Ketamine: 50 Years of Modulating the Mind

Linda Li, Phillip E Vlisides, Linda Li, Phillip E Vlisides

Abstract

Ketamine was introduced into clinical practice in the 1960s and continues to be both clinically useful and scientifically fascinating. With considerably diverse molecular targets and neurophysiological properties, ketamine's effects on the central nervous system remain incompletely understood. Investigators have leveraged the unique characteristics of ketamine to explore the invariant, fundamental mechanisms of anesthetic action. Emerging evidence indicates that ketamine-mediated anesthesia may occur via disruption of corticocortical information transfer in a frontal-to-parietal ("top down") distribution. This proposed mechanism of general anesthesia has since been demonstrated with anesthetics in other pharmacological classes as well. Ketamine remains invaluable to the fields of anesthesiology and critical care medicine, in large part due to its ability to maintain cardiorespiratory stability while providing effective sedation and analgesia. Furthermore, there may be an emerging role for ketamine in treatment of refractory depression and Post-Traumatic Stress Disorder. In this article, we review the history of ketamine, its pharmacology, putative mechanisms of action and current clinical applications.

Keywords: anesthesia; consciousness; depression; functional connectivity; ketamine; neuropharmacology; post-traumatic stress disorder.

Figures

Figure 1
Figure 1
Dr. Edward Domino as a young faculty at the University of Michigan. Dr. Domino is now in his 90s, an Emeritus Professor, and still active as a scientist in the field of neuropharmacology. Photograph provided courtesy of the University of Michigan Bentley Historical Library.
Figure 2
Figure 2
Structure of Ketamine.
Figure 3
Figure 3
Measures of directed connectivity after induction with Ketamine, adapted from Blain-Moraes et al. (2014). Graphical depiction of dominant feedback connectivity in the waking state that is neutralized after ketamine induction. Please see original article (Blain-Moraes et al., 2014) for additional information.

References

    1. Adam F., Chauvin M., Du Manoir B., Langlois M., Sessler D. I., Fletcher D. (2005). Small-dose ketamine infusion improves postoperative analgesia and rehabilitation after total knee arthroplasty. Anesth. Analg. 100, 475–480.
    1. Afridi S. K., Giffin N. J., Kaube H., Goadsby P. J. (2013). A randomized controlled trial of intranasal ketamine in migraine with prolonged aura. Neurology 80, 642–647.
    1. Ahern T. L., Herring A. A., Miller S., Frazee B. W. (2015). Low-Dose ketamine infusion for emergency department patients with severe pain. Pain Med. 16, 1402–1409.
    1. Akata T., Izumi K., Nakashima M. (2001). Mechanisms of direct inhibitory action of ketamine on vascular smooth muscle in mesenteric resistance arteries. Anesthesiology 95, 452–462.
    1. Akhavanakbari G., Mohamadian A., Entezariasl M. (2014). Evaluation the effects of adding ketamine to morphine in intravenous patient-controlled analgesia after orthopedic surgery. Perspect. Clin. Res. 5, 85–87.
    1. Albanèse J., Arnaud S., Rey M., Thomachot L., Alliez B., Martin C. (1997). Ketamine decreases intracranial pressure and electroencephalographic activity in traumatic brain injury patients during propofol sedation. Anesthesiology 87, 1328–1334.
    1. Aliman A. C., Piccioni M. A., Piccioni J. L., Oliva J. L., Auler Júnior J. O. (2011). Intraosseous anesthesia in hemodynamic studies in children with cardiopathy. Rev. Bras. Anestesiol. 61, 41–49.
    1. Allen J. Y., Macias C. G. (2005). The efficacy of ketamine in pediatric emergency department patients who present with acute severe asthma. Ann. Emerg. Med. 46, 43–50.
    1. Amin P., Roeland E., Atayee R. (2014). Case report: efficacy and tolerability of ketamine in opioid-refractory cancer pain. J. Pain Palliat. Care Pharmacother. 28, 233–242.
    1. Andolfatto G., Willman E. (2010). A prospective case series of pediatric procedural sedation and analgesia in the emergency department using single-syringe ketamine-propofol combination (ketofol). Acad. Emerg. Med. 17, 194–201.
    1. Andolfatto G., Willman E., Joo D., Miller P., Wong W. B., Koehn M., et al. . (2013). Intranasal ketamine for analgesia in the emergency department: a prospective observational series. Acad. Emerg. Med. 20, 1050–1054.
    1. Anis N. A., Berry S. C., Burton N. R., Lodge D. (1983). The dissociative anaesthetics, ketamine and phencyclidine, selectively reduce excitation of central mammalian neurones by N-methyl-aspartate. Br. J. Pharmacol. 79, 565–575.
    1. Ashkenazi I., Isakovich B., Kluger Y., Alfici R., Kessel B., Better O. S. (2005). Prehospital management of earthquake casualties buried under rubble. Prehosp. Disaster Med. 20, 122–133.
    1. Bar-Joseph G., Guilburd Y., Tamir A., Guilburd J. N. (2009). Effectiveness of ketamine in decreasing intracranial pressure in children with intracranial hypertension. J. Neurosurg. Pediatr. 4, 40–46.
    1. Baum V. C., Tecson M. E. (1991). Ketamine inhibits transsarcolemmal calcium entry in guinea pig myocardium: direct evidence by single cell voltage clamp. Anesth. Analg. 73, 804–807.
    1. Belluardo N., Mudò G., Dell’Albani P., Jiang X. H., Condorelli D. F. (1995). NMDA receptor-dependent and -independent immediate early gene expression induced by focal mechanical brain injury. Neurochem. Int. 26, 443–453.
    1. Bhargava R., Young K. D. (2007). Procedural pain management patterns in academic pediatric emergency departments. Acad. Emerg. Med. 14, 479–482.
    1. Blain-Moraes S., Lee U., Ku S., Noh G., Mashour G. A. (2014). Electroencephalographic effects of ketamine on power, cross-frequency coupling and connectivity in the alpha bandwidth. Front. Syst. Neurosci. 8:114.
    1. Blonk M. I., Koder B. G., van den Bemt P. M., Huygen F. J. (2010). Use of oral ketamine in chronic pain management: a review. Eur. J. Pain 14, 466–472.
    1. Bonhomme V., Vanhaudenhuyse A., Demertzi A., Bruno M. A., Jaquet O., Bahri M. A., et al. . (2016). Resting-state network-specific breakdown of functional connectivity during ketamine alteration of consciousness in volunteers. Anesthesiology 125, 873–888.
    1. Bourgoin A., Albanèse J., Léone M., Sampol-Manos E., Viviand X., Martin C. (2005). Effects of sufentanil or ketamine administered in target-controlled infusion on the cerebral hemodynamics of severely brain-injured patients. Crit. Care Med. 33, 1109–1113.
    1. Bourgoin A., Albanèse J., Wereszczynski N., Charbit M., Vialet R., Martin C. (2003). Safety of sedation with ketamine in severe head injury patients: comparison with sufentanil. Crit. Care Med. 31, 711–717.
    1. Brachman R. A., McGowan J. C., Perusini J. N., Lim S. C., Pham T. H., Faye C., et al. . (2016). Ketamine as a prophylactic against stress-induced depressive-like behavior. Biol. Psychiatry 79, 776–786.
    1. Brown E. N., Purdon P. L., Van Dort C. J. (2011). General anesthesia and altered states of arousal: a systems neuroscience analysis. Annu. Rev. Neurosci. 34, 601–628.
    1. Brown R. H., Wagner E. M. (1999). Mechanisms of bronchoprotection by anesthetic induction agents: propofol versus ketamine. Anesthesiology 90, 822–828.
    1. Canpolat D. G., Esmaoglu A., Tosun Z., Akn A., Boyaci A., Coruh A. (2012). Ketamine-propofol vs. ketamine-dexmedetomidine combinations in pediatric patients undergoing burn dressing changes. J. Burn Care Res. 33, 718–722.
    1. Carr D. B., Goudas L. C., Denman W. T., Brookoff D., Staats P. S., Brennen L., et al. . (2004). 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 108, 17–27.
    1. Carrier N., Kabbaj M. (2013). Sex differences in the antidepressant-like effects of ketamine. Neuropharmacology 70, 27–34.
    1. Cha M. H., Eom J. H., Lee Y. S., Kim W. Y., Park Y. C., Min S. H., et al. . (2012). Beneficial effects of adding ketamine to intravenous patient-controlled analgesia with fentanyl after the Nuss procedure in pediatric patients. Yonsei Med. J. 53, 427–432.
    1. Chazan S., Buda I., Nesher N., Paz J., Weinbroum A. A. (2010). Low-dose ketamine via intravenous patient-controlled analgesia device after various transthoracic procedures improves analgesia and patient and family satisfaction. Pain Manag. Nurs. 11, 169–176.
    1. Chazan S., Ekstein M. P., Marouani N., Weinbroum A. A. (2008). Ketamine for acute and subacute pain in opioid-tolerant patients. J. Opioid Manag. 4, 173–180.
    1. Chen X., Shu S., Bayliss D. A. (2009). HCN1 channel subunits are a molecular substrate for hypnotic actions of ketamine. J. Neurosci. 29, 600–609.
    1. Choi M., Lee S. H., Wang S. E., Ko S. Y., Song M., Choi J. S., et al. . (2015). Ketamine produces antidepressant-like effects through phosphorylation-dependent nuclear export of histone deacetylase 5 (HDAC5) in rats. Proc. Natl. Acad. Sci. U S A 112, 15755–15760.
    1. Chong C., Schug S. A., Page-Sharp M., Jenkins B., Ilett K. F. (2009). 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. 29, 317–324.
    1. Chu P. S., Ma W. K., Wong S. C., Chu R. W., Cheng C. H., Wong S., et al. . (2008). The destruction of the lower urinary tract by ketamine abuse: a new syndrome? BJU Int. 102, 1616–1622.
    1. Clements J. A., Nimmo W. S. (1981). Pharmacokinetics and analgesic effect of ketamine in man. Br. J. Anaesth. 53, 27–30.
    1. Clements J. A., Nimmo W. S., Grant I. S. (1982). Bioavailability, pharmacokinetics and analgesic activity of ketamine in humans. J. Pharm. Sci. 71, 539–542.
    1. Cohen M. L., Trevor A. J. (1974). On the cerebral accumulation of ketamine and the relationship between metabolism of the drug and its pharmacological effects. J. Pharmacol. Exp. Ther. 189, 351–358.
    1. Correll G. E., Maleki J., Gracely E. J., Muir J. J., Harbut R. E. (2004). Subanesthetic ketamine infusion therapy: a retrospective analysis of a novel therapeutic approach to complex regional pain syndrome. Pain Med. 5, 263–275.
    1. Corssen G., Domino E. F. (1966). Dissociative anesthesia: further pharmacologic studies and first clinical experience with the phencyclidine derivative CI-581. Anesth. Analg. 45, 29–40.
    1. Dahi-Taleghani M., Fazli B., Ghasemi M., Vosoughian M., Dabbagh A. (2014). Effect of intravenous patient controlled ketamine analgesiaon postoperative pain in opium abusers. Anesth. Pain Med. 4:e14129.
    1. Davey C. G., Harrison B. J., Yücel M., Allen N. B. (2012). Regionally specific alterations in functional connectivity of the anterior cingulate cortex in major depressive disorder. Psychol. Med. 42, 2071–2081.
    1. Demling R. H., Ellerbe S., Jarrett F. (1978). Ketamine anesthesia for tangenital excision of burn eschar: a burn unit procedure. J. Trauma 18, 269–270.
    1. Dewhirst E., Frazier W. J., Leder M., Fraser D. D., Tobias J. D. (2013). Cardiac arrest following ketamine administration for rapid sequence intubation. J. Intensive Care Med. 28, 375–379.
    1. Diaz F. A., Bianco J. A., Bello A., Beer N., Velarde H., Izquierdo J. P., et al. . (1976). Effects of ketamine on canine cardiovascular function. Br. J. Anaesth. 48, 941–946.
    1. DiazGranados N., Ibrahim L. A., Brutsche N. E., Ameli R., Henter I. D., Luckenbaugh D. A., et al. . (2010a). Rapid resolution of suicidal ideation after a single infusion of an N-methyl-D-aspartate antagonist in patients with treatment-resistant major depressive disorder. J. Clin. Psychiatry 71, 1605–1611.
    1. DiazGranados N., Ibrahim L., Brutsche N. E., Newberg A., Kronstein P., Khalife S., et al. . (2010b). A randomized add-on trial of an N-methyl-D-aspartate antagonist in treatment-resistant bipolar depression. Arch. Gen. Psychiatry 67, 793–802.
    1. Domino E. F. (1980). History and pharmacology of PCP and PCP-related analogs. J. Psychedelic Drugs 12, 223–227.
    1. Domino E. F. (2010). Taming the ketamine tiger. 1965. Anesthesiology 113, 678–684.
    1. Domino E. F., Chodoff P., Corssen G. (1965). Pharmacologic effects of CI-581, a new dissociative anesthetic, in man. Clin. Pharmacol. Ther. 6, 279–291.
    1. Domino E. F., Domino S. E., Smith R. E., Domino L. E., Goulet J. R., Domino K. E., et al. . (1984). Ketamine kinetics in unmedicated and diazepam-premedicated subjects. Clin. Pharmacol. Ther. 36, 645–653.
    1. Domino E. F., Luby E. D. (2012). Phencyclidine/schizophrenia: one view toward the past, the other to the future. Schizophr. Bull. 38, 914–919.
    1. Donoghue A. C., Roback M. G., Cullen K. R. (2015). Remission from behavioral dysregulation in a child with PTSD after receiving procedural ketamine. Pediatrics 136, e694–e696.
    1. Duman R. S., Voleti B. (2012). Signaling pathways underlying the pathophysiology and treatment of depression: novel mechanisms for rapid-acting agents. Trends Neurosci. 35, 47–56.
    1. Dundee J. W., Lilburn J. K. (1978). Ketamine-iorazepam. Attenuation of psychic sequelae of ketamine by lorazepam. Anaesthesia 33, 312–314.
    1. Eikermann M., Grosse-Sundrup M., Zaremba S., Henry M. E., Bittner E. A., Hoffmann U., et al. . (2012). Ketamine activates breathing and abolishes the coupling between loss of consciousness and upper airway dilator muscle dysfunction. Anesthesiology 116, 35–46.
    1. Feder A., Parides M. K., Murrough J. W., Perez A. M., Morgan J. E., Saxena S., et al. . (2014). Efficacy of intravenous ketamine for treatment of chronic posttraumatic stress disorder: a randomized clinical trial. JAMA Psychiatry 71, 681–688.
    1. Finch P. M., Knudsen L., Drummond P. D. (2009). Reduction of allodynia in patients with complex regional pain syndrome: a double-blind placebo-controlled trial of topical ketamine. Pain 146, 18–25.
    1. Finck A. D., Ngai S. H. (1982). Opiate receptor mediation of ketamine analgesia. Anesthesiology 56, 291–297.
    1. Fine P. G. (1999). Low-dose ketamine in the management of opioid nonresponsive terminal cancer pain. J. Pain Symptom Manage. 17, 296–300.
    1. Franceschelli A., Sens J., Herchick S., Thelen C., Pitychoutis P. M. (2015). Sex differences in the rapid and the sustained antidepressant-like effects of ketamine in stress-naive and “depressed” mice exposed to chronic mild stress. Neuroscience 290, 49–60.
    1. Frenkel C., Urban B. W. (1992). Molecular actions of racemic ketamine on human CNS sodium channels. Br. J. Anaesth. 69, 292–297.
    1. Garcia L. S., Comim C. M., Valvassori S. S., Réus G. Z., Barbosa L. M., Andreazza A. C., et al. . (2008). Acute administration of ketamine induces antidepressant-like effects in the forced swimming test and increases BDNF levels in the rat hippocampus. Prog. Neuropsychopharmacol. Biol. Psychiatry 32, 140–144.
    1. Gardner A. E., Olson B. E., Lichtiger M. (1971). Cerebrospinal-fluid pressure during dissociative anesthesia with ketamine. Anesthesiology 35, 226–228.
    1. Ghasemi M., Kazemi M. H., Yoosefi A., Ghasemi A., Paragomi P., Amini H., et al. . (2014). Rapid antidepressant effects of repeated doses of ketamine compared with electroconvulsive therapy in hospitalized patients with major depressive disorder. Psychiatry Res. 215, 355–361.
    1. Gill J. R., Stajic M. (2000). Ketamine in non-hospital and hospital deaths in New York City. J. Forensic Sci. 45, 655–658.
    1. Grant I. S., Nimmo W. S., Clements J. A. (1981). Pharmacokinetics and analgesic effects of i.m. and oral ketamine. Br. J. Anaesth. 53, 805–810.
    1. Graven-Nielsen T., Aspegren Kendall S., Henriksson K. G., Bengtsson M., Sörensen J., Johnson A., et al. . (2000). Ketamine reduces muscle pain, temporal summation and referred pain in fibromyalgia patients. Pain 85, 483–491.
    1. Green S. M., Johnson N. E. (1990). Ketamine sedation for pediatric procedures: part 2, review and implications. Ann. Emerg. Med. 19, 1033–1046.
    1. Green S. M., Roback M. G., Krauss B., Brown L., McGlone R. G., Agrawal D., et al. . (2009). Predictors of emesis and recovery agitation with emergency department ketamine sedation: an individual-patient data meta-analysis of 8,282 children. Ann. Emerg. Med. 54, 171.e4–180.e4.
    1. Green S. M., Roback M. G., Krauss B., Emergency Department Ketamine Meta-Analysis Study Group . (2010). Laryngospasm during emergency department ketamine sedation: a case-control study. Pediatr. Emerg. Care 26, 798–802.
    1. Green S. M., Roback M. G., Kennedy R. M., Krauss B. (2011). Clinical practice guideline for emergency department ketamine dissociative sedation: 2011 update. Ann. Emerg. Med. 57, 449–461.
    1. Greifenstein F. E., Devault M., Yoshitake J., Gajewski J. E. (1958). A study of a 1-aryl cyclo hexyl amine for anesthesia. Anesth. Analg. 37, 283–294.
    1. Guillou N., Tanguy M., Seguin P., Branger B., Campion J. P., Malledant Y. (2003). The effects of small-dose ketamine on morphine consumption in surgical intensive care unit patients after major abdominal surgery. Anesth. Analg. 97, 843–847.
    1. Haeseler G., Tetzlaff D., Bufler J., Dengler R., Münte S., Hecker H., et al. . (2003). Blockade of voltage-operated neuronal and skeletal muscle sodium channels by S+- and R--ketamine. Anesth. Analg. 96, 1019–1026.
    1. Haley-Andrews S. (2006). Ketamine: the sedative of choice in a busy pediatric emergency department. J. Emerg. Nurs. 32, 186–188.
    1. Hardy J., Quinn S., Fazekas B., Plummer J., Eckermann S., Agar M., et al. . (2012). Randomized, double-blind, placebo-controlled study to assess the efficacy and toxicity of subcutaneous ketamine in the management of cancer pain. J. Clin. Oncol. 30, 3611–3617.
    1. Harvey M., Sleigh J., Voss L., Jose J., Gamage S., Pruijn F., et al. . (2015). Development of rapidly metabolized and ultra-short-acting ketamine analogs. Anesth. Analg. 121, 925–933.
    1. Hashimoto K. (2016). Ketamine’s antidepressant action: beyond NMDA receptor inhibition. Expert Opin. Ther. Targets 20, 1389–1392.
    1. Hayashi Y., Kawaji K., Sun L., Zhang X., Koyano K., Yokoyama T., et al. . (2011). Microglial Ca2+-activated K+ channels are possible molecular targets for the analgesic effects of S-ketamine on neuropathic pain. J. Neurosci. 31, 17370–17382.
    1. Hertle D. N., Dreier J. P., Woitzik J., Hartings J. A., Bullock R., Okonkwo D. O., et al. . (2012). Effect of analgesics and sedatives on the occurrence of spreading depolarizations accompanying acute brain injury. Brain 135, 2390–2398.
    1. Hijazi Y., Boulieu R. (2002). Contribution of CYP3A4, CYP2B6 and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Drug Metab. Dispos. 30, 853–858.
    1. Homayoun H., Moghaddam B. (2007). NMDA receptor hypofunction produces opposite effects on prefrontal cortex interneurons and pyramidal neurons. J. Neurosci. 27, 11496–11500.
    1. Hondorp M. (1987). Burn care protocols: administration of ketamine. Feature protocol San Francisco general hospital, San Francisco, California. J. Burn Care Rehabil. 8, 148–149.
    1. Hopper A. B., Vilke G. M., Castillo E. M., Campillo A., Davie T., Wilson M. P. (2015). Ketamine use for acute agitation in the emergency department. J. Emerg. Med. 48, 712–719.
    1. Huge V., Lauchart M., Magerl W., Schelling G., Beyer A., Thieme D., et al. . (2010). Effects of low-dose intranasal (S)-ketamine in patients with neuropathic pain. Eur. J. Pain 14, 387–394.
    1. Ibrahim L., Diazgranados N., Luckenbaugh D. A., Machado-Vieira R., Baumann J., Mallinger A. G., et al. . (2011). Rapid decrease in depressive symptoms with an N-methyl-d-aspartate antagonist in ECT-resistant major depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 35, 1155–1159.
    1. Idvall J., Holasek J., Stenberg P. (1983). Rectal ketamine for induction of anaesthesia in children. Anaesthesia 38, 60–64.
    1. Irifune M., Sato T., Kamata Y., Nishikawa T., Dohi T., Kawahara M. (2000). Evidence for GABA(A) receptor agonistic properties of ketamine: convulsive and anesthetic behavioral models in mice. Anesth. Analg. 91, 230–236.
    1. Irnaten M., Wang J., Chang K. S., Andresen M. C., Mendelowitz D. (2002). Ketamine inhibits sodium currents in identified cardiac parasympathetic neurons in nucleus ambiguus. Anesthesiology 96, 659–666.
    1. Jabre P., Combes X., Lapostolle F., Dhaouadi M., Ricard-Hibon A., Vivien B., et al. . (2009). Etomidate versus ketamine for rapid sequence intubation in acutely ill patients: a multicentre randomised controlled trial. Lancet 374, 293–300.
    1. Jensen L. L., Handberg G., Helbo-Hansen H. S., Skaarup I., Lohse T., Munk T., et al. . (2008). No morphine sparing effect of ketamine added to morphine for patient-controlled intravenous analgesia after uterine artery embolization. Acta Anaesthesiol. Scand. 52, 479–486.
    1. Johnstone M., Evans V., Baigel S. (1959). Sernyl (CI-395) in clinical anaesthesia. Br. J. Anaesth. 31, 433–439.
    1. Jung I., Jung S. H. (2012). Vasorelaxant mechanisms of ketamine in rabbit renal artery. Korean J. Anesthesiol. 63, 533–539.
    1. Kannan T. R., Saxena A., Bhatnagar S., Barry A. (2002). Oral ketamine as an adjuvant to oral morphine for neuropathic pain in cancer patients. J. Pain Symptom. Manage. 23, 60–65.
    1. Kantrowitz J. T., Halberstam B., Gangwisch J. (2015). Single-dose ketamine followed by daily D-Cycloserine in treatment-resistant bipolar depression. J. Clin. Psychiatry 76, 737–738.
    1. Kaur S., Saroa R., Aggarwal S. (2015). Effect of intraoperative infusion of low-dose ketamine on management of postoperative analgesia. J. Nat. Sci. Biol. Med. 6, 378–382.
    1. Ketalar (1970). “U.S. food and drug administration, center for drug evaluation and research,” in Label and Approval History. Available online at:
    1. Kim S. H., Kim S. I., Ok S. Y., Park S. Y., Kim M. G., Lee S. J., et al. . (2013). Opioid sparing effect of low dose ketamine in patients with intravenous patient-controlled analgesia using fentanyl after lumbar spinal fusion surgery. Korean J. Anesthesiol. 64, 524–528.
    1. Kollender Y., Bickels J., Stocki D., Maruoani N., Chazan S., Nirkin A., et al. . (2008). Subanaesthetic ketamine spares postoperative morphine and controls pain better than standard morphine does alone in orthopaedic-oncological patients. Eur. J. Cancer 44, 954–962.
    1. Krystal J. H., Karper L. P., Seibyl J. P., Freeman G. K., Delaney R., Bremner J. D., et al. . (1994). Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive and neuroendocrine responses. Arch. Gen. Psychiatry 51, 199–214.
    1. Kubota T., Anzawa N., Hirota K., Yoshida H., Kushikata T., Matsuki A. (1999). Effects of ketamine and pentobarbital on noradrenaline release from the medial prefrontal cortex in rats. Can. J. Anaesth. 46, 388–392.
    1. Kundra P., Velayudhan S., Krishnamachari S., Gupta S. L. (2013). Oral ketamine and dexmedetomidine in adults’ burns wound dressing—A randomized double blind cross over study. Burns 39, 1150–1156.
    1. Kushikata T., Yoshida H., Kudo M., Kudo T., Kudo T., Hirota K. (2011). Role of coerulean noradrenergic neurones in general anaesthesia in rats. Br. J. Anaesth. 107, 924–929.
    1. Lally N., Nugent A. C., Luckenbaugh D. A., Ameli R., Roiser J. P., Zarate C. A. (2014). Anti-anhedonic effect of ketamine and its neural correlates in treatment-resistant bipolar depression. Transl Psychiatry 4:e469.
    1. Lally N., Nugent A. C., Luckenbaugh D. A., Niciu M. J., Roiser J. P., Zarate C. A., Jr. (2015). Neural correlates of change in major depressive disorder anhedonia following open-label ketamine. J. Psychopharmacol. 29, 596–607.
    1. Langsjo J. W., Maksimow A., Salmi E., Kaisti K., Aalto S., Oikonen V., et al. . (2005). S-ketamine anesthesia increases cerebral blood flow in excess of the metabolic needs in humans. Anesthesiology 103, 258–268.
    1. Larkin G. L., Beautrais A. L. (2011). A preliminary naturalistic study of low-dose ketamine for depression and suicide ideation in the emergency department. Int. J. Neuropsychopharmacol. 14, 1127–1131.
    1. Lee U., Ku S., Noh G., Baek S., Choi B., Mashour G. A. (2013). Disruption of frontal–parietal communication by ketamine, propofol and sevoflurane. Anesthesiology 118, 1264–1275.
    1. Liman S., Cheung C. W., Wong K. L., Tai W., Qiu Q., Ng K. F., et al. . (2015). Preventive treatment with ketamine attenuates the ischaemia-reperfusion response in a chronic postischaemia pain model. Oxid. Med. Cell. Longev. 2015:380403.
    1. Liu Y., Lin D., Wu B., Zhou W. (2016). Ketamine abuse potential and use disorder. Brain Res. Bull. 126, 68–73.
    1. Lo R. S., Krishnamoorthy R., Freeman J. G., Austin A. S. (2011). Cholestasis and biliary dilatation associated with chronic ketamine abuse: a case series. Singapore Med. J. 52, e52–e55.
    1. Lu J., Nelson L. E., Franks N., Maze M., Chamberlin N. L., Saper C. B. (2008). Role of endogenous sleep-wake and analgesic systems in anesthesia. J. Comp. Neurol. 508, 648–662.
    1. Lydic R., Baghdoyan H. A. (2002). Ketamine and MK-801 decrease acetylcholine release in the pontine reticular formation, slow breathing and disrupt sleep. Sleep 25, 617–622.
    1. Maddox V. H., Godefroi E. F., Parcell R. F. (1965). The synthesis of phencyclidine and other 1-arylcyclohexylamines. J. Med. Chem. 8, 230–235.
    1. Malinovsky J. M., Servin F., Cozian A., Lepage J. Y., Pinaud M. (1996). Ketamine and norketamine plasma concentrations after i.v., nasal and rectal administration in children. Br. J. Anaesth. 77, 203–207.
    1. Martin L. L., Bouchal R. L., Smith D. J. (1982). Ketamine inhibits serotonin uptake in vivo. Neuropharmacology 21, 113–118.
    1. Martin D., Lodge D. (1985). Ketamine acts as a non-competitive N-methyl-D-aspartate antagonist on frog spinal cord in vitro. Neuropharmacology 24, 999–1003.
    1. Mashour G. A. (2016). Network-level mechanisms of ketamine anesthesia. Anesthesiology 125, 830–831.
    1. Mei X. P., Wang W., Wang W., Zhu C., Chen L., Zhang T., et al. . (2010). Combining ketamine with astrocytic inhibitor as a potential analgesic strategy for neuropathic pain ketamine, astrocytic inhibitor and pain. Mol. Pain 6:50.
    1. Mercadante S., Arcuri E., Tirelli W., Casuccio A. (2000). Analgesic effect of intravenous ketamine in cancer patients on morphine therapy: a randomized, controlled, double-blind, crossover, double-dose study. J. Pain Symptom. Manage. 20, 246–252.
    1. Mercer S. J. (2009). ‘The drug of war’–a historical review of the use of Ketamine in military conflicts. J. R. Nav. Med. Serv. 95, 145–150.
    1. McCloud T. L., Caddy C., Jochim J., Rendell J. M., Diamond P.R., Shuttleworth C., et al. . (2015). Ketamine and other glutamate receptor modulators for depression in adults. Cochrane Database Syst. Rev. 9:CD011612.
    1. Michelet P., Guervilly C., Hélaine A., Avaro J. P., Blayac D., Gaillat F., et al. . (2007). Adding ketamine to morphine for patient-controlled analgesia after thoracic surgery: influence on morphine consumption, respiratory function and nocturnal desaturation. Br. J. Anaesth. 99, 396–403.
    1. Michelet D., Hilly J., Skhiri A., Abdat R., Diallo T., Brasher C., et al. . (2016). Opioid-sparing effect of ketamine in children: a meta-analysis and trial sequential analysis of published studies. Paediatr. Drugs [Epub ahead of print].
    1. Mikkelsen S., Ilkjaer S., Brennum J., Borgbjerg F. M., Dahl J. B. (1999). The effect of naloxone on ketamine-induced effects on hyperalgesia and ketamine-induced side effects in humans. Anesthesiology 90, 1539–1545.
    1. Miller A. C., Jamin C. T., Elamin E. M. (2011). Continuous intravenous infusion of ketamine for maintenance sedation. Minerva Anestesiol. 77, 812–820.
    1. Mitra S., Kazal S. (2015). Oral ketamine for phantom limb pain: an option for challenging cases. Indian J. Anaesth. 59, 446–448.
    1. Moore K. A., Kilbane E. M., Jones R., Kunsman G. W., Levine B., Smith M. (1997). Tissue distribution of ketamine in a mixed drug fatality. J. Forensic Sci. 42, 1183–1185.
    1. Morgan C. J., Muetzelfeldt L., Curran H. V. (2009). Ketamine use, cognition and psychological wellbeing: a comparison of frequent, infrequent and ex-users with polydrug and non-using controls. Addiction 104, 77–87.
    1. Morgan C. J., Muetzelfeldt L., Curran H. V. (2010). Consequences of chronic ketamine self-administration upon neurocognitive function and psychological wellbeing: a 1-year longitudinal study. Addiction 105, 121–133.
    1. Murrough J. W., Iosifescu D. V., Chang L. C., Al Jurdi R. K., Green C. E., Perez A. M., et al. . (2013). Antidepressant efficacy of ketamine in treatment-resistant major depression: a two-site randomized controlled trial. Am. J. Psychiatry 170, 1134–1142.
    1. Murrough J. W., Soleimani L., DeWilde K. E., Collins K. A., Lapidus K. A., Iacoviello B. M., et al. . (2015). Ketamine for rapid reduction of suicidal ideation: a randomized controlled trial. Psychol. Med. 45, 3571–3580.
    1. Muthukumaraswamy S. D., Shaw A. D., Jackson L. E., Hall J., Moran R., Saxena N. (2015). Evidence that subanesthetic doses of ketamine cause sustained disruptions of NMDA and AMPA-mediated frontoparietal connectivity in humans. J. Neurosci. 35, 11694–11706.
    1. Nesher N., Ekstein M. P., Paz Y., Marouani N., Chazan S., Weinbroum A. A. (2009). Morphine with adjuvant ketamine vs. higher dose of morphine alone for immediate postthoracotomy analgesia. Chest 136, 245–252.
    1. Nesher N., Serovian I., Marouani N., Chazan S., Weinbroum A. A. (2008). Ketamine spares morphine consumption after transthoracic lung and heart surgery without adverse hemodynamic effects. Pharmacol. Res. 58, 38–44.
    1. Niesters M., Aarts L., Sarton E., Dahan A. (2013). 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. 110, 1010–1016.
    1. Niesters M., Martini C., Dahan A. (2014). Ketamine for chronic pain: risks and benefits. Br. J. Clin. Pharmacol. 77, 357–367.
    1. Nishimura M., Sato K., Okada T., Yoshiya I., Schloss P., Shimada S., et al. . (1998). Ketamine inhibits monoamine transporters expressed in human embryonic kidney 293 cells. Anesthesiology 88, 768–774.
    1. Noppers I., Niesters M., Swartjes M., Bauer M., Aarts L., Geleijnse N., et al. . (2011). Absence of long-term analgesic effect from a short-term S-ketamine infusion on fibromyalgia pain: a randomized, prospective, double blind, active placebo-controlled trial. Eur. J. Pain 15, 942–949.
    1. Nugent A. C., Diazgranados N., Carlson P. J., Ibrahim L., Luckenbaugh D. A., Brutsche N., et al. . (2014). Neural correlates of rapid antidepressant response to ketamine in bipolar disorder. Bipolar Disord. 16, 119–128.
    1. Nugent A. C., Robinson S. E., Coppola R., Zarate C. A., Jr. (2016). Preliminary differences in resting state MEG functional connectivity pre- and post-ketamine in major depressive disorder. Psychiatry Res. 254, 56–66.
    1. Ohnesorge H., Feng Z., Zitta K., Steinfath M., Albrecht M., Bein B. (2013). Influence of clonidine and ketamine on m-RNA expression in a model of opioid-induced hyperalgesia in mice. PLoS One 8:e79567.
    1. Oliven A., O’Hearn D. J., Boudewyns A., Odeh M., De Backer W., van de P., et al. . (2003). Upper airway response to electrical stimulation of the genioglossus in obstructive sleep apnea. J. Appl. Physiol. (1985) 95, 2023–2029.
    1. Orser B. A., Pennefather P. S., MacDonald J. F. (1997). Multiple mechanisms of ketamine blockade of N-methyl-D-aspartate receptors. Anesthesiology 86, 903–917.
    1. Oye I., Paulsen O., Maurset A. (1992). Effects of ketamine on sensory perception: evidence for a role of N-methyl-D-aspartate receptors. J. Pharmacol. Exp. Ther. 260, 1209–1213.
    1. Pacheco D. F., Romero T. R. L., Duarte I. D. G. (2014). Central antinociception induced by ketamine is mediated by endogenous opioids and μ- and δ-opioid receptors. Brain Res. 1562, 69–75.
    1. Pal D., Hambrecht-Wiedbusch V. S., Silverstein B. H., Mashour G. A. (2015). Electroencephalographic coherence and cortical acetylcholine during ketamine-induced unconsciousness. Br. J. Anaesth. 114, 979–989.
    1. Pedraz J. L., Calvo M. B., Lanao J. M., Muriel C., Santos Lamas J., Domínguez-Gil A. (1989). Pharmacokinetics of rectal ketamine in children. Br. J. Anaesth. 63, 671–674.
    1. Perrine S. A., Ghoddoussi F., Michaels M. S., Sheikh I. S., McKelvey G., Galloway M. P. (2014). Ketamine reverses stress-induced depression-like behavior and increased GABA levels in the anterior cingulate: an 11.7 T 1H-MRS study in rats. Prog. Neuropsychopharmacol. Biol. Psychiatry 51, 9–15.
    1. Perumal D. K., Adhimoolam M., Selvaraj N., Lazarus S. P., Mohammed M. A. (2015). Midazolam premedication for Ketamine-induced emergence phenomenon: a prospective observational study. J. Res. Pharm. Pract. 4, 89–93.
    1. Petrenko A. B., Yamakura T., Fujiwara N., Askalany A. R., Baba H., Sakimura K. (2004). Reduced sensitivity to ketamine and pentobarbital in mice lacking the N-methyl-D-aspartate receptor GluRepsilon1 subunit. Anesth. Analg. 99, 1136–1140.
    1. Pomarol-Clotet E., Honey G. D., Murray G. K., Corlett P. R., Absalom A. R., Lee M., et al. . (2006). Psychological effects of ketamine in healthy volunteers. Phenomenological study. Br. J. Psychiatry 189, 173–179.
    1. Poon T. L., Wong K. F., Chan M. Y., Fung K. W., Chu S. K., Man C. W., et al. . (2010). Upper gastrointestinal problems in inhalational ketamine abusers. J. Dig. Dis. 11, 106–110.
    1. Reus G. Z., Abelaira H. M., dos Santos M. A., Carlessi A. S., Tomaz D. B., Neotti M. V., et al. . (2013). Ketamine and imipramine in the nucleus accumbens regulate histone deacetylation induced by maternal deprivation and are critical for associated behaviors. Behav. Brain Res. 256, 451–456.
    1. Reza F. M., Zahra F., Esmaeel F., Hossein A. (2010). Preemptive analgesic effect of ketamine in patients undergoing elective cesarean section. Clin. J. Pain 26, 223–226.
    1. Rodriguez C. I., Kegeles L. S., Levinson A., Ogden R. T., Mao X., Milak M. S., et al. . (2015). In vivo effects of ketamine on glutamate-glutamine and gamma-aminobutyric acid in obsessive-compulsive disorder: proof of concept. Psychiatry Res. 233, 141–147.
    1. Rolan P., Lim S., Sunderland V., Liu Y., Molnar V. (2014). The absolute bioavailability of racemic ketamine from a novel sublingual formulation. Br. J. Clin. Pharmacol. 77, 1011–1016.
    1. Sakai T., Sumikawa K. (2014). Phantom limb pain exacerbated by intravenous ketamine. J. Anesth. 28:643.
    1. Salas S., Frasca M., Planchet-Barraud B., Burucoa B., Pascal M., Lapiana J. M., et al. . (2012). Ketamine analgesic effect by continuous intravenous infusion in refractory cancer pain: considerations about the clinical research in palliative care. J. Palliat. Med. 15, 287–293.
    1. Sato Y., Kobayashi E., Hakamata Y., Kobahashi M., Wainai T., Murayama T., et al. . (2004). Chronopharmacological studies of ketamine in normal and NMDA epsilon1 receptor knockout mice. Br. J. Anaesth. 92, 859–864.
    1. Schmittner M. D., Vajkoczy S. L., Horn P., Bertsch T., Quintel M., Vajkoczy P., et al. . (2007). Effects of fentanyl and S(+)-ketamine on cerebral hemodynamics, gastrointestinal motility and need of vasopressors in patients with intracranial pathologies: a pilot study. J. Neurosurg. Anesthesiol. 19, 257–262.
    1. Schroeder K. E., Irwin Z. T., Gaidica M., Bentley J. N., Patil P. G., Mashour G. A., et al. . (2016). Disruption of corticocortical information transfer during ketamine anesthesia in the primate brain. Neuroimage 134, 459–465.
    1. Schwartzman R. J., Alexander G. M., Grothusen J. R., Paylor T., Reichenberger E., Perreault M. (2009). Outpatient intravenous ketamine for the treatment of complex regional pain syndrome: a double-blind placebo controlled study. Pain 147, 107–115.
    1. Sen H., Sizlan A., Yanarates O., Emirkadi H., Ozkan S., Dagli G., et al. . (2009). A comparison of gabapentin and ketamine in acute and chronic pain after hysterectomy. Anesth. Analg. 109, 1645–1650.
    1. Shanthanna H., Huilgol M., Manivackam V. K. (2010). Early and effective use of ketamine for treatment of phantom limb pain. Indian J. Anaesth. 54, 157–159.
    1. Shaprio H. M., Wyte S. R., Harris A. B. (1972). Ketamine anaesthesia in patients with intracranial pathology. Br. J. Anaesth. 44, 1200–1204.
    1. Sigtermans M. J., van Hilten J. J., Bauer M. C., Arbous M. S., Marinus J., Sarton E. Y., et al. . (2009). Ketamine produces effective and long-term pain relief in patients with complex regional pain syndrome type 1. Pain 145, 304–311.
    1. Sleigh J., Harvey M., Voss L., Denny B. (2014). Ketamine–More mechanisms of action than just NMDA blockade. Trends Anaesth. Crit. Care 4, 76–81.
    1. Smith D. J., Bouchal R. L., DeSanctis C. A., Monroe P. J., Amedro J. B., Perrotti J. M., et al. . (1987). Properties of the interaction between ketamine and opiate binding sites in vivo and in vitro. Neuropharmacology 26, 1253–1260.
    1. Strayer R. J., Nelson L. S. (2008). Adverse events associated with ketamine for procedural sedation in adults. Am. J. Emerg. Med. 26, 985–1028.
    1. Suppa E., Valente A., Catarci S., Zanfini B. A., Draisci G. (2012). A study of low-dose S-ketamine infusion as “preventive” pain treatment for cesarean section with spinal anesthesia: benefits and side effects. Minerva Anestesiol. 78, 774–781.
    1. Sveticic G., Farzanegan F., Zmoos P., Zmoos S., Eichenberger U., Curatolo M. (2008). Is the combination of morphine with ketamine better than morphine alone for postoperative intravenous patient-controlled analgesia? Anesth. Analg. 106, 287–293.
    1. Tan S., Rudd J. A., Yew D. T. (2011). Gene expression changes in GABA(A) receptors and cognition following chronic ketamine administration in mice. PLoS One 6:e21328.
    1. Tarumi Y., Watanabe S., Bruera E., Ishitani K. (2000). High-dose ketamine in the management of cancer-related neuropathic pain. J. Pain Symptom Manage. 19, 405–407.
    1. Thomson A. M., West D. C., Lodge D. (1985). An N-methylaspartate receptor-mediated synapse in rat cerebral cortex: a site of action of ketamine? Nature 313, 479–481.
    1. Traber D. L., Wilson R. D., Priano L. L. (1968). Differentiation of the cardiovascular effects of CI-581. Anesth. Analg. 47, 769–778.
    1. Traber D. L., Wilson R. D., Priano L. L. (1970). Blockade of the hypertensive response to ketamine. Anesth. Analg. 49, 420–426.
    1. Tsai T.-H., Cha T.-L., Lin C.-M., Tsao C.-W., Tang S.-H., Chuang F.-P., et al. . (2009). Ketamine-associated bladder dysfunction. Int. J. Urol. 16, 826–829.
    1. Wang X., Ding X., Tong Y., Zong J., Zhao X., Ren H., et al. . (2014). Ketamine does not increase intracranial pressure compared with opioids: meta-analysis of randomized controlled trials. J. Anesth. 28, 821–827.
    1. Waxman K., Shoemaker W. C., Lippmann M. (1980). Cardiovascular effects of anesthetic induction with ketamine. Anesth. Analg. 59, 355–358.
    1. Webb A. R., Skinner B. S., Leong S., Kolawole H., Crofts T., Taverner M., et al. . (2007). The addition of a small-dose ketamine infusion to tramadol for postoperative analgesia: a double-blinded, placebo-controlled, randomized trial after abdominal surgery. Anesth. Analg. 104, 912–917.
    1. Weber F., Wulf H., Gruber M., Biallas R. (2004). S-ketamine and s-norketamine plasma concentrations after nasal and i.v. administration in anesthetized children. Paediatr. Anaesth. 14, 983–988.
    1. Weiner A. L., Vieira L., McKay C. A., Bayer M. J. (2000). Ketamine abusers presenting to the emergency department: a case series. J. Emerg. Med. 18, 447–451.
    1. White P. F., Ham J., Way W. L., Trevor A. J. (1980). Pharmacology of ketamine isomers in surgical patients. Anesthesiology 52, 231–239.
    1. White P. F., Schuttler J., Shafer A., Stanski D. R., Horai Y., Trevor A. J. (1985). Comparative pharmacology of the ketamine isomers. Studies in volunteers. Br. J. Anaesth. 57, 197–203.
    1. Wieber J., Gugler R., Hengstmann J. H., Dengler H. J. (1975). Pharmacokinetics of ketamine in man. Anaesthesist 24, 260–263.
    1. Willman E. V., Andolfatto G. (2007). A prospective evaluation of “ketofol” (ketamine/propofol combination) for procedural sedation and analgesia in the emergency department. Ann. Emerg. Med. 49, 23–30.
    1. Wolff K., Winstock A. R. (2006). Ketamine : from medicine to misuse. CNS Drugs 20, 199–218.
    1. Wong J. J., O’Daly O., Mehta M. A., Young A. H., Stone J. M. (2016). Ketamine modulates subgenual cingulate connectivity with the memory-related neural circuit-a mechanism of relevance to resistant depression? PeerJ 4:e1710.
    1. Wong G. L., Tam Y. H., Ng C. F., Chan A. W., Choi P. C., Chu W. C., et al. . (2014). Liver injury is common among chronic abusers of ketamine. Clin. Gastroenterol. Hepatol. 12, 1759.e1–1762.e1.
    1. Wyte S. R., Shapiro H. M., Turner P., Harris A. B. (1972). Ketamine-induced intracranial hypertension. Anesthesiology 36, 174–176.
    1. Yamakage M., Hirshman C. A., Croxton T. L. (1995). Inhibitory effects of thiopental, ketamine and propofol on voltage-dependent Ca2+ channels in porcine tracheal smooth muscle cells. Anesthesiology 83, 1274–1282.
    1. Yamakura T., Chavez-Noriega L. E., Harris R. A. (2000). Subunit-dependent inhibition of human neuronal nicotinic acetylcholine receptors and other ligand-gated ion channels by dissociative anesthetics ketamine and dizocilpine. Anesthesiology 92, 1144–1153.
    1. Yamamura T., Harada K., Okamura A., Kemmotsu O. (1990). Is the site of action of ketamine anesthesia the N-methyl-D-aspartate receptor? Anesthesiology 72, 704–710.
    1. Yanagihara Y., Ohtani M., Kariya S., Uchino K., Hiraishi T., Ashizawa N., et al. . (2003). Plasma concentration profiles of ketamine and norketamine after administration of various ketamine preparations to healthy Japanese volunteers. Biopharm. Drug Dispos. 24, 37–43.
    1. Yang C., Hu Y. M., Zhou Z. Q., Zhang G. F., Yang J. J. (2013). Acute administration of ketamine in rats increases hippocampal BDNF and mTOR levels during forced swimming test. Ups. J. Med. Sci. 118, 3–8.
    1. Yang C. Y., Wong C. S., Chang J. Y., Ho S. T. (1996). Intrathecal ketamine reduces morphine requirements in patients with terminal cancer pain. Can. J. Anaesth. 43, 379–383.
    1. Yeaman F., Meek R., Egerton-Warburton D., Rosengarten P., Graudins A. (2014). Sub-dissociative-dose intranasal ketamine for moderate to severe pain in adult emergency department patients. Emerg. Med. Australas 26, 237–242.
    1. Yeom J. H., Chon M. S., Jeon W. J., Shim J. H. (2012). Peri-operative ketamine with the ambulatory elastometric infusion pump as an adjuvant to manage acute postoperative pain after spinal fusion in adults: a prospective randomized trial. Korean J. Anesthesiol. 63, 54–58.
    1. Yu W. L., Cho C. C., Lung P. F., Hung E. H., Hui J. W., Chau H. H., et al. . (2014). Ketamine-related cholangiopathy: a retrospective study on clinical and imaging findings. Abdom. Imaging 39, 1241–1246.
    1. Zakine J., Samarcq D., Lorne E., Moubarak M., Montravers P., Beloucif S., et al. . (2008). Postoperative ketamine administration decreases morphine consumption in major abdominal surgery: a prospective, randomized, double-blind, controlled study. Anesth. Analg. 106, 1856–1861.
    1. Zanos P., Moaddel R., Morris P. J., Georgiou P., Fischell J., Elmer G. I., et al. . (2016). NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature 533, 481–486.
    1. Zarate C. A., Jr., Singh J. B., Carlson P. J., Brutsche N. E., Ameli R., Luckenbaugh D. A., et al. . (2006). A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Arch. Gen. Psychiatry 63, 856–864.
    1. Zhang J. C., Li S. X., Hashimoto K. (2014). R (-)-ketamine shows greater potency and longer lasting antidepressant effects than S (+)-ketamine. Pharmacol. Biochem. Behav. 116, 137–141.
    1. Zhou C., Douglas J. E., Kumar N. N., Shu S., Bayliss D. A., Chen X. (2013). Forebrain HCN1 channels contribute to hypnotic actions of ketamine. Anesthesiology 118, 785–795.
    1. Zhou C., Liang P., Liu J., Ke B., Wang X., Li F., et al. . (2015). HCN1 channels contribute to the effects of amnesia and hypnosis but not immobility of volatile anesthetics. Anesth. Analg. 121, 661–666.
    1. Zhou J., Shaw S. G., Gilleece Y. (2013). Dilated common bile duct and deranged liver function tests associated with ketamine use in two HIV-positive MSM. Int. J. STD AIDS 24, 667–669.
    1. Zigman D., Blier P. (2013). Urgent ketamine infusion rapidly eliminated suicidal ideation for a patient with major depressive disorder: a case report. J. Clin. Psychopharmacol. 33, 270–272.
    1. Zsigmond E. K., Matsuki A., Kothary S. P., Jallad M. (1976). Arterial hypoxemia caused by intravenous ketamine. Anesth. Analg. 55, 311–314.

Source: PubMed

3
S'abonner