Skip to main content
Top
Published in: Clinical Pharmacokinetics 9/2016

01-09-2016 | Review Article

Ketamine: A Review of Clinical Pharmacokinetics and Pharmacodynamics in Anesthesia and Pain Therapy

Authors: Marko A. Peltoniemi, Nora M. Hagelberg, Klaus T. Olkkola, Teijo I. Saari

Published in: Clinical Pharmacokinetics | Issue 9/2016

Login to get access

Abstract

Ketamine is a phencyclidine derivative, which functions primarily as an antagonist of the N-methyl-d-aspartate receptor. It has no affinity for gamma-aminobutyric acid receptors in the central nervous system. Ketamine shows a chiral structure consisting of two optical isomers. It undergoes oxidative metabolism, mainly to norketamine by cytochrome P450 (CYP) 3A and CYP2B6 enzymes. The use of S-ketamine is increasing worldwide, since the S(+)-enantiomer has been postulated to be a four times more potent anesthetic and analgesic than the R(−)-enantiomer and approximately two times more effective than the racemic mixture of ketamine. Because of extensive first-pass metabolism, oral bioavailability is poor and ketamine is vulnerable to pharmacokinetic drug interactions. Sublingual and nasal formulations of ketamine are being developed, and especially nasal administration produces rapid maximum plasma ketamine concentrations with relatively high bioavailability. Ketamine produces hemodynamically stable anesthesia via central sympathetic stimulation without affecting respiratory function. Animal studies have shown that ketamine has neuroprotective properties, and there is no evidence of elevated intracranial pressure after ketamine dosing in humans. Low-dose perioperative ketamine may reduce opioid consumption and chronic postsurgical pain after specific surgical procedures. However, long-term analgesic effects of ketamine in chronic pain patients have not been demonstrated. Besides analgesic properties, ketamine has rapid-acting antidepressant effects, which may be useful in treating therapy-resistant depressive patients. Well-known psychotomimetic and cognitive adverse effects restrict the clinical usefulness of ketamine, even though fewer psychomimetic adverse effects have been reported with S-ketamine in comparison with the racemate. Safety issues in long-term use are yet to be resolved.
Literature
1.
go back to reference Domino EF. Taming the ketamine tiger. 1965. Anesthesiology. 2010;113:678–84.PubMed Domino EF. Taming the ketamine tiger. 1965. Anesthesiology. 2010;113:678–84.PubMed
2.
go back to reference Domino EF, Chodoff P, Corssen G. Pharmacologic effects of CI-581, a new dissociative anesthetic, in man. Clin Pharmacol Ther. 1965;6:279–91.PubMedCrossRef Domino EF, Chodoff P, Corssen G. Pharmacologic effects of CI-581, a new dissociative anesthetic, in man. Clin Pharmacol Ther. 1965;6:279–91.PubMedCrossRef
5.
go back to reference Weber F, Wulf H, Gruber M, Biallas R. S-ketamine and s-norketamine plasma concentrations after nasal and i.v. administration in anesthetized children. Paediatr Anaesth. 2004;14:983–8.PubMedCrossRef Weber F, Wulf H, Gruber M, Biallas R. S-ketamine and s-norketamine plasma concentrations after nasal and i.v. administration in anesthetized children. Paediatr Anaesth. 2004;14:983–8.PubMedCrossRef
6.
go back to reference Sigtermans MJ, van Hilten JJ, Bauer MCR, Arbous MS, Marinus J, Sarton EY, et al. Ketamine produces effective and long-term pain relief in patients with complex regional pain syndrome type 1. Pain. 2009;145:304–11.PubMedCrossRef Sigtermans MJ, van Hilten JJ, Bauer MCR, Arbous MS, Marinus J, Sarton EY, et al. Ketamine produces effective and long-term pain relief in patients with complex regional pain syndrome type 1. Pain. 2009;145:304–11.PubMedCrossRef
7.
go back to reference Chong C, Schug SA, Page-Sharp M, Jenkins B, Ilett KF. Development of a sublingual/oral formulation of ketamine for use in neuropathic pain: preliminary findings from a three-way randomized, crossover study. Clin Drug Investig. 2009;29:317–24.PubMedCrossRef Chong C, Schug SA, Page-Sharp M, Jenkins B, Ilett KF. Development of a sublingual/oral formulation of ketamine for use in neuropathic pain: preliminary findings from a three-way randomized, crossover study. Clin Drug Investig. 2009;29:317–24.PubMedCrossRef
8.
go back to reference Huge V, Lauchart M, Magerl W, Schelling G, Beyer A, Thieme D, et al. Effects of low-dose intranasal (S)-ketamine in patients with neuropathic pain. Eur J Pain. 2010;14:387–94.PubMedCrossRef Huge V, Lauchart M, Magerl W, Schelling G, Beyer A, Thieme D, et al. Effects of low-dose intranasal (S)-ketamine in patients with neuropathic pain. Eur J Pain. 2010;14:387–94.PubMedCrossRef
9.
go back to reference Riediger C, Haschke M, Bitter C, Fabbro T, Schaeren S, Urwyler A, et al. The analgesic effect of combined treatment with intranasal S-ketamine and intranasal midazolam compared with morphine patient-controlled analgesia in spinal surgery patients: a pilot study. J Pain Res. 2015;8:87–94.PubMedPubMedCentral Riediger C, Haschke M, Bitter C, Fabbro T, Schaeren S, Urwyler A, et al. The analgesic effect of combined treatment with intranasal S-ketamine and intranasal midazolam compared with morphine patient-controlled analgesia in spinal surgery patients: a pilot study. J Pain Res. 2015;8:87–94.PubMedPubMedCentral
10.
go back to reference Grant IS, Nimmo WS, Clements JA. Pharmacokinetics and analgesic effects of i.m. and oral ketamine. Br J Anaesth. 1981;53:805–10.PubMedCrossRef Grant IS, Nimmo WS, Clements JA. Pharmacokinetics and analgesic effects of i.m. and oral ketamine. Br J Anaesth. 1981;53:805–10.PubMedCrossRef
11.
go back to reference Clements JA, Nimmo WS, Grant IS. Bioavailability, pharmacokinetics, and analgesic activity of ketamine in humans. J Pharm Sci. 1982;71:539–42.PubMedCrossRef Clements JA, Nimmo WS, Grant IS. Bioavailability, pharmacokinetics, and analgesic activity of ketamine in humans. J Pharm Sci. 1982;71:539–42.PubMedCrossRef
12.
go back to reference Yanagihara Y, Ohtani M, Kariya S, Uchino K, Hiraishi T, Ashizawa N, et al. Plasma concentration profiles of ketamine and norketamine after administration of various ketamine preparations to healthy Japanese volunteers. Biopharm Drug Dispos. 2003;24:37–43.PubMedCrossRef Yanagihara Y, Ohtani M, Kariya S, Uchino K, Hiraishi T, Ashizawa N, et al. Plasma concentration profiles of ketamine and norketamine after administration of various ketamine preparations to healthy Japanese volunteers. Biopharm Drug Dispos. 2003;24:37–43.PubMedCrossRef
13.
go back to reference Peltoniemi MA, Saari TI, Hagelberg NM, Laine K, Kurkinen KJ, Neuvonen PJ, et al. Rifampicin has a profound effect on the pharmacokinetics of oral S-ketamine and less on intravenous S-ketamine. Basic Clin Pharmacol Toxicol. 2012;111:325–32.PubMedCrossRef Peltoniemi MA, Saari TI, Hagelberg NM, Laine K, Kurkinen KJ, Neuvonen PJ, et al. Rifampicin has a profound effect on the pharmacokinetics of oral S-ketamine and less on intravenous S-ketamine. Basic Clin Pharmacol Toxicol. 2012;111:325–32.PubMedCrossRef
14.
go back to reference Fanta S, Kinnunen M, Backman JT, Kalso E. Population pharmacokinetics of S-ketamine and norketamine in healthy volunteers after intravenous and oral dosing. Eur J Clin Pharmacol. 2015;71:441–7.PubMedCrossRef Fanta S, Kinnunen M, Backman JT, Kalso E. Population pharmacokinetics of S-ketamine and norketamine in healthy volunteers after intravenous and oral dosing. Eur J Clin Pharmacol. 2015;71:441–7.PubMedCrossRef
15.
go back to reference Hagelberg NM, Peltoniemi MA, Saari TI, Kurkinen KJ, Laine K, Neuvonen PJ, et al. Clarithromycin, a potent inhibitor of CYP3A, greatly increases exposure to oral S-ketamine. Eur J Pain. 2010;14:625–9.PubMedCrossRef Hagelberg NM, Peltoniemi MA, Saari TI, Kurkinen KJ, Laine K, Neuvonen PJ, et al. Clarithromycin, a potent inhibitor of CYP3A, greatly increases exposure to oral S-ketamine. Eur J Pain. 2010;14:625–9.PubMedCrossRef
16.
go back to reference White PF, Johnston RR, Pudwill CR. Interaction of ketamine and halothane in rats. Anesthesiology. 1975;42:179–86.PubMedCrossRef White PF, Johnston RR, Pudwill CR. Interaction of ketamine and halothane in rats. Anesthesiology. 1975;42:179–86.PubMedCrossRef
17.
go back to reference Leung LY, Baillie TA. Comparative pharmacology in the rat of ketamine and its two principal metabolites, norketamine and (Z)-6-hydroxynorketamine. J Med Chem. 1986;29:2396–9.PubMedCrossRef Leung LY, Baillie TA. Comparative pharmacology in the rat of ketamine and its two principal metabolites, norketamine and (Z)-6-hydroxynorketamine. J Med Chem. 1986;29:2396–9.PubMedCrossRef
18.
go back to reference Ebert B, Mikkelsen S, Thorkildsen C, Borgbjerg FM. Norketamine, the main metabolite of ketamine, is a non-competitive NMDA receptor antagonist in the rat cortex and spinal cord. Eur J Pharmacol. 1997;333:99–104.PubMedCrossRef Ebert B, Mikkelsen S, Thorkildsen C, Borgbjerg FM. Norketamine, the main metabolite of ketamine, is a non-competitive NMDA receptor antagonist in the rat cortex and spinal cord. Eur J Pharmacol. 1997;333:99–104.PubMedCrossRef
19.
go back to reference Holtman JR, Crooks PA, Johnson-Hardy JK, Hojomat M, Kleven M, Wala EP. Effects of norketamine enantiomers in rodent models of persistent pain. Pharmacol Biochem Behav. 2008;90:676–85.PubMedCrossRef Holtman JR, Crooks PA, Johnson-Hardy JK, Hojomat M, Kleven M, Wala EP. Effects of norketamine enantiomers in rodent models of persistent pain. Pharmacol Biochem Behav. 2008;90:676–85.PubMedCrossRef
20.
go back to reference Laskowski K, Stirling A, McKay WP, Lim HJ. A systematic review of intravenous ketamine for postoperative analgesia. Can J Anesth. 2011;58:911–23.PubMedCrossRef Laskowski K, Stirling A, McKay WP, Lim HJ. A systematic review of intravenous ketamine for postoperative analgesia. Can J Anesth. 2011;58:911–23.PubMedCrossRef
21.
go back to reference Adams HA, Werner C. From the racemate to the eutomer: (S)-ketamine. Renaissance of a substance? Anaesthesist. 1997;46:1026–42.PubMedCrossRef Adams HA, Werner C. From the racemate to the eutomer: (S)-ketamine. Renaissance of a substance? Anaesthesist. 1997;46:1026–42.PubMedCrossRef
22.
go back to reference Mion G, Villevieille T. Ketamine pharmacology: an update (pharmacodynamics and molecular aspects, recent findings). CNS Neurosci Ther. 2013;19:370–80.PubMedCrossRef Mion G, Villevieille T. Ketamine pharmacology: an update (pharmacodynamics and molecular aspects, recent findings). CNS Neurosci Ther. 2013;19:370–80.PubMedCrossRef
23.
go back to reference White PF, Ham J, Way WL, Trevor AJ. Pharmacology of ketamine isomers in surgical patients. Anesthesiology. 1980;52:231–9.PubMedCrossRef White PF, Ham J, Way WL, Trevor AJ. Pharmacology of ketamine isomers in surgical patients. Anesthesiology. 1980;52:231–9.PubMedCrossRef
24.
go back to reference Oye I, Paulsen O, Maurset A. Effects of ketamine on sensory perception: evidence for a role of N-methyl-d-aspartate receptors. J Pharmacol Exp Ther. 1992;260:1209–13.PubMed Oye I, Paulsen O, Maurset A. Effects of ketamine on sensory perception: evidence for a role of N-methyl-d-aspartate receptors. J Pharmacol Exp Ther. 1992;260:1209–13.PubMed
25.
go back to reference Arendt-Nielsen L, Nielsen J, Petersen-Felix S, Schnider TW, Zbinden AM. Effect of racemic mixture and the (S+)-isomer of ketamine on temporal and spatial summation of pain. Br J Anaesth. 1996;77:625–31.PubMedCrossRef Arendt-Nielsen L, Nielsen J, Petersen-Felix S, Schnider TW, Zbinden AM. Effect of racemic mixture and the (S+)-isomer of ketamine on temporal and spatial summation of pain. Br J Anaesth. 1996;77:625–31.PubMedCrossRef
26.
go back to reference Bell RF, Eccleston C, Kalso EA. Ketamine as an adjuvant to opioids for cancer pain. Cochrane Database Syst Rev. 2012;11:CD003351. Bell RF, Eccleston C, Kalso EA. Ketamine as an adjuvant to opioids for cancer pain. Cochrane Database Syst Rev. 2012;11:CD003351.
27.
go back to reference Bell RF, Dahl JB, Moore RA, Kalso E. Peri-operative ketamine for acute post-operative pain: a quantitative and qualitative systematic review (Cochrane review). Acta Anaesthesiol Scand. 2005;49:1405–28.PubMedCrossRef Bell RF, Dahl JB, Moore RA, Kalso E. Peri-operative ketamine for acute post-operative pain: a quantitative and qualitative systematic review (Cochrane review). Acta Anaesthesiol Scand. 2005;49:1405–28.PubMedCrossRef
28.
go back to reference Abdallah CG, Averill LA, Krystal JH. Ketamine as a promising prototype for a new generation of rapid-acting antidepressants. Ann N Y Acad Sci. 2015;1344:66–77.PubMedPubMedCentralCrossRef Abdallah CG, Averill LA, Krystal JH. Ketamine as a promising prototype for a new generation of rapid-acting antidepressants. Ann N Y Acad Sci. 2015;1344:66–77.PubMedPubMedCentralCrossRef
29.
go back to reference Fang Y, Wang X. Ketamine for the treatment of refractory status epilepticus. Seizure. 2015;30:14–20.PubMedCrossRef Fang Y, Wang X. Ketamine for the treatment of refractory status epilepticus. Seizure. 2015;30:14–20.PubMedCrossRef
30.
go back to reference Dayton PG, Stiller RL, Cook DR, Perel JM. The binding of ketamine to plasma proteins: emphasis on human plasma. Eur J Clin Pharmacol. 1983;24:825–31.PubMedCrossRef Dayton PG, Stiller RL, Cook DR, Perel JM. The binding of ketamine to plasma proteins: emphasis on human plasma. Eur J Clin Pharmacol. 1983;24:825–31.PubMedCrossRef
31.
go back to reference Hijazi Y, Bodonian C, Bolon M, Salord F, Boulieu R. Pharmacokinetics and haemodynamics of ketamine in intensive care patients with brain or spinal cord injury. Br J Anaesth. 2003;90:155–60.PubMedCrossRef Hijazi Y, Bodonian C, Bolon M, Salord F, Boulieu R. Pharmacokinetics and haemodynamics of ketamine in intensive care patients with brain or spinal cord injury. Br J Anaesth. 2003;90:155–60.PubMedCrossRef
32.
go back to reference Geisslinger G, Hering W, Thomann P, Knoll R, Kamp HD, Brune K. Pharmacokinetics and pharmacodynamics of ketamine enantiomers in surgical patients using a stereoselective analytical method. Br J Anaesth. 1993;70:666–71.PubMedCrossRef Geisslinger G, Hering W, Thomann P, Knoll R, Kamp HD, Brune K. Pharmacokinetics and pharmacodynamics of ketamine enantiomers in surgical patients using a stereoselective analytical method. Br J Anaesth. 1993;70:666–71.PubMedCrossRef
33.
go back to reference Dahan A, Olofsenl E, Sigtermans M, Noppers I, Niesters M, Aarts L, et al. Population pharmacokinetic-pharmacodynamic modeling of ketamine-induced pain relief of chronic pain. Eur J Pain. 2011;15:258–67.PubMedCrossRef Dahan A, Olofsenl E, Sigtermans M, Noppers I, Niesters M, Aarts L, et al. Population pharmacokinetic-pharmacodynamic modeling of ketamine-induced pain relief of chronic pain. Eur J Pain. 2011;15:258–67.PubMedCrossRef
34.
go back to reference Sigtermans M, Dahan A, Mooren R, Bauer M, Kest B, Sarton E, et al. S(+)-ketamine effect on experimental pain and cardiac output. Anesthesiology. 2009;111:892–903.PubMedCrossRef Sigtermans M, Dahan A, Mooren R, Bauer M, Kest B, Sarton E, et al. S(+)-ketamine effect on experimental pain and cardiac output. Anesthesiology. 2009;111:892–903.PubMedCrossRef
35.
go back to reference Persson J, Hasselström J, Maurset A, Oye I, Svensson JO, Almqvist O, et al. Pharmacokinetics and non-analgesic effects of S- and R-ketamines in healthy volunteers with normal and reduced metabolic capacity. Eur J Clin Pharmacol. 2002;57:869–75.PubMedCrossRef Persson J, Hasselström J, Maurset A, Oye I, Svensson JO, Almqvist O, et al. Pharmacokinetics and non-analgesic effects of S- and R-ketamines in healthy volunteers with normal and reduced metabolic capacity. Eur J Clin Pharmacol. 2002;57:869–75.PubMedCrossRef
36.
go back to reference White PF, Schüttler J, Shafer A, Stanski DR, Horai Y, Trevor AJ. Comparative pharmacology of the ketamine isomers. Studies in volunteers. Br J Anaesth. 1985;57:197–203.PubMedCrossRef White PF, Schüttler J, Shafer A, Stanski DR, Horai Y, Trevor AJ. Comparative pharmacology of the ketamine isomers. Studies in volunteers. Br J Anaesth. 1985;57:197–203.PubMedCrossRef
37.
go back to reference White M, de Graaff P, Renshof B, van Kan E, Dzoljic M. Pharmacokinetics of S(+) ketamine derived from target controlled infusion. Br J Anaesth. 2006;96:330–4.PubMedCrossRef White M, de Graaff P, Renshof B, van Kan E, Dzoljic M. Pharmacokinetics of S(+) ketamine derived from target controlled infusion. Br J Anaesth. 2006;96:330–4.PubMedCrossRef
38.
go back to reference Schüttler J, Stanski DR, White PF, Trevor AJ, Horai Y, Verotta D, et al. Pharmacodynamic modeling of the EEG effects of ketamine and its enantiomers in man. J Pharmacokinet Biopharm. 1987;15:241–53.PubMedCrossRef Schüttler J, Stanski DR, White PF, Trevor AJ, Horai Y, Verotta D, et al. Pharmacodynamic modeling of the EEG effects of ketamine and its enantiomers in man. J Pharmacokinet Biopharm. 1987;15:241–53.PubMedCrossRef
39.
go back to reference Ihmsen H, Geisslinger G, Schüttler J. Stereoselective pharmacokinetics of ketamine: R(−)-ketamine inhibits the elimination of S(+)-ketamine. Clin Pharmacol Ther. 2001;70:431–8.PubMedCrossRef Ihmsen H, Geisslinger G, Schüttler J. Stereoselective pharmacokinetics of ketamine: R(−)-ketamine inhibits the elimination of S(+)-ketamine. Clin Pharmacol Ther. 2001;70:431–8.PubMedCrossRef
40.
go back to reference Peltoniemi MA, Saari TI, Hagelberg NM, Laine K, Neuvonen PJ, Olkkola KT. St John’s wort greatly decreases the plasma concentrations of oral S-ketamine. Fundam Clin Pharmacol. 2012;26:743–50.PubMedCrossRef Peltoniemi MA, Saari TI, Hagelberg NM, Laine K, Neuvonen PJ, Olkkola KT. St John’s wort greatly decreases the plasma concentrations of oral S-ketamine. Fundam Clin Pharmacol. 2012;26:743–50.PubMedCrossRef
41.
go back to reference Peltoniemi MA, Saari TI, Hagelberg NM, Laine K, Neuvonen PJ, Olkkola KT. S-ketamine concentrations are greatly increased by grapefruit juice. Eur J Clin Pharmacol. 2012;68:979–86.PubMedCrossRef Peltoniemi MA, Saari TI, Hagelberg NM, Laine K, Neuvonen PJ, Olkkola KT. S-ketamine concentrations are greatly increased by grapefruit juice. Eur J Clin Pharmacol. 2012;68:979–86.PubMedCrossRef
42.
go back to reference Peltoniemi MA, Saari TI, Hagelberg NM, Reponen P, Turpeinen M, Laine K, et al. Exposure to oral S-ketamine is unaffected by itraconazole but greatly increased by ticlopidine. Clin Pharmacol Ther. 2011;90:296–302.PubMedCrossRef Peltoniemi MA, Saari TI, Hagelberg NM, Reponen P, Turpeinen M, Laine K, et al. Exposure to oral S-ketamine is unaffected by itraconazole but greatly increased by ticlopidine. Clin Pharmacol Ther. 2011;90:296–302.PubMedCrossRef
43.
go back to reference Woolf TF, Adams JD. Biotransformation of ketamine, (Z)-6-hydroxyketamine, and (E)-6-hydroxyketamine by rat, rabbit, and human liver microsomal preparations. Xenobiotica. 1987;17:839–47.PubMedCrossRef Woolf TF, Adams JD. Biotransformation of ketamine, (Z)-6-hydroxyketamine, and (E)-6-hydroxyketamine by rat, rabbit, and human liver microsomal preparations. Xenobiotica. 1987;17:839–47.PubMedCrossRef
44.
go back to reference Hijazi Y, Boulieu R. Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2002;30:853–8.PubMedCrossRef Hijazi Y, Boulieu R. Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2002;30:853–8.PubMedCrossRef
45.
go back to reference Kharasch ED, Labroo R. Metabolism of ketamine stereoisomers by human liver microsomes. Anesthesiology. 1992;77:1201–7.PubMedCrossRef Kharasch ED, Labroo R. Metabolism of ketamine stereoisomers by human liver microsomes. Anesthesiology. 1992;77:1201–7.PubMedCrossRef
46.
go back to reference Yanagihara Y, Kariya S, Ohtani M, Uchino K, Aoyama T, Yamamura Y, et al. Involvement of CYP2B6 in n-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2001;29:887–90.PubMed Yanagihara Y, Kariya S, Ohtani M, Uchino K, Aoyama T, Yamamura Y, et al. Involvement of CYP2B6 in n-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2001;29:887–90.PubMed
47.
go back to reference Noppers I, Olofsen E, Niesters M, Aarts L, Mooren R, Dahan A, et al. Effect of rifampicin on S-ketamine and S-norketamine plasma concentrations in healthy volunteers after intravenous S-ketamine administration. Anesthesiology. 2011;114:1435–45.PubMedPubMedCentralCrossRef Noppers I, Olofsen E, Niesters M, Aarts L, Mooren R, Dahan A, et al. Effect of rifampicin on S-ketamine and S-norketamine plasma concentrations in healthy volunteers after intravenous S-ketamine administration. Anesthesiology. 2011;114:1435–45.PubMedPubMedCentralCrossRef
48.
go back to reference Herd D, Anderson BJ. Ketamine disposition in children presenting for procedural sedation and analgesia in a children’s emergency department. Paediatr Anaesth. 2007;17:622–9.PubMedCrossRef Herd D, Anderson BJ. Ketamine disposition in children presenting for procedural sedation and analgesia in a children’s emergency department. Paediatr Anaesth. 2007;17:622–9.PubMedCrossRef
49.
go back to reference Herd DW, Anderson BJ, Keene NA, Holford NHG. Investigating the pharmacodynamics of ketamine in children. Paediatr Anaesth. 2008;18:36–42.PubMedCrossRef Herd DW, Anderson BJ, Keene NA, Holford NHG. Investigating the pharmacodynamics of ketamine in children. Paediatr Anaesth. 2008;18:36–42.PubMedCrossRef
50.
go back to reference Herd DW, Anderson BJ, Holford NHG. Modeling the norketamine metabolite in children and the implications for analgesia. Pediatr Anesth. 2007;17:831–40.CrossRef Herd DW, Anderson BJ, Holford NHG. Modeling the norketamine metabolite in children and the implications for analgesia. Pediatr Anesth. 2007;17:831–40.CrossRef
51.
go back to reference Dallimore D, Herd DW, Short T, Anderson BJ. Dosing ketamine for pediatric procedural sedation in the emergency department. Pediatr Emerg Care. 2008;24:529–33.PubMedCrossRef Dallimore D, Herd DW, Short T, Anderson BJ. Dosing ketamine for pediatric procedural sedation in the emergency department. Pediatr Emerg Care. 2008;24:529–33.PubMedCrossRef
52.
go back to reference Brunette KEJ, Anderson BJ, Thomas J, Wiesner L, Herd DW, Schulein S. Exploring the pharmacokinetics of oral ketamine in children undergoing burns procedures. Paediatr Anaesth. 2011;21:653–62.PubMedCrossRef Brunette KEJ, Anderson BJ, Thomas J, Wiesner L, Herd DW, Schulein S. Exploring the pharmacokinetics of oral ketamine in children undergoing burns procedures. Paediatr Anaesth. 2011;21:653–62.PubMedCrossRef
53.
go back to reference Elkomy MH, Drover DR, Hammer GB, Galinkin JL, Ramamoorthy C. Population pharmacokinetics of ketamine in children with heart disease. Int J Pharm. 2015;478:223–31.PubMedCrossRef Elkomy MH, Drover DR, Hammer GB, Galinkin JL, Ramamoorthy C. Population pharmacokinetics of ketamine in children with heart disease. Int J Pharm. 2015;478:223–31.PubMedCrossRef
54.
go back to reference Olofsen E, Noppers I, Niesters M, Kharasch E, Aarts L, Sarton E, et al. Estimation of the contribution of norketamine to ketamine-induced acute pain relief and neurocognitive impairment in healthy volunteers. Anesthesiology. 2012;117:353–64.PubMedPubMedCentralCrossRef Olofsen E, Noppers I, Niesters M, Kharasch E, Aarts L, Sarton E, et al. Estimation of the contribution of norketamine to ketamine-induced acute pain relief and neurocognitive impairment in healthy volunteers. Anesthesiology. 2012;117:353–64.PubMedPubMedCentralCrossRef
55.
go back to reference Zhao X, Venkata SLV, Moaddel R, Luckenbaugh DA, Brutsche NE, Ibrahim L, et al. Simultaneous population pharmacokinetic modelling of ketamine and three major metabolites in patients with treatment-resistant bipolar depression. Br J Clin Pharmacol. 2012;74:304–14.PubMedPubMedCentralCrossRef Zhao X, Venkata SLV, Moaddel R, Luckenbaugh DA, Brutsche NE, Ibrahim L, et al. Simultaneous population pharmacokinetic modelling of ketamine and three major metabolites in patients with treatment-resistant bipolar depression. Br J Clin Pharmacol. 2012;74:304–14.PubMedPubMedCentralCrossRef
56.
go back to reference Absalom AR, Lee M, Menon DK, Sharar SR, De Smet T, Halliday J, et al. Predictive performance of the Domino, Hijazi, and Clements models during low-dose target-controlled ketamine infusions in healthy volunteers. Br J Anaesth. 2007;98:615–23.PubMedCrossRef Absalom AR, Lee M, Menon DK, Sharar SR, De Smet T, Halliday J, et al. Predictive performance of the Domino, Hijazi, and Clements models during low-dose target-controlled ketamine infusions in healthy volunteers. Br J Anaesth. 2007;98:615–23.PubMedCrossRef
57.
go back to reference Jamsen KM, McLeay SC, Barras M, Green B. Reporting a population pharmacokinetic–pharmacodynamic study: a journal’s perspective. Clin Pharmacokinet. 2014;53:111–22.PubMedCrossRef Jamsen KM, McLeay SC, Barras M, Green B. Reporting a population pharmacokinetic–pharmacodynamic study: a journal’s perspective. Clin Pharmacokinet. 2014;53:111–22.PubMedCrossRef
58.
go back to reference Mould DR, Upton RN. Basic concepts in population modeling, simulation, and model-based drug development. CPT Pharmacomet Syst Pharmacol. 2012;1:e6.CrossRef Mould DR, Upton RN. Basic concepts in population modeling, simulation, and model-based drug development. CPT Pharmacomet Syst Pharmacol. 2012;1:e6.CrossRef
59.
go back to reference Kleinloog D, Uit den Boogaard A, Dahan A, Mooren R, Klaassen E, Stevens J, et al. Optimizing the glutamatergic challenge model for psychosis, using S+-ketamine to induce psychomimetic symptoms in healthy volunteers. J Psychopharmacol. 2015;29:401–13.PubMedCrossRef Kleinloog D, Uit den Boogaard A, Dahan A, Mooren R, Klaassen E, Stevens J, et al. Optimizing the glutamatergic challenge model for psychosis, using S+-ketamine to induce psychomimetic symptoms in healthy volunteers. J Psychopharmacol. 2015;29:401–13.PubMedCrossRef
60.
go back to reference Lodge D, Anis NA. Effects of ketamine and three other anaesthetics on spinal reflexes and inhibitions in the cat. Br J Anaesth. 1984;56:1143–51.PubMedCrossRef Lodge D, Anis NA. Effects of ketamine and three other anaesthetics on spinal reflexes and inhibitions in the cat. Br J Anaesth. 1984;56:1143–51.PubMedCrossRef
61.
go back to reference Anis NA, Berry SC, Burton NR, Lodge D. The dissociative anaesthetics, ketamine and phencyclidine, selectively reduce excitation of central mammalian neurones by N-methyl-aspartate. Br J Pharmacol. 1983;79:565–75.PubMedPubMedCentralCrossRef Anis NA, Berry SC, Burton NR, Lodge D. The dissociative anaesthetics, ketamine and phencyclidine, selectively reduce excitation of central mammalian neurones by N-methyl-aspartate. Br J Pharmacol. 1983;79:565–75.PubMedPubMedCentralCrossRef
62.
go back to reference Traynelis SF, Cull-Candy SG. Proton inhibition of N-methyl-d-aspartate receptors in cerebellar neurons. Nature. 1990;345:347–50.PubMedCrossRef Traynelis SF, Cull-Candy SG. Proton inhibition of N-methyl-d-aspartate receptors in cerebellar neurons. Nature. 1990;345:347–50.PubMedCrossRef
63.
go back to reference Fan W, Huang F, Wu Z, Zhu X, Li D, He H. The role of nitric oxide in orofacial pain. Nitric Oxide. 2012;26:32–7.PubMedCrossRef Fan W, Huang F, Wu Z, Zhu X, Li D, He H. The role of nitric oxide in orofacial pain. Nitric Oxide. 2012;26:32–7.PubMedCrossRef
64.
go back to reference Orser BA, Pennefather PS, MacDonald JF. Multiple mechanisms of ketamine blockade of N-methyl-d-aspartate receptors. Anesthesiology. 1997;86:903–17.PubMedCrossRef Orser BA, Pennefather PS, MacDonald JF. Multiple mechanisms of ketamine blockade of N-methyl-d-aspartate receptors. Anesthesiology. 1997;86:903–17.PubMedCrossRef
65.
go back to reference Pelissier T, Laurido C, Kramer V, Hernández A, Paeile C. Antinociceptive interactions of ketamine with morphine or methadone in mononeuropathic rats. Eur J Pharmacol. 2003;477:23–8.PubMedCrossRef Pelissier T, Laurido C, Kramer V, Hernández A, Paeile C. Antinociceptive interactions of ketamine with morphine or methadone in mononeuropathic rats. Eur J Pharmacol. 2003;477:23–8.PubMedCrossRef
66.
go back to reference Petersen-Felix S, Arendt-Nielsen L, Bak P, Roth D, Fischer M, Bjerring P, et al. Analgesic effect in humans of subanaesthetic isoflurane concentrations evaluated by experimentally induced pain. Br J Anaesth. 1995;75:55–60.PubMedCrossRef Petersen-Felix S, Arendt-Nielsen L, Bak P, Roth D, Fischer M, Bjerring P, et al. Analgesic effect in humans of subanaesthetic isoflurane concentrations evaluated by experimentally induced pain. Br J Anaesth. 1995;75:55–60.PubMedCrossRef
67.
go back to reference Bennett GJ. Update on the neurophysiology of pain transmission and modulation: focus on the NMDA-receptor. J Pain Symptom Manage. 2000;19:S2–6.PubMedCrossRef Bennett GJ. Update on the neurophysiology of pain transmission and modulation: focus on the NMDA-receptor. J Pain Symptom Manage. 2000;19:S2–6.PubMedCrossRef
68.
go back to reference Eide PK. Wind-up and the NMDA receptor complex from a clinical perspective. Eur J Pain. 2000;4:5–15.PubMedCrossRef Eide PK. Wind-up and the NMDA receptor complex from a clinical perspective. Eur J Pain. 2000;4:5–15.PubMedCrossRef
69.
go back to reference Smith DJ, Bouchal RL, deSanctis CA, Monroe PJ, Amedro JB, Perrotti JM, et al. Properties of the interaction between ketamine and opiate binding sites in vivo and in vitro. Neuropharmacology. 1987;26:1253–60.PubMedCrossRef Smith DJ, Bouchal RL, deSanctis CA, Monroe PJ, Amedro JB, Perrotti JM, et al. Properties of the interaction between ketamine and opiate binding sites in vivo and in vitro. Neuropharmacology. 1987;26:1253–60.PubMedCrossRef
70.
go back to reference Finck AD, Samaniego E, Ngai SH. Morphine tolerance decreases the analgesic effects of ketamine in mice. Anesthesiology. 1988;68:397–400.PubMedCrossRef Finck AD, Samaniego E, Ngai SH. Morphine tolerance decreases the analgesic effects of ketamine in mice. Anesthesiology. 1988;68:397–400.PubMedCrossRef
71.
go back to reference Hustveit O, Maurset A, Oye I. Interaction of the chiral forms of ketamine with opioid, phencyclidine, sigma and muscarinic receptors. Pharmacol Toxicol. 1995;77:355–9.PubMedCrossRef Hustveit O, Maurset A, Oye I. Interaction of the chiral forms of ketamine with opioid, phencyclidine, sigma and muscarinic receptors. Pharmacol Toxicol. 1995;77:355–9.PubMedCrossRef
72.
go back to reference Mikkelsen S, Ilkjaer S, Brennum J, Borgbjerg FM, Dahl JB. The effect of naloxone on ketamine-induced effects on hyperalgesia and ketamine-induced side effects in humans. Anesthesiology. 1999;90:1539–45.PubMedCrossRef Mikkelsen S, Ilkjaer S, Brennum J, Borgbjerg FM, Dahl JB. The effect of naloxone on ketamine-induced effects on hyperalgesia and ketamine-induced side effects in humans. Anesthesiology. 1999;90:1539–45.PubMedCrossRef
73.
go back to reference Nishimura M, Sato K, Okada T, Yoshiya I, Schloss P, Shimada S, et al. Ketamine inhibits monoamine transporters expressed in human embryonic kidney 293 cells. Anesthesiology. 1998;88:768–74.PubMedCrossRef Nishimura M, Sato K, Okada T, Yoshiya I, Schloss P, Shimada S, et al. Ketamine inhibits monoamine transporters expressed in human embryonic kidney 293 cells. Anesthesiology. 1998;88:768–74.PubMedCrossRef
74.
go back to reference Kohrs R, Durieux ME. Ketamine: teaching an old drug new tricks. Anesth Analg. 1998;87:1186–93.PubMed Kohrs R, Durieux ME. Ketamine: teaching an old drug new tricks. Anesth Analg. 1998;87:1186–93.PubMed
75.
go back to reference Levänen J, Mäkelä ML, Scheinin H. Dexmedetomidine premedication attenuates ketamine-induced cardiostimulatory effects and postanesthetic delirium. Anesthesiology. 1995;82:1117–25.PubMedCrossRef Levänen J, Mäkelä ML, Scheinin H. Dexmedetomidine premedication attenuates ketamine-induced cardiostimulatory effects and postanesthetic delirium. Anesthesiology. 1995;82:1117–25.PubMedCrossRef
76.
go back to reference Salmi E, Långsjö JW, Aalto S, Någren K, Metsähonkala L, Kaisti KK, et al. Subanesthetic ketamine does not affect 11C-flumazenil binding in humans. Anesth. Analg. 2005;101:722–5 (table of contents).PubMedCrossRef Salmi E, Långsjö JW, Aalto S, Någren K, Metsähonkala L, Kaisti KK, et al. Subanesthetic ketamine does not affect 11C-flumazenil binding in humans. Anesth. Analg. 2005;101:722–5 (table of contents).PubMedCrossRef
77.
go back to reference Shafer SL, Siegel LC, Cooke JE, Scott JC. Testing computer-controlled infusion pumps by simulation. Anesthesiology. 1988;68:261–6.PubMedCrossRef Shafer SL, Siegel LC, Cooke JE, Scott JC. Testing computer-controlled infusion pumps by simulation. Anesthesiology. 1988;68:261–6.PubMedCrossRef
78.
go back to reference Flood P, Krasowski MD. Intravenous anesthetics differentially modulate ligand-gated ion channels. Anesthesiology. 2000;92:1418–25.PubMedCrossRef Flood P, Krasowski MD. Intravenous anesthetics differentially modulate ligand-gated ion channels. Anesthesiology. 2000;92:1418–25.PubMedCrossRef
79.
go back to reference Kornhuber J, Mack-Burkhardt F, Kornhuber ME, Riederer P. [3H]MK-801 binding sites in post-mortem human frontal cortex. Eur J Pharmacol. 1989;162:483–90.PubMedCrossRef Kornhuber J, Mack-Burkhardt F, Kornhuber ME, Riederer P. [3H]MK-801 binding sites in post-mortem human frontal cortex. Eur J Pharmacol. 1989;162:483–90.PubMedCrossRef
80.
go back to reference Kapur S, Seeman P. NMDA receptor antagonists ketamine and PCP have direct effects on the dopamine D(2) and serotonin 5-HT(2)receptors-implications for models of schizophrenia. Mol Psychiatry. 2002;7:837–44.PubMedCrossRef Kapur S, Seeman P. NMDA receptor antagonists ketamine and PCP have direct effects on the dopamine D(2) and serotonin 5-HT(2)receptors-implications for models of schizophrenia. Mol Psychiatry. 2002;7:837–44.PubMedCrossRef
81.
go back to reference Toro-Matos A, Rendon-Platas AM, Avila-Valdez E, Villarreal-Guzman RA. Physostigmine antagonizes ketamine. Anesth Analg. 1980;59:764–7.PubMedCrossRef Toro-Matos A, Rendon-Platas AM, Avila-Valdez E, Villarreal-Guzman RA. Physostigmine antagonizes ketamine. Anesth Analg. 1980;59:764–7.PubMedCrossRef
82.
go back to reference Hamilton-Davies C, Bailie R, Restall J. Physostigmine in recovery from anaesthesia. Anaesthesia. 1995;50:456–8.PubMedCrossRef Hamilton-Davies C, Bailie R, Restall J. Physostigmine in recovery from anaesthesia. Anaesthesia. 1995;50:456–8.PubMedCrossRef
83.
go back to reference Drummond JC, Brebner J, Galloon S, Young PS. A randomized evaluation of the reversal of ketamine by physostigmine. Can Anaesth Soc J. 1979;26:288–95.PubMedCrossRef Drummond JC, Brebner J, Galloon S, Young PS. A randomized evaluation of the reversal of ketamine by physostigmine. Can Anaesth Soc J. 1979;26:288–95.PubMedCrossRef
84.
go back to reference Haeseler G, Tetzlaff D, Bufler J, Dengler R, Münte S, Hecker H, et al. Blockade of voltage-operated neuronal and skeletal muscle sodium channels by S(+)- and R(−)-ketamine. Anesth Analg. 2003;96:1019–26 (table of contents).PubMedCrossRef Haeseler G, Tetzlaff D, Bufler J, Dengler R, Münte S, Hecker H, et al. Blockade of voltage-operated neuronal and skeletal muscle sodium channels by S(+)- and R(−)-ketamine. Anesth Analg. 2003;96:1019–26 (table of contents).PubMedCrossRef
85.
go back to reference Servin FS, Sear JW. Pharmacokinetics of intravenous anesthetics. In: Evers AS, Maze M, Kharasch ED, editors. Anesthetic pharmacology: basic principles and clinical practice. Cambridge: Cambridge University Press; 2011. Servin FS, Sear JW. Pharmacokinetics of intravenous anesthetics. In: Evers AS, Maze M, Kharasch ED, editors. Anesthetic pharmacology: basic principles and clinical practice. Cambridge: Cambridge University Press; 2011.
86.
go back to reference Bowdle TA, Radant AD, Cowley DS, Kharasch ED, Strassman RJ, Roy-Byrne PP. Psychedelic effects of ketamine in healthy volunteers: relationship to steady-state plasma concentrations. Anesthesiology. 1998;88:82–8. Bowdle TA, Radant AD, Cowley DS, Kharasch ED, Strassman RJ, Roy-Byrne PP. Psychedelic effects of ketamine in healthy volunteers: relationship to steady-state plasma concentrations. Anesthesiology. 1998;88:82–8.
87.
go back to reference Clements JA, Nimmo WS. Pharmacokinetics and analgesic effect of ketamine in man. Br J Anaesth. 1981;53:27–30.PubMedCrossRef Clements JA, Nimmo WS. Pharmacokinetics and analgesic effect of ketamine in man. Br J Anaesth. 1981;53:27–30.PubMedCrossRef
88.
go back to reference Leung A, Wallace MS, Ridgeway B, Yaksh T. Concentration–effect relationship of intravenous alfentanil and ketamine on peripheral neurosensory thresholds, allodynia and hyperalgesia of neuropathic pain. Pain. 2001;91:177–87.PubMedCrossRef Leung A, Wallace MS, Ridgeway B, Yaksh T. Concentration–effect relationship of intravenous alfentanil and ketamine on peripheral neurosensory thresholds, allodynia and hyperalgesia of neuropathic pain. Pain. 2001;91:177–87.PubMedCrossRef
89.
go back to reference Himmelseher S, Pfenninger E. The clinical use of S-(+)-ketamine–a determination of its place. Anasthesiol Intensivmed Notfallmed Schmerzther. 1998;33:764–70.PubMedCrossRef Himmelseher S, Pfenninger E. The clinical use of S-(+)-ketamine–a determination of its place. Anasthesiol Intensivmed Notfallmed Schmerzther. 1998;33:764–70.PubMedCrossRef
90.
go back to reference Mathisen LC, Skjelbred P, Skoglund LA, Oye I. Effect of ketamine, an NMDA receptor inhibitor, in acute and chronic orofacial pain. Pain. 1995;61:215–20.PubMedCrossRef Mathisen LC, Skjelbred P, Skoglund LA, Oye I. Effect of ketamine, an NMDA receptor inhibitor, in acute and chronic orofacial pain. Pain. 1995;61:215–20.PubMedCrossRef
91.
go back to reference Green SM, Krauss B. Ketamine is a safe, effective, and appropriate technique for emergency department paediatric procedural sedation. Emerg Med J. 2004;21:271–2.PubMedPubMedCentralCrossRef Green SM, Krauss B. Ketamine is a safe, effective, and appropriate technique for emergency department paediatric procedural sedation. Emerg Med J. 2004;21:271–2.PubMedPubMedCentralCrossRef
92.
go back to reference Freye E, Sundermann S, Wilder-Smith OH. No inhibition of gastro-intestinal propulsion after propofol- or propofol/ketamine-N2O/O2 anaesthesia. A comparison of gastro-caecal transit after isoflurane anaesthesia. Acta Anaesthesiol Scand. 1998;42:664–9.PubMedCrossRef Freye E, Sundermann S, Wilder-Smith OH. No inhibition of gastro-intestinal propulsion after propofol- or propofol/ketamine-N2O/O2 anaesthesia. A comparison of gastro-caecal transit after isoflurane anaesthesia. Acta Anaesthesiol Scand. 1998;42:664–9.PubMedCrossRef
93.
go back to reference Jennings PA, Cameron P, Bernard S, Walker T, Jolley D, Fitzgerald M, et al. Morphine and ketamine is superior to morphine alone for out-of-hospital trauma analgesia: a randomized controlled trial. Ann Emerg Med. 2012;59:497–503.PubMedCrossRef Jennings PA, Cameron P, Bernard S, Walker T, Jolley D, Fitzgerald M, et al. Morphine and ketamine is superior to morphine alone for out-of-hospital trauma analgesia: a randomized controlled trial. Ann Emerg Med. 2012;59:497–503.PubMedCrossRef
94.
go back to reference Ahern TL, Herring AA, Anderson ES, Madia VA, Fahimi J, Frazee BW. The first 500: initial experience with widespread use of low-dose ketamine for acute pain management in the ED. Am J Emerg Med. 2015;33:197–201.PubMedCrossRef Ahern TL, Herring AA, Anderson ES, Madia VA, Fahimi J, Frazee BW. The first 500: initial experience with widespread use of low-dose ketamine for acute pain management in the ED. Am J Emerg Med. 2015;33:197–201.PubMedCrossRef
95.
go back to reference Beaudoin FL, Lin C, Guan W, Merchant RC. Low-dose ketamine improves pain relief in patients receiving intravenous opioids for acute pain in the emergency department: results of a randomized, double-blind, clinical trial. Acad Emerg Med. 2014;21:1193–202.PubMedCrossRef Beaudoin FL, Lin C, Guan W, Merchant RC. Low-dose ketamine improves pain relief in patients receiving intravenous opioids for acute pain in the emergency department: results of a randomized, double-blind, clinical trial. Acad Emerg Med. 2014;21:1193–202.PubMedCrossRef
96.
go back to reference Richebé P, Julien M, Brulotte V. Potential strategies for preventing chronic postoperative pain: a practical approach: continuing professional development. Anaesth: Can J; 2015. Richebé P, Julien M, Brulotte V. Potential strategies for preventing chronic postoperative pain: a practical approach: continuing professional development. Anaesth: Can J; 2015.
97.
go back to reference Schmid RL, Sandler AN, Katz J. Use and efficacy of low-dose ketamine in the management of acute postoperative pain: a review of current techniques and outcomes. Pain. 1999;82:111–25.PubMedCrossRef Schmid RL, Sandler AN, Katz J. Use and efficacy of low-dose ketamine in the management of acute postoperative pain: a review of current techniques and outcomes. Pain. 1999;82:111–25.PubMedCrossRef
98.
go back to reference Jouguelet-Lacoste J, La Colla L, Schilling D, Chelly JE. The use of intravenous infusion or single dose of low-dose ketamine for postoperative analgesia: a review of the current literature. Pain Med. 2015;16:383–403.PubMedCrossRef Jouguelet-Lacoste J, La Colla L, Schilling D, Chelly JE. The use of intravenous infusion or single dose of low-dose ketamine for postoperative analgesia: a review of the current literature. Pain Med. 2015;16:383–403.PubMedCrossRef
99.
go back to reference Elia N, Tramèr MR. Ketamine and postoperative pain—a quantitative systematic review of randomised trials. Pain. 2005;113:61–70.PubMedCrossRef Elia N, Tramèr MR. Ketamine and postoperative pain—a quantitative systematic review of randomised trials. Pain. 2005;113:61–70.PubMedCrossRef
100.
go back to reference Weinbroum AA. Non-opioid IV adjuvants in the perioperative period: pharmacological and clinical aspects of ketamine and gabapentinoids. Pharmacol Res. 2012;65:411–29.PubMedCrossRef Weinbroum AA. Non-opioid IV adjuvants in the perioperative period: pharmacological and clinical aspects of ketamine and gabapentinoids. Pharmacol Res. 2012;65:411–29.PubMedCrossRef
101.
go back to reference Ilkjaer S, Nikolajsen L, Hansen TM, Wernberg M, Brennum J, Dahl JB. Effect of i.v. ketamine in combination with epidural bupivacaine or epidural morphine on postoperative pain and wound tenderness after renal surgery. Br J Anaesth. 1998;81:707–12.PubMedCrossRef Ilkjaer S, Nikolajsen L, Hansen TM, Wernberg M, Brennum J, Dahl JB. Effect of i.v. ketamine in combination with epidural bupivacaine or epidural morphine on postoperative pain and wound tenderness after renal surgery. Br J Anaesth. 1998;81:707–12.PubMedCrossRef
102.
go back to reference Mathisen LC, Aasbø V, Raeder J. Lack of pre-emptive analgesic effect of (R)-ketamine in laparoscopic cholecystectomy. Acta Anaesthesiol Scand. 1999;43:220–4.PubMedCrossRef Mathisen LC, Aasbø V, Raeder J. Lack of pre-emptive analgesic effect of (R)-ketamine in laparoscopic cholecystectomy. Acta Anaesthesiol Scand. 1999;43:220–4.PubMedCrossRef
103.
go back to reference Bell RF, Dahl JB, Moore RA, Kalso E. Perioperative ketamine for acute postoperative pain. Cochrane Database Syst Rev. 2006;CD004603. Bell RF, Dahl JB, Moore RA, Kalso E. Perioperative ketamine for acute postoperative pain. Cochrane Database Syst Rev. 2006;CD004603.
104.
go back to reference Hans P, Dewandre P-Y, Brichant JF, Bonhomme V. Comparative effects of ketamine on bispectral index and spectral entropy of the electroencephalogram under sevoflurane anaesthesia. Br J Anaesth. 2005;94:336–40.PubMedCrossRef Hans P, Dewandre P-Y, Brichant JF, Bonhomme V. Comparative effects of ketamine on bispectral index and spectral entropy of the electroencephalogram under sevoflurane anaesthesia. Br J Anaesth. 2005;94:336–40.PubMedCrossRef
105.
go back to reference Neuhäuser C, Preiss V, Feurer M-K, Müller M, Scholz S, Kwapisz M, et al. Comparison of S-(+)-ketamine- with sufentanil-based anaesthesia for elective coronary artery bypass graft surgery: effect on troponin T levels. Br J Anaesth. 2008;100:765–71.PubMedCrossRef Neuhäuser C, Preiss V, Feurer M-K, Müller M, Scholz S, Kwapisz M, et al. Comparison of S-(+)-ketamine- with sufentanil-based anaesthesia for elective coronary artery bypass graft surgery: effect on troponin T levels. Br J Anaesth. 2008;100:765–71.PubMedCrossRef
106.
go back to reference Sprung J, Schuetz SM, Stewart RW, Moravec CS. Effects of ketamine on the contractility of failing and nonfailing human heart muscles in vitro. Anesthesiology. 1998;88:1202–10.PubMedCrossRef Sprung J, Schuetz SM, Stewart RW, Moravec CS. Effects of ketamine on the contractility of failing and nonfailing human heart muscles in vitro. Anesthesiology. 1998;88:1202–10.PubMedCrossRef
107.
go back to reference Lahtinen P, Kokki H, Hakala T, Hynynen M. S(+)-ketamine as an analgesic adjunct reduces opioid consumption after cardiac surgery. Anesth Analg. 2004;99:1295–301 (table of contents).PubMedCrossRef Lahtinen P, Kokki H, Hakala T, Hynynen M. S(+)-ketamine as an analgesic adjunct reduces opioid consumption after cardiac surgery. Anesth Analg. 2004;99:1295–301 (table of contents).PubMedCrossRef
108.
go back to reference Hillis LD, Smith PK, Anderson JL, Bittl JA, Bridges CR, Byrne JG, et al. Special articles: 2011 ACCF/AHA guideline for coronary artery bypass graft surgery: executive summary: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Anesth Analg. 2012;114:11–45.PubMedCrossRef Hillis LD, Smith PK, Anderson JL, Bittl JA, Bridges CR, Byrne JG, et al. Special articles: 2011 ACCF/AHA guideline for coronary artery bypass graft surgery: executive summary: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Anesth Analg. 2012;114:11–45.PubMedCrossRef
109.
go back to reference Kawasaki T, Ogata M, Kawasaki C, Ogata J, Inoue Y, Shigematsu A. Ketamine suppresses proinflammatory cytokine production in human whole blood in vitro. Anesth Analg. 1999;89:665–9.PubMed Kawasaki T, Ogata M, Kawasaki C, Ogata J, Inoue Y, Shigematsu A. Ketamine suppresses proinflammatory cytokine production in human whole blood in vitro. Anesth Analg. 1999;89:665–9.PubMed
110.
go back to reference Szekely A, Heindl B, Zahler S, Conzen PF, Becker BF. S(+)-ketamine, but not R(−)-ketamine, reduces postischemic adherence of neutrophils in the coronary system of isolated guinea pig hearts. Anesth Analg. 1999;88:1017–24.PubMedCrossRef Szekely A, Heindl B, Zahler S, Conzen PF, Becker BF. S(+)-ketamine, but not R(−)-ketamine, reduces postischemic adherence of neutrophils in the coronary system of isolated guinea pig hearts. Anesth Analg. 1999;88:1017–24.PubMedCrossRef
111.
go back to reference Zilberstein G, Levy R, Rachinsky M, Fisher A, Greemberg L, Shapira Y, et al. Ketamine attenuates neutrophil activation after cardiopulmonary bypass. Anesth Analg. 2002;95:531–6 (table of contents).PubMed Zilberstein G, Levy R, Rachinsky M, Fisher A, Greemberg L, Shapira Y, et al. Ketamine attenuates neutrophil activation after cardiopulmonary bypass. Anesth Analg. 2002;95:531–6 (table of contents).PubMed
112.
go back to reference Roytblat L, Talmor D, Rachinsky M, Greemberg L, Pekar A, Appelbaum A, et al. Ketamine attenuates the interleukin-6 response after cardiopulmonary bypass. Anesth Analg. 1998;87:266–71.PubMed Roytblat L, Talmor D, Rachinsky M, Greemberg L, Pekar A, Appelbaum A, et al. Ketamine attenuates the interleukin-6 response after cardiopulmonary bypass. Anesth Analg. 1998;87:266–71.PubMed
113.
go back to reference Olney JW, Labruyere J, Wang G, Wozniak DF, Price MT, Sesma MA. NMDA antagonist neurotoxicity: mechanism and prevention. Science. 1991;254:1515–8.PubMedCrossRef Olney JW, Labruyere J, Wang G, Wozniak DF, Price MT, Sesma MA. NMDA antagonist neurotoxicity: mechanism and prevention. Science. 1991;254:1515–8.PubMedCrossRef
114.
go back to reference Hayashi H, Dikkes P, Soriano SG. Repeated administration of ketamine may lead to neuronal degeneration in the developing rat brain. Paediatr Anaesth. 2002;12:770–4.PubMedCrossRef Hayashi H, Dikkes P, Soriano SG. Repeated administration of ketamine may lead to neuronal degeneration in the developing rat brain. Paediatr Anaesth. 2002;12:770–4.PubMedCrossRef
115.
go back to reference Jevtovic-Todorovic V, Benshoff N, Olney JW. Ketamine potentiates cerebrocortical damage induced by the common anaesthetic agent nitrous oxide in adult rats. Br J Pharmacol. 2000;130:1692–8.PubMedPubMedCentralCrossRef Jevtovic-Todorovic V, Benshoff N, Olney JW. Ketamine potentiates cerebrocortical damage induced by the common anaesthetic agent nitrous oxide in adult rats. Br J Pharmacol. 2000;130:1692–8.PubMedPubMedCentralCrossRef
116.
go back to reference Yan J, Jiang H. Dual effects of ketamine: neurotoxicity versus neuroprotection in anesthesia for the developing brain. J Neurosurg Anesthesiol. 2014;26:155–60.PubMedCrossRef Yan J, Jiang H. Dual effects of ketamine: neurotoxicity versus neuroprotection in anesthesia for the developing brain. J Neurosurg Anesthesiol. 2014;26:155–60.PubMedCrossRef
117.
go back to reference Hodgson PS, Neal JM, Pollock JE, Liu SS. The neurotoxicity of drugs given intrathecally (spinal). Anesth Analg. 1999;88:797–809.PubMedCrossRef Hodgson PS, Neal JM, Pollock JE, Liu SS. The neurotoxicity of drugs given intrathecally (spinal). Anesth Analg. 1999;88:797–809.PubMedCrossRef
118.
go back to reference Bai X, Yan Y, Canfield S, Muravyeva MY, Kikuchi C, Zaja I, et al. Ketamine enhances human neural stem cell proliferation and induces neuronal apoptosis via reactive oxygen species-mediated mitochondrial pathway. Anesth Analg. 2013;116:869–80.PubMedPubMedCentralCrossRef Bai X, Yan Y, Canfield S, Muravyeva MY, Kikuchi C, Zaja I, et al. Ketamine enhances human neural stem cell proliferation and induces neuronal apoptosis via reactive oxygen species-mediated mitochondrial pathway. Anesth Analg. 2013;116:869–80.PubMedPubMedCentralCrossRef
119.
go back to reference Yan J, Li Y, Zhang Y, Lu Y, Jiang H. Repeated exposure to anesthetic ketamine can negatively impact neurodevelopment in infants: a prospective preliminary clinical study. J Child Neurol. 2014;29:1333–8.PubMedCrossRef Yan J, Li Y, Zhang Y, Lu Y, Jiang H. Repeated exposure to anesthetic ketamine can negatively impact neurodevelopment in infants: a prospective preliminary clinical study. J Child Neurol. 2014;29:1333–8.PubMedCrossRef
120.
go back to reference Koerner IP, Brambrink AM. Brain protection by anesthetic agents. Curr Opin Anaesthesiol. 2006;19:481–6.PubMedCrossRef Koerner IP, Brambrink AM. Brain protection by anesthetic agents. Curr Opin Anaesthesiol. 2006;19:481–6.PubMedCrossRef
121.
go back to reference Sanders RD, Hassell J, Davidson AJ, Robertson NJ, Ma D. Impact of anaesthetics and surgery on neurodevelopment: an update. Br J Anaesth. 2013;110:i53–72.PubMedPubMedCentralCrossRef Sanders RD, Hassell J, Davidson AJ, Robertson NJ, Ma D. Impact of anaesthetics and surgery on neurodevelopment: an update. Br J Anaesth. 2013;110:i53–72.PubMedPubMedCentralCrossRef
122.
go back to reference Proescholdt M, Heimann A, Kempski O. Neuroprotection of S(+) ketamine isomer in global forebrain ischemia. Brain Res. 2001;904:245–51.PubMedCrossRef Proescholdt M, Heimann A, Kempski O. Neuroprotection of S(+) ketamine isomer in global forebrain ischemia. Brain Res. 2001;904:245–51.PubMedCrossRef
123.
go back to reference Hudetz JA, Pagel PS. Neuroprotection by ketamine: a review of the experimental and clinical evidence. J Cardiothorac Vasc Anesth. 2010;24:131–42.PubMedCrossRef Hudetz JA, Pagel PS. Neuroprotection by ketamine: a review of the experimental and clinical evidence. J Cardiothorac Vasc Anesth. 2010;24:131–42.PubMedCrossRef
124.
go back to reference Långsjö JW, Maksimow A, Salmi E, Kaisti K, Aalto S, Oikonen V, et al. S-ketamine anesthesia increases cerebral blood flow in excess of the metabolic needs in humans. Anesthesiology. 2005;103:258–68.PubMedCrossRef Långsjö JW, Maksimow A, Salmi E, Kaisti K, Aalto S, Oikonen V, et al. S-ketamine anesthesia increases cerebral blood flow in excess of the metabolic needs in humans. Anesthesiology. 2005;103:258–68.PubMedCrossRef
125.
go back to reference Långsjö JW, Kaisti KK, Aalto S, Hinkka S, Aantaa R, Oikonen V, et al. Effects of subanesthetic doses of ketamine on regional cerebral blood flow, oxygen consumption, and blood volume in humans. Anesthesiology. 2003;99:614–23.PubMedCrossRef Långsjö JW, Kaisti KK, Aalto S, Hinkka S, Aantaa R, Oikonen V, et al. Effects of subanesthetic doses of ketamine on regional cerebral blood flow, oxygen consumption, and blood volume in humans. Anesthesiology. 2003;99:614–23.PubMedCrossRef
126.
go back to reference Långsjö JW, Salmi E, Kaisti KK, Aalto S, Hinkka S, Aantaa R, et al. Effects of subanesthetic ketamine on regional cerebral glucose metabolism in humans. Anesthesiology. 2004;100:1065–71.PubMedCrossRef Långsjö JW, Salmi E, Kaisti KK, Aalto S, Hinkka S, Aantaa R, et al. Effects of subanesthetic ketamine on regional cerebral glucose metabolism in humans. Anesthesiology. 2004;100:1065–71.PubMedCrossRef
127.
go back to reference Himmelseher S, Durieux ME. Revising a dogma: ketamine for patients with neurological injury? Anesth. Analg. 2005;101:524–34 (table of contents).PubMedCrossRef Himmelseher S, Durieux ME. Revising a dogma: ketamine for patients with neurological injury? Anesth. Analg. 2005;101:524–34 (table of contents).PubMedCrossRef
128.
go back to reference Zeiler FA, Teitelbaum J, West M, Gillman LM. The ketamine effect on ICP in traumatic brain injury. Neurocrit Care. 2014;21:163–73.PubMedCrossRef Zeiler FA, Teitelbaum J, West M, Gillman LM. The ketamine effect on ICP in traumatic brain injury. Neurocrit Care. 2014;21:163–73.PubMedCrossRef
129.
go back to reference Kehlet H, Jensen TS, Woolf CJ. Persistent postsurgical pain: risk factors and prevention. Lancet (London, England). 2006;367:1618–25.CrossRef Kehlet H, Jensen TS, Woolf CJ. Persistent postsurgical pain: risk factors and prevention. Lancet (London, England). 2006;367:1618–25.CrossRef
130.
go back to reference McNicol ED, Schumann R, Haroutounian S. A systematic review and meta-analysis of ketamine for the prevention of persistent post-surgical pain. Acta Anaesthesiol Scand. 2014;58:1199–213.PubMedCrossRef McNicol ED, Schumann R, Haroutounian S. A systematic review and meta-analysis of ketamine for the prevention of persistent post-surgical pain. Acta Anaesthesiol Scand. 2014;58:1199–213.PubMedCrossRef
131.
go back to reference Humble SR, Dalton AJ, Li L. A systematic review of therapeutic interventions to reduce acute and chronic post-surgical pain after amputation, thoracotomy or mastectomy. Eur J Pain. 2015;19:451–65.PubMedCrossRef Humble SR, Dalton AJ, Li L. A systematic review of therapeutic interventions to reduce acute and chronic post-surgical pain after amputation, thoracotomy or mastectomy. Eur J Pain. 2015;19:451–65.PubMedCrossRef
132.
go back to reference Tena B, Gomar C, Rios J. Perioperative epidural or intravenous ketamine does not improve the effectiveness of thoracic epidural analgesia for acute and chronic pain after thoracotomy. Clin J Pain. 2014;30:490–500.PubMedCrossRef Tena B, Gomar C, Rios J. Perioperative epidural or intravenous ketamine does not improve the effectiveness of thoracic epidural analgesia for acute and chronic pain after thoracotomy. Clin J Pain. 2014;30:490–500.PubMedCrossRef
133.
go back to reference Hu J, Liao Q, Zhang F, Tong J, Ouyang W. Chronic postthoracotomy pain and perioperative ketamine infusion. J Pain Palliat Care Pharmacother. 2014;28:117–21.PubMedCrossRef Hu J, Liao Q, Zhang F, Tong J, Ouyang W. Chronic postthoracotomy pain and perioperative ketamine infusion. J Pain Palliat Care Pharmacother. 2014;28:117–21.PubMedCrossRef
136.
go back to reference Niesters M, Aarts L, Sarton E, Dahan A. Influence of ketamine and morphine on descending pain modulation in chronic pain patients: a randomized placebo-controlled cross-over proof-of-concept study. Br J Anaesth. 2013;110:1010–6.PubMedCrossRef Niesters M, Aarts L, Sarton E, Dahan A. Influence of ketamine and morphine on descending pain modulation in chronic pain patients: a randomized placebo-controlled cross-over proof-of-concept study. Br J Anaesth. 2013;110:1010–6.PubMedCrossRef
137.
go back to reference Niesters M, Dahan A, Swartjes M, Noppers I, Fillingim RB, Aarts L, et al. Effect of ketamine on endogenous pain modulation in healthy volunteers. Pain. 2011;152:656–63.PubMedCrossRef Niesters M, Dahan A, Swartjes M, Noppers I, Fillingim RB, Aarts L, et al. Effect of ketamine on endogenous pain modulation in healthy volunteers. Pain. 2011;152:656–63.PubMedCrossRef
138.
go back to reference Schwartzman RJ, Alexander GM, Grothusen JR, Paylor T, Reichenberger E, Perreault M. Outpatient intravenous ketamine for the treatment of complex regional pain syndrome: a double-blind placebo controlled study. Pain. 2009;147:107–15.PubMedCrossRef Schwartzman RJ, Alexander GM, Grothusen JR, Paylor T, Reichenberger E, Perreault M. Outpatient intravenous ketamine for the treatment of complex regional pain syndrome: a double-blind placebo controlled study. Pain. 2009;147:107–15.PubMedCrossRef
139.
go back to reference Finch PM, Knudsen L, Drummond PD. Reduction of allodynia in patients with complex regional pain syndrome: a double-blind placebo-controlled trial of topical ketamine. Pain 2009;146:18–25PubMedCrossRef Finch PM, Knudsen L, Drummond PD. Reduction of allodynia in patients with complex regional pain syndrome: a double-blind placebo-controlled trial of topical ketamine. Pain 2009;146:18–25PubMedCrossRef
140.
go back to reference Gewandter JS, Mohile SG, Heckler CE, Ryan JL, Kirshner JJ, Flynn PJ, et al. A phase III randomized, placebo-controlled study of topical amitriptyline and ketamine for chemotherapy-induced peripheral neuropathy (CIPN): a University of Rochester CCOP study of 462 cancer survivors. Support Care Cancer. 2014;22:1807–14.PubMedPubMedCentralCrossRef Gewandter JS, Mohile SG, Heckler CE, Ryan JL, Kirshner JJ, Flynn PJ, et al. A phase III randomized, placebo-controlled study of topical amitriptyline and ketamine for chemotherapy-induced peripheral neuropathy (CIPN): a University of Rochester CCOP study of 462 cancer survivors. Support Care Cancer. 2014;22:1807–14.PubMedPubMedCentralCrossRef
141.
go back to reference Bell RF. Ketamine for chronic noncancer pain: concerns regarding toxicity. Curr Opin Support Palliat Care. 2012;6:183–7.PubMedCrossRef Bell RF. Ketamine for chronic noncancer pain: concerns regarding toxicity. Curr Opin Support Palliat Care. 2012;6:183–7.PubMedCrossRef
142.
go back to reference Hardy J, Quinn S, Fazekas B, Plummer J, Eckermann S, Agar M, et al. Randomized, double-blind, placebo-controlled study to assess the efficacy and toxicity of subcutaneous ketamine in the management of cancer pain. J Clin Oncol. 2012;30:3611–7.PubMedCrossRef Hardy J, Quinn S, Fazekas B, Plummer J, Eckermann S, Agar M, et al. Randomized, double-blind, placebo-controlled study to assess the efficacy and toxicity of subcutaneous ketamine in the management of cancer pain. J Clin Oncol. 2012;30:3611–7.PubMedCrossRef
143.
go back to reference Rivosecchi RM, Rice MJ, Smithburger PL, Buckley MS, Coons JC, Kane-Gill SL. An evidence based systematic review of remifentanil associated opioid-induced hyperalgesia. Expert Opin Drug Saf. 2014;13:587–603.PubMedCrossRef Rivosecchi RM, Rice MJ, Smithburger PL, Buckley MS, Coons JC, Kane-Gill SL. An evidence based systematic review of remifentanil associated opioid-induced hyperalgesia. Expert Opin Drug Saf. 2014;13:587–603.PubMedCrossRef
144.
145.
go back to reference Segmiller F, Rüther T, Linhardt A, Padberg F, Berger M, Pogarell O, et al. Repeated S-ketamine infusions in therapy resistant depression: a case series. J Clin Pharmacol. 2013;53:996–8.PubMedCrossRef Segmiller F, Rüther T, Linhardt A, Padberg F, Berger M, Pogarell O, et al. Repeated S-ketamine infusions in therapy resistant depression: a case series. J Clin Pharmacol. 2013;53:996–8.PubMedCrossRef
146.
go back to reference Liu R-J, Fuchikami M, Dwyer JM, Lepack AE, Duman RS, Aghajanian GK. GSK-3 inhibition potentiates the synaptogenic and antidepressant-like effects of subthreshold doses of ketamine. Neuropsychopharmacology. 2013;38:2268–77.PubMedPubMedCentralCrossRef Liu R-J, Fuchikami M, Dwyer JM, Lepack AE, Duman RS, Aghajanian GK. GSK-3 inhibition potentiates the synaptogenic and antidepressant-like effects of subthreshold doses of ketamine. Neuropsychopharmacology. 2013;38:2268–77.PubMedPubMedCentralCrossRef
147.
go back to reference Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47:351–4.PubMedCrossRef Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000;47:351–4.PubMedCrossRef
148.
go back to reference aan het Rot M, Collins K, Murrough JW, Perez AM, Reich DL, Charney DS, et al. Safety and efficacy of repeated-dose intravenous ketamine for treatment-resistant depression. Biol Psychiatry. 2010;67:139–45.PubMedCrossRef aan het Rot M, Collins K, Murrough JW, Perez AM, Reich DL, Charney DS, et al. Safety and efficacy of repeated-dose intravenous ketamine for treatment-resistant depression. Biol Psychiatry. 2010;67:139–45.PubMedCrossRef
149.
go back to reference Zhang J-C, Li S-X, Hashimoto K. R(−)-ketamine shows greater potency and longer lasting antidepressant effects than S(+)-ketamine. Pharmacol Biochem Behav. 2014;116:137–41.PubMedCrossRef Zhang J-C, Li S-X, Hashimoto K. R(−)-ketamine shows greater potency and longer lasting antidepressant effects than S(+)-ketamine. Pharmacol Biochem Behav. 2014;116:137–41.PubMedCrossRef
150.
go back to reference Kranaster L, Kammerer-Ciernioch J, Hoyer C, Sartorius A. Clinically favourable effects of ketamine as an anaesthetic for electroconvulsive therapy: a retrospective study. Eur Arch Psychiatry Clin Neurosci. 2011;261:575–82.PubMedCrossRef Kranaster L, Kammerer-Ciernioch J, Hoyer C, Sartorius A. Clinically favourable effects of ketamine as an anaesthetic for electroconvulsive therapy: a retrospective study. Eur Arch Psychiatry Clin Neurosci. 2011;261:575–82.PubMedCrossRef
151.
go back to reference Rolan P, Lim S, Sunderland V, Liu Y, Molnar V. The absolute bioavailability of racemic ketamine from a novel sublingual formulation. Br J Clin Pharmacol. 2014;77:1011–6.PubMedCrossRef Rolan P, Lim S, Sunderland V, Liu Y, Molnar V. The absolute bioavailability of racemic ketamine from a novel sublingual formulation. Br J Clin Pharmacol. 2014;77:1011–6.PubMedCrossRef
152.
go back to reference Fitzgibbon D, Morgan D, Dockter D, Barry C, Kharasch ED. Initial pharmacokinetic, safety and efficacy evaluation of nasal morphine gluconate for breakthrough pain in cancer patients. Pain. 2003;106:309–15.PubMedCrossRef Fitzgibbon D, Morgan D, Dockter D, Barry C, Kharasch ED. Initial pharmacokinetic, safety and efficacy evaluation of nasal morphine gluconate for breakthrough pain in cancer patients. Pain. 2003;106:309–15.PubMedCrossRef
153.
go back to reference Carr DB, Goudas LC, Denman WT, Brookoff D, Staats PS, Brennen L, et al. Safety and efficacy of intranasal ketamine for the treatment of breakthrough pain in patients with chronic pain: a randomized, double-blind, placebo-controlled, crossover study. Pain. 2004;108:17–27.PubMedCrossRef Carr DB, Goudas LC, Denman WT, Brookoff D, Staats PS, Brennen L, et al. Safety and efficacy of intranasal ketamine for the treatment of breakthrough pain in patients with chronic pain: a randomized, double-blind, placebo-controlled, crossover study. Pain. 2004;108:17–27.PubMedCrossRef
154.
go back to reference Malinovsky JM, Servin F, Cozian A, Lepage JY, Pinaud M. Ketamine and norketamine plasma concentrations after i.v., nasal and rectal administration in children. Br J Anaesth. 1996;77:203–7.PubMedCrossRef Malinovsky JM, Servin F, Cozian A, Lepage JY, Pinaud M. Ketamine and norketamine plasma concentrations after i.v., nasal and rectal administration in children. Br J Anaesth. 1996;77:203–7.PubMedCrossRef
155.
go back to reference Winstock AR, Mitcheson L, Gillatt DA, Cottrell AM. The prevalence and natural history of urinary symptoms among recreational ketamine users. BJU Int. 2012;110:1762–6.PubMedCrossRef Winstock AR, Mitcheson L, Gillatt DA, Cottrell AM. The prevalence and natural history of urinary symptoms among recreational ketamine users. BJU Int. 2012;110:1762–6.PubMedCrossRef
156.
go back to reference Jhang J-F, Hsu Y-H, Kuo H-C. Possible pathophysiology of ketamine-related cystitis and associated treatment strategies. Int J Urol. 2015;22:816–25.PubMedCrossRef Jhang J-F, Hsu Y-H, Kuo H-C. Possible pathophysiology of ketamine-related cystitis and associated treatment strategies. Int J Urol. 2015;22:816–25.PubMedCrossRef
157.
go back to reference Liao Y, Tang J, Ma M, Wu Z, Yang M, Wang X, et al. Frontal white matter abnormalities following chronic ketamine use: a diffusion tensor imaging study. Brain. 2010;133:2115–22.PubMedCrossRef Liao Y, Tang J, Ma M, Wu Z, Yang M, Wang X, et al. Frontal white matter abnormalities following chronic ketamine use: a diffusion tensor imaging study. Brain. 2010;133:2115–22.PubMedCrossRef
158.
159.
go back to reference Brown L, Christian-Kopp S, Sherwin TS, Khan A, Barcega B, Denmark TK, et al. Adjunctive atropine is unnecessary during ketamine sedation in children. Acad Emerg Med. 2008;15:314–8.PubMedCrossRef Brown L, Christian-Kopp S, Sherwin TS, Khan A, Barcega B, Denmark TK, et al. Adjunctive atropine is unnecessary during ketamine sedation in children. Acad Emerg Med. 2008;15:314–8.PubMedCrossRef
Metadata
Title
Ketamine: A Review of Clinical Pharmacokinetics and Pharmacodynamics in Anesthesia and Pain Therapy
Authors
Marko A. Peltoniemi
Nora M. Hagelberg
Klaus T. Olkkola
Teijo I. Saari
Publication date
01-09-2016
Publisher
Springer International Publishing
Published in
Clinical Pharmacokinetics / Issue 9/2016
Print ISSN: 0312-5963
Electronic ISSN: 1179-1926
DOI
https://doi.org/10.1007/s40262-016-0383-6

Other articles of this Issue 9/2016

Clinical Pharmacokinetics 9/2016 Go to the issue