mecamylamine (2 μg) group compared with the saline/mecamylamine (2 μg) group. Mortality rates in the morphine/atropine (1 μg) and morphine/mecamylamine (2 μg) groups were significantly lower than those in the saline/atropine (1 μg) and saline/mecamylamine (2 μg) groups, respectively. Chronic neonatal morphine administration attenuated PTZ-induced seizures, reduced the mortality rate, and decreased the impact of the hippocampal cholinergic system on seizures and mortality rate in adult rats. Neonatal morphine exposure induces changes to μ-receptors that may lead to activation of GABAergic neurons in the hippocampus. This pathway may explain the anti-convulsant effects of morphine observed in our study.
Collapse
Affiliation(s)
- Ehsan Saboory
- Faculty of Medicine, Department of Physiology, Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | | | | | | |
Collapse
|
23
|
Bajic D, Berde CB, Commons KG. Periaqueductal gray neuroplasticity following chronic morphine varies with age: role of oxidative stress. Neuroscience 2012; 226:165-77. [PMID: 22999971 PMCID: PMC3489988 DOI: 10.1016/j.neuroscience.2012.09.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 09/09/2012] [Accepted: 09/11/2012] [Indexed: 12/30/2022]
Abstract
The development of tolerance to the antinociceptive effects of morphine has been associated with networks within ventrolateral periaqueductal gray (vlPAG) and separately, nitric oxide signaling. Furthermore, it is known that the mechanisms that underlie tolerance differ with age. In this study, we used a rat model of antinociceptive tolerance to morphine at two ages, postnatal day (PD) 7 and adult, to determine if changes in the vlPAG related to nitric oxide signaling produced by chronic morphine exposure were age-dependent. Three pharmacological groups were analyzed: control, acute morphine, and chronic morphine group. Either morphine (10mg/kg) or equal volume of normal saline was given subcutaneously twice daily for 6½ days. Animals were analyzed for morphine dose-response using Hot Plate test. The expression of several genes associated with nitric oxide metabolism was evaluated using rtPCR. In addition, the effect of morphine exposure on immunohistochemistry for Fos, and nNOS as well as nicotinamide adenine dinucleotide phosphate diaphorase (NADPH-d) reaction at the vlPAG were measured. In both age groups acute morphine activated Fos in the vlPAG, and this effect was attenuated by chronic morphine, specifically in the vlPAG at the level of the laterodorsal tegmental nucleus (LDTg). In adults, but not PD7 rats, chronic morphine administration was associated with activation of nitric oxide function. In contrast, changes in the gene expression of PD7 rats suggested superoxide and peroxide metabolisms may be engaged. These data indicate that there is supraspinal neuroplasticity following morphine administration as early as PD7. Furthermore, oxidative stress pathways associated with chronic morphine exposure appear age-specific.
Collapse
Affiliation(s)
- D Bajic
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, and Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
24
|
Neonatal Morphine Administration Leads to Changes in Hippocampal BDNF Levels and Antioxidant Enzyme Activity in the Adult Life of Rats. Neurochem Res 2012; 38:494-503. [DOI: 10.1007/s11064-012-0941-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 11/10/2012] [Accepted: 11/26/2012] [Indexed: 10/27/2022]
|
25
|
Gholami M, Saboory E. Morphine exposure induces age-dependent alterations in pentylenetetrazole-induced epileptic behaviors in prepubertal rats. Dev Psychobiol 2012; 55:881-7. [DOI: 10.1002/dev.21080] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 08/10/2012] [Indexed: 11/08/2022]
Affiliation(s)
- Morteza Gholami
- Faculty of Science; Department of Biology; University of Urmia; Urmia Iran
| | - Ehsan Saboory
- Neurophysiology Research Center; Urmia University of Medical Sciences; Urmia Iran
| |
Collapse
|
26
|
Walker SM, Yaksh TL. Neuraxial analgesia in neonates and infants: a review of clinical and preclinical strategies for the development of safety and efficacy data. Anesth Analg 2012; 115:638-62. [PMID: 22798528 DOI: 10.1213/ane.0b013e31826253f2] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neuraxial drugs provide robust pain control, have the potential to improve outcomes, and are an important component of the perioperative care of children. Opioids or clonidine improves analgesia when added to perioperative epidural infusions; analgesia is significantly prolonged by the addition of clonidine, ketamine, neostigmine, or tramadol to single-shot caudal injections of local anesthetic; and neonatal intrathecal anesthesia/analgesia is increasing in some centers. However, it is difficult to determine the relative risk-benefit of different techniques and drugs without detailed and sensitive data related to analgesia requirements, side effects, and follow-up. Current data related to benefits and complications in neonates and infants are summarized, but variability in current neuraxial drug use reflects the relative lack of high-quality evidence. Recent preclinical reports of adverse effects of general anesthetics on the developing brain have increased awareness of the potential benefit of neuraxial anesthesia/analgesia to avoid or reduce general anesthetic dose requirements. However, the developing spinal cord is also vulnerable to drug-related toxicity, and although there are well-established preclinical models and criteria for assessing spinal cord toxicity in adult animals, until recently there had been no systematic evaluation during early life. Therefore, in the second half of this review, we present preclinical data evaluating age-dependent changes in the pharmacodynamic response to different spinal analgesics, and recent studies evaluating spinal toxicity in specific developmental models. Finally, we advocate use of neuraxial drugs with the widest demonstrable safety margin and suggest minimum standards for preclinical evaluation before adoption of new analgesics or preparations into routine clinical practice.
Collapse
Affiliation(s)
- Suellen M Walker
- Portex Unit: Pain Research, UCL Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK.
| | | |
Collapse
|
27
|
Aubrun F, Mazoit JX, Riou B. Postoperative intravenous morphine titration. Br J Anaesth 2012; 108:193-201. [DOI: 10.1093/bja/aer458] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
28
|
Mohamad O, Chen D, Zhang L, Hofmann C, Wei L, Yu SP. Erythropoietin reduces neuronal cell death and hyperalgesia induced by peripheral inflammatory pain in neonatal rats. Mol Pain 2011; 7:51. [PMID: 21777449 PMCID: PMC3161880 DOI: 10.1186/1744-8069-7-51] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 07/21/2011] [Indexed: 11/26/2022] Open
Abstract
Painful stimuli during neonatal stage may affect brain development and contribute to abnormal behaviors in adulthood. Very few specific therapies are available for this developmental disorder. A better understanding of the mechanisms and consequences of painful stimuli during the neonatal period is essential for the development of effective therapies. In this study, we examined brain reactions in a neonatal rat model of peripheral inflammatory pain. We focused on the inflammatory insult-induced brain responses and delayed changes in behavior and pain sensation. Postnatal day 3 pups received formalin injections into the paws once a day for 3 days. The insult induced dysregulation of several inflammatory factors in the brain and caused selective neuronal cell death in the cortex, hippocampus and hypothalamus. On postnatal day 21, rats that received the inflammatory nociceptive insult exhibited increased local cerebral blood flow in the somatosensory cortex, hyperalgesia, and decreased exploratory behaviors. Based on these observations, we tested recombinant human erythropoietin (rhEPO) as a potential treatment to prevent the inflammatory pain-induced changes. rhEPO treatment (5,000 U/kg/day, i.p.), coupled to formalin injections, ameliorated neuronal cell death and normalized the inflammatory response. Rats that received formalin plus rhEPO exhibited normal levels of cerebral blood flow, pain sensitivity and exploratory behavior. Treatment with rhEPO also restored normal brain and body weights that were reduced in the formalin group. These data suggest that severe inflammatory pain has adverse effects on brain development and rhEPO may be a possible therapy for the prevention and treatment of this developmental disorder.
Collapse
Affiliation(s)
- Osama Mohamad
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
29
|
Anaesthetic Management of a 1-Month-Old Puppy Undergoing Lateral Thoracotomy for Vascular Ring Anomaly Correction. Case Rep Vet Med 2011. [DOI: 10.1155/2011/536064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A 1-month-old male flat-coated retriever was anaesthetized for correction of oesophageal constriction caused by a vascular ring anomaly. Anaesthesia was uneventfully induced with intravenous fentanyl, diazepam, and propofol and maintained with isoflurane in oxygen and air. An intercostal block with bupivacaine and lidocaine was performed, and additional analgesia with an infusion of fentanyl was provided. Fluid therapy consisted in 5% glucose in lactated Ringer’s solution and hetastarch 6%, which proved adequate to maintain normoglycemia and normovolemia. A lateral thoracotomy was performed, and the ligamentum arteriosum was ligated. Intraoperatively, heart rate (HR) varied between 120 and 180 beats min−1without accompanying changes in blood pressure. No arrhythmias were observed or bleeding occurred. The dog recovered uneventfully. Postoperative analgesia consisted in fentanyl infusion adjusted to the patient's requirement and metamizol. This paper describes for the first time the use of balanced anaesthesia and multimodal analgesia in a paediatric dog undergoing thoracotomy.
Collapse
|
30
|
Morphine exposure in early life increases nociceptive behavior in a rat formalin tonic pain model in adult life. Brain Res 2010; 1367:122-9. [PMID: 20977897 DOI: 10.1016/j.brainres.2010.10.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 10/11/2010] [Accepted: 10/13/2010] [Indexed: 11/23/2022]
Abstract
Considering the importance of a deeper understanding of the effect throughout life of opioid analgesia at birth, our objective was to determine whether morphine administration in early life, once a day for 7 days in 8-day-old rats, alters the nociceptive response over the short (P16), medium (P30), and long term (P60) and to evaluate which system is involved in the altered nociceptive response. The nociceptive responses were assessed by the formalin test, and the behavior analyzed was the total time spent in biting and flicking of the formalin-injected hindpaw, recorded during the first 5 min (phase I) and from 15-30 min (phase II). The morphine group showed no change in nociceptive response at P16, but at P30 and P60, the nociceptive response was increased in phase I, and in both phases, respectively. At P30 and P60, the animals received a non-steroidal anti-inflammatory drug (indomethacin) or NMDA receptor antagonist (ketamine) 30 min before the formalin test. The increase in the nociceptive response was completely reversed by ketamine, and partially by indomethacin. These results indicate that early morphine exposure causes an increase in the nociceptive response in adult life. It is possible that this lower nociception threshold is due to neuroadaptations in nociceptive circuits, such as the glutamatergic system. Thus, this work demonstrates the importance of evaluating clinical consequences related to early opioid administration and suggests a need for a novel design of agents that may counteract opiate-induced neuroplastic changes.
Collapse
|
31
|
Johnston CC, Fernandes AM, Campbell-Yeo M. Pain in neonates is different. Pain 2010; 152:S65-S73. [PMID: 20971562 DOI: 10.1016/j.pain.2010.10.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 09/30/2010] [Accepted: 10/05/2010] [Indexed: 12/16/2022]
Abstract
Pain processing and management in neonates, especially preterm neonates, differs from older populations. In this review, a brief background on pain processing in neonatal life, pain exposure in Neonatal Intensive Care Units (NICU), the consequences of untreated pain, and the difficulties in treating procedural pain pharmacologically will be presented. A more detailed review of non-pharmacological interventions for procedural pain in neonates will include sensory stimulation approaches, oral sweet solutions, and maternal interventions. Some possible mechanisms for the effectiveness of non-pharmacological interventions are offered. Finally, avenues of research into similar interventions as adjuvant therapies or drug-sparing effects in older populations are suggested.
Collapse
Affiliation(s)
- Celeste C Johnston
- McGill University School of Nursing, Montreal, Canada Coimbra School of Nursing, Coimbra, Portugal IWK Health Centre, Halifax, Canada
| | | | | |
Collapse
|
32
|
Mulla H. Understanding developmental pharmacodynamics: importance for drug development and clinical practice. Paediatr Drugs 2010; 12:223-33. [PMID: 20593907 DOI: 10.2165/11319220-000000000-00000] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Developmental pharmacodynamics is the study of age-related maturation of the structure and function of biologic systems and how this affects response to pharmacotherapy. This may manifest as a change in the potency, efficacy, or therapeutic range of a drug. The paucity of studies exploring developmental pharmacodynamics reflects the lack of suitable juvenile animal models and the ethical and practical constraints of conducting studies in children. However, where data from animal models are available, valuable insight has been gained into how response to therapy can change through the course of development. For example, animal neurodevelopmental models have revealed that temporal differences in the maturation of norepinephrine and serotonin neurotransmitter systems may explain the lack of efficacy of some antidepressants in children. GABA(A) receptors that switch from an excitatory to inhibitory mode during early development help to explain paradoxical seizures experienced by infants after exposure to benzodiazepines. The increased sensitivity of neonates to morphine may be due to increased postnatal expression of the mu opioid receptor. An age dependency to the pharmacokinetic-pharmacodynamic relationship has also been found in some clinical studies. For example, immunosuppressive effects of ciclosporin (cyclosporine) revealed markedly enhanced sensitivity in infants compared with older children and adults. A study of sotalol in the treatment of children with supraventricular tachycardia showed that neonates exhibited a higher sensitivity towards QTc interval prolongation compared with older children. However, the data are limited and efforts to increase and establish data on developmental pharmacodynamics are necessary to achieve optimal drug therapy in children and to ensure long-term success of pediatric drug development. This requires a dual 'bottom up' (ontogeny knowledge driven) and 'top down' (pediatric pharmacokinetic-pharmacodynamic studies) approach.
Collapse
Affiliation(s)
- Hussain Mulla
- Centre for Therapeutic Evaluation of Drugs in Children, University Hospitals of Leicester, Leicester, UK.
| |
Collapse
|
33
|
Liu MF, Lin KC, Chou YH, Lee TY. Using non-nutritive sucking and oral glucose solution with neonates to relieve pain: a randomised controlled trial. J Clin Nurs 2010; 19:1604-11. [PMID: 20384669 DOI: 10.1111/j.1365-2702.2009.03014.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
AIM To compare the efficacies of non-nutritive sucking and glucose solution as pain-relief interventions for neonates undergoing a venipuncture procedure. BACKGROUND Neonates may be subjected to painful procedures. The individual efficacies of non-nutritive sucking or oral glucose solution used alone on pain-relief for neonates still remain controversial. DESIGN A randomised, parallel-group controlled trial was designed. METHODS A total of 105 neonates were randomly assigned to one of the three groups: non-nutritive sucking, glucose solution, or control group. When a neonate underwent a venipuncture during a newborn screening test, his/her pain manifestations were videotaped and subsequently measured using the Neonatal Infant Pain Scale. Both the stability analysis and the analysis of repeated relationships were performed by generalised estimating equations. RESULTS The level of pain was regressed over the study variables (time and group) and covariate (gestational age). Both the non-nutritive sucking and glucose solution groups had significantly lower pain scores than the control group during the venipuncture and recovery phases. Furthermore, non-nutritive sucking seemed to be more effective than glucose solution. CONCLUSION Nurses can use a pain assessment tool and the pain-relief intervention to improve the quality of neonatal care in clinical practice. This study indicates that either non-nutritive sucking or glucose solution can effectively decrease the level of pain. RELEVANCE TO CLINICAL PRACTICE If a painful procedure on neonates is inevitable, simple, convenient and effective pain-relief methods such as non-nutritive sucking or glucose solution can be provided alone. Based on a neonate's condition, nurses can provide 2 ml of 25% glucose solution through a syringe for a breastfeeding infant before an invasive procedure if nipple confusion is the concern.
Collapse
Affiliation(s)
- Mei-Fang Liu
- Department of Nursing, National Taipei College of Nursing, Taipei, Taiwan, China
| | | | | | | |
Collapse
|
34
|
Abstract
Hyperalgesia and allodynia are frequent symptoms of disease and may be useful adaptations to protect vulnerable tissues. Both may, however, also emerge as diseases in their own right. Considerable progress has been made in developing clinically relevant animal models for identifying the most significant underlying mechanisms. This review deals with experimental models that are currently used to measure (sect. II) or to induce (sect. III) hyperalgesia and allodynia in animals. Induction and expression of hyperalgesia and allodynia are context sensitive. This is discussed in section IV. Neuronal and nonneuronal cell populations have been identified that are indispensable for the induction and/or the expression of hyperalgesia and allodynia as summarized in section V. This review focuses on highly topical spinal mechanisms of hyperalgesia and allodynia including intrinsic and synaptic plasticity, the modulation of inhibitory control (sect. VI), and neuroimmune interactions (sect. VII). The scientific use of language improves also in the field of pain research. Refined definitions of some technical terms including the new definitions of hyperalgesia and allodynia by the International Association for the Study of Pain are illustrated and annotated in section I.
Collapse
Affiliation(s)
- Jürgen Sandkühler
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
35
|
Oral Glucose and Parental Holding Preferable to Opioid in Pain Management in Preterm Infants. Clin J Pain 2009; 25:138-45. [DOI: 10.1097/ajp.0b013e318181ad81] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Fitzgerald M, Walker SM. Infant pain management: a developmental neurobiological approach. ACTA ACUST UNITED AC 2009; 5:35-50. [DOI: 10.1038/ncpneuro0984] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Accepted: 10/31/2008] [Indexed: 12/21/2022]
|
37
|
Ambrisko TD, Sleeper MM, Driessen B. ECG of the month. Arrythmia. J Am Vet Med Assoc 2008; 233:1548-50. [PMID: 19014285 DOI: 10.2460/javma.233.10.1548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Tamas D Ambrisko
- Department of Clinical Studies-New Bolton Center, George D. Widener Hospital for Large Animals, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Philadelphia, PA 19348-1692, USA
| | | | | |
Collapse
|
38
|
Sanders RD, Ma D, Brooks P, Maze M. Balancing paediatric anaesthesia: preclinical insights into analgesia, hypnosis, neuroprotection, and neurotoxicity. Br J Anaesth 2008; 101:597-609. [PMID: 18796440 DOI: 10.1093/bja/aen263] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Logistical and ethical reasons make conducting clinical research in paediatric practice difficult, and therefore safe and efficacious advances are dependent on good preclinical research. For example, notable advances have been made in preclinical studies of pain processing that correlate well with patient data. Other areas of paediatric anaesthetic research remain in their infancy including mechanisms of anaesthesia and anaesthetic neuroprotection and neurotoxicity. Animal data have identified the potential 'double-edged' sword of administering anaesthetic agents in the young; although these agents can be neuroprotective in certain circumstances, they can be neurotoxic in others. The potential for this toxicity must be balanced against the importance of providing adequate anaesthesia for which there can be no compromise. We review the current state of preclinical research in paediatric anaesthesia and identify areas which require further exploration in order to provide the foundations for well-conducted clinical trials.
Collapse
Affiliation(s)
- R D Sanders
- Department of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, London, UK.
| | | | | | | |
Collapse
|
39
|
Gioiosa L, Chen X, Watkins R, Umeda EA, Arnold AP. Sex chromosome complement affects nociception and analgesia in newborn mice. THE JOURNAL OF PAIN 2008; 9:962-9. [PMID: 18635401 DOI: 10.1016/j.jpain.2008.06.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 05/21/2008] [Accepted: 06/10/2008] [Indexed: 11/24/2022]
Abstract
UNLABELLED In animal studies of nociception, females are often more sensitive to painful stimuli, whereas males are often more sensitive to analgesia induced by mu-agonists. Sex differences are found even at birth, and in adulthood are likely caused, at least in part, by differences in levels of gonadal hormones. In this report, we investigate nociception and analgesia in neonatal mice and assess the contribution of the direct action of sex chromosome genes in hotplate and tail withdrawal tests. We used the 4 core genotypes mouse model, in which gonadal sex is independent of the complement of sex chromosomes (XX vs XY). Mice were tested at baseline and then injected with mu-opioid agonist morphine (10 mg/kg) or with the kappa-opioid agonist U50,488H (U50, 12.5 mg/kg) with or without the N-methyl-D-aspartate (NMDA) receptor antagonist MK-801 (0.1 mg/kg). On the day of birth, XX mice showed faster baseline latencies than XY in tail withdrawal, irrespective of their gonadal type. Gonadal males showed greater effects of morphine than gonadal females in the hotplate test, irrespective of their sex chromosome complement. U50 and morphine were effective analgesics in both tests, but MK-801 did not block the U50 effect. The results suggest that sex chromosome complement and gonadal secretions both contribute to sex differences in nociception and analgesia by the day of birth. PERSPECTIVE Sex differences in pain may stem not only from the action of gonadal hormones on pain circuits but from the sex-specific action of X and Y genes. Identification of sex chromosome genes causing sex differences could contribute to better pain therapy in females and males.
Collapse
Affiliation(s)
- Laura Gioiosa
- Department of Physiological Science and Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California 90095-1606, USA
| | | | | | | | | |
Collapse
|
40
|
Black AM, Pandya S, Clark D, Armstrong EA, Yager JY. Effect of caffeine and morphine on the developing pre-mature brain. Brain Res 2008; 1219:136-42. [DOI: 10.1016/j.brainres.2008.04.066] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Revised: 04/13/2008] [Accepted: 04/17/2008] [Indexed: 11/16/2022]
|
41
|
Rozisky JR, Dantas G, Adachi LS, Alves VS, Ferreira MBC, Sarkis JJF, Torres ILDS. Long-term effect of morphine administration in young rats on the analgesic opioid response in adult life. Int J Dev Neurosci 2008; 26:561-5. [PMID: 18579332 DOI: 10.1016/j.ijdevneu.2008.05.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Revised: 05/07/2008] [Accepted: 05/10/2008] [Indexed: 11/28/2022] Open
Abstract
Neonates, infants and children are often exposed to pain from invasive procedures during intensive care and during the post-operative period. Opioid anesthesia and post-operative opioid analgesia have been used in infants and result in clinical benefits. The objectives of this study were to verify the effect of repeated 5 microg morphine administration (subcutaneous), once a day for 7 days in 8-day-old rats, at P8 until P14. To verify the long-term effect of morphine, the animals were submitted to a second exposure of 5mg/kg (intraperitoneal) of morphine at P80 until P86. Animals that received morphine for 7 days, at P14 did not develop tolerance, however at P80, rats demonstrated greater morphine analgesia. At P86, after 7 days of morphine administration, animals showed classical tolerance. These findings may have important implications for the human neonate, suggesting a possible explanation for the differences in the requirements of morphine observed in the youngest patients.
Collapse
Affiliation(s)
- Joanna Ripoll Rozisky
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, 90050-170 Porto Alegre, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Significant advances in the assessment and management of acute pain in children have been made, and are supported by an increase in the availability and accessibility of evidence-based data. However, methodological and practical issues in the design and performance of clinical paediatric trials limit the quantity, and may influence the quality, of current data, which lags behind that available for adult practice. Collaborations within research networks, which incorporate both preclinical and clinical studies, may increase the feasibility and specificity of future trials. In early life, the developing nervous system responds differently to pain, analgesia, and injury, resulting in effects not seen in later life and which may have long-term consequences. Translational laboratory studies further our understanding of developmental changes in nociceptor pathway structure and function, analgesic pharmacodynamics, and the impact of different forms of injury. Chronic pain in children has a negative impact on quality of life, resulting in social and emotional consequences for both the child and the family. Despite age-related differences in many chronic pain conditions, such as neuropathic pain, management in children is often empirically based on data from studies in adults. There is a major need for further clinical research, training of health-care providers, and increased resources, to improve management and outcomes for children with chronic pain.
Collapse
Affiliation(s)
- S M Walker
- Portex Department of Anaesthesia, UCL Institute of Child Health and Great Ormond Street Hospital NHS Trust, 30 Guilford Street, London WC1N 1EH, UK.
| |
Collapse
|
43
|
El Sayed MF, Taddio A, Fallah S, De Silva N, Moore AM. Safety profile of morphine following surgery in neonates. J Perinatol 2007; 27:444-7. [PMID: 17592487 DOI: 10.1038/sj.jp.7211764] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To determine the effect of morphine on duration of mechanical ventilation, apnoea and hypotension among full-term neonates who underwent thoracic or abdominal surgery in a level III neonatal intensive care unit. METHOD Medical records of 82 infants were reviewed retrospectively and data including patient demographics, clinical diagnosis, type of surgery, postoperative opioid administration, duration of mechanical ventilation, hypotension, apnoea and pain scores (premature infant pain profile (PIPP) score) were collected. RESULT Sixty-two neonates (76%) received morphine following surgery as a continuous intravenous infusion during the postoperative period. Linear regression analysis showed that morphine dosage and duration were significantly associated with the duration of mechanical ventilation. An increase in morphine infusion rate by 10 microg kg(-1) h(-1) was associated with an increase in the duration of mechanical ventilation by 24 h (P<0.0001) and an increase in morphine duration of 1 hour was associated with a longer duration of mechanical ventilation by 38 min (P<0.0001). Logistic regression analysis showed no association between morphine infusion rate or duration and hypotension. Apnoea was not associated with morphine dosage or duration of infusion in neonates receiving morphine following extubation. Score on the PIPP correlated significantly with morphine infusion rate across time (r=0.47, P<0.01). CONCLUSION Postoperative morphine dose and duration may prolong the duration of mechanical ventilation but there are no significant dose-dependent effects on other parameters including apnoea or hypotension following extubation in term neonates. More research is needed to determine the safety profile of morphine for management of pain in non-ventilated neonates.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/therapeutic use
- Female
- Humans
- Infant, Newborn
- Infant, Newborn, Diseases/surgery
- Infusions, Intravenous
- Intensive Care Units, Neonatal
- Male
- Medical Records
- Morphine/administration & dosage
- Morphine/adverse effects
- Morphine/therapeutic use
- Ontario
- Pain, Postoperative/prevention & control
- Postoperative Complications
- Respiration, Artificial
- Retrospective Studies
- Safety
Collapse
Affiliation(s)
- M F El Sayed
- Department of Paediatrics, Division of Neonatology, Sick Kids, Toronto, ON, Canada.
| | | | | | | | | |
Collapse
|
44
|
Rouss K, Gerber A, Albisetti M, Hug M, Bernet V. Long-term subcutaneous morphine administration after surgery in newborns. J Perinat Med 2007; 35:79-81. [PMID: 17313316 DOI: 10.1515/jpm.2007.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AIM To analyze the management of newborns after major surgery receiving morphine subcutaneously and to identify possible side effects. METHODS Morphine was administered via a subcutaneous catheter (Insuflon) in 20 newborns after major surgery. Side effects like hypotension, pain during morphine administration and local infection were noted. Morphine dose was adjusted according to the hospital guidelines with the Neonatal Infant Pain Score (NIPS) and the Finnegan withdrawal score. RESULTS Surgery was performed at the median age of 38 5/7 weeks (range: 32 1/7-49 5/7 weeks). Before starting subcutaneous morphine administration, patients received intravenous morphine for a median of two weeks (range six days to seven weeks). All patients showed good pain relief with no severe side effects. Three patients reacted with crying to the first dose of subcutaneous morphine. No other side effects occurred. CONCLUSION Subcutaneous application of morphine with the Insuflon catheter is an alternative to intravenous treatment of postoperative pain in neonates. In this small group pain relief was good and side effects were harmless.
Collapse
Affiliation(s)
- Kerstin Rouss
- Department of Neonatology and Intensive Care, University Children's Hospital Zurich, Zurich, Switzerland
| | | | | | | | | |
Collapse
|
45
|
Spencer SJ, Martin S, Mouihate A, Pittman QJ. Early-life immune challenge: defining a critical window for effects on adult responses to immune challenge. Neuropsychopharmacology 2006; 31:1910-8. [PMID: 16395304 DOI: 10.1038/sj.npp.1301004] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Many aspects of mammalian physiology are functionally immature at birth and continue to develop throughout at least the first few weeks of life. Animals are therefore vulnerable during this time to environmental influences such as stress and challenges to the immune system that may permanently affect adult function. The adult immune system is uniquely sensitive to immune challenges encountered during the neonatal period, but it is unknown where the critical window for this programming lies. We subjected male Sprague-Dawley rats at postnatal day (P)7, P14, P21, and P28 to either a saline or lipopolysaccharide (LPS) injection and examined them in adulthood for differences in responses to a further LPS injection. Adult febrile and cyclooxygenase-2 responses to LPS were attenuated in rats given LPS at P14 and P21, but not in those treated at P7 or P28, while P7-LPS rats displayed lower adult body weights than those treated at other times. P28-LPS rats also tended to display enhanced anxiety in the elevated plus maze. In further experiments, we examined maternal-pup interactions, looking at the mothers' preference in two pup-retrieval tasks, and found no differences in maternal attention to LPS-treated pups. We therefore demonstrate a 'critical window' for the effects of a neonatal immune challenge on adult febrile responses to inflammation and suggest that there are other critical time points during development for the programming of adult physiology. We also show that the neonatal LPS effects on the adult immune system are not likely due to overt differences in maternal attention.
Collapse
Affiliation(s)
- Sarah J Spencer
- Department of Physiology and Biophysics, Faculty of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| | | | | | | |
Collapse
|
46
|
Abstract
Pain causes numerous physiological changes in neonates. All invasive procedures induce undesirable stress responses; theses responses can, however, be eliminated or reduced by a judicious use of analgesia. Even though a large number of analgesics and sedatives are currently available, most of them have not been studied in the neonate. At present, a precise understanding of the pharmacological mechanisms of analgesics is difficult because many interactions still remain unknown in the term and premature neonate. This article describes the main analgesics and sedative agents used in the neonate: morphine, fentanyl, sufentanil, alfentanil, nalbuphine, ketamine, midazolam, propofol, acetaminophen, and Emla cream. After a review of the literature regarding these drugs, some practical advices and suggestions for the treatment of procedure-induced pain, and background sedation/analgesia for ventilated neonates are given. It is also stated in this article that the best way to soothe pain in neonates is to combine non pharmacological and pharmacological strategies. At the national level, written guidelines should be prepared in order to improve pain management in the neonate.
Collapse
Affiliation(s)
- R Carbajal
- Centre National de Ressources de Lutte contre la Douleur, Hôpital d'Enfants Armand-Trousseau, 26, avenue du Docteur-Arnold-Netter, 75012 Paris, France.
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to highlight the important recent advances in this fast developing field of pain mechanisms. It is now recognized that acute tissue and neural injuries can result in nociceptor sensitization (primary hyperalgesia) and spinal cord hyperexcitability or central sensitization that results in secondary hyperalgesia and allodynia. RECENT FINDINGS New findings regarding peripheral and central sensitization are presented in this review. Newer ideas on pain modulation, pain states and pain syndromes, mechanisms of chronic pain, newer opioid and non opioid medications including newer N-methyl-D-asparate antagonists, cyclooxygenase-2 inhibitors and membrane stabilizing analgesics as well as pain control at the genetic level are discussed. SUMMARY The research of the last decade has focused on the biochemical and structural plasticity of the nervous system following tissue and nerve injury. The mechanisms involved in the transition from acute to chronic pain are complex with the involvement of interacting receptor systems and intracellular ion flux, second messenger systems, new synaptic connections and apoptosis.
Collapse
Affiliation(s)
- Nalini Vadivelu
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut 06824, USA.
| | | |
Collapse
|