Consensus Guidelines on the Use of Intravenous Ketamine Infusions for Chronic Pain From the American Society of Regional Anesthesia and Pain Medicine, the American Academy of Pain Medicine, and the American Society of Anesthesiologists

Steven P Cohen, Anuj Bhatia, Asokumar Buvanendran, Eric S Schwenk, Ajay D Wasan, Robert W Hurley, Eugene R Viscusi, Samer Narouze, Fred N Davis, Elspeth C Ritchie, Timothy R Lubenow, William M Hooten, Steven P Cohen, Anuj Bhatia, Asokumar Buvanendran, Eric S Schwenk, Ajay D Wasan, Robert W Hurley, Eugene R Viscusi, Samer Narouze, Fred N Davis, Elspeth C Ritchie, Timothy R Lubenow, William M Hooten

Abstract

Background: Over the past 2 decades, the use of intravenous ketamine infusions as a treatment for chronic pain has increased dramatically, with wide variation in patient selection, dosing, and monitoring. This has led to a chorus of calls from various sources for the development of consensus guidelines.

Methods: In November 2016, the charge for developing consensus guidelines was approved by the boards of directors of the American Society of Regional Anesthesia and Pain Medicine and, shortly thereafter, the American Academy of Pain Medicine. In late 2017, the completed document was sent to the American Society of Anesthesiologists' Committees on Pain Medicine and Standards and Practice Parameters, after which additional modifications were made. Panel members were selected by the committee chair and both boards of directors based on their expertise in evaluating clinical trials, past research experience, and clinical experience in developing protocols and treating patients with ketamine. Questions were developed and refined by the committee, and the groups responsible for addressing each question consisted of modules composed of 3 to 5 panel members in addition to the committee chair. Once a preliminary consensus was achieved, sections were sent to the entire panel, and further revisions were made. In addition to consensus guidelines, a comprehensive narrative review was performed, which formed part of the basis for guidelines.

Results: Guidelines were prepared for the following areas: indications; contraindications; whether there was evidence for a dose-response relationship, or a minimum or therapeutic dose range; whether oral ketamine or another N-methyl-D-aspartate receptor antagonist was a reasonable treatment option as a follow-up to infusions; preinfusion testing requirements; settings and personnel necessary to administer and monitor treatment; the use of preemptive and rescue medications to address adverse effects; and what constitutes a positive treatment response. The group was able to reach consensus on all questions.

Conclusions: Evidence supports the use of ketamine for chronic pain, but the level of evidence varies by condition and dose range. Most studies evaluating the efficacy of ketamine were small and uncontrolled and were either unblinded or ineffectively blinded. Adverse effects were few and the rate of serious adverse effects was similar to placebo in most studies, with higher dosages and more frequent infusions associated with greater risks. Larger studies, evaluating a wider variety of conditions, are needed to better quantify efficacy, improve patient selection, refine the therapeutic dose range, determine the effectiveness of nonintravenous ketamine alternatives, and develop a greater understanding of the long-term risks of repeated treatments.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

FIGURE 1
FIGURE 1
Graphical depiction of the relationship between ketamine dose and duration of analgesic benefit in randomized placebo-controlled trials that evaluated IV ketamine for chronic pain with a minimum of 48 hours' follow-up.,,,–, A trend line is been plotted to indicate the nature of this relationship.

References

    1. Global Burden of Disease Study 2013 Collaborators. Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2015;386:743–800.
    1. Chopra K, Arora V. An intricate relationship between pain and depression: clinical correlates, coactivation factors and therapeutic targets. Expert Opin Ther Targets. 2014;18:159–176.
    1. Radvansky BM, Puri S, Sifonios AN, Eloy JD, Le V. Ketamine-a narrative review of its uses in medicine. Am J Ther. 2016;23:e1414–e1426.
    1. Anderson P. Ketamine for chronic pain on the rise. . Published April 4, 2017. Accessed November 3, 2017.
    1. Sanacora G, Frye MA, McDonald W, et al. American Psychiatric Association (APA) Council of Research Task Force on Novel Biomarkers and Treatments. A consensus statement on the use of ketamine in the treatment of mood disorders. JAMA Psychiatry. 2017;74:399–405.
    1. U.S. Preventive Task Force. Grade definitions. Available at: . Accessed November 3, 2017.
    1. Helm Ii S, Simopoulos TT, Stojanovic M, Abdi S, El Terany MA. Effectiveness of thermal annular procedures in treating discogenic low back pain. Pain Physician. 2017;20:447–470.
    1. Oxford Centre for Evidence-Based Medicine—Levels of Evidence (March 2009). Available at: . Accessed January 15, 2018.
    1. The GRADE Working Group. GRADE. Available at: . Updated April 2016. Accessed January 18, 2018.
    1. Domino EF, Chodoff P, Corssen G. Pharmacologic effects of CI-581, a new dissociative anesthetic, in man. Clin Pharmacol Ther. 1965;6:279–291.
    1. Greifenstein FE, DeVault M, Yoshitake J, et al. A study of a 1-aryl cyclo hexyl amine for anesthesia. Anesth Analg. 1958:37:283–294.
    1. Maddox VH, Godefroi EF, Parcell RF. The synthesis of phencyclidine and other 1-arylcyclohexylamines. J Med Chem. 1965;8:230–235.
    1. Chen G, Ensor CR, Russell D, Bohner B. The pharmacology of 1-(1-phenylcyclohexyl) piperidine HCl. J Pharmacol Exp Ther. 1959;127:241–250.
    1. Mion G. History of anaesthesia: the ketamine story—past, present and future. Eur J Anaesthesiol. 2017;34:571–575.
    1. Murray CJ, Atkinson C, Bhalla K, et al. U.S. Burden of Disease Collaborators. The state of US health, 1990–2010: burden of diseases, injuries, and risk factors. JAMA. 2013;310:591–608.
    1. Institute of Medicine Committee on Advancing Pain Research, Care, and Education. Relieving Pain in America: A Blueprint for Transforming Prevention, Care, Education and Research. Washington, DC: The National Academies Press; 2011. Available at: . Accessed November 3, 2017.
    1. Leadley RM, Armstrong N, Lee YC, Allen A, Kleijnen J. Chronic diseases in the European Union: the prevalence and health cost implications of chronic pain. J Pain Palliat Care Pharmacother. 2012;26:310–325.
    1. Cohen SP, Mao J. Neuropathic pain: mechanisms and their clinical implications. BMJ. 2014;348:f7656.
    1. Clarke H, Bonin RP, Orser BA, Englesakis M, Wijeysundera DN, Katz J. The prevention of chronic postsurgical pain using gabapentin and pregabalin: a combined systematic review and meta-analysis. Anesth Analg. 2012;115:428–442.
    1. Cohen KL, Harris S. Efficacy and safety of nonsteroidal anti-inflammatory drugs in the therapy of diabetic neuropathy. Arch Intern Med. 1987;147:1442–1444.
    1. Graven-Nielsen T, Aspegren Kendall S, Henriksson KG, et al. Ketamine reduces muscle pain, temporal summation, and referred pain in fibromyalgia patients. Pain. 2000;85:483–491.
    1. Mercadante S, Arcuri E, Tirelli W, Casuccio A. Analgesic effect of intravenous ketamine in cancer patients on morphine therapy: a randomized, controlled, double-blind, crossover, double-dose study. J Pain Symptom Manage. 2000;20:246–252.
    1. Vo T, Rice AS, Dworkin RH. Non-steroidal anti-inflammatory drugs for neuropathic pain: how do we explain continued widespread use? Pain. 2009;143:169–171.
    1. Mak P, Broadbear JH, Kolosov A, Goodchild CS. Long-term antihyperalgesic and opioid-sparing effects of 5-day ketamine and morphine infusion (“burst ketamine”) in diabetic neuropathic rats. Pain Med. 2015;16:1781–1793.
    1. Castel A, Hélie P, Beaudry F, Vachon P. Bilateral central pain sensitization in rats following a unilateral thalamic lesion may be treated with high doses of ketamine. BMC Vet Res. 2013;9:59.
    1. Mehta AK, Halder S, Khanna N, Tandon OP, Singh UR, Sharma KK. Role of NMDA and opioid receptors in neuropathic pain induced by chronic constriction injury of sciatic nerve in rats. J Basic Clin Physiol Pharmacol. 2012;23:49–55.
    1. Le Roy C, Laboureyras E, Gavello-Baudy S, Chateauraynaud J, Laulin JP, Simonnet G. Endogenous opioids released during non-nociceptive environmental stress induce latent pain sensitization via a NMDA-dependent process. J Pain. 2011;12:1069–1079.
    1. Rivat C, Laulin JP, Corcuff JB, Célèrier E, Pain L, Simonnet G. Fentanyl enhancement of carrageenan-induced long-lasting hyperalgesia in rats: prevention by the N-methyl-d-aspartate receptor antagonist ketamine. Anesthesiology. 2002;96:381–391.
    1. Bouhassira D, Lantéri-Minet M, Attal N, Laurent B, Touboul C. Prevalence of chronic pain with neuropathic characteristics in the general population. Pain. 2008;136:380–387.
    1. Harifi G, Amine M, Ait Ouazar M, et al. Prevalence of chronic pain with neuropathic characteristics in the Moroccan general population: a national survey. Pain Med. 2013;14:287–292.
    1. Torrance N, Smith BH, Bennett MI, Lee AJ. The epidemiology of chronic pain of predominantly neuropathic origin. Results from a general population survey. J Pain. 2006;7:281–289.
    1. Finnerup NB, Haroutounian S, Kamerman P, et al. Neuropathic pain: an updated grading system for research and clinical practice. Pain. 2016;157:1599–1606.
    1. De Mos M, de Bruijn AG, Huygen FJ, Dieleman JP, Stricker BH, Sturkenboom MC. The incidence of complex regional pain syndrome: a population-based study. Pain. 2007;129:12–20.
    1. Sandroni P, Benrud-Larson LM, McClelland RL, Low PA. Complex regional pain syndrome type I: incidence and prevalence in Olmsted County, a population-based study. Pain. 2003;103:199–207.
    1. Smith BH, Torrance N, Bennett MI, Lee AJ. Health and quality of life associated with chronic pain of predominantly neuropathic origin in the community. Clin J Pain. 2007;23:143–149.
    1. Maher DP, Chen L, Mao J. Intravenous ketamine infusions for neuropathic pain management: a promising therapy in need of optimization. Anesth Analg. 2017;124:661–674.
    1. Patil S, Anitescu M. Efficacy of outpatient ketamine infusions in refractory chronic pain syndromes: a 5-year retrospective analysis. Pain Med. 2012;13:263–269.
    1. Cohen SP, Liao W, Gupta A, Plunkett A. Ketamine in pain management. Adv Psychosom Med. 2011;30:139–161.
    1. Orser BA, Pennefather PS, MacDonald JF. Multiple mechanisms of ketamine blockade of N-methyl-d-aspartate receptors. Anesthesiology. 1997;86:903–917.
    1. Chang CH, Hsiao YH, Chen YW, Yu YJ, Gean PW. Social isolation-induced increase in NMDA receptors in the hippocampus exacerbates emotional dysregulation in mice. Hippocampus. 2015;25:474–485.
    1. Duman RS, Li N, Liu RJ, Duric V, Aghajanian G. Signaling pathways underlying the rapid antidepressant actions of ketamine. Neuropharmacology. 2012;62:35–41.
    1. Trujillo KA, Akil H. Inhibition of opiate tolerance by non-competitive N-methyl-d-aspartate receptor antagonists. Brain Res. 1994;633:178–188.
    1. Zhou HY, Chen SR, Pan HL. Targeting N-methyl-d-aspartate receptors for treatment of neuropathic pain. Expert Rev Clin Pharmacol. 2011;4:379–388.
    1. 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;91:177–187.
    1. Casale R, Symeonidou Z, Bartolo M. Topical treatments for localized neuropathic pain. Curr Pain Headache Rep. 2017;21:15.
    1. Sarton E, Teppema LJ, Olievier C, et al. The involvement of the mu-opioid receptor in ketamine-induced respiratory depression and antinociception. Anesth Analg. 2001;93:1495–1500.
    1. Smith DJ, Perrotti JM, Mansell AL, et al. Ketamine analgesia is not related to an opiate action in the periaqueductal gray region of the rat brain. Pain. 1985;21:253–265.
    1. 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;110:1010–1016.
    1. Sinner B, Graf BM. Ketamine. Handb Exp Pharmacol. 2008;182:313–333.
    1. Seeman P, Ko F, Tallerico T. Dopamine receptor contribution to the action of PCP, LSD and ketamine psychotomimetics. Mol Psychiatry. 2005;10:877–883.
    1. Sleigh J, Harvey M, Voss L, Denny B. Ketamine-more mechanisms of action than just NMDA blockade. Trends Anaesth Crit Care. 2014;4:76–81.
    1. Carr DB, Goudas LC, Denman WT, 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.
    1. Rabi J. Topical ketamtene: a review of the history, mechanisms, uses, safety and future. Int J Pharm Compd. 2016;20:107–113.
    1. Lilius TO, Jokinen V, Neuvonen MS, Niemi M, Kalso EA, Rauhala PV. Ketamine coadministration attenuates morphine tolerance and leads to increased brain concentrations of both drugs in the rat. Br J Pharmacol. 2015;172:2799–2813.
    1. Wu L, Huang X, Sun L. The efficacy of N-methyl-d-aspartate receptor antagonists on improving the postoperative pain intensity and satisfaction after remifentanil-based anesthesia in adults: a meta-analysis. J Clin Anesth. 2015;27:311–324.
    1. Leal PC, Salomão R, Brunialti MK, Sakata RK. Evaluation of the effect of ketamine on remifentanil-induced hyperalgesia: a double-blind, randomized study. J Clin Anesth. 2015;27:331–337.
    1. Pestieau SR, Finkel JC, Junqueira MM, et al. Prolonged perioperative infusion of low-dose ketamine does not alter opioid use after pediatric scoliosis surgery. Paediatr Anaesth. 2014;24:582–590.
    1. Velly AM, Mohit S. Epidemiology of pain and relation to psychiatric disorders [published online ahead of print May 15, 2017]. Prog Neuropsychopharmacol Biol Psychiatry. 2017.
    1. Abdallah CG, Adams TG, Kelmendi B, Esterlis I, Sanacora G, Krystal JH. Ketamine's mechanism of action: a path to rapid-acting antidepressants. Depress Anxiety. 2016;33:689–697.
    1. Schoevers RA, Chaves TV, Balukova SM, Rot MA, Kortekaas R. Oral ketamine for the treatment of pain and treatment-resistant depression. Br J Psychiatry. 2016;208:108–113.
    1. Chilukuri H, Reddy NP, Pathapati RM, Manu AN, Jollu S, Shaik AB. Acute antidepressant effects of intramuscular versus intravenous ketamine. Indian J Psychol Med. 2014;36:71–76.
    1. Lapidus KA, Levitch CF, Perez AM, et al. A randomized controlled trial of intranasal ketamine in major depressive disorder. Biol Psychiatry. 2014;76:970–976.
    1. Beurel E, Song L, Jope RS. Inhibition of glycogen synthase kinase-3 is necessary for the rapid antidepressant effect of ketamine in mice. Mol Psychiatry. 2011;16:1068–1070.
    1. Li N, Lee B, Liu RJ, et al. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science. 2010;329:959–964.
    1. Li N, Liu RJ, Dwyer JM, et al. Glutamate N-methyl-d-aspartate receptor antagonists rapidly reverse behavioral and synaptic deficits caused by chronic stress exposure. Biol Psychiatry. 2011;69:754–761.
    1. Garcia LS, Comim CM, Valvassori SS, et al. 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. 2008;32:140–144.
    1. Maeng S, Zarate CA, Jr., Du J, et al. Cellular mechanisms underlying the antidepressant effects of ketamine: role of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors. Biol Psychiatry. 2008;63:349–352.
    1. Lener MS, Niciu MJ, Ballard ED, et al. Glutamate and gamma-aminobutyric acid systems in the pathophysiology of major depression and antidepressant response to ketamine. Biol Psychiatry. 2017;81:886–897.
    1. Scheuing L, Chiu CT, Liao HM, Chuang DM. Antidepressant mechanism of ketamine: perspective from preclinical studies. Front Neurosci. 2015;9:249.
    1. Feder A, Parides MK, Murrough JW, et al. Efficacy of intravenous ketamine for treatment of chronic posttraumatic stress disorder: a randomized clinical trial. JAMA Psychiatry. 2014;71:681–688.
    1. Burbiel JC. Primary prevention of posttraumatic stress disorder: drugs and implications. Mil Med Res. 2015;2:24.
    1. Nair J, Singh AS. The role of the glutamatergic system in posttraumatic stress disorder. CNS Spectr. 2008;13:585–591.
    1. Riaza Bermudo-Soriano C, Perez-Rodriguez MM, Vaquero-Lorenzo C, Baca-Garcia E. New perspectives in glutamate and anxiety. Pharmacol Biochem Behav. 2012;100:752–774.
    1. Himmelseher S, Pfenninger E. The clinical use of S-(+)-ketamine—a determination of its place. Anasthesiol Intensivmed Notfallmed Schmerzther. 1998;33:764–770.
    1. Pees C, Haas NA, Ewert P, Berger F, Lange PE. Comparison of analgesic/sedative effect of racemic ketamine and S(+)-ketamine during cardiac catheterization in newborns and children. Pediatr Cardiol. 2003;24:424–429.
    1. Peterbauer C, Larenza PM, Knobloch M, et al. Effects of a low dose infusion of racemic and S-ketamine on the nociceptive withdrawal reflex in standing ponies. Vet Anaesth Analg. 2008;35:414–423.
    1. Guará Sobrinho H, Garcia JB, Vasconcelos JW, Sousa JC, Ferro LS. Analgesic efficacy of the intra-articular administration of S(+)-ketamine in patients undergoing total knee arthroplasty. Rev Bras Anestesiol. 2012;62:665–675.
    1. Yang C, Shirayama Y, Zhang JC, et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry. 2015;5:e632.
    1. Engelhardt W. [Recovery and psychomimetic reactions following S-(+)-ketamine]. Anaesthesist. 1997;46(suppl 1):S38–S42.
    1. Zhang JC, Li SX, Hashimoto K. R(−)-ketamine shows greater potency and longer lasting antidepressant effects than S(+)-ketamine. Pharmacol Biochem Behav. 2014;116:1371–41.
    1. Chen X, Shu S, Bayliss DA. HCN1 channel subunits are a molecular substrate for hypnotic actions of ketamine. J Neurosci. 2009;29:600–609.
    1. Coates KM, Flood P. Ketamine and its preservative, benzethonium chloride, both inhibit human recombinant alpha7 and alpha4beta2 neuronal nicotinic acetylcholine receptors in Xenopus oocytes. Br J Pharmacol. 2001;134:871–879.
    1. Liy-Salmeron G, Meneses A. Effects of 5-HT drugs in prefrontal cortex during memory formation and the ketamine amnesia-model. Hippocampus. 2008;18:965–974.
    1. Synowiec AS, Singh DS, Yenugadhati V, Valeriano JP, Schramke CJ, Kelly KM. Ketamine use in the treatment of refractory status epilepticus. Epilepsy Res. 2013;105:183–188.
    1. Manocha A, Sharma KK, Mediratta PK. Possible mechanism of anticonvulsant effect of ketamine in mice. Indian J Exp Biol. 2001;39:1002–1008.
    1. Gálvez V, McGuirk L, Loo CK. The use of ketamine in ECT anaesthesia: a systematic review and critical commentary on efficacy, cognitive, safety and seizure outcomes. World J Biol Psychiatry. 2017;18:424–444.
    1. Hirshman CA, Krieger W, Littlejohn G, Lee R, Julien R. Ketamine-aminophylline-induced decrease in seizure threshold. Anesthesiology. 1982;56:464–467.
    1. Darimont PC, Jenkins LC. The influence of intravenous anaesthetics on enflurane-induced central nervous system seizure activity. Can Anaesth Soc J. 1977;24:42–56.
    1. Celesia GG, Chen RC, Bamforth BJ. Effects of ketamine in epilepsy. Neurology. 1975;25:169–172.
    1. Dimitrov A, Despodova TS. Case of convulsions with late onset after Ketalar anesthesia. [in Bulgarian] Akush Ginekol (Sofiia). 1976;15:229–230.
    1. Ketalar (ketamine hydrochloride) injection. Available at: . Accessed on November 11, 2017.
    1. Rao LK, Flaker AM, Friedel CC, Kharasch ED. Role of cytochrome P4502B6 polymorphisms in ketamine metabolism and clearance. Anesthesiology. 2016;125:1103–1112.
    1. Zheng X, Fang P, Bao SS, Lin D, Cai JP, Hu GX. Function of 38 variants CYP2C9 polymorphism on ketamine metabolism in vitro. J Pharmacol Sci. 2017;135:8–13.
    1. Singh NS, Zarate CA, Jr, Moaddel R, Bernier M, Wainer IW. What is hydroxynorketamine and what can it bring to neurotherapeutics? Expert Rev Neurother. 2014;14:1239–1242.
    1. Zanos P, Moaddel R, Morris PJ, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016;533:481–486.
    1. Kurdi MS, Theerth KA, Deva RS. Ketamine: current applications in anesthesia, pain, and critical care. Anesth Essays Res. 2014;8:283–290.
    1. Soliman MG, Brindle GF, Kuster G. Response to hypercapnia under ketamine anaesthesia. Can Anaesth Soc J. 1975;22:486–494.
    1. Fisher AD, Rippee B, Shehan H, Conklin C, Mabry RL. Prehospital analgesia with ketamine for combat wounds: a case series. J Spec Oper Med. 2014;14:11–17.
    1. 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;88:1202–1210.
    1. MacPherson RD, Woods D, Penfold J. Ketamine and midazolam delivered by patient-controlled analgesia in relieving pain associated with burns dressings. Clin J Pain. 2008;24:568–571.
    1. Lenze EJ, Farber NB, Kharasch E, et al. Ninety-six hour ketamine infusion with co-administered clonidine for treatment-resistant depression: a pilot randomised controlled trial. World J Biol Psychiatry. 2016;17:230–238.
    1. White MC, Karsli C. Long-term use of an intravenous ketamine infusion in a child with significant burns. Paediatr Anaesth. 2007;17:1102–1104.
    1. Domino EF. Taming the ketamine tiger. 1965. Anesthesiology. 2010;113:678–684.
    1. Laulin JP, Maurette P, Corcuff JB, Rivat C, Chauvin M, Simonnet G. The role of ketamine in preventing fentanyl-induced hyperalgesia and subsequent acute morphine tolerance. Anesth Analg. 2002;94:1263–1269.
    1. Richebé P, Rivat C, Rivalan B, Maurette P, Simonnet G. Low doses ketamine: antihyperalgesic drug, non-analgesic. [in French]. Ann Fr Anesth Reanim. 2005;24:1349–1359.
    1. Tawfic QA. A review of the use of ketamine in pain management. J Opioid Manag. 2013;9:379–388.
    1. Prommer EE. Ketamine for pain: an update of uses in palliative care. J Palliat Med. 2012;15:474–483.
    1. Ghasemi M, Phillips C, Trillo L, de Miguel Z, Das D, Salehi A. The role of NMDA receptors in the pathophysiology and treatment of mood disorders. Neurosci Biobehav Rev. 2014;47:336–358.
    1. Lin CH, Huang YJ, Lin CJ, Lane HY, Tsai GE. NMDA neurotransmission dysfunction in mild cognitive impairment and Alzheimer's disease. Curr Pharm Des. 2014;20:5169–5179.
    1. Alshuft HM, Condon LA, Dineen RA, Auer DP. Cerebral cortical thickness in chronic pain due to knee osteoarthritis: the effect of pain duration and pain sensitization. PLoS One. 2016;11:e0161687.
    1. De Kock M, Loix S, Lavand'homme P. Ketamine and peripheral inflammation. CNS Neurosci Ther. 2013;19:403–410.
    1. Vranken JH. Elucidation of pathophysiology and treatment of neuropathic pain. Cent Nerv Syst Agents Med Chem. 2012;12:304–314.
    1. Woolf CJ, Thompson SW. The induction and maintenance of central sensitization is dependent on N-methyl-d-aspartic acid receptor activation; implications for the treatment of post-injury pain hypersensitivity states. Pain. 1991;44:293–299.
    1. Seminowicz DA, Wideman TH, Naso L, et al. Effective treatment of chronic low back pain in humans reverses abnormal brain anatomy and function. J Neurosci. 2011;31:7540–7550.
    1. Čeko M, Shir Y, Ouellet JA, Ware MA, Stone LS, Seminowicz DA. Partial recovery of abnormal insula and dorsolateral prefrontal connectivity to cognitive networks in chronic low back pain after treatment. Hum Brain Mapp. 2015;36:2075–2092.
    1. Kregel J, Vuijk PJ, Descheemaeker F, et al. The Dutch Central Sensitization Inventory (CSI): factor analysis, discriminative power, and test-retest reliability. Clin J Pain. 2016;32:624–630.
    1. Eichenberger U, Neff F, Sveticic G, et al. Chronic phantom limb pain: the effects of calcitonin, ketamine, and their combination on pain and sensory thresholds. Anesth Analg. 2008;106:1265–1273.
    1. Kvarnström A, Karlsten R, Quiding H, Emanuelsson BM, Gordh T. The effectiveness of intravenous ketamine and lidocaine on peripheral neuropathic pain. Acta Anaesthesiol Scand. 2003;47:868–877.
    1. Kvarnström A, Karlsten R, Quiding H, Gordh T. The analgesic effect of intravenous ketamine and lidocaine on pain after spinal cord injury. Acta Anaesthesiol Scand. 2004;48:498–506.
    1. Baad-Hansen L, Juhl GI, Jensen TS, Brandsborg B, Svensson P. Differential effect of intravenous S-ketamine and fentanyl on atypical odontalgia and capsaicin-evoked pain. Pain. 2007;129:46–54.
    1. Nickel FT, Ott S, Möhringer S, et al. Effects of different anesthetics on pain processing in an experimental human pain model. Pain Pract. 2016;16:820–830.
    1. Kiefer RT, Rohr P, Ploppa A, et al. A pilot open-label study of the efficacy of subanesthetic isomeric S(+)-ketamine in refractory CRPS patients. Pain Med. 2008;9:44–54.
    1. 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;147:107–115.
    1. Tajerian M, Leu D, Yang P, Huang TT, Kingery WS, Clark JD. Differential efficacy of ketamine in the acute versus chronic stages of complex regional pain syndrome in mice. Anesthesiology. 2015;123:1435–1447.
    1. Zhang LM, Zhou WW, Ji YJ, et al. Anxiolytic effects of ketamine in animal models of posttraumatic stress disorder. Psychopharmacology (Berl). 2015;232:663–672.
    1. Juven-Wetzler A, Cohen H, Kaplan Z, Kohen A, Porat O, Zohar J. Immediate ketamine treatment does not prevent posttraumatic stress responses in an animal model for PTSD. Eur Neuropsychopharmacol. 2014;24:469–479.
    1. Morena M, Berardi A, Peloso A, et al. Effects of ketamine, dexmedetomidine and propofol anesthesia on emotional memory consolidation in rats: consequences for the development of post-traumatic stress disorder. Behav Brain Res. 2017;329:215–220.
    1. Scheidegger M, Henning A, Walter M, et al. Ketamine administration reduces amygdalo-hippocampal reactivity to emotional stimulation. Hum Brain Mapp. 2016;37:1941–1952.
    1. Li CT, Chen MH, Lin WC, et al. The effects of low-dose ketamine on the prefrontal cortex and amygdala in treatment-resistant depression: a randomized controlled study. Hum Brain Mapp. 2016;37:1080–1090.
    1. Hasler G, Drevets WC, Manji HK, Charney DS. Discovering endophenotypes for major depression. Neuropsychopharmacology. 2004;29:1765–1781.
    1. Stewart AM, Nguyen M, Poudel MK, et al. The failure of anxiolytic therapies in early clinical trials: what needs to be done. Expert Opin Investig Drugs. 2015;24:543–556.
    1. Khan A, Schwartz K. Study designs and outcomes in antidepressant clinical trials. Essent Psychopharmacol. 2005;6:221–226.
    1. Laskowski K, Stirling A, McKay WP, Lim HJ. A systematic review of intravenous ketamine for postoperative analgesia. Can J Anaesth. 2011;58:911–923.
    1. 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;49:1405–1428.
    1. Jouguelet-Lacoste J, LaColla 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.
    1. Elia N, Tramèr MR. Ketamine and postoperative pain—a quantitative systematic review of randomised trials. Pain. 2005;113:61–70.
    1. Elliott K, Minami N, Kolesnikov YA, Pasternak GW, Inturrisi CE. The NMDA receptor antagonists, LY274614 and MK-801, and the nitric oxide synthase inhibitor, NG-nitro-l-arginine, attenuate analgesic tolerance to the mu-opioid morphine but not to kappa opioids. Pain. 1994;56:69–75.
    1. Wenzel JT, Schwenk ES, Baratta JL, Viscusi ER. Managing opioid-tolerant patients in the perioperative surgical home. Anesthesiol Clin. 2016;34:287–301.
    1. Mahathanaruk M, Hitt J, de Leon-Casasola OA. Perioperative management of the opioid tolerant patient for orthopedic surgery. Anesthesiol Clin. 2014;32:923–932.
    1. Loftus RW, Yeager MP, Clark JA, et al. Intraoperative ketamine reduces perioperative opiate consumption in opiate-dependent patients with chronic back pain undergoing back surgery. Anesthesiology. 2010;113:639–646.
    1. Barreveld AM, Correll DJ, Liu X, et al. Ketamine decreases postoperative pain scores in patients taking opioids for chronic pain: results of a prospective, randomized, double-blind study. Pain Med. 2013;14:925–934.
    1. Urban MK, Ya deau JT, Wukovits B, Lipnitsky JY. Ketamine as an adjunct to postoperative pain management in opioid tolerant patients after spinal fusions: a prospective randomized trial. HSS J. 2008;4:62–65.
    1. Vaid P, Green T, Shinkaruk K, King-Shier K. Low-dose ketamine infusions for highly opioid-tolerant adults following spinal surgery: a retrospective before-and-after study. Pain Manag Nurs. 2016;17:150–158.
    1. Subramaniam K, Akhouri V, Glazer PA, et al. Intra- and postoperative very low dose intravenous ketamine infusion does not increase pain relief after major spine surgery in patients with preoperative narcotic analgesic intake. Pain Med. 2011;12:1276–1283.
    1. Assouline B, Tramèr MR, Kreienbühl L, Elia N. Benefit and harm of adding ketamine to an opioid in a patient-controlled analgesia device for the control of postoperative pain: systematic review and meta-analyses of randomized controlled trials with trial sequential analyses. Pain. 2016;157:2854–2864.
    1. Wang L, Johnston B, Kaushal A, Cheng D, Zhu F, Martin J. Ketamine added to morphine or hydromorphone patient-controlled analgesia for acute postoperative pain in adults: a systematic review and meta-analysis of randomized trials. Can J Anaesth. 2016;63:311–325.
    1. Uprety D, Baber A, Foy M. Ketamine infusion for sickle cell pain crisis refractory to opioids: a case report and review of literature. Ann Hematol. 2014;93:769–771.
    1. Jennings CA, Bobb BT, Noreika DM, Coyne PJ. Oral ketamine for sickle cell crisis pain refractory to opioids. J Pain Palliat Care Pharmacother. 2013;27:150–154.
    1. Meals CG, Mullican BD, Shaffer CM, Dangerfield PF, Ramirez RP. Ketamine infusion for sickle cell crisis pain in an adult. J Pain Symptom Manage. 2011;42:e7–e9.
    1. Tawfic QA, Faris AS, Kausalya R. The role of a low-dose ketamine-midazolam regimen in the management of severe painful crisis in patients with sickle cell disease. J Pain Symptom Manage. 2014;47:334–340.
    1. Losing AK, Jones JM, Keric A, Briggs SE, Leedahl DD. Ketamine infusion therapy as an alternative pain control strategy in patients with multi-trauma including rib fracture; case report and literature review. Bull Emerg Trauma. 2016;4:165–169.
    1. Max MB, Byas-Smith MG, Gracely RH, Bennett GJ. Intravenous infusion of the NMDA antagonist, ketamine, in chronic posttraumatic pain with allodynia: a double-blind comparison to alfentanil and placebo. Clin Neuropharmacol. 1995;18:360–368.
    1. Gottrup H, Bach FW, Juhl G, Jensen TS. Differential effect of ketamine and lidocaine on spontaneous and mechanical evoked pain in patients with nerve injury pain. Anesthesiology. 2006;104:527–536.
    1. Eide PK, Stubhaug A, Stenehjem AE. Central dysesthesia pain after traumatic spinal cord injury is dependent on N-methyl-d-aspartate receptor activation. Neurosurgery. 1995;37:1080–1087.
    1. Amr YM. Multi-day low dose ketamine infusion as adjuvant to oral gabapentin in spinal cord injury related chronic pain: a prospective, randomized, double blind trial. Pain Physician. 2010;13:245–249.
    1. Nikolajsen L, Hansen CL, Nielsen J, Keller J, Arendt-Nielsen L, Jensen TS. The effect of ketamine on phantom pain: a central neuropathic disorder maintained by peripheral input. Pain. 1996;67:69–77.
    1. Eide PK, Jørum E, Stubhaug A, Bremnes J, Breivik H. Relief of post-herpetic neuralgia with the N-methyl-d-aspartic acid receptor antagonist ketamine: a double-blind, cross-over comparison with morphine and placebo. Pain. 1994;58:347–354.
    1. Sorensen J, Bengtsson A, Backman E, Henriksson KG, Bengtsson M. Pain analysis in patients with fibromyalgia. Effects of intravenous morphine, lidocaine, and ketamine. Scand J Rheumatol. 1995;24:360–365.
    1. Sorensen J, Bengtsson A, Ahlner J, Henriksson KG, Ekselius L, Bengtsson M. Fibromyalgia—are there different mechanisms in the processing of pain? A double blind crossover comparison of analgesic drugs. J Rheumatol. 1997;24:1615–1621.
    1. Noppers I, Niesters M, Swartjes M, et al. 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. 2011;15:942–949.
    1. Sigtermans MJ, van Hilten JJ, Bauer MC, et al. Ketamine produces effective and long-term pain relief in patients with complex regional pain syndrome type 1. Pain. 2009;145:304–311.
    1. Mitchell AC, Fallon MT. A single infusion of intravenous ketamine improves pain relief in patients with critical limb ischaemia: results of a double blind randomised controlled trial. Pain. 2002;97:275–281.
    1. Persson J, Hasselstrom J, Wiklund B, Heller A, Svensson JO, Gustafsson LL. The analgesic effect of racemic ketamine in patients with chronic ischemic pain due to lower extremity arteriosclerosis obliterans. Acta Anaesthesiol Scand. 1998;42:750–758.
    1. Salas S, Frasca M, Planchet-Barraud B, et al. Ketamine analgesic effect by continuous intravenous infusion in refractory cancer pain: considerations about the clinical research in palliative care. J Palliat Med. 2012;15:287–293.
    1. Granata L, Niebergall H, Langner R, Agosti R, Sakellaris L. Ketamine I.V. for the treatment of cluster headaches: an observational study [in German]. Schmerz. 2016;30:286–288.
    1. Moisset X, Clavelou P, Lauxerois M, Dallel R, Picard P. Ketamine infusion combined with magnesium as a therapy for intractable chronic cluster headache: report of two cases. Headache. 2017;57:1261–1264.
    1. Pomeroy JL, Marmura MJ, Nahas SJ, Viscusi ER. Ketamine infusions for treatment refractory headache. Headache. 2017;57:276–282.
    1. Nicolodi M, Sicuteri F. Exploration of NMDA receptors in migraine: therapeutic and theoretic implications. Int J Clin Pharmacol Res. 1995;15:181–189.
    1. Afridi SK, Giffin NJ, Kaube H, Goadsby PJ. A randomized controlled trial of intranasal ketamine in migraine with prolonged aura. Neurology. 2013;80:642–647.
    1. Hrobjartsson A, Thomsen AS, Emanuelsson F, et al. Observer bias in randomised clinical trials with binary outcomes: systematic review of trials with both blinded and non-blinded outcome assessors. BMJ. 2012;344:e1119.
    1. Savovic J, Jones HE, Altman DG, et al. Influence of reported study design characteristics on intervention effect estimates from randomized, controlled trials. Ann Intern Med. 2012;157:429–438.
    1. Wood L, Egger M, Gluud LL, et al. Empirical evidence of bias in treatment effect estimates in controlled trials with different interventions and outcomes: meta-epidemiological study. BMJ. 2008;336:601–605.
    1. Gooding JM, Dimick AR, Tavakoli M, Corssen G. A physiologic analysis of cardiopulmonary responses to ketamine anesthesia in noncardiac patients. Anesth Analg. 1977;56:813–816.
    1. Waxman K, Shoemaker WC, Lippmann M. Cardiovascular effects of anesthetic induction with ketamine. Anesth Analg. 1980;59:355–358.
    1. Niesters M, Martini C, Dahan A. Ketamine for chronic pain: risks and benefits. Br J Clin Pharmacol. 2014;77:357–367.
    1. Craven R. Ketamine. Anaesthesia. 2007;62(suppl 1):48–53.
    1. Reich DL, Silvay G. Ketamine: an update on the first twenty-five years of clinical experience. Can J Anaesth. 1989;36:186–197.
    1. Gomes LM, Garcia JB, Ribamar JS, Jr, Nascimento AG. Neurotoxicity of subarachnoid preservative-free S(+)-ketamine in dogs. Pain Physician. 2011;14:83–90.
    1. Walker SM, Westin BD, Deumens R, Grafe M, Yaksh TL. Effects of intrathecal ketamine in the neonatal rat: evaluation of apoptosis and long-term functional outcome. Anesthesiology. 2010;113:147–159.
    1. Vranken JH, Troost D, de Haan P, et al. Severe Toxic damage to the rabbit spinal cord after intrathecal administration of preservative-free S(+)-ketamine. Anesthesiology. 2006;105:813–818.
    1. Rojas AC, Alves JG, Moreira EL, et al. The effects of subarachnoid administration of preservative-free S(+)-ketamine on spinal cord and meninges in dogs. Anesth Analg. 2012;114:450–455.
    1. Errando CL, Sifre C, Moliner S, et al. Subarachnoid ketamine in swine—pathological findings after repeated doses: acute toxicity study. Reg Anesth Pain Med. 1999;24:146–152.
    1. Deer TR, Pope AJ, Hayek SM, et al. The Poly-Analgesic Consensus Conference (PACC): recommendations for intrathecal drug delivery: guidance for improving safety and mitigating risks. Neuromodulation. 2017;20:155–176.
    1. Schwenk ES, Goldberg SF, Patel RD, et al. Adverse drug effects and preoperative medication factors related to perioperative low-dose ketamine infusions. Reg Anesth Pain Med. 2016;41:482–487.
    1. Wai MS, Chan WM, Zhang AQ, Wu Y, Yew DT. Long-term ketamine and ketamine plus alcohol treatments produced damages in liver and kidney. Hum Exp Toxicol. 2012;31:877–886.
    1. Bell RF. Ketamine for chronic noncancer pain: concerns regarding toxicity. Curr Opin Support Palliat Care. 2012;6:183–187.
    1. Rajandram R, Yap NY, Ong TA, et al. Oral ketamine induced pathological changes of the urinary tract in a rat model. Malays J Pathol. 2017;39:47–53.
    1. Wong GL, Tam YH, Ng CF, et al. Liver injury is common among chronic abusers of ketamine. Clin Gastroenterol Hepatol. 2014;12:1759–1762.
    1. Noppers IM, Niesters M, Aarts LP, et al. Drug-induced liver injury following a repeated course of ketamine treatment for chronic pain in CRPS type 1 patients: a report of 3 cases. Pain. 2011;152:2173–2178.
    1. Morgan CJ, Curran HV. Ketamine use: a review. Addiction. 2011;107:27–38.
    1. Jhang JF, Hsu YH, Kuo HC. Possible pathophysiology of ketamine-related cystitis and associated treatment strategies. Int J Urol. 2015;22:816–825.
    1. Shahani R, Streutker C, Dickson B, Stewart RJ. Ketamine-associated ulcerative cystitis: a new clinical entity. Urology. 2007;69:810–812.
    1. Chen CH, Lee MH, Chen YC, Lin MF. Ketamine-snorting associated cystitis. J Formos Med Assoc. 2011;110:787–791.
    1. Grégoire MC, MacLellan DL, Finley GA. A pediatric case of ketamine-associated cystitis (letter-to-the-editor re: Shahani R, Streutker C, Dickson B, et al. Ketamine-associated ulcerative cystitis: a new clinical entity. Urology 2007;69: 810–812). Urology. 2007;71:1232–1233.
    1. Drayna PC, Estrada C, Wang W, Saville BR, Arnold DH. Ketamine sedation is not associated with clinically meaningful elevation of intraocular pressure. Am J Emerg Med. 2012;30:1215–1218.
    1. Goldberg ME, Domsky R, Scaringe D, et al. Multi-day low dose ketamine infusion for the treatment of complex regional pain syndrome. Pain Physician. 2005;8:175–179.
    1. Backonja M, Arndt G, Gombar KA, Check B, Zimmermann M. Response of chronic neuropathic pain syndromes to ketamine: a preliminary study. Pain. 1994;56:51–57.
    1. Felsby S, Nielsen J, Arendt-Nielsen L, Jensen TS. NMDA receptor blockade in chronic neuropathic pain: a comparison of ketamine and magnesium chloride. Pain. 1996;64:283–291.
    1. Jørum E, Warncke T, Stubhaug A. Cold allodynia and hyperalgesia in neuropathic pain: the effect of N-methyl-d-aspartate (NMDA) receptor antagonist ketamine—a double-blind, cross-over comparison with alfentanil and placebo. Pain. 2003;101:229–235.
    1. American Society of Anesthesiologists. Practice guidelines for sedation and analgesia by non-anesthesiologists. Anesthesiology. 2002;96:1004–1017.
    1. Amr YM. Epidural ketamine in post spinal cord injury–related chronic pain. Anesth Essays Res. 2011;5:83–86.
    1. Ketamine hydrochloride—drug summary. Available at: . Accessed January 31, 2017.
    1. 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.
    1. Ahern TL, Herring AA, Miller S, Frazee BW. Low-dose ketamine infusion for emergency department patients with severe pain. Pain Med. 2015;16:1402–1409.
    1. Gorlin AW, Rosenfeld DM, Ramakrishna H. Intravenous sub-anesthetic ketamine for perioperative analgesia. J Anaesthesiol Clin Pharmacol. 2016;32:160–167.
    1. Dundee JW, Fee JP, Moore J, McIlroy PD, Wilson DB. Changes in serum enzyme levels following ketamine infusions. Anaesthesia. 1980;35:12–16.
    1. Murphy EJ. Acute pain management pharmacology for the patient with concurrent renal or hepatic disease. Anaesth Intensive Care. 2005;33:311–322.
    1. Cohen L, Athaide V, Wickham ME, Doyle-Waters MM, Rose NG, Hohl CM. The effect of ketamine on intracranial and cerebral perfusion pressure and health outcomes: a systematic review. Ann Emerg Med. 2015;65:43–51.
    1. Zeiler FA, Teitelbaum J, West M, Gillman LM. The ketamine effect on intracranial pressure in nontraumatic neurological illness. J Crit Care. 2014;29:1096–1106.
    1. Guerra F. Ketamine and angina. Anesthesiology. 1978;155.
    1. Ward J, Standage C. Angina pain precipitated by a continuous subcutaneous infusion of ketamine. J Pain Symptom Manage. 2003;25:6–7.
    1. Kraus C, Lanzenberger R, Kasper S. Ketamine for the treatment of depression. JAMA Psychiatry. 2017;74:970.
    1. Lahti AC, Holcomb HH, Medoff DR, Tamminga CA. Ketamine activates psychosis and alters limbic blood flow in schizophrenia. Neuroreport. 1995;6:869–872.
    1. Epocrates. Ketamine: contraindications/cautions. Available at: . Accessed January 18, 2018.
    1. WebMD. Who should not take ketamine HCL syringe? Available at: . Accessed January 18, 2018.
    1. Davis FA. Ketamine drug monograph. Available at: . Accessed January 18, 2018.
    1. Pal HR, Berry N, Kumar R, Ray R. Ketamine dependence. Anaesth Intensive Care. 2002;30:382–384.
    1. Chou R, Fanciullo GJ, Fine PG, Miaskowski C, Passik SD, Portenoy RK. Opioids for chronic noncancer pain: prediction and identification of aberrant drug-related behaviors: a review of the evidence for an American Pain Society and American Academy of Pain Medicine clinical practice guideline. J Pain. 2009;10:131–146.
    1. Yang M, Qian C, Liu Y. Suboptimal treatment of diabetic peripheral neuropathic pain in the United States. Pain Med. 2015;16:2075–2083.
    1. Xu Y, Hackett M, Carter G, et al. Effects of low-dose and very low-dose ketamine among patients with major depression: a systematic review and meta-analysis. Int J Neuropsychopharmacol. 2016;19:1–15.
    1. Coyle CM, Laws KR. The use of ketamine as an antidepressant: a systematic review and meta-analysis. Hum Psychopharmacol. 2015;30:152–163.
    1. Szymkowicz SM, Finnegan N, Dale RM. A 12-month naturalistic observation of three patients receiving repeat intravenous ketamine infusions for their treatment-resistant depression. J Affect Disord. 2013;147:416–420.
    1. Lauritsen C, Mazuera S, Lipton RB, Ashina S. Intravenous ketamine for subacute treatment of refractory chronic migraine: a case series. J Headache Pain. 2016;17:106.
    1. Huge V, Lauchart M, Magerl W, et al. Effects of low-dose intranasal (S)-ketamine in patients with neuropathic pain. Eur J Pain. 2010;14:387–394.
    1. Kiefer RT, Rohr P, Ploppa A, et al. Efficacy of ketamine in anesthetic dosage for the treatment of refractory complex regional pain syndrome: an open-label phase II study. Pain Med. 2008;9:1173–1201.
    1. Noppers I, Niesters M, Aarts L, Smith T, Sarton E, Dahan A. Ketamine for the treatment of chronic non-cancer pain. Expert Opin Pharmacother. 2010;11:2417–2429.
    1. Jafarinia M, Afarideh M, Tafakhori A, et al. Efficacy and safety of oral ketamine versus diclofenac to alleviate mild to moderate depression in chronic pain patients: a double-blind, randomized, controlled trial. J Affect Disord. 2016;204:1–8.
    1. Ishizuka P, Garcia JB, Sakata R, Issy A, Mülich SL. Assessment of oral S+ ketamine associated with morphine for the treatment of oncologic pain. Rev Bras Anestesiol. 2007;57:19–31.
    1. Lauretti G, Lima I, Reis M, Prado W, Pereira N. Oral ketamine and transdermal nitroglycerin as analgesic adjuvants to oral morphine therapy for cancer pain management. Anesthesiology. 1999;90:1528–1533.
    1. Haines DR, Gaines SP. N of 1 randomised controlled trials of oral ketamine in patients with chronic pain. Pain. 1999;83:283–287.
    1. Marchetti F, Coutaux A, Bellanger A, Magneux C, Bourgeois P, Mion G. Efficacy and safety of oral ketamine for the relief of intractable chronic pain: a retrospective 5-year study of 51 patients. Eur J Pain. 2015;19:984–993.
    1. Furuhashi-Yonaha A, Iida H, Asano T, Takeda T, Dohi S. Short- and long-term efficacy of oral ketamine in eight chronic-pain patients. Can J Anaesth. 2002;49:886–887.
    1. Giorgetti R, Marcotulli D, Tagliabracci A, Schifano F. Effects of ketamine on psychomotor, sensory and cognitive functions relevant for driving ability. Forensic Sci Int. 2015;252:127–142.
    1. Cohen SP, Chang AS, Larkin T, Mao J. The intravenous ketamine test: a predictive response tool for oral dextromethorphan treatment in neuropathic pain. Anesth Analg. 2004;99:1753–1759.
    1. Cohen SP, Verdolin MH, Chang AS, Kurihara C, Morlando BJ, Mao J. The intravenous ketamine test predicts subsequent response to an oral dextromethorphan treatment regimen in fibromyalgia patients. J Pain. 2006;7:391–398.
    1. Cohen SP, Wang S, Chen L, et al. An intravenous ketamine test as a predictive response tool in opioid-exposed patients with persistent pain. J Pain Symptom Manage. 2009;37:698–708.
    1. Cohen SP, Kapoor SG, Rathmell JP. Intravenous infusion tests have limited utility for selecting long-term drug therapy in patients with chronic pain: a systematic review. Anesthesiology. 2009;111:416–431.
    1. Collins S, Sigtermans M, Dahan A. NMDA receptor antagonists for the treatment of neuropathic pain. Pain Med. 2010;11:1726–1742.
    1. Aiyer R, Mehta N, Gungor S, Gulati A. A systematic review of NMDA receptor antagonists for treatment of neuropathic pain in clinical practice. Clin J Pain. 2018;34:450–467.
    1. Maneglia R, Cousin MT. A comparison between propofol and ketamine for anaesthesia in the elderly. Haemodynamic effects during induction and maintenance. Anaesthesia. 1988;43(suppl):109–111.
    1. Kiefer RT, Rohr P, Ploppa A, Altemeyer KH, Schwartzman RJ. Complete recovery from intractable complex regional pain syndrome, CRPS-type I, following anesthetic ketamine and midazolam. Pain Pract. 2007;7:147–150.
    1. Stukus KS, Przybylowicz RW, Backes CH, Jr, Cohen DM. Ventricular tachycardia after ketamine sedation for fracture reduction. Pediatr Emerg Care. 2014;30:730–732.
    1. Strayer RJ, Nelson LS. Adverse events associated with ketamine for procedural sedation in adults. Am J Emerg Med. 2008;26:985–1028.
    1. O'Connell NE, Wand BM, McAuley J, Marston L, Moseley GL. Interventions for treating pain and disability in adults with complex regional pain syndrome. Cochrane Database Syst Rev. 2013;30:CD009416.
    1. Correll GE, Maleki J, Gracely EJ, Muir JJ, Harbut RE. Subanesthetic ketamine infusion therapy: a retrospective analysis of a novel therapeutic approach to complex regional pain syndrome. Pain Med. 2004;5:263–275.
    1. American Society of Anesthesiologists. Statement on granting privileges for administration of moderate sedation to practitioners who are not anesthesia professionals (approved by the ASA House of Delegates on October 25, 2005, and reaffirmed on October 26, 2016). Available at: . Accessed July 19, 2017.
    1. Zorumski CF, Conway CR. Use of ketamine in clinical practice: a time for optimism and caution. JAMA Psychiatry. 2017;74:405–406.
    1. Mailis A, Taenzer P. Evidence-based guideline for neuropathic pain interventional treatments: spinal cord stimulation, intravenous infusions, epidural injections and nerve blocks. Pain Res Manag. 2012;17:150–158.
    1. Vollenweider FX, Leenders KL, Oye I, Hell D, Angst J. Differential psychopathology and patterns of cerebral glucose utilisation produced by (S)- and (R)-ketamine in healthy volunteers using positron emission tomography (PET). Eur Neuropsychopharmacol. 1997;7:25–38.
    1. Trivedi S, Kumar R, Tripathi AK, Mehta RK. A comparative study of dexmedetomidine and midazolam in reducing delirium caused by ketamine. J Clin Diagn Res. 2016;10:UC01–UC04.
    1. Krystal JH, Karper LP, Bennett A, et al. Interactive effects of subanesthetic ketamine and subhypnotic lorazepam in humans. Psychopharmacology (Berl). 1998;135:213–229.
    1. Sener S, Eken C, Schultz CH, Serinken M, Ozsarac M. Ketamine with and without midazolam for emergency department sedation in adults: a randomized controlled trial. Ann Emerg Med. 2011;57:109–114.
    1. National Library of Medicine Toxnet. Ketamine casrn:6740-88-1. Available at: . Accessed July 29, 2017.
    1. Dworkin RH, Turk DC, Wyrwich KW, et al. Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations. J Pain. 2008;9:105–121.
    1. Farrar JT, Young JP, Jr, LaMoreaux L, Werth JL, Poole RM. Clinical importance of changes in chronic pain intensity measured on an 11-point numerical pain rating scale. Pain. 2001;94:149–158.
    1. Beilin Y, Hossain S, Bodian CA. The numeric rating scale and labor epidural analgesia. Anesth Analg. 2003;96:1794–1798.
    1. Olsen MF, Bjerre E, Hansen MD, et al. Pain relief that matters to patients: systematic review of empirical studies assessing the minimum clinically important difference in acute pain. BMC Med. 2017;15:35.
    1. Bicket MC, Pasquina PF, Cohen SP. Which regional pain rating best predicts patient-reported improvement in lumbar radiculopathy? Pain Pract. 2017;17:1058–1065.
    1. Turk DC, Dworkin RH, Allen RR, et al. Core outcome domains for chronic pain clinical trials: IMMPACT recommendations. Pain. 2003;106:337–345.
    1. Copay AG, Glassman SD, Subach BR, Berven S, Schuler TC, Carreon LY. Minimum clinically important difference in lumbar spine surgery patients: a choice of methods using the Oswestry Disability Index, Medical Outcomes Study questionnaire Short Form 36, and pain scales. Spine J. 2008;8:968–974.
    1. Cohen SP, Hurley RW, Buckenmaier CC, 3rd, Kurihara C, Morlando B, Dragovich A. Randomized placebo-controlled study evaluating lateral branch radiofrequency denervation for sacroiliac joint pain. Anesthesiology. 2008;109:279–288.
    1. Cohen SP, Bicket MC, Jamison D, Wilkinson I, Rathmell JP. Epidural steroids: a comprehensive, evidence-based review. Reg Anesth Pain Med. 2013;38:175–200.
    1. Fishbain DA, Cole B, Lewis JE, Gao J. What is the evidence that neuropathic pain is present in chronic low back pain and soft tissue syndromes? An evidence-based structured review. Pain Med. 2014;15:4–15.
    1. Liu R, Kurihara C, Tsai HT, Silvestri PJ, Bennett MI, Cohen SP. Classification and treatment of chronic neck pain: a longitudinal cohort study. Reg Anesth Pain Med. 2017;42:52–61.
    1. Littlejohn G, Guymer E. Modulation of NMDA receptor activity in fibromyalgia. Biomedicines. 2017;5:pii:E15.
    1. Sackett DL, Rosenberg WM, Gray JA, Haynes RB, Richardson WS. Evidence based medicine: what it is and what it isn't. BMJ. 1996;312:71–72.
    1. Myers FA, Jr, Bluth MH, Cheung WW. Ketamine: a cause of urinary tract dysfunction. Clin Lab Med. 2016;36:721–744.
    1. Zuccoli ML, Muscella A, Fucile C, et al. Paliperidone for the treatment of ketamine-induced psychosis: a case report. Int J Psychiatry Med. 2014;48:103–108.

Source: PubMed

3
Sottoscrivi