Next Article in Journal
Alternative Arterial Access Routes for Endovascular Thrombectomy in Patients with Acute Ischemic Stroke: A Study from the MR CLEAN Registry
Previous Article in Journal
A Synchronous Robotic Resection of Colorectal Cancer and Liver Metastases—Our Initial Experience
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Narrative Review: Low-Dose Ketamine for Pain Management

by
Alessandro Riccardi
1,
Mario Guarino
2,
Sossio Serra
3,
Michele Domenico Spampinato
4,*,
Simone Vanni
5,
Dana Shiffer
6,
Antonio Voza
7,
Andrea Fabbri
8 and
Fabio De Iaco
9 on behalf of the Study and Research Center of the Italian Society of Emergency Medicine
1
Emergency Department, Ospedale di Imperia, 18100 Imperia, Italy
2
Emergency Department, Centro Traumatologico Ortopedico, Azienda Ospedaliera di Rilievo Nazionale dei Colli, 80131 Napoli, Italy
3
Emergency Department, Maurizio Bufalini Hospital, 47522 Cesena, Italy
4
Department of Translational Medicine and for Romagna, University of Ferrara, 44124 Ferrara, Italy
5
Dipartimento Emergenza e Area Critica, Azienda USL Toscana Centro Struttura Complessa di Medicina d’Urgenza, 50053 Empoli, Italy
6
Emergency Department, Humanitas University, Via Rita Levi Montalcini 4, 20089 Milan, Italy
7
Emergency Department, IRCCS Humanitas Research Hospital, 20089 Milan, Italy
8
Emergency Department, AUSL Romagna, Presidio Ospedaliero Morgagni-Pierantoni, 47121 Forlì, Italy
9
Emergency Department, Ospedale Maria Vittoria, 10144 Turin, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(9), 3256; https://doi.org/10.3390/jcm12093256
Submission received: 13 March 2023 / Revised: 14 April 2023 / Accepted: 28 April 2023 / Published: 2 May 2023
(This article belongs to the Section Emergency Medicine)

Abstract

:
Pain is the leading cause of medical consultations and occurs in 50–70% of emergency department visits. To date, several drugs have been used to manage pain. The clinical use of ketamine began in the 1960s and it immediately emerged as a manageable and safe drug for sedation and anesthesia. The analgesic properties of this drug were first reported shortly after its use; however, its psychomimetic effects have limited its use in emergency departments. Owing to the misuse and abuse of opioids in some countries worldwide, ketamine has become a versatile tool for sedation and analgesia. In this narrative review, ketamine’s role as an analgesic is discussed, with both known and new applications in various contexts (acute, chronic, and neuropathic pain), along with its strengths and weaknesses, especially in terms of psychomimetic, cardiovascular, and hepatic effects. Moreover, new scientific evidence has been reviewed on the use of additional drugs with ketamine, such as magnesium infusion for improving analgesia and clonidine for treating psychomimetic symptoms. Finally, this narrative review was refined by the experience of the Pain Group of the Italian Society of Emergency Medicine (SIMEU) in treating acute and chronic pain with acute manifestations in Italian Emergency Departments.

Graphical Abstract

1. Introduction

Since its introduction in the mid-1960s, ketamine has been a controversial topic. Initially, there was great enthusiasm about its potential uses. However, fears arose that hindered distribution and limited availability. Nevertheless, interest in ketamine has recently increased, especially in emergency medicine, psychiatry, and other professions [1]. Ketamine rapidly induces general anesthesia while preserving the patient’s protective reflexes and vital functions. However, the benefits of ketamine have somewhat diminished over time due to concerns about its psychomimetic effects [1,2]. This drug was originally developed by Craig Newlands and later synthesized by Calvin Stevens in 1962 [3,4]. The aim was to develop a safer and more manageable drug than phencyclidine (PCP), from which it was derived [5,6]. Soon after its development, the analgesic effect of ketamine was discovered. Research in 1965 demonstrated the analgesic effect of ketamine and suggested the use of subdissociative doses of ketamine during painful procedures in children [5,7]. In 1971, the analgesic effect of ketamine was further confirmed when it was observed that patients who underwent anesthesia with ketamine required less opioid medication and experienced better pain management [8,9]. By the mid-1970s, it became clear that different doses of ketamine had different effects [10]. A sub-dissociative dose of 0.6 mg/kg was identified as effective for analgesia, although fixed analgesic doses are still used [11]. The ability of ketamine to produce different effects depending on the dosage is unique among drugs [11]. However, concerns about its side effects have led to a gradual decline in its use [12]. Nevertheless, ketamine has recently experienced a resurgence in emergency departments, and its use is increasing rapidly. It is now used as a sedative, analgesic, antidepressant, and emergency rescue medication for super-refractory status epilepticus [1,13,14]. Due to its profile, ketamine is now considered one of the most important drugs for sedation and analgesia in critically ill patients [15] with haemodynamic instability [16,17].
The most used form of ketamine consists of a racemic mixture of two enantiomers, S-ketamine and R-ketamine, which contain an asymmetric carbon atom [6,18]. The two molecules have different antidepressant, psychomimetic, and analgesic effects [11,19,20,21]. However, this review focuses on racemic mixtures, as data on the specific use of either enantiomer have not been fully established [22]. The analgesic effects of low-dose ketamine are diverse, and its spread is partly due to the opioid crisis in the United States [23,24,25]. Low-dose ketamine has been shown to have an opioid-sparing effect and has been shown to reduce opioid tolerance [24]. Ketamine plays an important role in chronic pain in which the spinal cord is decompressed. In addition to its role as an analgesic in acute pain [26,27], ketamine can reduce hyperalgesia and allodynia in chronic pain [28,29]. Furthermore, there is increasing evidence of its antidepressant effect, which is particularly important in patients with chronic pain [30]. The analgesic role of ketamine metabolites has not been clearly established [18,31,32,33,34,35], but their antidepressant effects seem to be more evident [36]. Ketamine is essential when opioid analgesia is contraindicated or problematic [26], e.g., in elderly patients, patients with OSAS or COPD, patients on chronic opioid therapy, and patients with drug dependence or a history of drug dependence [37]. It is important to emphasise that pain is one of the most common reasons for emergency department visits [38]. The high safety profile of ketamine compared to the many adverse effects of opioids [39] suggests that prescription strategies for acute pain need to be redefined [40].
The objective of this narrative review was to explore the expanding role of ketamine as a unique analgesic drug to define the benefits and possible risks of a therapy gaining great interest [2]. This analysis addresses established and expanding indications for ketamine based on currently available evidence, focusing on areas of uncertainty and potential future developments. Therefore, a review of ketamine’s analgesic role is essential to provide a basis for further studies, as there are no established standards for dosing and administration [41]. An important aspect of the review is the investigation of adverse events, which are sometimes complicated by reports of neurological, bladder, or psychological damage in patients abusing ketamine as a substance. In addition, some studies frequently report various psychomimetic effects of ketamine at sub-dissociative doses, which presents a challenge in standardizing adverse events [42]. Finally, we referred to the experience of the Sedation and Analgesia in Urgency (SAU) group of the Italian Society of Emergency-Urgency Medicine (SIMEU) with the use of low-dose ketamine, which gives an insight into the situation in Italy [43,44].

2. Materials and Methods

To conduct this narrative review, we searched for articles published in English on PubMed Medline (last search 31 January 2023) without any time constraints on the pharmacokinetics, pharmacodynamics, and clinical applications of ketamine. Our search fields included keywords such as “ketamine”, “low dose ketamine”, “sub-dissociative ketamine”, “analgesia”, “pharmacokinetics”, “pharmacodynamics”, “neuropathic pain”, “chronic pain”, “cancer pain”, “opioid-sparing effect”, “opioid tolerance”, “OIH”, “headache”, “adverse effect”, “acute pain”, “infusional”, “antidepressant”, “abuse”, “topical anaesthetic”, and “intranasal”. This review addresses: (i) drug characteristics with emphasis on the antidepressant effect of ketamine and its effect on chronic pain, the interaction between ketamine and magnesium, the relationship between ketamine and the opioid system, and its anti-inflammatory properties; (ii) clinical use of ketamine with emphasis on postoperative pain, chronic and neuropathic pain, cancer-related pain, headache, and the use of ketamine as a local anesthetic; (iii) adverse events and their management; (iv) monitoring of patients treated with low-dose ketamine; (v) risk of abuse; and (vi) contraindications. Studies that addressed the sedative, dissociative, and antidepressant effects of ketamine were excluded.

3. Drug Characteristics

Pharmacokinetics
Ketamine is a highly lipophilic molecule with rapid distribution and immediate passage through the central nervous system. It has low plasma protein binding, ranging from 10% to 50%, an alpha half-life of 2–4 min and a beta half-life of 2–4 h [1,6,44]. Owing to its high liposolubility, it has a large volume of distribution, ranging from 160 to 550 litres [6]. The liver metabolizes ketamine via the cytochromes CYP 2B6 and CYP3A4, producing (R, S)-norketamine, which is converted to 6-hydroxynorketamine and 5,6-dehydronorketamine [45]. These metabolites have an extended half-life of up to 3 days and, according to various authors, provide prolonged analgesic and antidepressant effects, which will be discussed later [32,36,45,46]. Bioavailability and duration of action vary depending on the route of administration: with intravenous administration, bioavailability is 100% and maximum effect is achieved within 1–2 min [39,45,47]; with intramuscular administration, bioavailability is 93% and maximum effect is achieved within 5–10 min [39,45,47]; with oral administration, bioavailability is 16–29% and maximum effect is achieved within 20–120 min [45]. Intranasal administration shows a bioavailability of 35–50% [48,49,50], an analgesic effect with onset of action within 10 min, a time-to-peak effect of 10–14 min [51] and a duration of up to 60 min [52]. Oral administration of ketamine is considered less beneficial because of its lower bioavailability and significant hepatic first-pass effect [6,18]. As ketamine is one of the most commonly abused psychoactive substances worldwide, its oral route is not recommended because of its potential for abuse. The intranasal route of administration has been discussed in a separate section. Ketamine and its metabolites are excreted from the body mainly by the kidneys [45]. Women generally metabolize ketamine more rapidly (up to 20%) than men, whereas older people tend to metabolize it more slowly [45]. Ketamine is contraindicated during pregnancy and lactation [11,24]. Due to its short half-life, no dosage adjustment is required in patients with impaired renal function [29].
Owing to its pharmacokinetic properties, the intravenous route of administration is the most beneficial for acute pain. Ketamine has a faster onset of action than morphine but a shorter half-life [53].
Mechanism of action
The main mechanism of action of ketamine is to block glutamatergic neurons via its antagonistic effect on NMDA receptors [1,44]. It does this by non-competitively blocking the opening of glutamatergic channels, mainly in the prefrontal cortex and hippocampus [4,45]. Ketamine also activates the prefrontal cortex via blockade of inhibitory interneurons, which is one of the mechanisms responsible for its psychomimetic effects [3]. The effect of ketamine on NMDA receptors is unique in that it acts as an open-channel blocker. It blocks the calcium channel only when it is open and has no effect on the closed resting channel [47]. However, the analgesic effects of ketamine are diverse and multifaceted, with effects on dopaminergic [54,55,56], adrenergic [57,58], serotoninergic [59], opioid [58,60] and cholinergic [52,61] receptors. Ketamine acts on the latter by stimulating the nicotinic pathway and inhibiting the muscarinic pathway by blocking M1 receptors, which explains the mydriasis and sialorrhea observed at dissociative doses [52,61]. In addition, ketamine acts on spinal GABA interneurons [62].
Ketamine modulates the reuptake of serotonin, dopamine, and norepinephrine and causes a paradoxical increase in glutamate with stimulation of the descending inhibitory pathways [45,56]. This broad and diverse spectrum of action is essential for its antidepressant effect, which is of particular interest for chronic and neuropathic pain [1]. The blockade of NMDA receptors by ketamine is involved in reducing spinal cord exhaustion, which is a major contributor to the development of chronic pain [4]. Severe pain activates NMDA receptors with hyperexcitability of spinal interneurons in the posterior horn, leading to spinal cord wind-up and central sensitisation [63]. The paradoxical increase in glutamate is essential for the stimulation of medullary GABA inhibitors [45] and for the stimulation of AMPA receptors, which are crucial for the control of depressive symptoms [45,64,65]. Ketamine blocks the NMDA-Rs of GABAergic interneurons, leading to a paradoxical increase in extracellular glutamate and activation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAr), which stimulates the mammalian target of rapamycin complex-1 (mTORC1) signalling pathway, particularly in cortical excitatory pyramidal neurons [66]. AMPA blockade inhibits the antidepressant effect of ketamine [67,68]. Ketamine also has an anti-inflammatory effect by lowering the levels of IL-6 and TNF-alpha [69]. Increased levels of dopamine in the prefrontal, frontal, nucleus striatum, and nucleus accumbens are other causes of psychomimetic symptoms triggered by ketamine, which can mimic schizophrenic symptoms [70,71]. Serotonin also plays an important role in pain modulation, and blockade of 5-HT2 receptors in mouse models reduces the analgesic effect of ketamine [72].
In the past, great importance was placed on the analgesic role of ketamine metabolites, but this has since been revised [36]. However, experimental evidence in animal models suggests that norketamine plays an essential role in hyperpolarizing the HCN channels in the spinal cord and hippocampus, which is particularly important for antidepressant modulation by ketamine [45,46]. However, in animal models, HCN receptors appear to be involved in the analgesic effects [73,74,75,76]. The action of ketamine on opioid receptors does not appear to have a direct analgesic effect but does have a modulatory effect. Direct intrathecal antagonism of mu and delta receptors (but not kappa receptors) blocks the analgesic effect of ketamine, which is not affected by the parenteral administration of naloxone [77]. The effects of ketamine are extensive and are still largely misunderstood. It acts on sigma-1 receptors, L-type voltage-gated calcium channels, and voltage-gated sodium channels, but their exact functions and possible roles in analgesia are not yet known [1,4].
Ketamine reduces the reuptake of catecholamines at the neuronal level, resulting in increased levels of norepinephrine, dopamine, and serotonin, thus increasing the catecholaminergic tone [6]. However, the effects of ketamine on the cardiovascular system remain unclear. For example, ketamine has a negative inotropic effect only in patients with catecholamine deficiency due to direct myocyte blockade, as seen in patients with severe trauma or intensive care [4]. The neuronal interaction of ketamine is even more complex; while ketamine has raised concerns about neuronal damage in animal models [78,79], there is evidence of a neuroprotective effect of ketamine in the presence of acute stress [80,81,82,83]. Indeed, ketamine has been shown to protect against hyperammonemia-induced lethality in acute portosystemic encephalopathy through nitric oxide- and glutamate-mediated neuroprotection, with reduced neuronal oedema [84,85,86,87,88,89]. A similar neuroprotective effect was observed in patients with super-refractory epilepsy [14]. The exact mechanism by which ketamine exerts its neuroprotective effects is not yet fully understood. However, it has been suggested to increase neuronal calcium while inhibiting calmodulin activation, NO synthase, and NO production from L-arginine, resulting in a neuroprotective effect [90].
Ketamine has a broad spectrum of actions and undeniable benefits for the protection of respiratory function. Unlike opiates, ketamine does not induce respiratory depression [1]. In addition, unlike natural opiates such as morphine, which can cause bronchosthenosis in asthmatics [91], ketamine acts as a bronchodilator [18,92,93].
Ketamine’s antidepressant action and its effect on chronic pain
This review will not go into detail about the antidepressant effects of ketamine, but there is an interesting link between pain and depression: functional and neuroimaging studies have shown that ketamine reduces the activity of the insular cortex and thalamus, which are normally activated by pain [11,94,95]. Although the effect of ketamine on NMDAR receptors has not been fully elucidated, some observations have suggested that these receptors play a crucial role in the context of depression and chronic pain. Specifically, ketamine increases neuronal calcium via NMDAr blockade, which causes a secondary decrease in NMDA-R receptors via gene depression, thereby increasing levels of brain-derived neurotrophic factor (BDNF), which are low in mouse models of induced depression and whose levels are increased by ketamine [96]. In addition, ketamine has been shown to decrease receptor affinity for substance P, a neurotransmitter that increases in chronic pain and is one of the mechanisms underlying the loss of medullary pain inhibition [97,98]. In addition, ketamine appears to block acetylcholine muscarinic receptors (m1ChRs), which may also play a role in modulating chronic pain. Studies suggest that agonists of these receptors may increase the pain threshold [61,99]. In addition, animal studies have suggested that ketamine may modulate astrocytic and glial responses that play a role in chronic neuropathic pain [100,101].
As mentioned in the Introduction, the exact analgesic effect of ketamine metabolites is not yet fully understood. However, some studies suggest that these metabolites have analgesic properties equivalent to one-third of the analgesic effect of ketamine [33,34] and tend to accumulate over longer infusion periods, resulting in a sustained analgesic effect over several days [102,103].
The relationship between ketamine, serine, depression and chronic pain is of great interest because NMDA-Rs require D-serine or glycine as co-agonists, especially in neurons in the limbic region involved in the development of depression and chronic pain. Serine racemase produces D-serine in medullary interneurons, and its level increases during neuropathic pain, leading to the activation of NO synthase. Ketamine may also interfere with this level [104,105].
However, it is worth noting that depression and chronic pain are closely linked [106], and a drug that can effectively treat both conditions would be ideal [23,107,108,109]. Indeed, ketamine appears to have a stronger analgesic effect in patients with chronic pain and depression [110]. The first observation of the antidepressant effects of ketamine dates to the 1970s. However, it took many years to gain acceptance in this field [45], and the first major study on this topic was conducted in 2000 [111].
The mechanisms underlying the effects of ketamine are not fully understood. However, there is evidence of a synergistic effect between ketamine and lithium as antidepressants [112]. The paradoxical increase in extraneuronal glutamate by ketamine appears to disinhibit pyramidal neurons, activate AMPAs and TORC1s, increase levels of GABA-B and BdNF, and inhibit glycogen synthase kinase 3 (GSK-3B) in the brain, and lithium also affects GSK-3B. As a result, these two drugs can act synergistically to produce enhanced antidepressant effects [112].
Synergistic effects of ketamine and magnesium
The first observation of the analgesic effects of magnesium and ketamine dates to 1971 [113], and the relationship between magnesium and ketamine is intriguing. Magnesium binds to the NMDAR receptor channel at rest, whereas it does not bind when the channel is active [96]. Magnesium is the body’s NMDA receptor antagonist, and the binding site of ketamine to the same receptor is nearby [96]. The presence of magnesium increases the binding affinity of ketamine for NMDA receptors, and their association with analgesic purposes becomes even more interesting when one considers that brain magnesium levels are reduced in both depression and chronic pain [114]. The addition of 50 mg/kg magnesium to the analgesic ketamine bolus and 10 mg/kg/h magnesium to the ketamine infusion seems to enhance the analgesic efficacy of ketamine [115,116,117,118,119,120,121]. However, it is not yet known how long magnesium infusions should be administered. Magnesium also appears to improve haemodynamic stability [122].
Relationship between ketamine and the opioid system
As mentioned earlier, ketamine has a complex relationship with opioid receptors. By interacting with central and spinal opioid receptors and NMDA-R, it reduces opioid tolerance, opioid-induced hyperalgesia (OIH) and central sensitisation [6,24,63,123,124,125]. Although opiates reduce pain perception by activating mu receptors, they also activate NMDA receptors, leading to postsynaptic hyperexcitability, central tolerance, and sensitization. Ketamine has been shown to modulate and reduce these effects when combined with an NMDA antagonist such as MK-801 [3,126,127,128,129]. In addition, ketamine exerts a downstream effect by increasing opioid-induced phosphorylation of extracellular signal-regulated 1/2 kinase (ERK 1–2), so fewer opioids are required to achieve the desired therapeutic effect (opioid-sparing effect). This also helps reduce adverse events such as respiratory depression and vomiting [39,58,130,131,132,133,134,135].
Ketamine and its anti-inflammatory properties
Pain is an important inflammatory component, particularly in the postoperative setting [45]. Ketamine decreases IL-6, TNF alpha, CRP, and NO synthase levels. High levels of IL-6 are associated with poor postoperative outcomes [69]. Chronic postoperative pain can occur in up to 20% of all surgeries and 50–60% of surgeries involving nerve structures [136]. Postoperative neuropathic pain is caused by activated microglia in the spinal cord [3,69,136,137,138]. Therefore, the effect of ketamine on glial cells [100,101] can alleviate postoperative pain.

4. Clinical Settings

Low-dose ketamine has numerous applications in treating pain, including acute pain in the emergency department and postoperative, chronic, and neuropathic pain [1]. However, the use of ketamine for headaches and neoplastic pain is not well defined [1]. Ketamine is indicated for treating severe pain, but no benefit has been observed for moderate or mild pain [139]. Evidence for the efficacy of low-dose ketamine remains controversial, with studies providing mixed results [1,2,3,4]. Nevertheless, an increasing number of observations of specific receptor effects support the efficacy of ketamine. For example, PET studies have shown high ketamine binding in brain regions with high NMDA receptor density that are directly involved in pain modulation, such as the somatosensory cortex, insula, and anterior cingulate cortex [2,95,140]. The optimal analgesic dosage of ketamine varies widely in the literature, ranging from 0.15 to 0.5 mg/kg [45]. However, doses above 0.3 mg/kg can lead to psychomimetic symptoms, and 0.5 mg/kg is considered a subdissociative dose and is associated with a higher rate of adverse events [28,141,142,143]. As a result, many authors define safe and effective analgesic dosing as 0.15–0.3 mg/kg bolus, 0.15–0.3 mg/kg/h continuous infusion, 0.5–1 mg/kg intramuscular administration, and 1 mg/kg intranasal administration (see Table 1 [45]).
As described in the section on pharmacokinetics, oral administration of ketamine is not standardized, but doses of 0.5 mg/kg every 12 h are considered effective [144]. Other possible routes of administration include transdermal (25 mg/24 h), subcutaneous (0.05–0.15 mg/kg/h), and rectal (10 mg/kg) (Table 1) [145]. As the use of ketamine as an analgesic has increased, it is important to define its precise indications. While many meta-analyses and studies have shown that ketamine has a significant response rate comparable to that of opioids [146], comparison of studies and meta-analyses can be difficult due to differences in dosing, clinical setting, the presence or absence of other concomitant analgesic therapies, and difficulties in assessing and identifying chronic pain due to a lack of standardization [4].
The administration method can be tailored to the clinical setting.
  • isolated, repeatable administration for the treatment of acute pain [53]
  • continuous administration as an infusion, typically over less than 100 h, for the treatment of chronic, neuropathic, or certain forms of acute pain [53].
Continuous infusion administration takes advantage of the increased levels of ketamine metabolites [33,34,102,145,146] and their analgesic and antidepressant properties [45,46]. Although some hypothesise that the response to ketamine decreases with continuous infusions due to tachyphylaxis, there is no strong clinical evidence to support this [147]. Longer infusions (up to 100 h) resulted in a sustained analgesic response of 4 to 8 weeks, while infusions of 12 to 24 h resulted in a reduced but stable response of 7 to 10 days [41].
Intranasal administration at a dose of 1 mg/kg [52,148,149,150,151,152,153,154,155] has been shown to have few and limited side effects [156,157,158,159,160], and its efficacy and safety are comparable to intravenous ketamine therapy. Maximum blood concentration can be measured within 2 min of administration, with a peak at 30 min [159]. However, a possible side effect of intranasal administration is an unpleasant taste [160].
Treatment of acute pain with low-dose ketamine in the emergency department is perhaps the most well-tested indication [161,162,163]. However, meta-analyses have shown significant differences in adverse events when inconsistent and nearly subdissociated doses (0.5 to 0.6 mg/kg) are used [164,165,166], and comparison with other drugs with similar analgesic effects also makes it difficult to draw conclusions [167,168]. Ketamine appears to be an ideal analgesic in the prehospital setting because of its safety profile and the ability to administer it intranasally [169]. However, given the wide range of health profiles in prehospital emergency services, it is difficult to develop a standard of competence and authorisation for the administration of ketamine [170]. In patients with severe trauma, ketamine seems to be the better analgesic as it improves haemodynamic stability [171,172]. It is also useful when opioids are not an option due to hypotension [173,174,175], and it can be used safely in head injuries as it does not increase intracranial pressure [176,177,178,179,180,181,182]. Several studies in the emergency setting have shown that low-dose ketamine is non-inferior to morphine, fentanyl and other opioids [26,38,60,162,183]. The incidence of adverse reactions to ketamine administration varies according to dose and route of administration, but none of the adverse reactions have altered the patient’s course or led to discontinuation of treatment with ketamine [184]. Administration as a bolus is common in the emergency department, but continuous infusion is also possible and beneficial in some situations [185]. Treatment of acute pain in the emergency department must be rapid and effective, improve haemodynamic stability and reduce the risk of chronic pain [186]. Ketamine in analgesic doses appears to meet these criteria [6,141,164,164,165,166,187] and is extremely useful in patients with opioid dependence and tolerance, OIH or buprenorphine, methadone or naltrexone therapy [26] and in patients with exacerbated neuropathic pain [40]. Ketamine is also used to treat severe acute pain in patients with renal dysfunction and critically ill patients with hypoxic or hypercapnic respiratory failure [26,188].
Post-operative pain
The benefit of ketamine in reducing inflammation and IL-6 has not been demonstrated in clinical practice [189,190,191], although some authors have indirectly demonstrated its efficacy by reducing opioid use in the postoperative setting [140,192,193,194,195,196,197]. Due to differences in the surgical setting, procedures, timing of ketamine administration, and dosages, meta-analyses are difficult to compare, and a large study with 8000 participants has not yet been completed [198]. Nonetheless, ketamine’s efficacy as an analgesic for acute pain suggests that it can be used as an alternative to or in addition to opioids while reducing their adverse effects (sedation, respiratory depression, and vomiting). Furthermore, the potential benefits in postoperative management may be an additional factor, particularly in major surgery and especially when the nervous system is involved [64,134,198,199,200].
Chronic and neuropathic pain
The use of ketamine in analgesic doses for the treatment of chronic pain is of interest due to its previously highlighted effects on NMDA-R, the spinal cord, and the glia [3,63,100,101,201,202,203]. Although there is relatively little evidence for it at present, its potential use is very interesting [47]. Chronic pain not only causes severe disability but also imposes significant costs on society, which continue to rise [104,204,205]. The literature on chronic pain is complex due to varying definitions and a lack of standardisation, and the analysis of low-dose ketamine is no exception [34,206,207]. Analgesic-dose ketamine is used in a variety of clinical situations, but the most studied are post-herpetic neuralgia, complex regional pain syndrome type 1 (CRPS-1), fibromyalgia, and axonal neuropathies [103,208,209,210]. However, the strongest indications for its use seem to be chronic neuropathic pain that does not respond to other treatments [205,206,207], such as low back pain with chronic radiculopathy [90,211,212]. On the other hand, its use in trigeminal neuralgia is relatively poorly studied [213].
Chronic pain of any kind is maintained in part by events that ketamine counteracts [138]. These events include hyperactivity and disruption of NMDA-R neurons with central sensitisation [201,214,215,216], loss of descending inhibitory function of interneurons in the spinal cord [217,218], and activation of inflammatory cytokines in the spinal cord with central sensitisation [192,219].
Chronic pain impairs the inhibitory function of the spinal cord, and MRI and electrophysiological studies show that ketamine can help restore this function [140,187]. The modulation of chronic pain by ketamine occurs not only at the level of NMDA-R but also in other pathways that are not yet fully understood [220,221]. Given the close relationship between chronic pain and depression [17,222,223], the antidepressant effect of ketamine appears to play an important role in improving the quality of life of patients with chronic pain [34]. The link between chronic pain and a high risk of suicide should not be forgotten [223,224], and ketamine may modify these risks [90]. Speaking of quality of life, studies do not usually consider the effects (positive or negative) of analgesic therapy on various aspects of a patient’s life, such as driving ability, relationships, and depressed mood [41], which is improved by ketamine thanks to its antidepressant effect [90,200]. Studies suggest that ketamine may be effective in relieving chronic postoperative pain [225], although the best evidence has been observed for the treatment of complex regional pain syndrome [203,226]. Therefore, low-dose ketamine is recommended for complex chronic pain situations as there are few relative contraindications or risks associated with its use [227]. However, it is unclear which patients or types of pain benefit most from an analgesic dose of ketamine [228,229]. Studies suggest that a continuous infusion of ketamine over a longer period is associated with better pain control, even days after administration [103,202,203].
Cancer-related pain
Ketamine may be a viable option for the management of neoplastic pain in patients who cannot tolerate opioids or have opioid-induced hyperalgesia, resistant pain, or neuropathic pain components [230,231]. However, patients with neoplasms have unique clinical features, and the evidence for the use of ketamine in neoplastic pain is limited [47,232]. Nevertheless, the initial findings are promising [100] and deserve further research [233]. Undoubtedly, psychomimetic symptoms can be a major problem for these patients [234]. In response to the opioid crisis, ketamine has been successfully used in analgesic doses in cancer patients and in palliative care in several countries [234,235]. However, finding the right dosage for the individual patient can be challenging, as higher dosages are associated with stronger psychomimetic effects [232,233,234,235,236]. Unlike opioids, where the dosage is increased gradually to achieve the desired effect, ketamine dosage should remain constant to avoid changes in clinical effect [11]. Neoplastic pain is also difficult to manage, and a distinction must be made between palliative treatment and the treatment of active cancer [237]. Intravenous administration is preferred as subcutaneous administration is ineffective [238]. Similar to chronic pain, the antidepressant effect of ketamine is thought to be a key element in its analgesic effect, which may help to control or reduce the disabling symptoms of cancer patients or palliative care patients [239].
Headache
Because of the various clinical pictures [240] and the possible forms of drug abuse [241], the treatment of headaches is particularly complex. Nevertheless, ketamine appears to be effective in controlling migraine [242], although the evidence is not particularly convincing [243]. It is important to note that the timing of migraine treatment is critical, as pathophysiology and therapies vary between the prodromal phase with aura (vasospasm) and the headache phase (vasodilation), where ketamine shows good efficacy [244]. While more studies are needed to fully understand the use of ketamine in analgesic dosing for the treatment of headaches, the initial results are interesting because they offer an additional therapeutic option [245,246].
Ketamine as a local anaesthetic
The idea of using locally administered ketamine to block nerve conduction was proposed as early as 1975, similar to other clinical applications [247]. Later, the direct action of ketamine on sodium channels at peripheral nerve terminals was theorised [248]. Although ketamine does not play a significant role in pain management, the idea of using ketamine to induce local anaesthesia is intriguing and deserves further investigation [249,250,251,252].

5. Adverse Events

Analysis of the literature on the adverse effects of low-dose ketamine is challenging, as most of the reported incidents are due to higher doses used for sedation during procedures or to the persistent abuse of ketamine as an illicit substance [25,26,53]. In general, current evidence suggests that ketamine in analgesic doses has a better safety profile than opioids [26,38,253,254]. Psychomimetic effects occur with the use of ketamine in analgesic doses. They are generally dose-dependent and are most observed at doses above 0.35 mg/kg [3,45] (especially at doses between 0.4 and 0.7 mg/kg) [24,47]. These effects include depersonalisation, conceptual and mental disorganisation, hallucinations, and cognitive or emotional blunting [255,256]. They usually disappear within 30 min after the first dose, especially at high doses, or after the start of a continuous infusion [3]. Longer infusions are less likely to produce these effects [202]. While some studies use high doses (0.5 mg/kg), which already have psychomimetic effects [255], other studies show worsening of schizophrenia symptoms even at doses as low as 0.12 mg/kg [256], which is because of ketamine on prefrontal dopamine receptors [70,71], similar to PCP [47]. However, the contraindication to low-dose ketamine in schizophrenia should be put into perspective, as deterioration of the condition usually resolves within 90 min after the end of ketamine administration or infusion [256]. Interestingly, the psychomimetic effects are less frequent and more intense in individuals with depression who have elevated NMDA-Rs [100,257]. Ketamine may also affect memory, but this is based on abuse studies in which the effect of the drug cannot be distinguished from other confounding factors. This effect does not apply to the use of ketamine in analgesic doses, especially when administered acutely or in isolation, but must be considered in prolonged therapy [45]. Analgesic doses of ketamine are associated with several other side effects, including Ketamine may also influence memory, but this is based on abuse studies where the effect of the drug cannot be distinguished from other confounding factors. This effect does not apply to the use of ketamine in analgesic doses, especially when administered acutely or in isolation, but must be considered in longer-term therapy [45]. Analgesic doses of ketamine are associated with several other side effects, including nystagmus and dizziness, which are among the most common side effects that resolve on their own [45]. Tachycardia and blood pressure elevations are uncommon with analgesic doses and usually resolve on their own within 30 min of administration [3,45]. Diplopia and mydriasis are possible but usually minor and resolve spontaneously, even with prolonged infusion [45]. Vomiting occurs less frequently with ketamine than with opioids [26,53,175], but is possible, especially with intramuscular administration [13,258]. Sialorrhoea is more common in young people when ketamine is administered in dissociative doses. It is uncommon at analgesic doses and can be treated with atropine (0–01 mg/kg) [44]. Although transaminase elevations have never been observed with isolated administration, even in patients with severe hepatopathy [34], they do occur with prolonged infusions of more than 100 h duration [23,39,259,260]. However, normalisation of transaminase levels after discontinuation of therapy is also observed with longer infusions [259].
Oral administration of ketamine is associated with a higher rate of adverse events, particularly psychomimetic effects, especially at the beginning of therapy [261]. This is because initial bioavailability cannot be predicted from patient to patient and thus the effective and safe dose cannot be determined in advance. On the other hand, subcutaneous administration can cause irritation and pain [237].
Deaths directly attributable to the abuse of ketamine are very rare (if traumatic causes are excluded) because the therapeutic range of ketamine is very wide. In rats, the lethal dose of ketamine is well above 40 mg/kg [45], while in humans it is about 60 mg/kg, or 4.2 g in 70 kg subjects [42]. In fact, no medical deaths from ketamine are known in the literature, and non-fatal cases have been reported with the administration of up to 10 times the maximum analgesic dose [4]. Even in paediatric cases treated with an overdose of 5–100 times the therapeutic dissociation dose, no apparent harm occurred, apart from protracted dissociation [257,262].
Other effects reported with chronic abuse do not suggest analgesic use of ketamine, such as erosive cystitis [6,45,47,263,264] or memory impairment and decreased cognitive abilities [4,6].
One of the main concerns limiting the therapeutic use of ketamine is the possibility of neurotoxicity. Olney lesions, i.e., cytoplasmic vacuolar lesions observed in brainstem neurons of rats treated with NMDA-R antagonists such as ketamine, phencyclidine and MK801, have been observed with continuous high-dose infusions above 20 mg/kg [78,79]. However, these lesions regress within 24 h after discontinuation of the infusion [45]. On the other hand, ketamine has been found to have a neuroprotective effect in acute conditions such as head injury [81,82,181], where neuronal damage is mediated by an increase in glutamate, which stimulates NMDA-Rs, leading to a further increase in glutamate [11,265], and ketamine can interrupt this vicious cycle. Ketamine can interrupt this vicious cycle. Ketamine also reduces TNF-alpha-mediated damage in the hippocampus, as occurs in acute inflammation [83,266]. We have already mentioned the neuroprotective role of ketamine in super-refractory status epilepticus [14] and acute portosystemic encephalopathy [84,85,86,87,88,89], but it also seems to exert a neuroprotective effect in ischaemic stroke [267].
In summary, neither animal models nor illicit drug abuse data on ketamine can be used to automatically assess the adverse effects of ketamine when used in analgesic doses. The reason for this is that in the first case, the models and doses are neither consistent nor fully transferable to humans. In addition, numerous confounding factors must be considered when analysing data on ketamine abuse [4,40,268,269]. Meta-analyses are also limited because dosing and definitions of adverse events are not consistent, making it difficult to draw definitive conclusions [270].

6. Management of Adverse Events of Low-Dose Ketamine

Adverse events associated with low-dose ketamine include psychomimetic symptoms, which may be mild or moderate and can be well controlled by benzodiazepines such as diazepam, bromazepam or midazolam [271,272]. These drugs are well tolerated and have a low risk of other adverse events [271,272]. Clonidine, a central alpha-2-adrenergic agonist with sedative effects, has been hypothesised to counteract the central and peripheral dopaminergic stimulus of ketamine and reduce both psychomimetic and vascular symptoms [34,38,202]. The combination of ketamine with dexmedetomidine is also promising [273,274,275,276]. Preparing the patient for possible psychomimetic side effects and managing them in a calm environment can reduce the magnitude of these effects and transform them from unpleasant to acceptable or even pleasant [34]. Inadequate preparation and use of doses greater than 0.35 mg/kg may result in unpleasant dreams for up to 3 days after administration [277]. A thorough knowledge of the specifics of ketamine is therefore essential, as the experience and knowledge of those administering it can reduce the magnitude of adverse events, especially psychomimetic ones [278,279]. Vomiting can be adequately controlled with ondansetron [38].

7. Monitoring the Patient Treated with Low-Dose Ketamine

Ketamine is a dissociative dose that requires continuous monitoring. However, there is no established standard for analgesic dosing. ECG monitoring before and, if necessary, during administration is recommended, as is blood pressure monitoring before and, if necessary, at intervals during treatment. There is currently no evidence for continuous digital saturation monitoring [17].

8. Risk of Abuse

The risk of abuse of ketamine in analgesic doses is generally considered to be low [279,280,281], although the exact risk is not known [282]. Although ketamine is one of the most popular substances of abuse [4,283], it is impossible to infer risk from the literature in this area because of the variation in doses and routes of administration. In addition, data on plasma concentrations in people who abuse ketamine recreationally are lacking [45]. Animal models suggest that continuous administration of ketamine leads to tolerance and dependence [47]. In humans, it leads to tolerance and psychological dependence, but not physical dependence, as discontinuation does not produce withdrawal symptoms [284]. However, given the high social risk, a high threshold of attention to this drug needs to be maintained and its use controlled, especially about the psychological dependence maintained by dopamine [42,100,285].
It is important to note that ketamine is also used to treat and control other addictions. Due to its effect on NDMA-Rs, it can delay the onset of alcohol and heroin withdrawal syndromes, reduce cocaine addiction, and promote neuroplasticity by remodeling pathological neural circuits [286]. Ketamine also blocks and modifies pathologically reinforcing mnemonic circuits and exerts significant antidepressant effects [286]. In addition, ketamine interacts with the opiate system, reduces tolerance, has opioid-sparing effects, reduces OIH and combats dependence [6,24,63,124,125,126]. Given these properties, the potential of ketamine to induce dependence must be evaluated considering its ability to control other addictions [280,281,287].

9. Contraindications

There are some absolute contraindications to the use of ketamine. These include (i) administration within the first three months of life due to possible neuronal damage during development as shown in animal models [202]; (ii) during pregnancy as suggested by research studies [11,24]; (iii) in patients with hepatic porphyria [11]; (iv) and in patients, there are some absolute contraindications to the use of ketamine. These include (i) administration within the first three months of life due to possible neuronal damage during development, as shown in animal models [202]; (ii) during pregnancy, as suggested by research studies [11,24]; (iii) in patients with hepatic porphyria [11]; (iv) and in patients with a known allergy to ketamine [24]. Other contraindications can be derived from the dissociative dosages, but they do not automatically apply to the analgesic dosages [11]. Therefore, these contraindications may be relative rather than absolute [17]. Caution should be exercised in patients with severe and uncontrolled hypertension [3], persistent heart failure [24], psychosis [45] and glaucoma (based on animal models only) [11]. Head trauma is no longer considered a contraindication [179,180,181,182]. However, these contraindications are rarely observed in patients treated with low-dose ketamine [39].
Liver failure must be considered separately. Acute and isolated administration of ketamine does not worsen liver function, even in patients with advanced cirrhosis [34]. However, continuous infusions may cause an increase in transaminases that are self-limiting [282] but require increased caution in patients with cirrhosis [26,39,259,260].

10. Conclusions

Ketamine has been rediscovered by emergency medicine [1] for a variety of indications ranging from procedural sedation [5,258] to rapid and safe immobilisation of patients with psychosis or excited delirium [288]. This is partly due to its ability to exert neuroprotective effects in various situations [289]. The antidepressant effects of ketamine [3,22,290], which have been explored in psychiatry, may also be of undeniable benefit in pain management.
Evidence for the efficacy of low-dose ketamine in various clinical settings is accumulating in the literature [2,3,4,200]. However, the large methodological differences make it difficult to compare studies [172]. For acute pain, non-inferiority to opioids is now well established [162], and ketamine is among the safest options for critically ill patients or those with haemodynamic instability [163]. The ability to administer it intranasally makes it very practical in emergency situations [148].
In chronic and neuropathic pain, the main advantage of ketamine in modulating pain is its simultaneous action on different signalling pathways, although these are not yet fully identified or understood. Ketamine may prevent hyperactivity of the opioid system (leading to tolerance, OIH and central sensitisation), exert an opioid-sparing effect, reduce the pain response, counteract hyperalgesia and allodynia, reduce spinal cord wind-up and restore descending inhibitory pathways. Ketamine in continuous infusions plays an important role as an antidepressant, which is essential for the treatment of patients with chronic pain [2,3,4,162,163,164]. Ketamine is also essential in patients with opiate tolerance or dependence, OIH, dependence on other substances, methadone, buprenorphine, and naltrexone therapy [211,231].
For palliative therapies and the treatment of neoplastic pain, the evidence is still limited, as it is for the treatment of headache, but there is evidence of considerable interest [236].
More extensive studies are needed to define the role of ketamine more precisely. However, the Sedo-Analgesia in Urgenza (SAU) group of the Società Italiana Medicina d’Emergenza-Urgenza (SIMEU) has been using ketamine in analgesic doses for many years, and our experience confirms what is reported in the literature. Ketamine is an excellent drug for the treatment of severe pain in acute cases, but also has remarkable benefits in chronic pain infusions, especially in neuropathic pain such as radiculopathy. Unfortunately, there is currently no clinical evidence to predict individual patient responses. Knowledge of the specifics of the drug and appropriate management are critical to improving the management of adverse events and reducing their prevalence. In our experience, adverse events are kept within limits and rarely limit therapy. Training in analgesic therapy with ketamine is essential for proper use. The SIMEU SAU group has trained more than 5000 physicians and nurses in various areas over the past decade, including low-dose analgesic therapy with ketamine [291]. We have carried out surveys that have shown an increase in the use of ketamine in analgesic doses, and retrospective analyses are underway, and prospective observational studies are planned that will serve to define more precisely the fields of application of ketamine in analgesic doses.

11. Future Directions

While the pharmacodynamic properties of the drug and its increasing use in emergency medicine are encouraging, rigorous research is still needed. Identifying clinical factors that can predict a patient’s response to ketamine will help clinicians determine the most appropriate treatment option. Given the opioid crisis, such studies are more urgent than ever. Future research should also investigate ketamine enantiomers and the development of molecules with more targeted analgesic effects and fewer psychomimetic side effects. Nevertheless, all healthcare providers involved in the treatment of acute, chronic, neuropathic, or neoplastic pain need to be aware of this treatment option and be able to manage its unique side effects. Educational campaigns conducted by scientific societies can be an effective means of achieving this goal quickly and efficiently.

Author Contributions

Conceptualization, A.R., M.G., S.S., M.D.S. and F.D.I.; Methodology, M.D.S. and A.R.; software, D.S.; validation, A.R. and F.D.I.; formal analysis, A.V. and M.D.S.; investigation, A.V. and S.V.; resources, A.F. and M.G.; data curation, A.F. and S.V.; writing—original draft preparation, A.R.; writing—review and editing, A.V., D.S. and M.D.S.; visualization, A.V. and S.V.; supervision, A.R. and F.D.I.; project administration, M.D.S.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

Not Applicable.

Acknowledgments

The authors want to thank the Study and Research center of the Italian Society for Emergency Medicine (SIMEU), Italy, for their support and involvement in this study, and for their collaboration in the infrastructures, consumables and inventoriable material necessary to carry out the study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kohtala, S. Ketamine-50 years in use: From anesthesia to rapid antidepressant effects and neurobiological mechanisms. Pharmacol. Rep. 2021, 73, 323–345. [Google Scholar] [CrossRef]
  2. Persson, J. Ketamine in pain management. CNS Neurosci. Ther. 2013, 19, 396–402. [Google Scholar] [CrossRef] [PubMed]
  3. Subramanian, S.; Haroutounian, S.; Palanca, B.J.A.; Lenze, E.J. Ketamine as a therapeutic agent for depression and pain: Mechanisms and evidence. J. Neurol. Sci. 2022, 434, 120152. [Google Scholar] [CrossRef] [PubMed]
  4. Cohen, S.P.; Liao, W.; Gupta, A.; Plunkett, A. Ketamine in pain management. Adv. Psychosom. Med. 2011, 30, 139–161. [Google Scholar] [CrossRef] [PubMed]
  5. Corssen, G.; Domino, E.F. Dissociative anesthesia: Further pharmacologic studies and first clinical experience with the phencyclidine derivative CI-581. Anesth. Analg. 1966, 45, 29–40. [Google Scholar] [CrossRef] [PubMed]
  6. Peltoniemi, M.A.; Hagelberg, N.M.; Olkkola, K.T.; Saari, T.I. Ketamine: A Review of Clinical Pharmacokinetics and Pharmacodynamics in Anesthesia and Pain Therapy. Clin. Pharmacokinet. 2016, 55, 1059–1077. [Google Scholar] [CrossRef] [PubMed]
  7. Oye, I. Ketamine analgesia, NMDA receptors and the gates of perception. Acta Anaesthesiol. Scand. 1998, 42, 747–749. [Google Scholar] [CrossRef] [PubMed]
  8. Bovill, J.G.; Dundee, J.W. Alterations in response to somatic pain associated with anaesthesia. XX. Ketamine. Br. J Anaesth. 1971, 43, 496–499. [Google Scholar] [CrossRef]
  9. Sadove, M.S.; Shulman, M.; Hatano, S.; Fevold, N. Analgesic effects of ketamine administered in subdissociative doses. Anesth. Analg. 1971, 50, 452–457. [Google Scholar] [CrossRef]
  10. Ito, Y.; Ichiyanagi, K. Post-operative pain relief with ketamine infusion. Anaesthesia 1974, 29, 222–226. [Google Scholar] [CrossRef]
  11. Sinner, B.; Graf, B.M. Ketamine; Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2008; pp. 313–333. [Google Scholar] [CrossRef]
  12. Domino, E.F. Taming the ketamine tiger. 1965. Anesthesiology 2010, 113, 678–684. [Google Scholar] [CrossRef] [PubMed]
  13. Gao, M.; Rejaei, D.; Liu, H. Ketamine use in current clinical practice. Acta Pharmacol. Sin. 2016, 37, 865–872. [Google Scholar] [CrossRef]
  14. Fujikawa, D.G. Neuroprotective effect of ketamine administered after status epilepticus onset. Epilepsia 1995, 36, 186–195. [Google Scholar] [CrossRef] [PubMed]
  15. Erstad, B.L.; Patanwala, A.E. Ketamine for analgosedation in critically ill patients. J. Crit. Care 2016, 35, 145–149. [Google Scholar] [CrossRef] [PubMed]
  16. Hosseinzadeh, H.; Eidy, M.; Golzari, S.E.; Vasebi, M. Hemodynamic Stability during Induction of Anesthesia in ElderlyPatients: Propofol + Ketamine versus Propofol + Etomidate. J. Cardiovasc. Thorac. Res. 2013, 5, 51–54. [Google Scholar] [CrossRef]
  17. Cohen, S.P.; Bhatia, A.; Buvanendran, A.; Schwenk, E.S.; Wasan, A.D.; Hurley, R.W.; Viscusi, E.R.; Narouze, S.; Davis, F.N.; Ritchie, E.C.; et al. 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. Reg. Anesth. Pain Med. 2018, 43, 521–546. [Google Scholar] [CrossRef] [PubMed]
  18. Barrett, W.; Buxhoeveden, M.; Dhillon, S. Ketamine: A versatile tool for anesthesia and analgesia. Curr. Opin. Anaesthesiol. 2020, 33, 633–638. [Google Scholar] [CrossRef]
  19. Jelen, L.A.; Young, A.H.; Stone, J.M. Ketamine: A tale of two enantiomers. J. Psychopharmacol. 2021, 35, 109–123. [Google Scholar] [CrossRef]
  20. Hartvig, P.; Valtysson, J.; Antoni, G.; Westerberg, G.; Långström, B.; Ratti Moberg, E.; Oye, I. Brain kinetics of (R)- and (S)-[N-methyl-11C]ketamine in the rhesus monkey studied by positron emission tomography (PET). Nucl. Med. Biol. 1994, 21, 927–934. [Google Scholar] [CrossRef]
  21. White, P.F.; Ham, J.; Way, W.L.; Trevor, A.J. Pharmacology of ketamine isomers in surgical patients. Anesthesiology 1980, 52, 231–239. [Google Scholar] [CrossRef]
  22. Han, Y.; Li, P.; Miao, M.; Tao, Y.; Kang, X.; Zhang, J. S-ketamine as an adjuvant in patient-controlled intravenous analgesia for preventing postpartum depression: A randomized controlled trial. BMC Anesthesiol. 2022, 22, 49. [Google Scholar] [CrossRef]
  23. Doan, L.V.; Wang, J. An Update on the Basic and Clinical Science of Ketamine Analgesia. Clin. J. Pain 2018, 34, 1077–1088. [Google Scholar] [CrossRef]
  24. Pourmand, A.; Mazer-Amirshahi, M.; Royall, C.; Alhawas, R.; Shesser, R. Low dose ketamine use in the emergency department, a new direction in pain management. Am. J. Emerg. Med. 2017, 35, 918–921. [Google Scholar] [CrossRef] [PubMed]
  25. Crumb, M.W.; Bryant, C.; Atkinson, T.J. Emerging Trends in Pain Medication Management: Back to the Future: A Focus on Ketamine. Am. J. Med. 2018, 131, 883–886. [Google Scholar] [CrossRef] [PubMed]
  26. Balzer, N.; McLeod, S.L.; Walsh, C.; Grewal, K. Low-dose Ketamine for Acute Pain Control in the Emergency Department: A Systematic Review and Meta-analysis. Acad. Emerg. Med. 2021, 28, 444–454. [Google Scholar] [CrossRef]
  27. 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. [Google Scholar] [CrossRef] [PubMed]
  28. Sheikh, S.; Hendry, P. The Expanding Role of Ketamine in the Emergency Department. Drugs 2018, 78, 727–735. [Google Scholar] [CrossRef] [PubMed]
  29. Visser, E.; Schug, S.A. The role of ketamine in pain management. Biomed. Pharmacother. 2006, 60, 341–348. [Google Scholar] [CrossRef]
  30. Paul, R.K.; Singh, N.S.; Khadeer, M.; Moaddel, R.; Sanghvi, M.; Green, C.E.; O’Loughlin, K.; Torjman, M.C.; Bernier, M.; Wainer, I.W. (R,S)-Ketamine metabolites (R,S)-norketamine and (2S,6S)-hydroxynorketamine increase the mammalian target of rapamycin function. Anesthesiology 2014, 121, 149–159. [Google Scholar] [CrossRef]
  31. Kroin, J.S.; Das, V.; Moric, M.; Buvanendran, A. Efficacy of the ketamine metabolite (2R,6R)-hydroxynorketamine in mice models of pain. Reg. Anesth. Pain Med. 2019, 44, 111–117. [Google Scholar] [CrossRef]
  32. Olofsen, E.; Noppers, I.; Niesters, M.; Kharasch, E.; Aarts, L.; Sarton, E.; Dahan, A. Estimation of the contribution of norketamine to ketamine-induced acute pain relief and neurocognitive impairment in healthy volunteers. Anesthesiology 2012, 117, 353–364. [Google Scholar] [CrossRef]
  33. Fanta, S.; Kinnunen, M.; Backman, J.T.; Kalso, E. Population pharmacokinetics of S-ketamine and norketamine in healthy volunteers after intravenous and oral dosing. Eur. J. Clin. Pharmacol. 2015, 71, 441–447. [Google Scholar] [CrossRef] [PubMed]
  34. Niesters, M.; Martini, C.; Dahan, A. Ketamine for chronic pain: Risks and benefits. Br. J. Clin. Pharmacol. 2014, 77, 357–367. [Google Scholar] [CrossRef] [PubMed]
  35. Lilius, T.O.; Viisanen, H.; Jokinen, V.; Niemi, M.; Kalso, E.A.; Rauhala, P.V. Interactions of (2S,6S;2R,6R)-Hydroxynorketamine, a Secondary Metabolite of (R,S)-Ketamine, with Morphine. Basic. Clin. Pharmacol. Toxicol. 2018, 122, 481–488. [Google Scholar] [CrossRef]
  36. Schwenk, E.S.; Pradhan, B.; Nalamasu, R.; Stolle, L.; Wainer, I.W.; Cirullo, M.; Olson, A.; Pergolizzi, J.V.; Torjman, M.C.; Viscusi, E.R. Ketamine in the Past, Present, and Future: Mechanisms, Metabolites, and Toxicity. Curr. Pain Headache Rep. 2021, 25, 57, Erratum in Curr. Pain Headache Rep. 2021, 25, 62. [Google Scholar] [CrossRef]
  37. Hawk, K.; D’Onofrio, G. Emergency department screening and interventions for substance use disorders. Addict. Sci. Clin. Pract. 2019, 13, 18, Erratum in Addict. Sci. Clin. Pract. 2019, 14, 26.. [Google Scholar] [CrossRef] [PubMed]
  38. Miller, J.P.; Schauer, S.G.; Ganem, V.J.; Bebarta, V.S. Low-dose ketamine vs morphine for acute pain in the ED: A randomized controlled trial. Am. J. Emerg. Med. 2015, 33, 402–408. [Google Scholar] [CrossRef]
  39. Davis, W.D.; Davis, K.A.; Hooper, K. The Use of Ketamine for the Management of Acute Pain in the Emergency Department. Adv. Emerg. Nurs. J. 2019, 41, 111–121. [Google Scholar] [CrossRef]
  40. Schwenk, E.S.; Viscusi, E.R.; Buvanendran, A.; Hurley, R.W.; Wasan, A.D.; Narouze, S.; Bhatia, A.; Davis, F.N.; Hooten, W.M.; Cohen, S.P. Consensus Guidelines on the Use of Intravenous Ketamine Infusions for Acute Pain Management from the American Society of Regional Anesthesia and Pain Medicine, the American Academy of Pain Medicine, and the American Society of Anesthesiologists. Reg. Anesth. Pain Med. 2018, 43, 456–466. [Google Scholar] [CrossRef]
  41. Orhurhu, V.; Orhurhu, M.S.; Bhatia, A.; Cohen, S.P. Ketamine Infusions for Chronic Pain: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Anesth. Analg. 2019, 129, 241–254. [Google Scholar] [CrossRef]
  42. Morgan, C.J.; Curran, H.V.; Independent Scientific Committee on Drugs. Ketamine use: A review. Addiction 2012, 107, 27–38. [Google Scholar] [CrossRef]
  43. Di Mascio, A.; Bossini, B.; Barbi, E.; Benini, F.; Cozzi, G. Use of ketamine by paediatricians in Italian paediatric emergency departments: A missed opportunity? Eur. J. Pediatr. 2019, 178, 587–591. [Google Scholar] [CrossRef]
  44. De Iaco, F.; Gandolfo, E.; Guarino, M.; Riccardi, A.; Saggese, M.P.; Serra, S. Terapia Del Dolore in Urgenza E Sedazione Procedurale: Manuale SAU; Independently Published: Traverse, MI, USA, 2022; ISBN 979-8819663554. [Google Scholar]
  45. Zanos, P.; Moaddel, R.; Morris, P.J.; Riggs, L.M.; Highland, J.N.; Georgiou, P.; Pereira, E.F.R.; Albuquerque, E.X.; Thomas, C.J.; Zarate, C.A., Jr.; et al. Ketamine and Ketamine Metabolite Pharmacology: Insights into Therapeutic Mechanisms. Pharmacol. Rev. 2018, 70, 621–660, Erratum in Pharmacol. Rev. 2018, 70, 879. [Google Scholar] [CrossRef]
  46. Shaffer, C.L.; Dutra, J.K.; Tseng, W.C.; Weber, M.L.; Bogart, L.J.; Hales, K.; Pang, J.; Volfson, D.; Am Ende, C.W.; Green, M.E.; et al. Pharmacological evaluation of clinically relevant concentrations of (2R,6R)-hydroxynorketamine. Neuropharmacology 2019, 153, 73–81. [Google Scholar] [CrossRef]
  47. Nowacka, A.; Borczyk, M. Ketamine applications beyond anesthesia—A literature review. Eur. J. Pharmacol. 2019, 860, 172547. [Google Scholar] [CrossRef] [PubMed]
  48. Malinovsky, J.M.; Servin, F.; Cozian, A.; Lepage, J.Y.; Pinaud, M. Ketamine and norketamine plasma concentrations after i.v., nasal and rectal administration in children. Br. J. Anaesth. 1996, 77, 203–207. [Google Scholar] [CrossRef] [PubMed]
  49. Miller, M.; Kruit, N.; Heldreich, C.; Ware, S.; Habig, K.; Reid, C.; Burns, B. Hemodynamic response after rapid sequence induction with ketamine in out-of-hospital patients at risk of shock as defined by the shock index. Ann. Emerg. Med. 2016, 68, 181–188. [Google Scholar] [CrossRef]
  50. Nielsen, B.N.; Friis, S.M.; Rømsing, J.; Schmiegelow, K.; Anderson, B.J.; Ferreirós, N.; Labocha, S.; Henneberg, S. Intranasal sufentanil/ketamine analgesia in children. Paediatr. Anaesth. 2014, 24, 170–180. [Google Scholar] [CrossRef]
  51. Huge, V.; Lauchart, M.; Magerl, W.; Schelling, G.; Beyer, A.; Thieme, D.; Azad, S.C. Effects of low-dose intranasal (S)-ketamine in patients with neuropathic pain. Eur. J. Pain 2010, 14, 387–394. [Google Scholar] [CrossRef] [PubMed]
  52. Carr, D.B.; Goudas, L.C.; Denman, W.T.; Brookoff, D.; Staats, P.S.; Brennen, L.; Green, G.; Albin, R.; Hamilton, D.; Rogers, M.C.; 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. [Google Scholar] [CrossRef] [PubMed]
  53. Ahern, T.L.; Herring, A.A.; Miller, S.; Frazee, B.W. Low-Dose Ketamine Infusion for Emergency Department Patients with Severe Pain. Pain Med. 2015, 16, 1402–1409. [Google Scholar] [CrossRef] [PubMed]
  54. Marcus, D.J.; Bruchas, M.R. Where ketamine and dopamine collide. eLife 2021, 10, e70148. [Google Scholar] [CrossRef] [PubMed]
  55. Kapur, S.; Seeman, P. NMDA receptor antagonists ketamine and PCP have direct effects on the dopamine D(2) and serotonin 5-HT(2)receptors-implications for models of schizophrenia. Mol. Psychiatry. 2002, 7, 837–844. [Google Scholar] [CrossRef] [PubMed]
  56. Seeman, P.; Guan, H.C.; Hirbec, H. Dopamine D2High receptors stimulated by phencyclidines, lysergic acid diethylamide, salvinorin A, and modafinil. Synapse 2009, 63, 698–704. [Google Scholar] [CrossRef]
  57. Thurston, T.A.; Mathew, B.P. In vitro myocardial depression by ketamine or thiopental is dependent on the underlying beta-adrenergic tone. Acta Anaesthesiol. Scand. 1996, 40, 338–341. [Google Scholar] [CrossRef] [PubMed]
  58. Gupta, A.; Devi, L.A.; Gomes, I. Potentiation of μ-opioid receptor-mediated signaling by ketamine. J. Neurochem. 2011, 119, 294–302. [Google Scholar] [CrossRef] [PubMed]
  59. Aleksandrova, L.R.; Phillips, A.G. Neuroplasticity as a convergent mechanism of ketamine and classical psychedelics. Trends Pharmacol. Sci. 2021, 42, 929–942. [Google Scholar] [CrossRef]
  60. Lee, E.N.; Lee, J.H. The Effects of Low-Dose Ketamine on Acute Pain in an Emergency Setting: A Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0165461. [Google Scholar] [CrossRef]
  61. Durieux, M.E. Inhibition by ketamine of muscarinic acetylcholine receptor function. Anesth. Analg. 1995, 81, 57–62. [Google Scholar] [CrossRef]
  62. Gerhard, D.M.; Pothula, S.; Liu, R.J.; Wu, M.; Li, X.Y.; Girgenti, M.J.; Taylor, S.R.; Duman, C.H.; Delpire, E.; Picciotto, M.; et al. GABA interneurons are the cellular trigger for ketamine’s rapid antidepressant actions. J. Clin. Investig. 2020, 130, 1336–1349. [Google Scholar] [CrossRef]
  63. Subramaniam, K.; Subramaniam, B.; Steinbrook, R.A. Ketamine as adjuvant analgesic to opioids: A quantitative and qualitative systematic review. Anesth. Analg. 2004, 99, 482–495. [Google Scholar] [CrossRef]
  64. Matveychuk, D.; Thomas, R.K.; Swainson, J.; Khullar, A.; MacKay, M.A.; Baker, G.B.; Dursun, S.M. Ketamine as an antidepressant: Overview of its mechanisms of action and potential predictive biomarkers. Ther. Adv. Psychopharmacol. 2020, 10, 2045125320916657. [Google Scholar] [CrossRef]
  65. Singh, N.S.; Zarate, C.A., Jr.; Moaddel, R.; Bernier, M.; Wainer, I.W. What is hydroxynorketamine and what can it bring to neurotherapeutics? Expert Rev. Neurother. 2014, 14, 1239–1242. [Google Scholar] [CrossRef] [PubMed]
  66. Miller, O.H.; Moran, J.T.; Hall, B.J. Two cellular hypotheses explaining the initiation of ketamine’s antidepressant actions: Direct inhibition and disinhibition. Neuropharmacology 2016, 100, 17–26. [Google Scholar] [CrossRef] [PubMed]
  67. Zanos, P.; Moaddel, R.; Morris, P.J.; Georgiou, P.; Fischell, J.; Elmer, G.I.; Alkondon, M.; Yuan, P.; Pribut, H.J.; Singh, N.S.; et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature 2016, 533, 481–486. [Google Scholar] [CrossRef]
  68. Highland, J.N.; Zanos, P.; Riggs, L.M.; Georgiou, P.; Clark, S.M.; Morris, P.J.; Moaddel, R.; Thomas, C.J.; Zarate, C.A., Jr.; Pereira, E.F.R.; et al. Hydroxynorketamines: Pharmacology and Potential Therapeutic Applications. Pharmacol. Rev. 2021, 73, 763–791. [Google Scholar] [CrossRef]
  69. Shaked, G.; Czeiger, D.; Dukhno, O.; Levy, I.; Artru, A.A.; Shapira, Y.; Douvdevani, A. Ketamine improves survival and suppresses IL-6 and TNFalpha production in a model of Gram-negative bacterial sepsis in rats. Resuscitation 2004, 62, 237–242. [Google Scholar] [CrossRef] [PubMed]
  70. Kokkinou, M.; Irvine, E.E.; Bonsall, D.R.; Natesan, S.; Wells, L.A.; Smith, M.; Glegola, J.; Paul, E.J.; Tossell, K.; Veronese, M.; et al. Reproducing the dopamine pathophysiology of schizophrenia and approaches to ameliorate it: A translational imaging study with ketamine. Mol. Psychiatry. 2021, 26, 2562–2576. [Google Scholar] [CrossRef]
  71. Kokkinou, M.; Ashok, A.H.; Howes, O.D. The effects of ketamine on dopaminergic function: Meta-analysis and review of the implications for neuropsychiatric disorders. Mol. Psychiatry. 2018, 23, 59–69. [Google Scholar] [CrossRef]
  72. Crisp, T.; Perrotti, J.M.; Smith, D.L.; Stafinsky, J.L.; Smith, D.J. The local monoaminergic dependency of spinal ketamine. Eur. J. Pharmacol. 1991, 194, 167–172. [Google Scholar] [CrossRef]
  73. Takasu, K.; Ono, H.; Tanabe, M. Spinal hyperpolarization-activated cyclic nucleotide-gated cation channels at primary afferent terminals contribute to chronic pain. Pain 2010, 151, 87–96. [Google Scholar] [CrossRef]
  74. Zhang, S.; You, Z.; Wang, S.; Yang, J.; Yang, L.; Sun, Y.; Mi, W.; Yang, L.; McCabe, M.F.; Shen, S.; et al. Neuropeptide S modulates the amygdaloidal HCN activities (Ih) in rats: Implication in chronic pain. Neuropharmacology 2016, 105, 420–433. [Google Scholar] [CrossRef]
  75. Du, L.; Wang, S.J.; Cui, J.; He, W.J.; Ruan, H.Z. The role of HCN channels within the periaqueductal gray in neuropathic pain. Brain Res. 2013, 1500, 36–44. [Google Scholar] [CrossRef]
  76. Du, L.; Wang, S.J.; Cui, J.; He, W.J.; Ruan, H.Z. Inhibition of HCN channels within the periaqueductal gray attenuates neuropathic pain in rats. Behav. Neurosci. 2013, 127, 325–329. [Google Scholar] [CrossRef]
  77. Mikkelsen, S.; Ilkjaer, S.; Brennum, J.; Borgbjerg, F.M.; Dahl, J.B. The effect of naloxone on ketamine-induced effects on hyperalgesia and ketamine-induced side effects in humans. Anesthesiology 1999, 90, 1539–1545. [Google Scholar] [CrossRef] [PubMed]
  78. Olney, J.W.; Labruyere, J.; Price, M.T. Pathological changes induced in cerebrocortical neurons by phencyclidine and related drugs. Science 1989, 244, 1360–1362. [Google Scholar] [CrossRef] [PubMed]
  79. Olney, J.W.; Labruyere, J.; Wang, G.; Wozniak, D.F.; Price, M.T.; Sesma, M.A. NMDA antagonist neurotoxicity: Mechanism and prevention. Science 1991, 254, 1515–1518. [Google Scholar] [CrossRef] [PubMed]
  80. Yan, J.; Li, Y.R.; Zhang, Y.; Lu, Y.; Jiang, H. Repeated exposure to anesthetic ketamine can negatively impact neurodevelopment in infants: A prospective preliminary clinical study. J. Child Neurol. 2014, 29, 1333–1338. [Google Scholar] [CrossRef] [PubMed]
  81. Church, J.; Zeman, S.; Lodge, D. The neuroprotective action of ketamine and MK-801 after transient cerebral ischemia in rats. Anesthesiology 1988, 69, 702–709. [Google Scholar] [CrossRef]
  82. Spandou, E.; Karkavelas, G.; Soubasi, V.; Avgovstides-Savvopoulou, P.; Loizidis, T.; Guiba-Tziampiri, O. Effect of ketamine on hypoxic-ischemic brain damage in newborn rats. Brain Res. 1999, 819, 1–7. [Google Scholar] [CrossRef]
  83. Wang, L.; Deng, B.; Yan, P.; Wu, H.; Li, C.; Zhu, H.; Du, J.; Hou, L. Neuroprotective effect of ketamine against TNF-α-induced necroptosis in hippocampal neurons. J. Cell. Mol. Med. 2021, 25, 3449–3459. [Google Scholar] [CrossRef] [PubMed]
  84. Monfort, P.; Kosenko, E.; Erceg, S.; Canales, J.J.; Felipo, V. Molecular mechanism of acute ammonia toxicity: Role of NMDA receptors. Neurochem. Int. 2002, 41, 95–102. [Google Scholar] [CrossRef] [PubMed]
  85. Chan, P.H.; Chu, L. Ketamine protects cultured astrocytes from glutamate-induced swelling. Brain Res. 1989, 487, 380–383. [Google Scholar] [CrossRef]
  86. Rodrigo, R.; Cauli, O.; Boix, J.; ElMlili, N.; Agusti, A.; Felipo, V. Role of NMDA receptors in acute liver failure and ammonia toxicity: Therapeutical implications. Neurochem. Int. 2009, 55, 113–118. [Google Scholar] [CrossRef] [PubMed]
  87. Cauli, O.; Rodrigo, R.; Boix, J.; Piedrafita, B.; Agusti, A.; Felipo, V. Acute liver failure-induced death of rats is delayed or prevented by blocking NMDA receptors in brain. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 295, G503–G511. [Google Scholar] [CrossRef]
  88. Boscán, P.L.; Piña-Crespo, J.C.; Daló, N.L. Ketamine reduces lethality on the acute ammonia intoxication in mice. Investig. Clin. 1996, 37, 129–135. [Google Scholar]
  89. Romero, T.R.; Galdino, G.S.; Silva, G.C.; Resende, L.C.; Perez, A.C.; Côrtes, S.F.; Duarte, I.D. Ketamine activates the L-arginine/Nitric oxide/cyclic guanosine monophosphate pathway to induce peripheral antinociception in rats. Anesth. Analg. 2011, 113, 1254–1259. [Google Scholar] [CrossRef]
  90. Kamp, J.; Van Velzen, M.; Olofsen, E.; Boon, M.; Dahan, A.; Niesters, M. Pharmacokinetic and pharmacodynamic considerations for NMDA-receptor antagonist ketamine in the treatment of chronic neuropathic pain: An update of the most recent literature. Expert Opin. Drug Metab. Toxicol. 2019, 15, 1033–1041. [Google Scholar] [CrossRef]
  91. Hlavaty, L.; Hansma, P.; Sung, L. Contribution of opiates in sudden asthma deaths. Am. J. Forensic Med. Pathol. 2015, 36, 49–52. [Google Scholar] [CrossRef]
  92. Kurdi, M.S.; Theerth, K.A.; Deva, R.S. Ketamine: Current applications in anesthesia, pain, and critical care. Anesth. Essays Res. 2014, 8, 283–290. [Google Scholar] [CrossRef]
  93. Rehder, K.J. Adjunct Therapies for Refractory Status Asthmaticus in Children. Respir. Care. 2017, 62, 849–865. [Google Scholar] [CrossRef] [PubMed]
  94. Rogers, R.; Wise, R.G.; Painter, D.J.; Longe, S.E.; Tracey, I. An investigation to dissociate the analgesic and anesthetic properties of ketamine using functional magnetic resonance imaging. Anesthesiology 2004, 100, 292–301. [Google Scholar] [CrossRef]
  95. Sprenger, T.; Valet, M.; Woltmann, R.; Zimmer, C.; Freynhagen, R.; Kochs, E.F.; Tölle, T.R.; Wagner, K.J. Imaging pain modulation by subanesthetic S-(+)-ketamine. Anesth. Analg. 2006, 103, 729–737. [Google Scholar] [CrossRef]
  96. Iacobucci, G.J.; Visnjevac, O.; Pourafkari, L.; Nader, N.D. Ketamine: An Update on Cellular and Subcellular Mechanisms with Implications for Clinical Practice. Pain Physician 2017, 20, E285–E301. [Google Scholar] [PubMed]
  97. Abbadie, C.; Brown, J.L.; Mantyh, P.W.; Basbaum, A.I. Spinal cord substance P receptor immunoreactivity increases in both inflammatory and nerve injury models of persistent pain. Neuroscience 1996, 70, 201–209. [Google Scholar] [CrossRef] [PubMed]
  98. Okamoto, T.; Minami, K.; Uezono, Y.; Ogata, J.; Shiraishi, M.; Shigematsu, A.; Ueta, Y. The inhibitory effects of ketamine and pentobarbital on substance p receptors expressed in Xenopus oocytes. Anesth. Analg. 2003, 97, 104–110. [Google Scholar] [CrossRef] [PubMed]
  99. Bartolini, A.; Ghelardini, C.; Fantetti, L.; Malcangio, M.; Malmberg-Aiello, P.; Giotti, A. Role of muscarinic receptor subtypes in central antinociception. Br. J. Pharmacol. 1992, 105, 77–82. [Google Scholar] [CrossRef]
  100. Culp, C.; Kim, H.K.; Abdi, S. Ketamine Use for Cancer and Chronic Pain Management. Front. Pharmacol. 2021, 11, 599721. [Google Scholar] [CrossRef] [PubMed]
  101. Mika, J.; Osikowicz, M.; Rojewska, E.; Korostynski, M.; Wawrzczak-Bargiela, A.; Przewlocki, R.; Przewlocka, B. Differential activation of spinal microglial and astroglial cells in a mouse model of peripheral neuropathic pain. Eur. J. Pharmacol. 2009, 623, 65–72. [Google Scholar] [CrossRef]
  102. Sigtermans, M.J.; van Hilten, J.J.; Bauer, M.C.R.; Arbous, S.M.; Marinus, J.; Sarton, E.Y.; Dahan, A. Ketamine produces effective and long-term pain relief in patients with Complex Regional Pain Syndrome Type 1. Pain 2009, 145, 304–311. [Google Scholar] [CrossRef]
  103. Sigtermans, M.; Dahan, A.; Mooren, R.; Bauer, M.; Kest, B.; Sarton, E.; Olofsen, E. S(+)-ketamine effect on experimental pain and cardiac output: A population pharmacokinetic-pharmacodynamic modeling study in healthy volunteers. Anesthesiology 2009, 111, 892–903. [Google Scholar] [CrossRef] [PubMed]
  104. Lefèvre, Y.; Amadio, A.; Vincent, P.; Descheemaeker, A.; Oliet, S.H.; Dallel, R.; Voisin, D.L. Neuropathic pain depends upon D-serine co-activation of spinal NMDA receptors in rats. Neurosci. Lett. 2015, 603, 42–47. [Google Scholar] [CrossRef] [PubMed]
  105. Choi, S.R.; Roh, D.H.; Yoon, S.Y.; Choi, H.S.; Kang, S.Y.; Han, H.J.; Beitz, A.J.; Lee, J.H. Astrocyte D-serine modulates the activation of neuronal NOS leading to the development of mechanical allodynia in peripheral neuropathy. Mol. Pain 2019, 15, 1744806919843046. [Google Scholar] [CrossRef]
  106. Sheng, J.; Liu, S.; Wang, Y.; Cui, R.; Zhang, X. The Link between Depression and Chronic Pain: Neural Mechanisms in the Brain. Neural. Plast. 2017, 2017, 9724371. [Google Scholar] [CrossRef]
  107. Nicholas, M.K.; Coulston, C.M.; Asghari, A.; Malhi, G.S. Depressive symptoms in patients with chronic pain. Med. J. Aust. 2009, 190, S66–S70. [Google Scholar] [CrossRef]
  108. Lerman, S.F.; Rudich, Z.; Brill, S.; Shalev, H.; Shahar, G. Longitudinal associations between depression, anxiety, pain, and pain-related disability in chronic pain patients. Psychosom. Med. 2015, 77, 333–341. [Google Scholar] [CrossRef] [PubMed]
  109. Wang, J.; Goffer, Y.; Xu, D.; Tukey, D.S.; Shamir, D.B.; Eberle, S.E.; Zou, A.H.; Blanck, T.J.; Ziff, E.B. A single subanesthetic dose of ketamine relieves depression-like behaviors induced by neuropathic pain in rats. Anesthesiology 2011, 115, 812–821. [Google Scholar] [CrossRef]
  110. Zhou, Y.; Wang, C.; Lan, X.; Li, H.; Chao, Z.; Ning, Y. Plasma inflammatory cytokines and treatment-resistant depression with comorbid pain: Improvement by ketamine. J. Neuroinflammation 2021, 18, 200. [Google Scholar] [CrossRef] [PubMed]
  111. Berman, R.M.; Cappiello, A.; Anand, A.; Oren, D.A.; Heninger, G.R.; Charney, D.S.; Krystal, J.H. Antidepressant effects of ketamine in depressed patients. Biol. Psychiatry 2000, 47, 351–354. [Google Scholar] [CrossRef]
  112. Scheuing, L.; Chiu, C.T.; Liao, H.M.; Chuang, D.M. Antidepressant mechanism of ketamine: Perspective from preclinical studies. Front. Neurosci. 2015, 9, 249. [Google Scholar] [CrossRef]
  113. Weisman, H. Anesthesia for pediatric ophthalmology. Ann. Ophthalmol. 1971, 3, 229–232. [Google Scholar] [PubMed]
  114. Kotermanski, S.E.; Johnson, J.W. Mg2+ imparts NMDA receptor subtype selectivity to the Alzheimer’s drug memantine. J. Neurosci. 2009, 29, 2774–2779. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, H.T.; Hollmann, M.W.; Liu, W.H.; Hoenemann, C.W.; Durieux, M.E. Modulation of NMDA receptor function by ketamine and magnesium: Part I. Anesth. Analg. 2001, 92, 1173–1181. [Google Scholar] [CrossRef] [PubMed]
  116. Hollmann, M.W.; Liu, H.T.; Hoenemann, C.W.; Liu, W.H.; Durieux, M.E. Modulation of NMDA receptor function by ketamine and magnesium. Part II: Interactions with volatile anesthetics. Anesth. Analg. 2001, 92, 1182–1191. [Google Scholar] [CrossRef] [PubMed]
  117. Savic Vujovic, K.R.; Vuckovic, S.; Srebro, D.; Medic, B.; Stojanovic, R.; Vucetic, C.; Prostran, M. A synergistic interaction between magnesium sulphate and ketamine on the inhibition of acute nociception in rats. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 2503–2509. [Google Scholar]
  118. Varas, V.; Bertinelli, P.; Carrasco, P.; Souper, N.; Álvarez, P.; Danilla, S.; Egaña, J.I.; Penna, A.; Sepúlveda, S.; Arancibia, V.; et al. Intraoperative Ketamine and Magnesium Therapy to Control Postoperative Pain After Abdominoplasty and/or Liposuction: A Clinical Randomized Trial. J. Pain Res. 2020, 13, 2937–2946. [Google Scholar] [CrossRef]
  119. Crosby, V.; Wilcock, A.; Corcoran, R. The safety and efficacy of a single dose (500 mg or 1 g) of intravenous magnesium sulfate in neuropathic pain poorly responsive to strong opioid analgesics in patients with cancer. J. Pain Symptom Manag. 2000, 19, 35–39. [Google Scholar] [CrossRef]
  120. Begon, S.; Pickering, G.; Eschalier, A.; Dubray, C. Magnesium and MK-801 have a similar effect in two experimental models of neuropathic pain. Brain Res. 2000, 887, 436–439. [Google Scholar] [CrossRef]
  121. Delage, N.; Morel, V.; Picard, P.; Marcaillou, F.; Pereira, B.; Pickering, G. Effect of ketamine combined with magnesium sulfate in neuropathic pain patients (KETAPAIN): Study protocol for a randomized controlled trial. Trials 2017, 18, 517. [Google Scholar] [CrossRef]
  122. Forget, P.; Cata, J. Stable anesthesia with alternative to opioids: Are ketamine and magnesium helpful in stabilizing hemodynamics during surgery? A systematic review and meta-analyses of randomized controlled trials. Best Pr. Res. Clin. Anaesthesiol. 2017, 31, 523–531. [Google Scholar] [CrossRef]
  123. Mak, P.; Broadbear, J.H.; Kolosov, A.; Goodchild, C.S. 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. [Google Scholar] [CrossRef] [PubMed]
  124. Lilius, T.O.; Jokinen, V.; Neuvonen, M.S.; Niemi, M.; Kalso, E.A.; Rauhala, P.V. Ketamine coadministration attenuates morphine tolerance and leads to increased brain concentrations of both drugs in the rat. Br. J. Pharmacol. 2015, 172, 2799–2813. [Google Scholar] [CrossRef] [PubMed]
  125. Klein, T.; Magerl, W.; Nickel, U.; Hopf, H.C.; Sandkühler, J.; Treede, R.D. Effects of the NMDA-receptor antagonist ketamine on perceptual correlates of long-term potentiation within the nociceptive system. Neuropharmacology 2007, 52, 655–661. [Google Scholar] [CrossRef] [PubMed]
  126. Mao, J.; Price, D.D.; Mayer, D.J. Mechanisms of hyperalgesia and morphine tolerance: A current view of their possible interactions. Pain 1995, 62, 259–274. [Google Scholar] [CrossRef] [PubMed]
  127. Dickenson, A.H. NMDA receptor antagonists: Interactions with opioids. Acta Anaesthesiol. Scand. 1997, 41 Pt 2, 112–115. [Google Scholar] [CrossRef] [PubMed]
  128. Laulin, J.P.; Maurette, P.; Corcuff, J.B.; 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. [Google Scholar] [CrossRef]
  129. Trujillo, K.A.; Akil, H. Inhibition of opiate tolerance by non-competitive N-methyl-D-aspartate receptor antagonists. Brain Res. 1994, 633, 178–188. [Google Scholar] [CrossRef]
  130. Riddell, J.M.; Trummel, J.M.; Onakpoya, I.J. Low-dose ketamine in painful orthopaedic surgery: A systematic review and meta-analysis. Br. J. Anaesth. 2019, 123, 325–334. [Google Scholar] [CrossRef]
  131. Bi, Y.; Ye, Y.; Zhu, Y.; Ma, J.; Zhang, X.; Liu, B. The Effect of Ketamine on Acute and Chronic Wound Pain in Patients Undergoing Breast Surgery: A Meta-Analysis and Systematic Review. Pain Pract. 2021, 21, 316–332. [Google Scholar] [CrossRef]
  132. 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. [Google Scholar] [CrossRef]
  133. Brinck, E.C.; Tiippana, E.; Heesen, M.; Bell, R.F.; Straube, S.; Moore, R.A.; Kontinen, V. Perioperative intravenous ketamine for acute postoperative pain in adults. Cochrane Database Syst. Rev. 2018, 12, CD012033. [Google Scholar] [CrossRef] [PubMed]
  134. Gorlin, A.W.; Rosenfeld, D.M.; Ramakrishna, H. Intravenous sub-anesthetic ketamine for perioperative analgesia. J. Anaesthesiol. Clin. Pharmacol. 2016, 32, 160–167. [Google Scholar] [CrossRef]
  135. Radvansky, B.M.; Shah, K.; Parikh, A.; Sifonios, A.N.; Le, V.; Eloy, J.D. Role of ketamine in acute postoperative pain management: A narrative review. Biomed Res. Int. 2015, 2015, 749837. [Google Scholar] [CrossRef] [PubMed]
  136. Yu, M.; Shao, D.; Yang, R.; Feng, X.; Zhu, S.; Xu, J. Effects of ketamine on pulmonary inflammatory responses and survival in rats exposed to polymicrobial sepsis. J. Pharm. Pharm. Sci. 2007, 10, 434–442. [Google Scholar] [CrossRef] [PubMed]
  137. Hayashi, Y.; Kawaji, K.; Sun, L.; Zhang, X.; Koyano, K.; Yokoyama, T.; Kohsaka, S.; Inoue, K.; Nakanishi, H. Microglial Ca(2+)-activated K(+) channels are possible molecular targets for the analgesic effects of S-ketamine on neuropathic pain. J. Neurosci. 2011, 31, 17370–17382. [Google Scholar] [CrossRef]
  138. Watkins, L.R.; Milligan, E.D.; Maier, S.F. Glial activation: A driving force for pathological pain. Trends Neurosci. 2001, 24, 450–455. [Google Scholar] [CrossRef]
  139. Laskowski, K.; Stirling, A.; McKay, W.P.; Lim, H.J. A systematic review of intravenous ketamine for postoperative analgesia. Can. J. Anaesth. 2011, 58, 911–923. [Google Scholar] [CrossRef] [PubMed]
  140. Niesters, M.; Khalili-Mahani, N.; Martini, C.; Aarts, L.; van Gerven, J.; van Buchem, M.A.; Dahan, A.; Rombouts, S. Effect of subanesthetic ketamine on intrinsic functional brain connectivity: A placebo-controlled functional magnetic resonance imaging study in healthy male volunteers. Anesthesiology 2012, 117, 868–877. [Google Scholar] [CrossRef]
  141. Weinbroum, A.A. A single small dose of postoperative ketamine provides rapid and sustained improvement in morphine analgesia in the presence of morphine-resistant pain. Anesth. Analg. 2003, 96, 789–795. [Google Scholar] [CrossRef]
  142. Nesher, N.; Serovian, I.; Marouani, N.; Chazan, S.; Weinbroum, A.A. Ketamine spares morphine consumption after transthoracic lung and heart surgery without adverse hemodynamic effects. Pharmacol. Res. 2008, 58, 38–44. [Google Scholar] [CrossRef]
  143. Hartvig, P.; Larsson, E.; Joachimsson, P.O. Postoperative analgesia and sedation following pediatric cardiac surgery using a constant infusion of ketamine. J. Cardiothorac. Vasc. Anesth. 1993, 7, 148–153. [Google Scholar] [CrossRef] [PubMed]
  144. Lauretti, G.R.; Lima, I.C.; Reis, M.P.; Prado, W.A.; Pereira, N.L. Oral ketamine and transdermal nitroglycerin as analgesic adjuvants to oral morphine therapy for cancer pain management. Anesthesiology 1999, 90, 1528–1533. [Google Scholar] [CrossRef] [PubMed]
  145. Velzen, M.V.; Dahan, J.D.C.; van Dorp, E.L.A.; Mogil, J.S.; Hooijmans, C.R.; Dahan, A. Efficacy of ketamine in relieving neuropathic pain: A systematic review and meta-analysis of animal studies. Pain 2021, 162, 2320–2330. [Google Scholar] [CrossRef] [PubMed]
  146. Backonja, M.; Arndt, G.; Gombar, K.A.; Check, B.; Zimmermann, M. Response of chronic neuropathic pain syndromes to ketamine: A preliminary study. Pain 1994, 56, 51–57, Erratum in Pain 1994, 58, 433. [Google Scholar] [CrossRef] [PubMed]
  147. Livingston, A.; Waterman, A.E. The development of tolerance to ketamine in rats and the significance of hepatic metabolism. Br. J. Pharmacol. 1978, 64, 63–69. [Google Scholar] [CrossRef]
  148. Shimonovich, S.; Gigi, R.; Shapira, A.; Sarig-Meth, T.; Nadav, D.; Rozenek, M.; West, D.; Halpern, P. Intranasal ketamine for acute traumatic pain in the Emergency Department: A prospective, randomized clinical trial of efficacy and safety. BMC Emerg. Med. 2016, 16, 43. [Google Scholar] [CrossRef] [PubMed]
  149. Holdgate, A.; Cao, A.; Lo, K.M. The implementation of intranasal fentanyl for children in a mixed adult and pediatric emergency department reduces time to analgesic administration. Acad. Emerg. Med. 2010, 17, 214–217. [Google Scholar] [CrossRef] [PubMed]
  150. Bailey, A.M.; Baum, R.A.; Horn, K.; Lewis, T.; Morizio, K.; Schultz, A.; Weant, K.; Justice, S.N. Review of Intranasally Administered Medications for Use in the Emergency Department. J. Emerg. Med. 2017, 53, 38–48. [Google Scholar] [CrossRef]
  151. Del Pizzo, J.; Callahan, J.M. Intranasal medications in pediatric emergency medicine. Pediatr. Emerg. Care 2014, 30, 496–501; quiz 502–504. [Google Scholar] [CrossRef]
  152. Li, X.; Hua, G.C.; Peng, F. Efficacy of intranasal ketamine for acute pain management in adults: A systematic review and meta-analysis. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 3286–3295. [Google Scholar] [CrossRef]
  153. Poonai, N.; Canton, K.; Ali, S.; Hendrikx, S.; Shah, A.; Miller, M.; Joubert, G.; Rieder, M.; Hartling, L. Intranasal ketamine for procedural sedation and analgesia in children: A systematic review. PLoS ONE 2017, 12, e0173253. [Google Scholar] [CrossRef]
  154. Whitley, G.A.; Pilbery, R. Pre-hospital intranasal analgesia for children suffering pain: A rapid evidence review. Br. Paramed. J. 2019, 4, 24–34. [Google Scholar] [CrossRef] [PubMed]
  155. Oliveira, J.E.; Silva, L.; Lee, J.Y.; Bellolio, F.; Homme, J.L.; Anderson, J.L. Intranasal ketamine for acute pain management in children: A systematic review and meta-analysis. Am. J. Emerg. Med. 2020, 38, 1860–1866. [Google Scholar] [CrossRef]
  156. Frey, T.M.; Florin, T.A.; Caruso, M.; Zhang, N.; Zhang, Y.; Mittiga, M.R. Effect of Intranasal Ketamine vs Fentanyl on Pain Reduction for Extremity Injuries in Children: The PRIME Randomized Clinical Trial. JAMA Pediatr. 2019, 173, 140–146. [Google Scholar] [CrossRef] [PubMed]
  157. Rocchio, R.J.; Ward, K.E. Intranasal Ketamine for Acute Pain. Clin. J. Pain 2021, 37, 295–300. [Google Scholar] [CrossRef] [PubMed]
  158. Ferguson, C.L.; Beckett, R.D. Intranasal Ketamine for Treatment of Acute Pain in Pediatrics: A Systematic Review. Pediatr. Emerg. Care 2020, 36, e476–e481. [Google Scholar] [CrossRef] [PubMed]
  159. Andolfatto, G.; Willman, E.; Joo, D.; Miller, P.; Wong, W.B.; Koehn, M.; Dobson, R.; Angus, E.; Moadebi, S. Intranasal ketamine for analgesia in the emergency department: A prospective observational series. Acad. Emerg. Med. 2013, 20, 1050–1054. [Google Scholar] [CrossRef]
  160. Reynolds, S.L.; Bryant, K.K.; Studnek, J.R.; Hogg, M.; Dunn, C.; Templin, M.A.; Moore, C.G.; Young, J.R.; Walker, K.R.; Runyon, M.S. Randomized Controlled Feasibility Trial of Intranasal Ketamine Compared to Intranasal Fentanyl for Analgesia in Children with Suspected Extremity Fractures. Acad. Emerg. Med. 2017, 24, 1430–1440. [Google Scholar] [CrossRef]
  161. Beaudoin, F.L.; Lin, C.; Guan, W.; Merchant, R.C. Low-dose ketamine improves pain relief in patients receiving intravenous opioids for acute pain in the emergency department: Results of a randomized, double-blind, clinical trial. Acad. Emerg. Med. 2014, 21, 1193–1202. [Google Scholar] [CrossRef]
  162. Motov, S.; Rosenbaum, S.; Vilke, G.M.; Nakajima, Y. Is There a Role for Intravenous Subdissociative-Dose Ketamine Administered as an Adjunct to Opioids or as a Single Agent for Acute Pain Management in the Emergency Department? J. Emerg. Med. 2016, 51, 752–757. [Google Scholar] [CrossRef]
  163. Duhaime, M.J.; Wolfson, A.B. Ketamine Versus Opioids for Acute Pain in the Emergency Department. Acad. Emerg. Med. 2020, 27, 781–782. [Google Scholar] [CrossRef]
  164. Karlow, N.; Schlaepfer, C.H.; Stoll, C.R.T.; Doering, M.; Carpenter, C.R.; Colditz, G.A.; Motov, S.; Miller, J.; Schwarz, E.S. A Systematic Review and Meta-analysis of Ketamine as an Alternative to Opioids for Acute Pain in the Emergency Department. Acad. Emerg. Med. 2018, 25, 1086–1097. [Google Scholar] [CrossRef] [PubMed]
  165. Ghate, G.; Clark, E.; Vaillancourt, C. Systematic review of the use of low-dose ketamine for analgesia in the emergency department. Can. J. Emerg. Med. 2018, 20, 36–45. [Google Scholar] [CrossRef] [PubMed]
  166. Sin, B.; Ternas, T.; Motov, S.M. The use of subdissociative-dose ketamine for acute pain in the emergency department. Acad. Emerg. Med. 2015, 22, 251–257. [Google Scholar] [CrossRef] [PubMed]
  167. Gurnani, A.; Sharma, P.K.; Rautela, R.S.; Bhattacharya, A. Analgesia for acute musculoskeletal trauma: Low-dose subcutaneous infusion of ketamine. Anaesth. Intensive Care. 1996, 24, 32–36. [Google Scholar] [CrossRef]
  168. Jennings, P.A.; Cameron, P.; Bernard, S. Ketamine as an analgesic in the pre-hospital setting: A systematic review. Acta Anaesthesiol. Scand. 2011, 55, 638–643. [Google Scholar] [CrossRef]
  169. Bansal, A.; Miller, M.; Ferguson, I.; Burns, B. Ketamine as a Prehospital Analgesic: A Systematic Review. Prehosp. Disaster Med. 2020, 35, 314–321. [Google Scholar] [CrossRef]
  170. Sandberg, M.; Hyldmo, P.K.; Kongstad, P.; Dahl Friesgaard, K.; Raatiniemi, L.; Larsen, R.; Magnusson, V.; Rognås, L.; Kurola, J.; Rehn, M.; et al. Ketamine for the treatment of prehospital acute pain: A systematic review of benefit and harm. BMJ Open 2020, 10, e038134. [Google Scholar] [CrossRef]
  171. Bredmose, P.P.; Grier, G.; Davies, G.E.; Lockey, D.J. Pre-hospital use of ketamine in paediatric trauma. Acta Anaesthesiol. Scand. 2009, 53, 543–545. [Google Scholar] [CrossRef]
  172. Yousefifard, M.; Askarian-Amiri, S.; Rafiei Alavi, S.N.; Sadeghi, M.; Saberian, P.; Baratloo, A.; Talebian, M.T. The Efficacy of Ketamine Administration in Prehospital Pain Management of Trauma Patients; a Systematic Review and Meta-Analysis. Arch. Acad. Emerg. Med. 2019, 8, e1. [Google Scholar]
  173. Tran, K.P.; Nguyen, Q.; Truong, X.N.; Le, V.; Le, V.P.; Mai, N.; Husum, H.; Losvik, O.K. A comparison of ketamine and morphine analgesia in prehospital trauma care: A cluster randomized clinical trial in rural Quang Tri province, Vietnam. Prehosp. Emerg. Care 2014, 18, 257–264. [Google Scholar] [CrossRef] [PubMed]
  174. Husum, H. Effects of early prehospital life support to war injured: The battle of Jalalabad, Afghanistan. Prehosp. Disaster. Med. 1999, 14, 75–80. [Google Scholar] [CrossRef] [PubMed]
  175. Husum, H.; Gilbert, M.; Wisborg, T.; Van Heng, Y.; Murad, M. Rural prehospital trauma systems improve trauma outcome in low-income countries: A prospective study from North Iraq and Cambodia. J. Trauma. 2003, 54, 1188–1196. [Google Scholar] [CrossRef] [PubMed]
  176. Bourgoin, A.; Albanèse, J.; Léone, M.; Sampol-Manos, E.; Viviand, X.; Martin, C. Effects of sufentanil or ketamine administered in target-controlled infusion on the cerebral hemodynamics of severely brain-injured patients. Crit. Care Med. 2005, 33, 1109–1113. [Google Scholar] [CrossRef] [PubMed]
  177. Bourgoin, A.; Albanèse, J.; Wereszczynski, N.; Charbit, M.; Vialet, R.; Martin, C. Safety of sedation with ketamine in severe head injury patients: Comparison with sufentanil. Crit. Care Med. 2003, 31, 711–717. [Google Scholar] [CrossRef] [PubMed]
  178. Albanèse, J.; Arnaud, S.; Rey, M.; Thomachot, L.; Alliez, B.; Martin, C. Ketamine decreases intracranial pressure and electroencephalographic activity in traumatic brain injury patients during propofol sedation. Anesthesiology 1997, 87, 1328–1334. [Google Scholar] [CrossRef] [PubMed]
  179. Green, S.M.; Andolfatto, G.; Krauss, B.S. Ketamine and intracranial pressure: No contraindication except hydrocephalus. Ann. Emerg. Med. 2015, 65, 52–54. [Google Scholar] [CrossRef]
  180. Loflin, R.; Koyfman, A. When used for sedation, does ketamine increase intracranial pressure more than fentanyl or sufentanil? Ann. Emerg. Med. 2015, 65, 55–56. [Google Scholar] [CrossRef]
  181. Zeiler, F.A.; Teitelbaum, J.; West, M.; Gillman, L.M. The ketamine effect on intracranial pressure in nontraumatic neurological illness. J. Crit. Care. 2014, 29, 1096–1106. [Google Scholar] [CrossRef]
  182. Zeiler, F.A.; Teitelbaum, J.; West, M.; Gillman, L.M. The ketamine effect on ICP in traumatic brain injury. Neurocrit. Care 2014, 21, 163–173. [Google Scholar] [CrossRef]
  183. Naghipour, B.; Pouraghaei, M.; Tabatabaey, A.; Arjmand, A.; Faridaalaee, G. Comparison of Ketamine and Tramadol in Management of Acute Pain; a Systematic Review. Arch. Acad. Emerg. Med. 2020, 8, e68. [Google Scholar]
  184. Moradi, M.M.; Moradi, M.M.; Safaie, A.; Baratloo, A.; Payandemehr, P. Sub dissociative dose of ketamine with haloperidol versus fentanyl on pain reduction in patients with acute pain in the emergency department; a randomized clinical trial. Am. J. Emerg. Med. 2022, 54, 165–171. [Google Scholar] [CrossRef]
  185. Snijdelaar, D.G.; Cornelisse, H.B.; Schmid, R.L.; Katz, J. A randomised, controlled study of peri-operative low dose s(+)-ketamine in combination with postoperative patient-controlled s(+)-ketamine and morphine after radical prostatectomy. Anaesthesia 2004, 59, 222–228. [Google Scholar] [CrossRef] [PubMed]
  186. Turturro, M.A. Pain, priorities, and prehospital care. Prehosp. Emerg. Care 2002, 6, 486–488. [Google Scholar] [CrossRef] [PubMed]
  187. 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. [Google Scholar] [CrossRef] [PubMed]
  188. Davison, S.N. Clinical Pharmacology Considerations in Pain Management in Patients with Advanced Kidney Failure. Clin. J. Am. Soc. Nephrol. 2019, 14, 917–931. [Google Scholar] [CrossRef] [PubMed]
  189. Schmid, R.L.; Sandler, A.N.; Katz, J. Use and efficacy of low-dose ketamine in the management of acute postoperative pain: A review of current techniques and outcomes. Pain 1999, 82, 111–125. [Google Scholar] [CrossRef]
  190. Moyse, D.W.; Kaye, A.D.; Diaz, J.H.; Qadri, M.Y.; Lindsay, D.; Pyati, S. Perioperative Ketamine Administration for Thoracotomy Pain. Pain Physician 2017, 20, 173–184. [Google Scholar] [PubMed]
  191. Wang, X.; Lin, C.; Lan, L.; Liu, J. Perioperative intravenous S-ketamine for acute postoperative pain in adults: A systematic review and meta-analysis. J. Clin. Anesth. 2021, 68, 110071. [Google Scholar] [CrossRef]
  192. Chumbley, G.M.; Thompson, L.; Swatman, J.E.; Urch, C. Ketamine infusion for 96 h after thoracotomy: Effects on acute and persistent pain. Eur. J. Pain 2019, 23, 985–993. [Google Scholar] [CrossRef]
  193. Bornemann-Cimenti, H.; Wejbora, M.; Michaeli, K.; Edler, A.; Sandner-Kiesling, A. The effects of minimal-dose versus low-dose S-ketamine on opioid consumption, hyperalgesia, and postoperative delirium: A triple-blinded, randomized, active- and placebo-controlled clinical trial. Minerva Anestesiol. 2016, 82, 1069–1076. [Google Scholar]
  194. Pendi, A.; Field, R.; Farhan, S.D.; Eichler, M.; Bederman, S.S. Perioperative Ketamine for Analgesia in Spine Surgery: A Meta-analysis of Randomized Controlled Trials. Spine 2018, 43, E299–E307. [Google Scholar] [CrossRef]
  195. Bell, R.F.; Dahl, J.B.; Moore, R.A.; Kalso, E. Perioperative ketamine for acute postoperative pain. Cochrane Database Syst. Rev. 2006, 7, CD004603, Update in Cochrane Database Syst. Rev. 2015, 7, CD004603. [Google Scholar] [CrossRef]
  196. Zakine, J.; Samarcq, D.; Lorne, E.; Moubarak, M.; Montravers, P.; Beloucif, S.; Dupont, H. Postoperative ketamine administration decreases morphine consumption in major abdominal surgery: A prospective, randomized, double-blind, controlled study. Anesth. Analg. 2008, 106, 1856–1861. [Google Scholar] [CrossRef]
  197. Jouguelet-Lacoste, J.; La Colla, L.; Schilling, D.; Chelly, J.E. 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. [Google Scholar] [CrossRef]
  198. Schug, S.A.; Peyton, P. Does perioperative ketamine have a role in the prevention of chronic postsurgical pain: The ROCKet trial. Br. J. Pain 2017, 11, 166–168. [Google Scholar] [CrossRef] [PubMed]
  199. Correll, D. Chronic postoperative pain: Recent findings in understanding and management. F1000Research 2017, 6, 1054. [Google Scholar] [CrossRef] [PubMed]
  200. Clark, J.D. Ketamine for Chronic Pain: Old Drug New Trick? Anesthesiology 2020, 133, 13–15. [Google Scholar] [CrossRef] [PubMed]
  201. Petrenko, A.B.; Yamakura, T.; Baba, H.; Shimoji, K. The role of N-methyl-D-aspartate (NMDA) receptors in pain: A review. Anesth. Analg. 2003, 97, 1108–1116. [Google Scholar] [CrossRef]
  202. 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. [Google Scholar] [CrossRef]
  203. Schwartzman, R.J.; Alexander, G.M.; Grothusen, J.R.; 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. [Google Scholar] [CrossRef]
  204. Israel, J.E.; St Pierre, S.; Ellis, E.; Hanukaai, J.S.; Noor, N.; Varrassi, G.; Wells, M.; Kaye, A.D. Ketamine for the Treatment of Chronic Pain: A Comprehensive Review. Health Psychol. Res. 2021, 9, 25535. [Google Scholar] [CrossRef]
  205. Maher, D.P.; Chen, L.; Mao, J. Intravenous Ketamine Infusions for Neuropathic Pain Management: A Promising Therapy in Need of Optimization. Anesth. Analg. 2017, 124, 661–674. [Google Scholar] [CrossRef]
  206. Eide, P.K.; 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. [Google Scholar] [CrossRef]
  207. Kvarnström, A.; Karlsten, R.; Quiding, H.; Emanuelsson, B.M.; Gordh, T. The effectiveness of intravenous ketamine and lidocaine on peripheral neuropathic pain. Acta Anaesthesiol. Scand. 2003, 47, 868–877. [Google Scholar] [CrossRef] [PubMed]
  208. Hocking, G.; Cousins, M.J. Ketamine in chronic pain management: An evidence-based review. Anesth. Analg. 2003, 97, 1730–1739. [Google Scholar] [CrossRef] [PubMed]
  209. Shteamer, J.W.; Callaway, M.A.; Patel, P.; Singh, V. How effective is ketamine in the management of chronic neuropathic pain? Pain Manag. 2019, 9, 517–519. [Google Scholar] [CrossRef]
  210. Guimarães Pereira, J.E.; Ferreira Gomes Pereira, L.; Mercante Linhares, R.; Darcy Alves Bersot, C.; Aslanidis, T.; Ashmawi, H.A. Efficacy and Safety of Ketamine in the Treatment of Neuropathic Pain: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Pain Res. 2022, 15, 1011–1037. [Google Scholar] [CrossRef] [PubMed]
  211. Loftus, R.W.; Yeager, M.P.; Clark, J.A.; Brown, J.R.; Abdu, W.A.; Sengupta, D.K.; Beach, M.L. Intraoperative ketamine reduces perioperative opiate consumption in opiate-dependent patients with chronic back pain undergoing back surgery. Anesthesiology 2010, 113, 639–646. [Google Scholar] [CrossRef] [PubMed]
  212. Amr, Y.M. 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. [Google Scholar] [CrossRef]
  213. McMullin, P.R.; Hynes, A.T.; Arefin, M.A.; Saeed, M.; Gandhavadi, S.; Arefin, N.; Eckmann, M.S. Infusion Therapy in the Treatment of Neuropathic Pain. Curr. Pain Headache Rep. 2022, 26, 693–699. [Google Scholar] [CrossRef] [PubMed]
  214. 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. [Google Scholar] [CrossRef]
  215. Bennett, G.J. Update on the neurophysiology of pain transmission and modulation: Focus on the NMDA-receptor. J. Pain Symptom Manag. 2000, 19 (Suppl. S1), S2–S6. [Google Scholar] [CrossRef] [PubMed]
  216. Woolf, C.J.; Thompson, S.W.N. 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. [Google Scholar] [CrossRef]
  217. Costigan, M.; Scholz, J.; Woolf, C.J. Neuropathic pain: A maladaptive response of the nervous system to damage. Annu. Rev. Neurosci. 2009, 32, 1–32. [Google Scholar] [CrossRef] [PubMed]
  218. Ossipov, M.H.; Dussor, G.O.; Porreca, F. Central modulation of pain. J. Clin. Investig. 2010, 120, 3779–3787. [Google Scholar] [CrossRef]
  219. Marchand, F.; Perretti, M.; McMahon, S.B. Role of the immune system in chronic pain. Nat. Rev. Neurosci. 2005, 6, 521–532. [Google Scholar] [CrossRef]
  220. Wolff, K.; Winstock, A.R. Ketamine: From medicine to misuse. CNS Drugs 2006, 20, 199–218. [Google Scholar] [CrossRef]
  221. Yang, Y.; Maher, D.P.; Cohen, S.P. Emerging concepts on the use of ketamine for chronic pain. Expert Rev. Clin. Pharmacol. 2020, 13, 135–146. [Google Scholar] [CrossRef]
  222. Baldwin, M.; Boilini, H.; Lamvu, G. Chronic Pain and Suicide: Is There a Role for Ketamine? Mil. Med. 2017, 182, 1746–1748. [Google Scholar] [CrossRef]
  223. Hassett, A.L.; Aquino, J.K.; Ilgen, M.A. The risk of suicide mortality in chronic pain patients. Curr. Pain Headache Rep. 2014, 18, 436. [Google Scholar] [CrossRef] [PubMed]
  224. Tang, N.K.; Crane, C. Suicidality in chronic pain: A review of the prevalence, risk factors and psychological links. Psychol. Med. 2006, 36, 575–586. [Google Scholar] [CrossRef] [PubMed]
  225. McNicol, E.D.; Schumann, R.; Haroutounian, S. A systematic review and meta-analysis of ketamine for the prevention of persistent post-surgical pain. Acta Anaesthesiol. Scand. 2014, 58, 1199–1213. [Google Scholar] [CrossRef] [PubMed]
  226. Zhao, J.; Wang, Y.; Wang, D. The Effect of Ketamine Infusion in the Treatment of Complex Regional Pain Syndrome: A Systemic Review and Meta-analysis. Curr. Pain Headache Rep. 2018, 22, 12. [Google Scholar] [CrossRef]
  227. Voute, M.; Riant, T.; Amodéo, J.M.; André, G.; Barmaki, M.; Collard, O.; Colomb, C.; Créac’h, C.; Deleens, R.; Delorme, C.; et al. Ketamine in chronic pain: A Delphi survey. Eur. J. Pain 2022, 26, 873–887. [Google Scholar] [CrossRef]
  228. Corriger, A.; Voute, M.; Lambert, C.; Pereira, B.; Pickering, G.; OKAPI Consortium. Ketamine for refractory chronic pain: A 1-year follow-up study. Pain 2022, 163, 690–701. [Google Scholar] [CrossRef]
  229. Dahan, A.; van Velzen, M.; Niesters, M. Ketamine for neuropathic pain: A tiger that won’t bite? Br. J. Anaesth. 2020, 125, e275–e276. [Google Scholar] [CrossRef]
  230. Jonkman, K.; van de Donk, T.; Dahan, A. Ketamine for cancer pain: What is the evidence? Curr. Opin. Support. Palliat. Care 2017, 11, 88–92. [Google Scholar] [CrossRef]
  231. Kissin, I.; Bright, C.A.; Bradley, E.L., Jr. The effect of ketamine on opioid-induced acute tolerance: Can it explain reduction of opioid consumption with ketamine-opioid analgesic combinations? Anesth. Analg. 2000, 91, 1483–1488. [Google Scholar] [CrossRef]
  232. Bell, R.F.; Eccleston, C.; Kalso, E.A. Ketamine as an adjuvant to opioids for cancer pain. Cochrane Database Syst. Rev. 2017, 6, CD003351. [Google Scholar] [CrossRef]
  233. Courade, M.; Bertrand, A.; Guerrini-Rousseau, L.; Pagnier, A.; Levy, D.; Lervat, C.; Cojean, N.; Ribrault, A.; Dugue, S.; Thouvenin, S.; et al. Low-dose ketamine adjuvant treatment for refractory pain in children, adolescents and young adults with cancer: A pilot study. BMJ Support. Palliat. Care 2022, 12, e656–e663. [Google Scholar] [CrossRef]
  234. Bell, R.F.; Eccleston, C.; Kalso, E. Ketamine as adjuvant to opioids for cancer pain. A qualitative systematic review. J. Pain Symptom Manag. 2003, 26, 867–875. [Google Scholar] [CrossRef] [PubMed]
  235. Legge, J.; Ball, N.; Elliott, D.P. The potential role of ketamine in hospice analgesia: A literature review. Consult. Pharm. 2006, 21, 51–57. [Google Scholar] [CrossRef] [PubMed]
  236. Prommer, E.E. Ketamine for pain: An update of uses in palliative care. J. Palliat. Med. 2012, 15, 474–483. [Google Scholar] [CrossRef] [PubMed]
  237. Bredlau, A.L.; Thakur, R.; Korones, D.N.; Dworkin, R.H. Ketamine for pain in adults and children with cancer: A systematic review and synthesis of the literature. Pain Med. 2013, 14, 1505–1517. [Google Scholar] [CrossRef]
  238. Hardy, J.; Quinn, S.; Fazekas, B.; Plummer, J.; Eckermann, S.; Agar, M.; Spruyt, O.; Rowett, D.; Currow, D.C. Randomized, double-blind, placebo-controlled study to assess the efficacy and toxicity of subcutaneous ketamine in the management of cancer pain. J. Clin. Oncol. 2012, 30, 3611–3617. [Google Scholar] [CrossRef]
  239. Goldman, N.; Frankenthaler, M.; Klepacz, L. The Efficacy of Ketamine in the Palliative Care Setting: A Comprehensive Review of the Literature. J. Palliat. Med. 2019, 22, 1154–1161. [Google Scholar] [CrossRef]
  240. Giamberardino, M.A.; Affaitati, G.; Costantini, R.; Guglielmetti, M.; Martelletti, P. Acute headache management in emergency department. A narrative review. Intern. Emerg. Med. 2020, 15, 109–117. [Google Scholar] [CrossRef]
  241. Takahashi, T.T.; Ornello, R.; Quatrosi, G.; Torrente, A.; Albanese, M.; Vigneri, S.; Guglielmetti, M.; Maria De Marco, C.; Dutordoir, C.; Colangeli, E.; et al. Medication overuse and drug addiction: A narrative review from addiction perspective. J. Headache Pain 2021, 22, 32, Erratum in J. Headache Pain 2021, 22, 64. [Google Scholar] [CrossRef]
  242. Lauritsen, C.; Mazuera, S.; Lipton, R.B.; Ashina, S. Intravenous ketamine for subacute treatment of refractory chronic migraine: A case series. J. Headache Pain 2016, 17, 106. [Google Scholar] [CrossRef]
  243. Chah, N.; Jones, M.; Milord, S.; Al-Eryani, K.; Enciso, R. Efficacy of ketamine in the treatment of migraines and other unspecified primary headache disorders compared to placebo and other interventions: A systematic review. J. Dent. Anesth. Pain Med. 2021, 21, 413–429. [Google Scholar] [CrossRef] [PubMed]
  244. Sarvari, H.R.; Baigrezaii, H.; Nazarianpirdosti, M.; Meysami, A.; Safari-Faramani, R. Comparison of the efficacy of intranasal ketamine versus intravenous ketorolac on acute non-traumatic headaches: A randomized double-blind clinical trial. Head Face Med. 2022, 18, 1. [Google Scholar] [CrossRef]
  245. Naeem, F.; Schramm, C.; Friedman, B.W. Emergent management of primary headache: A review of current literature. Curr. Opin. Neurol. 2018, 31, 286–290. [Google Scholar] [CrossRef] [PubMed]
  246. Rashed, A.; Mazer-Amirshahi, M.; Pourmand, A. Current Approach to Undifferentiated Headache Management in the Emergency Department. Curr. Pain Headache Rep. 2019, 23, 26. [Google Scholar] [CrossRef]
  247. Weber, W.V.; Jawalekar, K.S.; Jawalekar, S.R. The effect of ketamine on nerve conduction in isolated sciatic nerves of the toad. Neurosci. Lett. 1975, 1, 115–120. [Google Scholar] [CrossRef] [PubMed]
  248. Frenkel, C.; Urban, B.W. Molecular actions of racemic ketamine on human CNS sodium channels. Br. J. Anaesth. 1992, 69, 292–297. [Google Scholar] [CrossRef]
  249. Abdel-Ghaffar, H.S.; Kalefa, M.A.; Imbaby, A.S. Efficacy of ketamine as an adjunct to lidocaine in intravenous regional anesthesia. Reg. Anesth. Pain Med. 2014, 39, 418–422. [Google Scholar] [CrossRef]
  250. Heydari, F.; Khalilian, S.; Golshani, K.; Majidinejad, S.; Masoumi, B.; Massoumi, A. Topical ketamine as a local anesthetic agent in reducing venipuncture pain: A randomized controlled trial. Am. J. Emerg. Med. 2021, 48, 48–53. [Google Scholar] [CrossRef]
  251. Wang, H.Z.; Wang, L.Y.; Liang, H.H.; Fan, Y.T.; Song, X.R.; She, Y.J. Effect of caudal ketamine on minimum local anesthetic concentration of ropivacaine in children: A prospective randomized trial. BMC Anesthesiol. 2020, 20, 144. [Google Scholar] [CrossRef]
  252. Tverskoy, M.; Oren, M.; Vaskovich, M.; Dashkovsky, I.; Kissin, I. Ketamine enhances local anesthetic and analgesic effects of bupivacaine by peripheral mechanism: A study in postoperative patients. Neurosci. Lett. 1996, 215, 5–8. [Google Scholar] [CrossRef]
  253. Majidinejad, S.; Esmailian, M.; Emadi, M. Comparison of Intravenous Ketamine with Morphine in Pain Relief of Long Bones Fractures: A Double Blind Randomized Clinical Trial. Emerg 2014, 2, 77–80. [Google Scholar]
  254. Motov, S.; Mann, S.; Drapkin, J.; Butt, M.; Likourezos, A.; Yetter, E.; Brady, J.; Rothberger, N.; Gohel, A.; Flom, P.; et al. Intravenous subdissociative-dose ketamine versus morphine for acute geriatric pain in the Emergency Department: A randomized controlled trial. Am. J. Emerg. Med. 2019, 37, 220–227. [Google Scholar] [CrossRef] [PubMed]
  255. Krystal, J.H.; Karper, L.P.; Seibyl, J.P.; Freeman, G.K.; Delaney, R.; Bremner, J.D.; Heninger, G.R.; Bowers, M.B., Jr.; Charney, D.S. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch. Gen. Psychiatry 1994, 51, 199–214. [Google Scholar] [CrossRef] [PubMed]
  256. Lahti, A.C.; Weiler, M.A.; Tamara Michaelidis, B.A.; Parwani, A.; Tamminga, C.A. Effects of ketamine in normal and schizophrenic volunteers. Neuropsychopharmacology 2001, 25, 455–467. [Google Scholar] [CrossRef] [PubMed]
  257. Stoker, A.D.; Rosenfeld, D.M.; Buras, M.R.; Alvord, J.M.; Gorlin, A.W. Evaluation of Clinical Factors Associated with Adverse Drug Events in Patients Receiving Sub-Anesthetic Ketamine Infusions. J. Pain Res. 2019, 12, 3413–3421. [Google Scholar] [CrossRef]
  258. Green, S.M.; Roback, M.G.; Kennedy, R.M.; Krauss, B. Clinical practice guideline for emergency department ketamine dissociative sedation: 2011 update. Ann. Emerg. Med. 2011, 57, 449–461. [Google Scholar] [CrossRef]
  259. Noppers, I.M.; Niesters, M.; Aarts, L.P.H.J.; Bauer, M.C.R.; Drewes, A.M.; Dahan, A.; Sarton, E.Y. 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. [Google Scholar] [CrossRef] [PubMed]
  260. Kiefer, R.T.; Rohr, P.; Ploppa, A.; Dieterich, H.J.; Grothusen, J.; Koffler, S.; Altemeyer, K.H.; Unertl, K.; Schwartzman, R.J. 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. [Google Scholar] [CrossRef] [PubMed]
  261. Blonk, M.I.; Koder, B.G.; van den Bemt, P.M.; Huygen, F.J. Use of oral ketamine in chronic pain management: A review. Eur. J. Pain 2010, 14, 466–472. [Google Scholar] [CrossRef]
  262. Green, S.M.; Clark, R.; Hostetler, M.A.; Cohen, M.; Carlson, D.; Rothrock, S.G. Inadvertent ketamine overdose in children: Clinical manifestations and outcome. Ann. Emerg. Med. 1999, 34 Pt 1, 492–497. [Google Scholar] [CrossRef]
  263. Lin, C.C.; Lin, A.T.; Yang, A.H.; Chen, K.K. Microvascular Injury in Ketamine-Induced Bladder Dysfunction. PLoS ONE 2016, 11, e0160578. [Google Scholar] [CrossRef]
  264. Gray, T.; Dass, M. Ketamine cystitis: An emerging diagnostic and therapeutic challenge. Br. J. Hosp. Med. 2012, 73, 576–579. [Google Scholar] [CrossRef]
  265. Himmelseher, S.; Durieux, M.E. Ketamine for perioperative pain management. Anesthesiology 2005, 102, 211–220. [Google Scholar] [CrossRef] [PubMed]
  266. Anand, K.J.; Garg, S.; Rovnaghi, C.R.; Narsinghani, U.; Bhutta, A.T.; Hall, R.W. Ketamine reduces the cell death following inflammatory pain in newborn rat brain. Pediatr. Res. 2007, 62, 283–290. [Google Scholar] [CrossRef]
  267. Bell, J.D. In Vogue: Ketamine for Neuroprotection in Acute Neurologic Injury. Anesth. Analg. 2017, 124, 1237–1243. [Google Scholar] [CrossRef]
  268. Bates, M.L.S.; Trujillo, K.A. Long-lasting effects of repeated ketamine administration in adult and adolescent rats. Behav. Brain Res. 2019, 369, 111928. [Google Scholar] [CrossRef] [PubMed]
  269. Green, S.M.; Coté, C.J. Ketamine and neurotoxicity: Clinical perspectives and implications for emergency medicine. Ann. Emerg. Med. 2009, 54, 181–190. [Google Scholar] [CrossRef]
  270. Short, B.; Fong, J.; Galvez, V.; Shelker, W.; Loo, C.K. Side-effects associated with ketamine use in depression: A systematic review. Lancet Psychiatry 2018, 5, 65–78. [Google Scholar] [CrossRef] [PubMed]
  271. Strayer, R.J.; Nelson, L.S. Adverse events associated with ketamine for procedural sedation in adults. Am. J. Emerg. Med. 2008, 26, 985–1028, Erratum in Am. J. Emerg. Med. 2009, 27, 512. [Google Scholar] [CrossRef] [PubMed]
  272. Coppel, D.L.; Bovill, J.G.; Dundee, J.W. The taming of ketamine. Anaesthesia 1973, 28, 293–296. [Google Scholar] [CrossRef]
  273. Tobias, J.D. Dexmedetomidine and ketamine: An effective alternative for procedural sedation? Pediatr. Crit. Care Med. 2012, 13, 423–427. [Google Scholar] [CrossRef]
  274. Azizkhani, R.; Kouhestani, S.; Heydari, F.; Majidinejad, S. A comparative study of dexmedetomidine and propofol to prevent recovery agitation in adults undergoing procedural sedation with ketamine: A randomized double-blind clinical trial. Am. J. Emerg. Med. 2021, 50, 167–172. [Google Scholar] [CrossRef] [PubMed]
  275. Azizkhani, R.; Kouhestani, S.; Heydari, F.; Esmailian, M.; Feizi, A.; Gourtani, B.K.; Safavi, M. Comparison of the effects of dexmedetomidine and propofol in reducing recovery agitation in pediatric patients after ketamine procedural sedation in emergency department. J. Res. Med. Sci. 2021, 26, 61. [Google Scholar] [CrossRef]
  276. Bubeníková-Valesová, V.; Horácek, J.; Vrajová, M.; Höschl, C. Models of schizophrenia in humans and animals based on inhibition of NMDA receptors. Neurosci. Biobehav. Rev. 2008, 32, 1014–1023. [Google Scholar] [CrossRef] [PubMed]
  277. Blagrove, M.; Morgan, C.J.; Curran, H.V.; Bromley, L.; Brandner, B. The incidence of unpleasant dreams after sub-anaesthetic ketamine. Psychopharmacology 2009, 203, 109–120. [Google Scholar] [CrossRef] [PubMed]
  278. Allen, C.A.; Ivester, J.R., Jr. Ketamine for Pain Management-Side Effects & Potential Adverse Events. Pain Manag. Nurs. 2017, 18, 372–377. [Google Scholar] [CrossRef]
  279. Liu, Y.; Lin, D.; Wu, B.; Zhou, W. Ketamine abuse potential and use disorder. Brain Res. Bull. 2016, 126 Pt 1, 68–73. [Google Scholar] [CrossRef]
  280. Walsh, Z.; Mollaahmetoglu, O.M.; Rootman, J.; Golsof, S.; Keeler, J.; Marsh, B.; Nutt, D.J.; Morgan, C.J.A. Ketamine for the treatment of mental health and substance use disorders: Comprehensive systematic review. BJPsych Open 2021, 8, e19, Erratum in BJPsych Open 2022, 8, e29. [Google Scholar] [CrossRef] [PubMed]
  281. Worrell, S.D.; Gould, T.J. Therapeutic potential of ketamine for alcohol use disorder. Neurosci. Biobehav. Rev. 2021, 126, 573–589. [Google Scholar] [CrossRef]
  282. Pal, H.R.; Berry, N.; Kumar, R.; Ray, R. Ketamine dependence. Anaesth. Intensive Care 2002, 30, 382–384. [Google Scholar] [CrossRef]
  283. Gable, R.S. Acute toxic effects of club drugs. J. Psychoact. Drugs 2004, 36, 303–313. [Google Scholar] [CrossRef]
  284. Jansen, K.L.; Darracot-Cankovic, R. The nonmedical use of ketamine, part two: A review of problem use and dependence. J. Psychoact. Drugs 2001, 33, 151–158. [Google Scholar] [CrossRef] [PubMed]
  285. Moghaddam, B.; Adams, B.; Verma, A.; Daly, D. Activation of glutamatergic neurotransmission by ketamine: A novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. J. Neurosci. 1997, 17, 2921–2927. [Google Scholar] [CrossRef] [PubMed]
  286. Ivan Ezquerra-Romano, I.; Lawn, W.; Krupitsky, E.; Morgan, C.J.A. Ketamine for the treatment of addiction: Evidence and potential mechanisms. Neuropharmacology 2018, 142, 72–82. [Google Scholar] [CrossRef] [PubMed]
  287. Cvrcek, P. Side effects of ketamine in the long-term treatment of neuropathic pain. Pain Med. 2008, 9, 253–257. [Google Scholar] [CrossRef]
  288. Gottlieb, M.; Long, B.; Koyfman, A. Approach to the Agitated Emergency Department Patient. J. Emerg. Med. 2018, 54, 447–457. [Google Scholar] [CrossRef]
  289. Cauli, O.; González-Usano, A.; Cabrera-Pastor, A.; Gimenez-Garzó, C.; López-Larrubia, P.; Ruiz-Sauri, A.; Hernández-Rabaza, V.; Duszczyk, M.; Malek, M.; Lazarewicz, J.W.; et al. Blocking NMDA receptors delays death in rats with acute liver failure by dual protective mechanisms in kidney and brain. Neuromolecular. Med. 2014, 16, 360–375. [Google Scholar] [CrossRef]
  290. Lindefors, N.; Barati, S.; O’Connor, W.T. Differential effects of single and repeated ketamine administration on dopamine, serotonin and GABA transmission in rat medial prefrontal cortex. Brain Res. 1997, 759, 205–212. [Google Scholar] [CrossRef]
  291. De Iaco, F.; Riccardi, A.; Guarino, M.; Gandolfo, E.; Di Ricatti, G.; Saggese, M.P.; Cibinel, G.A.; Carbone, G. Il dolore non può aspettare. Campagna formativa SIMEU sul dolore acuto in Medicina d’Emergenza Urgenza. In Fighting Pain—Combattere Il Dolore; HPS—Health Publishing & Services: Milano, Italy, 2014; Volume 1. [Google Scholar]
Table 1. Analgesic doses of ketamine administered via different routes.
Table 1. Analgesic doses of ketamine administered via different routes.
LOW DOSE KETAMINE (ANALGESIC)
Administration RouteDose
Intravenous0.15–0.3 mg/kg bolus
0.15–0.3 mg/kg/h infusion
Intramuscolar0.5–1 mg/kg
Intranasal1 mg/kg
Oral0.5 mg/kg every 12 h
Transdermal25 mg/24 h
Subcutaneous0.05–0.15 mg/kg/h
Rectal10 mg/kg
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Riccardi, A.; Guarino, M.; Serra, S.; Spampinato, M.D.; Vanni, S.; Shiffer, D.; Voza, A.; Fabbri, A.; De Iaco, F., on behalf of the Study and Research Center of the Italian Society of Emergency Medicine. Narrative Review: Low-Dose Ketamine for Pain Management. J. Clin. Med. 2023, 12, 3256. https://doi.org/10.3390/jcm12093256

AMA Style

Riccardi A, Guarino M, Serra S, Spampinato MD, Vanni S, Shiffer D, Voza A, Fabbri A, De Iaco F on behalf of the Study and Research Center of the Italian Society of Emergency Medicine. Narrative Review: Low-Dose Ketamine for Pain Management. Journal of Clinical Medicine. 2023; 12(9):3256. https://doi.org/10.3390/jcm12093256

Chicago/Turabian Style

Riccardi, Alessandro, Mario Guarino, Sossio Serra, Michele Domenico Spampinato, Simone Vanni, Dana Shiffer, Antonio Voza, Andrea Fabbri, and Fabio De Iaco on behalf of the Study and Research Center of the Italian Society of Emergency Medicine. 2023. "Narrative Review: Low-Dose Ketamine for Pain Management" Journal of Clinical Medicine 12, no. 9: 3256. https://doi.org/10.3390/jcm12093256

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop