Next Article in Journal
Ribosomally Synthesized and Post-Translationally Modified Peptides Assembled by ThiF-like Adenylyltransferases: Recent Advances and Future Perspectives
Previous Article in Journal
Exploring the Structural Design, Antibacterial Activity, and Molecular Docking of Newly Synthesized Zn(II) Complexes with NNO-Donor Carbazate Ligands
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Ketamine—From an Anesthetic to a Psychiatric Drug: Mechanisms of Action, Clinical Applications and Potential Risks

by
Ewa Gibuła-Tarłowska
1,
Anna Wiszniewska
2,
Magdalena Turyk
2,
Paulina Szymczyk
2,
Jolanta H. Kotlińska
1 and
Ewa Kędzierska
1,*
1
Department of Pharmacology and Pharmacodynamics, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland
2
Student Scientific Club “Synapse”, Department of Pharmacology and Pharmacodynamics, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland
*
Author to whom correspondence should be addressed.
Molecules 2025, 30(13), 2824; https://doi.org/10.3390/molecules30132824
Submission received: 24 April 2025 / Revised: 30 May 2025 / Accepted: 13 June 2025 / Published: 30 June 2025

Abstract

Ketamine, originally developed as an anesthetic, is gaining increasing attention due to its multifaceted pharmacological properties. In addition to its use in anesthesia, ketamine exerts potent analgesic effects via N-methyl-D-aspartate (NMDA) receptor antagonism, modulating pain perception and reducing central sensitization, particularly in chronic and neuropathic pain conditions. Emerging evidence also supports ketamine’s potential in the treatment of substance use disorder, where it may disrupt maladaptive reward-related memories and promote neuroplasticity which facilitates behavioral change. Moreover, in recent years, S-ketamine has shown rapid and potent antidepressant effects, especially in treatment-resistant depression (TRD), probably due to increased glutamatergic signaling, synaptic plasticity and the release of neurotrophic factors. This heterogeneous therapeutic profile positions ketamine as a unique agent at the interface of anesthesia, pain management, addiction medicine and psychiatry, warranting further exploration into its mechanism and long-term effectiveness.

1. Basic Information

The medical history of ketamine begins in the 1950s, when a new class of anesthetic drugs—cyclohexylamines—was developed. The first representative of this group to be discovered was phencyclidine (PCP). However, due to its long duration of action, phencyclidine caused severe postoperative delirium in humans, which motivated scientists to search for a short-acting derivative with a more favorable pharmacological profile. Then in 1962, ketamine (2-(2-chlorophenyl)-2-(methylamino)cyclohexanone) was obtained by Professor Calvin Lee Stevens. One of the numerous derivatives of arylcyclohexylamine, ketamine showed unique anesthetic properties, combined with analgesic and amnestic effects and a short duration of action, making it a promising anesthetic drug [1]. The researchers found that, in contrast to its precursor, it did not impact blood pressure and did not cause major respiratory depression. In addition, it showed a number of advantages, such as a low incidence of postoperative nausea or vomiting and a lower psychomimetic potential. As a result of its observed effects, in 1970, the Food and Drug Administration (FDA) approved ketamine as an anesthetic drug for use in children and adults [2].
The etymology of the compound name ‘ketamine’ refers to its chemical structure, in which a ketone and an amine fragment can be distinguished. It is an arylcycloalkylamine consisting of a chlorophenyl ring linked to a cyclohexanone ring. Furthermore, ketamine constitutes a chiral molecule and exists as two enantiomeric forms—S-ketamine and R-ketamine—which differ in their pharmacological properties, potency and duration of action due to their distinct spatial configurations [3]. S-ketamine shows approximately three-times more powerful analgesic and anesthetic effects [4,5] with a three-times greater affinity for N-methyl-D-aspartate (NMDA) receptors [6,7]. However, commercially, it is available in a racemic form containing equal parts of both enantiomers in the form of the hydrochloride salt for parenteral administration [1,3,8] and as a single S-ketamine used intranasally, as this is cheaper [3,4].
Initially, ketamine was employed exclusively as an anesthetic. Its unique dissociative effects and partial agonism at mu opioid receptors allow for painful procedures to be performed in a continuous state of sedation and comfort [9,10,11]. The term ‘dissociative anesthetic’, which is used to describe ketamine, suggests that it induces a state of detachment from the environment in the patient, with an accompanying distortion of visual and auditory stimuli [1,12]. This state has often been described as floating in space and a lack of feeling in the arms and legs. Unfortunately, such effects have contributed to attempts at the recreational and criminal use of this drug [1]. Particularly, subanesthetic doses of ketamine can induce psychedelic effects which manifest as hallucinations and enhancements in sensory perception. Furthermore, it causes memory impairment, which increases according to the administered dose [13,14,15]. However, scientists have focused primarily on the potential therapeutic applications of ketamine. It has been noted that when administered at a subdissociative dose (0.1 to 0.4 mg/kg intravenously, i.v.), otherwise known as ‘low-dose ketamine’ (LDK), it has analgesic and sedative properties, so it can be effective in treating chronic, postoperative and neuropathic pain [3,12]. In this way, it is used either alone or as an adjunct to other pain relief medications. It is safe and effective in combination with injectable nonsteroidal pain medications, as well as opioids. Furthermore, as concerns about opioid use have grown, it has become more widely accepted [16,17]. The dosage determines the application and resulting effects of the drug, leading to variations in the prescribing protocol. This versatility renders ketamine valuable in both anesthesia and pain management. Currently, ketamine is an alternative hospital drug for the relief of postoperative pain and to prevent the exacerbation of acute or chronic pain when standard therapy fails. In addition, ketamine reduces opioid tolerance and prevents opioid-induced hypersensitivity (OIH), which can develop after the chronic use of opioids [12,15,18]. Since these conditions are mainly caused by the activation of NMDA receptors by opioids, they are reversed by ketamine administration [19]. Another extremely interesting finding was the antidepressant effect of ketamine. The rapid improvement observed in patient mood in the 1970s [1] now constitutes one of the main lines of research on the use of ketamine as an effective antidepressant [20]. Particularly important is the fact that the antidepressant action appears very quickly and persists for about a week even after a single administration. Both enantiomers of ketamine are rapidly metabolized in the liver by cytochrome P450 enzymes [21,22]. In the first step, S-ketamine and R-ketamine are demethylated by CYP3A4 or CYP2B6 to the main metabolites: S-norketamine and R-norketamine [23,24]. These compounds are then converted to dehydronorketamines (DHNKs) (S-DHNK and R-DHNK) or hydroxyketamines (HNKs) (S-HNK and R-HNK), respectively [25,26]. Additionally, cytochrome CYP2A6 catalyzes the hydroxylation of norketamines at position 6, forming (2S,6S)-HNK and (2R,6R)-HNK (Figure 1). Although the liver is considered the main site of ketamine metabolism, the kidneys, intestines and lungs are also potential metabolism sites [27]. The metabolites are excreted and eliminated in the urine and bile [28]. Furthermore, age-dependent differences in ketamine metabolism are observed. In pediatric cases, higher doses are required due to their accelerated metabolism. Conversely, in elderly individuals, a slower metabolic rate necessitates lower doses [29]. In view of the information presented above, it is essential to note the pharmacological differences in the activity of enantiomers and active metabolites of ketamine. While R-norketamine, an R-ketamine metabolite, does not show antidepressant activity, S-norketamine, the main metabolite of S-ketamine, induces a comparable antidepressant effect to the mother compound, with a slightly lower affinity for NMDA receptors. Furthermore, behavioral tests showed no potential for abuse or psychotomimetic effects compared to S-ketamine, making it a safer alternative in the treatment of depression. Nevertheless, further studies are needed to determine its potential in terms of this therapeutic indication [30,31].

2. Pharmacokinetic Properties

In addition to i.v., ketamine is commonly given via the intramuscular (i.m.) and subcutaneous (s.c.) routes. Ketamine demonstrates good solubility in both water and lipids; thus, it may be administered orally, rectally and even intranasally [2,3]. This is an important property, as in many clinical cases it may be impossible or dangerous to administer the drug by one of these available routes, necessitating the choice of another route. However, contradicting those advantages, routes of drug administration vary in bioavailability. Following i.v. administration, ketamine exhibits a rapid onset of action and peak plasma concentrations. Moreover, it rapidly enters the brain and then redistributes to peripheral tissues. The recommended initial i.v. dose of ketamine ranges from 1 to 4.5 mg/kg and should be administered over 60 s to individuals aged 16 and older. An average dose of 2 mg/kg typically induces analgesic or dissociative effects lasting about 5 to 15 min; the onset of action is within 10 to 30 s. When i.v. access is difficult, in emergency situations, i.m. or s.c. injections are used as an alternative. The i.m. route of administration, with an effective dosage of 8–10 mg/kg, provides a high bioavailability of 93% and results in peak plasma concentrations within 5 to 30 min [32,33]. However, oral ketamine has a much lower bioavailability of about 17% to 29% due to extensive first-pass hepatic metabolism, whereas the intranasal and rectal administration of ketamine demonstrate bioavailabilities of 45% to 50% and 25% to 30%, respectively [8] (Table 1). It is understandable that the selection of the appropriate route of administration of ketamine depends on many factors, including the patient’s condition, the type of procedure and the available resources. Through i.v., ketamine is employed for the induction of general anesthesia, acute pain control and diagnostic procedures, which guarantees a rapid onset of action, precise dosing and the possibility of continuous infusion. A separate method is the intranasal administration of S-ketamine in TRD, described later in the manuscript.

3. Molecular Mechanism of Action of Ketamine and Its Enantiomers (S-, R- and Racemic)

The mechanism of action of ketamine is multidirectional and not fully understood. Its primary mechanism involves non-selective and non-competitive antagonism of glutamatergic ionotropic NMDA receptors widely distributed in the central nervous system (CNS). NMDA receptors are heterotetrameric complexes typically composed of two GluN1 subunits combined with two regulatory GluN2 subunits (GluN2A-D), and sometimes GluN3 subunits. The specific composition of these subunits determines the receptor’s functional properties, including ion conductance, gating kinetics and sensitivity to pharmacological agents. NMDA receptors play a key role in synaptic transmission, plasticity and learning and memory processes [49,50]. Furthermore, ketamine differentially inhibits NMDA receptors depending on their subunit composition, with receptors containing GluN2C/D subunits displaying distinct sensitivity compared to those with GluN2A/B subunits [35,36,37,38,39,51]. At low concentrations, ketamine induces the allosteric inhibition of NMDA receptors. This allosteric action occurs through the stabilization of receptor conformations that reduce receptor activation independently of the channel-blocking effect, likely by binding to sites outside the ion channel pore. This mechanism, as described by [52], contributes to the fine-tuning of NMDA receptor activity and may underlie ketamine’s unique pharmacological profile, including its rapid anesthetic and antidepressant properties [52]. Ketamine also interacts with sigma receptors, particularly sigma-1 receptors that are chaperone proteins located in the endoplasmic reticulum membrane which are involved in neuroprotection, synaptic plasticity and cellular stress responses. Sigma-1 receptors modulate calcium signaling, ion channel activity and mitochondrial function. Ketamine acts as an agonist of these receptors, and this interaction may contribute to its antidepressant and neuroprotective effects, notably through the regulation of neurotrophic factors like brain-derived neurotrophic factor (BDNF) and the modulation of stress-related pathways [53,54]. Ketamine modulates adrenergic and serotoninergic receptors, influencing neurotransmitter release and mood regulation, thereby complementing its rapid antidepressant actions [55]. It also blocks voltage-gated calcium and potassium channels, further affecting neuronal excitability and synaptic signaling [40,41,56]. Acting as an H1 receptor antagonist, ketamine contributes to sedation and anesthesia [56].
Moreover, ketamine exhibits antihistaminic and anti-inflammatory (immunosuppressive) properties, as it can inhibit histamine release from mast cells, likely through the modulation of calcium-dependent processes. This antihistaminic action may play a role in reducing inflammatory responses during surgical procedures and chronic pain treatments [57]. Ketamine demonstrates the aforementioned significant anti-inflammatory effects by suppressing pro-inflammatory cytokine production, including interleukin-6 (IL-6), tumor necrosis factor alpha (TNFα) and Interleukin-1β (IL-1β). In addition, it can modulate key inflammatory signaling pathways, such as the inhibition of nuclear kappa B (NF-κB) activation, thereby downregulating genes involved in inflammatory responses. These properties are particularly relevant in conditions like chronic pain and depression, where inflammation plays a central role [30,58]. Furthermore, ketamine reduces the activation of macrophages and microglia, which may explain its neuroprotective properties in conditions associated with neuroinflammation [58,59].
Other pharmacological targets of ketamine include the inhibition of nicotinic acetylcholine receptors (nAChRs), particularly α7-nAChRs, which are involved in inflammatory and cognitive processes. This action may contribute to ketamine’s effects on memory and its anti-inflammatory profile [60]. Ketamine also interacts with opioid receptors, primarily as a weak partial agonist at µ-opioid receptors, which may enhance its analgesic properties, although the clinical relevance of this interaction remains debated [61]. These diverse actions highlight ketamine’s complex pharmacological profile, extending beyond NMDA receptor antagonism to include the modulation of sigma receptors, immune and histaminergic systems and additional molecular targets. This multifaceted activity likely underpins its wide-ranging therapeutic effects in clinical practice.
However, in this case, we observe high activity not only of the racemic form but also of the single enantiomers. S-ketamine has an approximately two-to-three-times greater affinity for NMDA receptors than R-ketamine, making it more potent as an anesthetic and analgesic [6]. Its higher potency allows for lower dosing, potentially reducing side effects while maintaining therapeutic efficacy. S-ketamine has also demonstrated a rapid antidepressant effect and is approved for use in TRD under the name ‘esketamine’ [62]. In addition to its primary effects on NMDA receptors, S-ketamine has been shown to affect α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor activity, contributing to synaptic plasticity and neuroprotection. This mechanism is thought to underlie its rapid antidepressant properties [46].
Although R-ketamine is less potent at NMDA receptors compared to its S-counterpart, it exhibits unique pharmacodynamic properties, with preclinical studies suggesting that R-ketamine may provide longer-lasting antidepressant effects due to stronger modulation of synaptic plasticity and neurogenesis [63]. Similarly, S-ketamine enhances synaptic plasticity and neurogenesis by increasing BDNF release and activating the mTOR signaling pathway, which supports synaptic remodeling and new neuronal formation, contributing to its rapid therapeutic effects [62,64].
The racemic mixture of ketamine contains both the S- and R-enantiomers, combining their respective pharmacological properties. This formulation is widely used in clinical practice for analgesia and pain management, as it provides a balanced profile of rapid analgesic, sedative and antidepressant effects [8]. The complementary actions of both enantiomers may also contribute to ketamine’s efficacy in conditions such as neuropathic pain and chronic pain syndromes. Clinically, the racemic form offers the advantage of utilizing the high affinity of S-ketamine for the NMDA receptor to achieve rapid effects, while incorporating the potentially longer-lasting therapeutic actions of R-ketamine. This duality may enhance the overall efficacy of ketamine, particularly in complex or refractory cases [62].

4. Clinical Uses in Anesthesiology

Focusing on the use of ketamine as an anesthetic, it should be noted that it is well entrenched in the induction and maintenance of general anesthesia, both inside and outside the operating room, as well as in emergency states. Ketamine is highly effective for short medical procedures that do not require skeletal muscle relaxation and can be used as a pre-anesthetic for the induction of general anesthesia in combination with other drugs, such as nitrous oxide in patients of different ages, ranging from 3 months of age [29] onwards. Of particular importance is the fact that it maintains proper skeletal muscle tone, preserves systemic vascular resistance and cardiac efficiency and does not affect oxygen saturation: it maintains proper pharyngeal and laryngeal reflexes and enables spontaneous breathing [65]. Moreover, ketamine maintains the reflexes of the respiratory system and exhibits bronchodilatatory effects—which makes it safe for patients with active bronchospasm and reactive airway disease [33,66], as well as asthma [67]. The drug is effective in states where hemodynamic balance is required—in trauma, sepsis or cyanotic heart disease. Ketamine is likewise valuable in total i.v. anesthesia for patients with cardiogenic shock, hypovolemia and pericardial tamponade [33,49]. Furthermore, it may be used as an LDK alongside other drugs, such as opiates, benzodiazepines or propofol, which allows a reduction in the dosage of administered drugs by up to 50%, which equals a lower risk of side effects [66]. However, it should be emphasized that the onset and duration of the drug’s effect varies depending on the route of administration. Moreover, the differences in the activity of the individual stereoisomeric forms are significant. S-ketamine has an approximately two-to-three-times greater affinity for NMDA receptors than R-ketamine and it has been confirmed to have a greater anesthetic potency and faster clearance, which results in a shorter recovery time [6]. In the randomized, double-blind study of Geisslinger et al. [68], when a racemic mixture was given, a statistically significantly lower clearance and volume of distribution was observed for R-ketamine compared with S-ketamine; therefore, the body eliminates R-ketamine more slowly and it likely remains more in the circulation than in the tissues, whereas S-ketamine’s pharmacokinetics are more predictable and stable, regardless of whether it is administered alone or as part of a racemic mixture; it has a faster onset of action and is also eliminated faster, allowing the patient to regain psychomotor skills more quickly [68,69].
Racemic ketamine is known worldwide and has been used in medicine since the 1960s. It has been clinically tested; its properties, dosage and profile of action have been determined; and it is also inexpensive. The better parameters of S-ketamine, i.e., greater potency and a better profile of side effects, are of little importance in anesthetic practice, because many groups of drugs are utilized in such procedures, and these mutually intensify their effects in the mechanism of hyperadditive synergism, and the side effects are less significant when the patient remains unconscious [5,8,70,71]. S-ketamine is relatively new, is patented in some countries (e.g., Germany, Austria) and is significantly more expensive, which is why racemic ketamine is still the drug of choice in situations where there is an urgent need for anesthesia [72].

5. Risks and Benefits

In view of the considerable amount of controversy surrounding ketamine, the risk–benefit factors need to be evaluated before application as an anesthetic (Table 2). Generally, it has been observed that ketamine possesses neurotoxic potential connected to neuronal cell apoptosis and necrosis [73]. However, the research results are contradictory, with one stating ketamine’s toxicity has not been demonstrated in humans—only in select animal models [73]—and the other reporting the negative effect as ‘dose-dependent’ [74]. As mentioned previously, the anesthetic’s bronchodilatatory property makes it a well-established drug for patients with airway diseases, such as asthma and bronchospasms. Nonetheless, the administration of ketamine should be monitored, as the medicine may carry the risk of respiratory hazards and blockage. In addition, it can cause cardiovascular perturbations and psychomimetic effects; hence, blood pressure and electrocardiogram measurements should also be taken during operations [73,74]. It is crucial to mention the limitations of ketamine use in light of its dissociative as well as pro-convulsant properties. Administration of the drug may be dangerous to patients with epilepsy (can cause seizures) and schizophrenia (may induce or worsen psychosis). In addition, during recovery, the patient may be characterized by excessive agitation caused by hallucinations after ketamine anesthesia. Furthermore, the use of this drug can be particularly problematic during certain treatments and operations, as the patient anesthetized with ketamine can make spontaneous and involuntary movements [32]. The anesthetic can also induce hallucinations, delirium and vivid dreams. Likewise, it distorts sensory input to the upper centers of the CNS, affecting pain and emotional responses, as well as memory [66,75]. It also increases cerebral blood flow, which has been a factor in ruling out the administration of the anesthetic in patients with cranial injuries. However, studies show that this effect can be negated by controlling ventilation and sedation during anesthesia, and by administering midazolam along with ketamine. This pairing has even been shown to be more effective than the combination of fentanyl and midazolam [32].
As a conclusion, it should be emphasized that despite its good reputation and popularity over the years, today, ketamine is no longer the preferred primary drug for anesthesia. However, its wide spectrum of advantages still makes it a valuable co-anesthetic. In modern medicine, the use of ketamine should be preceded by an assessment of benefits and risks on an individual basis for the patient [74]. Very valuable sources of information in this context are public assessment reports. These reports, published by the European Medicines Agency (EMA), provide patients, healthcare providers and public health institutions with comprehensive insight into data regarding the effectiveness and safety of drugs. Currently, EMA publishes European Public Assessment Reports (EPAR), including the report for Spravato® ((S)-ketamine nasal spray) which was approved by the European Commission in 2019 (European Medicines Agency. Spravato (S-ketamine): European Public Assessment Report, 2019. Available from https://www.ema.europa.eu/en/medicines/human/EPAR/spravato, accessed on 26 May 2025). Other authorized medicinal products, such as Ketalar® or Ketanest®, contain racemic ketamine and do not have EPAR. This indicates that they were not assessed through the centralized EMA procedure but were authorized either via national procedures or through the Mutual Recognition Procedure (MRP) in individual European Union member countries. In such cases, information on these medicines can be found through national regulatory authorities or pharmaceutical databases.

6. Ketamine in Pain Management

Ketamine plays a crucial role in pain management primarily by modulating NMDA receptor activity, thereby reducing central sensitization, interrupting the wind-up phenomenon and diminishing the formation of pain memory. At the spinal cord level, NMDA receptor activation contributes to the development of central sensitization and the wind-up phenomenon leading to an increased transmission of pain signals to higher brain centers [76]. NMDA receptors are also involved in the formation of pain memories through mechanisms of long-term potentiation (LTP). Activation of these receptors in brain regions such as the anterior cingulate cortex leads to lasting changes in synaptic transmission, which can result in chronic pain sensations, even after the original stimulus has subsided [60]. Ketamine acts by reducing central sensitization, the wind-up phenomenon and pain memory. Central sensitization occurs when the nervous system becomes hypersensitive to pain due to prolonged stimulation. This causes an exaggerated pain response, even to stimuli that normally do not cause pain [76,77]. Ketamine helps to prevent this by blocking NMDA receptors, which are key mediators of central sensitization. In this way, it reduces hyperexcitability in pain pathways, helping to control acute and chronic pain. The wind-up phenomenon is a process in which the repeated stimulation of pain pathways leads to an increased pain response over time. It occurs due to the continuous activation of NMDA receptors in the spinal cord, which amplifies pain signals and may contribute to the presence of acute pain, its progression to chronic pain, increased pain sensitivity (hyperalgesia) and even allodynia, when stimuli that are normally painless, such as touch, can be perceived as painful [78,79]. Studies have shown that ketamine has mild-to-moderate analgesic effects for a variety of chronic pain conditions (e.g., neuropathic pain, central pain syndromes, headaches and temporomandibular joint disorders) [63,79] (Table 3). Pain memory (the persistence of pain perception even after the initial injury has healed) is a common feature of chronic pain. Ketamine’s ability to modulate neuronal plasticity in areas of the brain and spinal cord responsible for pain processing plays a significant role in ‘resetting’ pain pathways, thereby reducing the persistence of chronic pain [80].
Ketamine also interacts with opioid receptors, particularly the μ-opioid receptor. Studies have shown that ketamine can potentiate μ-opioid receptor-dependent signaling, enhancing the analgesic effects of opioids, which may explain the ability of ketamine to improve the efficacy of opioids and reduce postoperative opioid requirements. Perioperative subanesthetic doses of ketamine were effective in postoperative pain control, reducing opioid requirements, while displaying minimal adverse effects [81,82]. This was seen in breast, obstetric, bariatric and orthopedic surgeries, while minimal effect was evident in pediatric and thoracic surgeries [83]. Another advantage of LDK is that it can reverse opioid tolerance, allowing for more effective pain relief [67,68,69,70] and reduce or oppose the clinical features of opioid-induced hyperalgesia (OIH), in which opioids paradoxically increase pain in patients receiving long-term opioid therapy [82,84,85]. The occurrence of OIH is associated with changes in the conformation of opioid receptors [86]. Several mechanisms of the development of this phenomenon are considered, with the glutamatergic mechanism associated with the activation of NMDA receptors being extremely important. NMDA receptor stimulation can occur under the influence of opioids [69,70,71,72] which, by binding to μ receptors, activate protein kinase C (PKC), which causes phosphorylation of the NMDA receptor. The NMDA receptor, at rest, is blocked by magnesium ions and its phosphorylation causes the attenuation of the magnesium blockade and an increase in the influence of calcium ions [87]. This phenomenon causes an increase in pain conduction [86].
Table 3. The selected examples of the use of ketamine in different types of pain.
Table 3. The selected examples of the use of ketamine in different types of pain.
Type of PainAdministration Regimen/Key FeaturesLiterature
AcutePost-traumatic pain in prehospital settings
fentanyl combined with low-dose
ketamine (0.25–0.3 mg/kg) was administered,
resulting in more effective and safer analgesia.
[88,89,90]
Postoperative pain in orthopedic surgery
ketamine was administered as an i.v. bolus at a dose of 0.5 mg/kg, followed by a continuous i.v. infusion at 0.25 mg/kg/h, which resulted in a
reduction in hyperalgesia areas without side effects.
[90,91,92]
Pain in renal colic
ketamine was administered intranasally
at a dose of 1 mg/kg, which resulted
in a reduction in pain intensity.
[91,92,93]
Pain after third molar extraction
a single intranasal dose of ketamine (50 mg)
provided significant pain relief
lasting for 3 h post-administration.
[43,88,94]
Pain in acute traumatic conditions in children
ketamine was administered intranasally 0.7 mg/kg, with the option of an additional 0.3 mg/kg bolus if pain exceeded 50 mm on the VAS scale; this regimen resulted in effective pain control with minimal
and transient adverse effects.
[95,96,97]
Chronic and neuropathicChronic pain—cancer-related (opioid-resistant)
i.v. infusion of ketamine; dosage 0.25–0.6 mg/kg
for 4–6 h daily for several days; reduction in pain
in opioid-resistant patients and improvement
in quality of life.
[98,99,100]
Chronic pain—CRPS (complex regional pain syndrome)
daily infusions of ketamine 0.35 mg/kg i.v. for 4 h a day for 10 days results in long-lasting pain relief (lasting up to 12 weeks) and improved limb function.
[54,94,101]
Neuropathic pain—chronic (e.g., diabetes, neuralgia)
i.v. infusion of ketamine 0.1–0.5 mg/kg/h for 4–6 h; reduction in pain intensity, improved quality of life, possibility of reducing opioid dosage.
[102,103,104]
Neuropathic pain—postherpetic neuralgia
i.v. infusion of 0.1–0.5 mg/kg/h for 4–6 h; reduction in pain and improvement in patient functioning.
[97,103,104]
PerioperativeOrthopedic surgery—arthroplasties
i.v. bolus 0.5 mg/kg during induction of anesthesia, followed by continuous infusion at 0.25 mg/kg/h, which resulted in a reduction in pain management and reduced opioid therapy.
[69,105,106]
Spinal surgery
i.v. bolus dose of 0.15–0.25 mg/kg prior to anesthesia induction, resulting in reduced pain.
[107,108,109]
Laparoscopic procedures
Low-dose ketamine—0.25 mg/kg, i.v. as a single bolus or by continuous infusion, resulting
in improved pain control.
[110,111,112]
Orthopedic limb surgery
Subanalgesic doses of ketamine—i.v. bolus
0.3 mg/kg followed by i.v. infusion 0.2 mg/kg/h, which provides a strong analgesic effect.
[113,114,115]
CancerBreakthrough cancer pain
for sudden, severe pain, an additional dose
of ketamine can be given sublingually or intranasally 10–50 mg; rapid pain relief, especially for patients who do not tolerate opioids well.
[116,117,118]
Advanced cancer pain—palliative therapy
ketamine can be used as an add-on to opioids to help control pain: i.v., either as a continuous infusion
or as a bolus (single dose): continuous infusion
0.1–0.3 mg/kg per hour or bolus 0.25 mg/kg
every 8 h; pain relief and improved quality of life, especially in patients with opioid resistance.
[119,120,121]
In 2020, ketamine was approved by the World Health Organization (WHO) as a drug for the treatment of refractory neuropathic pain, the use of which is intended to reduce the doses of opioids used and serve as an alternative for people with an increased tolerance to opioids. However, it should be remembered that ketamine is not a first-line agent; it is a strong drug, intended for people who do not benefit from other therapeutic methods.

7. Ketamine as Potential Therapy for Addiction

Despite long-term pharmacological research on the creation of new substances, addiction still remains an unresolved important social and clinical problem. Furthermore, due to the complex nature of this disease, currently available drugs possess only limited effectiveness. This therapy is significantly complicated by the common co-occurrence of depression with addictions [122,123,124,125,126,127]. It is well known that untreated depression may cause or intensify addiction, and taking psychoactive substances may intensify or even lead to the development of depression. Furthermore, high levels of depression and anxiety may predispose not only to addiction, but also to relapse even after a long period of abstinence [126,128,129,130]. Therefore, scientists and clinicians pay attention to the possible benefits of using antidepressants in the treatment of addictions, as drugs enhancing abstinence. It seems that ketamine could be an interesting alternative in the treatment of addictions, especially since in many cases, conventional antidepressants do not reduce alcohol consumption [131,132]. This is mainly explained by the fact that the effects of these drugs are delayed in time.
As confirmation of these assumptions, recent decades have brought many new reports about the use of ketamine beyond its previous indications. Increasing number of studies suggest that it has a multifaceted effect that can be used, among other purposes, in the treatment or at least alleviation of some symptoms of addiction. Moreover, ketamine is characterized by properties different from most typical antidepressants and has a unique mechanism of action, which suggests that it may improve the ability to establish and maintain abstinence in substance use disorders.
It was well known that diminished glutamatergic synaptic transmission and reduced plasticity are associated with addiction [133,134]. Therefore, ketamine, which acts primarily by influencing glutamatergic transmission, may be particularly helpful in this therapy. Thus far, it has been suggested that the possible mechanisms by which ketamine may have a beneficial effect in addiction include the following: (1) enhancement of neuroplasticity and neurogenesis, (2) disruption of relevant functional neural networks, (3) treating depressive symptoms, (4) blocking re-consolidation of drug-related memories, (5) provoking mystical experiences, (6) enhancing psychological therapy efficacy.
Existing models suggest that ketamine’s blockade of NMDA receptors increases synaptogenesis by stimulating protein synthesis and the insertion of AMPA receptors [83,133,135]. Hence, ketamine’s effects help to reverse the glutamatergic changes associated with depression and addiction. The fact that the administration of rapamycin (an mTOR antagonist) blocks both the ketamine-induced reduction in alcohol intake [135] and the antidepressant effects [84,136] suggests that the underlying mechanism of both effects might be the same: synaptogenesis. Moreover, a previous study showed that ketamine’s metabolites, (R,S)-norketamine and (2S,6S)-hydroxynorketamine, contribute to ketamine-mediated increase in mTOR signaling both in vivo in rats and in vitro [137]. Therefore, this represents a mechanism by which ketamine could redress an imbalance in addiction. However, there are also contradictory results published by Zanos et al. [125], which indicate that antidepressant effects are independent of mTOR levels. Therefore, further research is needed to assess the relationship between changes in synaptogenesis and any effects of ketamine administration.
In addition, it has been shown that addictions are connected to disorders of neurogenesis, especially in cortical and hippocampal structures which may influence levels of self-administration and the vulnerability to relapse [138,139]. This process may be related, among others, to a reduction in BDNF serum levels [140] observed, among others, in groups of people addicted to cocaine and heroin [141,142,143]. Ketamine has been previously shown to affect the level of BDNF, and this mechanism is one of the key factors in its antidepressant action [39,91,92,144]. Hence, its use may also be helpful in combating addictions through this mechanism [145,146].
Ketamine also disrupts drug-cue memories via the manipulation of reconsolidation processes [93,94,95,96,97,143,147]. Recent reviews have suggested that ketamine may be able to disrupt maladaptive appetitive memories [93,94,95]. It is interesting that many authors indicate the specific involvement of mystical experience as induced by ketamine in the therapeutic mechanism [93,94,95,96].
This suggests that the combination of ketamine with motivational enhancement therapy may be an effective pharmacotherapy for initiating and sustaining abstinence. Indeed, this type of therapy, known as ‘Ketamine Psychedelic Therapy’ (KPT), has been used, among others, in patients with drug addiction, but also in people with other types of addictions (e.g., food addiction) and mental disorders [148,149]. It was suggested that ketamine can provide a unique mental state that facilitates and enriches therapeutic experiences, which in turn may improve efficacy and lengthen treatment effects [35,95]. The authors suggest that the subjective psychedelic experience seemed to help the addict to undergo a cathartic process, improve relationships with the world and other people, maintain positive psychological changes and enhance self-awareness and personal growth [149,150,151,152,153]. These changes are considered as favorable for promoting abstinence. Furthermore, patients feel less depressed and anxious, more self-confident and more emotionally open. It should be emphasized that ketamine infusions facilitate psychological therapy presumably due an increase in synaptogenesis and neurogenesis, and thus improved learning of relapse-reducing strategies [154]. However, some researchers suggest that despite the very promising effects observed during the use of KPT, it should be pointed out that a ketamine-induced psychedelic experience may have only marginal and transitory beneficial effects in and of itself, no beneficial effects at all or may be harmful when ketamine is used in uncontrolled settings recreationally, hence leading to significant medical problems and addiction [155,156,157]. These findings suggest a new usefulness for ketamine in facilitating addiction treatment and reducing the risk of relapse, namely, by maintaining motivation for sobriety even in the face of stressors and challenges.
Collectively, these studies reveal that ketamine may improve the ability to establish and maintain abstinence in substance use disorder. The presented findings (Table 4) may have a significant impact on the development of new treatments of addiction, as addiction is a complex condition that currently presents challenges for successful treatment.
In addition to the indicated action in reducing the effects of addiction, ketamine is characterized by having a much more favorable dosing method compared to classic pharmacotherapy. Daily administration is not needed, and this significantly improves treatment, since it is less stigmatizing than the requirement to take daily medication. The primary problem may be the route of its administration. Most published data concern administration via injection, which significantly limits its use and increases the costs of treatment. The search for a less invasive method of administration that would also limit the need to use highly specialized centers and would therefore be available to a wider group of patients is still ongoing. Currently, it seems that intranasal administration will be the optimal method of administration and widely expand the availability of ketamine treatment [173]. Furthermore, in the case of attempts to introduce ketamine into the pharmacotherapy of addictions, evaluations of the optimal dose and frequency schedules are also needed. Most of the studies to date have used prior depression trial dosages of 0.5–0.8 mg/kg i.v. ketamine, although a few studies utilized doses of 2–2.5 mg/kg i.m.
However, despite its many advantages, the effects of ketamine require further research for the following reasons:
  • Some of the presented studies used only small populations of naïve individuals, lacked inactive placebo groups or were relatively homogeneous in terms of ethnicity, age and gender. Thus, the effects of ketamine administration early in life have not yet been clearly established, and it is possible that ketamine administration to adolescents for the treatment of depression may lead to an increased risk of addiction later in life.
  • In the case of ethanol addiction, a very important element is the occurrence of potential interactions and co-dependency especially in that ketamine has become popular as a recreational drug, sometimes used with alcoholic beverages or stimulants.
  • The issue of the addictive effect of ketamine and the possibility of its therapeutic use in controlled conditions without causing addiction also remains unresolved.
However, as previously stated, extensive clinical trials are essential for determining the optimal dosing strategies, identifying biomarkers linked to therapeutic outcomes and evaluating the long-term risks associated with repeated administration. Despite these challenges, the available data represent a promising advancement in understanding the potential role of ketamine in addiction treatment.

8. Present Primary Pharmacotherapy of Depression

Depressive disorders represent one of the greatest challenges of modern medicine and pharmacy. According to data released in 2021, depression affects approximately 280 million people worldwide and is one of the most common causes of life-limiting disability [174]. However, its pharmacotherapy is still problematic due to the frequent unresponsiveness of patients to the available treatments [175]. Treatment includes mainly drugs belonging to the selective serotonin reuptake inhibitor (SSRI) and serotonin norepinephrine reuptake inhibitor (SNRI) groups, with the mechanism of actions of these corresponding to monoaminergic theory. However, the disadvantages held by these drugs may affect the efficacy of pharmacotherapy, leading to discontinuation, as well as impairing some areas of functioning. Such drugs are characterized by latency of action, the potential for worsening of the patient’s condition at the start of pharmacotherapy and sexual or cognitive dysfunction. Furthermore, drugs from the SSRI group may prolong bleeding time and induce teratogenic effects. Despite their relative efficacy, they are met with treatment non-response in some patients [174]. It is estimated that more than 30% of all patients with major depressive disorder (MDD) do not achieve satisfactory treatment results, and this outcome is associated with a reduced quality of life and increased deaths from suicide. Furthermore, it is noted that depression resistance to at least two antidepressants indicated by standard pharmacotherapy—TRD—is an increasingly occurring problem and a serious challenge for pharmacotherapy [176]. The exact causes determining treatment resistance are still not known, but, presumably, inflammation within the CNS and impaired neuroplasticity are involved in this process [40,177]. The lack of efficacy of conventional therapy leads to a search for drugs with new therapeutic targets. Currently, drugs targeting the glutamatergic and γ-aminobutyric acid (GABA)-ergic systems are being widely investigated [174]. Attention is also focused on drugs involved in abolishing inflammation in the CNS [178]. The discovery of the antidepressant effect of ketamine has led to the development of research into its use in this therapeutic direction [1].

9. The Mechanism of Antidepressant Activity of Ketamine

In the context of antidepressant action, ketamine selectively blocks NMDA receptors on GABA-ergic interneurons, which results in the stimulation of various neurotransmission in other regions involved in mood regulation (Figure 2). The decrease in GABA release reduces the inhibitory effect of GABA on the glutamatergic system and causes increased presynaptic glutamate secretion. Glutamate then stimulates AMPA receptors [7] and this effect may be responsible for the antidepressant activity of ketamine (but not other antidepressants, suggesting a unique mechanism of action) since the administration of an AMPA receptor antagonist inhibits this effect in behavioral tests. The inhibition of NMDA receptor-dependent spontaneous impulsation in the hippocampus may also be associated with antidepressant effects. This action leads to changes in protein synthesis and increased neurotransmission in the CA1 region of the hippocampus. Ketamine can inhibit extrasynaptic NMDA receptors on pyramidal neurons and thus prevent their activation by decreasing glutamate levels in the extracellular space. This leads to disinhibition of the mechanistic target for rapamycin (mTOR) protein synthesis, which ultimately induces an antidepressant effect. Furthermore, NMDA receptors on pyramidal neurons mainly contain GluN2B subunits, and in mice lacking GluN2B, no antidepressant effect was observed. Additionally, in humans, the administration of selective inhibitors of the GluN2B subunit of NMDA receptors induces antidepressant effects [8]. The antagonism of NMDA receptors leads to the inhibition of lateral habenula (LHb) neurons’ activity [8]. This region is involved in the formation of reward, aversion emotion and cognitive processes [179] and may be activated under aversive environmental stressors, which significantly promote the manifestation of depressive symptoms. The inhibition of LHb activity by ketamine has been found in vitro, while in vivo studies in rats have shown an acute antidepressant effect correlated with these properties [8].
However, there are also studies demonstrating mechanisms independent of NMDA receptor inhibition [7], as other NMDA receptor antagonists do not show antidepressant effects as rapid and intense as those of ketamine [8,180]. In addition, partial agonists such as rapistenol and D-cycloserine are devoid of ketamine’s side effects associated with NMDA receptor inhibition. Furthermore, R-ketamine has an approximately four-fold lower affinity for NMDA receptors than S-ketamine, but induces more potent and longer-lasting antidepressant effects. Researchers also highlight the role of its metabolite S-HNK, while chemically modified ketamine, with reduced metabolism to S-HNK and retaining affinity for the NMDA receptor, induced no antidepressant effects in behavioral tests. Additionally, in this study, a stronger antidepressant effect was observed in rat females with higher brain exposure to S-HNK, while there were no differences in ketamine concentrations between the two genders [8,180].
Other neurochemical pathways involving elongation factor kinase 2 in eukaryotic cells (eEF2K) and BDNF levels are also thought to be involved in this mechanism of action [8]. Ketamine can increase BDNF levels by affecting eEF2K activity. It has been demonstrated that BDNF is essential for the antidepressant activity of ketamine, and mutations in genes crucial for the secretion and activity of this factor in humans and mice resulted in a lack of antidepressant effect. Furthermore, it determines neuroplasticity and influences the formation of synaptic connections. Active eEF2K, regulated by NMDA receptor activity, inhibits the substrate elongation factor in eukaryotic cells (eEF2) which inhibit the translation of BDNF. Ketamine, by inhibiting spontaneous NMDA receptor neurotransmission, deactivates eEF2K, while subsequently activating eEF2 and suppressing protein synthesis. Previous behavioral tests showed that the administration of eEF2K inhibitors to mice reduced depressive symptoms. In addition, no increase in drug-induced BDNF levels was observed in animals lacking genes involved in eEF2K expression. However, attention is drawn to the finding that the effect of ketamine in this case may also be NMDA receptor-independent. The peripheral administration of the ketamine metabolite HNK also causes eEF2 activation and an associated increase in BDNF levels in NMDA receptor-independent mechanisms. Protein synthesis can also be affected by mTOR kinase which is involved in the regulation of neurogenesis, initiates protein translation and synthesis and is crucial for the regulation of synaptic plasticity. Its action is dictated by activation of the mTOR1 complex [8]. The induction of pathways involving mTOR can occur as a result of an increase in BDNF levels, the activation of the receptor for this factor or the activation of tropomyosin kinase (TrkB) receptors by BDNF [8,180]. Interestingly, it was observed that the administration of a selective mTOR inhibitor abolishes the antidepressant effect of ketamine and the synaptic changes. It is thought that activation of the mTOR pathway may be connected to the inhibition of another factor—glycogen synthesis kinase-3 (GSK-3). Mutation in GSK-3-relevant genes prevents activation of the mTOR pathway, but, interestingly, this is not observed after the administration of GSK-3 inhibitors. This situation may be explained by attributing ketamine’s function to the activation of AKT kinase (protein kinase B), which, in part, regulates GSK-3 activity, since it was observed that the inhibition of AKT activity abolishes the antidepressant effect of ketamine (inhibiting GSK-3 and mTOR activity) [8].

10. Current Position of Ketamine in the Treatment of Depression

Both the racemic form and its two independent enantiomers produce surprising improvements in patient mood. As a result, Johnson & Johnson successfully formulated S-ketamine in an intranasal form (Spravato®), which was subsequently approved by the FDA in 2019 for the treatment of TRD in the USA and Europe [179]. In 2020, it was approved also for the treatment of MDD with associated suicidal thoughts or behavior [181]. Furthermore, the i.v. administration of racemic ketamine is used off-label for the treatment of depression [182]. Therefore, S-ketamine is not a drug of first or second choice, but is recommended in situations where time is of the essence, e.g., suicide risk or hospitalization. Nonetheless, its great advantage is its fast antidepressant effects, observed after a few hours or days. In contrast, the lack of sufficient long-term data on its safety and effectiveness raises concerns. Spravato® is only used in adult patients and they self-administer it under direct supervision to reduce the symptoms of depression, together with another antidepressant. It is prescribed when the patient has tried at least two other antidepressants without improvement (Figure 3). A doctor or other healthcare professional will monitor the patient each time they use Spravato®. The preparation contains 28 mg of S-ketamine per device (dispenser). During each session, the patient receives from one to three dispensers. The dose is determined individually, most often it is 56 mg, i.e., two dispensers. Administration takes a few minutes (the medicine is administered every 5 min). The patient remains in the facility under observation, for at least 2 h, until any side effects such as disturbances of consciousness and blood pressure, dizziness and headaches, nausea, vomiting, or anxiety disappear. It is not recommended for patients to drive or operate any machines until the end of the day after the administration of the drug. The entire treatment is supervised by a psychiatrist [183].

11. The Antidepressant Effect of Ketamine in Animal Studies

In animal studies, the antidepressant effect of ketamine has been shown in a variety of behavioral tests [114]. In a group of rats receiving ketamine or desipramine intraperitoneally (i.p.), Koike and colleagues observed a decrease in the number of failures in the learned helplessness test and a reduction in the time of immobility in the tail suspended test (TST) 30 min after administration. However, only the administration of ketamine obtained similar results in tests performed 72 h after administration, indicating its long duration of action compared to desipramine. Furthermore, ketamine did not affect the locomotor activity of the animals [185]. Subsequently, Jiang et al. demonstrated the antidepressant effect of ketamine in rats 24 h after administration in the standard penetration test (SPT), FST, elevated plus maze test (EPM) and water maze test (WMT), but the effect did not persist with long-term treatment after 7 weeks [186]. However, it should be emphasized that in a group of growing rats, the drug showed stronger and longer-lasting effects as determined by increased locomotion compared to adult rodents, which may suggest a higher potential for addiction in juveniles [187]. In mice, an antidepressant effect was observed in the forced swim test (FST) [64]. In addition, ketamine administration increased saccharose preference in test animals, indicating a reduction in the anhedonia often associated with depression. Moreover, a comparative study of the two enantiomers showed a stronger antidepressant effect of R-ketamine than S-ketamine. In addition, for S-ketamine, a single administration caused a decrease in PV-positive cells in the brain which is associated with psychotomimetic effects and impaired cognitive function, but the changes were not observed for R-ketamine. This evidence may indicate a potential role for R-ketamine in the treatment of depression since it does not induce psychotomimetic effects [63]. Interestingly, in animal models, some researchers have demonstrated neurotoxic effects of ketamine, but no such effects have been found in humans. Furthermore, many of the results from animal studies are inconclusive and have not been reflected in subsequent clinical studies, and thus should be interpreted with caution [188].

12. Ketamine in Clinical Trials

Analysis of the clinical trials conducted to date demonstrates the efficacy of ketamine in the treatment of TRD [189]. The antidepressant effect in most patients begins within 24 h of administration and persists for approximately 1 week [190]. Satisfactory effects of pharmacotherapy have been found in both groups: low- and high-resistance patients treated with standard antidepressants. For intravenous ketamine, the effective antidepressant effect in people with lower levels of resistance (resistance up to three antidepressants) is achieved at a dose of 0.5 mg/kg. However, in a group of resistance above three drugs, it is suggested that the dose needs to be increased to 0.75 mg/kg to achieve a long-lasting effect. Studies conducted in different age groups showed an increase in efficacy in patients with an earlier age of onset (<55 years). However, patients in the 65–74 age range also showed satisfactory improvement. On the contrary, a decrease in the effect of ketamine was observed in the age group > 75. These studies indicate a better prognosis when ketamine treatment is initiated earlier when resistance to standard treatment regimens is suspected [189]. Ketamine has also been shown to effectively abolish suicidal thoughts often associated with depression and incidents of hospitalization. To reduce anxiety, standard treatments include benzodiazepines such as midazolam, which some patients do not respond to. Patients unresponsive to midazolam have been shown to experience rapid improvement after i.v. ketamine. The researchers concluded that the use of ketamine would significantly reduce the time and number of hospitalizations and associated costs [191]. However, in the study of Zhang and Hashimoto, 2022 [181], patients showed a similar response to treatment after i.v., s.c. and i.m. administration. After oral administration, the antidepressant effects were slower and observed only after 14 days of use, which may be related to the lower bioavailability, while a slightly higher bioavailability is reported with sublingual administration [190]. Notwithstanding, this form of administration requires further study, but the results to date indicate its efficacy and safety [192].
Additionally, clinical trials using S-ketamine as a treatment were run for patients enrolled in the Risk Evaluation and Mitigation Strategy (REMS) program [193], and the efficacy of each of the doses currently in use was confirmed in phase II clinical trials. These were multicenter, double-blind, placebo-controlled studies. Evaluation was based on the difference in pre- and post-treatment scores on the Montgomery–Asberg Depression Rating Scale (MADRS) [193]. Phase III studies evaluated the efficacy and safety of S-ketamine spray together with a newly included oral antidepressant [193]. The initiative was supported by the FDA based on patient experience and preference data and an analysis of possible harms and benefits. The data came from patients entered in the phase III trial who had already had experience with ketamine and from patients who had not taken it, and was obtained through an online patient portal [189]. Subsequently, the short-term TRANSFORM studies were aimed at assessing efficacy and safety. Depression severity was assessed using the MADRS scale. The TRANSFORM-1 and TRANSFORM-2 study additionally considered the patients’ level of irritability, which affects 30–50% of all patients struggling with a major depressive episode and exacerbates the escalation of suicidal thoughts. The level of irritability was assessed using the 7-item Generalized Anxiety Disorder Scale (GAD-7). The newly included oral antidepressants were duloxetine, escitalopram, sertraline and extended-release venlafaxine [177]. The TRANSFORM-3 study was conducted following the same regimen as TRANSFORM-2 with flexible dosing and included only elderly subjects aged ≥65 [193,194,195,196,197] (Table 5 and Table 6).
A greater improvement was observed in the group receiving S-ketamine plus another antidepressant than in the placebo group. It was more pronounced in those aged 18–64 than in those > 64 years. Additionally, S-ketamine was more effective in those with an earlier age of onset (<55) [196,197]. Likewise, extended clinical trials have assessed the efficacy and safety of long-term S-ketamine use, including the potential for addiction and withdrawal symptoms [193,198].
SUSTAIN-1 focused on assessing long-term efficacy [193]. Basic information about the study conducted is assembled in Table 7. The recurrence of relapse after S-ketamine treatment is shown in Table 8.
The researchers estimated that treatment with S-ketamine reduces the risk of relapse by 51% in those with stable remission and by 70% in those with a stable response, compared to placebo [190].
SUSTAIN-2 was conducted on adults of all ages using flexible S-ketamine dosing [198]. Basic information about the study conducted is assembled in Table 9. A summary of the important results and adverse effects is provided in Table 9, Table 10 and Table 11.
In this study, patients were also administered a second antidepressant. The trial included an induction phase (4 weeks) in which S-ketamine was administered twice a week, a maintenance phase (48 weeks) in which patients received this drug once a week and then a follow-up phase of up to one year in which patients did not receive S-ketamine and continued treatment with another antidepressant [124]. S-ketamine proved to be effective in treating depression, but, importantly, some patients only achieved remission during the maintenance phase [124]. Similarly, in the ASPIRE and ASPIRE II long-term study, not all patients showed an early response to treatment. The greatest response was found on day 25 of the study (74.6%). After 24 h, only 35.3% of all patients showed a response. In the retrospective, multicenter observational study REAL-ESK without a placebo, 38% of all patients not showing a response after one month, had a remission after three months of treatment [126]. This shows that S-ketamine, which initially appears to be ineffective in some people, may produce satisfactory results after prolonged use, and that a lack of response after the initial doses is not an indication for discontinuing pharmacotherapy [127].
In all short-term studies, adverse effects were more frequent in groups receiving S-ketamine plus an antidepressant than in the placebo group, and these effects were classified as mild or moderate. There were no significant differences between the 56 mg S-ketamine dose group and the 84 mg dose group [120]. The most common symptoms in patients aged 18–64 are shown in Table 11.
In the TRANSFORM-3 study, adverse effects were reported by 70.8% of all patients receiving S-ketamine and 60% of all patients in the placebo group. The most common reports were dizziness (20.8%), nausea (18.1%) and dissociation (12.5%). An increase in blood pressure occurred in 12.5% of all patients in the S-ketamine plus antidepressant group and resolved over a 2 h period in most patients. There were no changes in laboratory parameters, electrocardiogram or nasal mucosa. Moreover, no cases of bladder interstitial inflammation occurred [197]. The adverse effects in the long-term study were similar to those observed in the short-term study and had the same severity (Table 12 and Table 13) [198,199,200,201].
It should be emphasized that none of the patients asked for an increase in the dose of the drug and there was no recorded desire to obtain the drug on their own (Table 10) [127]. Two patients died during the study; however, this was found to be unrelated to the treatment [201].
The prospective STRIVE evaluation examined overall patient satisfaction with S-ketamine treatment in terms of mood and effects on functioning. It included patients who had previously participated in clinical trials such as TRANSFORM and SUSTAIN. Patients gave positive feedback on S-ketamine treatment and judged it significantly better compared to previous conventional pharmacotherapy [202]. One study [203] extended the spectrum of research to use ketamine in the prevention of postpartum depression. The results show a decrease in postpartum depression in women who received a low dose of ketamine i.v. (0.5 mg/kg + 2 mL physiological salt) after the postpartum period. The 138 women were divided into two equal groups. The control group received physiological salt only. Women who received ketamine had lower Edinburgh Postnatal Depression Scale (EPDS) scores and significantly fewer incidences of postnatal depression. Ketamine also appeared to be safe and there were no significant differences in side effects between the two groups [132].
Undoubtedly, S-ketamine in the form of a nasal spray is of great interest and so far is the only form registered for the treatment of depression [182]. It can be used by patients with the assistance of medical staff and, in the future, offers the possibility for use in outpatient therapy [199]. Despite the proven efficacy of S-ketamine in clinical trials, treatment-related side effects are still an issue. Recently, R-ketamine has become of interest to researchers [105]. Although it has a lower affinity for NMDA receptors, in rodent studies, it showed a stronger and longer-lasting antidepressant effect than racemic ketamine and S-ketamine. Importantly, this fact did not correlate with an increase in the potency of adverse effects. R-ketamine showed a lower psychotomimetic and dissociative potential than S-ketamine [181,204], presumably as a result of its lower ability to stimulate dopamine release [30]. Based on the available results, it is believed that the antidepressant effect of R-ketamine is not related to NMDA antagonism [181,204], since R-ketamine induces activity within different brain structures than S-ketamine, (R,S)-ketamine and an NMDA antagonist in magnetic resonance imaging (fMR) [205]. However, in the first open pilot study in patients with MDD, R-ketamine showed higher response and remission rates compared to (R,S)-ketamine and S-ketamine. Furthermore, it induced only minor side effects observed as a transient visual impairment and dizziness. Notwithstanding, due to the small size of the study group and the lack of a reference sample, further studies on its clinical efficacy and safety are needed, but, nevertheless, the results obtained seem promising [204].

13. Summary

Ketamine, originally used for anesthesia, has gained significant attention in recent years for its potential in treating pain, depression and addiction. In the context of pain management, ketamine may be an alternative treatment for patients with opioid-resistant pain. Additionally, ketamine produces rapid and robust antidepressant effects, especially in patients with TRD. Current research has indicated that LDK can provide fast-acting relief, often within hours, through the modulation of NMDA receptors and increased synaptic plasticity. This has positioned ketamine as a promising therapeutic option for patients who do not respond to conventional antidepressants. In the realm of addiction, ketamine’s ability to disrupt maladaptive neural circuits offers hope for treatment strategies in substance use disorders. Studies suggest that ketamine-assisted psychotherapy supports a reduction in craving and promotes abstinence, making it a potential adjunct in addiction treatment; however, further clinical trials are needed to confirm this efficacy.
Despite these promising findings, further studies are essential to fully understand the long-term effects of ketamine administration, including its safety profile and potential for abuse. Investigating its mechanisms at a molecular level (including the role of its metabolites), the sustainability of its therapeutic effects over time and its neuroprotective effects could open new treatment possibilities for patients with limited therapeutic options.

Author Contributions

Conceptualization, E.G.-T. and E.K.; writing—original draft preparation, E.G.-T., A.W., M.T., P.S. and E.K.; writing—review and editing: E.G.-T., J.H.K. and E.K.; visualization: E.G.-T. and E.K.; supervision: E.G.-T., J.H.K. and E.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Medical University of Lublin (grant number DS 19).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Domino, E.F.; Warner, D.S. Taming the ketamine tiger. Anesthesiology 2010, 113, 678–684. [Google Scholar] [CrossRef] [PubMed]
  2. Ross, S. Ketamine and addiction. Prim. Psychiatry 2008, 15, 61–69. [Google Scholar]
  3. Radvansky, B.M.; Puri, S.; Sifonios, A.N.; Eloy, J.D.; Le, V. Ketamine-A narrative review of its uses in medicine. Am. J. Ther. 2016, 23, 1414–1426. [Google Scholar] [CrossRef]
  4. Adams, H.A. Wirkmechanismen von Ketamin Mechanisms of action of ketamine. Anaesthesiol. Reanim. 1998, 23, 60–63. [Google Scholar]
  5. White, P.F.; Schüttler, J.; Shafer, A.; Stanski, D.R.; Horai, Y.; Trevor, A.J. Comparative pharmacology of the ketamine isomers. Studies in volunteers. Br. J. Anaesth. 1985, 57, 197–203. [Google Scholar] [CrossRef]
  6. Ebert, B. Differential pharmacology of ketamine enantiomers. Anesth. Analg. 1997, 85, 119–124. [Google Scholar]
  7. Colla, M.; Scheerer, H.; Weidt, S.; Seifritz, E.; Kronenberg, G. Novel insights into the neurobiology of the antidepressant response from ketamine research: A mini review. Front. Behav. Neurosci. 2021, 15, 759466. [Google Scholar] [CrossRef] [PubMed]
  8. Zanos, P. Ketamine and ketamine metabolite pharmacology: Insights into therapeutic mechanisms. Trends Pharmacol. Sci. 2018, 39, 591–601. [Google Scholar] [CrossRef]
  9. Nichols, K.A.; Paciullo, C.A. Subdissociative Ketamine Use in the Emergency Department. Adv. Emerg. Nurs. J. 2019, 41, 15–22. [Google Scholar] [CrossRef]
  10. BinKharfi, M.; AlSagre, A. BET 2: Safety and efficacy of low-dose ketamine versus opioids for acute pain management in the ED. Emerg. Med. J. 2019, 36, 128–129. [Google Scholar] [CrossRef]
  11. Kapur, A.; Kapur, V. Conscious Sedation in Dentistry. Ann. Maxillofac. Surg. 2018, 8, 320–323. [Google Scholar] [CrossRef] [PubMed]
  12. 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] [PubMed]
  13. Lim, D.K. Ketamine associated psychedelic effects and dependence. Singap. Med. 2003, 44, 31–34. [Google Scholar]
  14. Zou, L.; Tian, S.Y.; Quan, X.; Ye, T.H. Psychodelic effects of subanesthetic doses of ketamine. Zhongguo Yi XueKe Xue Yuan Xue Bao 2009, 31, 68–72. [Google Scholar]
  15. Sørensen, A.G.; Barnung, S.; Rasmussen, L.S. Ketamingenopdagetafbådelægerogmisbrugere [Ketamine is used again by both physicians and addicts]. Ugeskr. Laeger 2011, 173, 2123–2126. [Google Scholar] [PubMed]
  16. Sin, B.; Ternas, T.; Motov, S.M. The use of subdissociative-dose ketamine for acute pain in the emergency department. Acad. Med. 2015, 22, 251–257. [Google Scholar] [CrossRef]
  17. Lumanauw, D.D.; Youn, S.; Horeczko, T.; Yadav, K.; Tanen, D.A. Subdissociative-dose Ketamine Is Effective for Treating Acute Exacerbations of Chronic Pain. Acad. Med. 2019, 26, 1044–1051. [Google Scholar] [CrossRef]
  18. Puri, L.; Morgan, K.J.; Anghelescu, D.L. Ketamine and lidocaine infusions decrease opioid consumption during vaso-occlusive crisis in adolescents with sickle cell disease. Curr. Opin. Support. Palliat. Care 2019, 13, 402–407. [Google Scholar] [CrossRef]
  19. Mion, G. History of anaesthesia: The ketamine story—Past, present and future. Eur. J. Anaesthesiol. 2017, 34, 571–575. [Google Scholar] [CrossRef]
  20. Potter, D.E.; Choudhury, M. Ketamine: Repurposing and redefining a multifaceted drug. Drug Discov. Today 2014, 19, 1848–1854. [Google Scholar] [CrossRef]
  21. Desta, Z.; Moaddel, R.; Ogburn, E.T.; Xu, C.; Ramamoorthy, A.; Venkata, S.L.; Sanghvi, M.; Goldberg, M.E.; Torjman, M.C.; Wainer, I.W. Stereoselective and regiospecific hydroxylation of ketamine and norketamine. Xenobiotica 2012, 42, 1076–1087. [Google Scholar] [CrossRef] [PubMed]
  22. Rao, L.K.; Flaker, A.M.; Friedel, C.C.; Kharasch, E.D. Role of Cytochrome P4502B6 Polymorphisms in Ketamine Metabolism and Clearance. Anesthesiology 2016, 125, 1103–1112. [Google Scholar] [CrossRef] [PubMed]
  23. Andrade, C. Ketamine for Depression, 5: Potential Pharmacokinetic and Pharmacodynamic Drug Interactions. J. Clin. Psychiatry 2017, 78, 858–861. [Google Scholar] [CrossRef] [PubMed]
  24. ILangmia, M.; Just, K.S.; Yamoune, S.; Müller, J.P.; Stingl, J.C. Pharmacogenetic and drug interaction aspects on ketamine safety in its use as antidepressant—Implications for precision dosing in a global perspective. Br. J. Clin. Pharmacol. 2022, 88, 5149–5165. [Google Scholar] [CrossRef]
  25. Hijazi, Y.; Boulieu, R. Protein binding of ketamine and its active metabolites to human serum. Eur. J. Clin. Pharmacol. 2002, 58, 37–40. [Google Scholar] [CrossRef]
  26. Yanagihara, Y.; Kariya, S.; Ohtani, M.; Uchino, K.; Aoyama, T.; Yamamura, Y.; Iga, T. Involvement of CYP2B6 in n-demethylation of ketamine in human liver microsomes. Drug Metab. Dispos. 2001, 29, 887–890. [Google Scholar]
  27. Edwards, S.R.; Mather, L.E. Tissue uptake of ketamine and norketamine enantiomers in the rat: Indirect evidence for extrahepatic metabolic inversion. Life Sci. 2001, 69, 2051–2066. [Google Scholar] [CrossRef]
  28. Dinis-Oliveira, R.J. Metabolism and metabolomics of ketamine: A toxicological approach. Forensic Sci. Res. 2017, 2, 2–10. [Google Scholar] [CrossRef]
  29. Rosenbaum, S.B.; Gupta, V.; Patel, P.; Palacios, J.L. Ketamine. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024; Volume 30. [Google Scholar]
  30. Hashimoto, K. Molecular mechanisms of antidepressant actions of ketamine enantiomers and their metabolites. J. Pharmacol. 2019, 176, 1452–1456. [Google Scholar]
  31. Yang, C.; Kobayashi, S.; Nakao, K.; Dong, C.; Han, M.; Qu, Y.; Ren, Q.; Zhang, J.C.; Ma, M.; Toki, H.; et al. AMPA receptor activation-independent antidepressant actions of ketamine metabolite (S)-norketamine. Biol. Psychiatry 2018, 84, 591–600. [Google Scholar] [CrossRef]
  32. Craven, R. Ketamine. Anaesthesia 2007, 62, 48–53. [Google Scholar] [CrossRef] [PubMed]
  33. Barrett, W.; Buxhoeveden, M.; Dhillon, S. Ketamine: A versatile tool for anesthesia and analgesia. Curr. Opin. Anaesthesiol. 2020, 33, 633–638. [Google Scholar] [CrossRef]
  34. Weber, F.; Wulf, H.; Gruber, M.; Biallas, R. S-ketamine and s-norketamine plasma concentrations after nasal and i.v. administration in anesthetized children. Paediatr. Anaesth. 2004, 14, 983–988. [Google Scholar] [CrossRef] [PubMed]
  35. Clemens, J.; Nimmo, W.; Grant, I. Bioavailability, pharmacokinetics, and analgesic activity of ketamine in humans. Pharm. Sci. 1982, 71, 539–542. [Google Scholar] [CrossRef]
  36. Hasan, M.; Modess, C.; Roustom, T.; Dokter, A.; Grube, M.; Link, A.; Rey, H.; Adler, S.; Meissner, K.; Siegmund, W. Chiral Pharmacokinetics and Metabolite Profile of Prolonged-release Ketamine Tablets in Healthy Human Subjects. Anesthesiology 2021, 35, 326–339. [Google Scholar] [CrossRef]
  37. Chong, C.; Schug, S.A.; Page-Sharp, M.; Jenkins, B.; Ilett, K.F. Development of a sublingual/oral formulation of ketamine for use in neuropathic pain: Preliminary findings from a three-way randomized, crossover study. Clin. Drug Investig. 2019, 29, 317–324. [Google Scholar] [CrossRef] [PubMed]
  38. Blonk, M.I.; Koder, B.G.; 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]
  39. Rolan, P.; Lim, S.; Sunderland, V.; Liu, Y.; Molnar, V. The absolute bioavailability of racemic ketamine from a novel sublingual formulation. Br. J. Clin. Pharmacol. 2014, 77, 1011–1016. [Google Scholar] [CrossRef]
  40. Yanagihara, Y.; Ohtani, M.; Kariya, S.; Uchino, K.; Hiraishi, T.; Ashizawa, N.; Aoyama, T.; Yamamura, Y.; Yamada, Y.; Iga, T. Plasma concentration profiles of ketamine and norketamine after administration of various ketamine preparations to healthy Japanese volunteers. Biopharm. Drug Dispos. 2003, 24, 37–43. [Google Scholar] [CrossRef]
  41. Malinovsky, J.M.; Servin, F.; Cozian, A.; Lepage, J.Y.; Pinaud, M. Ketamine and norketamine plasma concentrations after iv, nasal and rectal administration in children. Br. J. Anaesth. 1996, 77, 203–207. [Google Scholar] [CrossRef]
  42. 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]
  43. 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]
  44. Yeaman, F.; Meek, R.; Egerton-Warburton, D.; Rosengarten, P.; Graudins, A. Sub-dissociative-dose intranasal ketamine for moderate to severe pain in adult emergency department patients. Emerg. Med. 2014, 26, 237–242. [Google Scholar] [CrossRef]
  45. 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]
  46. Bitter, C. Pharmacokinetics and Pharmacodynamics of Nasally Applied s-Ketamine. Ph.D. Thesis, The University of Basel, Basel, Switzerland, 2010. Available online: https://edoc.unibas.ch/1310/1/20110314_1408_DissCB_e_version.pdf (accessed on 16 December 2014).
  47. Peltoniemi, S.; Hagelberg, L.; Kurkinen, K.J.; Neuvonen, P.J.; Olkkola, K.T. Rifampicin has a profound effect on the pharmacokinetics of oral S-ketamine and less on intravenous S-ketamine. Basic Clin. Pharmacol. Toxicol. 2012, 111, 325–332. [Google Scholar] [CrossRef] [PubMed]
  48. Perez-Ruixo, C.; Rossenu, S.; Zannikos, P.; Nandy, P.; Singh, J.; Drevets, W.C. Perez-Ruixo, J.J. Population Pharmacokinetics of Esketamine Nasal Spray and its Metabolite Noresketamine in Healthy Subjects and Patients with Treatment-Resistant Depression. Clin. Pharmacokinet. 2021, 60, 501–516. [Google Scholar] [CrossRef]
  49. Paoletti, P. NMDA receptor subunits: Function and pharmacology. Curr. Opin. Pharmacol. 2013, 13, 34–41. [Google Scholar]
  50. Hansen, K.B. Structural determinants of NMDA receptor function. Nat. Neurosci. 2016, 19, 1281–1291. [Google Scholar]
  51. Johnson, J.W. Mechanisms of NMDA receptor inhibition by ketamine and its metabolites. J. Neurosci. 2009, 29, 8563–8571. [Google Scholar]
  52. Abott, J.A.; Wen, H.; Liu, B.; Gupta, S.S.; Iacobucci, G.J.; Zheng, W.; Popescu, G.K. Allosteric inhibition of NMDA receptors by low dose ketamine. Mol. Psychiatry 2025, 30, 1009–1018. [Google Scholar] [CrossRef]
  53. Hayashi, T. Sigma-1 receptor chaperones regulate the ER-mitochondrion interface and mitochondrial function. Science 2007, 316, 585–589. [Google Scholar]
  54. Maurice, T.; Su, T.P. The pharmacology of sigma-1 receptors. Pharmacol. Rev. 2009, 61, 518–537. [Google Scholar] [CrossRef]
  55. Lepack, A.E.; Fuchikami, M.; Dwyer, J.M.; Banasr, M.; Duman, R.S. BDNF Release Is Required for the Behavioral Actions of Ketamine. Int. J. Neuropsychopharmacol. 2015, 18, pyu033. [Google Scholar] [CrossRef]
  56. Yamakura, T.; Chavez-Noriega, L.E.; Harris, R.A. Subunit-dependent inhibition of human neuronal nicotinic acetylcholine receptors and other ligand-gated ion channels by dissociative anesthetics ketamine and dizocilpine. Anesthesiology 2000, 92, 1144–1153. [Google Scholar] [CrossRef] [PubMed]
  57. Laurence, E. The histamine-releasing effect of ketamine on mast cells: Mechanisms and clinical implications. Anesthesiology 2013, 119, 734–744. [Google Scholar]
  58. Loix, S. The anti-inflammatory and immunomodulatory effects of ketamine: A review. Acta Anaesthesiol. Belg. 2011, 62, 47–58. [Google Scholar]
  59. Chang, Y. Ketamine reduces lipopolysaccharide-induced neuroinflammation in the rat brain. Anesth. Analg. 2009, 109, 1544–1551. [Google Scholar]
  60. Moaddel, R. Ketamine and its metabolites: Effects on α7-nicotinic receptor inhibition. J. Med. Chem. 2013, 56, 8232–8236. [Google Scholar]
  61. Hirota, K. Interaction of ketamine with opioid receptors in the central nervous system: Mechanisms and implications. Anesth. Analg. 1999, 89, 1–5. [Google Scholar]
  62. Duman, R.S. Ketamine and rapid-acting antidepressants: A new era in the battle against depression. Nat. Rev. Drug Discov. 2016, 15, 285–295. [Google Scholar] [CrossRef]
  63. Yang, C. Comparison of the antidepressant effects of R-ketamine and S-ketamine in animal models of depression. Pharmacol. Biochem. Behav. 2015, 138, 22–26. [Google Scholar]
  64. 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 34 ketamine metabolites. Nature 2016, 533, 481–486. [Google Scholar] [CrossRef] [PubMed]
  65. Suleiman, A.; Santer, P.; Munoz-Acuna, R.; Hammer, M.; Schaefer, M.S.; Wachtendorf, L.J.; Rumyantsev, S.; Berra, L.; Chamadia, S.; Johnson-Akeju, O.; et al. Effects of Ketamine Infusion on Breathing and Encephalography in Spontaneously Breathing ICU Patients. J. Intensive Care Med. 2023, 38, 299–306. [Google Scholar] [CrossRef]
  66. Gales, A.; Maxwell, S.K. Recent evidence and current uses. ATOTW 2018, 381, 1–7. [Google Scholar]
  67. Świątkiewicz, G. Social sciences in “Alkoholizm i Narkomania”–25 years of experience. AiN 2013, 26, 221–230. [Google Scholar]
  68. Geisslinger, G.; Hering, W.; Thomann, P.; Knoll, R.; Kamp, H.D.; Brune, K. Pharmacokinetics and pharmacodynamics of ketamine enantiomers in surgical patients using a stereoselective analytical method. Br. J. Anaesth. 1993, 70, 666–671. [Google Scholar] [CrossRef] [PubMed]
  69. Wang, J.; Huang, J.; Yang, S.; Cui, C.; Ye, L.; Wang, S.Y.; Yang, G.P.; Pei, Q. Pharmacokinetics and Safety of S-ketamine in Chinese Patients Undergoing Painless Gastroscopy in Comparison with Ketamine: A Randomized, Open-Label Clinical Study. Drug Dev. Ther. 2019, 13, 4135–4144. [Google Scholar] [CrossRef]
  70. Gao, M.; Rejaei, D.; Liu, H. Ketamine use in current clinical practice. Acta Pharmacol. Sin. 2016, 37, 865–872. [Google Scholar] [CrossRef] [PubMed]
  71. 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]
  72. Trimmel, H.; Helbok, R.; Staudinger, T.; Jaksch, W.; Messerer, B.; Schöchl, H.; Likar, R. S(+)-ketamine: Current trends in emergency and intensive care medicine. Wien. Klin. Wochenschr. 2018, 130, 356–366. [Google Scholar] [CrossRef]
  73. Kung, J.; Meisner, R.C.; Berg, S.; Ellis, D.B. Ketamine: A review of an established yet often underappreciated medication. Newsl. J. APSF 2020, 35, 33–68. [Google Scholar]
  74. Stoicea, N.; Versteeg, G.; Florescu, D.; Joseph, N.; Fiorda-Diaz, J.; Navarrete, V.; Bergese, S.D. Ketamine-Based Anesthetic Protocols and Evoked Potential Monitoring: A Risk/Benefit Overview. Front. Neurosci. 2016, 10, 37. [Google Scholar] [CrossRef] [PubMed]
  75. 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] [PubMed]
  76. Woolf, C.J. Central sensitization: Implications for the diagnosis and treatment of pain. Pain 2011, 152, 2–15. [Google Scholar] [CrossRef]
  77. 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]
  78. Feizerfan, A.; Sheh, G. Transition from acute to chronic pain. Contin. Educ. Anaesth. Crit. Care Pain 2015, 15, 98–102. [Google Scholar] [CrossRef]
  79. Tsui, P.Y.; Chu, M.C. Ketamine: An old drug revitalized in pain medicine. BJA Educ. 2017, 17, 84–87. [Google Scholar] [CrossRef]
  80. Niesters, M.; Martini, C.; Dahan, A. Ketamine for chronic pain: Risks and benefits. Br. J. Clin. Pharmacol. 2014, 77, 357–367. [Google Scholar] [CrossRef]
  81. Bell, R.F.; Dahl, J.B.; Moore, R.A.; Kalso, E. Perioperative ketamine for acute postoperative pain. Cochrane Database Syst. Rev. 2006, 5, CD004603. [Google Scholar]
  82. Choi, E.; Lee, H.; Park, H.S.; Lee, G.Y.; Kim, Y.J.; Baik, H.J. Effect of intraoperative infusion of ketamine on remifentanil-induced hyperalgesia. Korean J. Anesth. 2015, 68, 476–480. [Google Scholar] [CrossRef]
  83. Khan, Z.; Hameed, M.; Khan, F.A. Current role of perioperative intravenous ketamine: A narrative review. Anesthesiol. Perioper. Sci. 2023, 1, 36. [Google Scholar] [CrossRef]
  84. Martinez, M.R.; Garmon, E.H.; Starling, G.D.; Sheth, M.A. Ketamine as an Analgesic Adjunct for Opioid-Induced Hyperalgesia in a Patient With a Sickle Cell Pain Episode. Ochsner J. 2022, 22, 281–284. [Google Scholar] [CrossRef] [PubMed]
  85. Cairns, J.; Feng, T.L.; Ong, M.C. Continuous ketamine infusion for the management of opioid-induced hyperalgesia following amputation. BMJ Case Rep. 2024, 17, 255333. [Google Scholar] [CrossRef]
  86. Żylicz, Z.; Krajnik, M. Hiperalgezja opioidowa jako problem w leczeniu bólu. Mechanizmy powstawania, rozpoznanie i leczenie. Med. Paliatywna W Praktyce 2007, 1, 16–23. [Google Scholar]
  87. Koppert, W. Opioid-inducedhyperalgesia–Pathophysiology and clinicalrelevance. Acute Pain 2007, 9, 21–34. [Google Scholar] [CrossRef]
  88. McMullan, J.T.; Droege, C.A.; Chard, K.M.; Otten, E.J.; Hart, K.W.; Lindsell, C.J. Out-of-Hospital Intranasal Ketamine as an Adjunct to Fentanyl for the Treatment of Acute Traumatic Pain: A Randomized Clinical Trial. Ann. Emerg. Med. 2024, 84, 363–373. [Google Scholar] [CrossRef] [PubMed]
  89. McMullan, J.; Droege, C.; Strilka, R.; Hart, K.; Lindsell, C. Intranasal Ketamine as an Adjunct to Fentanyl for the Prehospital Treatment of Acute Traumatic Pain: Design and Rationale of a Randomized Controlled Trial. Prehosp. Emerg. Care 2021, 25, 519–529. [Google Scholar] [CrossRef]
  90. Yousefifard, M. The Efficacy of Ketamine Administration in Prehospital Pain Management of Trauma Patients; a Systematic Review and Meta-Analysis. Arch. Acad. Emerg. Med. 2020, 8, 1. [Google Scholar] [CrossRef]
  91. Mozafari, J.; Verki, M.M.; Motamed, H.; Sabouhi, A.; Tirandaz, F. Intranasal ketamine versus intravenous morphine for pain management in patients with renal colic: A double-blind, randomized, controlled trial. Am. J. Emerg. Med. 2020, 38, 2132–2136. [Google Scholar] [CrossRef]
  92. Farnia, M.R.; Jalali, A.; Vahidi, E.; Momeni, M.; Seyedhosseini, J.; Saeedi, M. Comparison of intranasal ketamine versus IV morphine in reducing pain in patients with renal colic. Am. J. Emerg. Med. 2017, 35, 434–437. [Google Scholar] [CrossRef]
  93. Jalili, A.; Farnia, M.R.; Momeni, M.; Saeedi, M. Comparing intranasal ketamine with intravenous fentanyl in reducing pain in patients with renal colic: A double-blind randomized clinical trial. Am. J. Emerg. Med. 2020, 38, 549–553. [Google Scholar] [CrossRef]
  94. Schwartzman, S.A.; Alexander, M.M.; Kravitz, S.A.; Shah, S.A.; Kravitz, M.J. A dose-escalation clinical trial of intranasal ketamine for uncontrolled cancer-related pain. Pharmacotherapy 2022, 42, 321–329. [Google Scholar] [CrossRef]
  95. Shrestha, S.; Shrestha, R.; Shrestha, S.; Shrestha, B. Intranasal ketamine for the treatment of patients with acute pain in the emergency department. J. Nepal. Health Res. Counc. 2016, 14, 197–201. [Google Scholar] [CrossRef]
  96. 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]
  97. Reynolds, S.L. 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]
  98. 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] [PubMed]
  99. Park, K.; Kim, J.J.; Oh, S.B.; Oh, S.Y.; Hong, Y.J.; Kim, S.J.; Park, E.J.; Choi, N.; Shin, S.H.; Kim, S.; et al. A Phase II Study About Efficacy and Safety of the Continuous IntraVenous Infusion of Ketamine as Adjuvant to Opioids in Terminally Ill Cancer Patients With Refractory Cancer Pain (CIVIK Trial). Am. J. Hosp. Palliat. Care 2025, 42, 244–252. [Google Scholar] [CrossRef] [PubMed]
  100. Orhurhu, V.J.; Roberts, J.S.; Ly, N.; Cohen, S.P. Ketamine in Acute and Chronic Pain Management. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  101. Chitneni, A.; Patil, A.; Dalal, S.; Ghorayeb, J.H.; Pham, Y.N.; Grigoropoulos, G. Use of Ketamine Infusions for Treatment of Complex Regional Pain Syndrome: A Systematic Review. Cureus 2021, 13, e18910. [Google Scholar] [CrossRef] [PubMed]
  102. Pereira, J.E.G.; Pereira, L.F.G.; Linhares, R.M.; Bersot, C.D.A.; 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]
  103. Bruna-Mejias, A.; Baeza, V.; Gamboa, J.; Baez Flores, B.; San Martin, J.; Astorga, C.; Leyton, J.; Nova-Baeza, P.; Orellana-Donoso, M.; Suazo-Santibañez, A.; et al. Use of Ketamine in Patients with Multifactorial Neuropathic Pain: A Systematic Review and Meta-Analysis. Pharmaceuticals 2024, 17, 1165. [Google Scholar] [CrossRef]
  104. Mitchell, A.C.; Fallon, M.T. A single infusion of intravenous ketamine improves pain relief in patients with critical limb ischaemia: Results of a double blind randomised controlled trial. Pain 2002, 10, 12044624. [Google Scholar] [CrossRef] [PubMed]
  105. Li, Q. S-Ketamine reduces the risk of rebound pain in patients following total knee arthroplasty: A randomized controlled trial. Drug Des. Dev. Ther. 2025, 19, 2315–2327. [Google Scholar] [CrossRef]
  106. Gumelar, W. The effectiveness of intraoperative ketamine and fentanyl as preemptive analgesia assessed with qNOX Score. J. Anestesi Indonesia. 2021, 13, 67–77. [Google Scholar] [CrossRef]
  107. 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, 299–307. [Google Scholar] [CrossRef]
  108. Nielsen, R.V. Intraoperative Ketamine Reduces Immediate Postoperative Opioid Consumption After Spinal Fusion Surgery in Chronic Pain Patients with Opioid Dependency: A Randomized, Blinded Trial. Pain 2017, 158, 463–470. [Google Scholar] [CrossRef]
  109. Park, P.J.; Makhni, M.C.; Cerpa, M.; Lehman, R.A.; Lenke, L.G. Intravenous Ketamine for Postoperative Pain Control in Spine Surgery: A Randomized Controlled Trial. J. Spine Surg. 2020, 6, 546–556. [Google Scholar] [CrossRef] [PubMed]
  110. 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]
  111. Jain, S.; Nazir, N.; Mustafi, S.M. Effect of Low-dose Ketamine on Inflammatory Markers, Perioperative Pain, and Chronic Pain After Laparoscopic Inguinal Hernia Surgery. Turk. J. Anaesthesiol. Reanim. 2024, 52, 200–207. [Google Scholar] [CrossRef]
  112. Xu, Y.; He, L.; Liu, S.; Zhang, C.; Ai, Y. Intraoperative Intravenous Low-dose Esketamine Improves Quality of Early Recovery After Laparoscopic Radical Resection of Colorectal Cancer: A Prospective, Randomized Controlled Trial. PLoS ONE 2023, 18, 0286590. [Google Scholar] [CrossRef]
  113. Zhao, L.; Li, Z.; Jin, B.; Hou, N.; Yang, H. Safety and efficacy of low-dose esketamine in laparoscopic cholecystectomy: A prospective, double-blind randomized controlled trial. BMC Anesthesiol. 2024, 24, 47. [Google Scholar] [CrossRef]
  114. Sukantarat, N.; Uthaiwan, K. Comparison of postoperative pain scores between intraoperative intravenous ketamine and placebo in patients undergoing unilateral total knee arthroplasty under general anesthesia: A double-blind randomized controlled trial. Thai J. Anesth. 2024, 50, 1–8. [Google Scholar]
  115. Chen, H.; Zhi, J.; Wang, L.; Jin, Z.; Xu, J.; Xing, F.; Wen, C.; Wang, Q.; Chen, C.; Li, W.; et al. Subanesthetic Dose of Esketamine Improves the Sedative and Analgesic Effects of Dexmedetomidine and Remifentanil in Liposuction Anesthesia: A Prospective, Double-Blinded, Randomized Controlled Trial. Drug Des. Dev. Ther. 2024, 20, 3645–3658. [Google Scholar] [CrossRef]
  116. Sutoh, K.; Sanuki, N.; Sakaki, T.; Imai, R. Specific induction of TaAAPT1, an ER- and Golgi-localized ECPT-type aminoalcoholphosphotransferase, results in preferential accumulation of the phosphatidylethanolamine membrane phospholipid during cold acclimation in wheat. Plant Mol. Biol. 2010, 72, 519–531. [Google Scholar] [CrossRef]
  117. Mercadante, S.; Villari, P.; Ferrera, P. Burst ketamine to reverse opioid tolerance in cancer pain. J. Pain Symptom Manag. 2023, 25, 302–305. [Google Scholar] [CrossRef]
  118. Singh, V.; Gillespie, T.W.; Harvey, R.D. Intranasal Ketamine and Its Potential Role in Cancer-Related Pain. Pharmacotherapy 2018, 38, 390–401. [Google Scholar] [CrossRef]
  119. Hocking, G.; Cousins, M.J. Ketamine in chronic pain management: An evidence-based review. Anesth. Analg. 2003, 97, 1730–1739. [Google Scholar] [CrossRef] [PubMed]
  120. Fine, P.G. Low-dose ketamine in the management of opioid nonresponsive terminal cancer pain. J. Pain Symptom Manag. 1999, 17, 296–300. [Google Scholar] [CrossRef] [PubMed]
  121. Forth, N.; Nguyen, M.; Grech, A. A Case Report of Subanesthetic Ketamine Bolus and Infusion for Opioid Refractory Cancer Pain. J. Palliat. Med. 2022, 25, 1161–1165. [Google Scholar] [CrossRef]
  122. Hasin, D.S.; Goodwin, R.D.; Stinson, F.S.; Grant, B.F. Epidemiology of major depressive disorder: Results from the National Epidemiologic Survey on Alcoholism and Related Conditions. Arch. Gen. Psychiatry 2005, 62, 1097–1106. [Google Scholar] [CrossRef]
  123. Kessler, R.C.; Berglund, P.; Demler, O.; Jin, R.; Koretz, D.; Merikangas, K.R.; Rush, A.J.; Walters, E.E.; Wang, P.S. The epidemiology of major depressive disorder: Results from the National Comorbidity Survey Replication (NCS-R). JAMA 2003, 289, 3095–3105. [Google Scholar] [CrossRef]
  124. Grant, B.F.; Stinson, F.S.; Dawson, D.A.; Chou, S.P.; Dufour, M.C.; Compton, W.; Pickering, R.P.; Kaplan, K. Prevalence and co-occurrence of substance use disorders and independent mood and anxiety disorders: Results from the national epidemiologic survey on alcohol and related conditions. Arch. Gen. Psychiatry 2004, 61, 807–816. [Google Scholar] [CrossRef]
  125. Curran, H.V.; Freeman, T.P.; Mokrysz, C.; Lewis, D.A.; Morgan, C.J.; Parsons, L.H. Keep off the grass? Cannabis, cognition and addiction. Nat. Rev. Neurosci. 2016, 17, 293–306. [Google Scholar] [CrossRef] [PubMed]
  126. Markou, A.; Kosten, T.R.; Koob, G.F. Neurobiological similarities in depression and drug dependence: A self-medication hypothesis. Neuropsychopharmacology 1998, 18, 135–174. [Google Scholar] [CrossRef]
  127. Quello, S.B.; Brady, K.T.; Sonne, S.C. Mood disorders and substance use disorder: A complex comorbidity. Sci. Pract. Perspect. 2005, 3, 13–21. [Google Scholar] [CrossRef]
  128. Greenfield, S.F.; Weiss, R.D.; Muenz, L.R. The effect of depression on return to drinking: A prospective study. Arch. Gen. Psychiatry 1998, 55, 259–265. [Google Scholar] [CrossRef] [PubMed]
  129. White, A.M.; Jordan, J.D.; Schroeder, K.M.; Acheson, S.K.; Georgi, B.D.; Sauls, G.; Ellington, R.R.; Swartzwelder, H.S. Predictors of Relapse During Treatment and Treatment Completion Among Marijuana-Dependent Adolescents in an Intensive Outpatient Substance Abuse Program Aaron. Subst. Abus. 2004, 25, 53–59. [Google Scholar] [CrossRef]
  130. Willinger, U.; Lenzinger, E.; Hornik, K.; Fischer, G.; Schönbeck, G.; Aschauer, H.N.; Meszaros, K. Anxiety as a predictor of relapse in detoxified alcohol dependent patients. Alcohol Alcohol. 2002, 37, 609–612. [Google Scholar] [CrossRef] [PubMed]
  131. Mason, B.J.; Kocsis, J.H.; Ritvo, E.C.; Cutler, R.B. A double blind, placebo controlled trial of desipramine for primary alcohol dependence stratified on the presence or absence of major depression. JAMA 1996, 275, 761–767. [Google Scholar] [CrossRef]
  132. Pettinati, H.M. Antidepressant treatment of co-occurring depression and 31 alcohol dependence. Biol. Psychiatry 2004, 56, 785–792. [Google Scholar] [CrossRef]
  133. Kalivas, P.W.; Kalivas, P.W.; Volkow, N.D. The glutamate homeostasis hypothesis of addiction. Nat. Rev. Neurosci. 2009, 10, 561–572. [Google Scholar] [CrossRef]
  134. Kalivas, P.W.; Volkow, N.D. The neural basis of addiction: A pathology of motivation and choice. Am. J. Psychiatry 2005, 162, 1403–1413. [Google Scholar] [CrossRef]
  135. Sabino, V.; Narayan, A.R.; Zeric, T.; Steardo, L.; Cottone, P. mTOR activation is required for the anti-alcohol effect of ketamine, but not memantine, in alcohol-preferring rats. Behav. Brain Res. 2013, 247, 9–16. [Google Scholar] [CrossRef] [PubMed]
  136. Li, N. mTOR-dependent synapse formation underlies the rapid antidepressant effects of NMDA antagonists. Science 2010, 329, 959–964. [Google Scholar] [CrossRef] [PubMed]
  137. 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] [PubMed]
  138. Mandyam, C.D.; Koob, G.F. The addicted brain craves new neurons: Putative role for adult-born progenitors in promoting recovery. Trends Neurosci. 2012, 35, 250–260. [Google Scholar] [CrossRef]
  139. Noonan, M.A.; Bulin, S.E.; Fuller, D.C.; Eisch, A.J. Reduction of adult hippocampal neurogenesis confers vulnerability in an animal model of cocaine addiction. J. Neurosci. 2010, 30, 304–315. [Google Scholar] [CrossRef]
  140. Zanardini, R.; Fontana, A.; Pagano, R.; Mazzaro, E.; Bergamasco, F.; Romagnosi, G.; Gennarelli, M.; Bocchio-Chiavetto, L. Alterations of brain-derived neurotrophic factor serum levels in patients with alcohol dependence. Alcohol. Clin. Exp. Res. 2011, 35, 1529–1533. [Google Scholar] [CrossRef]
  141. Angelucci, F.; Ricci, V.; Pomponi, M.; Conte, G.; Mathé, A.A.; Attilio Tonali, P.; Bria, P. Chronic heroin and cocaine abuse is associated with decreased serum concentrations of the nerve growth factor and brain-derived neurotrophic factor. J. Psychopharmacol. 2007, 21, 820–825. [Google Scholar] [CrossRef]
  142. Corominas-Roso, M.; Roncero, C.; Eiroa-Orosa, F.J.; Gonzalvo, B.; Grau-Lopez, L.; Ribases, M.; Rodriguez-Cintas, L.; Sánchez-Mora, C.; Ramos-Quiroga, J.A.; Casas, M. Brain-derived neurotrophic factor serum levels in cocaine-dependent patients during early abstinence. Eur. Neuropsychopharmacol. 2013, 23, 1078–1084. [Google Scholar] [CrossRef]
  143. Sönmez, M.B.; Görgülü, Y.; Çınar, R.K.; Kılıç, E.K.; Ünal, A.; Vardar, M. Alterations of BDNF and GDNF serum levels in alcohol-addicted patients during alcohol withdrawal. Eur. J. Psychiatry 2016, 30, 109–118. [Google Scholar]
  144. Duncan, W.C., Jr.; Zarate, C.A., Jr. Ketamine, sleep, and depression: Current status and new questions. Curr. Psychiatry Rep. 2013, 15, 394. [Google Scholar] [CrossRef]
  145. Autry, A.E.; Adachi, M.; Nosyreva, E.; Na, E.S.; Los, M.F.; Cheng, P.F.; Kavalali, E.T.; Monteggia, L.M. NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses. Nature 2011, 475, 91–95. [Google Scholar] [CrossRef] [PubMed]
  146. Ota, K.T.; Duman, R.S. Environmental and pharmacological modulations of cellular plasticity: Role in the pathophysiology and treatment of depression. Neurobiol. Dis. 2013, 57, 28–37. [Google Scholar] [CrossRef]
  147. Corlett, P.R.; Cambridge, V.; Gardner, J.M.; Piggot, J.S.; Turner, D.C.; Everitt, J.C.; Arana, F.S.; Morgan, H.L.; Milton, A.L.; Lee, J.L.; et al. Ketamine effects on memory reconsolidation favor a learning model of delusions. PLoS ONE 2013, 8, e65088. [Google Scholar] [CrossRef] [PubMed]
  148. Kolp, E.; Friedman, H.L.; Young, M.S. Ketamine enhanced psychotherapy: Preliminary clinical observations on its effectiveness in treating alcoholism. Humanist. Psychol. 2006, 34, 399–422. [Google Scholar] [CrossRef]
  149. Krupitsky, E.M.; Grinenko, A.Y. Ketamine psychedelic therapy (KPT): A review of the results of ten years of research. J. Psychoact. Drugs 1997, 29, 165–183. [Google Scholar] [CrossRef] [PubMed]
  150. Dakwar, E.; Anerella, C.; Hart, C.L. Therapeutic infusions of ketamine: Do the psychoactive effects matter? Drug Alcohol. Depend. 2014, 136, 153–157. [Google Scholar] [CrossRef]
  151. Kolp, E.; Friedman, H.L.; Krupitsky, E.; Jansen, K.; Sylvester, M.; Young, M.S.; Kolp, A. Ketamine psychedelic psychotherapy: Focus on its pharmacology, phenomenology, and clinical applications. Int. J. Transpers. Stud. 2014, 33, 84–140. [Google Scholar] [CrossRef]
  152. Kolp, E.; Krupitsky, E.; Young, M.S.; Jansen, K.; Friedman, H.; O’Connor, L.A. Ketamine-enhanced psychotherapy: Preliminary clinical observations on its effects in treating death anxiety. Int. J. Transpers. Stud. 2007, 26, 1–17. [Google Scholar] [CrossRef]
  153. Krupitsky, E.; Burakov, A.; Romanova, T. Ketamine psychotherapy for heroin addiction: Immediate effects and two-year follow-up. J. Subst. Abuse Treat. 2002, 23, 273–283. [Google Scholar] [CrossRef]
  154. Giese, K.P. Memory Mechanisms in Health and Disease: Mechanistic Basis of Memory; World Scientific: Singapore, 2012. [Google Scholar]
  155. Jansen, K.L.R. A Review of the Nonmedical Use of Ketamine: Use, Users and Consequences. J. Psychoact. Drugs 2000, 32, 419–433. [Google Scholar] [CrossRef]
  156. 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]
  157. Ricuarte, G.; McCann, U. Recognition and management of complications of new recreational drug use. Lancet 2005, 365, 2137–2145. [Google Scholar] [CrossRef]
  158. Dakwar, E.; Levin, F.; Hart, C.L.; Basaraba, C.; Choi, J.; Pavlicova, M. A single ketamine infusion combined with motivational enhancement therapy for alcohol use disorder: A randomized midazolam-controlled pilot trial. Am. J. Psychiatry 2020, 177, 125–133. [Google Scholar] [CrossRef]
  159. Das, R.K.; Gale, G.; Walsh, K. Ketamine can reduce harmful drinking by pharmacologically rewriting drinking memories. Nat. Commun. 2019, 10, 5187. [Google Scholar] [CrossRef] [PubMed]
  160. Grabski, M.; McAndrew, A.; Lawn, W. Adjunctive ketamine with relapse prevention-based psychological therapy in the treatment of alcohol use disorder. Am. J. Psychiatry 2022, 179, 152–162. [Google Scholar] [CrossRef] [PubMed]
  161. Krupitsky, E.M.; Grinenko, A.Y.; Berkaliev, T.N.; Paley, A.I.; Petrov, V.N.; Moshkov, K.A.; Borodkin, Y.S. The combination of psychedelic and aversive approaches in alcoholism treatment: The affective contra-attribution method. Alcohol. Treat. Q. 1992, 9, 99–105. [Google Scholar] [CrossRef]
  162. Pizon, A.F.; Lynch, M.J.; Benedict, N.J.; Yanta, J.H.; Frisch, A.; Menke, N.B.; Swartzentruber, G.S.; King, A.M.; Abesamis, M.G.; Kane-Gill, S.L. Adjunct ketamine use in the management of severe ethanol withdrawal. Crit. Care Med. 2018, 46, e768–e771. [Google Scholar] [CrossRef]
  163. Rothberg, R.L.; Azhari, N.; Haug, N.A.; Dakwar, E. Mystical-type experiences occasioned by ketamine mediate its impact on at-risk drinking: Results from a randomized, controlled trial. J. Psychopharmacol. 2021, 35, 150–158. [Google Scholar] [CrossRef]
  164. Shah, P.; McDowell, M.; Ebisu, R.; Hanif, T.; Toerne, T. Adjunctive use of ketamine for benzodiazepine-resistant severe alcohol withdrawal: A retrospective evaluation. J. Med. Toxicol. 2018, 14, 229–236. [Google Scholar] [CrossRef]
  165. Yoon, G.; Petrakis, I.L.; Krystal, J.H. Association of combined naltrexone and ketamine with depressive symptoms in a case series of patients with depression and alcohol use disorder. JAMA Psychiatry 2019, 76, 337–338. [Google Scholar] [CrossRef] [PubMed]
  166. Wong, A.; Benedict, N.J.; Armahizer, M.J.; Kane-Gill, S.L. Evaluation of adjunctive ketamine to benzodiazepines for management of alcohol withdrawal syndrome. Ann. Pharmacother. 2015, 49, 14–19. [Google Scholar] [CrossRef]
  167. Dakwar, E.; Hart, C.L.; Levin, F.R. Cocaine self-administration disrupted by the N-methyl-D-aspartate receptor antagonist ketamine: A randomized, crossover trial. Mol. Psychiatry 2017, 22, 76–81. [Google Scholar] [CrossRef] [PubMed]
  168. Dakwar, E.; Nunes, E.V.; Hart, C.L.; Foltin, R.W.; Mathew, S.J.; Carpenter, K.M.; Choi, C.J.J.; Basaraba, C.N.; Pavlicova, M.; Levin, F.R. A Single Ketamine Infusion Combined With Mindfulness-Based Behavioral Modification to Treat Cocaine Dependence: A Randomized Clinical Trial. Am. J. Psychiatry 2019, 176, 923–930. [Google Scholar] [CrossRef] [PubMed]
  169. Jovaisa, T.; Laurinenas, G.; Vosylius, S.; Sipylaite, J.; Badaras, R.; Ivaskevicius, J. Effects of ketamine on precipitated opiate withdrawal. Medicina 2006, 42, 625–634. [Google Scholar]
  170. Lalanne, L.; Nicot, C.; Lang, J.P.; Bertschy, G.; Salvat, E. Experience of the use of Ketamine to manage opioid withdrawal in an addicted woman: A case report. BMC Psychiatry 2016, 16, 395. [Google Scholar] [CrossRef]
  171. Omoigui, S.; Hashmat, F.; Bernardo, Z. Use of Ketamine in Ameliorating Opioid Withdrawal Symptoms During an Induction Phase of Buprenorphine. Open Pain. J. 2011, 4, 1–3. [Google Scholar] [CrossRef]
  172. Pradhan, B.; Rossi, G. Combining Ketamine, Brain Stimulation (rTMS) and Mindfulness Therapy (TIMBER) for Opioid Addiction. Cureus 2020, 12, e11798. [Google Scholar] [CrossRef]
  173. Lapidus, K.A.; Levitch, C.F.; Perez, A.M.; Brallier, J.W.; Parides, M.K.; Soleimani, L.; Feder, A.; Iosifescu, D.V.; Charney, D.S.; Murrough, J.W. A randomized controlled trial of intranasal ketamine in major depressive disorder. Biol. Psychiatry 2014, 76, 970–976. [Google Scholar] [CrossRef]
  174. Stachowicz, K.; Sowa-Kućma, M. The treatment of depression—Searching for new ideas. Front. Pharmacol. 2022, 13, 988648. [Google Scholar] [CrossRef]
  175. Hashimoto, K. Molecular mechanisms of the rapid-acting and long-lasting antidepressant actions of (R)-ketamine. Biochem. Pharmacol. 2020, 177, 113935. [Google Scholar] [CrossRef] [PubMed]
  176. Amasi-Hartoonian, N.; Pariante, C.M.; Cattaneo, A.; Sforzini, L. Understanding treatment-resistant depression using «omics» techniques: A systematic review. J. Affect. Disord. 2022, 318, 423–455. [Google Scholar] [CrossRef]
  177. Bhatt, S.; Devadoss, T.; Jha, N.K.; Baidya, M.; Gupta, G.; Chellappan, D.K.; Singh, S.K.; Dua, K. Targeting inflammation: A potential approach for the treatment of depression. Metab. Brain Dis. 2023, 38, 45–59. [Google Scholar] [CrossRef] [PubMed]
  178. Murdaca, G.; Paladin, F.; Casciaro, M.; Vicario, C.M.; Gangemi, S.; Martino, G. Neuro-Inflammaging and psychopathological distress. Biomedicines 2022, 10, 2133. [Google Scholar] [CrossRef] [PubMed]
  179. Baker, P.M.; Jhou, T.; Li, B.; Matsumoto, M.; Mizumori, S.J.; Stephenson-Jones, M.; Vicentic, A. The lateral habenula circuitry: Reward processing and cognitive control. J. Neurosci. 2016, 36, 11482–11488. [Google Scholar] [CrossRef]
  180. Strasburger, S.E.; Bhimani, P.M.; Kaabe, J.H.; Krysiak, J.T.; Nanchanatt, D.L.; Nguyen, T.N.; Pough, K.A.; Prince, T.A.; Ramsey, N.S.; Savsani, K.H.; et al. What is the mechanism of ketamine’s rapid-onset antidepressant effect? A concise overview of the surprisingly large number of possibilities. J. Clin. Pharm. Ther. 2017, 42, 147–154. [Google Scholar] [CrossRef]
  181. Zhang, J.C.; Yao, W.; Hashimoto, K. Arketamine, a new rapid-acting antidepressant: A historical review and future directions. Neuropharmacology 2022, 218, 109219. [Google Scholar] [CrossRef] [PubMed]
  182. Brendle, M.; Robison, R.; Malone, D.C. Cost-effectiveness of esketamine nasal spray compared to intravenous ketamine for patients with treatment-resistant depression in the US utilizing clinical trial efficacy and real-world effectiveness estimates. J. Affect. Disord. 2022, 319, 388–396. [Google Scholar] [CrossRef]
  183. European Medicines Agency (EMA). Spravato: Summary of Product Characteristics. Available online: https://www.ema.europa.eu/en/documents/product-information/spravato-epar-product-information_en.pdf (accessed on 18 May 2025).
  184. Available online: https://euromedis.pl/najnowszy-lek-przeciwdepresyjny-spravato-leczenie-dostepne-dla-pacjentow-w-centrum-medycznym-euromedis/ (accessed on 1 May 2025).
  185. Koike, H.; Iijima, M.; Chaki, S. Involvement of AMPA receptor in both the rapid and sustained antidepressant-like effects of ketamine in animal models of depression. Behav. Brain Res. 2011, 224, 107–111. [Google Scholar] [CrossRef]
  186. Jiang, Y.; Wang, Y.; Sun, X.; Lian, B.; Sun, H.; Wang, G.; Du, Z.; Li, Q.; Sun, L. Short- and long-term antidepressant effects of ketamine in a rat chronic unpredictable stress model. Brain Behav. 2017, 7, 00749. [Google Scholar] [CrossRef]
  187. 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]
  188. Fitzgerald, P.J.; Yen, J.Y.; Watson, B.O. Stress-sensitive antidepressant-like effects of ketamine in the mouse forced swim test. PLoS ONE 2019, 14, 0215554. [Google Scholar] [CrossRef]
  189. Levinta, A.; Meshkat, S.; McIntyre, R.S.; Ho, C.; Lui, L.M.W.; Lee, Y.; Mansur, R.B.; Teopiz, K.M.; Rodrigues, N.B.; Di Vincenzo, J.D.; et al. The association between stage of treatment-resistant depression and clinical utility of ketamine/esketamine: A systematic review. J. Affect. Disord. 2022, 318, 139–149. [Google Scholar] [CrossRef]
  190. Daly, E.J.; Trivedi, M.H.; Janik, A.; Li, H.; Zhang, Y.; Li, X.; Lane, R.; Lim, P.; Duca, A.R.; Hough, D.; et al. Efficacy of esketamine nasal spray plus oral antidepressant treatment for relapse prevention in patients with treatment-resistant depression: A randomized clinical trial. JAMA Psychiatry 2019, 76, 893–903. [Google Scholar] [CrossRef] [PubMed]
  191. Alario, A.A.; Niciu, M.J. (Es)Ketamine for suicidal ideation and behavior: Clinical efficacy. Chronic Stress 2022, 6, 24705470221128017. [Google Scholar] [CrossRef]
  192. Hassan, K.; Struthers, W.M.; Sankarabhotla, A.; Daid, P. Safety, effectiveness and tolerability of sublingual ketamine in depression and anxiety: A retrospective study of off-label, at-home use. Front. Psychiatry 2022, 13, 992624. [Google Scholar] [CrossRef]
  193. Khorassani, F.; Talreja, O. Intranasal esketamine: A novel drug for treatment-resistant depression. Am. J. Health Syst. Pharm. 2020, 77, 1382–1388. [Google Scholar] [CrossRef] [PubMed]
  194. Popova, V.; Daly, E.J.; Trivedi, M.; Cooper, K.; Lane, R.; Lim, P.; Mazzucco, C.; Hough, D.; Thase, M.E.; Shelton, R.C.; et al. Efficacy and safety of flexibly dosed esketamine nasal spray combined with a newly initiated oral antidepressant in treatment-resistant depression: A randomized double-blind active-controlled study. Am. J. Psychiatry 2019, 176, 428–438. [Google Scholar] [CrossRef]
  195. Fedgchin, M.; Trivedi, M.; Daly, E.J.; Melkote, R.; Lane, R.; Lim, P.; Vitagliano, D.; Blier, P.; Fava, M.; Liebowitz, M.; et al. Efficacy and safety of fixed-dose esketamine nasal spray combined with a new oral antidepressant in treatment-resistant depression: Results of a randomized, double-blind, active-controlled study TRANSFORM-1. Int. J. Neuropsychopharmacol. 2019, 22, 616–630. [Google Scholar] [CrossRef]
  196. Jha, M.K.; Williamson, D.J.; Magharehabed, G.; Turkoz, I.; Daly, E.J.; Trivedi, M.H. Intranasal esketamine effectively treats treatment-resistant depression in adults regardless of baseline irritability. J. Affect. Disord. 2022, 321, 153–160. [Google Scholar] [CrossRef]
  197. Ochs-Ross, R.; Daly, E.J.; Zhang, Y.; Lane, R.; Lim, P.; Morrison, R.L.; Hough, D.; Manji, H.; Drevets, W.C.; Sanacora, G.; et al. Efficacy and safety of esketamine nasal spray plus an oral antidepressant in elderly patients with treatment-resistant depression—TRANSFORM-3. Am. J. Geriatr. Psychiatry 2020, 28, 121–141. [Google Scholar] [CrossRef]
  198. Jeon, H.J.; Ju, P.C.; Sulaiman, A.H.; Aziz, S.A.; Paik, J.W.; Tan, W.; Bai, D.; Li, C.T. Long-term safety and efficacy of esketamine nasal spray plus an oral antidepressant in patients with treatment-resistant depression—An Asian sub-group analysis from the SUSTAIN-2 Study. Clin. Psychopharmacol. Neurosci. 2022, 20, 70–86. [Google Scholar] [CrossRef] [PubMed]
  199. Martinotti, G.; Vita, A.; Fagiolini, A.; Maina, G.; Bertolino, A.; Dell’Osso, B.; Siracusano, A.; Clerici, M.; Bellomo, A.; Sani, G.; et al. Real-world experience of esketamine use to manage treatment-resistant depression: A multicentric study on safety and effectiveness REAL-ESK study. J. Affect. Disord. 2022, 319, 646–654. [Google Scholar] [CrossRef]
  200. Turkoz, I.; Lopena, O.; Salvadore, G.; Sanacora, G.; Shelton, R.; Fu, D.J. Treatment response to esketamine nasal spray in patients with major depressive disorder and acute suicidal ideation or behavior without evidence of early response: A pooled post hoc analysis of ASPIRE. CNS Spectr. 2022, 28, 482–488. [Google Scholar] [CrossRef]
  201. Wajs, E.; Aluisio, L.; Holder, R.; Daly, E.J.; Lane, R.; Lim, P.; George, J.E.; Morrison, R.L.; Sanacora, G.; Young, A.H.; et al. Esketamine nasal spray plus oral antidepressant in patients with treatment-resistant depression: Assessment of long-term safety in a phase 3, open-label study (SUSTAIN-2). J. Clin. Psychiatry 2020, 81, 19m12891. [Google Scholar] [CrossRef] [PubMed]
  202. Starr, H.L.; Abell, J.; Larish, A.; Lewis, S.; DeMuro, C.; Gogate, J.; Jamieson, C.; Daly, E.; Zaki, N.; Kramer, M. Self-reported review of the value of esketamine in patients with treatment-resistant depression: Understanding the patient experience in the STRIVE Study. Psychiatry Res. 2020, 293, 113376. [Google Scholar] [CrossRef] [PubMed]
  203. Wang, Y.; Zhang, Q.; Dai, X.; Xiao, G.; Luo, H. Effect of low-dose esketamine on pain control and postpartum depression after cesarean section: A retrospective cohort study. Ann. Palliat. Med. 2022, 11, 45–57. [Google Scholar] [CrossRef]
  204. Leal, G.C.; Bandeira, I.D.; Correia-Melo, F.S.; Telles, M.; Mello, R.P.; Vieira, F.; Lima, C.S.; Jesus-Nunes, A.P.; Guerreiro-Costa, L.N.F.; Marback, R.F.; et al. Intravenous arketamine for treatment-resistant depression: Open-label pilot study. Eur. Arch. Psychiatry Clin. Neurosci. 2021, 271, 577–582. [Google Scholar] [CrossRef]
  205. Masaki, Y.; Kashiwagi, Y.; Watabe, H.; Abe, K. (R)- and (S)-ketamine induce differential fMRI responses in conscious rats. Synapse 2019, 73, 22126. [Google Scholar] [CrossRef]
Figure 1. Metabolism of ketamine enantiomers.
Figure 1. Metabolism of ketamine enantiomers.
Molecules 30 02824 g001
Figure 2. Mechanism of action of ketamine, adaptation based on [19].
Figure 2. Mechanism of action of ketamine, adaptation based on [19].
Molecules 30 02824 g002
Figure 3. Method of Spravato® administration [184].
Figure 3. Method of Spravato® administration [184].
Molecules 30 02824 g003
Table 1. Basic pharmacokinetic parameters of ketamine enantiomers (S-, R- and racemic) [34,35,36,37,38].
Table 1. Basic pharmacokinetic parameters of ketamine enantiomers (S-, R- and racemic) [34,35,36,37,38].
Ketamine EnantiomerRoute of
Administration
Dose (mg/kg)Bioavailability (%)Literature
(S-, R-)-
ketamine
i.v.1–4.5 100 [34]
i.m.8–10 93[32,33,35]
p.o.0.25–0.5 17–29[36,37,38,39]
intranasal0.3–9.045–50[8,40,41,42,43,44]
S-ketaminei.v.0.125–0.3100[40,45,46,47,48]
i.m.0.27100
p.o.0.25–0.38–24
intranasal0.3370
R-ketaminei.v. lack of data[49,50,51]
i.m.0.593
p.o lack of data
intranasal lack of data
Table 2. Use, contraindications and notes for ketamine [32,66,73,74,75].
Table 2. Use, contraindications and notes for ketamine [32,66,73,74,75].
IndicationsContraindicationsAdditional Notes
Anesthesia:
Sedation (mechanical ventilation)
Procedural sedation
Induction and maintenance of
general anesthesia
Anesthesia for patients with respiratory conditions
Anesthesia where hemodynamic
balance is required
Subanesthesia
Psychosis
Poorly controlled hypertension
Severe liver dysfunction
Epilepsy
Cranial injuries (not fully studied)
Schizophrenia
Required during administration and procedures: blood pressure, electrocardiogram and respiratory measurements
Analgesia:
Acute pain
Chronic pain
Neuropathic pain
Perioperative pain
Cancer pain
Severe liver dysfunction
Active substance abuse
Ulcerative cystitis
Significant coronary disease
Psychosis
Simultaneous administration
of benzodiazepines or α2-adrenoceptor agonists may be required
Precaution against chronic postoperative pain—not
effective
Table 4. Basic characteristics about the included studies.
Table 4. Basic characteristics about the included studies.
AuthorSchedule of Ketamine
Administration
Principal Conclusions
Ethanol addiction
Dakwar, E. et al., 2020 [158]i.v. administration 0.71 mg/kg
+ motivational enhancement
therapy/psychotherapy
increased likelihood of abstinence, delayed time to relapse and reduced likelihood of heavy drinking days compared to midazolam
Das et al., 2019 [159]i.v. 0.5 mg/kg for 10 daysreduction in the reinforcing effects of ethanol, reduction in number
of drinking days per week and volume of consumed alcohol
Grabski et al., 2022 [160]i.v. 0.8 mg/kg + psychotherapyincrease the number
of abstinent days
Kolp et al., 2006 [148]i.m. administration
2–3 mg/kg + psychotherapy
extending the period
of alcohol abstinence
Krupitsky et al., 1992 [161]i.m. 3 mg/kg + psychotherapyextending the period
of alcohol abstinence
Krupitsky and Grinenko, 1997 [149]i.m. 2.5 mg/kg + psychotherapyextending the period of alcohol abstinence and reduced
risk of relapse
Pizon et al., 2018 [162]i.v. 0.15–0.3 mg/kg/h
+ bolus (0.3 mg/kg) + conventional withdrawal treatment
reduction in benzodiazepine
requirements, decrease likelihood
of intubation and a shorter length of stay in the intensive care unit (ICU)
Rothberg et al., 2021 [163]i.v. 0.71 mg/kg + motivational
enhancement therapy
increased probability of abstinence, delayed time to relapse, decreased likelihood of heavy drinking days compared to midazolam
Shah et al., 2018 [164]i.v. 0.75 mg/kg + conventional withdrawal treatmentenhanced symptom control for benzodiazepine-refractory patients and reduced infusion requirements
Yoon et al., 2019 [165]i.v. 0.5 mg/kg once a week for 4 weeks + naltrexone 380 mgreduced alcohol craving
and consumption
Wong et al., 2015 [166]i.v. median infusion 0.20 mg/kg/h + conventional withdrawal
treatment with a standardized treatment protocol (benzodiazepine + dexmedetomidine + phenobarbital + propofol ± antipsychotics + clonidine + intubation)
reduction in short-term
benzodiazepine dose requirements in patients with alcohol withdrawal
Cocaine addiction
Dakwar et al., 2014 [150]3 × i.v. 0.41 mg/kg or 0.71 mg/kgenhanced motivation to quit and dampened cue-induced craving
Dakwar et al., 2017 [167]i.v. 0.11 mg/kg 2-min bolus
+ 0.60 mg/kg 50 min
decreased cocaine self-administration
Dakwar et al., 2018 [168]i.v. 0.71 mg/kgdecreased cocaine self-administration, cocaine
use and craving
Dakwar et al., 2019 [158]i.v. 0.5 mg/kg promoted abstinence, diminished craving and reduced risk of relapse
Opioid addiction
Jovaiša et al., 2006 [169]i.v. 0.5 mg/kgbetter control of withdrawal
symptoms with no effects
on treatment of opiate dependence after 4 months
Krupitsky et al., 2002 [153]i.m. 0.2 or 2.0 mg/kg
+ psychotherapy
increased rate of abstinence within the first two years of follow-up, reduction in craving for heroin, positive change in nonverbal
unconscious emotional attitudes
Lalanne et al., 2016 [170]oral administration, 1 mg/kg reduction in dosage of opioid
painkillers without withdrawal
symptoms
Omoigui et al., 2011 [171]i.v. 5 mg/kgeffective treatment for the opioid withdrawal symptoms and pain during transition to buprenorphine
Pradhan and Rossi, 2020 [172]i.v. 0.75 mg/kgCombination therapy with
ketamine, rTMS and TIMBER
is feasible in patients with opioid
addiction, reduces craving
and promotes abstinence
Table 5. Essential information about the TRANSFORM clinical trial.
Table 5. Essential information about the TRANSFORM clinical trial.
TRANSFORM-1TRANSFORM-2TRANSFORM-3
Features of a clinical trialRandomized, double-blinded and placebo controlled
Number of respondents
included in the analysis
324223137
Age of respondents18–6418–64≥65
Basic selection criterionRecurrent MDD or an episode of MDD lasting ≥ 2 years without psychotic featuresRecurrent MDD or an episode of MDD lasting ≥ 2 years without psychotic featuresMDD without
psychotic features and resistant to ≥2 different AD
Dosage in groups56 mg or 84 mg of
S-ketamine + AD,
placebo + AD
56 mg or 84 mg of
S-ketamine + AD,
placebo + AD
28 mg, 56 mg
or 84 mg of S-ketamine + AD,
placebo + AD
Duration of treatment phase28 days28 days28 days
Table 6. Significant results from clinical trials TRANSFORM.
Table 6. Significant results from clinical trials TRANSFORM.
TRANSFORM-1TRANSFORM-2TRANSFORM-3
Initial average score on the MADRS37.5537.1535.2
Mean change in MADRS scoreS-ketamine−18.9−21.4−10
Placebo −14.8−17−6.3
Mean change in irritability on the 7-GAD scaleS-ketamine−7.4−7.9-
Placebo−6−6.8
Summary response and remission by day 28 S-ketamine53.6%69.3%44.4%
Placebo38.9%52%20%
Table 7. Essential information about the SUSTAIN-1 clinical trial.
Table 7. Essential information about the SUSTAIN-1 clinical trial.
SUSTAIN-1
Main objective of the clinical trialLong-term effectiveness
Features of the clinical trialRandomized, double-blinded, placebo controlled
Number
of respondents
705
Age of respondents18–64
Basic selection
criterion
Recurrent MDD or an episode of MDD lasting ≥ 2 years without psychotic features. No suicidal thoughts or behavior.
Duration analyzedInduction phase (4 weeks)
Optimization phase (12 weeks)
Sustaining phase (variable duration)
DosageVariable dosage 56 mg or 84 mg S-ketamine with an AD.
Induction phase—twice a week
Optimization phase—one or two times a week
Sustaining phase—AD only
Table 8. Relapse in the group with stable remission and stable response.
Table 8. Relapse in the group with stable remission and stable response.
RelapseS-ketamine + ADPlacebo + AD
Patients with stable remission [%]26.745.3
Mean time to relapse in stable remission in days63588
Patients with stable response [%]25.857.6
Table 9. Essential information about the SUSTAIN-2 clinical trial.
Table 9. Essential information about the SUSTAIN-2 clinical trial.
SUSTAIN-2
Main objective of the clinical trialLong-term safety and effectiveness
Features of the clinical trialNon-randomized, no placebo
Number of respondents802
Age of respondents≥18
Basic selection criterionMDD without psychotic features and resistant to ≥2 different antidepressants. No suicidal thoughts or behavior.
Duration analyzedInduction phase (4 weeks)
Sustaining phase (48 weeks)
Observation phase (up to one year)
DosageFlexible dosage of 28 mg (in age of ≥65), 56 mg or 84 mg S-ketamine with an AD
Induction phase—twice a week
Sustaining phase—one or two times a week
Observation phase—AD only
Table 10. Changes in MADRS and 7-GAD score in the SUSTAIN-2 clinical trial.
Table 10. Changes in MADRS and 7-GAD score in the SUSTAIN-2 clinical trial.
Result
Mean baseline MADRS score31.4 ± 5.39
Mean change in the MADRS score−16.4
Clinical response of respondents (↓MADRS ≥ 50%)Induction phase78.4%
Optimization/observation phase76.5%
Remission of respondents (MADRS ≤ 12)Induction phase47.2%
Optimization/observation phase58.2%
Mean change in 7-GAD scoreInduction phase−5.9
Optimization/observation phase0.2
Table 11. Summary of mean incidence of significant adverse event in TRANSFORM-1 and TRANSFORM-2 clinical trials. AD—antidepressant.
Table 11. Summary of mean incidence of significant adverse event in TRANSFORM-1 and TRANSFORM-2 clinical trials. AD—antidepressant.
Total S-Ketamine + AD (343 Participants)Total Placebo + AD
(222 Participants)
Incidence of Adverse Event [%]
Adverse event27.88.5
Nausea26.53.6
Dissociation22.66.7
Dizziness20.217.1
Headache16.39
Somnolence20.213.5
Dysgeusia8.92.2
Blood pressure increased27.88.5
Table 12. Frequency of TEAE and TEAE suggesting abuse in the SUSTAIN-2 clinical trial.
Table 12. Frequency of TEAE and TEAE suggesting abuse in the SUSTAIN-2 clinical trial.
Adverse Effects[%]
Prevalence TEAE90.1
≥1 serious TEAE14.7
Serious TEAE associated with increased pressure12.8
TEAE suggesting abuse53.5
OverdoseNot reported
Abuse of S-ketamine
Request of increased dosage
Attempting to get the drug
Table 13. Most common adverse effects in patients in the SUSTAIN-2 clinical trial during S-ketamine use.
Table 13. Most common adverse effects in patients in the SUSTAIN-2 clinical trial during S-ketamine use.
[%]
Dizziness32.9
Headache24.9
Cognitive impairmentNot reported
Somnolence16.7
Dysgeusia11.8
Dissociation27.6
Nausea25.1
Vomiting10.8
Urinary tract infection8.1
Bladder inflammationNot reported
Increase in blood pressure9.4
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

Gibuła-Tarłowska, E.; Wiszniewska, A.; Turyk, M.; Szymczyk, P.; Kotlińska, J.H.; Kędzierska, E. Ketamine—From an Anesthetic to a Psychiatric Drug: Mechanisms of Action, Clinical Applications and Potential Risks. Molecules 2025, 30, 2824. https://doi.org/10.3390/molecules30132824

AMA Style

Gibuła-Tarłowska E, Wiszniewska A, Turyk M, Szymczyk P, Kotlińska JH, Kędzierska E. Ketamine—From an Anesthetic to a Psychiatric Drug: Mechanisms of Action, Clinical Applications and Potential Risks. Molecules. 2025; 30(13):2824. https://doi.org/10.3390/molecules30132824

Chicago/Turabian Style

Gibuła-Tarłowska, Ewa, Anna Wiszniewska, Magdalena Turyk, Paulina Szymczyk, Jolanta H. Kotlińska, and Ewa Kędzierska. 2025. "Ketamine—From an Anesthetic to a Psychiatric Drug: Mechanisms of Action, Clinical Applications and Potential Risks" Molecules 30, no. 13: 2824. https://doi.org/10.3390/molecules30132824

APA Style

Gibuła-Tarłowska, E., Wiszniewska, A., Turyk, M., Szymczyk, P., Kotlińska, J. H., & Kędzierska, E. (2025). Ketamine—From an Anesthetic to a Psychiatric Drug: Mechanisms of Action, Clinical Applications and Potential Risks. Molecules, 30(13), 2824. https://doi.org/10.3390/molecules30132824

Article Metrics

Back to TopTop