Skip to main content

Advertisement

Log in

Anesthetic Exposure During Early Childhood and Neurodevelopmental Outcomes: Our Current Understanding

  • Anesthesia, Pain Management and Long-term Outcomes (VNR Gottumukkala and ER Mariano, Section Editors)
  • Published:
Current Anesthesiology Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

The question of anesthetic neurotoxicity emerged two decades ago, but controversy about whether anesthetics cause neurodevelopmental problems in children remains. Interpretation of the published literature is complicated by a paucity of randomized controlled trials and heterogeneity of the published studies. This review summarizes our current understanding and discusses potential sources of study bias and methods to better understand and address issues contributing to bias.

Recent Findings

Recent clinical studies of anesthetic neurotoxicity and meta-analyses of the published studies have reported that children exposed to anesthesia have worse neurodevelopmental outcome scores than unexposed children, particularly in domains of executive function and behavior.

Summary

While anesthetic-exposed children report worse neurodevelopmental outcomes, whether these differences are caused by the anesthesia, or other factors such baseline disease, surgical inflammation, or physiologic disturbances, remains a subject of intense debate. To answer this question, further well-designed studies will be required.

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 •• Of major importance

  1. Rabbitts JA, Groenewald CB, Moriarty JP, Flick R. Epidemiology of ambulatory anesthesia for children in the United States: 2006 and 1996. Anesth Analg. 2010;111(4):1011–5.

    Article  PubMed  Google Scholar 

  2. Tzong KY, Han S, Roh A, Ing C. Epidemiology of pediatric surgical admissions in US children: data From the HCUP Kids Inpatient Database. J Neurosurg Anesthesiol. 2012;24(4):391–5.

    Article  PubMed  Google Scholar 

  3. FDA Drug Safety Communication: FDA review results in new warnings about using general anesthetics and sedation drugs in young children and pregnant women [12–14–2016]. U.S. Food and Drug Administration. http://www.fda.gov/Drugs/DrugSafety/ucm532356.htm?source=govdelivery&utm_medium=email&utm_source=govdelivery. Accessed January 3, 2017.

  4. Jevtovic-Todorovic V, Hartman RE, Izumi Y, et al. Early exposure to common anesthetic agents causes widespread neurodegeneration in the developing rat brain and persistent learning deficits. J Neurosci. 2003;23(3):876–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Vutskits L, Xie Z. Lasting impact of general anaesthesia on the brain: mechanisms and relevance. Nat Rev Neurosci. 2016;17(11):705–17.

    Article  CAS  PubMed  Google Scholar 

  6. Doberschuetz N, Dewitz R, Rolle U, Schlösser R, Allendorf A. Follow-up of children with gastrointestinal malformations and postnatal surgery and anesthesia: evaluation at two years of age. Neonatol. 2016;110(1):8–13.

    Article  CAS  Google Scholar 

  7. Davidson AJ, Disma N, de Graaff JC, et al. Neurodevelopmental outcome at 2 years of age after general anaesthesia and awake-regional anaesthesia in infancy (GAS): an international multicentre, randomised controlled trial. Lancet. 2016;387(10015):239–50.

    Article  PubMed  Google Scholar 

  8. McCann ME, de Graaff JC, Dorris L, et al. Neurodevelopmental outcome at 5 years of age after general anaesthesia or awake-regional anaesthesia in infancy (GAS): an international, multicentre, randomised, controlled equivalence trial. Lancet. 2019;393(10172):664–77.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Ing CH, DiMaggio CJ, Whitehouse AJ, et al. Neurodevelopmental outcomes after initial childhood anesthetic exposure between ages 3 and 10 years. J Neurosurg Anesthesiol. 2014;26(4):377–86.

    Article  PubMed  Google Scholar 

  10. Zhang Q, Peng Y, Wang Y. Long-duration general anesthesia influences the intelligence of school age children. BMC Anesthesiol. 2017;17(1):170.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Graham MR, Brownell M, Chateau DG, Dragan RD, Burchill C, Fransoo RR. Neurodevelopmental assessment in kindergarten in children exposed to general anesthesia before the age of 4 years: a retrospective matched cohort study. Anesthesiol. 2016;125(4):667–77.

    Article  CAS  Google Scholar 

  12. Ing C, Sun M, Olfson M, et al. Age at exposure to surgery and anesthesia in children and association with mental disorder diagnosis. Anesth Analg. 2017;125(6):1988–98.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Ing C, Landau R, DeStephano D, et al. Prenatal exposure to general anesthesia and childhood behavioral deficit. Anesth Analg. 2021;133(3):595–605. This study analyzed data from the Raine Study, an observational cohort study of children born in Perth, Western Australia with 2 generations of participations. They authors reported increased externalizing behavioral problems in childhood due to prenatal exposure to general anesthetics.

  14. Bleeser T, Devroe S, Lucas N, et al. Neurodevelopmental outcomes after prenatal exposure to anaesthesia for maternal surgery: a propensity-score weighted bidirectional cohort study. Anaesthesia. 2022. This bidirectional cohort study is the largest to date and was conducted using data between 2001 and 2018. The authors found no evidence that there is an association between prenatal exposure to anesthesia and neurodevelopmental outcomes in children.

  15. Wilder RT, Flick RP, Sprung J, et al. Early exposure to anesthesia and learning disabilities in a population-based birth cohort. Anesthesiol. 2009;110(4):796–804.

    Article  Google Scholar 

  16. Flick RP, Katusic SK, Colligan RC, et al. Cognitive and behavioral outcomes after early exposure to anesthesia and surgery. Pediatrics. 2011;128(5):e1053-1061.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Hu D, Flick RP, Zaccariello MJ, et al. Association between exposure of young children to procedures requiring general anesthesia and learning and behavioral outcomes in a population-based birth cohort. Anesthesiol. 2017;127(2):227–40.

    Article  Google Scholar 

  18. Sprung J, Flick RP, Katusic SK, et al. Attention-deficit/hyperactivity disorder after early exposure to procedures requiring general anesthesia. Mayo Clin Proc. 2012;87(2):120–9.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Shi Y, Hu D, Rodgers EL, et al. Epidemiology of general anesthesia prior to age 3 in a population-based birth cohort. Paediatr Anaesth. 2018;28(6):513–9.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Moser JJ, Archer DP, Walker AM, et al. Association of sedation and anesthesia on cognitive outcomes in very premature infants: a retrospective observational study. Can J Anesth. 2023;70(1):56–68.

    Article  CAS  PubMed  Google Scholar 

  21. Puia-Dumitrescu M, Comstock BA, Li S, et al. Assessment of 2-year neurodevelopmental outcomes in extremely preterm infants receiving opioids and benzodiazepines. JAMA Netw Open. 2021;4(7): e2115998.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Simpao AF, Randazzo IR, Chittams JL, et al. Anesthesia and sedation exposure and neurodevelopmental outcomes in infants undergoing congenital cardiac surgery: a retrospective cohort study. Anesthesiology. 2023;139(4):393–404. https://doi.org/10.1097/ALN.0000000000004684.

    Article  PubMed  Google Scholar 

  23. Andropoulos DB, Ahmad HB, Haq T, et al. The association between brain injury, perioperative anesthetic exposure, and 12-month neurodevelopmental outcomes after neonatal cardiac surgery: a retrospective cohort study. Paediatr Anaesth. 2014;24(3):266–74.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Warner DO, Zaccariello MJ, Katusic SK, et al. Neuropsychological and behavioral outcomes after exposure of young children to procedures requiring general anesthesia: the Mayo Anesthesia Safety in Kids (MASK) study. Anesthesiology. 2018;129(1):89–105.

    Article  PubMed  Google Scholar 

  25. Sun LS, Li G, Miller TL, et al. Association between a single general anesthesia exposure before age 36 months and neurocognitive outcomes in later childhood. JAMA. 2016;315(21):2312–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ing C, Jackson WM, Zaccariello MJ, et al. Prospectively assessed neurodevelopmental outcomes in studies of anaesthetic neurotoxicity in children: a systematic review and meta-analysis. Br J Anaesth. 2021;126(2):433–444. This meta-analysis showed that exposure to general anesthesia was associated with increases in parental reports of behavioral problems, but no difference in general intelligence.

  27. Reighard C, Junaid S, Jackson WM, et al. Anesthetic exposure during childhood and neurodevelopmental outcomes: a systematic review and meta-analysis. JAMA Netw Open. 2022;5(6):e2217427. This meta-analysis of 31 studies found that anesthetic exposure during childhood and subsequent neurodevelopmental deficits differed based on neurodevelopmental domain.

  28. Pikwer A, Yang B, Granstrom M, Mattsson N, Sadr-Azodi O. General anesthesia in early childhood and possible association with autism: a population-based matched cohort study. Minerva Anestesiol. 2023;89(1–2):22–31. This population-based cohort analyzed data collected between 2001 and 2014 on children between 0 to 5 who were exposed to anesthesia and found an increased risk of autism or autism spectrum disorder.

  29. Laporta ML, Sprung J, Fejedelem CA, et al. Association between exposure of children to general anesthesia and autism spectrum disorder. J Autism Dev Disord. 2022;52(10):4301–4310. This nested and matched-case control study analyzed data from the population-based birth cohort of children born in Olmsted County, MN from 1976 to 2000. The authors found that autism spectrum disorder was not associated with exposure to general anesthesia in children after adjusting for covariates.

  30. Salaun JP, Chagnot A, Cachia A, et al. Consequences of general anesthesia in infancy on behavior and brain structure. Anesth Analg. 2023;136(2):240–250. This study conducted independent preclinical and clinical analyses to test whether exposure to general anesthesia could cause behavioral and structural changes in the developing brain. Both mouse and human models had changes in brain structure; mouse model revealed exacerbations in fear response whereas the human model revealed lower emotional control in subjects exposed to general anesthesia compared to unexposed controls. These findings corroborate the hypothesis that early life exposure to general anesthesia can have lasting changes in behavior and brain structure.

  31. Rozeske RR, Jercog D, Karalis N, et al. Prefrontal-periaqueductal gray-projecting neurons mediate context fear discrimination. Neuron. 2018;97(4):898.

    Article  CAS  PubMed  Google Scholar 

  32. Ho YC, Lin TB, Hsieh MC, et al. Periaqueductal gray glutamatergic transmission governs chronic stress-induced depression. Neuropsychopharmacol. 2018;43(2):302–12.

    Article  CAS  Google Scholar 

  33. Satomoto M, Satoh Y, Terui K, et al. Neonatal exposure to sevoflurane induces abnormal social behaviors and deficits in fear conditioning in mice. Anesthesiol. 2009;110(3):628–37.

    Article  CAS  Google Scholar 

  34. Luby JL, Barch D, Whalen D, Tillman R, Belden A. Association between early life adversity and risk for poor emotional and physical health in adolescence a putative mechanistic neurodevelopmental pathway. Jama Pediatr. 2017;171(12):1168–75.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Zhang FF, Peng W, Sweeney JA, Jia ZY, Gong QY. Brain structure alterations in depression: Psychoradiological evidence. Cns Neurosci Ther. 2018;24(11):994–1003.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Greenland S, Pearl J, Robins JM. Causal diagrams for epidemiologic research. Epidemiol. 1999;10(1):37–48.

    Article  CAS  Google Scholar 

  37. Ma H, Jm R. Causal Inference: What If. Boca Raton: Chapman & Hall/CRC; 2020.

    Google Scholar 

  38. Morgan SL, Winship C. Counterfactuals and Causal Inference: Methods and Principles for Social Research, 2nd Edition. Anal Method Soc Res. 2015:1–499. https://doi.org/10.1017/CBO9781107587991

  39. Shrier I, Platt RW. Reducing bias through directed acyclic graphs. Bmc Med Res Methodol. 2008;8. https://doi.org/10.1186/1471-2288-8-70

  40. Schisterman EF, Cole SR, Platt RW. Overadjustment bias and unnecessary adjustment in epidemiologic studies. Epidemiol. 2009;20(4):488–95.

    Article  Google Scholar 

  41. Westreich D, Greenland S. The table 2 fallacy: presenting and interpreting confounder and modifier coefficients. Am J Epidemiol. 2013;177(4):292–8.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Krishnamoorthy V, Wong DJN, Wilson M, et al. Causal inference in perioperative medicine observational research: part 1, a graphical introduction. Br J Anaesth. 2020;125(3):393–7.

    Article  PubMed  Google Scholar 

  43. Vutskits L, Davidson A. Clinical investigations on anesthesia-induced developmental neurotoxicity: the knowns, the unknowns and future prospects. Best Pract Res Clin Anaesthesiol. 2023;37(1):40–51.

    Article  PubMed  Google Scholar 

  44. Deaton A, Cartwright N. Understanding and misunderstanding randomized controlled trials. Soc Sci Med. 2018;210:2–21.

    Article  PubMed  Google Scholar 

  45. Pearl J, Glymour M, Jewell NP. Causal inference in statistics: a primer. John Wiley & Sons; 2016.

  46. Pitts AJ, Fowler CR. Comparison of open-source software for producing directed acyclic graphs. 2023. https://arxiv.org/abs/2305.12006. Accessed 24 Jul 2023.

  47. Ji Q, Zhang HY, Geng JB. What drives natural gas prices in the United States?–A directed acyclic graph approach. Energ Econ. 2018;69:79–88.

    Article  Google Scholar 

  48. Imbens GW. Potential outcome and directed acyclic graph approaches to causality: relevance for empirical practice in economics. J Econ Lit. 2020;58(4):1129–79.

    Article  Google Scholar 

  49. Yang ZH, Zhao YL. Energy consumption, carbon emissions, and economic growth in India: evidence from directed acyclic graphs. Econ Model. 2014;38:533–40.

    Article  Google Scholar 

  50. Huynh QL, Thi TLL. applying the directed acyclic graph to examine the factors related to the adoption of e-learning. Journal of Knowledge Management, Economics and Information Technology. 2014;4(1).

  51. Iwata H, Wakabayashi T, Kato R. The dawn of directed acyclic graphs in primary care research and education. J Gen Fam Med. 2023;24(4):274–5.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Costa JD, Bernardini F, Artigas D, Viterbo J. Mining direct acyclic graphs to find frequent substructures - an experimental analysis on educational data. Inform Sci. 2019;482:266–78.

    Article  Google Scholar 

  53. Winship C, Knight C. The causal implications of mechanistic thinking: identification using Directed Acyclic Graphs (DAGs). In: Handbook of Causal Analysis for Social Research. Dordrecht: Springer Science & Business Media; 2013. p. 275-99.

  54. Changpetch P, Haughton D. Sociological mechanisms underlying alcohol, tobacco, and gambling: a causal mediation analysis. Thail Statist. 2018;16(1):56–63.

    Google Scholar 

  55. Kohler U, Class F, Sawert T. Control variable selection in applied quantitative sociology: a critical review. Eur Sociol Rev. 2023. https://doi.org/10.1093/esr/jcac078

  56. Gongola A, Bradshaw JC. Directed acyclic graphs in surgical research. J Surg Res. 2023;282:285–8.

    Article  PubMed  Google Scholar 

  57. Tennant PWG, Murray EJ, Arnold KF, et al. Use of directed acyclic graphs (DAGs) to identify confounders in applied health research: review and recommendations. Int J Epidemiol. 2021;50(2):620–32.

    Article  PubMed  Google Scholar 

  58. Gaskell AL, Sleigh JW. An introduction to causal diagrams for anesthesiology research. Anesthesiol. 2020;132(5):951–967. This paper discusses directed acyclic graphs (DAGs) and their utility in anesthesiology research.

  59. Walkden GJ, Pickering AE, Gill H. Assessing Long-term Neurodevelopmental Outcome Following General Anesthesia in Early Childhood: Challenges and Opportunities. Anesth Analg. 2019;128(4):681-694.

  60. Hansen TG, Pedersen JK, Henneberg SW, et al. Academic performance in adolescence after inguinal hernia repair in infancy: a nationwide cohort study. Anesthesiol. 2011;114(5):1076–85.

    Article  Google Scholar 

  61. Ing C, DiMaggio C, Whitehouse A, et al. Long-term differences in language and cognitive function after childhood exposure to anesthesia. Pediatrics. 2012;130(3):e476-485.

    Article  PubMed  Google Scholar 

  62. DiMaggio C, Sun LS, Li G. Early childhood exposure to anesthesia and risk of developmental and behavioral disorders in a sibling birth cohort. Anesth Analg. 2011;113(5):1143–51.

    Article  PubMed  PubMed Central  Google Scholar 

  63. O’Leary JD, Janus M, Duku E, et al. Influence of surgical procedures and general anesthesia on child development before primary school entry among matched sibling pairs. JAMA Pediatr. 2019;173(1):29–36.

    Article  PubMed  Google Scholar 

  64. Bartels M, Althoff RR, Boomsma DI. Anesthesia and cognitive performance in children: no evidence for a causal relationship. Twin Res Hum Genet. 2009;12(3):246–53.

    Article  PubMed  Google Scholar 

  65. Jacola LM, Anghelescu DL, Hall L, et al. Anesthesia exposure during therapy predicts neurocognitive outcomes in survivors of childhood medulloblastoma. J Pediatr. 2020;223:141-147.e144.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Partanen M, Anghelescu DL, Hall L, et al. Longitudinal associations between exposure to anesthesia and neurocognitive functioning in pediatric medulloblastoma. Eur J Cancer. 2021;148:103–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Xiao A, Feng Y, Yu S, Xu C, Chen J, Wang T, Xiao W. General anesthesia in children and long-term neurodevelopmental deficits: a systematic review. Front Mol Neurosci. 2022;15:972025. https://doi.org/10.3389/fnmol.2022.972025

  68. Messeri A, Caprilli S, Busoni P. Anaesthesia induction in children: a psychological evaluation of the efficiency of parents’ presence. Pediatr Anesth. 2004;14(7):551–6.

    Article  Google Scholar 

  69. Lerwick JL. Psychosocial implications of pediatric surgical hospitalization. Semin Pediatr Surg. 2013;22(3):129–33.

    Article  PubMed  Google Scholar 

  70. Kain ZN, Caldwell-Andrews AA, Maranets I, et al. Preoperative anxiety and emergence delirium and postoperative maladaptive behaviors. Anesth Analg. 2004;99(6):1648–54.

    Article  PubMed  Google Scholar 

  71. Chieng YJS, Chan WCS, Klainin-Yobas P, He HG. Perioperative anxiety and postoperative pain in children and adolescents undergoing elective surgical procedures: a quantitative systematic review. J Adv Nurs. 2014;70(2):243–55.

    Article  PubMed  Google Scholar 

  72. Kain ZN. Postoperative maladaptive behavioral changes in children: incidence, risks factors and interventions. Acta Anaesthesiol Belg. 2000;51(4):217–26.

    CAS  PubMed  Google Scholar 

  73. Kj A, Fm S. Can adverse neonatal experiences alter brain development and subsequent behavior? Biol Neonate. 2000;77(2):69–82.

    Article  Google Scholar 

  74. Anand KJS, Aranda JV, Berde CB, et al. Summary proceedings from the neonatal pain-control group. Pediatrics. 2006;117(3):S9–22.

    Article  PubMed  Google Scholar 

  75. Berardi N, Pizzorusso T, Maffei L. Critical periods during sensory development. Curr Opin Neurobiol. 2000;10(1):138–45.

    Article  CAS  PubMed  Google Scholar 

  76. Fitzgerald M, Jennings E. The postnatal development of spinal sensory processing. P Natl Acad Sci USA. 1999;96(14):7719–22.

    Article  ADS  CAS  Google Scholar 

  77. A T. Pain management for neonatal circumcision. Paediatr Drugs. 2001;3(2):101–11.

  78. Hermann C, Hohmeister J, Demirakca S, Zohsel K, Flor H. Long-term alteration of pain sensitivity in school-aged children with early pain experiences. Pain. 2006;125(3):278–85.

    Article  PubMed  Google Scholar 

  79. Ward CG, Loepke AW. Anesthetics and sedatives: toxic or protective for the developing brain? Pharmacol Res. 2012;65(3):271–4.

    Article  CAS  PubMed  Google Scholar 

  80. Rovnaghi CR, Garg S, Hall RW, Bhutta AT, Anand KJS. Ketamine analgesia for inflammatory pain in neonatal rats: a factorial randomized trial examining long-term effects. Behav Brain Funct. 2008;4. https://doi.org/10.1186/1744-9081-4-35

  81. Anand KJS, Garg S, Rovnaghi CR, Narsinghani U, Bhutta AT, Hall RW. Ketamine reduces the cell death following inflammatory pain in newborn rat brain. Pediatr Res. 2007;62(3):283–90.

    Article  CAS  PubMed  Google Scholar 

  82. Avramescu S, Wang DS, Lecker I, et al. Inflammation increases neuronal sensitivity to general anesthetics. Anesthesiol. 2016;124(2):417–27.

    Article  CAS  Google Scholar 

  83. Knuesel I, Chicha L, Britschgi M, et al. Maternal immune activation and abnormal brain development across CNS disorders. Nat Rev Neurol. 2014;10(11):643–60.

    Article  CAS  PubMed  Google Scholar 

  84. Kuban KCK, O’Shea M, Allred EN, et al. The breadth and type of systemic inflammation and the risk of adverse neurological outcomes in extremely low gestation newborns. Pediatr Neurol. 2015;52(1):42–8.

    Article  PubMed  Google Scholar 

  85. Jiang NM, Cowan M, Moonah SN, Petri WA. The impact of systemic inflammation on neurodevelopment. Trends Mol Med. 2018;24(9):794–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. McCann ME, Schouten ANJ. Beyond survival; influences of blood pressure, cerebral perfusion and anesthesia on neurodevelopment. Pediatr Anesth. 2014;24(1):68–73.

    Article  Google Scholar 

  87. Weiss M, Bissonnette B, Engelhardt T, Soriano S. Anesthetists rather than anesthetics are the threat to baby brains. Pediatr Anesth. 2013;23(10):881–2.

    Article  Google Scholar 

  88. McCann ME, Lee JK, Inder T. Beyond anesthesia toxicity: anesthetic considerations to lessen the risk of neonatal neurological injury. Anesth Analg. 2019;129(5):1354–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. de Graaff JC, Pasma W, van Buuren S, et al. Reference values for noninvasive blood pressure in children during anesthesia: a multicentered retrospective observational cohort study. Anesthesiol. 2016;125(5):904–13.

    Article  Google Scholar 

  90. McCann ME, Withington DE, Arnup SJ, et al. Differences in blood pressure in infants after general anesthesia compared to awake regional anesthesia (GAS study-a prospective randomized trial). Anesth Analg. 2017;125(3):837–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Ing C, Sun LS, Friend AF, et al. Differences in intraoperative hemodynamics between spinal and general anesthesia in infants undergoing pyloromyotomy. Paediatr Anaesth. 2017;27(7):733–41.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Maheshwari K, Ahuja S, Khanna AK, et al. Association between perioperative hypotension and delirium in postoperative critically ill patients: a retrospective cohort analysis. Anesth Analg. 2020;130(3):636–43.

    Article  PubMed  Google Scholar 

  93. Sun LY, Wijeysundera DN, Tait GA, Beattie WS. Association of intraoperative hypotension with acute kidney injury after elective noncardiac surgery. Anesthesiol. 2015;123(3):515–23.

    Article  Google Scholar 

  94. van Waes JA, van Klei WA, Wijeysundera DN, van Wolfswinkel L, Lindsay TF, Beattie WS. Association between intraoperative hypotension and myocardial injury after vascular surgery. Anesthesiol. 2016;124(1):35–44.

    Article  Google Scholar 

  95. Hansen TG, Lonnqvist PA. The rise and fall of anaesthesia-related neurotoxicity and the immature developing human brain. Acta Anaesthesiol Scand. 2016;60(3):280–3.

    Article  CAS  PubMed  Google Scholar 

  96. Gleich SJ, Shi Y, Flick R, et al. Hypotension and adverse neurodevelopmental outcomes among children with multiple exposures to general anesthesia: subanalysis of the Mayo Anesthesia Safety in Kids (MASK) Study. Paediatr Anaesth. 2021;31(3):282–289. This paper was a sub-analysis of the widely cited MASK study and found no evidence to support the hypothesis that physiologic disturbances such as intraoperative hypotension was associated with adverse neurodevelopmental outcomes in children exposed to multiple anesthetics before age 3.

  97. VanderWeele TJ. Explanation in causal inference : methods for mediation and interaction. New York: Oxford University Press; 2015.

    Google Scholar 

  98. Robins JM, Greenland S. Identifiability and exchangeability for direct and indirect effects. Epidemiol. 1992;3(2):143–55.

    Article  CAS  Google Scholar 

  99. Pearl J. Direct and indirect effects. In: Proceedings of the Seventeenth conference on Uncertainty in artificial intelligence (UAI 2001). Morgan Kaufmann Publishers Inc.; 2001. p. 411–420.

  100. Shi B, Choirat C, Coull BA, VanderWeele TJ, Valeri L. CMAverse: a suite of functions for reproducible causal mediation analyses. Epidemiol. 2021;32(5):e20–2. https://doi.org/10.1097/EDE.0000000000001378.

    Article  Google Scholar 

  101. Tingley D, Yamamoto T, Hirose K, Keele L, Imai K. mediation: R package for causal mediation analysis. J Stat Softw. Aug 2014;59(5). https://doi.org/10.18637/jss.v059.i05

Download references

Author information

Authors and Affiliations

Authors

Contributions

T.S. and C.I. wrote the main manuscript text. A.P. and C.M. also contributed to writing and editing the manuscript. A.P. prepared Figure 1. All authors reviewed the manuscript.

Corresponding author

Correspondence to Caleb Ing.

Ethics declarations

Conflict of Interest and Financial Disclosure Statement

CI and CM are supported by the Agency for Healthcare Research and Quality (AHRQ) under award number R01HS026493. AP is supported by the National Institute on Drug Abuse (NIDA) under award number T32DA031099. The content is solely the responsibility of the author and does not necessarily represent the official views of the AHRQ or NIDA. There are no commercial associations, or any other conditions posing a conflict of interest to report for any of the above authors.

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

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Singh, T., Pitts, A., Miles, C. et al. Anesthetic Exposure During Early Childhood and Neurodevelopmental Outcomes: Our Current Understanding. Curr Anesthesiol Rep 14, 15–24 (2024). https://doi.org/10.1007/s40140-023-00592-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40140-023-00592-y

Keywords

Navigation