1
|
Baudat M, Joosten EAJ, Simons SHP. Repetitive daily oxytocin treatment reduces weight gain but not acute neonatal procedural pain. Pediatr Res 2024:10.1038/s41390-024-03680-9. [PMID: 39523390 DOI: 10.1038/s41390-024-03680-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/09/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND While the incidence of neonatal intensive care unit (NICU) admission steadily increases, neonatology lacks evidence of a safe, effective, and preventive analgesic for treating procedural pain. Given its role in nociception and promoting healthy neurodevelopment, the endogenous neuropeptide oxytocin (OT) emerges as a promising candidate. METHODS This study investigates the use of daily repeated subcutaneous OT (1 mg/kg) treatment in an established model of neonatal repetitive procedural pain and assesses the effectivity of OT treatment on mechanical sensitivity and body weight. RESULTS Contrary to our hypothesis repeated daily OT treatment did not prevent the development of mechanical hypersensitivity following needle pricks. Furthermore, treatment with OT diminished body weight gain in neonatal pups, a major side effect observed throughout the neonatal week. These results highlight the unique nature of the maturing nociceptive system that makes the identification and selection of analgesic options for the treatment of acute neonatal procedural pain a major challenge. CONCLUSION In conclusion, our preclinical results do not support the use of repeated OT for acute pain relief in the NICU, and the side effects on body weight gain raise concerns about the use of OT in the NICU. IMPACT Repeated daily OT treatment inhibits weight gain in neonatal rat pus. Repetitive daily OT administration does not prevent the development of mechanical hypersensitivity in a model of neonatal procedural pain. Future research must focus on the unique physiology of the developing nociceptive system to establish safe, effective and protective treatment of neonatal procedural pain.
Collapse
Affiliation(s)
- Mathilde Baudat
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre+, Maastricht, the Netherlands.
- Department of Translational Neuroscience, Institute of Mental Health and Neuroscience Research, Maastricht University, Maastricht, the Netherlands.
| | - Elbert A J Joosten
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre+, Maastricht, the Netherlands
- Department of Translational Neuroscience, Institute of Mental Health and Neuroscience Research, Maastricht University, Maastricht, the Netherlands
| | - Sinno H P Simons
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus University Medical Centre Rotterdam- Sophia Children Hospital, Rotterdam, the Netherlands
| |
Collapse
|
2
|
Lim JY, Ker CJ, Lai NM, Romantsik O, Fiander M, Tan K. Dexmedetomidine for analgesia and sedation in newborn infants receiving mechanical ventilation. Cochrane Database Syst Rev 2024; 5:CD012361. [PMID: 38695625 PMCID: PMC11064761 DOI: 10.1002/14651858.cd012361.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
BACKGROUND Dexmedetomidine is a selective alpha-2 agonist with minimal impact on the haemodynamic profile. It is thought to be safer than morphine or stronger opioids, which are drugs currently used for analgesia and sedation in newborn infants. Dexmedetomidine is increasingly being used in children and infants despite not being licenced for analgesia in this group. OBJECTIVES To determine the overall effectiveness and safety of dexmedetomidine for sedation and analgesia in newborn infants receiving mechanical ventilation compared with other non-opioids, opioids, or placebo. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, CINAHL, and two trial registries in September 2023. SELECTION CRITERIA We planned to include randomised controlled trials (RCTs) and quasi-RCTs evaluating the effectiveness of dexmedetomidine compared with other non-opioids, opioids, or placebo for sedation and analgesia in neonates (aged under four weeks) requiring mechanical ventilation. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Our primary outcomes were level of sedation and level of analgesia. Our secondary outcomes included days on mechanical ventilation, number of infants requiring additional medication for sedation or analgesia (or both), hypotension, neonatal mortality, and neurodevelopmental outcomes. We planned to use GRADE to assess the certainty of evidence for each outcome. MAIN RESULTS We identified no eligible studies for inclusion. We identified four ongoing studies, two of which appear to be eligible for inclusion; they will compare dexmedetomidine with fentanyl in newborn infants requiring surgery. We listed the other two studies as awaiting classification pending assessment of full reports. One study will compare dexmedetomidine with morphine in asphyxiated newborns undergoing hypothermia, and the other (mixed population, age up to three years) will evaluate dexmedetomidine versus ketamine plus dexmedetomidine for echocardiography. The planned sample size of the four studies ranges from 40 to 200 neonates. Data from these studies may provide some evidence for dexmedetomidine efficacy and safety. AUTHORS' CONCLUSIONS Despite the increasing use of dexmedetomidine, there is insufficient evidence supporting its routine use for analgesia and sedation in newborn infants on mechanical ventilation. Furthermore, data on dexmedetomidine safety are scarce, and there are no data available on its long-term effects. Future studies should address the efficacy, safety, and long-term effects of dexmedetomidine as a single drug therapy for sedation and analgesia in newborn infants.
Collapse
Affiliation(s)
- Jia Yi Lim
- Department of Paediatrics, Monash University, Melbourne, Australia
| | - Chin Jin Ker
- Department of Paediatrics, Monash University, Melbourne, Australia
| | - Nai Ming Lai
- School of Medicine, Taylor's University, Subang Jaya, Malaysia
| | - Olga Romantsik
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | | | - Kenneth Tan
- Department of Paediatrics, Monash University, Melbourne, Australia
- Monash Newborn, Monash Medical Centre, Clayton, Melbourne, Australia
| |
Collapse
|
3
|
Pirlotte S, Beeckman K, Ooms I, Cools F. Non-pharmacological interventions for the prevention of pain during endotracheal suctioning in ventilated neonates. Cochrane Database Syst Rev 2024; 1:CD013353. [PMID: 38235838 PMCID: PMC10795104 DOI: 10.1002/14651858.cd013353.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
BACKGROUND Pain, when treated inadequately, puts preterm infants at a greater risk of developing clinical and behavioural sequelae because of their immature pain system. Preterm infants in need of intensive care are repeatedly and persistently exposed to noxious stimuli, and this happens during a critical window of their brain development with peak rates of brain growth, exuberant synaptogenesis and the developmental regulation of specific receptor populations. Nearly two-thirds of infants born at less than 29 weeks' gestation require mechanical ventilation for some duration during the newborn period. These neonates are endotracheally intubated and require repeated endotracheal suctioning. Endotracheal suctioning is identified as one of the most frequent and most painful procedures in premature infants, causing moderate to severe pain. Even with improved nursing performance and standard procedures based on neonatal needs, endotracheal suctioning remains associated with mild pain. OBJECTIVES To evaluate the benefits and harms of non-pharmacological interventions for the prevention of pain during endotracheal suctioning in mechanically ventilated neonates. Non-pharmacological interventions were compared to no intervention, standard care or another non-pharmacological intervention. SEARCH METHODS We conducted searches in June 2023 in the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE via PubMed, Embase, CINAHL and three trial registries. We searched the reference lists of related systematic reviews, and of studies selected for inclusion. SELECTION CRITERIA We included randomised controlled trials (RCTs), quasi-RCTs and cluster-RCTs that included term and preterm neonates who were mechanically ventilated via endotracheal tube or via tracheostomy tube and required endotracheal suctioning performed by doctors, nurses, physiotherapists or other healthcare professionals. DATA COLLECTION AND ANALYSIS Our main outcome measures were validated composite pain scores (including a combination of behavioural, physiological and contextual indicators). Secondary outcomes included separate physiological and behavioural pain indicators. We used standard methodological procedures expected by Cochrane. For continuous outcome measures, we used a fixed-effect model and reported mean differences (MDs) with 95% confidence intervals (CIs). For categorical outcomes, we reported the typical risk ratio (RR) and risk difference (RD) and 95% CIs. We assessed risk of bias using the Cochrane RoB 1 tool, and assessed the certainty of the evidence using GRADE. MAIN RESULTS We included eight RCTs (nine reports), which enroled 386 infants, in our review. Five of the eight studies were included in a meta-analysis. All studies enrolled preterm neonates. Facilitated tucking versus standard care (four studies) Facilitated tucking probably reduces Premature Infant Pain Profile (PIPP) score during endotracheal suctioning (MD -2.76, 95% CI 3.57 to 1.96; I² = 82%; 4 studies, 148 infants; moderate-certainty evidence). Facilitated tucking probably has little or no effect during endotracheal suctioning on: heart rate (MD -3.06 beats per minute (bpm), 95% CI -9.33 to 3.21; I² = 0%; 2 studies, 80 infants; low-certainty evidence); oxygen saturation (MD 0.87, 95% CI -1.33 to 3.08; I² = 0%; 2 studies, 80 infants; low-certainty evidence); or stress and defensive behaviours (SDB) (MD -1.20, 95% CI -3.47 to 1.07; 1 study, 20 infants; low-certainty evidence). Facilitated tucking may result in a slight increase in self-regulatory behaviours (SRB) during endotracheal suctioning (MD 0.90, 95% CI 0.20 to 1.60; 1 study, 20 infants; low-certainty evidence). No studies reported intraventricular haemorrhage (IVH). Familiar odour versus standard care (one study) Familiar odour during endotracheal suctioning probably has little or no effect on: PIPP score (MD -0.30, 95% CI -2.15 to 1.55; 1 study, 40 infants; low-certainty evidence); heart rate (MD -6.30 bpm, 95% CI -16.04 to 3.44; 1 study, 40 infants; low-certainty evidence); or oxygen saturation during endotracheal suctioning (MD -0.80, 95% CI -4.82 to 3.22; 1 study, 40 infants; low-certainty evidence). No studies reported SRB, SDB or IVH. White noise (one study) White noise during endotracheal suctioning probably has little or no effect on PIPP (MD -0.65, 95% CI -2.51 to 1.21; 1 study, 40 infants; low-certainty evidence); heart rate (MD -1.85 bpm, 95% CI -11.46 to 7.76; 1 study, 40 infants; low-certainty evidence); or oxygen saturation (MD 2.25, 95% CI -2.03 to 6.53; 1 study, 40 infants; low-certainty evidence). No studies reported SRB, SDB or IVH. AUTHORS' CONCLUSIONS Facilitated tucking / four-handed care / gentle human touch probably reduces PIPP score. The evidence of a single study suggests that facilitated tucking / four-handed care / gentle human touch slightly increases self-regulatory and approach behaviours during endotracheal suctioning. Based on a single study, familiar odour and white noise have little or no effect on any of the outcomes compared to no intervention. The use of expressed breast milk or oral sucrose suggests that there is no discernible advantage of one method over the other for reducing pain during endotracheal suctioning. None of the studies reported on any of the prespecified secondary outcomes of adverse events.
Collapse
Affiliation(s)
| | - Katrien Beeckman
- Midwifery Research, Education and Policymaking, Universiteit Antwerpen, Brussel, Belgium
| | - Isabel Ooms
- Physiotherapy and Neonatology, UZ Brussel, Jette, Belgium
| | - Filip Cools
- Neonatology, UZ Brussel, Jette, Belgium
- CEBAM, Belgian Centre for Evidence-Based Medicine, Leuven, Belgium
- Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
4
|
Luzzati M, Coviello C, De Veye HS, Dudink J, Lammertink F, Dani C, Koopmans C, Benders M, Tataranno ML. Morphine exposure and neurodevelopmental outcome in infants born extremely preterm. Dev Med Child Neurol 2023; 65:1053-1060. [PMID: 36649164 DOI: 10.1111/dmcn.15510] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 01/18/2023]
Abstract
AIM To investigate the association between morphine exposure in the neonatal period and neurodevelopment at 2 and 5 years of age while controlling for potential confounders. METHOD We performed a retrospective, single-centre cohort study on 106 infants (60 males, 46 females; mean gestational age 26 weeks [SD 1]) born extremely preterm (gestational age < 28 weeks). Morphine administration was expressed as cumulative dose (mg/kg) until term-equivalent age. Neurodevelopmental outcome was assessed at 2 years with the Bayley Scales of Infant and Toddler Development, Third Edition, Dutch version and at 5 years with the Wechsler Preschool and Primary Scale of Intelligence, Third Edition, Dutch version. Multiple linear regression analysis was used to assess the association between morphine exposure and outcome. RESULTS Sixty-four out of 106 (60.4%) infants included in the study received morphine. Morphine exposure was not associated with poorer motor, cognitive, and language subscores of the Bayley Scales of Infant and Toddler Development, Third Edition, Dutch version at 2 years. Morphine exposure was associated with lower Full-Scale IQ scores (p = 0.008, B = -9.3, 95% confidence interval [CI] = -15.6 to -3.1) and Performance IQ scores (p = 0.005, B = -17.5, 95% CI = -27.9 to -7) at 5 years of age. INTERPRETATION Morphine exposure in infants born preterm is associated with poorer Full-Scale IQ and Performance IQ at 5 years. Individualized morphine administration is advised in infants born extremely preterm.
Collapse
Affiliation(s)
- Michele Luzzati
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Division of Neonatology, University of Florence, Florence, Italy
| | | | - Henriette Swarenburg De Veye
- Department of Neonatology, Division of Perinatology and Gynecology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jeroen Dudink
- Department of Neonatology, Division of Perinatology and Gynecology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Femke Lammertink
- Department of Neonatology, Division of Perinatology and Gynecology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Carlo Dani
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Division of Neonatology, University of Florence, Florence, Italy
| | - Corine Koopmans
- Department of Neonatology, Division of Perinatology and Gynecology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Manon Benders
- Department of Neonatology, Division of Perinatology and Gynecology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Maria Luisa Tataranno
- Department of Neonatology, Division of Perinatology and Gynecology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
5
|
Kinoshita M, Olsson E, Borys F, Bruschettini M. Opioids for procedural pain in neonates. Cochrane Database Syst Rev 2023; 6:CD015056. [PMID: 37350685 PMCID: PMC10292809 DOI: 10.1002/14651858.cd015056.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
BACKGROUND Neonates might be exposed to numerous painful procedures due to diagnostic reasons, therapeutic interventions, or surgical procedures. Options for pain management include opioids, non-pharmacological interventions, and other drugs. Morphine, fentanyl, and remifentanil are the opioids most often used in neonates. However, negative impact of opioids on the structure and function of the developing brain has been reported. OBJECTIVES To evaluate the benefits and harms of opioids in term or preterm neonates exposed to procedural pain, compared to placebo or no drug, non-pharmacological intervention, other analgesics or sedatives, other opioids, or the same opioid administered by a different route. SEARCH METHODS We used standard, extensive Cochrane search methods. The latest search date was December 2021. SELECTION CRITERIA We included randomized controlled trials conducted in preterm and term infants of a postmenstrual age (PMA) up to 46 weeks and 0 days exposed to procedural pain where opioids were compared to 1) placebo or no drug; 2) non-pharmacological intervention; 3) other analgesics or sedatives; 4) other opioids; or 5) the same opioid administered by a different route. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Our primary outcomes were pain assessed with validated methods and any harms. We used a fixed-effect model with risk ratio (RR) for dichotomous data and mean difference (MD) for continuous data, and their confidence intervals (CI). We used GRADE to assess the certainty of the evidence for each outcome. MAIN RESULTS We included 13 independent studies (enrolling 823 newborn infants): seven studies compared opioids to no treatment or placebo (the main comparison in this review), two studies to oral sweet solution or non-pharmacological intervention, and five studies (of which two were part of the same study) to other analgesics and sedatives. All studies were performed in a hospital setting. Opioids compared to placebo or no drug Compared to placebo, opioids probably reduce pain score assessed with the Premature Infant Pain Profile (PIPP)/PIPP-Revised (PIPP-R) scale during the procedure (MD -2.58, 95% CI -3.12 to -2.03; 199 participants, 3 studies; moderate-certainty evidence); may reduce Neonatal Infant Pain Scale (NIPS) during the procedure (MD -1.97, 95% CI -2.46 to -1.48; 102 participants, 2 studies; low-certainty evidence); and may result in little to no difference in pain score assessed with the Douleur Aiguë du Nouveau-né (DAN) scale one to two hours after the procedure (MD -0.20, 95% CI -2.21 to 1.81; 42 participants, 1 study; low-certainty evidence). The evidence is very uncertain about the effect of opioids on pain score assessed with the PIPP/PIPP-R scale up to 30 minutes after the procedure (MD 0.14, 95% CI -0.17 to 0.45; 123 participants, 2 studies; very low-certainty evidence) or one to two hours after the procedure (MD -0.83, 95% CI -2.42 to 0.75; 54 participants, 2 studies; very low-certainty evidence). The evidence is very uncertain about the effect of opioids on episodes of bradycardia (RR 3.19, 95% CI 0.14 to 72.69; 172 participants, 3 studies; very low-certainty evidence). Opioids may result in an increase in episodes of apnea compared to placebo (RR 3.15, 95% CI 1.08 to 9.16; 199 participants, 3 studies; low-certainty evidence): with one study reporting a concerning increase in severe apnea (RR 7.44, 95% CI 0.42 to 132.95; 31 participants, 1 study; very low-certainty). The evidence is very uncertain about the effect of opioids on episodes of hypotension (RR not estimable, risk difference 0.00, 95% CI -0.06 to 0.06; 88 participants, 2 studies; very low-certainty evidence). No studies reported parent satisfaction with care provided in the neonatal intensive care unit (NICU). Opioids compared to non-pharmacological intervention The evidence is very uncertain about the effect of opioids on pain score assessed with the Crying Requires oxygen Increased vital signs Expression Sleep (CRIES) scale during the procedure when compared to facilitated tucking (MD -4.62, 95% CI -6.38 to -2.86; 100 participants, 1 study; very low-certainty evidence) or sensorial stimulation (MD 0.32, 95% CI -1.13 to 1.77; 100 participants, 1 study; very low-certainty evidence). The other main outcomes were not reported. Opioids compared to other analgesics or sedatives The evidence is very uncertain about the effect of opioids on pain score assessed with the PIPP/PIPP-R during the procedure (MD -0.29, 95% CI -1.58 to 1.01; 124 participants, 2 studies; very low-certainty evidence); up to 30 minutes after the procedure (MD -1.10, 95% CI -2.82 to 0.62; 12 participants, 1 study; very low-certainty evidence); and one to two hours after the procedure (MD -0.17, 95% CI -2.22 to 1.88; 12 participants, 1 study; very low-certainty evidence). No studies reported any harms. The evidence is very uncertain about the effect of opioids on episodes of apnea during (RR 3.27, 95% CI 0.85 to 12.58; 124 participants, 2 studies; very low-certainty evidence) and after the procedure (RR 2.71, 95% CI 0.11 to 64.96; 124 participants, 2 studies; very low-certainty evidence) and on hypotension (RR 1.34, 95% CI 0.32 to 5.59; 204 participants, 3 studies; very low-certainty evidence). The other main outcomes were not reported. We identified no studies comparing different opioids (e.g. morphine versus fentanyl) or different routes for administration of the same opioid (e.g. morphine enterally versus morphine intravenously). AUTHORS' CONCLUSIONS Compared to placebo, opioids probably reduce pain score assessed with PIPP/PIPP-R scale during the procedure; may reduce NIPS during the procedure; and may result in little to no difference in DAN one to two hours after the procedure. The evidence is very uncertain about the effect of opioids on pain assessed with other pain scores or at different time points. The evidence is very uncertain about the effect of opioids on episodes of bradycardia, hypotension or severe apnea. Opioids may result in an increase in episodes of apnea. No studies reported parent satisfaction with care provided in the NICU. The evidence is very uncertain about the effect of opioids on any outcome when compared to non-pharmacological interventions or to other analgesics. We identified no studies comparing opioids to other opioids or comparing different routes of administration of the same opioid.
Collapse
Affiliation(s)
- Mari Kinoshita
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Emma Olsson
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Franciszek Borys
- II Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Matteo Bruschettini
- Paediatrics, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Department of Research and Education, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
6
|
Abstract
BACKGROUND Neonates might be exposed to numerous painful procedures due to diagnostic reasons, therapeutic interventions, or surgical procedures. Options for pain management include opioids, non-pharmacological interventions, and other drugs. Morphine, fentanyl, and remifentanil are the opioids most often used in neonates. However, negative impact of opioids on the structure and function of the developing brain has been reported. OBJECTIVES To evaluate the benefits and harms of opioids in term or preterm neonates exposed to procedural pain, compared to placebo or no drug, non-pharmacological intervention, other analgesics or sedatives, other opioids, or the same opioid administered by a different route. SEARCH METHODS We used standard, extensive Cochrane search methods. The latest search date was December 2021. SELECTION CRITERIA We included randomized controlled trials conducted in preterm and term infants of a postmenstrual age (PMA) up to 46 weeks and 0 days exposed to procedural pain where opioids were compared to 1) placebo or no drug; 2) non-pharmacological intervention; 3) other analgesics or sedatives; 4) other opioids; or 5) the same opioid administered by a different route. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Our primary outcomes were pain assessed with validated methods and any harms. We used a fixed-effect model with risk ratio (RR) for dichotomous data and mean difference (MD) for continuous data, and their confidence intervals (CI). We used GRADE to assess the certainty of the evidence for each outcome. MAIN RESULTS We included 13 independent studies (enrolling 823 newborn infants): seven studies compared opioids to no treatment or placebo (the main comparison in this review), two studies to oral sweet solution or non-pharmacological intervention, and five studies (of which two were part of the same study) to other analgesics and sedatives. All studies were performed in a hospital setting. Opioids compared to placebo or no drug Compared to placebo, opioids probably reduce pain score assessed with the Premature Infant Pain Profile (PIPP)/PIPP-Revised (PIPP-R) scale during the procedure (MD -2.58, 95% CI -3.12 to -2.03; 199 participants, 3 studies; moderate-certainty evidence); may reduce Neonatal Infant Pain Scale (NIPS) during the procedure (MD -1.97, 95% CI -2.46 to -1.48; 102 participants, 2 studies; low-certainty evidence); and may result in little to no difference in pain score assessed with the Douleur Aiguë du Nouveau-né (DAN) scale one to two hours after the procedure (MD -0.20, 95% CI -2.21 to 1.81; 42 participants, 1 study; low-certainty evidence). The evidence is very uncertain about the effect of opioids on pain score assessed with the PIPP/PIPP-R scale up to 30 minutes after the procedure (MD 0.14, 95% CI -0.17 to 0.45; 123 participants, 2 studies; very low-certainty evidence) or one to two hours after the procedure (MD -0.83, 95% CI -2.42 to 0.75; 54 participants, 2 studies; very low-certainty evidence). No studies reported any harms. The evidence is very uncertain about the effect of opioids on episodes of bradycardia (RR 3.19, 95% CI 0.14 to 72.69; 172 participants, 3 studies; very low-certainty evidence). Opioids may result in an increase in episodes of apnea compared to placebo (RR 3.15, 95% CI 1.08 to 9.16; 199 participants, 3 studies; low-certainty evidence). The evidence is very uncertain about the effect of opioids on episodes of hypotension (RR not estimable, risk difference 0.00, 95% CI -0.06 to 0.06; 88 participants, 2 studies; very low-certainty evidence). No studies reported parent satisfaction with care provided in the neonatal intensive care unit (NICU). Opioids compared to non-pharmacological intervention The evidence is very uncertain about the effect of opioids on pain score assessed with the Crying Requires oxygen Increased vital signs Expression Sleep (CRIES) scale during the procedure when compared to facilitated tucking (MD -4.62, 95% CI -6.38 to -2.86; 100 participants, 1 study; very low-certainty evidence) or sensorial stimulation (MD 0.32, 95% CI -1.13 to 1.77; 100 participants, 1 study; very low-certainty evidence). The other main outcomes were not reported. Opioids compared to other analgesics or sedatives The evidence is very uncertain about the effect of opioids on pain score assessed with the PIPP/PIPP-R during the procedure (MD -0.29, 95% CI -1.58 to 1.01; 124 participants, 2 studies; very low-certainty evidence); up to 30 minutes after the procedure (MD -1.10, 95% CI -2.82 to 0.62; 12 participants, 1 study; very low-certainty evidence); and one to two hours after the procedure (MD -0.17, 95% CI -2.22 to 1.88; 12 participants, 1 study; very low-certainty evidence). No studies reported any harms. The evidence is very uncertain about the effect of opioids on episodes of apnea during (RR 3.27, 95% CI 0.85 to 12.58; 124 participants, 2 studies; very low-certainty evidence) and after the procedure (RR 2.71, 95% CI 0.11 to 64.96; 124 participants, 2 studies; very low-certainty evidence) and on hypotension (RR 1.34, 95% CI 0.32 to 5.59; 204 participants, 3 studies; very low-certainty evidence). The other main outcomes were not reported. We identified no studies comparing different opioids (e.g. morphine versus fentanyl) or different routes for administration of the same opioid (e.g. morphine enterally versus morphine intravenously). AUTHORS' CONCLUSIONS Compared to placebo, opioids probably reduce pain score assessed with PIPP/PIPP-R scale during the procedure; may reduce NIPS during the procedure; and may result in little to no difference in DAN one to two hours after the procedure. The evidence is very uncertain about the effect of opioids on pain assessed with other pain scores or at different time points. No studies reported if any harms occurred. The evidence is very uncertain about the effect of opioids on episodes of bradycardia or hypotension. Opioids may result in an increase in episodes of apnea. No studies reported parent satisfaction with care provided in the NICU. The evidence is very uncertain about the effect of opioids on any outcome when compared to non-pharmacological interventions or to other analgesics. We identified no studies comparing opioids to other opioids or comparing different routes of administration of the same opioid.
Collapse
Affiliation(s)
- Mari Kinoshita
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Emma Olsson
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Franciszek Borys
- II Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Matteo Bruschettini
- Paediatrics, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Department of Research and Education, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
7
|
Campbell-Yeo M, Benoit B, Newman A, Johnston C, Bardouille T, Stevens B, Jiang A. The influence of skin-to-skin contact on Cortical Activity during Painful procedures in preterm infants in the neonatal intensive care unit (iCAP mini): study protocol for a randomized control trial. Trials 2022; 23:512. [PMID: 35725632 PMCID: PMC9208173 DOI: 10.1186/s13063-022-06424-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/24/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Strong evidence suggests that maternal-infant skin-to-skin contact (SSC) is effective in reducing behavioural responses to pain. Given the multi-sensory benefits of SSC, it is highly likely that SSC provided during pain in early life may reduce pain-induced brain activity. The aim of this study is to examine the effect of SSC compared to 24% sucrose on pain-induced activity in the preterm infant brain during a medically required heel lance. Secondary objectives include determining (a) differences between behavioural pain response and noxious-related brain activity during heel lance and (b) rate of adverse events across groups. METHODS We will randomly assign 126 babies (32 to 36 completed weeks gestational age) admitted to the neonatal intensive care unit, and their mothers within the first seven days of age to receive (i) SSC plus sterile water and (ii) 24% oral sucrose. Each baby will receive a medically indicated heel lance, following a no treatment baseline period. The primary outcome is noxious-related brain activity measured using an electroencephalogram (EEG) pain-specific event-related potential. Secondary outcomes include pain intensity measured using a bio-behavioural infant pain assessment tool (Premature Infant Pain Profile-Revised) and rate of adverse events. DISCUSSION This will be the first clinical trial to compare the effect of SSC and 24% sucrose on pain-induced brain activity in the preterm infant brain during a clinical noxious stimulus, measured using EEG. Given the negative neurodevelopmental outcomes associated with unmanaged pain, it is imperative that preterm babies receive the most effective pain-reducing treatments to improve their health outcomes. Our findings will have important implications in informing optimal pain assessment and management in preterm infants. TRIAL REGISTRATION ClinicalTrials.gov NCT03745963 . Registered on November 19, 2018.
Collapse
Affiliation(s)
- Marsha Campbell-Yeo
- School of Nursing, Faculty of Health, Dalhousie University and IWK Health, Halifax, NS Canada
| | - Britney Benoit
- Rankin School of Nursing, St. Francis Xavier University, Antigonish, NS Canada
| | - Aaron Newman
- Faculty of Science, Department of Psychology & Neuroscience, Dalhousie University, Halifax, NS Canada
| | | | - Tim Bardouille
- Department of Physics & Atmospheric Science, Dalhousie University, Halifax, NS Canada
| | - Bonnie Stevens
- Lawrence S Bloomberg Faculty of Nursing, University of Toronto and Child Health Evaluative Sciences Program, Research Institute, The Hospital for Sick Children (SickKids), Toronto, ON Canada
| | | |
Collapse
|
8
|
Sykes AG, Oviedo P, Rooney AS, Gollin G. An assessment of dexmedetomidine as an opioid-sparing agent after neonatal open thoracic and abdominal operations. J Perinatol 2022; 42:307-312. [PMID: 34312472 DOI: 10.1038/s41372-021-01175-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To evaluate the efficacy of dexmedetomidine as an opioid-sparing agent in infants following open thoracic or abdominal operations. METHODS Retrospective review of postoperative neonates who received IV acetaminophen with or without dexmedetomidine. The primary outcome was opioid dosage within the first ten postoperative days. Secondary outcomes included times to extubation, full feedings and discharge. RESULTS 112 infants met inclusion criteria. Those managed with dexmedetomidine received 1.8-4.3 times more opioid on postoperative days 1-3, had longer times to extubation and trended towards longer lengths of hospital stay than infants who were not. Opioid was dosed >0.2 ME/kg on only 23% of days when the acetaminophen dose was >40 mg/kg/day and 10% of days when the acetaminophen dose was >45 mg/kg. CONCLUSION Dexmedetomidine may not be opioid sparing after major operations in neonates and its use delays recovery. IV acetaminophen dosed at 40 mg/kg/day or greater may yield the most substantial opioid-sparing effect.
Collapse
Affiliation(s)
| | - Parisa Oviedo
- University of California San Diego School of Medicine, San Diego, CA, USA
| | | | | |
Collapse
|
9
|
Affiliation(s)
- Mari Kinoshita
- Department of Pediatrics; Clinical Sciences Lund, Lund University; Lund Sweden
| | - Emma Olsson
- Department of Pediatrics, Faculty of Medicine and Health; Örebro University; Örebro Sweden
| | | | - Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics; Lund University, Skåne University Hospital; Lund Sweden
- Cochrane Sweden; Lund University, Skåne University Hospital; Lund Sweden
| |
Collapse
|
10
|
Marchal A, Melchior M, Dufour A, Poisbeau P, Zores C, Kuhn P. Pain Behavioural Response to Acoustic and Light Environmental Changes in Very Preterm Infants. CHILDREN (BASEL, SWITZERLAND) 2021; 8:children8121081. [PMID: 34943277 PMCID: PMC8700556 DOI: 10.3390/children8121081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/12/2021] [Accepted: 11/19/2021] [Indexed: 04/12/2023]
Abstract
Noise and high light illumination in the neonatal intensive care unit (NICU) are recognized as stressors that could alter the well-being and development of vulnerable preterm infants. This prospective observational study evaluated the pain behaviours of very preterm infants (VPIs) to sound peaks (SPs) and light levels variations (LLVs) in the NICU. We measured spontaneously occurring SPs and LLVs in the incubators of 26 VPIs over 10 h. Their behavioural responses were analysed through video recordings using the "Douleur Aigue du Nouveau-né" (DAN) scale. We compared the maximum DAN scores before and after environmental stimuli and the percentage of VPIs with a score ≥ 3 according to the type of stimuli. A total of 591 SPs and 278 LLVs were analysed. SPs of 5 to 15 dBA and LLVs significantly increased the maximum DAN scores compared to baseline. The occurrence of DAN scores ≥ 3 increased with both stressors, with a total of 16% of SPs and 8% of LLVs leading to quantifiable pain behaviour. Altogether, this study shows that VPIs are sensitive to SPs and LLVs, with a slighter higher sensitivity to SPs. The mechanisms leading to pain behaviours induced by noise and light changes should be evaluated further in the context of VPIs brain development. Our results provide further arguments to optimize the NICU sensory environment of neonatal units and to adapt it to the expectations and sensory abilities of VPIs.
Collapse
Affiliation(s)
- Audrey Marchal
- Service de Médecine et Réanimation du Nouveau-né, Hôpital de Hautepierre, Centre Hospitalier Universitaire de Strasbourg, 67000 Strasbourg, France; (A.M.); (C.Z.)
| | - Meggane Melchior
- Institut des Neurosciences Cellulaires et Intégratives (INCI, CNRS UPR-3212), Centre National de la Recherche Scientifique, 67000 Strasbourg, France; (M.M.); (P.P.)
| | - André Dufour
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA, CNRS UMR-7364), Centre National de la Recherche Scientifique, Université de Strasbourg, 67000 Strasbourg, France;
| | - Pierrick Poisbeau
- Institut des Neurosciences Cellulaires et Intégratives (INCI, CNRS UPR-3212), Centre National de la Recherche Scientifique, 67000 Strasbourg, France; (M.M.); (P.P.)
| | - Claire Zores
- Service de Médecine et Réanimation du Nouveau-né, Hôpital de Hautepierre, Centre Hospitalier Universitaire de Strasbourg, 67000 Strasbourg, France; (A.M.); (C.Z.)
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA, CNRS UMR-7364), Centre National de la Recherche Scientifique, Université de Strasbourg, 67000 Strasbourg, France;
| | - Pierre Kuhn
- Service de Médecine et Réanimation du Nouveau-né, Hôpital de Hautepierre, Centre Hospitalier Universitaire de Strasbourg, 67000 Strasbourg, France; (A.M.); (C.Z.)
- Institut des Neurosciences Cellulaires et Intégratives (INCI, CNRS UPR-3212), Centre National de la Recherche Scientifique, 67000 Strasbourg, France; (M.M.); (P.P.)
- Correspondence: ; Tel.: +33-388127779
| |
Collapse
|
11
|
Chambelland A, Devos C, Casagrande F, Chiaverini C. Topical ropivacaine for analgesia of aplasia cutis congenita in newborns with hereditary epidermolysis bullosa. Orphanet J Rare Dis 2020; 15:338. [PMID: 33261637 PMCID: PMC7708204 DOI: 10.1186/s13023-020-01605-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 11/03/2020] [Indexed: 11/13/2022] Open
Abstract
Aplasia cutis congenita (ACC) in patients with hereditary epidermolysis bullosa (EB) is often associated with major pain. We report our experience with using topical ropivacaine during dressing in newborns with ACC. Eight full-term newborns with EB and ACC were hospitalized in a neonatal intensive care unit for severe pain during dressing despite the use of paracetamol, opioids (n = 8) or ketamine (n = 7). Topical xylocaine was poorly tolerated and not effective. Ropivacaine 2 mg/ml was used directly in contact with the ACC, with a maximum 1 mg/kg/day, which enabled care without the child crying. No immediate or late systemic toxicity was observed. Topical ropivacaine 0.2% appears to be an interesting topical analgesic, with good clinical tolerance and rapid action, in newborns with ACC and EB. These data need to be confirmed in a prospective study including pharmacokinetics evaluations.
Collapse
Affiliation(s)
- A Chambelland
- CRMPR Sud, Department of Dermatology, Université Côte D'Azur, CHU de Nice, Archet 2, 151 route de Saint-Antoine, 06200, Nice, France
| | - C Devos
- Department of Algology, Université Côte D'Azur, CHU de Nice, Nice, France
| | - F Casagrande
- Department of Neonatal Reanimation, Université Côte D'Azur,CHU de Nice, Nice, France
| | - C Chiaverini
- CRMPR Sud, Department of Dermatology, Université Côte D'Azur, CHU de Nice, Archet 2, 151 route de Saint-Antoine, 06200, Nice, France.
| |
Collapse
|
12
|
Neonatal morphine exposure and maternal deprivation alter nociceptive response and central biomarkers' levels throughout the life of rats. Neurosci Lett 2020; 738:135350. [PMID: 32889004 DOI: 10.1016/j.neulet.2020.135350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 11/20/2022]
Abstract
In the present study, we investigated the effect of repeated neonatal morphine exposure and/or maternal deprivation(MD) on the nociceptive response and central biomarkers' BDNF, IL-1β, and IL-4 levels at postnatal days 16(PND16), 30(PND30), and 60(PND60). At birth, the litters were standardized to contain 8 pups/dam (n = 58). From PND1 to PND10, the pups of the deprived groups were separated daily from their mothers for 3 h and divided into 5 groups: control(C), saline(S), morphine(M), deprived-saline(DS), and deprived-morphine(DM). The pups received subcutaneous injections of saline/morphine (5 μg) in the mid-scapular area between PND8 and PND14. Nociceptive responses were assessed by hot plate(HP) and tail-flick(TFL) tests and biomarker levels by ELISA. Thermal hyperalgesia(HP) was found in all assessments for the M, DS, and DM groups, and a decrease in nociceptive threshold(TFL) was found in the DS group at PND16; M and DM groups at PND30; and M, DS, and DM groups at PND60. There were interactions between treatment/deprivation/timepoint in all central biomarkers' levels. The current study indicates that neonatal exposure to morphine and MD, which occurs in the pediatric ICU, can alter the nociceptive and neuroinflammatory responses.
Collapse
|
13
|
Al-Mouqdad MM, Khalil TM, Asfour SS. Retrospective study of short-term complications associated with early morphine use in intubated premature infants. Sci Rep 2020; 10:10874. [PMID: 32616894 PMCID: PMC7331726 DOI: 10.1038/s41598-020-67891-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/10/2020] [Indexed: 11/15/2022] Open
Abstract
Relieving neonatal pain is essential for the management of premature infants. Morphine is the most frequently used analgesic in neonatal intensive care. Here we report the relationship between early morphine infusion and the composite outcome of intraventricular hemorrhage and/or death in intubated premature infants. Infants (gestational age ≤ 32 weeks and birth weight < 1,500 g) intubated on admission were retrospectively evaluated in a large tertiary neonatal intensive care unit. Modified log-Poisson regression with robust variance estimator and Cox regression was applied to adjust the relative risk for infants’ outcomes. Of 420 premature infants, 230 (54.7%) received continuous morphine infusion in the first 72 h. Of these, 153 were < 28 gestational weeks; of the 190 patients who did not receive morphine, 63 were < 28 gestational weeks. The analysis revealed that infants < 28 gestational weeks who received morphine were significantly associated with an increased risk for IVH and/or death [adjusted relative risk (aRR) 1.37, 95% confidence interval (CI) 1.1–1.71)], and mortality (aRR 1.83, 95% CI 1.17–2.89). Moreover, in infants < 28 gestational weeks, survival was low in those infants who were exposed to morphine infusion in the first 72 h (hazard ratio 2.11; 95% CI 1.19–3.73). Early morphine infusion is associated with an increased risk for IVH and/or death; however, further studies are required to verify our findings.
Collapse
Affiliation(s)
- Mountasser M Al-Mouqdad
- Neonatal Intensive Care, NICU Department, Hospital of Paediatrics, King Saud Medical City, Al Imam Abdul Aziz Ibn Muhammad Ibn Saud, Riyadh, 12746, Saudi Arabia.
| | - Thanaa M Khalil
- Obstetric and Gynecology Department, Maternity Hospital, King Saud Medical City, Riyadh, Saudi Arabia
| | - Suzan S Asfour
- Clinical Pharmacy Department, Pharmaceutical Care Services, King Saud Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Dallefeld SH, Smith PB, Crenshaw EG, Daniel KR, Gilleskie ML, Smith DS, Balevic S, Greenberg RG, Chu V, Clark R, Kumar KR, Zimmerman KO. Comparative safety profile of chloral hydrate versus other sedatives for procedural sedation in hospitalized infants. J Neonatal Perinatal Med 2020; 13:159-165. [PMID: 32538879 DOI: 10.3233/npm-190214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Given the limited available evidence on chloral hydrate safety in neonatal populations and the discrepancy in chloral hydrate acceptance between the US and other countries, we sought to clarify the safety profile of chloral hydrate compared to other sedatives in hospitalized infants. METHODS We included all infants <120 days of life who underwent a minor procedure and were administered chloral hydrate, clonidine, clonazepam, dexmedetomidine, diazepam, ketamine, lorazepam, midazolam, propofol, or pentobarbital on the day of the procedure. We characterized the distribution of infant characteristics and evaluated the relationship between drug administration and any adverse event. We performed propensity score matching, regression adjustment (RA), and inverse probability weighting (IPW) to ensure comparison of similar infants and to account for confounding by indication and residual bias. Results were assessed for robustness to analytical technique by reanalyzing the main outcomes with multivariate logistic regression, a doubly robust IPW with RA model, and a doubly robust augmented IPW model with bias-correction. RESULTS Of 650 infants, 497 (76%) received chloral hydrate, 79 (12%) received midazolam, 54 (8%) received lorazepam, and 15 (2%) received pentobarbital. Adverse events occurred in 41 (6%) infants. Using propensity score matching, chloral hydrate was associated with a decreased risk of an adverse event compared to other sedatives, risk difference (95% confidence interval) of -12.79 (-18.61, -6.98), p < 0.001. All other statistical methods resulted in similar findings. CONCLUSION Administration of chloral hydrate to hospitalized infants undergoing minor procedures is associated with a lower risk for adverse events compared to other sedatives.
Collapse
Affiliation(s)
- S H Dallefeld
- Pediatric Intensive Care Unit, Dell Children's Medical Center of Central Texas, Austin, TX, USA.,Department of Pediatrics, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - P B Smith
- Department of Pediatrics, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - E G Crenshaw
- Department of Pediatrics, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - K R Daniel
- Department of Pediatrics, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - M L Gilleskie
- Department of Pediatrics, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - D S Smith
- Department of Pediatrics, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - S Balevic
- Department of Pediatrics, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - R G Greenberg
- Department of Pediatrics, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Vivian Chu
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - R Clark
- Pediatrix Medical Group, Inc, Sunrise, FL, USA
| | - K R Kumar
- Department of Pediatrics, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - K O Zimmerman
- Department of Pediatrics, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| |
Collapse
|
15
|
Chen Y, Tong Y, Xue Z, Cheng Y, Li X. Evaluation of the Reliability and Validity of the Behavioral Indicators of Infant Pain Scale in Chinese Neonates. Pain Manag Nurs 2020; 21:456-461. [PMID: 32088094 DOI: 10.1016/j.pmn.2020.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 11/13/2019] [Accepted: 01/12/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Behavioral Indicators of Infant Pain scale (BIIP) has been shown to be a good tool to assess pain in infants. AIMS This paper aimed to translate BIIP into Chinese and evaluate its reliability and validity for neonates in China. DESIGN A prospective observational study. SETTING AND PARTICIPANTS: A convenience sample of 396 neonates (preterm and term infants) were recruited from neonatal intensive care units in China from July to October 2016. METHODS The BIIP was translated and adjusted for semantic adaption.396 neonates were assessed during 3 phases of blood collection from an artery/vein. A video camera was positioned for a close-up view of the face and body. The neonates' pain was rated independently by 2 nurses who were trained and familiar with the Chinese version of BIIP (C-BIIP)and FLACC (Facial expression, Legs, Activity, Crying and Consolability). RESULTS The internal consistency were 0.904 (preterm) and 0.895 (term). The test-retest reliability were 0.947 (preterm) and 0.938 (term) and the interclass correlation coefficients were 0.921 to 0.959 (preterm) and 0.921 to 0.959 (term). The correlations between the C-BIIP and FLACC were high (preterm: r = 0.948, term: r = 0.896). Using the C-BIIP, the 3 phases of blood collection were found to be statistically different (preterm: F = 635.76, term: F = 675.54; P < 0.001), which showed that the construct validity of C-BIIP was good. CONCLUSION The BIIP is a reliable and valid tool to assess pain in term and preterm neonates in China.
Collapse
Affiliation(s)
- Yi Chen
- Medical College, Hangzhou Normal University, Hangzhou, China; Leshan Vocational and Technical College, Leshan, China
| | - Yingge Tong
- Medical College, Hangzhou Normal University, Hangzhou, China.
| | - Zihao Xue
- Medical College, Hangzhou Normal University, Hangzhou, China
| | - Yan Cheng
- People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - XiaoYan Li
- Faculty of Medicine and Health, Lishui University, Lishui, China
| |
Collapse
|
16
|
Opioids: A Review of Pharmacokinetics and Pharmacodynamics in Neonates, Infants, and Children. Eur J Drug Metab Pharmacokinet 2020; 44:591-609. [PMID: 31006834 DOI: 10.1007/s13318-019-00552-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Pain management in the pediatric population is complex for many reasons. Mild pain is usually managed quite well with oral acetaminophen or ibuprofen. Situations involving more severe pain often require the use of an opioid, which may be administered by many different routes, depending on clinical necessity. Acute and chronic disease states, as well as the constantly changing maturational process, produce unique challenges at every level of pediatrics in dosing and management of all medications, especially with regard to high-risk opioids. Although there has been significant progress in the understanding of opioid pharmacokinetics and pharmacodynamics in neonates, infants, children, and adolescents, somewhat limited data exist from which necessary information, concerning the safe and effective use of these agents, may be drawn. The evidence here provided is intended to be helpful in directing the practitioner to patient-specific reasons for preferring one opioid over another. As our knowledge of opioids and their effects has grown, it has become clear that older medications like codeine and meperidine (pethidine) have very limited use in pediatrics. This review provides pharmacokinetic and pharmacodynamic evidence on the currently available opioids: morphine, fentanyl (and derivatives), codeine, meperidine, oxycodone, hydrocodone, hydromorphone, methadone, buprenorphine, butorphanol, nalbuphine, pentazocin, ketobemidone, tramadol, piritramide, naloxone and naltrexone. Morphine, being the most studied opioid analgesic, is the standard against which all others are compared. Pharmacokinetic parameters of morphine that have been found in neonates, i.e., higher volume of distribution, immature metabolic processes that develop at various rates, elimination that is variable based on age and weight, as well as treated and untreated disease processes, are an example of all opioids in the population discussed in this review. Outside the premature and neonatal population, the use of opioids in infants, children, and adolescents quickly begins to resemble the established values found in adults. As such, the concerns (risks) of these medications become comparable to those seen in adults.
Collapse
|
17
|
Mascarenhas CJ, Liu R, Barr GA. Effects of plant-derived analgesic compounds sinomenine and salvinorin A in infant rats. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2020; 18:174-180. [PMID: 31992510 DOI: 10.1016/j.joim.2020.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 07/03/2019] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Premature and ill neonates undergo painful but medically necessary procedures while hospitalized. Although opiate drugs are administered as analgesics, problems associated with their side effects, tolerance, and potential dependence necessitate research into alternative pain-relieving medications. Here we test two plant-derived compounds in infant rats: sinomenine, which targets the Mas-related G-protein-coupled receptor member X2 opioid receptor; and salvinorin A, which is a κ opioid receptor agonist. In adult animals both sinomenine and salvinorin A are analgesic, but neither has been tested in infants. METHODS We used the formalin and thermal plantar tests in rats 7 and 21 days of age (PN7 and PN21) for behavioral signs of pain. In addition, brain sections were stained using Fos immunohistochemistry to examine patterns of brain activation in the midbrain periaqueductal gray and the paraventricular nucleus of the hypothalamus. RESULTS Sinomenine was analgesic in both the formalin and thermal tests on animals 21 days of age. At PN7 only the highest dose elevated response latencies in the thermal test and there were no effects of sinomenine in the formalin test. Analysis of Fos expression in the sinomenine-treated animals showed no drug effect, in contrast to the behavioral results. Salvinorin A was analgesic in the formalin test only at the highest dose at 21 days of age but not in the thermal test at either age. CONCLUSION The increased modest effectiveness of sinomenine in older animals and the minimum salvinorin A drug effect suggest that the compounds act on sites that develop during the preweaning period (sinomenine) or after weaning (salvinorin A).
Collapse
Affiliation(s)
- Conrad J Mascarenhas
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Renyu Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gordon A Barr
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Anesthesiology and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Gan KML, Oei JL, Quah-Smith I, Kamar AA, Lordudass AAD, Liem KD, Lindrea KB, Daly M, Gaunker N, Mangat AK, Yaskina M, Schmölzer GM. Magnetic Non-invasive Auricular Acupuncture During Eye-Exam for Retinopathy of Prematurity in Preterm Infants: A Multicentre Randomized Controlled Trial. Front Pediatr 2020; 8:615008. [PMID: 33425820 PMCID: PMC7786201 DOI: 10.3389/fped.2020.615008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/25/2020] [Indexed: 11/17/2022] Open
Abstract
Background: Eye exam for Retinopathy of prematurity (ROP) is a painful procedure and pharmacological analgesia might be ineffective. We hypothesized that magnetic auricular acupuncture (MAA) compared to placebo will decrease pain during ROP exam in preterm infants. Methods: Multicentre randomized controlled trial conducted in three hospitals (Australia, Canada, and Malaysia). Eligibility: >32 weeks, ROP exam, not sedated, and parental consent. A total of 100 infants were randomized (1:1) to MAA (n = 50) or placebo (n = 50). MAA stickers or placebo were placed on both ears by an unblinded investigator. Pain was assessed using the Premature Infant Pain Profile. Primary analyses were by intention-to-treat. ClinicalTrials.gov:NCT03650621. Findings: The mean (standard deviation, SD) gestation, birthweight, and postnatal age were (MAA 28(3) vs. placebo 28(2) weeks; MAA 1,057(455) vs. placebo 952(273) g; MAA 7(3) vs. placebo 7(3) weeks. Placebo infants had significantly higher PIPP scores during [mean difference 1.6 points (95%CI 0.1-3.1)] and 1 h mean difference 1.5 points (95%CI 0.7-2.2) after the procedure (p < 0.03). Heart rate was lower (173(22) vs. 184(18)/min) and oxygen saturations were higher (93.8(6.2) vs. 91.7(6.1)%, p = 0.05) in MAA infants. No adverse effects. Interpretation: MAA may reduce physiological pain responses during and after ROP exam in preterm infants. Assessment of long-term effects are warranted. Clinical trial registration: www.ClinicalTrials.gov, identifier: NCT03650621.
Collapse
Affiliation(s)
- Kimberly M L Gan
- Faculty of Medicine, University of New South Wales, Kensington, NSW, Australia.,School of Women's and Children's Health, University of New South Wales, Kensington, NSW, Australia
| | - Ju-Lee Oei
- School of Women's and Children's Health, University of New South Wales, Kensington, NSW, Australia.,Department of Newborn Care, The Royal Hospital for Women, Randwick, NSW, Australia
| | - Im Quah-Smith
- School of Women's and Children's Health, University of New South Wales, Kensington, NSW, Australia.,Roseville Wellness Group, Roseville, NSW, Australia
| | - Azanna A Kamar
- Neonatology Unit, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | | | - Kian D Liem
- Department of Neonatology, Radboudumc Amalia Children's Hospital, Nijmegen, Netherlands
| | - Kwee Bee Lindrea
- Department of Newborn Care, The Royal Hospital for Women, Randwick, NSW, Australia
| | - Mary Daly
- Department of Newborn Care, The Royal Hospital for Women, Randwick, NSW, Australia
| | - Nilima Gaunker
- Department of Newborn Care, The Royal Hospital for Women, Randwick, NSW, Australia
| | - Avneet K Mangat
- Neonatal Research Unit, Centre for the Studies of Asphyxia and Resuscitation, Royal Alexandra Hospital, Edmonton, AB, Canada.,Department of Paediatrics, University of Alberta, Edmonton, AB, Canada
| | - Maryna Yaskina
- Women and Children's Health Research Institute (WCHRI), University of Alberta, Edmonton, AB, Canada
| | - Georg M Schmölzer
- Neonatal Research Unit, Centre for the Studies of Asphyxia and Resuscitation, Royal Alexandra Hospital, Edmonton, AB, Canada.,Department of Paediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
19
|
Kasala S, Briyal S, Prazad P, Ranjan AK, Stefanov G, Donovan R, Gulati A. Exposure to Morphine and Caffeine Induces Apoptosis and Mitochondrial Dysfunction in a Neonatal Rat Brain. Front Pediatr 2020; 8:593. [PMID: 33042927 PMCID: PMC7530195 DOI: 10.3389/fped.2020.00593] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/11/2020] [Indexed: 11/23/2022] Open
Abstract
Background: Preterm infants experience rapid brain growth during early post-natal life making them vulnerable to drugs acting on central nervous system. Morphine is administered to premature neonates for pain control and caffeine for apnea of prematurity. Simultaneous use of morphine and caffeine is common in the neonatal intensive care unit. Prior studies have shown acute neurotoxicity with this combination, however, little information is available on the mechanisms mediating the neurotoxic effects. The objective of this study was to determine the effects of morphine and caffeine, independently and in combination on mitochondrial dysfunction (Drp1 and Mfn2), neural apoptosis (Bcl-2, Bax, and cell damage) and endothelin (ET) receptors (ETA and ETB) in neonatal rat brain. Methods: Male and female rat pups were grouped separately and were divided into four different subgroups on the basis of treatments-saline (Control), morphine (MOR), caffeine (CAFF), and morphine + caffeine (M+C) treatment. Pups in MOR group were injected with 2 mg/kg morphine, CAFF group received 100 mg/kg caffeine, and M+C group received both morphine (2 mg/kg) and caffeine (100 mg/kg), subcutaneously on postnatal days (PND) 3-6. Pups were euthanized at PND 7, 14, or 28. Brains were isolated and analyzed for mitochondrial dysfunction, apoptosis markers, cell damage, and ET receptor expression via immunofluorescence and western blot analyses. Results: M+C showed a significantly higher expression of Bax compared to CAFF or MOR alone at PND 7, 14, 28 in female pups (p < 0.05) and at PND 7, 14 in male pups (p < 0.05). Significantly (p < 0.05) increased expression of Drp1, Bax, and suppressed expression of Mfn2, Bcl-2 at PND 7, 14, 28 in all the treatment groups compared to the control was observed in both genders. No significant difference in the expression of ETA and ETB receptors in male or female pups was seen at PND 7, 14, and 28. Conclusion: Concurrent use of morphine and caffeine during the first week of life increases apoptosis and cell damage in the developing brain compared to individual use of caffeine and morphine.
Collapse
Affiliation(s)
- Sweatha Kasala
- Division of Neonatology, Department of Pediatrics, Advocate Children's Hospital, Park Ridge, IL, United States
| | - Seema Briyal
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Preetha Prazad
- Division of Neonatology, Department of Pediatrics, Advocate Children's Hospital, Park Ridge, IL, United States
| | - Amaresh K Ranjan
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Gospodin Stefanov
- Division of Neonatology, Department of Pediatrics, Advocate Children's Hospital, Park Ridge, IL, United States
| | - Ramona Donovan
- Advocate Aurora Research Institute, Park Ridge, IL, United States
| | - Anil Gulati
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States.,Pharmazz Inc. Research and Development, Willowbrook, IL, United States
| |
Collapse
|
20
|
Abstract
Abstract
Purpose
The changes in physiological functions as children grow and organ systems mature result in pharmacokinetic alterations throughout childhood. These alterations in children result in absorption, distribution, metabolism, and excretion of drugs that are different from those seen in the typical adult diseased population.
Summary
Changes in gastrointestinal motility and gastric pH in neonates and infants affect the absorption rate and bioavailability of drugs. Skin absorption rate and extent can be altered by different skin structures and perfusion in young children. Intramuscular and rectal absorption become less predictable in children due to erratic absorption site perfusion and other factors. Children’s body compositions also differ greatly from that in adults. Water-soluble drugs distribute more extensively in newborns due to larger water content than in older children and adults. Drug elimination and excretion are also affected in pediatric population due to differences in liver and renal function. Immature enzyme development and renal function result in reduced clearance of drugs in young children. There are limited pharmacokinetic data available for many drugs used in children.
Conclusion
Considering the changes in pharmacokinetics in children can help pharmacists optimize the dosing and monitoring of drugs and do the best they can to help this vulnerable population.
Collapse
|
21
|
Abstract
Measuring brain activity in infants provides an objective surrogate approach with which to infer pain perception following noxious events. Here we discuss different approaches which can be used to measure noxious-evoked brain activity, and discuss how these measures can be used to assess the analgesic efficacy of pharmacological and non-pharmacological interventions. We review factors that can modulate noxious-evoked brain activity, which may impact infant pain experience, including gestational age, sex, prior pain, stress, and illness.
Collapse
Affiliation(s)
- Deniz Gursul
- Department of Paediatrics, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, United Kingdom
| | - Caroline Hartley
- Department of Paediatrics, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, United Kingdom
| | - Rebeccah Slater
- Department of Paediatrics, University of Oxford, John Radcliffe Hospital, Headington, Oxford, OX3 9DU, United Kingdom.
| |
Collapse
|
22
|
Norman E, Kindblom JM, Rane A, Berg A, Schubert U, Hallberg B, Fellman V. Individual variations in fentanyl pharmacokinetics and pharmacodynamics in preterm infants. Acta Paediatr 2019; 108:1441-1446. [PMID: 30721546 DOI: 10.1111/apa.14744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/02/2018] [Accepted: 02/01/2019] [Indexed: 12/24/2022]
Abstract
AIM Fentanyl pharmacokinetics and pharmacodynamics are lacking in preterm infants. Our aim was to study these and their relation with a new formulation of fentanyl 5 μg/mL for procedural pain. METHODS Preterm infants were given 0.5 (n = 20, median gestational age 26.5; range 23.3-34.1 weeks) and 2 μg/kg (n = 8, 27.4; 25.3-30.7 weeks) fentanyl, respectively, before skin-breaking procedures or tracheal intubation. Blood samples were collected after ten minutes, two, four, eight and 24 hours. Physiologic parameters were monitored and pain scores assessed. RESULTS The median fentanyl concentrations were 0.18, 0.15, 0.15 and 0.57, 0.37, 0.35 ng/mL at 15-31 minutes, two and four hours and the half-lives were 1.6 to 20.5 or 4.1 to 32.6 hours for the low- and high-dose groups, respectively. A significant correlation was seen between weight at study inclusion and half-life (Spearman's r = -0.9, p < 0.001), volume of distribution (r = -0.8, p < 0.01) and clearance (r = -0.9, p < 0.01) in the low-dose group (n = 9). Pain assessment results were not correlated to pharmacokinetic variables. Fentanyl was well tolerated. CONCLUSION The inter-individual variation of fentanyl pharmacokinetics is large in preterm infants, and the dose of 0.5 μg/kg seems not effective for skin-breaking procedures.
Collapse
Affiliation(s)
- Elisabeth Norman
- Department of Clinical Sciences, Pediatrics, Lund Lund University Lund Sweden
- Neonatology Skåne University Hospital Lund Sweden
| | | | - Anders Rane
- Division of Clinical Pharmacology Karolinska University Hospital (Huddinge site) and Karolinska Institutet Stockholm Sweden
| | - Ann‐Cathrine Berg
- Department of Clinical Sciences, Pediatrics, Lund Lund University Lund Sweden
- Neonatology Skåne University Hospital Lund Sweden
| | - Ulf Schubert
- Department of Neonatology Karolinska University Hospital and Karolinska Institutet CLINTEC Stockholm Sweden
| | - Boubou Hallberg
- Department of Neonatology Karolinska University Hospital and Karolinska Institutet CLINTEC Stockholm Sweden
| | - Vineta Fellman
- Department of Clinical Sciences, Pediatrics, Lund Lund University Lund Sweden
- Neonatology Skåne University Hospital Lund Sweden
- Children′s Hospital University of Helsinki Helsinki Finland
| |
Collapse
|
23
|
Abstract
Palliative care concentrates on preventing and relieving suffering by reducing the severity of disease symptoms. Consistent treatment of pain and distress must therefore be an integral component of every palliative care concept. In this review non-pharmacological and pharmacological measures for pain and distress management in the context of palliative neonatal care are summarised. Furthermore, recommendations are given focusing on two special palliative neonatal care settings: compassionate extubation and withdrawing artificial nutrition and hydration.
Collapse
Affiliation(s)
- Lars Garten
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Christoph Bührer
- Department of Neonatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
24
|
Pirlotte S, Beeckman K, Ooms I, Van Rompaey B, Cools F. Non-pharmacological interventions for the prevention of pain during endotracheal suctioning in ventilated neonates. Hippokratia 2019. [DOI: 10.1002/14651858.cd013353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
| | - Katrien Beeckman
- Universiteit Antwerpen; Midwifery Research, Education and Policymaking; Brussel Belgium
| | - Isabel Ooms
- UZ Brussel; Physiotherapy and Neonatology; Jette Belgium
| | - Bart Van Rompaey
- University of Antwerp; Department of Nursing and Midwifery; Antwerp Belgium
| | - Filip Cools
- CEBAM, Belgian Centre for Evidence-Based Medicine; Kapucijnenvoer 33, blok J, bus 7001 Leuven Vlaams-Brabant Belgium 3000
| |
Collapse
|
25
|
Pirlotte S, Beeckman K, Ooms I, Van Rompaey B, Cools F. Pharmacological interventions for the prevention of pain during endotracheal suctioning in ventilated neonates. Hippokratia 2019. [DOI: 10.1002/14651858.cd013355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
| | - Katrien Beeckman
- Universiteit Antwerpen; Midwifery Research, Education and Policymaking; Brussel Belgium
| | - Isabel Ooms
- UZ Brussel; Physiotherapy and Neonatology; Jette Belgium
| | - Bart Van Rompaey
- University of Antwerp; Department of Nursing and Midwifery; Antwerp Belgium
| | - Filip Cools
- UZ Brussel; Department Neonatology; Laarbeeklaan Brussels Belgium
| |
Collapse
|
26
|
Williams N, MacLean K, Guan L, Collet JP, Holsti L. Pilot Testing a Robot for Reducing Pain in Hospitalized Preterm Infants. OTJR-OCCUPATION PARTICIPATION AND HEALTH 2019; 39:108-115. [PMID: 30770034 DOI: 10.1177/1539449218825436] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Optimizing neurodevelopment is a key goal of neonatal occupational therapy. In preterm infants, repeated procedural pain is associated with adverse effects on neurodevelopment long term. Calmer is a robot designed to reduce infant pain. The objective of this study was to examine the effects of Calmer on heart rate variability (HRV) during routine blood collection in preterm infants. In a randomized controlled pilot trial, 10 infants were assigned to either standard care ( n = 5, facilitated tucking [FT]) or Calmer treatment ( n = 5). HRV was recorded continuously and quantified using the area (power) of the spectrum in high and low frequency (HF: 0.15-0.40Hz/ms2; LF: 0.04-0.15 Hz/ms2) regions. Changes in HRV during three, 2-min phases (Baseline, Heel Poke, and Recovery) were compared between groups. Calmer infants had 90% greater parasympathetic activation ([PS] reduced stress) during Baseline, 82% greater PS activation during Poke, and 24% greater PS activation during Recovery than FT infants. Calmer reduced physiological preterm infant pain reactivity during blood collection.
Collapse
Affiliation(s)
| | - Karon MacLean
- The University of British Columbia, Vancouver, Canada
| | - Ling Guan
- The University of British Columbia, Vancouver, Canada
| | | | - Liisa Holsti
- The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
- BC Women’s Hospital Research Institute, Vancouver, Canada
| |
Collapse
|
27
|
A Systematic Review of Behavioral and Environmental Interventions for Procedural Pain Management in Preterm Infants. J Pediatr Nurs 2019; 44:22-30. [PMID: 30683278 DOI: 10.1016/j.pedn.2018.10.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 10/07/2018] [Accepted: 10/08/2018] [Indexed: 11/23/2022]
Abstract
PROBLEM Current research suggests behavioral and environmental interventions to prevent neonatal pain prior to an invasive procedure are rarely administered and seldom documented. The aim of this study was to systematically review findings from published randomized controlled trials that tested the effects of behavioral and environmental procedural pain management interventions on behavioral pain response in preterm infants. ELIGIBILITY CRITERIA Randomized controlled trials examining the effects of behavioral and environmental pain management interventions on behavioral pain response in preterm infants were identified. Articles accepted for inclusion met the following criteria: English language, original, peer refereed, randomized controlled clinical trials published within the past 5 years, study sample: preterm infants, setting: neonatal intensive care units, study intervention behavioral and environmental, outcome pain measurement score from valid and reliable pain scale. SAMPLE Fourteen randomized controlled trials from a literature search of PubMed and Medline databases were included in this review. RESULTS Across all age groups, facilitated tucking, oral sucrose, and kangaroo care decreased behavioral and physiologic pain response alone and in combination with other behavioral and environmental interventions. CONCLUSION Among preterm infants, facilitated tucking, oral sucrose, and kangaroo care significantly mitigates biobehavioral pain response associated with acutely painful procedures. IMPLICATIONS Evidence suggests that behavioral and environmental interventions can decrease biobehavioral pain response associated with acutely painful procedures in preterm infants. This review highlights the need for rigorous studies to help healthcare providers to build a tailored pain treatment plan for preterm infants.
Collapse
|
28
|
Campbell-Yeo M, Benoit B. Use of morphine before retinopathy of prematurity examinations. Lancet 2018; 392:2521-2523. [PMID: 30509742 DOI: 10.1016/s0140-6736(18)32324-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 09/13/2018] [Indexed: 10/27/2022]
Affiliation(s)
| | - Britney Benoit
- Dalhousie University School of Nursing, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
29
|
Šapina M, Kośmider M, Kramarić K, Garcin M, Adelson PD, Pirić M, Milas K, Brdarić D. Asymmetric detrended fluctuation analysis in neonatal stress. Physiol Meas 2018; 39:085006. [PMID: 30019692 DOI: 10.1088/1361-6579/aad425] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To detect stress in newborns by observing heart rate (HR) variability utilizing an asymmetric detrended fluctuation analysis (ADFA), we sought to determine the fractal structure of the series of inter-beat intervals, so as to distinguish the periods of acceleration of the HR from decelerations. Thus, two scaling exponents, α + and α -, representing decelerations and accelerations respectively, are obtained. APPROACH Forty healthy term newborns were included in this study, undergoing two different types of stress stimuli: routine heel lance blood sampling for metabolic screening purposes, and its simulation by applying dull pressure on the heel. MAIN RESULTS It appears that when newborns face stress, the scaling exponent related to accelerations significantly increases and becomes higher than the deceleration scaling exponent. To test the diagnostic properties of the scaling exponents, an ROC curve analysis was applied; α - showed good diagnostic performance with an AUC between 0.626 and 0.826, depending on the length of the time series. The joint use of α + and α - further increased the diagnostic performance, in particular for shorter series of RR intervals, with an AUC between 0.691 and 0.833. SIGNIFICANCE ADFA, particularly of the acceleration scaling exponent, may be a useful clinical diagnostic tool for monitoring neonatal stress.
Collapse
Affiliation(s)
- Matej Šapina
- University hospital Osijek, Pediatric Clinic, J. Huttlera 4, 31000 Osijek, Croatia. Medical faculty Osijek, Osijek, Cara Hadrijana 10E, 31000 Osijek, Croatia. Faculty of Dental medicine and Health, Crkvena 21, 31000 Osijek, Croatia
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Purpose of review Pain management presents a major challenge in neonatal care. Newborn infants who require medical treatment can undergo frequent invasive procedures during a critical period of neurodevelopment. However, adequate analgesic provision is infrequently and inconsistently provided for acute noxious procedures because of limited and conflicting evidence regarding analgesic efficacy and safety of most commonly used pharmacological agents. Here, we review recent advances in the measurement of infant pain and discuss clinical trials that assess the efficacy of pharmacological analgesia in infants. Recent findings Recently developed measures of noxious-evoked brain activity are sensitive to analgesic modulation, providing an objective quantitative outcome measure that can be used in clinical trials of analgesics. Summary Noxious stimulation evokes changes in activity across all levels of the infant nervous system, including reflex activity, altered brain activity and behaviour, and long-lasting changes in infant physiological stability. A multimodal approach is needed if we are to identify efficacious and well tolerated analgesic treatments. Well designed clinical trials are urgently required to improve analgesic provision in the infant population.
Collapse
|
31
|
Acupuncture in the neonatal intensive care unit-using ancient medicine to help today's babies: a review. J Perinatol 2017; 37:749-756. [PMID: 27977013 DOI: 10.1038/jp.2016.227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/21/2016] [Accepted: 11/01/2016] [Indexed: 11/08/2022]
Abstract
Acupuncture has been used for thousands of years in Eastern medicine for a variety of conditions and illnesses, including pain. Neonatal intensive care, on the other hand, is a relatively new branch of medicine that has emerged as the pivotal influence in increasing survival of critically ill newborn infants only within the last 50 years. Unfortunately, pain is an inevitable part of treatment in a neonatal intensive care unit (NICU). The control and prevention of pain remains a major issue for clinicians despite recognition and understanding of the myriad of short- and long-term problems that are associated with both pain and its treatment within the NICU environment. In this review, we examine the potential role of acupuncture to decrease and treat pain in babies requiring neonatal intensive care and discuss future therapeutic and research implications for the use of this ancient therapy within the modern environment of the NICU.
Collapse
|
32
|
Romantsik O, Calevo MG, Norman E, Bruschettini M. Clonidine for sedation and analgesia for neonates receiving mechanical ventilation. Cochrane Database Syst Rev 2017; 5:CD012468. [PMID: 28488361 PMCID: PMC6481534 DOI: 10.1002/14651858.cd012468.pub2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although routine administration of pharmacologic sedation or analgesia during mechanical ventilation in preterm neonates is not recommended, its use in clinical practice remains common. Alpha-2 agonists, mainly clonidine and dexmedetomidine, are used as adjunctive (or alternative) sedative agents alongside opioids and benzodiazepines. Clonidine has not been systematically assessed for use in neonatal sedation during ventilation. OBJECTIVES To assess whether clonidine administered to term and preterm newborn infants receiving mechanical ventilation reduces morbidity and mortality rates. To compare the intervention versus placebo, no treatment, and dexmedetomidine; and to assess the safety of clonidine infusion for potential harms.To perform subgroup analyses according to gestational age; birth weight; administration method (infusion or bolus therapy); dose, duration, and route of clonidine administration; and pharmacologic sedation as a co-intervention. SEARCH METHODS We used the standard search strategy of the Cochrane Neonatal Review Group to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2016, Issue 12) in the Cochrane Library, MEDLINE via PubMed (1966 to January 10, 2017), Embase (1980 to January 10, 2017), and the Cumulative Index to Nursing and Allied Health Literature (CINAHL; 1982 to January 10, 2017). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomized controlled trials and quasi-randomized trials. SELECTION CRITERIA We searched for randomized controlled trials, quasi-randomized controlled trials, and cluster trials comparing clonidine versus placebo, no treatment, or dexmedetomidine administered to term and preterm newborns receiving mechanical ventilation via an endotracheal tube. DATA COLLECTION AND ANALYSIS For the included trial, two review authors independently extracted data (e.g. number of participants, birth weight, gestational age, all-cause death during initial hospitalization, duration of respiratory support, sedation scale, duration of hospital stay) and assessed risk of bias (e.g. adequacy of randomization, blinding, completeness of follow-up). This review considered primary outcomes of all-cause neonatal death, all-cause death during initial hospitalization, and duration of mechanical ventilation in days. MAIN RESULTS One trial, which included 112 infants, met the inclusion criteria for this review. Term newborn infants on mechanical ventilation with the need for continuous analgesia and sedation with fentanyl and midazolam were eligible for enrollment during the first 96 hours of ventilation. Study authors administered clonidine 1 μg/kg/h or placebo on day 4 after intubation.We found no differences between the two groups in all-cause death during hospitalization (risk ratio [RR] 0.69, 95% confidence interval [CI] 0.12 to 3.98). The quality of the evidence supporting these findings is low owing to imprecision of the estimates (one study; few events). The median (interquartile range) duration of mechanical ventilation was 7.1 days (5.7 to 9.1 days) in the clonidine group and 5.8 days (4.9 to 7.9 days) in the placebo group, respectively (P = 0.070). Among secondary outcomes, we found no differences in terms of duration of stay in the intensive care unit. Sedation scale values (COMFORT) and analgesia scores (Hartwig) during the first 72 hours of infusion of study medication were lower in the clonidine group than in the placebo group. AUTHORS' CONCLUSIONS At present, evidence is insufficient to show the efficacy and safety of clonidine for sedation and analgesia in term and preterm newborn infants receiving mechanical ventilation.
Collapse
Affiliation(s)
- Olga Romantsik
- Lund University, Skåne University HospitalDepartment of PaediatricsLundSweden
| | - Maria Grazia Calevo
- Istituto Giannina GasliniEpidemiology, Biostatistics and Committees UnitGenoaItaly16147
| | - Elisabeth Norman
- Lund University, Skane University HospitalDepartment of PaediatricsLundSweden
| | - Matteo Bruschettini
- Lund University, Skåne University HospitalDepartment of PaediatricsLundSweden
- Skåne University HospitalResearch & Development, Section for HTA AnalysisLasarettsgatan 40LundSweden
| | | |
Collapse
|
33
|
Johnston C, Campbell‐Yeo M, Disher T, Benoit B, Fernandes A, Streiner D, Inglis D, Zee R. Skin-to-skin care for procedural pain in neonates. Cochrane Database Syst Rev 2017; 2:CD008435. [PMID: 28205208 PMCID: PMC6464258 DOI: 10.1002/14651858.cd008435.pub3] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Skin-to-skin care (SSC), often referred to as 'kangaroo care' (KC) due to its similarity with marsupial behaviour of ventral maternal-infant contact, is one non-pharmacological intervention for pain control in infants. OBJECTIVES The primary objectives were to determine the effect of SSC alone on pain from medical or nursing procedures in neonates compared to no intervention, sucrose or other analgesics, or additions to simple SSC such as rocking; and to determine the effects of the amount of SSC (duration in minutes), method of administration (e.g. who provided the SSC) of SSC in reducing pain from medical or nursing procedures in neonatesThe secondary objectives were to determine the safety of SSC care for relieving procedural pain in infants; and to compare the SSC effect in different postmenstrual age subgroups of infants. SEARCH METHODS For this update, we used the standard search strategy of the Cochrane Neonatal Review group to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2016, Issue 1); MEDLINE via PubMed (1966 to 25 February 2016); Embase (1980 to 25 February 2016); and CINAHL (1982 to 25 February 2016). We also searched clinical trials' databases, conference proceedings, and the reference lists of retrieved articles for randomized controlled trials and quasi-randomized trials. SELECTION CRITERIA Studies with randomisation or quasi-randomisation, double- or single-blinded, involving term infants (≥ 37 completed weeks' postmenstrual age (PMA) to a maximum of 44 weeks' PMA and preterm infants (< 37 completed weeks PMA) receiving SSC for painful procedures conducted by healthcare professionals. DATA COLLECTION AND ANALYSIS The main outcome measures were physiological or behavioural pain indicators and composite pain scores. A mean difference (MD) with 95% confidence interval (CI) using a fixed-effect model was reported for continuous outcome measures. We included variations on type of tissue-damaging procedure, provider of care, and duration of SSC. MAIN RESULTS Twenty-five studies (n = 2001 infants) were included. Nineteen studies (n = 1065) used heel lance as the painful procedure, one study combined venepuncture and heel stick (n = 50), three used intramuscular injection (n = 776), one used 'vaccination' (n = 60), and one used tape removal (n = 50). The studies were generally strong and had low or uncertain risk of bias. Blinding of the intervention was not possible, making them subject to high risk, depending on the method of scoring outcomes.Seventeen studies (n = 810) compared SSC to a no-treatment control. Although 15 studies measured heart rate during painful procedures, data from only five studies (n = 161) could be combined for a mean difference (MD) of -10.78 beats per minute (95% CI -13.63 to -7.93) favouring SSC. Meta-analysis of four studies (n = 120) showed no difference in heart rate following the painful procedure (MD 0.08, 95% CI -4.39 to 4.55). Two studies (n = 38) reported heart rate variability with no significant differences. Two studies (n = 101) in a meta-analysis on oxygen saturation at 30 and 60 seconds following the painful procedure did not show a difference. Duration of crying meta-analysis was performed on four studies (n = 133): two (n = 33) investigated response to heel lance (MD = -34.16, 95% CI -42.86 to -25.45), and two (n = 100) following IM injection (MD = -8.83, 95% CI -14.63 to -3.02), favouring SSC. Five studies, one consisting of two substudies (n = 267), used the Premature Infant Pain Profile (PIPP) as a primary outcome, which favoured SCC at 30 seconds (MD -3.21, 95% CI -3.94 to -2.47), at 60 seconds (3 studies; n = 156) (MD -1.64, 95% CI -2.86 to -0.43), and at 90 seconds (n = 156) (MD -1.28, 95% CI -2.53 to -0.04); but at 120 seconds there was no difference (n = 156) (MD 0.07, 95% CI -1.11 to 1.25). No studies on return of heart rate to baseline level, cortisol levels, and facial actions could be combined for meta-analysis findings.Eight studies compared SSC to another intervention with or without a no-treatment control. Two cross-over studies (n = 80) compared mother versus other provider (father, another female) on PIPP scores at 30, 60, 90, and 120 seconds with no significant difference. When SSC was compared to other interventions, there were not enough similar studies to pool results in an analysis. One study compared SSC (n = 640) with and without dextrose and found that the combination was most effective and that SSC alone was more effective than dextrose alone. Similarly, in another study SSC was more effective than oral glucose for heart rate (n = 95). SSC either in combination with breastfeeding or alone was favoured over a no-treatment control, but not different to breastfeeding. One study compared SSC alone and in combination with both sucrose and breastfeeding on heart rate (HR), NIPS scores, and crying time (n = 127). The combinations were more effective than SSC alone for NIPS and crying. Expressed breast milk was compared to SSC in one study (n = 50) and found both equally effective on PIPP scores. There were not enough participants with similar outcomes and painful procedures to compare age groups or duration of SSC. No adverse events were reported in any of the studies. AUTHORS' CONCLUSIONS SSC appears to be effective as measured by composite pain indicators with both physiological and behavioural indicators and, independently, using heart rate and crying time; and safe for a single painful procedure. Purely behavioural indicators tended to favour SSC but with facial actions there is greater possibility of observers not being blinded. Physiological indicators were mixed although the common measure of heart rate favoured SSC. Two studies compared mother-providers to others, with non-significant results. There was more heterogeneity in the studies with behavioural or composite outcomes. There is a need for replication studies that use similar, clearly defined outcomes. Studies examining optimal duration of SSC, gestational age groups, repeated use, and long-term effects of SSC are needed. Of interest would be to study synergistic effects of SSC with other interventions.
Collapse
Affiliation(s)
| | - Marsha Campbell‐Yeo
- IWK Health CentreNeonatal Intensive Care Unit5850/5980 University AvenuePO Box 9700HalifaxNSCanadaB3K 6R8
| | | | | | - Ananda Fernandes
- Coimbra College of NursingDepartment of Child HealthAv. BissayaBarretoAp. 55CoimbraPortugal3001‐901
| | - David Streiner
- McMaster UniversityDepartment of Psychiatry and Behavioural Neurociences100 West 5th StreetRoom B‐366HamiltonONCanadaL8N 3K7
| | - Darlene Inglis
- IWK Health CentreNeonatal Intensive Care Unit5850/5980 University AvenuePO Box 9700HalifaxNSCanadaB3K 6R8
| | - Rebekah Zee
- IWK Health CentreNeonatal Intensive Care Unit5850/5980 University AvenuePO Box 9700HalifaxNSCanadaB3K 6R8
| | | |
Collapse
|
34
|
Roofthooft DWE, Simons SHP, van Lingen RA, Tibboel D, van den Anker JN, Reiss IKH, van Dijk M. Randomized Controlled Trial Comparing Different Single Doses of Intravenous Paracetamol for Placement of Peripherally Inserted Central Catheters in Preterm Infants. Neonatology 2017; 112:150-158. [PMID: 28558384 PMCID: PMC5637290 DOI: 10.1159/000468975] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/20/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND The availability of a safe and effective pharmacological therapy to reduce procedural pain in preterm infants is limited. The effective analgesic single dose of intravenous paracetamol in preterm infants is unknown. Comparative studies on efficacy of different paracetamol doses in preterm infants are lacking. OBJECTIVES To determine the analgesic effects of different single intravenous paracetamol doses on pain from peripherally inserted central catheter (PICC) placement in preterm infants. METHODS In a blinded randomized controlled trial, the analgesic effects of 10-, 15-, and 20-mg/kg single-dose intravenous paracetamol before PICC placement were compared in neonates with a gestational age <32 weeks. Secondly, a separate age-matched nonrandomized control group receiving oral sucrose was included. Pain was assessed with the Premature Infant Pain Profile (PIPP) and the COMFORTneo score. Peak plasma concentrations of paracetamol were determined. RESULTS A total of 60 patients were included in the paracetamol dose groups (median gestational age = 27.8, IQR: 25.7-29.2 weeks). PIPP scores were comparable: median = 8 (IQR: 6-10.5), 7 (IQR: 6-9), and 8 (IQR: 6-10) for the 10-, 15-, and 20-mg/kg paracetamol groups, respectively (p = 0.94). COMFORTneo scores were not statistically different between the different paracetamol dose groups (p = 0.35). All randomized subjects, except for 3 who received 10 mg/kg of paracetamol, had peak paracetamol concentrations >9 mg/L. PIPP (p = 0.78) and COMFORTneo (p = 0.08) scores were also comparable between paracetamol- and sucrose-treated patients. CONCLUSIONS We found no analgesic benefit from intravenous paracetamol studied in different single doses over sucrose for PICC placement in preterm infants. Paracetamol is not a suitable analgesic for this procedure in preterm infants.
Collapse
Affiliation(s)
- Daniella W E Roofthooft
- Division of Neonatology, Department of Pediatrics, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
35
|
Borenstein-Levin L, Synnes A, Grunau RE, Miller SP, Yoon EW, Shah PS. Narcotics and Sedative Use in Preterm Neonates. J Pediatr 2017; 180:92-98.e1. [PMID: 27614931 DOI: 10.1016/j.jpeds.2016.08.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/15/2016] [Accepted: 08/09/2016] [Indexed: 01/26/2023]
Abstract
OBJECTIVES To evaluate patterns of narcotic and sedative use in neonatal intensive care units (NICUs) across Canada using data collected by the Canadian Neonatal Network. STUDY DESIGN We conducted a retrospective observational cohort study of preterm neonates at <33 weeks' gestation and admitted to a participating Canadian Neonatal Network NICU. The proportion of all neonates who received sedative(s), narcotic(s), or either sedative(s), narcotic(s), or both during their NICU stay was calculated for each year. Because opioids are used for premedication before intubation, only continuous infusions of a narcotic drug were included. Variation in narcotics and sedative usage between sites in 2014 was determined using logistic regression analysis, with adjustment for gestational age, surgery, and mechanical ventilation. RESULTS Of 20 744 neonates, 29% of neonates received a narcotic, a sedative, or both; 23% received a narcotic and 17% a sedative. Although no clinically significant changes in drug exposure were documented during the 5-year period, there were statistically significant differences in narcotic and sedative use between sites, ranging from 3% to 41% for narcotic and 2% to 48% for sedative use (aORs 0.2-5.7 and 0.1-15, respectively, P < .05). CONCLUSIONS Exposure to narcotic or sedative agents is highly variable in preterm neonates across Canada despite concerns of adverse outcomes associated with these drugs. The tremendous variation in practice suggests that further research on their current usage, as well as identifying optimal practice procedures is warranted.
Collapse
Affiliation(s)
- Liron Borenstein-Levin
- British Columbia's Women's Hospital and Health Center, Vancouver, British Columbia, Canada
| | - Anne Synnes
- British Columbia's Women's Hospital and Health Center, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada; Child and Family Research Institute, Vancouver, British Columbia, Canada.
| | - Ruth E Grunau
- British Columbia's Women's Hospital and Health Center, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada; Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Steven P Miller
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada; Child and Family Research Institute, Vancouver, British Columbia, Canada; Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada; Department of Pediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Eugene W Yoon
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Prakesh S Shah
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
36
|
Mitchell AJ, Hall RW, Golianu B, Yates C, Williams DK, Chang J, Anand KJS. Does noninvasive electrical stimulation of acupuncture points reduce heelstick pain in neonates? Acta Paediatr 2016; 105:1434-1439. [PMID: 27607517 DOI: 10.1111/apa.13581] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/22/2016] [Accepted: 09/05/2016] [Indexed: 11/28/2022]
Abstract
AIM Noninvasive electrical stimulation at acupuncture points (NESAP) for analgesia is used in children, but has not been widely studied in neonates. The purpose of this study was to determine whether NESAP alone or in combination with sucrose relieved heelstick pain in neonates. METHODS Term neonates (n = 162) receiving routine heelsticks for newborn screening were enrolled following parental consent. All infants received facilitated tucking and non-nutritive sucking. Neonates were randomised to standard care, sucrose, NESAP or sucrose plus NESAP. NESAP (3.5 mA, 10 Hz) or sham was administered over four acupuncture points. The Premature Infant Pain Profile (PIPP), heart rate variability (HRV) and salivary cortisol were used to measure heelstick pain. RESULTS PIPP scores among all four treatment groups increased during heelstick, F (9,119) = 1.95, p = 0.05 and NESAP therapy had no significant effect on PIPP scores. However, PIPP scores from baseline to heelstick increased the most in the two groups not receiving sucrose (p < 0.01). Mean PIPP scores remained below five during the heelstick in all four groups, indicating minimal or no pain. Differences in HRV and salivary cortisol among groups were insignificant. CONCLUSION NESAP at 3.5 mA, 10 Hz is not effective in relieving pain during heelsticks in neonates.
Collapse
Affiliation(s)
- Anita J. Mitchell
- College of Nursing; University of Arkansas for Medical Sciences; Little Rock AR USA
| | - Richard W. Hall
- Department of Pediatrics and Neonatology; University of Arkansas for Medical Sciences; Little Rock AR USA
| | - Brenda Golianu
- Stanford University and Lucile Packard Children's Hospital Stanford; Palo Alto CA USA
| | - Charlotte Yates
- Research Faculty at University of Arkansas for Medical Sciences; Department of Physical Therapy; University of Central Arkansas; Conway AR USA
| | | | - Jason Chang
- University of Arkansas for Medical Sciences; Little Rock AR USA
| | | |
Collapse
|
37
|
Effect of Vibration on Pain Response to Heel Lance: A Pilot Randomized Control Trial. Adv Neonatal Care 2016; 16:439-448. [PMID: 27533335 DOI: 10.1097/anc.0000000000000315] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Applied mechanical vibration in pediatric and adult populations has been shown to be an effective analgesic for acute and chronic pain, including needle pain. Studies among the neonatal population are lacking. According to the Gate Control Theory, it is expected that applied mechanical vibration will have a summative effect with standard nonpharmacologic pain control strategies, reducing behavioral and physiologic pain responses to heel lancing. PURPOSE To determine the safety and efficacy of mechanical vibration for relief of heel lance pain among neonates. METHODS In this parallel design randomized controlled trial, eligible enrolled term or term-corrected neonates (n = 56) in a level IV neonatal intensive care unit were randomized to receive either sucrose and swaddling or sucrose, swaddling, and vibration for heel lance analgesia. Vibration was applied using a handheld battery-powered vibrator (Norco MiniVibrator, Hz = 92) to the lateral aspect of the lower leg along the sural dermatome throughout the heel lance procedure. Neonatal Pain, Agitation, and Sedation Scale (N-PASS) scores, heart rate, and oxygen saturations were collected at defined intervals surrounding heel lancing. RESULTS Infants in the vibration group (n = 30) had significantly lower N-PASS scores and more stable heart rates during heel stick (P = .006, P = .037) and 2 minutes after heel lance (P = .002, P = .016) than those in the nonvibration group. There were no adverse behavioral or physiologic responses to applied vibration in the sample. IMPLICATIONS FOR PRACTICE AND RESEARCH Applied mechanical vibration is a safe and effective method for managing heel lance pain. This pilot study suggests that mechanical vibration warrants further exploration as a nonpharmacologic pain management tool among the neonatal population.
Collapse
|
38
|
Slater R, Hartley C, Moultrie F, Adams E, Juszczak E, Rogers R, Norman JE, Patel C, Stanbury K, Hoskin A, Green G. A blinded randomised placebo-controlled trial investigating the efficacy of morphine analgesia for procedural pain in infants: Trial protocol. Wellcome Open Res 2016; 1:7. [PMID: 28066825 PMCID: PMC5218543 DOI: 10.12688/wellcomeopenres.10005.2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infant pain has both immediate and long-term negative consequences, yet in clinical practice it is often undertreated. To date, few pain-relieving drugs have been tested in infants. Morphine is a potent analgesic that provides effective pain relief in adults, but there is inconclusive evidence for its effectiveness in infants. The purpose of this study is to establish whether oral morphine provides effective analgesia for procedural pain in infants. A blinded, placebo-controlled, parallel-group randomized, phase II, clinical trial will be undertaken to determine whether morphine sulphate administered orally prior to clinically-required retinopathy of prematurity (ROP) screening and heel lancing provides effective analgesia.
156 infants between 34 and 42 weeks' gestational age who require a clinical heel lance and ROP screening on the same test occasion will be included in the trial. Infants will be randomised to receive either a single dose of morphine sulphate (100 μg/kg) or placebo. Each infant will be monitored for 48 hours and safety data will be collected during the 24 hours following drug administration. The primary outcome will be the Premature Infant Pain Profile-revised (PIPP-R) score 30 seconds after ROP screening. The co-primary outcome will be the magnitude of nociceptive-specific brain activity evoked by a clinically-required heel lance. Infant clinical stability will be assessed by comparing the number of episodes of bradycardia, tachycardia, desaturation and apnoea, and changes in respiratory support requirements in the 24-hour periods before and after the clinical intervention. In addition, drug safety will be assessed by considering the occurrence of apnoeic and hypotensive episodes requiring intervention in the 24-hour period following drug administration. This study has been published as an Accepted Protocol Summary by The Lancet.
Collapse
Affiliation(s)
| | | | - Fiona Moultrie
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Eleri Adams
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Ed Juszczak
- National Perinatal Epidemiology Unit (NPEU), University of Oxford, Oxford, UK
| | - Richard Rogers
- Nuffield Department of Anaesthetics, John Radcliffe Hospital, Oxford, UK
| | - Jane E Norman
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, Edinburgh, UK
| | - Chetan Patel
- Oxford Eye Hospital, John Radcliffe Hospital, Oxford, UK
| | - Kayleigh Stanbury
- National Perinatal Epidemiology Unit (NPEU), University of Oxford, Oxford, UK
| | - Amy Hoskin
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
39
|
Slater R, Hartley C, Moultrie F, Adams E, Juszczak E, Rogers R, Norman JE, Patel C, Stanbury K, Hoskin A, Green G. A blinded randomised placebo-controlled trial investigating the efficacy of morphine analgesia for procedural pain in infants: Trial protocol. Wellcome Open Res 2016. [PMID: 28066825 DOI: 10.12688/wellcomeopenres.10005.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infant pain has both immediate and long-term negative consequences, yet in clinical practice it is often undertreated. To date, few pain-relieving drugs have been tested in infants. Morphine is a potent analgesic that provides effective pain relief in adults, but there is inconclusive evidence for its effectiveness in infants. The purpose of this study is to establish whether oral morphine provides effective analgesia for procedural pain in infants. A blinded, placebo-controlled, parallel-group randomized, phase II, clinical trial will be undertaken to determine whether morphine sulphate administered orally prior to clinically-required retinopathy of prematurity (ROP) screening and heel lancing provides effective analgesia.
156 infants between 34 and 42 weeks' gestational age who require a clinical heel lance and ROP screening on the same test occasion will be included in the trial. Infants will be randomised to receive either a single dose of morphine sulphate (100 μg/kg) or placebo. Each infant will be monitored for 48 hours and safety data will be collected during the 24 hours following drug administration. The primary outcome will be the Premature Infant Pain Profile-revised (PIPP-R) score 30 seconds after ROP screening. The co-primary outcome will be the magnitude of nociceptive-specific brain activity evoked by a clinically-required heel lance. Infant clinical stability will be assessed by comparing the number of episodes of bradycardia, tachycardia, desaturation and apnoea, and changes in respiratory support requirements in the 24-hour periods before and after the clinical intervention. In addition, drug safety will be assessed by considering the occurrence of apnoeic and hypotensive episodes requiring intervention in the 24-hour period following drug administration. This study has been published as an Accepted Protocol Summary by The Lancet.
Collapse
Affiliation(s)
| | | | - Fiona Moultrie
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Eleri Adams
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Ed Juszczak
- National Perinatal Epidemiology Unit (NPEU), University of Oxford, Oxford, UK
| | - Richard Rogers
- Nuffield Department of Anaesthetics, John Radcliffe Hospital, Oxford, UK
| | - Jane E Norman
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, Edinburgh, UK
| | - Chetan Patel
- Oxford Eye Hospital, John Radcliffe Hospital, Oxford, UK
| | - Kayleigh Stanbury
- National Perinatal Epidemiology Unit (NPEU), University of Oxford, Oxford, UK
| | - Amy Hoskin
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
40
|
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
|
41
|
Gao H, Gao H, Xu G, Li M, Du S, Li F, Zhang H, Wang D. Efficacy and safety of repeated oral sucrose for repeated procedural pain in neonates: A systematic review. Int J Nurs Stud 2016; 62:118-25. [DOI: 10.1016/j.ijnurstu.2016.07.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 07/15/2016] [Accepted: 07/15/2016] [Indexed: 11/28/2022]
|
42
|
Ibrahim M, Jones LJ, Lai NM, Tan K. Dexmedetomidine for analgesia and sedation in newborn infants receiving mechanical ventilation. Cochrane Database Syst Rev 2016; 2016:CD012361. [PMCID: PMC6457690 DOI: 10.1002/14651858.cd012361] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
This is the protocol for a review and there is no abstract. The objectives are as follows: To determine the overall effectiveness and safety of dexmedetomidine for sedation and analgesia in newborn infants receiving mechanical ventilation compared with other non‐opioids, opioids or placebo. We will perform subgroup analyses according to method of dexmedetomidine administration; dose of dexmedetomidine; age of initiation of dexmedetomidine; indication for mechanical ventilation; gestational age; and duration of treatment.
Collapse
Affiliation(s)
- Masitah Ibrahim
- Monash Medical CentreMonash Newborn246 Clayton RoadClayton, MelbourneAustralia3168
| | - Lisa J Jones
- University of SydneyCentral Clinical School, Discipline of Obstetrics, Gynaecology and NeonatologySydneyAustralia
| | - Nai Ming Lai
- Taylor's UniversitySchool of MedicineSubang JayaMalaysia
| | - Kenneth Tan
- Monash UniversityDepartment of Paediatrics246 Clayton RoadClaytonMelbourneAustraliaVIC 3168
| |
Collapse
|
43
|
Warnock FF, Craig KD, Bakeman R, Castral T, Mirlashari J. The relationship of prenatal maternal depression or anxiety to maternal caregiving behavior and infant behavior self-regulation during infant heel lance: an ethological time-based study of behavior. BMC Pregnancy Childbirth 2016; 16:264. [PMID: 27604153 PMCID: PMC5477804 DOI: 10.1186/s12884-016-1050-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 08/25/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Sensitive and responsive maternal caregiving behavior strengthens infant self-regulatory capacities (HL), but this regulatory role may be diminished in some mothers with second-trimester prenatal exposure to depression and/ or anxiety (MDA). This study examined maternal and infant behavior during infant heel lance (HL) when mothers had or did not have MDA. Ethological methods and micro-analytic approaches capable of distinguishing and comparing time-based patterning in maternal and infant behavior were used to clarify biological mechanisms, such as MDA, that may underlie observed behavior. Aims were to examine group differences in caregiving behavior between mothers with and without MDA 5 min Pre-HL and 5 min Post-H, and relationships between MDA, maternal caregiving behavior and infant pain behavior self-regulation, concurrently. METHODS At second trimester, mothers were assessed for symptoms of mild-severe depression or anxiety. Mothers whose scores exceeded predetermined cut-off scores on one or more of the mental health measures were allocated to the MDA-exposure group, those below to the non-MDA-exposure group. Reliable observers, blinded to MDA status and study phases, coded video records of the caregiving behavior of each study mother for the full duration of the 5 min Pre-HL and 5 min Post-HL study phases. Group differences and associations between mean measures of maternal mental health scores, time-based measures of maternal behavior, and time-based measures of infant pain behavior regulation (previously coded) were concurrently analyzed using comparative and correlational statistics. RESULTS MDA-exposed mothers spent significantly more time not embracing, engaging or responding to infant cues than maternal controls Pre-HL and Post-HL. MDA was associated with atypical maternal caregiving behavior, which in turn was related to atypical infant pain behavior self-regulation during and after the HL. CONCLUSION Our findings have implication for practice. We recommend inclusion of mothers with MDA and their infants in interventions that strengthen the early mother-infant interaction and mother's regulatory caregiving role. MDA and maternal caregiving behavior must be considered in future infant pain studies to examine if they confound effectiveness of mother driven caregiving interventions for neonatal pain. We highlight the importance of examining maternal mental health throughout the perinatal and postnatal trajectory, and particularly the newborn period.
Collapse
Affiliation(s)
- Fay F Warnock
- Developmental Neurosciences, Child and Family Research Institute, L408, 4480 Oak Street, Vancouver, BC, Canada. .,University of British Columbia (BC), School of Nursing, Vancouver, BC, Canada.
| | - Kenneth D Craig
- Department of Psychology, University of British Columbia (BC), Vancouver, BC, Canada
| | - Roger Bakeman
- Georgia State University, Atlanta, Georgia, 30303, USA
| | - Thaila Castral
- University of Goiás Faculty of Nursing, Goiânia, GO, Brazil
| | - Jila Mirlashari
- School of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Knøsgaard KR, Foster DJR, Kreilgaard M, Sverrisdóttir E, Upton RN, van den Anker JN. Pharmacokinetic models of morphine and its metabolites in neonates:: Systematic comparisons of models from the literature, and development of a new meta-model. Eur J Pharm Sci 2016; 92:117-30. [PMID: 27373670 DOI: 10.1016/j.ejps.2016.06.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 11/19/2022]
Abstract
Morphine is commonly used for pain management in preterm neonates. The aims of this study were to compare published models of neonatal pharmacokinetics of morphine and its metabolites with a new dataset, and to combine the characteristics of the best predictive models to design a meta-model for morphine and its metabolites in preterm neonates. Moreover, the concentration-analgesia relationship for morphine in this clinical setting was also investigated. A population of 30 preterm neonates (gestational age: 23-32weeks) received a loading dose of morphine (50-100μg/kg), followed by a continuous infusion (5-10μg/kg/h) until analgesia was no longer required. Pain was assessed using the Premature Infant Pain Profile. Five published population models were compared using numerical and graphical tests of goodness-of-fit and predictive performance. Population modelling was conducted using NONMEM® and the $PRIOR subroutine to describe the time-course of plasma concentrations of morphine, morphine-3-glucuronide, and morphine-6-glucuronide, and the concentration-analgesia relationship for morphine. No published model adequately described morphine concentrations in this new dataset. Previously published population pharmacokinetic models of morphine, morphine-3-glucuronide, and morphine-6-glucuronide were combined into a meta-model. The meta-model provided an adequate description of the time-course of morphine and the concentrations of its metabolites in preterm neonates. Allometric weight scaling was applied to all clearance and volume terms. Maturation of morphine clearance was described as a function of postmenstrual age, while maturation of metabolite elimination was described as a function of postnatal age. A clear relationship between morphine concentrations and pain score was not established.
Collapse
Affiliation(s)
- Katrine Rørbæk Knøsgaard
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - David John Richard Foster
- Australian Centre for Pharmacometrics and Sansom Institute, School of Pharmaceutical and Medical Sciences, City East Campus, North Terrace, University of South Australia, Adelaide, SA 5000, Australia
| | - Mads Kreilgaard
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Eva Sverrisdóttir
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Richard Neil Upton
- Australian Centre for Pharmacometrics and Sansom Institute, School of Pharmaceutical and Medical Sciences, City East Campus, North Terrace, University of South Australia, Adelaide, SA 5000, Australia.
| | - Johannes N van den Anker
- Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA; Division of Paediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, Switzerland; Intensive Care and Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
45
|
Epidemiology and neonatal pain management of heelsticks in intensive care units: EPIPPAIN 2, a prospective observational study. Int J Nurs Stud 2016; 59:79-88. [DOI: 10.1016/j.ijnurstu.2016.03.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/29/2016] [Accepted: 03/22/2016] [Indexed: 11/18/2022]
|
46
|
Courtois E, Cimerman P, Dubuche V, Goiset MF, Orfèvre C, Lagarde A, Sgaggero B, Guiot C, Goussot M, Huraux E, Nanquette MC, Butel C, Ferreira AM, Lacoste S, Séjourné S, Jolly V, Lajoie G, Maillard V, Guedj R, Chappuy H, Carbajal R. The burden of venipuncture pain in neonatal intensive care units: EPIPPAIN 2, a prospective observational study. Int J Nurs Stud 2016; 57:48-59. [DOI: 10.1016/j.ijnurstu.2016.01.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/27/2016] [Accepted: 01/30/2016] [Indexed: 10/22/2022]
|
47
|
Zwicker JG, Miller SP, Grunau RE, Chau V, Brant R, Studholme C, Liu M, Synnes A, Poskitt KJ, Stiver ML, Tam EWY. Smaller Cerebellar Growth and Poorer Neurodevelopmental Outcomes in Very Preterm Infants Exposed to Neonatal Morphine. J Pediatr 2016; 172:81-87.e2. [PMID: 26763312 PMCID: PMC5462546 DOI: 10.1016/j.jpeds.2015.12.024] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 11/10/2015] [Accepted: 12/09/2015] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To examine the relationship between morphine exposure and growth of the cerebellum and cerebrum in very preterm neonates from early in life to term-equivalent age, as well as to examine morphine exposure and brain volumes in relation to neurodevelopmental outcomes at 18 months corrected age (CA). STUDY DESIGN A prospective cohort of 136 very preterm neonates (24-32 weeks gestational age) was serially scanned with magnetic resonance imaging near birth and at term-equivalent age for volumetric measurements of the cerebellum and cerebrum. Motor outcomes were assessed with the Peabody Developmental Motor Scales, Second Edition and cognitive outcomes with the Bayley Scales of Infant and Toddler Development, Third Edition at 18 months CA. Generalized least squares models and linear regression models were used to assess relationships between morphine exposure, brain volumes, and neurodevelopmental outcomes. RESULTS A 10-fold increase in morphine exposure was associated with a 5.5% decrease in cerebellar volume, after adjustment for multiple clinical confounders and total brain volume (P = .04). When infants exposed to glucocorticoids were excluded, the association of morphine was more pronounced, with an 8.1% decrease in cerebellar volume. Morphine exposure was not associated with cerebral volume (P = .30). Greater morphine exposure also predicted poorer motor (P < .001) and cognitive outcomes (P = .006) at 18 months CA, an association mediated, in part, by slower brain growth. CONCLUSIONS Morphine exposure in very preterm neonates is independently associated with impaired cerebellar growth in the neonatal period and poorer neurodevelopmental outcomes in early childhood. Alternatives to better manage pain in preterm neonates that optimize brain development and functional outcomes are urgently needed.
Collapse
Affiliation(s)
- Jill G Zwicker
- Department of Occupational Science and Occupational Therapy, University of British Columbia, Vancouver, Canada; Department of Pediatrics, University of British Columbia, Vancouver, Canada; Child and Family Research Institute, Vancouver, Canada.
| | - Steven P Miller
- Department of Pediatrics, University of British Columbia, Vancouver, Canada,Child & Family Research Institute, Vancouver, Canada,Department of Pediatrics, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Ruth E Grunau
- Department of Pediatrics, University of British Columbia, Vancouver, Canada,Child & Family Research Institute, Vancouver, Canada,British Columbia Women’s Hospital, Vancouver, Canada
| | - Vann Chau
- Department of Pediatrics, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Rollin Brant
- Child & Family Research Institute, Vancouver, Canada,Department of Statistics, University of British Columbia, Vancouver, Canada
| | - Colin Studholme
- Departments of Bioengineering & Pediatrics, University of Washington, Seattle, USA
| | - Mengyuan Liu
- Departments of Bioengineering & Pediatrics, University of Washington, Seattle, USA
| | - Anne Synnes
- Department of Pediatrics, University of British Columbia, Vancouver, Canada,Child & Family Research Institute, Vancouver, Canada,British Columbia Women’s Hospital, Vancouver, Canada
| | - Kenneth J Poskitt
- Child & Family Research Institute, Vancouver, Canada,Department of Radiology, University of British Columbia, Vancouver, Canada
| | - Mikaela L Stiver
- Department of Pediatrics, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Emily WY Tam
- Department of Pediatrics, Hospital for Sick Children and University of Toronto, Toronto, Canada
| |
Collapse
|
48
|
Witt N, Coynor S, Edwards C, Bradshaw H. A Guide to Pain Assessment and Management in the Neonate. CURRENT EMERGENCY AND HOSPITAL MEDICINE REPORTS 2016; 4:1-10. [PMID: 27073748 PMCID: PMC4819510 DOI: 10.1007/s40138-016-0089-y] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Newborn infants experience acute pain with various medical procedures. Evidence demonstrates that controlling pain in the newborn period is beneficial, improving physiologic, behavioral, and hormonal outcomes. Multiple validated scoring systems exist to assess pain in a neonate; however, there is no standardized or universal approach for pain management. Healthcare facilities should establish a neonatal pain control program. The first step is to minimize the total number of painful iatrogenic events when possible. If a procedure cannot be avoided, a tiered approach to manage pain using environmental, non-pharmacologic, and pharmacologic modalities is recommended. This systematic approach should decrease acute neonatal pain, poor outcomes, and provider and parent dissatisfaction.
Collapse
Affiliation(s)
- Norina Witt
- />Department of Pediatrics, The University of Arizona, PO Box 245073, Tuscon, 85724 AZ USA
| | - Seth Coynor
- />Department of Pediatrics, The University of Arizona, PO Box 245073, Tuscon, 85724 AZ USA
| | - Christopher Edwards
- />Departments of Pediatrics and Emergency Medicine, The University of Arizona, PO Box 245057, Tucson, 85724-5057 AZ USA
- />College of Pharmacy, The University of Arizona, Department of Pharmacy Services, Banner-University Medical Center, PO Box 210202, Tucson, AZ USA
| | - Hans Bradshaw
- />Departments of Pediatrics and Emergency Medicine, The University of Arizona, PO Box 245057, Tucson, 85724-5057 AZ USA
| |
Collapse
|
49
|
Pediatric Cardiac Intensive Care Society 2014 Consensus Statement: Pharmacotherapies in Cardiac Critical Care: Sedation, Analgesia and Muscle Relaxant. Pediatr Crit Care Med 2016; 17:S3-S15. [PMID: 26945327 DOI: 10.1097/pcc.0000000000000619] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE This article reviews pharmacotherapies currently available to manage sedation, analgesia, and neuromuscular blockade for pediatric cardiac critical patients. DATA SOURCES The knowledge base of an expert panel of pharmacists, cardiac anesthesiologists, and a cardiac critical care physician involved in the care of pediatric cardiac critical patients was combined with a comprehensive search of the medical literature to generate the data source. STUDY SELECTION The panel examined all studies relevant to management of sedation, analgesia, and neuromuscular blockade in pediatric cardiac critical patients. DATA EXTRACTION Each member of the panel was assigned a specific subset of the studies relevant to their particular area of expertise (pharmacokinetics, pharmacodynamics, and clinical care) to review and analyze. DATA SYNTHESIS The panel members each crafted a comprehensive summary of the literature relevant to their area of expertise. The panel, as a whole, then collaborated to cohesively summarize all the available, relevant literature. CONCLUSIONS In the cardiac ICU, management of the cardiac patient requires an individualized sedative and analgesic strategy that maintains hemodynamic stability. Multiple pharmacological therapies exist to achieve these goals and should be selected based on the patient's underlying physiology, hemodynamic vulnerabilities, desired level of sedation and analgesia, and the projected short- or long-term recovery trajectory.
Collapse
|
50
|
Dezhdar S, Jahanpour F, Firouz Bakht S, Ostovar A. The Effects of Kangaroo Mother Care and Swaddling on Venipuncture Pain in Premature Neonates: A Randomized Clinical Trial. IRANIAN RED CRESCENT MEDICAL JOURNAL 2016; 18:e29649. [PMID: 27274399 PMCID: PMC4894081 DOI: 10.5812/ircmj.29649] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 09/24/2015] [Accepted: 10/19/2015] [Indexed: 01/03/2023]
Abstract
Background Hospitalized premature babies often undergo various painful procedures. Kangaroo mother care (KMC) and swaddling are two pain reduction methods. Objectives This study was undertaken to compare the effects of swaddling and KMC on pain during venous sampling in premature neonates. Patients and Methods This study was performed as a randomized clinical trial on 90 premature neonates. The neonates were divided into three groups using a random allocation block. The three groups were group A (swaddling), group B (KMC), and group C (control). In all three groups, the heart rate and arterial oxygen saturation were measured and recorded in time intervals of 30 seconds before, during, and 30, 60, 90, and 120 seconds after blood sampling. The neonate’s face was video recorded and assessed using the premature infant pain profile (PIPP) at time intervals of 30 seconds. The data was analyzed using the t-test, chi-square test, Repeated Measure analysis of variance (ANOVA), Kruskal-Wallis, Post-hoc, and Bonferroni test. Results The findings revealed that pain was reduced to a great extent in the swaddling and KMC methods compared to the control group. However, there was no significant difference between KMC and swaddling (P ≥ 0.05). Conclusions The results of this study indicate that there is no meaningful difference between swaddling and KMC on physiological indexes and pain in neonates. Therefore, the swaddling method may be a good substitute for KMC.
Collapse
Affiliation(s)
- Shahin Dezhdar
- Bushehr University of Medical Sciences, Bushehr, IR Iran
| | - Faezeh Jahanpour
- Nursing and Midwifery Faculty, Bushehr University of Medical Sciences, Bushehr, IR Iran
- Corresponding Author: Faezeh Jahanpour, Nursing and Midwifery Faculty, Bushehr University of Medical Sciences, Bushehr, IR Iran. Tel: +98-9177720846, Fax: +98-7714550187, E-mail:
| | | | - Afshin Ostovar
- The Persian Gulf Tropical Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, IR Iran
| |
Collapse
|