OPEN Foundation

Search
Close this search box.

Ketamine

A Double-Blinded, Randomized, Placebo-Controlled Sub-Dissociative Dose Ketamine Pilot Study in the Treatment of Acute Depression and Suicidality in a Military Emergency Department Setting

Abstract

Background: Rates of completed suicide in the military have increased. Options are limited for acute relief of depression and suicidal ideation. Traditional treatments’ effects take weeks to months. A novel, rapid, therapeutic target has emerged with the N-methyl-D-aspartate antagonist ketamine. Previous studies suggest that a single dose of intravenous (IV) ketamine rapidly alleviates depression and suicidality.

Methods: In this proof of concept study, an active duty convenience sample population presenting to the emergency department (ED) meeting criteria for inpatient psychiatric admission as a result of depression and suicidal thinking were randomized to receive either a subdissociative dose (0.2 mg/kg) of IV ketamine or equivalent volume of normal saline (placebo). Subjects were evaluated for symptoms throughout a 4-hour ED course, at hospital discharge, and 2 weeks postdischarge.

Results: Methodological problems limited analyzable data to 10 subjects. Two of three who received ketamine experienced dramatic decreases in suicidality and hopelessness within 40 minutes. No such improvements were seen in any of seven controls over the 4-hour observation in the ED. At discharge from the hospital, there was no clinically significant difference. No subjects described adverse symptoms.

Conclusion: Despite methodology difficulties noted in this pilot study, there was statistical improvement in intervention group versus controls.

Burger, J., Capobianco, M., Lovern, R., Boche, B., Ross, E., Darracq, M. A., & McLay, R. (2016). A Double-Blinded, Randomized, Placebo-Controlled Sub-Dissociative Dose Ketamine Pilot Study in the Treatment of Acute Depression and Suicidality in a Military Emergency Department Setting. Military Medicine, 181(10), 1195-1199. 10.7205/MILMED-D-15-00431
Link to full text

Clinical and biological predictors of ketamine response in treatment-resistant major depression: Review

Abstract

OBJECTIVE: The aim of this review was to determine the clinical and biological predictors of the ketamine response.

METHODS: A systematic research on PubMed and PsycINFO database was performed without limits on year of publication.

RESULTS: The main predictive factors of ketamine response, which were found in different studies, were (i) a family history of alcohol dependence, (ii) unipolar depressive disorder, and (iii) neurocognitive impairments, especially a slower processing speed. Many other predictive factors were identified, but not replicated, such as personal history of alcohol dependence, no antecedent of suicide attempt, anxiety symptoms. Some biological factors were also found such as markers of neural plasticity (slow wave activity, brain-derived neurotrophic factor Val66Met polymorphism, expression of Shank 3 protein), other neurologic factors (anterior cingulate activity, concentration of glutamine/glutamate), inflammatory factors (IL-6 concentration) or metabolic factors (concentration of B12 vitamin, D- and L-serine, alterations in the mitochondrial β-oxidation of fatty acids). This review had several limits: (i) patients had exclusively resistant major depressive episodes which represent a sub-type of depression and not all depression, (ii) response criteria were more frequently assessed than remission criteria, it was therefore difficult to conclude that these predictors were similar, and finally (iii) many studies used a very small number of patients.

CONCLUSIONS: In conclusion, this review found that some predictors of ketamine response, like basal activity of anterior cingulate or vitamin B12 concentration, were identical to other therapeutics used in major depressive episode. These factors could be more specific to the major depressive episode and not to the ketamine response. Others, like family history of alcohol dependence, body mass index, or D- and L-serine were different from the other therapeutics. Neurocognitive impairments like slower speed processing or alterations in attention tests were also predictive to a good response. These predictive factors could be more specific to ketamine. With these different predictor factors (clinical and biological), it could be interesting to develop clinical strategies to personalize ketamine’s administration.

Romeo, B., Choucha, W., Fossati, P., & Rotge, J. Y. (2016). [fusion_builder_container hundred_percent=”yes” overflow=”visible”][fusion_builder_row][fusion_builder_column type=”1_1″ background_position=”left top” background_color=”” border_size=”” border_color=”” border_style=”solid” spacing=”yes” background_image=”” background_repeat=”no-repeat” padding=”” margin_top=”0px” margin_bottom=”0px” class=”” id=”” animation_type=”” animation_speed=”0.3″ animation_direction=”left” hide_on_mobile=”no” center_content=”no” min_height=”none”][Clinical and biological predictors of ketamine response in treatment-resistant major depression: Review]. L’Encephale. 10.1016/j.encep.2016.06.005
Link to full text

[/fusion_builder_column][/fusion_builder_row][/fusion_builder_container]

Ketamine Treatment and Global Brain Connectivity in Major Depression

Abstract

Capitalizing on recent advances in resting state functional connectivity magnetic resonance imaging (rs-fcMRI) and the distinctive paradigm of rapid mood normalization following ketamine treatment, the current study investigated intrinsic brain networks in major depressive disorder (MDD) during a depressive episode and following treatment with ketamine. Medication-free patients with MDD and healthy control subjects (HC) completed baseline rs-fcMRI. MDD patients received a single infusion of ketamine and underwent repeated rs-fcMRI at 24 h post-treatment. Global brain connectivity with global signal regression (GBCr) values were computed as the average of correlations of each voxel with all other gray matter voxels in the brain. MDD group showed reduced GBCr in the prefrontal cortex (PFC), but increased GBCr in the posterior cingulate, precuneus, lingual gyrus, and cerebellum. Ketamine significantly increased GBCr in the PFC and reduced GBCr in the cerebellum. At baseline, 2174 voxels of altered GBCr were identified, but only 310 voxels significantly differed relative to controls following treatment (corrected α<0.05). Responders to ketamine showed increased GBCr in the lateral PFC, caudate, and insula. Followup seed-based analyses illustrated a pattern of dysconnectivity between the PFC/subcortex and the rest of the brain in MDD, which appeared to normalize post-ketamine. The extent of the functional dysconnectivity identified in MDD and the swift and robust normalization following treatment, suggest that GBCr may serve as a treatment response biomarker for the development of rapid acting antidepressants. The data also identified unique prefrontal and striatal circuitry as putative marker of successful treatment and target for antidepressants development.

Abdallah, C. G., Averill, L. A., Collins, K. A., Geha, P., Schwartz, J., Averill, C., … & Iosifescu, D. V. (2016). Ketamine Treatment and Global Brain Connectivity in Major Depression. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 10.1038/npp.2016.186

Link to full text

Ketamine: Future Treatment For Unresponsive Depression?

Abstract

Major Depressive Disorder (MDD) is a debilitating mental health condition which accounts for a significant portion of worldwide disability. Historically, the suggested pharmacotherapy to treat MDD have been monoaminergic-acting antidepressants, such as SSRIs or SNRIs. These drugs can provide relief, but often take weeks to noticeably improve depressive symptoms and are not always effective, leading to a condition known as Treatment-Resistant Depression (TRD). It is believed that 50% MDD sufferers in Ireland suffer from TRD, and thus the development of improved pharmacotherapies is necessary. One emerging therapy is low dose, intravenous (R-S)-Ketamine (ketamine). While the molecular basis of ketamine’s therapeutic effect has not been fully determined, it has shown to effectively and swiftly mitigate the symptoms of TRD. Barriers do exist preventing the legal prescription of ketamine, including its questionable safety profile and risk of inducing dependence. Despite this, ketamine remains a promising pharmacotherapy for TRD and further investigation is required.

Frere, M., & Tepper, J. (2016). Ketamine: Future Treatment For Unresponsive Depression?. Irish Medical Journal. 10147/620895
Link to full text

Ketamine for Depression: An Update

Abstract

A decade has now passed since research into the antidepressant effects of ketamine began in earnest, after the clinical trial reported by Zarate et al. in 2006 (1). In that proof-of-concept study, 18 medication-free patients with treatment-resistant major depressive disorder (TRD) showed a large reduction in core depressive symptoms within hours of receiving a single low-dose 0.5 mg/kg intravenous infusion of ketamine as measured by the 21-item Hamilton Depression Rating Scale compared with saline placebo.

Murrough, J. W. (2016). Ketamine for Depression: An Update. Biological Psychiatry, 80(6), 416-418. http://dx.doi.org/10.1016/j.biopsych.2016.07.005
Link to full text

New use for an old drug: oral ketamine for treatment-resistant depression

Abstract

Treatment-resistant depression (TRD) is a disabling disorder that can interfere with a patient’s capacity to understand and participate in medical care and thus negatively impact individual morbidity and mortality. Hospitalised patients with TRD may require rapid alleviation of severe symptomatology, particularly when suicidal or if unable to participate in care decisions. Ketamine is well known for its anaesthetic effects and its use as a ‘street’ drug; however, its action as an N-methyl-D-aspartate receptor antagonist makes ketamine a potential therapy for TRD. The majority of studies investigating ketamine for TRD have used intravenous drug delivery, demonstrating benefit for rapid alleviation and sustained response of depression symptoms. Oral ketamine for urgent alleviation of TRD symptoms is less reported. We describe rapid alleviation of severe TRD with oral ketamine in a severely ill postoperative hospitalised patient, and review the current literature on ‘off-label’ use of ketamine for treatment of refractory depression.

Swiatek, K. M., Jordan, K., & Coffman, J. (2016). New use for an old drug: oral ketamine for treatment-resistant depression. BMJ Case Reports, 2016, bcr2016216088. 10.1136/bcr-2016-216088

Link to full text

Ketamine could be the first rapid-acting antidepressant medication

Ketamine has been used for over half a century as an anaesthetic, but interest has been steadily growing in its ability to rapidly decrease depressive symptoms. This interest has culminated in many studies attempting to elucidate its antidepressant mechanism, and in turn, these studies have contributed to our understanding of depressive disorders. Ketamine was first synthesised in 1962 as a dissociative anaesthetic, inducing a state of conscious sedation in which patients are awake, but cognitively dissociated from their pain (Young et al., 2011). In 1970, the Food and Drug Administration (FDA) approved ketamine as an anaesthetic, but throughout the 1970s ‘special K’, as it was known on the street, was gaining popularity as a recreational drug. The drug became notorious for its ability in high doses to lead users down a ‘K-hole’ – or a state of complete bodily dissociation (Muetzelfeldt et al., 2008). In 1999, the FDA scheduled the drug in the United Stated (US), banning non-medical use. While ketamine’s use and abuse was being argued over by policy makers, a team of scientists began to investigate ketamine as an antidepressant (Berman et al., 2000). Following this first human trial of ketamine as an antidepressant, the drug quickly garnered interest in the field of mental health, where since the publication of the fourth edition of the “Diagnostic and Statistical Manual of Mental Disorders” (DSM-IV) in 1994, depression was considered as a unitary concept and psychiatric disorder.

Depression and the pharmacological response

Major depressive disorder (MDD) is the most prevalent mental disorder, affecting roughly 16% of the world’s population at some point in their lives (Kessler et al., 2005). The dominant pharmacological hypothesis came about by trying to understand why certain monoaminergic-targeted medications seemed to alleviate depressive symptoms over time. The monoamine hypothesis of depression describes the disorder as the dysregulation of a group of monoaminergic neurotransmitters in the brain, specifically, the transport of dopamine, adrenaline, noradrenaline and serotonin into and out of synapses (Hirschfeld, 2000). First-line treatment of depression involves selective norepinephrine or serotonin reuptake inhibitors (SNRI/SSRI). These drugs inhibit the transport of key neurotransmitters out of the synapse between neurons. Unfortunately, less than half of those who suffer from MDD respond to monoamine-targeted medication, and for those who do, it takes at least two weeks and often longer for any symptom relief to become clinically noticeable (Kishimoto et al., 2016). Some studies have also linked SSRI treatment to an increased risk of suicide attempts and completed suicides (Fergusson et al., 2005; Healy, 2003). There is clearly a need for a more rapid and efficacious treatment of this debilitating disorder.

Ketamine as an antidepressant

Ketamine is rare in that it is a psychoactive substance that is classified in the U.S. in Schedule III under the Controlled Substances Act (Marshall, 1999) and available to be prescribed by physicians, making research accessible; however, the drug’s status as a non-patentable substance has proven to be a hurdle to funding research. Nevertheless, research into ketamine as an antidepressant has been ongoing since 2000 (Berman et al., 2000) and resulted in over 1500 studies. Studies have found that single intravenous infusions of ketamine at doses ranging from 0.1-0.5 mg/kg over 40 minutes show robust efficacy in short-term relief of MDD symptoms (Kishimoto et al., 2016). Symptoms decrease within 40-60 minutes and during the first 24 hours suicidality is also rapidly decreased. Interestingly, reduced suicidality was found to be a specific effect of ketamine, as it was also found in patients who did not respond to the antidepressant effects of the drug (Ryan et al., 2014). The promise for ketamine lies in its rapid-acting antidepressant and antisuicidal effects, as few current treatments achieve clinical significance in such a short time frame. Remission of depressive symptoms, on the other hand, only lasts between five and eight days, but can be extended to months through administration of repeated infusions (Murrough et al., 2013). This technique is somewhat controversial as the effects of repeated exposure to sub-anaesthetic doses of ketamine are yet to be known. Some patients who receive these sub-anaesthetic doses of ketamine report mild side-effects including headaches, dizziness and nausea, as well as dissociative effects and mild psychotomimetic experiences; however, these effects are transient and rarely outlast the time in which the drug is pharmacologically active (about 4 hours) (Coyle & Laws, 2015).

The glutamate theory of depression

Ketamine is a chemically promiscuous substance that interacts with many neurotransmitters in the brain, including the monoamines (Frohlich & Van Horn, 2015). Much of the research, however, has attributed ketamine’s antidepressant effects to its activation of the glutamate system. Glutamate is an excitatory neurotransmitter found in about 50% of synapses in the mammalian brain (all the monoamines together are found in only 15-20%) (Zarate & Niciu, 2015). It is the primary system by which neurons fire and communicate with one another and, as such, may be more pertinent to rapid changing of mood than monoaminergic systems.

The mechanism is complicated, but ketamine modulates glutamate by blocking, and thereby inhibiting, N-methyl-D-aspartate receptors (NMDAr) on interneurons. When active, these interneurons inhibit glutamatergic neurons in mood relevant brain areas; however, when blocked by ketamine, the disinhibition of these neurons leads to an increase in glutamatergic synaptic transmission in brain areas responsible for mood. Ultimately, the increase in glutamate activates a cascade of effects that results in neuroplasticity or neural regeneration – morphological changes to neurons in these brain areas (Kavalali & Monteggia, 2012). According to this hypothesis, the drug begins by causing chemical changes which result in non-chemical, morphological changes. It is these physical changes in the brain that could explain the persisting antidepressant effects of the drug once its pharmacological activity has ended. While NMDAr are believed to mediate these beneficial effects, recent research comparing ketamine with selective NMDAr antagonists shows that the selective NMDAr antagonists are not nearly as effective as ketamine in treating depression (Kishimoto et al., 2016; Sanacora & Schatzburg, 2015; Zanos et al., 2016). This suggests the possibility of an alternate mechanism of action.

Competing mechanisms

Excitement about ketamine as a wonder drug for depression should be tempered by a number of safety concerns about the medication. Firstly, it can induce psychedelic experiences which could be dangerous for those with a family history of psychotic disorders. Many pharmacologists, physicians and chemists have been searching for ways of achieving anti-depressant effects separate from the psychedelic experiences by adjusting doses to sub-anaesthetic levels, or exploring metabolites and stereoisomers of ketamine. Other more psychotherapeutically oriented researchers claim that the psychedelic and dissociative effects of the drug are part and parcel of its success in treating depression when combined with therapy, and that stripping the drug treatment of these mind-altering effects is akin to practicing homeopathy (Wolfson, 2014).

One such psychiatrist, Terrence Early (2014), suggests that ketamine works via the mechanism of ‘action-facilitated emotional learning.’ According to Early, patients on ketamine dissociate from their bodies, and are thus able to remember negatively charged emotional memories or trauma without the anxiety that would normally accompany these memories. Ketamine attenuates the anxiety response when trauma is revisited in therapy, and this in turn allows these memories to gradually become manageable. The age-old debate between minimising the psychedelic effects of psychedelics and embracing them for their therapeutic potential is a complex one that is present throughout the literature of psychedelic science, and involves political narratives of the war on drugs and freedom of thought.

Broadening the scope of treatment

Looking ahead to the broadened use of ketamine outside of hospital settings, addiction liability is an issue that worries some researchers (Sanacora & Schatzburg, 2015; Zhang et al., 2016), especially considering that nearly one-third of people who suffer from depression also meet criteria for substance use disorders (Davis et al., 2008). To date, trials involving ketamine almost always exclude a comorbid substance abuse disorder and this means we have very little data regarding ketamine’s addiction potential for this significant population of depressed patients. Ketamine addiction is well documented, but only at doses above 1 mg/kg (Newport et al., 2015). Current trials treating depression typically use doses of 0.5 mg/kg and never over 1.0 mg/kg. Nevertheless, the medical field has an embarrassing history of creating addictions through prescription medications like laudanum, heroin and cocaine, and we don’t have to look very far to see the current epidemic of prescription opiate addiction. Because of the drug’s short half-life, in order to achieve remission of longer than one week, repeated doses of ketamine are required, potentially increasing the likelihood of tolerance and addiction to the medication. Currently there is a paucity of research on the adverse effects of long-term repeated ketamine usage.

Ketamine is currently approved by the FDA via intravenous (IV) or intramuscular (IM) routes for large-dose anaesthesiology. This requires the presence of an anaesthesiologist and must take place in a hospital setting. This expensive, invasive and highly medicalised treatment model shows little regard for set and setting, which plays such an important role in ensuring meaningful psychedelic experiences. It has been shown that patients who received ketamine in electroconvulsive therapy rooms have worse outcomes than patients who received the medication in comfortable, relaxed settings (Ryan et al., 2014).

Alternate routes of ketamine administration have been developed and are currently being researched, including intranasal, subcutaneous, oral and sublingual (Lara et al., 2013; Mathews et al., 2012; Opler et al., 2016); however, bioavailability of ketamine is less than 50% for oral, subcutaneous and intranasal routes of administration compared to 93% for the more invasive routes (Clements et al., 1982). The antidepressant response to these alternate routes is also lower than IV or IM administration (Ryan et al., 2014). Sub-anaesthetic doses, not requiring the presence of anaesthesiologists and which can be administered in more comfortable settings, have shown antidepressant efficacy (Berman et al., 2000; Zarate et al., 2006). If off-label prescriptions are being written and administered, informed consent and integration in a therapeutic treatment are important set and setting factors for maximising the effect of the medication.

On the horizon

An exciting new article was published in Nature in May 2016, which claims to have found the key metabolite of ketamine responsible for the sustained antidepressant effects. The compound of interest was tested using animal models and found to be non-addictive and non-psychotomimetic (Zanos et al., 2016). Ketamine as it is generally administered is a racemic mixture of S-ketamine and R-ketamine (both left- and right-handed molecules in more or less equal parts). The body converts both of these enantiomers into a number of metabolites. S-ketamine and its metabolites are known to have three to four times greater affinity for NMDAr than R-forms, leading researchers to believe that S-ketamine could be used in smaller doses to achieve similarly potent effects; however, Zanos and colleagues identified (2R, 6R)-hydroxynorketamine (R-HNK) – a metabolite of ketamine with two right-handed chiral centres – as essential to the potent antidepressant effect.

Unexpectedly, R-HNK does not bind to or inhibit NMDAr, calling into question the NMDAr hypothesis of ketamine. While the target of R-HNK is not yet known, it was shown that R-HNK increases α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAr)-mediated postsynaptic potentials in the hippocampus, even after the drug’s pharmacological activity has ended. It is the upregulation of these excitatory glutamatergic AMPAr that is hypothesised to be responsible for the longer-lasting antidepressant effects of ketamine. This novel NMDAr-independent, non-addictive and non-psychotomimetic antidepressant mechanism is an exciting find (Zanos et al., 2016), but this research needs to be replicated and scaled up to human trials before any firm conclusions can be drawn as to its efficacy in treating depressive disorders.

Ketamine has emerged as a first-in-class rapid-acting antidepressant medication with a unique mechanism of action that differentiates it from the current psychiatric tools for depression. We may be on the brink of next-generation rapid-acting antidepressant medication; however, excitement about ketamine’s antidepressant benefits should be tempered by issues of safety, including adverse psychotomimetic effects, abuse potential, and costly invasive routes of administration. Before the FDA approves ketamine as a medication for MDD in broader clinical contexts, research into adverse effects of prolonged use needs to be done along with the standardisation of optimal dosing, route of administration and frequency of ketamine administration. Even in the early days of research, and with these safety concerns in mind, ketamine’s ability to rapidly decrease depressive and suicidal symptoms allows physicians to ethically treat the most severe cases of depression in emergency room contexts. At the very least, it can give clinicians time to implement alternative therapies and allow for the slower-onset, first-line treatments to reach efficacy.

References

Berman, R. M., Cappiello, A., Anand, A., Oren, D. A., Heninger, G. R., Charney, D. S., & Krystal, J. H. (2000). Antidepressant effects of ketamine in depressed patients. Society of Biological Psychiatry. 47. 351-354. doi: 10.1016/S0006-3223(99)00230-9

Clements, J., Nimmo, W., & Grant, I. (1982). Bioavailability, pharmacokinetics, and analgesic activity of ketamine in humans. Journal of Pharmaceutical Sciences. 71(5). 539-542. doi: 10.1002/jps.2600710516

Coyle, C. M., & Laws, K. R. (2015). The use of ketamine as an antidepressant: a systematic review and meta-analysis. Human Psychopharmacology: Clinical and Experimental, 30(3). 152-163. doi: 10.1002/hup.2475

Davis, L., Uezato, A., Newell, J. m., & Frazier, E. (2008). Major depression and comorbid substance use disorders. Current Opinions in Psychiatry. 21(1). 14-18. doi: 10.1097/YCO.0b013e3282f32408

Early, T. S. (2014). Making ketamine work in the long run. International Journal of Transpersonal Studies 33(2). 141-150.

Fergusson D., Doucette, S., Glass, K. C., Shapiro, S., Healy, D., Hebert, P., & Hutton, B. (2005). Association between suicide attempts and selective serotonin reuptake inhibitors: Systematic review of randomised controlled trials. British Medical Journal, 330. 396. doi: 10.1136/bmj330.7488.396

Frohlich, J. & Van Horn, J. D. (2014). Reviewing the ketamine model for schizophrenia. Journal of Psychopharmacology, 28(4). 287-302. doi: 10.1177/0269881113512909jp.sagepub.com

Healy, D. (2003). Lines of evidence on the risks of suicide with selective serotonin reuptake inhibitors. Psychotherapy and Psychosomatics. 72(2). 71-79. doi: 10.1159/000068691

Hirschfeld, R. M. (2000) History and evolution of the monoamine hypothesis of depression. The Journal of Clinical Psychiatry. 61 (suppl 6). 4-6

Kavalali, E. T., & Monteggia, L. M. (2012). Synaptic mechanisms underlying rapid antidepressant action of ketamine. Journal of American Psychiatry, 169(11). 1150-1156. doi: 10.3389/fphar.2013.00161

Kessler, R, C., Berglund, P., Demier, O., Jing, R., Merikangas, K. R., & Walters, E. E. (2005). Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the national comorbiditysurvey replication. Archives of General Psychiatry, 62(6). 593-602. doi:10.1001/archpsyc.62.6.593.

Kishimoto, T., Chawla, J. M., Hagi, K., Zarate, C. A., Kane, J. M., Bauer, M., & Correll, C. U. (2016). Single-dose infusion ketamine and non-ketamine N-methyl-D-aspartate receptor antagonists for unipolar and bipolar depression: a meta-analysis of efficacy, safety and time trajectories.  Psychological Medicine, 46. 1459-1472. doi:10.1017/S0033291716000064

Lara, D. R., Biosol, L. W., & Munari, L. R. (2013). Antidepressant, mood stabilizing and precognitive effects of very low dose sublingual ketamine in refractory unipolar and bipolar depression. The International Journey of Neuropsychopharmacology. 16(9). 2111-2117. doi: 10.1017/S1461145713000485

Marshall, D. R. (1999). Schedules of controlled substances: Placement of ketamine into schedule III. Federal Register. 64 (133). 37673-37675. docid: fr13jy99-7

Mathews, S. J., Shah, A., Lapidus, K., Clark, C., Jarun, N., Ostermeyer, B., & Murrough, J. W. (2012). Ketamine for treatment resistant unipolar depression. CNS Drugs. 26(3). 189-204. doi: 10.2165/11599770-000000000-00000

Muetzelfeldt, L., Kamboj, S. K., Rees, H., Taylor, J., Morgan, C. J.,& Curran, H. V. (2008). Journey through the K-hole: Phenomenological aspects of ketamine use. Drug and Alcohol Dependence, 95(3). 219-229. doi:10.1016/j.drugalcdep.2008.01.024

Murrough, J. W., Perez, A. M., Pillemer, S., Stern, J., Parides. M. K., aan het Rot, M., Collins, K. A., Mathew, S. J., Charney, D. S., & Iosifescu, D. V. (2013). Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression. Biological Psychiatry, 74(4). 250-256. doi: 10.1016/j.biopsych.2012.06.022

Newport, D. J., Carpenter, L. L., McDonald, W. M., Potash, J. B., Tohen, M., Nemeroff, C. B., The APA Council of Research Task Force on Novel Biomarkers and Treatments. (2015). Ketamine and other NMDA antagonists: Early clinical trials and possible mechanisms of depression. The American Journal of Psychiatry. 172(10). 950 -966. doi: 10.1176/appi.ajp.2015.15040465

Opler, L.A., Opler, M. G. A., & Arnsten, A. F. T. (2016). Ameliorating treatment-refractory depression with intranasal ketamine: potential NMDA receptor actions in the pain circuitry representing mental anguish. CNS Spectrums. 21(1). 12-22. doi: 10.1017/S1092852914000686

Ryan, W. C., Marta, C, J. & Koek, R. J. (2014). Ketamine and depression: A review. International Journal of Transpersonal Studies. 33(2). 40-74.

Sanacora, G., & Schatzberg, A. F. (2015) Ketamine: Promising path or false prophecy in the development of novel therapeutics for mood disorders? Neuropsychopharmacology, 40. 259-267. doi:10.1038/npp.2014.261.

Wolfson, P. E. (2014). Ketamine-Its history, uses, pharmacology, therapeutic practice, and exploration of its potential as a novel treatment for depression. International Journal of Transpersonal Studies. 33(2). 33-39.

Young, M., Kolp, E. & Friedman, H. (2011). Ketamine. In M.A. Kleiman & J. E. Hawdon (Eds.), Encyclopedia of drug policy (2). 451. doi:10.4135/9781412976961.n194

Zanos, P., Moaddel, R., Morris, P. J., Georgiou, P., Fischell, J., Elmer, G. I., Alkondon, M., Yuan, P., Pribut, H. J., Singh, N. S., Dossou, K. S., Fang, Y., Huang, X., Mayo, C. L., Wainer, I. W., Albuquerque, E. X., Thompson, S. M., Thomas, C. J., Zarate Jr. C. A., & Gould, T. D. (2016). NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 533(7604). 481-486. doi: 10.1038/nature17998.

Zarate, C. A., Singh, J. B., Carlson, P. J., Brutsche, N. E., Ameli, R., Luckenbaugh, D. A., Charney, D. S., & Manji, H. K. (2006). A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression. Archives of General Psychiatry. 63(8). 856-864. doi: 10.1001/archpsyc.63.8.856

Zarate, C. A. & Niciu, M. (2015). Ketamine for depression: Evidence, challenges and promise. World Psychiatry, 14(3). 348-350. doi:10.1002/wps.20269.

Zhang, M. W., Harris, K. M., & Ho, R. C. (2016). Is off-label repeat prescription of ketamine as a rapid antidepressant safe? Controversies, ethical concerns, and legal implications. BMC Medical Ethics. 17(4). 1-8. doi: 10.1186/s12910-016-0087-3

Neurovascular Plasticity of the Hippocampus One Week after a Single Dose of Ketamine in Genetic Rat Model of Depression

Abstract

Glutamatergic system and the structural plasticity hypothesis are principal components for rapid and sustained antidepressant effects of novel antidepressant therapeutics. This study represents the first investigation of the structural plasticity of the hippocampus as one of the main contributed mechanisms to the sustained anti-depressive effect of ketamine. Flinders Sensitive Line (FSL) and Flinders Resistant Line (FRL) rats were given a single intraperitoneal injection of ketamine (15 mg/kg) or saline 7 days before perfusion-fixed. The optical fractionator method was used to estimate the total number of neurons in the granular cell layer. Microvessel length in the molecular layer of DG was evaluated with global spatial sampling method. By use of the physical disector method, the number of synapses was estimated. The volume of the hippocampus was larger in the FRL-vehicle rats compared with FSL-vehicle group and in FSL-ketamine versus FSL-vehicle rats (P < 0.05). The number of non-perforated synapses was significantly higher in the FSL-ketamine versus FSL-vehicle group, (P = 0.01). A significant effect of ketamine on enhancement of the number of neurons in DG in FSL rats was observed (P = 0.01). The total length of the microvessels 1 week after ketamine treatment in the FSL rats significantly increased (P  < 0.05). Our results indicate that neurovascular changes of hippocampus could be one of the possible mechanisms underlying the sustained antidepressant effect of ketamine by reversing alteration of the number of the excitatory synapses, neuronal number and length of the microvessels in the hippocampus.

Ardalan, M., Wegener, G., Polsinelli, B., Madsen, T. M., & Nyengaard, J. R. (2016). Neurovascular Plasticity of the Hippocampus One Week after a Single Dose of Ketamine in Genetic Rat Model of Depression. Hippocampus. http://dx.doi.org/10.1002/hipo.22617
Link to full text

Efficacy and safety of ketamine in bipolar depression: A systematic review

Abstract

The depression is the most prevalent state throughout the life of the bipolar patient. Ketamine has been shown to be an effective and rapid treatment for depression. The objective of the present work is to perform a systematic review on the efficacy and safety of ketamine as treatment of bipolar depression, as well as its different patterns of administration. The search found 10 relevant manuscripts that met the inclusion criteria: one clinical trial, 5 cohort studies, and 4 case reports. Intravenous infusion was used in 60% of the studies. According to data, ketamine seems to be an effective and safe treatment for bipolar depression, although the length of its effect is short. Adverse effects observed generally occurred at the time of infusion, and tended to completely disappear within 1-2h. Therefore, more studies are necessary to explore new patterns of administration, as well as on its safety and adverse effects.

Alberich, S., Martínez-Cengotitabengoa, M., López, P., Zorrilla, I., Núñez, N., Vieta, E., & González-Pinto, A. (2016). Efficacy and safety of ketamine in bipolar depression: A systematic review. Revista de psiquiatria y salud mental. 10.1016/j.rpsm.2016.05.005

Link to full text

Online Event - Psychedelic Care in Recreational Settings - 3 October 2024

X

interested in becoming a trained psychedelic-assisted therapist?