CC BY-NC-ND 4.0 · J Neuroanaesth Crit Care 2018; 05(02): 77-82
DOI: 10.1055/s-0038-1654746
Review Article
Indian Society of Neuroanaesthesiology and Critical Care

Ketamine: A Neuroanesthesiologist's Friend or Foe?

Ankur Luthra
1   Department of Anaesthesia and Intensive Care, Postgraduate Institute of Medical Education and Research, Chandigarh, India
,
Girija Prasad Rath
2   Department of Neuroanaesthesiology and Critical Care, All India Institute of Medical Sciences, New Delhi, India
› Institutsangaben
Weitere Informationen

Publikationsverlauf

Received: 22. Februar 2018

Accepted: 16. April 2018

Publikationsdatum:
24. Mai 2018 (online)

Abstract

Ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, introduced into clinical practice six decades ago, has often been the worst fear of a neuroanesthesiologist due to its concerns of causing a rise in intracranial pressure and increasing cerebral blood volume. However, the recent literature clarifies that it may have a beneficial role, even in neurosurgical patients, because of its propensity to cause neuroprotection through antiglutamatergic action on the NMDA receptors in the ischemic brain. Apart from having an anticonvulsive role in refractory status epilepticus, its inherent property of increasing blood flow to the ischemic areas offers protection to the penumbral zone, thus preventing secondary brain injuries. Also, it has been widely used as an analgesic at subanesthetic doses post spine surgeries. Recently, enough scientific evidence has been published in favor of ketamine establishing the fact that it does not cause a rise in intracranial pressure when the patient is mechanically ventilated and normocarbia is maintained. It has also been used during evoked potential monitoring as it amplifies signals even under general anesthesia and proves to be quite effective during neuromonitoring. However, its adverse effects of increasing muscle tone, excessive salivation, emergence delirium, agitation, and “out of body” experiences with long-term psychomimetic effects and potential to cause addiction have precluded its widespread use.

This review on ketamine summarizes the benefits of using it in neurosurgical anesthesia with the aim of removing the fears we had in the past, which did not seem to be evidence based.

 
  • References

  • 1 Domino EF. Taming the ketamine tiger. 1965. Anesthesiology 2010; 113 (03) 678-684
  • 2 Salt TE, Wilson DG, Prasad SK. Antagonism of N-methylaspartate and synaptic responses of neurones in the rat ventrobasal thalamus by ketamine and MK-801. Br J Pharmacol 1988; 94 (02) 443-448
  • 3 Durieux ME. Inhibition by ketamine of muscarinic acetylcholine receptor function. Anesth Analg 1995; 81 (01) 57-62
  • 4 Smith DJ, Bouchal RL, deSanctis CA. et al. Properties of the interaction between ketamine and opiate binding sites in vivo and in vitro. Neuropharmacology 1987; 26 (09) 1253-1260
  • 5 Crisp T, Perrotti JM, Smith DL, Stafinsky JL, Smith DJ. The local monoaminergic dependency of spinal ketamine. Eur J Pharmacol 1991; 194 (02) (03) 167-172
  • 6 Baum VC, Tecson ME. Ketamine inhibits transsarcolemmal calcium entry in guinea pig myocardium: direct evidence by single cell voltage clamp. Anesth Analg 1991; 73 (06) 804-807
  • 7 Lundy PM, Lockwood PA, Thompson G, Frew R. Differential effects of ketamine isomers on neuronal and extraneuronal catecholamine uptake mechanisms. Anesthesiology 1986; 64 (03) 359-363
  • 8 Hirota K, Lambert DG. Ketamine: its mechanism(s) of action and unusual clinical uses. Br J Anaesth 1996; 77 (04) 441-444
  • 9 Kolenda H, Gremmelt A, Rading S, Braun U, Markakis E. Ketamine for analgosedative therapy in intensive care treatment of head-injured patients. Acta Neurochir (Wien) 1996; 138 (10) 1193-1199
  • 10 Urwin SC, Menon DK. Comparative tolerability of sedative agents in head-injured adults. Drug Saf 2004; 27 (02) 107-133
  • 11 Hudetz JA, Pagel PS. Neuroprotection by ketamine: a review of the experimental and clinical evidence. J Cardiothorac Vasc Anesth 2010; 24 (01) 131-142
  • 12 Långsjö JW, Kaisti KK, Aalto S. et al. Effects of subanesthetic doses of ketamine on regional cerebral blood flow, oxygen consumption, and blood volume in humans. Anesthesiology 2003; 99 (03) 614-623
  • 13 Bar-Joseph G, Guilburd Y, Tamir A, Guilburd JN. Effectiveness of ketamine in decreasing intracranial pressure in children with intracranial hypertension. J Neurosurg Pediatr 2009; 4 (01) 40-46
  • 14 Bourgoin A, Albanèse J, Wereszczynski N, Charbit M, Vialet R, Martin C. Safety of sedation with ketamine in severe head injury patients: comparison with sufentanil. Crit Care Med 2003; 31 (03) 711-717
  • 15 Nagase K, Iida H, Ohata H, Dohi S. Ketamine, not propofol, attenuates cerebrovascular response to carbon dioxide in humans with isoflurane anesthesia. J Clin Anesth 2001; 13 (08) 551-555
  • 16 Sakai K, Cho S, Fukusaki M, Shibata O, Sumikawa K. The effects of propofol with and without ketamine on human cerebral blood flow velocity and CO(2) response. Anesth Analg 2000; 90 (02) 377-382
  • 17 Hardingham GE. Coupling of the NMDA receptor to neuroprotective and neurodestructive events. Biochem Soc Trans 2009; 37 (Pt (06) 1147-1160
  • 18 Lafon-Cazal M, Perez V, Bockaert J, Marin P. Akt mediates the anti-apoptotic effect of NMDA but not that induced by potassium depolarization in cultured cerebellar granule cells. Eur J Neurosci 2002; 16 (04) 575-583
  • 19 Soriano FX, Papadia S, Hofmann F, Hardingham NR, Bading H, Hardingham GE. Preconditioning doses of NMDA promote neuroprotection by enhancing neuronal excitability. J Neurosci 2006; 26 (17) 4509-4518
  • 20 Lau D, Bading H. Synaptic activity-mediated suppression of p53 and induction of nuclear calcium-regulated neuroprotective genes promote survival through inhibition of mitochondrial permeability transition. J Neurosci 2009; 29 (14) 4420-4429
  • 21 Al-Mubarak B, Soriano FX, Hardingham GE. Synaptic NMDAR activity suppresses FOXO1 expression via a cis-acting FOXO binding site: FOXO1 is a FOXO target gene. Channels (Austin) 2009; 3 (04) 233-238
  • 22 Xu J, Kurup P, Zhang Y. et al. Extrasynaptic NMDA receptors couple preferentially to excitotoxicity via calpain-mediated cleavage of STEP. J Neurosci 2009; 29 (29) 9330-9343
  • 23 Hardingham GE, Fukunaga Y, Bading H. Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. Nat Neurosci 2002; 5 (05) 405-414
  • 24 Liu Y, Zhang G, Gao C, Hou X. NMDA receptor activation results in tyrosine phosphorylation of NMDA receptor subunit 2A(NR2A) and interaction of Pyk2 and Src with NR2A after transient cerebral ischemia and reperfusion. Brain Res 2001; 909 (01) (2) 51-58
  • 25 Lan JY, Skeberdis VA, Jover T. et al. Protein kinase C modulates NMDA receptor trafficking and gating. Nat Neurosci 2001; 4 (04) 382-390
  • 26 Ankarcrona M, Dypbukt JM, Bonfoco E. et al. Glutamate-induced neuronal death: a succession of necrosis or apoptosis depending on mitochondrial function. Neuron 1995; 15 (04) 961-973
  • 27 Dick O, Bading H. Synaptic activity and nuclear calcium signaling protect hippocampal neurons from death signal-associated nuclear translocation of FoxO3a induced by extrasynaptic N-methyl-D-aspartate receptors. J Biol Chem 2010; 285 (25) 19354-19361
  • 28 Grathwohl KW, Black IH, Spinella PC. et al. Total intravenous anesthesia including ketamine versus volatile gas anesthesia for combat-related operative traumatic brain injury. Anesthesiology 2008; 109 (01) 44-53
  • 29 Cohen L, Athaide V, Wickham ME, Doyle-Waters MM, Rose NGW, Hohl CM. The effect of ketamine on intracranial and cerebral perfusion pressure and health outcomes: a systematic review. Ann Emerg Med 2015; 65 (01) 43-51.e2
  • 30 Zeiler FA, Teitelbaum J, West M, Gillman LM. The ketamine effect on ICP in traumatic brain injury. Neurocrit Care 2014; 21 (01) 163-173
  • 31 Bucher J, Koyfman A. Intubation of the neurologically injured patient. J Emerg Med 2015; 49 (06) 920-927
  • 32 Jabre P, Combes X, Lapostolle F. et al; KETASED Collaborative Study Group. Etomidate versus ketamine for rapid sequence intubation in acutely ill patients: a multicentre randomised controlled trial. Lancet 2009; 374 9686 293-300
  • 33 Dorsch NW, King MT. A review of cerebral vasospasm in aneurysmal subarachnoid haemorrhage. Part I: Incidence and effects. J Clin Neurosci 1994; 1 (01) 19-26
  • 34 Kassell NF, Torner JC, Haley Jr EC, Jane JA, Adams HP, Kongable GL. The International Cooperative Study on the Timing of Aneurysm Surgery. Part 1: overall management results. J Neurosurg 1990; 73 (01) 18-36
  • 35 Leao A. Spreading depression of activity in the cerebral cortex. J Neurophysiol 1944; 7: 359
  • 36 Carlson AP, Abbas M, Alunday R, Qeadank F, Shuttleworth CW. 375 ketamine inhibits cortical spreading depolarization in acute brain injury: a prospective randomized multiple crossover trial. Neurosurgery 2017; 64 e1 288-289
  • 37 Gakuba C, Gauberti M, Mazighi M, Defer G, Hanouz JL, Vivien D. Preclinical evidence toward the use of ketamine for recombinant tissue-type plasminogen activator-mediated thrombolysis under anesthesia or sedation. Stroke 2011; 42 (10) 2947-2949
  • 38 Nemergut EC, Durieux ME, Missaghi NB, Himmelseher S. Pain management after craniotomy. Best Pract Res Clin Anaesthesiol 2007; 21 (04) 557-573
  • 39 Pendi A, Field R, Farhan SD, Eichler M, Bederman SS. Perioperative ketamine for analgesia in spine surgery: a meta-analysis of randomized controlled trials. Spine 2018; 43 (05) E299-E307
  • 40 Hudetz JA, Iqbal Z, Gandhi SD. et al. Ketamine attenuates post-operative cognitive dysfunction after cardiac surgery. Acta Anaesthesiol Scand 2009; 53 (07) 864-872
  • 41 Lee KH, Kim JY, Kim JW, Park JS, Lee KW, Jeon SY. Influence of ketamine on early postoperative cognitive function after orthopedic surgery in elderly patients. Anesth Pain Med 2015; 5 (05) e28844
  • 42 Synowiec AS, Singh DS, Yenugadhati V, Valeriano JP, Schramke CJ, Kelly KM. Ketamine use in the treatment of refractory status epilepticus. Epilepsy Res 2013; 105 (01) (2) 183-188
  • 43 Fang Y, Wang X. Ketamine for the treatment of refractory status epilepticus. Seizure 2015; 30: 14-20
  • 44 Rysz A, Bachański M, Bidziński J, Bacia T. [The comparison of ketamine with methohexital and thiopental in the intraoperative EEG in drug-resistant epilepsy] [in Polish]. Neurol Neurochir Pol 1998; 32 (02) Suppl 2) 237-245
  • 45 Kano T, Shimoji K. The effects of ketamine and neuroleptanalgesia on the evoked electrospinogram and electromyogram in man. Anesthesiology 1974; 40 (03) 241-246
  • 46 Schubert A, Licina MG, Lineberry PJ. The effect of ketamine on human somatosensory evoked potentials and its modification by nitrous oxide. Anesthesiology 1990; 72 (01) 33-39
  • 47 Glassman SD, Shields CB, Linden RD, Zhang YP, Nixon AR, Johnson JR. Anesthetic effects on motor evoked potentials in dogs. Spine 1993; 18 (08) 1083-1089
  • 48 Schmidt A, Ryding E, Akeson J. Racemic ketamine does not abolish cerebrovascular autoregulation in the pig. Acta Anaesthesiol Scand 2003; 47 (05) 569-575
  • 49 Engelhard K, Werner C, Möllenberg O, Kochs E. S(+)-ketamine/propofol maintain dynamic cerebrovascular autoregulation in humans. Can J Anaesth 2001; 48 (10) 1034-1039
  • 50 Loflin R, Koyfman A. When used for sedation, does ketamine increase intracranial pressure more than fentanyl or sufentanil?. Ann Emerg Med 2015; 65 (01) 55-56
  • 51 Albanèse J, Arnaud S, Rey M, Thomachot L, Alliez B, Martin C. Ketamine decreases intracranial pressure and electroencephalographic activity in traumatic brain injury patients during propofol sedation. Anesthesiology 1997; 87 (06) 1328-1334
  • 52 Caricato A, Tersali A, Pitoni S. et al. Racemic ketamine in adult head injury patients: use in endotracheal suctioning. Crit Care 2013; 17 (06) R267
  • 53 Vick PG, Lamer TJ. Treatment of central post-stroke pain with oral ketamine. Pain 2001; 92 (01) (2) 311-313
  • 54 O'Brien SL, Pangarkar S, Prager J. The use of ketamine in neuropathic pain. Curr Phys Med Rehabil Rep 2014; 2: 128-145