Skip to main content

Advertisement

Log in

Adequate Pain Management and Sedation in the Neonate: a Fine Balance

  • Neonatology (T Thorkelsson, Section Editor)
  • Published:
Current Treatment Options in Pediatrics Aims and scope Submit manuscript

Abstract

Purpose of review

With heightened awareness of long-term neurocognitive effects of pain and stress in the neonatal population, increased focus has been placed on pain assessment as well as potential effects of routinely administered analgesics/sedatives in our neonatal intensive care units (NICUs). Our goal was to review recently published literature on current pain assessment practices, novel approaches to management, pharmacogenomics in the neonate, and risks of commonly used medications.

Recent findings

There is an increasing trend in the administration of analgesics and sedatives in NICUs, despite reports of the negative effects these medications may have on neonatal brain development. As in the adult world, studies are finding that pharmacogenomics has the potential to play a significant role in adequate pain control and sedation in the individual neonatal patient. Due to increased reports of adverse effects of commonly used medications such as morphine and midazolam, new pharmacologic agents are being evaluated in the neonatal population as possibly safer alternatives.

Summary

There have been significant advances in our knowledge of pain and sedation in the neonate, ranging from a better understanding of long-term neurocognitive effects of pain in neonatal brain development to the role of pharmacogenomics in the NICU setting. These developments have opened the door for continued learning and investigation into the ideal management of neonatal pain and sedation.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance

  1. Borenstein-Levin L, Synnes A, Grunau RE, Miller SP, Yoon EW, Shah PS, et al. Narcotics and sedative use in preterm neonates. J Pediatr. 2017;180:92–98 e91. https://doi.org/10.1016/j.jpeds.2016.08.031.

    Article  PubMed  Google Scholar 

  2. Kinney HC. The near-term (late preterm) human brain and risk for periventricular leukomalacia: a review. Semin Perinatol. 2006;30(2):81–8. https://doi.org/10.1053/j.semperi.2006.02.006.

    Article  PubMed  Google Scholar 

  3. Guo T, Winterburn JL, Pipitone J, Duerden EG, Park MT, Chau V, et al. Automatic segmentation of the hippocampus for preterm neonates from early-in-life to term-equivalent age. Neuroimage Clin. 2015;9:176–93. https://doi.org/10.1016/j.nicl.2015.07.019.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Hall RW, Anand KJS. Physiology of pain and stress in the newborn. NeoReviews. 2005;6(2):e61–8. https://doi.org/10.1542/neo.6-2-e61.

    Article  Google Scholar 

  5. Carbajal R, Eriksson M, Courtois E, Boyle E, Avila-Alvarez A, Andersen RD, et al. Sedation and analgesia practices in neonatal intensive care units (EUROPAIN): results from a prospective cohort study. Lancet Respir Med. 2015;3(10):796–812. https://doi.org/10.1016/S2213-2600(15)00331-8.

    Article  PubMed  Google Scholar 

  6. Carter BS, Brunkhorst J. Neonatal pain management. Semin Perinatol. 2017;41(2):111–6. https://doi.org/10.1053/j.semperi.2016.11.001.

    Article  PubMed  Google Scholar 

  7. Lewis T, Erfe BL, Ezell T, Gauda E. Pharmacoepidemiology of opiate use in the neonatal ICU: increasing cumulative doses and iatrogenic opiate withdrawal. J Opioid Manag. 2015;11(4):305–12. https://doi.org/10.5055/jom.2015.0279.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Zimmerman KO, Smith PB, Benjamin DK, Laughon M, Clark R, Traube C, et al. Sedation, analgesia, and paralysis during mechanical ventilation of premature infants. J Pediatr. 2017;180:99–104 e101. https://doi.org/10.1016/j.jpeds.2016.07.001.

    Article  PubMed  Google Scholar 

  9. 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.

    PubMed  Google Scholar 

  10. Liu Y, Huang X, Luo B, Peng W. Effects of combined oral sucrose and nonnutritive sucking (NNS) on procedural pain of NICU newborns, 2001 to 2016: a PRISMA-compliant systematic review and meta-analysis. Medicine (Baltimore). 2017;96(6):e6108. https://doi.org/10.1097/MD.0000000000006108.

    Article  Google Scholar 

  11. Johnston C, Campbell-Yeo M, Disher T, Benoit B, Fernandes A, Streiner D, et al. Skin-to-skin care for procedural pain in neonates. Cochrane Database Syst Rev. 2017;2:CD008435.

    PubMed  Google Scholar 

  12. Shah PS, Herbozo C, Aliwalas LL, Shah VS. Breastfeeding or breast milk for procedural pain in neonates. Cochrane Database Syst Rev. 2012;12:CD004950.

    PubMed  Google Scholar 

  13. Badr LK, Abdallah B, Kahale L. A meta-analysis of preterm infant massage: an ancient practice with contemporary applications. MCN Am J Matern Child Nurs. 2015;40(6):344–58. https://doi.org/10.1097/NMC.0000000000000177.

    Article  PubMed  Google Scholar 

  14. Baudesson de Chanville A, Brevaut-Malaty V, Garbi A, Tosello B, Baumstarck K, Gire C. Analgesic effect of maternal human milk odor on premature neonates: a randomized controlled trial. J Hum Lact. 2017;33(2):300–8. https://doi.org/10.1177/0890334417693225.

    Article  PubMed  Google Scholar 

  15. Committee On F, Newborn, Section On A, Pain M. Prevention and management of procedural pain in the neonate: an update. Pediatrics. 2016;137(2):e20154271. https://doi.org/10.1542/peds.2015-4271.

    Article  Google Scholar 

  16. Hummel P. Psychometric evaluation of the neonatal pain, agitation, and sedation (N-PASS) scale in infants and children up to age 36 months. Pediatr Nurs. 2017;43(4):175–84.

    Google Scholar 

  17. • Anand KJS, Eriksson M, Boyle EM, Avila-Alvarez A, Andersen RD, Sarafidis K, et al. Assessment of continuous pain in newborns admitted to NICUs in 18 European countries. Acta Paediatr. 2017;106(8):1248–59. https://doi.org/10.1111/apa.13810This is a multicenter prospective observational study that spans over 18 European countries that evaluated the current state of pain management within 243 NICUs. They tracked the use of mechanical ventilation, sedation, analgesia, neuromuscular blockade, and frequency of pain assessments in 6648 neonates over an 8-month period. They found that only 31.8% of the total neonates received assessments of continuous pain at least one time during their admission. Only 10% of these neonates received daily assessments of continuous pain. The assessments of continuous pain varied between countries, occuring most commonly in France (100%). The NICUs with increased surgical admissions, pain management guidelines, and nursing champions performed assessments of continous pain more frequently.

    Article  PubMed  Google Scholar 

  18. Rana D, Bellflower B, Sahni J, Kaplan AJ, Owens NT, Arrindell EL Jr, et al. Reduced narcotic and sedative utilization in a NICU after implementation of pain management guidelines. J Perinatol. 2017;37(9):1038–42. https://doi.org/10.1038/jp.2017.88.

    Article  CAS  PubMed  Google Scholar 

  19. Deindl P, Giordano V, Fuiko R, Waldhoer T, Unterasinger L, Berger A, et al. The implementation of systematic pain and sedation management has no impact on outcome in extremely preterm infants. Acta Paediatr. 2016;105(7):798–805. https://doi.org/10.1111/apa.13334.

    Article  PubMed  Google Scholar 

  20. Moultrie F, Slater R, Hartley C. Improving the treatment of infant pain. Curr Opin Support Palliat Care. 2017;11(2):112–7. https://doi.org/10.1097/SPC.0000000000000270.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Leeder JS, Kearns GL. Interpreting pharmacogenetic data in the developing neonate: the challenge of hitting a moving target. Clin Pharmacol Ther. 2012;92(4):434–6. https://doi.org/10.1038/clpt.2012.130.

    CAS  PubMed  Google Scholar 

  22. Smits A, van den Anker J, Allegaert K. Clinical pharmacology of analgosedatives in neonates: ways to improve their safe and effective use. J Pharm Pharmacol. 2017;69(4):350–60. https://doi.org/10.1111/jphp.12599.

    Article  CAS  PubMed  Google Scholar 

  23. Elens L, Norman E, Matic M, Rane A, Fellman V, van Schaik RH. Genetic predisposition to poor opioid response in preterm infants: impact of KCNJ6 and COMT polymorphisms on pain relief after endotracheal intubation. Ther Drug Monit. 2016;38(4):525–33. https://doi.org/10.1097/FTD.0000000000000301.

    Article  CAS  PubMed  Google Scholar 

  24. Allegaert K, van den Anker JN. Neonatal pain management: still in search for the Holy Grail. Int J Clin Pharmacol Ther. 2016;54(7):514–23. https://doi.org/10.5414/CP202561.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. • Zwicker JG, Miller SP, Grunau RE, Chau V, Brant R, Studholme C, et al. Smaller cerebellar growth and poorer neurodevelopmental outcomes in very preterm infants exposed to neonatal morphine. J Pediatr. 2016;172:81–87 e82. https://doi.org/10.1016/j.jpeds.2015.12.024This study utilized magnetric resonance imaging (MRI) in order to evaluate early brain development in a prospective cohort of 188 preterm infants (24–32 weeks of gestational age) admitted to the neonatal intensive care unit. All participants underwent brain MRI scans without sedation within the first weeks of life and at term-equivalent age. At 18-month corrected age, 136 of the original participants received standardized motor and cognitive assessments. Increased morphine exposure was associated with a decrease in cerebellar volume, after adjustments were made for total brain volume and various clinical confounders. They also found that morphine exposure was significantly associated with poor motor and cognitive outcomes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kocek M, Wilcox R, Crank C, Patra K. Evaluation of the relationship between opioid exposure in extremely low birth weight infants in the neonatal intensive care unit and neurodevelopmental outcome at 2 years. Early Hum Dev. 2016;92:29–32. https://doi.org/10.1016/j.earlhumdev.2015.11.001.

    Article  CAS  PubMed  Google Scholar 

  27. Ng E, Taddio A, Ohlsson A. Intravenous midazolam infusion for sedation of infants in the neonatal intensive care unit. Cochrane Database Syst Rev. 2017;1:CD002052.

    PubMed  Google Scholar 

  28. Duerden EG, Guo T, Dodbiba L, Chakravarty MM, Chau V, Poskitt KJ, et al. Midazolam dose correlates with abnormal hippocampal growth and neurodevelopmental outcome in preterm infants. Ann Neurol. 2016;79(4):548–59. https://doi.org/10.1002/ana.24601.

    Article  CAS  PubMed  Google Scholar 

  29. Harma A, Aikio O, Hallman M, Saarela T. Intravenous paracetamol decreases requirements of morphine in very preterm infants. J Pediatr. 2016;168:36–40. https://doi.org/10.1016/j.jpeds.2015.08.003.

    Article  CAS  PubMed  Google Scholar 

  30. 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.

    PubMed  Google Scholar 

  31. Sifringer M, von Haefen C, Krain M, Paeschke N, Bendix I, Buhrer C, et al. Neuroprotective effect of dexmedetomidine on hyperoxia-induced toxicity in the neonatal rat brain. Oxidative Med Cell Longev. 2015;2015:530371.

    Article  Google Scholar 

  32. Sacha GL, Foreman MG, Kyllonen K, Rodriguez RJ. The use of gabapentin for pain and agitation in neonates and infants in a neonatal ICU. J Pediatr Pharmacol Therapeut. 2017;22(3):207–11. https://doi.org/10.5863/1551-6776-22.3.207.

    Article  Google Scholar 

  33. Edwards L, DeMeo S, Hornik CD, Cotton CM, Smith PB, Pizoli C, et al. Gabapentin us in the neonatal intensive care unit. J Pediatr. 2016;169:310–2. https://doi.org/10.1016/j.jpeds.2015.10.013.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Brian S. Carter MD.

Ethics declarations

Conflict of Interest

Lauren Cummings declares that she has no conflict of interest. Tamorah Lewis declares that she has no conflict of interest. Brian S. Carter declares that he has no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

This article is part of the Topical Collection on Neonatology

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cummings, L., Lewis, T. & Carter, B.S. Adequate Pain Management and Sedation in the Neonate: a Fine Balance. Curr Treat Options Peds 4, 108–118 (2018). https://doi.org/10.1007/s40746-018-0109-0

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40746-018-0109-0

Keywords

Navigation