Neonatal pain

Suellen M Walker, Suellen M Walker

Abstract

Effective management of procedural and postoperative pain in neonates is required to minimize acute physiological and behavioral distress and may also improve acute and long-term outcomes. Painful stimuli activate nociceptive pathways, from the periphery to the cortex, in neonates and behavioral responses form the basis for validated pain assessment tools. However, there is an increasing awareness of the need to not only reduce acute behavioral responses to pain in neonates, but also to protect the developing nervous system from persistent sensitization of pain pathways and potential damaging effects of altered neural activity on central nervous system development. Analgesic requirements are influenced by age-related changes in both pharmacokinetic and pharmacodynamic response, and increasing data are available to guide safe and effective dosing with opioids and paracetamol. Regional analgesic techniques provide effective perioperative analgesia, but higher complication rates in neonates emphasize the importance of monitoring and choice of the most appropriate drug and dose. There have been significant improvements in the understanding and management of neonatal pain, but additional research evidence will further reduce the need to extrapolate data from older age groups. Translation into improved clinical care will continue to depend on an integrated approach to implementation that encompasses assessment and titration against individual response, education and training, and audit and feedback.

Keywords: NICU; neonate; neurodevelopment; opioids; pain; regional analgesia.

© 2013 The Authors. Pediatric Anesthesia published by John Wiley & Sons Ltd.

References

    1. Association of Paediatric Anaesthetists of Great Britain and Ireland. Good practice in postoperative and procedural pain management, 2nd edition. Pediatr Anesth. 2012;22(Suppl 1):1–79.
    1. Macintyre PE, Schug SA, Scott DA, et al. Acute Pain Management: Scientific Evidence. 3rd edn. Melbourne: ANZCA & FPM; 2010.
    1. Groenewald CB, Rabbitts JA, Schroeder DR, et al. Prevalence of moderate-severe pain in hospitalized children. Pediatr Anesth. 2012;22:661–668.
    1. Stevens BJ, Harrison D, Rashotte J, et al. Pain assessment and intensity in hospitalized children in Canada. J Pain. 2012;13:857–865.
    1. Foster J, Spence K, Henderson-Smart D, et al. Procedural pain in neonates in Australian hospitals: a survey update of practices. J Paediatr Child Health. 2013;49:E35–E39.
    1. Lago P, Boccuzzo G, Garetti E, et al. Pain management during invasive procedures at Italian NICUs: has anything changed in the last five years? J Matern Fetal Neonatal Med. 2013;26:303–305.
    1. Deindl P, Unterasinger L, Kappler G, et al. Successful implementation of a neonatal pain and sedation protocol at 2 NICUs. Pediatrics. 2013;132:e211–e218.
    1. Howard RF, Lloyd-Thomas A, Thomas M, et al. Nurse-controlled analgesia (NCA) following major surgery in 10,000 patients in a children's hospital. Pediatr Anesth. 2010;20:126–134.
    1. Fitzgerald M, Walker SM. Infant pain management: a developmental neurobiological approach. Nat Clin Pract Neurol. 2009;5:35–50.
    1. Baccei ML. Modulation of developing dorsal horn synapses by tissue injury. Ann N Y Acad Sci. 2010;1198:159–167.
    1. Koch SC, Tochiki KK, Hirschberg S, et al. C-fiber activity-dependent maturation of glycinergic inhibition in the spinal dorsal horn of the postnatal rat. Proc Natl Acad Sci USA. 2012;109:12201–12206.
    1. Fitzgerald M. The development of nociceptive circuits. Nat Rev Neurosci. 2005;6:507–520.
    1. Slater R, Cornelissen L, Fabrizi L, et al. Oral sucrose as an analgesic drug for procedural pain in newborn infants: a randomised controlled trial. Lancet. 2010;376:1225–1232.
    1. Fitzgerald M, Millard C, MacIntosh N. Hyperalgesia in premature infants. Lancet. 1988;1:292.
    1. Andrews K, Fitzgerald M. Wound sensitivity as a measure of analgesic effects following surgery in human neonates and infants. Pain. 2002;99:185–195.
    1. Slater R, Cantarella A, Gallella S, et al. Cortical pain responses in human infants. J Neurosci. 2006;26:3662–3666.
    1. Bartocci M, Bergqvist LL, Lagercrantz H, et al. Pain activates cortical areas in the preterm newborn brain. Pain. 2006;122:109–117.
    1. Slater R, Worley A, Fabrizi L, et al. Evoked potentials generated by noxious stimulation in the human infant brain. Eur J Pain. 2010;14:321–326.
    1. McKeever S, Johnston L, Davidson A. A review of the utility of EEG depth of anaesthesia monitors in the paediatric intensive care environment. Intensive Crit Care Nurs. 2012;28:294–303.
    1. Fabrizi L, Slater R, Worley A, et al. A shift in sensory processing that enables the developing human brain to discriminate touch from pain. Curr Biol. 2011;21:1552–1558.
    1. Slater R, Fabrizi L, Worley A, et al. Premature infants display increased noxious-evoked neuronal activity in the brain compared to healthy age-matched term-born infants. Neuroimage. 2010;52:583–589.
    1. Maxwell LG, Malavolta CP, Fraga MV. Assessment of pain in the neonate. Clin Perinatol. 2013;40:457–469.
    1. Royal College of Nursing. The Recognition and Assessment of Acute Pain in Children. Clinical Practice Guidelines. London: RCN; 2009.
    1. Stevens B, Johnston C, Petryshen P, et al. Premature infant pain profile: development and initial validation. Clin J Pain. 1996;12:13–22.
    1. Merkel SI, Voepel-Lewis T, Shayevitz JR, et al. The FLACC: a behavioral scale for scoring postoperative pain in young children. Pediatr Nurs. 1997;23:293–297.
    1. Ambuel B, Hamlett KW, Marx CM, et al. Assessing distress in pediatric intensive care environments: the COMFORT scale. J Pediatr Psychol. 1992;17:95–109.
    1. van Dijk M, de Boer JB, Koot HM, et al. The reliability and validity of the COMFORT scale as a postoperative pain instrument in 0 to 3-year-old infants. Pain. 2000;84:367–377.
    1. Anand KJ, Sippell WG, Aynsley-Green A. Randomised trial of fentanyl anaesthesia in preterm babies undergoing surgery: effects on the stress response. Lancet. 1987;1:62–66.
    1. Walker SM. Biological and neurodevelopmental implications of neonatal pain. Clin Perinatol. 2013;40:471–491.
    1. Holsti L, Grunau RE, Shany E. Assessing pain in preterm infants in the neonatal intensive care unit: moving to a ‘brain-oriented’ approach. Pain Manag. 2011;1:171–179.
    1. Stevens B, Yamada J, Lee GY, et al. Sucrose for analgesia in newborn infants undergoing painful procedures. Cochrane Database Syst Rev. 2013;1:CD001069.
    1. Taddio A, Shah V, Atenafu E, et al. Influence of repeated painful procedures and sucrose analgesia on the development of hyperalgesia in newborn infants. Pain. 2009;144:43–48.
    1. Taddio A, Katz J, Ilersich AL, et al. Effect of neonatal circumcision on pain response during subsequent routine vaccination. Lancet. 1997;349:599–603.
    1. Peters JW, Schouw R, Anand KJ, et al. Does neonatal surgery lead to increased pain sensitivity in later childhood? Pain. 2005;114:444–454.
    1. Hermann C, Hohmeister J, Demirakca S, et al. Long-term alteration of pain sensitivity in school-aged children with early pain experiences. Pain. 2006;125:278–285.
    1. Hohmeister J, Demirakca S, Zohsel K, et al. Responses to pain in school-aged children with experience in a neonatal intensive care unit: cognitive aspects and maternal influences. Eur J Pain. 2009;13:94–101.
    1. Walker SM, Franck LS, Fitzgerald M, et al. Long-term impact of neonatal intensive care and surgery on somatosensory perception in children born extremely preterm. Pain. 2009;141:79–87.
    1. Ririe DG, Vernon TL, Tobin JR, et al. Age-dependent responses to thermal hyperalgesia and mechanical allodynia in a rat model of acute postoperative pain. Anesthesiology. 2003;99:443–448.
    1. Ririe DG, Bremner LR, Fitzgerald M. Comparison of the immediate effects of surgical incision on dorsal horn neuronal receptive field size and responses during postnatal development. Anesthesiology. 2008;109:698–706.
    1. Walker SM, Tochiki KK, Fitzgerald M. Hindpaw incision in early life increases the hyperalgesic response to repeat surgical injury: critical period and dependence on initial afferent activity. Pain. 2009;147:99–106.
    1. Li J, Walker SM, Fitzgerald M, et al. Activity-dependent modulation of glutamatergic signaling in the developing rat dorsal horn by early tissue injury. J Neurophysiol. 2009;102:2208–2219.
    1. Li J, Blankenship ML, Baccei ML. Deficits in glycinergic inhibition within adult spinal nociceptive circuits after neonatal tissue damage. Pain. 2013;154:1129–1139.
    1. Li J, Baccei ML. Neonatal tissue damage facilitates nociceptive synaptic input to the developing superficial dorsal horn via NGF-dependent mechanisms. Pain. 2011;152:1846–1855.
    1. Beggs S, Currie G, Salter MW, et al. Priming of adult pain responses by neonatal pain experience: maintenance by central neuroimmune activity. Brain. 2012;135:404–417.
    1. Ririe DG, Barclay D, Prout H, et al. Preoperative sciatic nerve block decreases mechanical allodynia more in young rats: is preemptive analgesia developmentally modulated? Anesth Analg. 2004;99:140–145.
    1. Grunau RE, Whitfield MF, Petrie-Thomas J, et al. Neonatal pain, parenting stress and interaction, in relation to cognitive and motor development at 8 and 18 months in preterm infants. Pain. 2009;143:138–146.
    1. Vinall J, Miller SP, Chau V, et al. Neonatal pain in relation to postnatal growth in infants born very preterm. Pain. 2012;153:1374–1381.
    1. Brummelte S, Grunau RE, Chau V, et al. Procedural pain and brain development in premature newborns. Ann Neurol. 2012;71:385–396.
    1. Zwicker JG, Grunau RE, Adams E, et al. Score for neonatal acute physiology-II and neonatal pain predict corticospinal tract development in premature newborns. Pediatr Neurol. 2013;48:123–129. e121.
    1. Filan PM, Hunt RW, Anderson PJ, et al. Neurologic outcomes in very preterm infants undergoing surgery. J Pediatr. 2012;160:409–414.
    1. Kabra NS, Schmidt B, Roberts RS, et al. Neurosensory impairment after surgical closure of patent ductus arteriosus in extremely low birth weight infants: results from the Trial of Indomethacin Prophylaxis in Preterms. J Pediatr. 2007;150:229–234. e221.
    1. Rees CM, Pierro A, Eaton S. Neurodevelopmental outcomes of neonates with medically and surgically treated necrotizing enterocolitis. Arch Dis Child Fetal Neonatal Ed. 2007;92:F193–F198.
    1. Schulzke SM, Deshpande GC, Patole SK. Neurodevelopmental outcomes of very low-birth-weight infants with necrotizing enterocolitis: a systematic review of observational studies. Arch Pediatr Adolesc Med. 2007;161:583–590.
    1. Anjari M, Counsell SJ, Srinivasan L, et al. The association of lung disease with cerebral white matter abnormalities in preterm infants. Pediatrics. 2009;124:268–276.
    1. Hall RW, Kronsberg SS, Barton BA, et al. Morphine, hypotension, and adverse outcomes among preterm neonates: who's to blame? Secondary results from the NEOPAIN trial. Pediatrics. 2005;115:1351–1359.
    1. Bouwmeester NJ, Hop WC, van Dijk M, et al. Postoperative pain in the neonate: age-related differences in morphine requirements and metabolism. Intensive Care Med. 2003;29:2009–2015.
    1. Taylor J, Liley A, Anderson BJ. The relationship between age and morphine infusion rate in children. Pediatr Anesth. 2013;23:40–44.
    1. Holford NH, Ma SC, Anderson BJ. Prediction of morphine dose in humans. Pediatr Anesth. 2012;22:209–222.
    1. Anderson BJ. Pharmacology in the very young: anaesthetic implications. Eur J Anaesthesiol. 2012;29:261–270.
    1. Wang C, Sadhavisvam S, Krekels EH, et al. Developmental changes in morphine clearance across the entire paediatric age range are best described by a bodyweight-dependent exponent model. Clin Drug Investig. 2013;33:523–534.
    1. Nandi R, Beacham D, Middleton J, et al. The functional expression of mu opioid receptors on sensory neurons is developmentally regulated; morphine analgesia is less selective in the neonate. Pain. 2004;111:38–50.
    1. Marsh D, Dickenson A, Hatch D, et al. Epidural opioid analgesia in infant rats II: responses to carrageenan and capsaicin. Pain. 1999;82:33–38.
    1. Marsh D, Dickenson A, Hatch D, et al. Epidural opioid analgesia in infant rats I: mechanical and heat responses. Pain. 1999;82:23–32.
    1. Westin BD, Walker SM, Deumens R, et al. Validation of a preclinical spinal safety model: effects of intrathecal morphine in the neonatal rat. Anesthesiology. 2010;113:183–199.
    1. Beland B, Fitzgerald M. Mu-and delta-opioid receptors are downregulated in the largest diameter primary sensory neurons during postnatal development in rats. Pain. 2001;90:143–150.
    1. Rahman W, Dashwood MR, Fitzgerald M, et al. Postnatal development of multiple opioid receptors in the spinal cord and development of spinal morphine analgesia. Brain Res Dev Brain Res. 1998;108:239–254.
    1. Windh RT, Kuhn CM. Increased sensitivity to mu opiate antinociception in the neonatal rat despite weaker receptor-guanyl nucleotide binding protein coupling. J Pharmacol Exp Ther. 1995;273:1353–1360.
    1. Morton NS, Errera A. APA national audit of pediatric opioid infusions. Pediatr Anesth. 2010;20:119–125.
    1. Hume-Smith H, McCormack J, Montgomery C, et al. The effect of age on the dose of remifentanil for tracheal intubation in infants and children. Pediatr Anesth. 2010;20:19–27.
    1. Allegaert K, Tibboel D, van den Anker J. Pharmacological treatment of neonatal pain: in search of a new equipoise. Semin Fetal Neonatal Med. 2013;18:42–47.
    1. Welzing L, Oberthuer A, Junghaenel S, et al. Remifentanil/midazolam versus fentanyl/midazolam for analgesia and sedation of mechanically ventilated neonates and young infants: a randomized controlled trial. Intensive Care Med. 2012;38:1017–1024.
    1. Sammartino M, Garra R, Sbaraglia F, et al. Experience of remifentanil in extremely low-birth-weight babies undergoing laparotomy. Pediatr Neonatol. 2011;52:176–179.
    1. Norman E, Wikstrom S, Rosen I, et al. Premedication for intubation with morphine causes prolonged depression of electrocortical background activity in preterm infants. Pediatr Res. 2013;73:87–94.
    1. Lago P, Tiozzo C, Boccuzzo G, et al. Remifentanil for percutaneous intravenous central catheter placement in preterm infant: a randomized controlled trial. Pediatr Anesth. 2008;18:736–744.
    1. Allegaert K. The clinical pharmacology of short acting analgo-sedatives in neonates. Curr Clin Pharmacol. 2011;6:222–226.
    1. Hudak ML, Tan RC, et al. Committee On Drugs. Neonatal drug withdrawal. Pediatrics. 2012;129:e540–e560.
    1. Cramton RE, Gruchala NE. Babies breaking bad: neonatal and iatrogenic withdrawal syndromes. Curr Opin Pediatr. 2013;25:532–542.
    1. Anand KJ, Willson DF, Berger J, et al. Tolerance and withdrawal from prolonged opioid use in critically ill children. Pediatrics. 2010;125:e1208–e1225.
    1. Jenkins IA, Playfor SD, Bevan C, et al. Current United Kingdom sedation practice in pediatric intensive care. Pediatr Anesth. 2007;17:675–683.
    1. Franck LS, Scoppettuolo LA, Wypij D, et al. Validity and generalizability of the Withdrawal Assessment Tool-1 (WAT-1) for monitoring iatrogenic withdrawal syndrome in pediatric patients. Pain. 2012;153:142–148.
    1. Anand KJ, Hall RW, Desai N, et al. Effects of morphine analgesia in ventilated preterm neonates: primary outcomes from the NEOPAIN randomised trial. Lancet. 2004;363:1673–1682.
    1. Bellu R, de Waal KA, Zanini R. Opioids for neonates receiving mechanical ventilation. Cochrane Database Syst Rev. 2008;(1):CD004212.
    1. Roze JC, Denizot S, Carbajal R, et al. Prolonged sedation and/or analgesia and 5-year neurodevelopment outcome in very preterm infants: results from the EPIPAGE cohort. Arch Pediatr Adolesc Med. 2008;162:728–733.
    1. Suarez A, Knoppert DC, Lee DS, et al. Opioid infusions in the neonatal intensive care unit. J Pediatr Pharmacol Ther. 2010;15:142–146.
    1. de Graaf J, van Lingen RA, Simons SH, et al. Long-term effects of routine morphine infusion in mechanically ventilated neonates on children's functioning: five-year follow-up of a randomized controlled trial. Pain. 2011;152:1391–1397.
    1. de Graaf J, van Lingen RA, Valkenburg AJ, et al. Does neonatal morphine use affect neuropsychological outcomes at 8 to 9 years of age? Pain. 2013;154:449–458.
    1. Jevtovic-Todorovic V, Absalom AR, Blomgren K, et al. Anaesthetic neurotoxicity and neuroplasticity: an expert group report and statement based on the BJA Salzburg Seminar. Br J Anaesth. 2013;111:143–151.
    1. Sanders RD, Hassell J, Davidson AJ, et al. Impact of anaesthetics and surgery on neurodevelopment: an update. Br J Anaesth. 2013;110(Suppl 1):i53–i72.
    1. Stratmann G. Review article: neurotoxicity of anesthetic drugs in the developing brain. Anesth Analg. 2011;113:1170–1179.
    1. Bajic D, Berde CB, Commons KG. Periaqueductal gray neuroplasticity following chronic morphine varies with age: role of oxidative stress. Neuroscience. 2012;226:165–177.
    1. Bajic D, Commons KG, Soriano SG. Morphine-enhanced apoptosis in selective brain regions of neonatal rats. Int J Dev Neurosci. 2013;31:258–266.
    1. Lim G, Wang S, Lim JA, et al. Activity of adenylyl cyclase and protein kinase A contributes to morphine-induced spinal apoptosis. Neurosci Lett. 2005;389:104–108.
    1. Rizzi S, Ori C, Jevtovic-Todorovic V. Timing versus duration: determinants of anesthesia-induced developmental apoptosis in the young mammalian brain. Ann N Y Acad Sci. 2010;1199:43–51.
    1. Rizzi S, Carter LB, Ori C, et al. Clinical anesthesia causes permanent damage to the fetal guinea pig brain. Brain Pathol. 2008;18:198–210.
    1. Duhrsen L, Simons SH, Dzietko M, et al. Effects of repetitive exposure to pain and morphine treatment on the neonatal rat brain. Neonatology. 2013;103:35–43.
    1. Hansen TG, O'Brien K, Morton NS, et al. Plasma paracetamol concentrations and pharmacokinetics following rectal administration in neonates and young infants. Acta Anaesthesiol Scand. 1999;43:855–859.
    1. Allegaert K, Palmer GM, Anderson BJ. The pharmacokinetics of intravenous paracetamol in neonates: size matters most. Arch Dis Child. 2011;96:575–580.
    1. Palmer GM, Atkins M, Anderson BJ, et al. I.V. acetaminophen pharmacokinetics in neonates after multiple doses. Br J Anaesth. 2008;101:523–530.
    1. Anderson BJ, Allegaert K. Intravenous neonatal paracetamol dosing: the magic of 10 days. Pediatr Anesth. 2009;19:289–295.
    1. Anderson BJ. Paracetamol (Acetaminophen): mechanisms of action. Paediatr Anaesth. 2008;18:915–921.
    1. Smith HS. Potential analgesic mechanisms of acetaminophen. Pain Physician. 2009;12:269–280.
    1. Engstrom Ruud L, Wilhelms DB, Eskilsson A, et al. Acetaminophen reduces lipopolysaccharide-induced fever by inhibiting cyclooxygenase-2. Neuropharmacology. 2013;71:124–129.
    1. Ruggieri V, Vitale G, Pini LA, et al. Differential involvement of opioidergic and serotonergic systems in the antinociceptive activity of N-arachidonoyl-phenolamine (AM404) in the rat: comparison with paracetamol. Naunyn Schmiedebergs Arch Pharmacol. 2008;377:219–229.
    1. Dogrul A, Seyrek M, Akgul EO, et al. Systemic paracetamol-induced analgesic and antihyperalgesic effects through activation of descending serotonergic pathways involving spinal 5-HT(7) receptors. Eur J Pharmacol. 2012;677:93–101.
    1. Crawley B, Saito O, Malkmus S, et al. Acetaminophen prevents hyperalgesia in central pain cascade. Neurosci Lett. 2008;442:50–53.
    1. Malmberg AB, Yaksh TL. Antinociceptive actions of spinal nonsteroidal anti-inflammatory agents on the formalin test in the rat. J Pharmacol Exp Ther. 1992;263:136–146.
    1. Shah V, Taddio A, Ohlsson A. Randomised controlled trial of paracetamol for heel prick pain in neonates. Arch Dis Child Fetal Neonatal Ed. 1998;79:F209–F211.
    1. Allegaert K, Naulaers G, Vanhaesebrouck S, et al. The paracetamol concentration-effect relation in neonates. Pediatr Anesth. 2013;23:45–50.
    1. Wong I, St John-Green C, Walker SM. Opioid-sparing effects of perioperative paracetamol and nonsteroidal anti-inflammatory drugs (NSAIDs) in children. Pediatr Anesth. 2013;23:475–495.
    1. van der Marel CD, Peters JW, Bouwmeester NJ, et al. Rectal acetaminophen does not reduce morphine consumption after major surgery in young infants. Br J Anaesth. 2007;98:372–379.
    1. Ceelie I, de Wildt SN, van Dijk M, et al. Effect of intravenous paracetamol on postoperative morphine requirements in neonates and infants undergoing major noncardiac surgery: a randomized controlled trial. JAMA. 2013;309:149–154.
    1. Oncel MY, Yurttutan S, Degirmencioglu H, et al. Intravenous paracetamol treatment in the management of patent ductus arteriosus in extremely low birth weight infants. Neonatology. 2013;103:166–169.
    1. Allegaert K, Anderson B, Simons S, et al. Paracetamol to induce ductus arteriosus closure: is it valid? Arch Dis Child. 2013;98:462–466.
    1. Oncel MY, Yurttutan S, Uras N, et al. An alternative drug (paracetamol) in the management of patent ductus arteriosus in ibuprofen-resistant or contraindicated preterm infants. Arch Dis Child Fetal Neonatal Ed. 2013;98:F94.
    1. Allegaert K, Naulaers G. Haemodynamics of intravenous paracetamol in neonates. Eur J Clin Pharmacol. 2010;66:855–858.
    1. Nevin DG, Shung J. Intravenous paracetamol overdose in a preterm infant during anesthesia. Pediatr Anesth. 2010;20:105–107.
    1. Porta R, Sanchez L, Nicolas M, et al. Lack of toxicity after paracetamol overdose in a extremely preterm neonate. Eur J Clin Pharmacol. 2012;68:901–902.
    1. Beringer RM, Thompson JP, Parry S, et al. Intravenous paracetamol overdose: two case reports and a change to national treatment guidelines. Arch Dis Child. 2011;96:307–308.
    1. Ji P, Wang Y, Li Z, et al. Regulatory review of acetaminophen clinical pharmacology in young pediatric patients. J Pharm Sci. 2012;101:4383–4389.
    1. Etminan M, Sadatsafavi M, Jafari S, et al. Acetaminophen use and the risk of asthma in children and adults: a systematic review and metaanalysis. Chest. 2009;136:1316–1323.
    1. Beasley R, Clayton T, Crane J, et al. Association between paracetamol use in infancy and childhood, and risk of asthma, rhinoconjunctivitis, and eczema in children aged 6-7 years: analysis from Phase Three of the ISAAC programme. Lancet. 2008;372:1039–1048.
    1. Schnabel E, Heinrich J, Group LS. Respiratory tract infections and not paracetamol medication during infancy are associated with asthma development in childhood. J Allergy Clin Immunol. 2010;126:1071–1073.
    1. Lowe AJ, Carlin JB, Bennett CM, et al. Paracetamol use in early life and asthma: prospective birth cohort study. BMJ. 2010;341:c4616.
    1. Headley J, Northstone K. Medication administered to children from 0 to 7.5 years in the Avon Longitudinal Study of Parents and Children (ALSPAC) Eur J Clin Pharmacol. 2007;63:189–195.
    1. Lonnqvist PA. Themed issue ‘Pediatric Regional Anesthesia’-starting 2012 with a bang! Pediatr Anesth. 2012;22:1–2.
    1. Lonnqvist PA. Regional anaesthesia and analgesia in the neonate. Best Pract Res Clin Anaesthesiol. 2010;24:309–321.
    1. Walker SM, Yaksh TL. Neuraxial analgesia in neonates and infants: a review of clinical and preclinical strategies for the development of safety and efficacy data. Anesth Analg. 2012;115:638–662.
    1. Brady-Fryer B, Wiebe N, Lander JA. Pain relief for neonatal circumcision. Cochrane Database Syst Rev. 2004;(4):CD004217.
    1. Ecoffey C, Lacroix F, Giaufre E, et al. Epidemiology and morbidity of regional anesthesia in children: a follow-up one-year prospective survey of the French-Language Society of Paediatric Anaesthesiologists (ADARPEF) Pediatr Anesth. 2010;20:1061–1069.
    1. Mai CL, Young MJ, Quraishi SA. Clinical implications of the transversus abdominis plane block in pediatric anesthesia. Pediatr Anesth. 2012;22:831–840.
    1. Leelanukrom R, Suraseranivongse S, Boonrukwanich V, et al. Effect of wound infiltration with bupivacaine on postoperative analgesia in neonates and infants undergoing major abdominal surgery: a pilot randomized controlled trial. J Anesth. 2012;26:541–544.
    1. Rodriguez-Navarro AJ, Berde CB, Wiedmaier G, et al. Comparison of neosaxitoxin versus bupivacaine via port infiltration for postoperative analgesia following laparoscopic cholecystectomy: a randomized, double-blind trial. Reg Anesth Pain Med. 2011;36:103–109.
    1. Roberson DP, Binshtok AM, Blasl F, et al. Targeting of sodium channel blockers into nociceptors to produce long-duration analgesia: a systematic study and review. Br J Pharmacol. 2011;164:48–58.
    1. Berde CB, Athiraman U, Yahalom B, et al. Tetrodotoxin-bupivacaine-epinephrine combinations for prolonged local anesthesia. Mar Drugs. 2011;9:2717–2728.
    1. Kokki H. Spinal blocks. Pediatr Anesth. 2012;22:56–64.
    1. Johr M, Berger TM. Caudal blocks. Pediatr Anesth. 2012;22:44–50.
    1. Bosenberg A. Benefits of regional anesthesia in children. Pediatr Anesth. 2012;22:10–18.
    1. Llewellyn N, Moriarty A. The national pediatric epidural audit. Pediatr Anesth. 2007;17:520–533.
    1. Polaner DM, Taenzer AH, Walker BJ, et al. Pediatric Regional Anesthesia Network (PRAN): a multi-institutional study of the use and incidence of complications of pediatric regional anesthesia. Anesth Analg. 2012;115:1353–1364.
    1. Wong GK, Arab AA, Chew SC, et al. Major complications related to epidural analgesia in children: a 15-year audit of 3,152 epidurals. Can J Anaesth. 2013;60:355–363.
    1. Flandin-Blety C, Barrier G. Accidents following extradural analgesia in children. The results of a retrospective study. Pediatr Anesth. 1995;5:41–46.
    1. van Niekerk J, Bax-Vermeire BM, Geurts JW, et al. Epidurography in premature infants. Anaesthesia. 1990;45:722–725.
    1. Yahalom B, Athiraman U, Soriano SG, et al. Spinal anesthesia in infant rats: development of a model and assessment of neurologic outcomes. Anesthesiology. 2011;114:1325–1335.
    1. Sanders RD, Xu J, Shu Y, et al. General anesthetics induce apoptotic neurodegeneration in the neonatal rat spinal cord. Anesth Analg. 2008;106:1708–1711.
    1. Lonnqvist PA, Walker SM. Ketamine as an adjunct to caudal block in neonates and infants: is it time to re-evaluate? Br J Anaesth. 2012;109:138–140.
    1. Lonnqvist PA. Adjuncts to caudal block in children–Quo vadis? Br J Anaesth. 2005;95:431–433.
    1. Hamurtekin E, Fitzsimmons BL, Shubayev VI, et al. Evaluation of spinal toxicity and long-term spinal reflex function after intrathecal levobupivacaine in the neonatal rat. Anesthesiology. 2013;119:142–155.
    1. Walker SM, Grafe M, Yaksh TL. Intrathecal clonidine in the neonatal rat: dose-dependent analgesia and evaluation of spinal apoptosis and toxicity. Anesth Analg. 2012;115:450–460.
    1. Walker SM, Westin BD, Deumens R, et al. Effects of intrathecal ketamine in the neonatal rat: evaluation of apoptosis and long-term functional outcome. Anesthesiology. 2010;113:147–159.
    1. Yaksh TL, Tozier N, Horais KA, et al. Toxicology profile of N-methyl-D-aspartate antagonists delivered by intrathecal infusion in the canine model. Anesthesiology. 2008;108:938–949.
    1. Eich C, Strauss J. Prompt and powerful effect of a practice guideline on caudal additives. Pediatr Anesth. 2009;19:271–272.
    1. Morton NS. The pain-free ward: myth or reality. Pediatr Anesth. 2012;22:527–529.
    1. Zhu LM, Stinson J, Palozzi L, et al. Improvements in pain outcomes in a Canadian pediatric teaching hospital following implementation of a multifaceted knowledge translation initiative. Pain Res Manag. 2012;17:173–179.

Source: PubMed

3
Subscribe