1Department of Pharmacology, Matoshri College of Pharmacy, Eklahare, Nashik, Maharashtra, India
2Department of Pharmacology, K. V. N. Naik S. P. Sanstha's, Institute of Pharmaceutical Education & Research, Nashik, Maharashtra, India
Ketamine, originally synthesized in the early 1960s as a safer alternative to phencyclidine (PCP), has since evolved into a multifaceted agent with applications extending far beyond its initial use as a dissociative anesthetic in surgical settings. With a unique pharmacological profile characterized by N-methyl-D-aspartate (NMDA) receptor antagonism, ketamine induces rapid anesthesia, analgesia, and dissociation. However, over the past two decades, it has garnered significant attention for its rapid-acting antidepressant effects, particularly in individuals with treatment-resistant depression (TRD), a condition that fails to respond to conventional antidepressants. Clinical trials and real-world data have demonstrated that low-dose ketamine, administered intravenously or intranasally, can produce profound and sustained improvements in mood within hours, a stark contrast to the delayed onset of traditional antidepressants.The therapeutic promise of ketamine has extended into the realms of post-traumatic stress disorder (PTSD), bipolar depression, anxiety disorders, and suicidal ideation, positioning it as a breakthrough in psychiatric medicine. Nevertheless, ketamine’s benefits are tempered by concerns surrounding its safety, potential for abuse, and long-term neuropsychiatric effects. As a Schedule III controlled substance, ketamine is also misused recreationally, known colloquially as “Special K,” for its hallucinogenic and dissociative properties. Chronic use has been associated with cognitive deficits, urinary tract dysfunction, and addiction, necessitating careful monitoring in both clinical and non-clinical contexts.This review paper provides a comprehensive examination of ketamine’s evolving role in modern medicine by exploring its pharmacodynamics and pharmacokinetics, mechanisms of antidepressant action, and evidence from clinical studies. It also delves into its misuse potential, societal impact, ethical considerations in off-label psychiatric use, and regulatory challenges. By synthesizing data from preclinical and clinical research, the paper seeks to present a balanced perspective on ketamine’s dual identity—as a therapeutic breakthrough and a drug of misuse—highlighting the imperative for responsible clinical integration and ongoing research.
Ketamine (2-(2-chlorophenyl)-2-(methylamino)cyclohexanone) stands as one of the most intriguing and versatile compounds in modern pharmacology. Synthesized in 1962 and approved for medical use in 1970, ketamine was initially introduced as a dissociative anesthetic with a better safety profile than its predecessor, phencyclidine (PCP). Its rapid onset, profound analgesic properties, and minimal respiratory depression made it a valuable anesthetic, especially in battlefield and emergency settings. Unlike many traditional anesthetics, ketamine produces a trance-like state characterized by profound analgesia, sedation, and amnesia, while maintaining airway reflexes and cardiovascular stability. These properties positioned ketamine as a vital tool in surgical and critical care settings, particularly in resource-limited environments. In recent decades, ketamine’s clinical profile has expanded dramatically beyond its traditional anesthetic role. A growing body of research has uncovered its powerful antidepressant effects, especially in individuals suffering from treatment-resistant depression (TRD), where conventional pharmacotherapies often fail. Low-dose intravenous ketamine and, more recently, intranasal esketamine (an enantiomer of ketamine), have shown rapid and robust antidepressant activity, sometimes within hours of administration. These findings have revolutionized the approach to mood disorders and prompted a re-evaluation of the neurobiology of depression itself. The emerging role of ketamine in psychiatry now includes applications for anxiety disorders, bipolar depression, post-traumatic stress disorder (PTSD), and suicidal ideation, with ongoing clinical trials exploring even broader indications. The pharmacological underpinnings of ketamine’s effects are multifaceted. Primarily, ketamine acts as a non-competitive antagonist of the N-methyl-D-aspartate (NMDA) receptor, a subtype of glutamate receptor involved in synaptic plasticity, learning, and memory. This mechanism not only accounts for its anesthetic and dissociative effects but also underlies its rapid antidepressant action, possibly through enhancement of synaptogenesis and neurotrophic signaling pathways, such as brain-derived neurotrophic factor (BDNF) and mammalian target of rapamycin (mTOR). However, ketamine’s pharmacodynamics extend beyond NMDA receptor antagonism, involving interactions with opioid receptors, monoaminergic systems, and inflammatory pathways—highlighting its complex and still not fully understood mechanism of action. Despite these promising therapeutic applications, ketamine’s profile is shadowed by its significant potential for misuse and dependence. Recreational users seek its hallucinogenic and dissociative effects, often referring to experiences such as the “K-hole,” a state of extreme dissociation. Chronic recreational use has been associated with a range of health issues, including cognitive impairment, urinary tract dysfunction (ketamine-induced ulcerative cystitis), hepatic toxicity, and psychological dependence. These risks complicate ketamine’s integration into routine psychiatric practice and raise ethical, regulatory, and societal concerns. As ketamine straddles the line between a revolutionary therapeutic agent and a substance of abuse, its role in medicine remains complex and dynamic. The regulatory landscape continues to evolve in response to its rising medical use and concurrent misuse. In some countries, ketamine clinics have emerged, offering outpatient treatment for depression, anxiety, and chronic pain. At the same time, law enforcement and public health officials grapple with increasing illicit use. This review aims to present a comprehensive and critical analysis of ketamine’s multifaceted identity in modern medicine. It explores the historical development of the drug, its pharmacological mechanisms, clinical applications, and therapeutic efficacy. It also addresses the risks associated with misuse, dependence, and long-term health outcomes. Through an examination of current research, clinical practices, and ongoing debates, this paper provides an in-depth understanding of ketamine’s medical evolution and its implications for the future of psychiatric and anesthetic care.
Ketamine in Modern Medicine
2. Historical Overview
2.1 Discovery and Synthesis
Ketamine was first synthesized in 1962 by Dr. Calvin L. Stevens, a chemist at Parke-Davis Laboratories. It was developed as part of the search for a safer alternative to phencyclidine (PCP), a dissociative anesthetic known for its powerful analgesic properties but also for its severe side effects including intense hallucinations, delirium, and neurotoxicity [2]. Ketamine’s molecular structure was designed to retain PCP’s anesthetic and analgesic efficacy while minimizing adverse effects. Its unique pharmacological profile—producing dissociation without deep respiratory depression—made it an attractive candidate for clinical use. Early animal studies confirmed ketamine’s safety and efficacy as an anesthetic and analgesic, paving the way for human trials.
2.2 Early Medical Use
Ketamine received FDA approval for human use in 1970 and quickly became a popular anesthetic agent, particularly in situations requiring rapid and effective sedation with minimal respiratory compromise. Its use expanded rapidly in surgical settings due to its ability to induce a trance-like state known as “dissociative anesthesia,” where patients remain conscious but detached from pain and external stimuli. During the Vietnam War (1965–1973), ketamine gained particular prominence as a battlefield anesthetic. Its rapid onset, cardiovascular stability, and relative safety in field conditions made it invaluable for trauma care in combat zones where conventional anesthetics were impractical or unavailable [3]. Beyond anesthesia, ketamine was explored for use in emergency medicine, pediatrics, and veterinary medicine, solidifying its status as a versatile drug with a wide safety margin.
2.3 Evolution into Psychiatry
The psychiatric potential of ketamine emerged much later. In the late 1990s and early 2000s, researchers noticed that patients administered ketamine for anesthesia or pain relief sometimes experienced rapid improvements in mood and relief from depressive symptoms [4]. This serendipitous observation led to formal investigations into ketamine’s effects on mental health disorders, particularly treatment-resistant depression (TRD). Unlike traditional antidepressants, which often take weeks to show efficacy, ketamine demonstrated rapid antidepressant effects within hours of administration. Early clinical trials began focusing on low-dose intravenous ketamine infusions as a novel treatment approach for major depressive disorder (MDD) and bipolar depression. Subsequent research revealed that ketamine acts primarily as an NMDA receptor antagonist, modulating glutamate transmission in the brain and triggering neuroplasticity pathways that may underlie its rapid mood-elevating effects. Ketamine’s emergence in psychiatry also spurred the development of esketamine, the S-enantiomer of ketamine, which received FDA approval in 2019 as a nasal spray for TRD, further validating ketamine’s therapeutic potential. Over the past two decades, ketamine’s role has expanded beyond anesthesia into a promising treatment for mood disorders, PTSD, and chronic pain syndromes, reflecting a major shift in its clinical use and societal perception.
3.1 NMDA Receptor Antagonism
Ketamine’s primary and most studied pharmacological action is its role as a non-competitive antagonist at the N-methyl-D-aspartate (NMDA) receptor, a subtype of glutamate receptor found abundantly throughout the central nervous system [5]. By binding to the phencyclidine site within the ion channel of the NMDA receptor, ketamine blocks the influx of calcium ions, thereby reducing excitatory neurotransmission. This blockade leads to a disruption of normal synaptic activity, which is believed to underlie its anesthetic and dissociative effects. The NMDA receptor antagonism is particularly relevant in the modulation of pain pathways and is central to ketamine’s rapid antidepressant properties. Unlike traditional NMDA antagonists, ketamine’s effects are transient and dose-dependent, with subanesthetic doses selectively modulating synaptic activity without causing profound unconsciousness. This selective inhibition disrupts pathological glutamate excitotoxicity, which is implicated in neurodegenerative diseases and mood disorders, offering a neuroprotective advantage.
3.2 AMPA Receptor Stimulation
Interestingly, ketamine’s inhibition of NMDA receptors leads to a paradoxical increase in extracellular glutamate levels. This increase is thought to result from disinhibition of presynaptic neurons, which enhances the release of glutamate into the synaptic cleft [6]. The excess glutamate then stimulates alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, another class of ionotropic glutamate receptors. Activation of AMPA receptors initiates downstream intracellular signaling cascades critical for synaptic plasticity, including calcium influx and activation of kinases that promote the strengthening of synaptic connections. This cascade is believed to be essential for the rapid antidepressant effects of ketamine, as it facilitates neuroplastic changes in the prefrontal cortex and hippocampus, regions associated with mood regulation and cognitive function. Enhanced AMPA receptor throughput may restore impaired synaptic connectivity observed in depression and other neuropsychiatric conditions.
3.3 Opioid and Monoaminergic Systems
Beyond its glutamatergic actions, ketamine interacts with multiple neurotransmitter systems, contributing to its complex pharmacodynamics. Ketamine exhibits affinity for opioid receptors, particularly the mu and kappa subtypes, which may partly explain its analgesic properties [7]. This interaction is subtle but significant, as it complements NMDA receptor blockade in reducing pain perception without the respiratory depression typical of classical opioids. Additionally, ketamine influences monoaminergic neurotransmission by modulating dopamine and serotonin pathways. It increases dopamine release in the prefrontal cortex and striatum, potentially enhancing motivation and reward processing. Similarly, ketamine affects serotonergic transmission, which plays a crucial role in mood regulation. These effects collectively contribute to ketamine’s antidepressant efficacy and may help alleviate symptoms such as anhedonia and low motivation commonly seen in treatment-resistant depression.
3.4 Neurotrophic and Anti-inflammatory Effects
Ketamine’s influence extends beyond neurotransmitter systems to the molecular and cellular levels, where it promotes neurotrophic and anti-inflammatory processes. One of the key molecular pathways activated by ketamine is the mammalian target of rapamycin (mTOR) signaling pathway, which regulates protein synthesis necessary for synaptic growth and remodeling [8]. Ketamine rapidly increases levels of brain-derived neurotrophic factor (BDNF), a protein vital for neuronal survival, differentiation, and synaptic plasticity. This enhancement of BDNF and activation of mTOR signaling facilitate the formation of new synaptic connections and dendritic spine growth, processes that are often impaired in depression and chronic stress. In parallel, ketamine exhibits anti-inflammatory properties by reducing pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), which are elevated in many psychiatric and neurodegenerative disorders. This immunomodulatory action may contribute to its therapeutic effects in depression, where inflammation is increasingly recognized as a contributing factor. Together, these neurotrophic and anti-inflammatory effects position ketamine as a unique therapeutic agent capable of reversing neurobiological deficits associated with mood disorders and chronic pain.
4. Ketamine in Anesthesia and Emergency Medicine
4.1 General Anesthesia
Ketamine has long been a cornerstone in the practice of general anesthesia, particularly valued for its unique pharmacological properties that distinguish it from other anesthetics. Unlike many anesthetics that suppress cardiovascular function, ketamine stimulates the sympathetic nervous system, resulting in increased heart rate, blood pressure, and cardiac output. These effects make ketamine especially useful in patients with cardiovascular instability or shock, where maintaining hemodynamic stability is critical [9]. Additionally, ketamine possesses potent bronchodilatory properties, which render it a preferred agent in patients with reactive airway diseases such as asthma or chronic obstructive pulmonary disease (COPD). Its ability to maintain spontaneous respiration and airway reflexes, even at anesthetic doses, reduces the risk of respiratory depression—a common concern with other anesthetics. Clinicians utilize ketamine for both induction and maintenance of anesthesia in a wide variety of surgical procedures, particularly when intubation and mechanical ventilation pose additional risks.
4.2 Procedural Sedation
In emergency medicine, ketamine is widely used for procedural sedation and analgesia (PSA) due to its rapid onset and favorable safety profile [10]. It allows patients to tolerate painful or anxiety-provoking procedures while maintaining airway patency, spontaneous breathing, and protective airway reflexes. This is particularly advantageous in pediatric populations and trauma patients, who are often at increased risk of respiratory compromise. Ketamine’s dissociative anesthesia enables rapid sedation without the deep unconsciousness associated with other sedatives, facilitating quicker recovery times. Its analgesic properties further reduce the need for opioid administration during procedures. Emergency physicians commonly use ketamine for fracture reductions, laceration repairs, and other minor surgical interventions, especially when intravenous access is limited or patient cooperation is poor.
4.3 Battlefield and Remote Applications
Ketamine’s stability under a variety of environmental conditions, ease of administration via multiple routes (intravenous, intramuscular, intranasal), and wide therapeutic index have made it an indispensable anesthetic in battlefield medicine and remote healthcare settings [11]. During the Vietnam War, ketamine was extensively employed to provide rapid anesthesia and analgesia for wounded soldiers in field hospitals and during evacuation. Its capacity to preserve cardiovascular and respiratory function, combined with minimal monitoring requirements, allows its use in austere environments where advanced anesthesia equipment is unavailable. Modern military medicine continues to rely on ketamine for trauma care, including pre-hospital emergency medical services, combat medics, and disaster response teams. In remote or rural healthcare settings, ketamine offers a practical solution for pain management and sedation where resources and specialist care are limited.
4.4 Pediatric Applications
Ketamine has established itself as a leading agent for pediatric anesthesia and sedation due to its safety and efficacy profile [12]. Children undergoing short diagnostic or therapeutic procedures benefit from ketamine’s rapid onset, profound analgesia, and relatively minimal cardiorespiratory depression. Its ability to maintain airway reflexes reduces the risk of aspiration, a critical consideration in pediatric sedation. The drug’s short duration of action and rapid recovery enable efficient turnover in busy clinical settings such as emergency departments, dental surgeries, and imaging studies. Additionally, ketamine’s amnestic effects help minimize psychological trauma associated with medical procedures in young patients. Despite some potential side effects such as emergence reactions or increased salivation, careful dosing and adjunctive medications help mitigate these issues, making ketamine a preferred choice in pediatric anesthesia worldwide.
4.5 Veterinary Applications
Ketamine is extensively used in veterinary medicine across a variety of species and clinical indications [13]. It serves as a primary anesthetic agent for surgical procedures, diagnostic interventions, and immobilization of animals ranging from domestic pets to large wildlife. The drug’s wide margin of safety, rapid onset, and versatility in administration routes make it highly suitable for field and clinical use. Veterinarians often combine ketamine with sedatives or muscle relaxants to tailor anesthesia depth and duration to the specific needs of the animal. Its analgesic properties also improve postoperative comfort. Ketamine’s role in veterinary anesthesia mirrors its importance in human medicine, with ongoing research aimed at optimizing protocols to enhance animal welfare and procedural outcomes.
5.1 Emergence in Psychiatry
Ketamine’s transition from an anesthetic to a psychiatric treatment represents one of the most significant breakthroughs in neuropsychiatry over the past few decades. The initial serendipitous observation of its antidepressant effects occurred in the late 1990s and early 2000s, when clinicians noticed that patients receiving ketamine for anesthesia or chronic pain reported rapid mood improvements [14]. This was particularly notable in individuals suffering from treatment-resistant depression (TRD), a subset of depressive disorders unresponsive to conventional antidepressants such as selective serotonin reuptake inhibitors (SSRIs) and tricyclics. Subsequent controlled clinical trials confirmed that a single subanesthetic dose of ketamine administered intravenously could produce robust antidepressant effects within hours, often lasting up to a week. This rapid onset of action marked a stark contrast to traditional antidepressants, which typically require several weeks to achieve clinical efficacy, thereby positioning ketamine as a potential lifesaving option, especially in acutely suicidal patients.
5.2 Mechanistic Insights
Ketamine’s antidepressant actions are intricately linked to its unique modulation of glutamatergic neurotransmission. The drug’s antagonism of the NMDA receptor leads to a surge in extracellular glutamate, which activates AMPA receptors on postsynaptic neurons. This glutamate-AMPA receptor interaction initiates a cascade of intracellular signaling that culminates in enhanced synaptogenesis— the formation of new synaptic connections—particularly in the prefrontal cortex and hippocampus [15]. These brain regions are critical for regulating mood, cognition, and emotional processing, and are often impaired in depression. Ketamine also activates the mammalian target of rapamycin (mTOR) pathway and elevates brain-derived neurotrophic factor (BDNF) levels, both of which are essential for neuroplasticity and synaptic remodeling. Together, these mechanisms help reverse the synaptic deficits induced by chronic stress and depression, underpinning the rapid and sustained antidepressant effects observed clinically.
5.3 Routes of Administration
Ketamine’s versatility is reflected in its multiple routes of administration, each with distinct pharmacokinetic and clinical implications:
5.4 Duration and Maintenance
While a single ketamine infusion can produce rapid antidepressant effects, these benefits typically persist for approximately one week, necessitating repeated dosing to maintain symptom relief [16]. Maintenance therapy regimens vary widely, with some protocols recommending biweekly or monthly infusions depending on patient response and tolerability. The optimal frequency and duration of maintenance ketamine treatment remain areas of active investigation, as standardized clinical guidelines are still evolving. Long-term management also involves integrating ketamine therapy with psychotherapeutic interventions and conventional antidepressants to sustain remission and prevent relapse. Careful monitoring during maintenance is essential to mitigate potential risks such as tolerance, side effects, or abuse.
5.5 Safety and Side Effects
Ketamine administration is generally well-tolerated when used in controlled clinical environments, but it is not without risks. Short-term side effects commonly include transient dissociative symptoms (e.g., feelings of detachment or unreality), nausea, dizziness, elevated blood pressure, and perceptual disturbances [17]. These effects typically resolve within hours after administration. Long-term safety data are limited but raise concerns about potential cognitive impairments, urinary tract toxicity (ketamine cystitis), and neurotoxicity with repeated or high-dose exposure. Additionally, ketamine’s psychoactive properties carry a risk for misuse and dependence, warranting stringent patient selection and monitoring. Current research is focused on understanding the long-term neuropsychological effects and establishing safety parameters for chronic use.
5.6 Comparison with Conventional Antidepressants
Ketamine stands apart from conventional antidepressants such as SSRIs, serotonin-norepinephrine reuptake inhibitors (SNRIs), and monoamine oxidase inhibitors (MAOIs), primarily due to its rapid onset of action. Traditional antidepressants often require 4 to 6 weeks or longer to achieve significant clinical improvement, a delay that poses serious risks in severely depressed or suicidal patients [18]. In contrast, ketamine’s rapid mood-elevating effects can manifest within hours, making it a vital option for acute crisis intervention. Furthermore, ketamine is effective in patients who have failed multiple antidepressant trials, addressing a critical unmet need in psychiatry. However, ketamine’s effects are typically transient, and it is not yet considered a standalone long-term treatment. Conventional antidepressants, despite their slower onset, remain essential for sustained maintenance therapy and relapse prevention, often used alongside ketamine to enhance and prolong therapeutic outcomes.
6. Ketamine in Pain Management
6.1 Chronic and Neuropathic Pain
Ketamine has emerged as a valuable therapeutic agent in the management of chronic and neuropathic pain syndromes, conditions often resistant to conventional analgesics. Chronic Regional Pain Syndrome (CRPS), fibromyalgia, and spinal cord injury-associated neuropathic pain are among the most studied indications where ketamine demonstrates efficacy [19]. These conditions involve pathological central sensitization, where the nervous system becomes hyper-responsive to pain stimuli, resulting in persistent and often debilitating pain. Ketamine’s NMDA receptor antagonism disrupts this sensitization process by inhibiting excitatory neurotransmission and reducing the amplification of pain signals within the spinal cord and brain. Beyond receptor blockade, ketamine modulates descending inhibitory pain pathways and enhances endogenous analgesic mechanisms, increasing overall pain tolerance. Clinical protocols often involve low-dose intravenous infusions administered over several days or weeks, which can lead to sustained reductions in pain intensity and improvements in functional status. Despite its promising effects, variability in patient response and concerns about long-term safety require individualized treatment planning and close monitoring.
6.2 Perioperative Use
In the perioperative setting, ketamine has gained prominence as an adjunct to opioid analgesia, particularly in surgeries associated with severe postoperative pain or in patients with opioid tolerance [20]. Low-dose ketamine infusions administered intraoperatively or immediately postoperatively reduce the requirement for opioids, thereby minimizing opioid-related side effects such as respiratory depression, nausea, and constipation. This opioid-sparing effect is especially beneficial in chronic pain patients undergoing surgery, as their baseline opioid tolerance often necessitates higher doses for adequate analgesia. Additionally, ketamine’s analgesic properties improve pain control during the critical early postoperative period, which can facilitate earlier mobilization and reduce the risk of chronic postsurgical pain development. Various anesthetic protocols incorporate ketamine as part of multimodal analgesia, combining it with regional anesthesia and non-opioid medications to optimize patient outcomes.
6.3 Cancer and Palliative Care
Ketamine’s role in cancer pain management and palliative care has been increasingly recognized, particularly for patients experiencing refractory pain unresponsive to escalating doses of opioids [21]. In advanced malignancies, pain mechanisms are often complex, involving nociceptive, neuropathic, and inflammatory components. Ketamine’s multifaceted actions allow it to address several of these mechanisms simultaneously. By reducing central sensitization and modulating inflammatory pathways, ketamine enhances analgesia without the respiratory depressive effects commonly associated with high-dose opioids. This opioid-sparing effect not only improves pain control but also mitigates opioid-induced side effects, thereby improving patients’ quality of life during end-of-life care. Ketamine can be administered via multiple routes including intravenous, oral, subcutaneous, or intrathecal, allowing for flexible dosing tailored to patient needs and care settings. While evidence supports its use in refractory cancer pain, further clinical trials are needed to standardize dosing regimens and define long-term safety in this vulnerable population.
6.4 Anti-inflammatory Role
Recent research has illuminated ketamine’s anti-inflammatory properties, which may contribute to its analgesic and antidepressant effects [22]. Ketamine inhibits the production and release of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β), which are implicated in the pathogenesis of chronic pain and depression. By modulating the immune response, ketamine helps attenuate neuroinflammation within the central nervous system, a recognized driver of persistent pain and mood disorders. This immunomodulatory effect enhances neuronal survival and promotes neuroplasticity, aligning with its observed benefits in neuropathic pain and depressive symptoms. Understanding ketamine’s role in inflammation opens potential avenues for novel therapeutic applications in neuroimmune disorders and highlights the drug’s complex pharmacodynamic profile beyond traditional anesthetic and analgesic mechanisms.
7. Recreational Use and Misuse
7.1 Patterns of Recreational Use
Ketamine’s unique pharmacological profile, characterized by dissociative and hallucinogenic effects, has contributed to its widespread popularity as a recreational drug. Since the late 20th century, ketamine has become a staple in club and rave scenes worldwide, often referred to colloquially as “K,” “Special K,” or “Kit-Kat.” Recreational users typically consume ketamine in powdered form, insufflating (snorting) the substance to achieve rapid onset of effects. It is also used in liquid form for injection or, less commonly, swallowed or smoked. The drug induces an altered state of consciousness, characterized by feelings of detachment from the environment and self (dissociation), visual and auditory hallucinations, and euphoria. At higher doses, users may experience what is known as the “K-hole,” a profound dissociative state resembling out-of-body experiences or near-anesthetic unconsciousness. The unpredictable nature of the effects, coupled with polydrug use common in recreational settings, increases the risks associated with ketamine misuse [23].
7.2 Health Consequences
Chronic and high-dose recreational use of ketamine is associated with a range of adverse health outcomes affecting multiple organ systems. One of the most well-documented complications is ketamine-induced urinary tract toxicity, often termed “ketamine bladder syndrome.” This condition is characterized by severe urinary frequency, urgency, pain, and hematuria, progressing to ulcerative cystitis and even irreversible bladder fibrosis in some cases. The exact pathophysiology is not fully understood but is believed to involve direct toxicity to the urothelium and inflammatory processes [24]. Long-term misuse may also lead to cognitive decline, particularly impairments in memory, attention, and executive function, which may be partially reversible with cessation but could persist in some individuals. Hepatic damage, including elevated liver enzymes and, rarely, more severe hepatotoxicity, has also been reported, possibly due to the drug’s metabolic burden and oxidative stress. Psychological dependence on ketamine can develop, characterized by cravings, compulsive use despite negative consequences, and withdrawal symptoms such as anxiety, tremors, and cravings upon cessation. The dissociative and hallucinogenic properties that attract recreational users may paradoxically exacerbate underlying psychiatric disorders or precipitate psychotic episodes, especially in vulnerable populations.
7.3 Legal Status
Recognizing ketamine’s medical utility alongside its abuse potential, governments worldwide have implemented regulatory controls to balance access and misuse prevention. In the United States, ketamine is classified as a Schedule III controlled substance, indicating moderate to low potential for physical and psychological dependence but recognized medical uses [25]. This scheduling imposes restrictions on manufacturing, prescribing, dispensing, and possession outside authorized medical contexts. Internationally, ketamine’s legal status varies but generally falls under controlled substance frameworks, with many countries restricting non-medical possession and trafficking. Despite these controls, illicit ketamine manufacture and distribution persist, particularly in regions with limited regulatory enforcement. Law enforcement efforts, combined with public health policies, strive to curb illicit ketamine availability while ensuring legitimate medical use is not impeded.
7.4 Social and Public Health Implications
The increasing prevalence of ketamine misuse, especially among adolescents and young adults, poses significant social and public health challenges [26]. Recreational use is often normalized in nightlife and music festival cultures, complicating prevention efforts. The health consequences of misuse, including physical morbidity and mental health deterioration, place additional burdens on healthcare systems. Moreover, ketamine’s potential to cause cognitive and psychological harm affects educational attainment, employment, and social functioning, perpetuating cycles of disadvantage. Addressing these issues requires a multifaceted approach involving public awareness campaigns, school-based drug education, harm reduction strategies, and accessible treatment services for substance use disorders. Policymakers must balance regulatory enforcement with harm minimization, ensuring that punitive measures do not drive users away from seeking help. Collaboration between healthcare providers, law enforcement, community organizations, and researchers is essential to develop evidence-based interventions tailored to at-risk populations. Emerging technologies, such as drug checking and digital outreach, also offer innovative avenues for early intervention and education.
8. Ethical and Clinical Considerations
8.1 Commercialization and Clinics
The rapid rise of ketamine clinics across the United States and globally has introduced new ethical dilemmas concerning the commercialization of medical treatments, particularly those with evolving evidence bases. While many of these clinics are founded by well-intentioned clinicians, the for-profit nature of some centers has raised concerns about over prescription, insufficient medical oversight, and the exploitation of vulnerable patients. Many clinics offer off-label ketamine treatments for psychiatric conditions such as depression, anxiety, PTSD, and even chronic pain, sometimes without the comprehensive multidisciplinary support needed for safe administration. Unlike FDA-approved medications such as esketamine (Spravato), which are administered under controlled conditions, off-label use of racemic ketamine in these clinics may lack robust protocols or psychiatric follow-up. Some centers advertise ketamine as a near-cure for mood disorders, contributing to therapeutic misconceptions. Ethical concerns also include whether patients are receiving adequate psychological assessment, integration therapy, or informed guidance before and after treatments [27]. The commodification of ketamine risks prioritizing profit over patient welfare, necessitating clear regulations and accreditation standards to safeguard clinical integrity.
8.2 Informed Consent
Administering ketamine for psychiatric or off-label use requires a nuanced approach to informed consent, ensuring that patients are fully aware of the benefits, risks, and limitations of the therapy. Unlike traditional antidepressants, ketamine’s effects are rapid but often short-lived, requiring repeated treatments with still-uncertain long-term safety profiles. Patients must understand the experimental nature of some uses, especially those not yet supported by large-scale randomized controlled trials. Informed consent should include discussions about:
8.3 Guidelines and Training
The expansion of ketamine use for mental health necessitates urgent development of standardized treatment guidelines and comprehensive training programs for healthcare providers. While some professional bodies, such as the American Psychiatric Association and the American Society of Anesthesiologists, have begun to outline best practices, consensus on proper screening, dosing regimens, monitoring procedures, and treatment duration remains incomplete. Training must go beyond basic pharmacology, equipping providers with skills in managing adverse events, recognizing misuse or dependency, and offering psychological support during and after administration. Psychiatrists, anesthesiologists, pain specialists, and primary care physicians must be brought into a shared educational framework to ensure continuity and safety of care. In the absence of unified guidelines, considerable variation exists in clinical practice, increasing the risk of inconsistent outcomes and patient harm. Credentialing systems and institutional oversight mechanisms are essential to maintaining a high standard of care in ketamine administration [29].
8.4 Access and Equity
Despite its potential to transform psychiatric treatment, ketamine therapy remains financially inaccessible to many patients, particularly those from marginalized communities. Insurance coverage is limited for off-label uses, forcing most patients to pay out of pocket—often at significant cost. The high price of repeated sessions, coupled with travel and time commitments, disproportionately excludes low-income populations, thereby exacerbating existing health disparities. Furthermore, clinics are often concentrated in urban centers, creating geographical barriers for rural and underserved populations. Ensuring equitable access to ketamine therapy requires systemic policy interventions. These may include:
9. Future Directions
9.1 Drug Development
As ketamine continues to demonstrate promise in treating psychiatric and pain disorders, researchers are intensifying efforts to develop next-generation compounds that preserve its therapeutic benefits while minimizing adverse effects. A major focus is on stereoisomer optimization—particularly arketamine, the R-enantiomer of ketamine, which has shown potential for antidepressant efficacy with a more favorable safety profile than esketamine (the S-enantiomer). Arketamine may exhibit reduced psychotomimetic and dissociative side effects, making it more tolerable for long-term clinical use [31]. Another promising area of research involves ketamine metabolites, especially (2R,6R)-hydroxynorketamine (HNK), which may mediate antidepressant effects independently of NMDA receptor antagonism. HNK is being explored for its ability to enhance synaptic plasticity without producing sedation, addiction potential, or hallucinations. The goal is to develop rapid-acting antidepressants that retain the neuroplastic and mood-stabilizing benefits of ketamine, while eliminating or drastically reducing its liability for abuse and cognitive side effects. Additionally, formulations such as slow-release implants, transdermal patches, and oral lozenges are being tested to improve administration convenience, compliance, and dosing precision.
9.2 Combination with Psychotherapy
Emerging evidence supports the integration of ketamine administration with psychotherapy, a model known as ketamine-assisted psychotherapy (KAP). Unlike standalone pharmacological treatments, KAP emphasizes the importance of therapeutic context, with ketamine sessions often paired with preparatory and integrative psychotherapy. The altered states induced by ketamine—marked by increased emotional openness, diminished psychological defenses, and heightened suggestibility—are believed to enhance engagement with deeply rooted traumas, anxiety, addiction, and treatment-resistant depression [32]. This synergy allows patients to confront and process repressed memories or maladaptive thought patterns in a therapeutically guided environment. Clinical studies suggest that KAP may produce more durable improvements in mood and functioning compared to ketamine or psychotherapy alone. Current protocols are being refined across various disciplines, including psychiatry, psychology, and psychedelic medicine, to optimize timing, dosage, and therapeutic approaches. Standardizing therapist training and integrating ethical and cultural competencies are key challenges in expanding the responsible use of KAP in mainstream mental health care.
9.3 Personalized Medicine
The variability in individual responses to ketamine therapy underscores the growing need for personalized medicine approaches. Not all patients benefit equally, and side effects may differ significantly based on genetic, neurochemical, and psychosocial factors. Researchers are now investigating genetic biomarkers, such as polymorphisms in genes related to the glutamatergic system (e.g., BDNF Val66Met), to predict ketamine responsiveness and tolerability [33]. Neuroimaging techniques, including fMRI and PET scans, are being used to identify functional brain signatures that correlate with clinical outcomes. These tools may help clinicians tailor treatments by selecting optimal dosing strategies, routes of administration, and treatment intervals. Moreover, individualized risk assessments can guide the management of patients at higher risk of adverse effects, such as those with a personal or family history of psychosis or substance use disorders. The future of ketamine therapy will likely involve integrated diagnostic platforms that combine genomic, neuroimaging, and clinical data to maximize therapeutic outcomes while minimizing risks.
9.4 AI and Digital Monitoring
The application of artificial intelligence (AI) and digital health technologies is transforming the landscape of ketamine treatment delivery and monitoring. AI-driven tools are being employed to analyze complex patient data sets in real time, enabling precision dosing, automated adverse event detection, and treatment response prediction [34]. Mobile health applications, wearable devices, and remote monitoring platforms can track physiological and psychological changes during and after ketamine sessions, offering insights into individual responses and facilitating early identification of complications. Some platforms integrate natural language processing to evaluate patient-reported outcomes or journal entries for signs of improvement or distress. These digital innovations not only enhance clinical safety but also support scalability by enabling telehealth-based ketamine treatments, particularly in underserved or remote areas. The integration of AI with electronic health records (EHRs) allows for continuous feedback loops, helping providers make data-informed decisions and refine therapeutic protocols dynamically. As ketamine moves further into mainstream use, AI and digital tools will be essential for ensuring responsible, responsive, and personalized care delivery.
CONCLUSION
Ketamine stands at the crossroads of innovation and controversy in modern medicine. Originally developed as a dissociative anesthetic, it has evolved into a multifaceted therapeutic agent with profound implications for anesthesiology, psychiatry, pain management, and beyond. Its unique pharmacological profile, particularly its rapid-acting antidepressant and analgesic properties, has revolutionized treatment paradigms for conditions historically deemed refractory to standard interventions, such as treatment-resistant depression, chronic pain syndromes, and severe post-traumatic stress disorder. The emergence of ketamine-based therapies offers a rare and vital advancement in fields that have seen limited breakthroughs over the past few decades. However, with its therapeutic promise comes significant concern. Ketamine’s dissociative and hallucinogenic effects, while clinically useful in controlled environments, are also what make it prone to recreational misuse. The increase in non-medical use, particularly among young adults and within nightlife subcultures, has led to serious public health challenges, including cognitive impairment, urinary tract dysfunction, and psychological dependence. This dual nature—as both a life-enhancing medicine and a potentially harmful substance—underscores the importance of a balanced, well-informed approach to its clinical application. From an ethical and regulatory standpoint, the rapid proliferation of ketamine clinics and off-label use calls for urgent standardization. Comprehensive treatment guidelines, enhanced physician training, and robust patient education are essential to ensure safety and effectiveness. Informed consent processes must clearly communicate both the known benefits and the uncertainties that remain, particularly regarding long-term safety and the risk of dependence. Furthermore, equitable access must be prioritized, as the high cost of ketamine therapies currently restricts availability to a privileged few, thereby exacerbating existing disparities in mental health care. Looking forward, ketamine’s future in medicine will be shaped by ongoing scientific discovery and innovation. Advances in drug development, such as novel enantiomers and non-dissociative metabolites, promise to refine its therapeutic index. The integration of ketamine with psychotherapy, personalized medicine tools, and AI-driven digital health platforms offers exciting opportunities to enhance patient outcomes and monitor safety. Nonetheless, these advancements must be pursued within a framework of rigorous clinical trials, ethical oversight, and transparent policy development. In conclusion, ketamine is a potent symbol of medicine’s evolving complexity—where a single compound can offer profound healing while posing serious risks. Its journey from the battlefield to the psychiatrist’s office encapsulates both the progress and the pitfalls of contemporary pharmacology. To fully realize its potential, the medical community must commit to responsible stewardship: embracing innovation while safeguarding against misuse, ensuring access while maintaining equity, and expanding treatment options while upholding the highest standards of patient care. Only through such a comprehensive and cautious approach can ketamine fulfill its promise as both a therapeutic breakthrough and a model for the future of integrative medicine.
REFERENCE
Rahul Chaudhari*, Mohan Nikam, Krushna Bharwad, Onkar Khatode, Aniruddh Thorat, Pratik Bhabad, Ketamine in Modern Medicine: Antidepressant Breakthrough, Anesthetic Staple, or Substance of Misuse, Int. J. Sci. R. Tech., 2025, 2 (5), 644-657. https://doi.org/10.5281/zenodo.15560054