The influence of pain, agitation, and their management on the immature brain

Christopher McPherson, Steven P Miller, Mohamed El-Dib, An N Massaro, Terrie E Inder, Christopher McPherson, Steven P Miller, Mohamed El-Dib, An N Massaro, Terrie E Inder

Abstract

Preterm infants are exposed to frequent painful procedures and agitating stimuli over the many weeks of their hospitalization in the neonatal intensive care unit (NICU). The adverse neurobiological impact of pain and stress in the preterm infant has been well documented, including neuroimaging and neurobehavioral outcomes. Although many tools have been validated to assess acute pain, few methods are available to assess chronic pain or agitation (a clinical manifestation of neonatal stress). Both nonpharmacologic and pharmacologic approaches are used to reduce the negative impact of pain and agitation in the preterm infant, with concerns emerging over the adverse effects of analgesia and sedatives. Considering benefits and risks of available treatments, units must develop a stepwise algorithm to prevent, assess, and treat pain. Nonpharmacologic interventions should be consistently utilized prior to mild to moderately painful procedures. Sucrose may be utilized judiciously as an adjunctive therapy for minor painful procedures. Rapidly acting opioids (fentanyl or remifentanil) form the backbone of analgesia for moderately painful procedures. Chronic sedation during invasive mechanical ventilation represents an ongoing challenge; appropriate containment and an optimal environment should be standard; when indicated, low-dose morphine infusion may be utilized cautiously and dexmedetomidine infusion may be considered as an emerging adjunct.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Growth of the thalamus associated with neonatal pain: The left image illustrates a deformation-based morphometry t-statistic map reflecting reductions in thalamus volume associated with early skin-breaking procedures overlaid on a magnetic resonance image developed as a T1-weighted image template from a median postmenstrual age of 32 weeks. The right scatterplot illustrates the association between volume loss in the lateral thalamus and an increasing exposure to skin-breaking procedures between birth and magnetic resonance imaging. Reproduced with permission from Duerden et al..

References

    1. Synnes AR, et al. School entry age outcomes for infants with birth weight </= 800 grams. J. Pediatr. 2010;157:989–994.
    1. Schmidt B, et al. Prediction of late death or disability at age 5 years using a count of 3 neonatal morbidities in very low birth weight infants. J. Pediatr. 2015;167:982–986.
    1. Synnes A, et al. Determinants of developmental outcomes in a very preterm Canadian cohort. Arch. Dis. Child Fetal Neonatal Ed. 2017;102:F235–F234.
    1. Guo T, et al. Quantitative assessment of white matter injury in preterm neonates: association with outcomes. Neurology. 2017;88:614–622.
    1. Back SA, Miller SP. Brain injury in premature neonates: a primary cerebral dysmaturation disorder? Ann. Neurol. 2014;75:469–486.
    1. Anand KJ. Clinical importance of pain and stress in preterm neonates. Biol. Neonate. 1998;73:1–9.
    1. Anand KJ, Craig KD. New perspectives on the definition of pain. Pain. 1996;67:3–6.
    1. Anand KJ, Hickey PR. Pain and its effects in the human neonate and fetus. N. Engl. J. Med. 1987;317:1321–1329.
    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. Fitzgerald M, Shaw A, MacIntosh N. Postnatal development of the cutaneous flexor reflex: comparative study of preterm infants and newborn rat pups. Dev. Med. Child Neurol. 1988;30:520–526.
    1. Anand KJ. Neonatal stress responses to anesthesia and surgery. Clin. Perinatol. 1990;17:207–214.
    1. Fitzgerald M, Millard C, McIntosh N. Cutaneous hypersensitivity following peripheral tissue damage in newborn infants and its reversal with topical anaesthesia. Pain. 1989;39:31–36.
    1. Committee on Fetus and NewbornSection on Anesthesiology and Pain Medicine. Prevention and management of procedural pain in the neonate: an update. Pediatrics. 2016;137:e20154271.
    1. Disma N, et al. Anesthesia and the developing brain: a way forward for laboratory and clinical research. Paediatr. Anaesth. 2018;28:758–763.
    1. Durrmeyer X, et al. Premedication for neonatal endotracheal intubation: results from the epidemiology of procedural pain in neonates study. Pediatr. Crit. Care Med. 2013;14:e169–e175.
    1. Hatfield LA, Murphy N, Karp K, Polomano RC. A systematic review of behavioral and environmental interventions for procedural pain management in preterm infants. J. Pediatr. Nurs. 2019;44:22–30.
    1. Ranger M, et al. Adverse behavioral changes in adult mice following neonatal repeated exposure to pain and sucrose. Front. Psychol. 2018;9:2394.
    1. Zimmerman KO, et al. Sedation, analgesia, and paralysis during mechanical ventilation of premature infants. J. Pediatr. 2017;180:99–104.
    1. Miller SP, Ferriero DM. From selective vulnerability to connectivity: insights from newborn brain imaging. Trends Neurosci. 2009;32:496–505.
    1. Woodward LJ, Anderson PJ, Austin NC, Howard K, Inder TE. Neonatal MRI to predict neurodevelopmental outcomes in preterm infants. N. Engl. J. Med. 2006;355:685–694.
    1. Buser JR, et al. Arrested preoligodendrocyte maturation contributes to myelination failure in premature infants. Ann. Neurol. 2012;71:93–109.
    1. Zwicker JG, et al. Smaller cerebellar growth and poorer neurodevelopmental outcomes in very preterm infants exposed to neonatal morphine. J. Pediatr. 2016;172:81–87.
    1. Schneider J, et al. Procedural pain and oral glucose in preterm neonates: brain development and sex-specific effects. Pain. 2018;159:515–525.
    1. Duerden EG, et al. Midazolam dose correlates with abnormal hippocampal growth and neurodevelopmental outcome in preterm infants. Ann. Neurol. 2016;79:548–559.
    1. Duerden EG, et al. Early procedural pain is associated with regionally-specific alterations in thalamic development in preterm neonates. J. Neurosci. 2018;38:878–886.
    1. Thompson DK, et al. Neonate hippocampal volumes: prematurity, perinatal predictors, and 2-year outcome. Ann. Neurol. 2008;63:642–651.
    1. Toulmin H, et al. Specialization and integration of functional thalamocortical connectivity in the human infant. Proc. Natl Acad. Sci. USA. 2015;112:6485–6490.
    1. Brummelte S, et al. Procedural pain and brain development in premature newborns. Ann. Neurol. 2012;71:385–396.
    1. Zwicker JG, et al. Score for neonatal acute physiology-II and neonatal pain predict corticospinal tract development in premature newborns. Pediatr. Neurol. 2013;48:123–129.
    1. Smith GC, et al. Neonatal intensive care unit stress is associated with brain development in preterm infants. Ann. Neurol. 2011;70:541–549.
    1. Ranger M, et al. Neonatal pain-related stress predicts cortical thickness at age 7 years in children born very preterm. PLoS ONE. 2013;8:e76702.
    1. Chau CMY, et al. Hippocampus, amygdala, and thalamus volumes in very preterm children at 8 years: neonatal pain and genetic variation. Front. Behav. Neurosci. 2019;13:51.
    1. Vinall J, et al. Invasive procedures in preterm children: brain and cognitive development at school age. Pediatrics. 2014;133:412–421.
    1. Doesburg, S. M. et al. Neonatal pain-related stress, functional cortical activity and visual–perceptual abilities in school-age children born at extremely low gestational age. Pain154, 1946–1952 (2013).
    1. Vinall J, et al. Slower postnatal growth is associated with delayed cerebral cortical maturation in preterm newborns. Sci. Transl. Med. 2013;5:168ra8.
    1. Grunau, R. E. & Craig, K. D. Neonatal Facial Coding System Revised: Training Manual (University of British Columbia, Vancouver, 2010).
    1. Gibbins S, et al. Validation of the premature infant pain profile-revised (PIPP-R) Early Hum. Dev. 2014;90:189–193.
    1. Hummel P, Puchalski M, Creech SD, Weiss MG. Clinical reliability and validity of the N-PASS: neonatal pain, agitation and sedation scale with prolonged pain. J. Perinatol. 2008;28:55–60.
    1. Lawrence J, et al. The development of a tool to assess neonatal pain. Neonatal Netw. 1993;12:59–66.
    1. Cignacco E, Mueller R, Hamers JP, Gessler P. Pain assessment in the neonate using the Bernese Pain Scale for Neonates. Early Hum. Dev. 2004;78:125–131.
    1. Kappesser J, et al. Pain-specific reactions or indicators of a general stress response?: Investigating the discriminant validity of 5 well-established neonatal pain assessment tools. Clin. J. Pain. 2019;35:101–110.
    1. Benoit B, Martin-Misener R, Newman A, Latimer M, Campbell-Yeo M. Neurophysiological assessment of acute pain in infants: a scoping review of research methods. Acta Paediatr. 2017;106:1053–1066.
    1. Fabrizi L, 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, 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. Bartocci M, Bergqvist LL, Lagercrantz H, Anand KJ. Pain activates cortical areas in the preterm newborn brain. Pain. 2006;122:109–117.
    1. Munsters J, Wallstrom L, Agren J, Norsted T, Sindelar R. Skin conductance measurements as pain assessment in newborn infants born at 22-27 weeks gestational age at different postnatal age. Early Hum. Dev. 2012;88:21–26.
    1. Jansen J, Beijers R, Riksen-Walraven M, de Weerth C. Cortisol reactivity in young infants. Psychoneuroendocrinology. 2010;35:329–338.
    1. Roue JM, et al. Multi-modal pain assessment: are near-infrared spectroscopy, skin conductance, salivary cortisol, physiologic parameters, and Neonatal Facial Coding System interrelated during venepuncture in healthy, term neonates. J. Pain Res. 2018;11:2257–2267.
    1. Pena-Bautista, C. et al. Non-invasive monitoring of stress biomarkers in the newborn period. Semin. Fetal Neonatal Med. 24, 101002 (2019).
    1. Pillai Riddell, R. R. et al. Non-pharmacological management of infant and young child procedural pain. Cochrane Database Syst. Rev. CD006275 (2015). 10.1002/14651858.CD006275.pub2.
    1. Carbajal R, et al. Epidemiology and treatment of painful procedures in neonates in intensive care units. JAMA. 2008;300:60–70.
    1. Bellieni CV, Johnston CC. Analgesia, nil or placebo to babies, in trials that test new analgesic treatments for procedural pain. Acta Paediatr. 2016;105:129–136.
    1. Stevens B, Yamada J, Ohlsson A, Haliburton S, Shorkey A. Sucrose for analgesia in newborn infants undergoing painful procedures. Cochrane Database Syst. Rev. 2016;7:CD001069.
    1. de Freitas RL, Kubler JM, Elias-Filho DH, Coimbra NC. Antinociception induced by acute oral administration of sweet substance in young and adult rodents: the role of endogenous opioid peptides chemical mediators and mu(1)-opioid receptors. Pharmacol. Biochem. Behav. 2012;101:265–270.
    1. Blass E, Fitzgerald E, Kehoe P. Interactions between sucrose, pain and isolation distress. Pharmacol. Biochem. Behav. 1987;26:483–489.
    1. Taddio A, Shah V, Shah P, Katz J. Beta-endorphin concentration after administration of sucrose in preterm infants. Arch. Pediatr. Adolesc. Med. 2003;157:1071–1074.
    1. Gradin M, Schollin J. The role of endogenous opioids in mediating pain reduction by orally administered glucose among newborns. Pediatrics. 2005;115:1004–1007.
    1. Hajnal A, Smith GP, Norgren R. Oral sucrose stimulation increases accumbens dopamine in the rat. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2004;286:R31–R37.
    1. Jones PG, Dunlop J. Targeting the cholinergic system as a therapeutic strategy for the treatment of pain. Neuropharmacology. 2007;53:197–206.
    1. Reboucas EC, et al. Effect of the blockade of mu1-opioid and 5HT2A-serotonergic/alpha1-noradrenergic receptors on sweet-substance-induced analgesia. Psychopharmacology. 2005;179:349–355.
    1. Stevens B, et al. The minimally effective dose of sucrose for procedural pain relief in neonates: a randomized controlled trial. BMC Pediatr. 2018;18:85.
    1. Bauer K, Ketteler J, Hellwig M, Laurenz M, Versmold H. Oral glucose before venepuncture relieves neonates of pain, but stress is still evidenced by increase in oxygen consumption, energy expenditure, and heart rate. Pediatr. Res. 2004;55:695–700.
    1. Slater R, et al. Oral sucrose as an analgesic drug for procedural pain in newborn infants: a randomised controlled trial. Lancet. 2010;376:1225–1232.
    1. Taddio A, Shah V, Atenafu E, Katz J. Influence of repeated painful procedures and sucrose analgesia on the development of hyperalgesia in newborn infants. Pain. 2009;144:43–48.
    1. Durrmeyer X, Vutskits L, Anand KJ, Rimensberger PC. Use of analgesic and sedative drugs in the NICU: integrating clinical trials and laboratory data. Pediatr. Res. 2010;67:117–127.
    1. Tremblay S, et al. Repeated exposure to sucrose for procedural pain in mouse pups leads to long-term widespread brain alterations. Pain. 2017;158:1586–1598.
    1. Johnston CC, et al. Routine sucrose analgesia during the first week of life in neonates younger than 31 weeks’ postconceptional age. Pediatrics. 2002;110:523–528.
    1. Ranger M, Johnston CC, Anand KJ. Current controversies regarding pain assessment in neonates. Semin. Perinatol. 2007;31:283–288.
    1. McPherson C, Grunau RE. Neonatal pain control and neurologic effects of anesthetics and sedatives in preterm infants. Clin. Perinatol. 2014;41:209–227.
    1. Berde CB, et al. Anesthesia and analgesia during and after surgery in neonates. Clin. Ther. 2005;27:900–921.
    1. Kumar P, Denson SE, Mancuso TJ, Committee on Fetus and Newborn Section on Anesthesiology and Pain Medicine. Premedication for nonemergency endotracheal intubation in the neonate. Pediatrics. 2010;125:608–615.
    1. Ancora G, et al. Evidence-based clinical guidelines on analgesia and sedation in newborn infants undergoing assisted ventilation and endotracheal intubation. Acta Paediatr. 2019;108:208–217.
    1. Anand KJ, et al. Analgesia and sedation in preterm neonates who require ventilatory support: results from the NOPAIN trial. Neonatal Outcome and Prolonged Analgesia in Neonates. Arch. Pediatr. Adolesc. Med. 1999;153:331–338.
    1. van Straaten HL, Rademaker CM, de Vries LS. Comparison of the effect of midazolam or vecuronium on blood pressure and cerebral blood flow velocity in the premature newborn. Dev. Pharmacol. Ther. 1992;19:191–195.
    1. Anand KJ, et al. Effects of morphine analgesia in ventilated preterm neonates: primary outcomes from the NEOPAIN randomised trial. Lancet. 2004;363:1673–1682.
    1. Bhandari V, Bergqvist LL, Kronsberg SS, Barton BA, Anand KJ. Morphine administration and short-term pulmonary outcomes among ventilated preterm infants. Pediatrics. 2005;116:352–359.
    1. Menon G, et al. Morphine analgesia and gastrointestinal morbidity in preterm infants: secondary results from the NEOPAIN trial. Arch. Dis. Child Fetal Neonatal Ed. 2008;93:F362–F367.
    1. Rao R, et al. Neurobehavior of preterm infants at 36 weeks postconception as a function of morphine analgesia. Am. J. Perinatol. 2007;24:511–517.
    1. Ferguson SA, Ward WL, Paule MG, Hall RW, Anand KJ. A pilot study of preemptive morphine analgesia in preterm neonates: effects on head circumference, social behavior, and response latencies in early childhood. Neurotoxicol. Teratol. 2012;34:47–55.
    1. Simons SH, et al. Routine morphine infusion in preterm newborns who received ventilatory support: a randomized controlled trial. J. Am. Med. Assoc. 2003;290:2419–2427.
    1. de Graaf J, 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, et al. Does neonatal morphine use affect neuropsychological outcomes at 8 to 9 years of age? Pain. 2013;154:449–458.
    1. Steinhorn R, et al. Neonatal morphine exposure in very preterm infants—cerebral development and outcomes. J. Pediatr. 2015;166:1200–1207.
    1. Chau CMY, et al. Morphine biotransformation genes and neonatal clinical factors predicted behaviour problems in very preterm children at 18 months. EBioMedicine. 2019;40:655–662.
    1. Saarenmaa E, Neuvonen PJ, Fellman V. Gestational age and birth weight effects on plasma clearance of fentanyl in newborn infants. J. Pediatr. 2000;136:767–770.
    1. Ancora G, et al. Efficacy and safety of continuous infusion of fentanyl for pain control in preterm newborns on mechanical ventilation. J. Pediatr. 2013;163:645–651.
    1. Ancora G, et al. Follow-up at the corrected age of 24 months of preterm newborns receiving continuous infusion of fentanyl for pain control during mechanical ventilation. Pain. 2017;158:840–845.
    1. McPherson C, et al. Brain injury and development in preterm infants exposed to fentanyl. Ann. Pharmacother. 2015;49:1291–1297.
    1. Völler, S. et al. Rapidly maturing fentanyl clearance in preterm neonates. Arch. Dis. Child-Fetal Neonatal Ed. 10.1136/archdischild-2018-315920 (2019).
    1. O'Mara K, et al. Dexmedetomidine versus standard therapy with fentanyl for sedation in mechanically ventilated premature neonates. J. Pediatr. Pharm. Ther. 2012;17:252–262.
    1. Chrysostomou C, et al. A phase II/III, multicenter, safety, efficacy, and pharmacokinetic study of dexmedetomidine in preterm and term neonates. J. Pediatr. 2014;164:276–282.
    1. Laudenbach V, et al. Effects of alpha(2)-adrenoceptor agonists on perinatal excitotoxic brain injury: comparison of clonidine and dexmedetomidine. Anesthesiology. 2002;96:134–141.

Source: PubMed

3
S'abonner