1
|
Jobnah S, Latifeh Y, Al Kabani D, Youssef LA. Ketamine and chronic treatment-resistant depression: real-world practice and after relapse. BMC Psychiatry 2024; 24:745. [PMID: 39468512 PMCID: PMC11520900 DOI: 10.1186/s12888-024-06203-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Chronic treatment-resistant depression (TRD) poses a major challenge for clinicians. Ketamine has shown a rapid but short-lived antidepressant effect in several studies involving TRD patients with different demographic and clinical profiles. Our study aimed to assess the antidepressant effect of serial infusion sessions of ketamine in patients with chronic TRD and evaluate the severity of symptoms after relapse and the general psychiatric health of the responding patients. METHODS In this single arm open-label study, six infusions of ketamine at 0.5 mg/kg were administered to chronic TRD patients for approximately two weeks. Response and remission rates, side effects, adverse events and after-relapse symptoms were evaluated, and patients were followed for three months. RESULTS 23 patients underwent at least one infusion session, and 18 patients completed the six sessions. Twelve (66.67%) patients responded to the treatment at some point, and 11 (61.11%) patients maintained response after the end of the treatment protocol. One infusion was not sufficient to achieve a response (P > 0.9999, z = 1.81), and more than half of the responders met the response criteria after the third infusion. Only one patient (5.56%) achieved remission at the end of the infusion phase. All but one ketamine responders relapsed within one month after the end of the treatment. There was no statistical difference between baseline and after-relapse MADRS scores (P = 0.7886, 95% CI=-5.512-4.312, R2 = 0,008411). However, a high incidence of serious adverse events related to suicidality was evident; one of the non-responding patients attempted suicide and several attempts to sedate this patient with benzodiazepines failed. Two responding patients ended up with a suicidal attempt or severe suicidal thoughts. CONCLUSIONS Introducing rapid-acting antidepressant to manage TRD patients in clinical practice demands further investigation, and the benefit-to-harm ratio should be assessed in the light of the increased risk of suicidality.
Collapse
Affiliation(s)
- Sumaya Jobnah
- Program of Clinical and Hospital Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Damascus University, Damascus, Syrian Arab Republic
| | - Youssef Latifeh
- Department of Internal Medicine, Division of Psychiatry, Al-Mouwasat University Hospital, Damascus University, Damascus, Syrian Arab Republic
| | - Dina Al Kabani
- Department of Internal Medicine, Division of Psychiatry, Al-Mouwasat University Hospital, Damascus University, Damascus, Syrian Arab Republic
| | - Lama A Youssef
- Program of Clinical and Hospital Pharmacy, Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Damascus University, Damascus, Syrian Arab Republic.
| |
Collapse
|
2
|
De Jager JE, Boesjes R, Roelandt GHJ, Koliaki I, Sommer IEC, Schoevers RA, Nuninga JO. Shared effects of electroconvulsive shocks and ketamine on neuroplasticity: A systematic review of animal models of depression. Neurosci Biobehav Rev 2024; 164:105796. [PMID: 38981574 DOI: 10.1016/j.neubiorev.2024.105796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
Electroconvulsive shocks (ECS) and ketamine are antidepressant treatments with a relatively fast onset of therapeutic effects compared to conventional medication and psychotherapy. While the exact neurobiological mechanisms underlying the antidepressant response of ECS and ketamine are unknown, both interventions are associated with neuroplasticity. Restoration of neuroplasticity may be a shared mechanism underlying the antidepressant efficacy of these interventions. In this systematic review, literature of animal models of depression is summarized to examine the possible role of neuroplasticity in ECS and ketamine on a molecular, neuronal, synaptic and functional level, and specifically to what extent these mechanisms are shared between both interventions. The results highlight that hippocampal neurogenesis and brain-derived neurotrophic factor (BDNF) levels are consistently increased after ECS and ketamine. Moreover, both interventions positively affect glutamatergic neurotransmission, astrocyte and neuronal morphology, synaptic density, vasculature and functional plasticity. However, a small number of studies investigated these processes after ECS. Understanding the shared fundamental mechanisms of fast-acting antidepressants can contribute to the development of novel therapeutic approaches for patients with severe depression.
Collapse
Affiliation(s)
- Jesca E De Jager
- Department of Biomedical Sciences, Brain Center, University Medical Center, Groningen, the Netherlands.
| | - Rutger Boesjes
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Gijs H J Roelandt
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Ilektra Koliaki
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Iris E C Sommer
- Department of Biomedical Sciences, Brain Center, University Medical Center, Groningen, the Netherlands
| | - Robert A Schoevers
- University Centre of Psychiatry, University Medical Center Groningen, the Netherlands
| | - Jasper O Nuninga
- Department of Biomedical Sciences, Brain Center, University Medical Center, Groningen, the Netherlands; University Medical Centre Utrecht, Department of Psychiatry, the Netherlands
| |
Collapse
|
3
|
Smith-Apeldoorn SY, Veraart JKE, Kamphuis J, Spijker J, van der Meij A, van Asselt ADI, Aan Het Rot M, Schoevers RA. Oral esketamine in patients with treatment-resistant depression: a double-blind, randomized, placebo-controlled trial with open-label extension. Mol Psychiatry 2024; 29:2657-2665. [PMID: 38523183 DOI: 10.1038/s41380-024-02478-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/23/2024] [Accepted: 02/05/2024] [Indexed: 03/26/2024]
Abstract
About one-third of patients with depression do not achieve adequate response to current treatment options. Although intravenous and intranasal administrations of (es)ketamine have shown antidepressant properties, their accessibility and scalability are limited. We investigated the efficacy, safety, and tolerability of generic oral esketamine in patients with treatment-resistant depression (TRD) in a randomized placebo-controlled trial with open-label extension. This study consisted of 1) a six-week fixed low-dose treatment phase during which 111 participants received oral esketamine 30 mg or placebo three times a day; 2) a four-week wash-out phase; and 3) an optional six-week open-label individually titrated treatment phase during which participants received 0.5 to 3.0 mg/kg oral esketamine two times a week. The primary outcome measure was change in depressive symptom severity, assessed with the Hamilton Depression Rating Scale (HDRS17), from baseline to 6 weeks. Fixed low-dose oral esketamine when compared to placebo had no benefit on the HDRS17 total score (p = 0.626). Except for dizziness and sleep hallucinations scores, which were higher in the esketamine arm, we found no significant difference in safety and tolerability aspects. During the open-label individually titrated treatment phase, the mean HDRS17 score decreased from 21.0 (SD 5.09) to 15.1 (SD 7.27) (mean difference -6.0, 95% CI -7.71 to -4.29, p < 0.001). Our results suggest that fixed low-dose esketamine is not effective in TRD. In contrast, individually titrated higher doses of oral esketamine might have antidepressant properties.
Collapse
Affiliation(s)
- Sanne Y Smith-Apeldoorn
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Jolien K E Veraart
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Psychiatry, PsyQ Haaglanden, Parnassia Psychiatric Institute, The Hague, The Netherlands
| | - Jeanine Kamphuis
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Spijker
- Depression Expertise Center, Pro Persona Mental Health Care, Nijmegen, The Netherlands
- Behavioral Science Institute, Radboud University, Nijmegen, The Netherlands
| | | | - Antoinette D I van Asselt
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Health Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marije Aan Het Rot
- Department of Psychology, University of Groningen, Groningen, The Netherlands
- School of Behavioral and Cognitive Neurosciences, University of Groningen, Groningen, The Netherlands
| | - Robert A Schoevers
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- School of Behavioral and Cognitive Neurosciences, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Ohtani Y, Tani H, Nomoto-Takahashi K, Yatomi T, Yonezawa K, Tomiyama S, Nagai N, Kusudo K, Honda S, Moriyama S, Nakajima S, Yamada T, Morisaki H, Iwabuchi Y, Jinzaki M, Yoshimura K, Eiro T, Tsugawa S, Ichijo S, Fujimoto Y, Miyazaki T, Takahashi T, Uchida H. Efficacy and safety of intravenous ketamine treatment in Japanese patients with treatment-resistant depression: A double-blind, randomized, placebo-controlled trial. Psychiatry Clin Neurosci 2024. [PMID: 39210712 DOI: 10.1111/pcn.13734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/17/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024]
Abstract
AIM Although the antidepressant effect of ketamine on treatment-resistant depression (TRD) has been frequently reported in North American and European countries, evidence is scarce among the Asian population. We aimed to evaluate the efficacy and safety of intravenous ketamine in Japanese patients with TRD. METHODS In this double-blind randomized placebo-controlled trial, 34 Japanese patients with TRD were randomized to receive either intravenous ketamine (0.5 mg/kg) or placebo, administered over 40 min, twice a week, for 2 weeks. The primary outcome was the change in the Montgomery Åsberg Depression Rating Scale (MADRS) total score from baseline to post-treatment. Secondary outcomes included changes in other depressive symptomatology scores and remission, response, and partial response rates. We also examined the association between baseline clinical demographic characteristics and changes in the MADRS total score. RESULTS Intention-to-treat analysis indicated no significant difference in the decrease in MADRS total score between the groups (-8.1 ± 10.0 vs -2.5 ± 5.2, t[32] = 2.02, P = 0.052), whereas per-protocol analysis showed a significant reduction in the ketamine group compared to the placebo group (-9.1 ± 10.2 vs -2.7 ± 5.3, t[29] = 2.22, P = 0.034). No significant group differences were observed in other outcomes. Adverse events were more frequent in the ketamine group than in the placebo group, and no serious adverse events were reported. A higher baseline MADRS total score and body mass index were associated with a greater reduction in the MADRS total score. CONCLUSION Intravenous ketamine outperformed placebo in Japanese patients with TRD who completed the study, suggesting that ketamine could alleviate depressive symptoms of TRD across diverse ethnic populations.
Collapse
Affiliation(s)
- Yohei Ohtani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Tani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | | | - Taisuke Yatomi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kengo Yonezawa
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Sota Tomiyama
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhiro Nagai
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
- Department of Psychiatry, Minami-Hanno Hospital, Saitama, Japan
| | - Keisuke Kusudo
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shiori Honda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Sotaro Moriyama
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Takashige Yamada
- Department of Anesthesiology, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Morisaki
- Department of Anesthesiology, Keio University School of Medicine, Tokyo, Japan
| | - Yu Iwabuchi
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Kimio Yoshimura
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Eiro
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sakiko Tsugawa
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sadamitsu Ichijo
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yu Fujimoto
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoyuki Miyazaki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takuya Takahashi
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Pan Y, Gorenflo MP, Davis PB, Kaelber DC, De Luca S, Xu R. Suicidal ideation following ketamine prescription in patients with recurrent major depressive disorder: a nation-wide cohort study. Transl Psychiatry 2024; 14:327. [PMID: 39122686 PMCID: PMC11315997 DOI: 10.1038/s41398-024-03033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/14/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
Ketamine has gained attention for its effective treatment for patients with major depressive disorder (MDD) and suicidal ideation; Despite numerous studies presenting the rapid efficacy, long-term benefit in real-world populations remains poorly characterized. This is a retrospective cohort study using TriNetX US Collaborative Network, a platform aggregating electronic health records (EHRs) data from 108 million patients from 62 health care organizations in the US, and the study population includes 514,988 patients with a diagnosis of recurrent MDD who were prescribed relevant treatment in their EHRs. The prescription of ketamine was associated with significantly decreased risk of suicidal ideation compared to the prescription of other common antidepressants: HR = 0.63 (95% CI: 0.53-0.76) at 1 day - 7 days, 0.67 (95% CI: 0.59-0.77) at 1 day - 30 days, 0.69 (95% CI: 0.62-0.77) at 1 day - 90 days, 0.74 (95% CI: 0.67-0.81) at 1 day - 180 days, and 0.78 (95% CI: 0.69-0.83) at 1 day - 270 days. This trend was especially robust among adults over 24 years of age, females, males, and White patients with recurrent MDD. This study provides real-world evidence that ketamine has long-term benefits in mitigating suicidal ideation in patients with recurrent MDD. Future work should focus on optimizing dosage regimens for ketamine, understanding the mechanism, and the difference in various demographic subpopulations.
Collapse
Affiliation(s)
- Yiheng Pan
- Center for Artificial Intelligence in Drug Discovery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Computer and Data Science, Case Western Reserve University, Cleveland, OH, USA
| | - Maria P Gorenflo
- Center for Artificial Intelligence in Drug Discovery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Pamela B Davis
- Center for Community Health Integration, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - David C Kaelber
- Center for Clinical Informatics Research and Education, The MetroHealth System, Cleveland, OH, USA
| | - Susan De Luca
- Population Health Research Institute, The MetroHealth System, Cleveland, OH, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
6
|
Liu Y, Yang J, Liu Y. Ketamine and electroconvulsive therapy for severe depression: A network meta-analysis of efficacy and safety. J Psychiatr Res 2024; 175:218-226. [PMID: 38744161 DOI: 10.1016/j.jpsychires.2024.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/21/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Ketamine, electroconvulsive therapy (ECT), and their combination are effective for treating severe depression, but few large-scale studies have compared these. METHODS We searched databases for randomized controlled trials (RCTs) using ketamine, ECT, ketamine + ECT, or placebo for severe depression. Standardized measures were efficacy outcomes. Risk of bias was assessed. Stata and ADDIS were used for network meta-analysis (NMA) comparing efficacy and adverse reactions post-treatment. This study was registered on PROSPERO (CRD42023476740). RESULTS 17 RCTs with 1370 patients were included. NMA showed ECT and ketamine improved Hamilton Depression Rating Scale (HDRS) versus placebo; other comparisons not significant. Rank probabilities showed highest probability for ECT, followed by ketamine + ECT, ketamine, placebo. No differences in Montgomery-Asberg Depression Rating Scale (MADRS); highest rank probability again for ECT, followed by ketamine + ECT, ketamine, placebo. CONCLUSIONS Analysis suggests ECT superior to ketamine and their combination for improving depressive severity, but individualized treatment selection warranted. Higher adverse reactions with ketamine + ECT need further study for optimized combined use.
Collapse
Affiliation(s)
- Yecun Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiguo Yang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yuanxiang Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
7
|
Aykan D, Genc M, Unal G. Environmental enrichment enhances the antidepressant effect of ketamine and ameliorates spatial memory deficits in adult rats. Pharmacol Biochem Behav 2024; 240:173790. [PMID: 38761992 DOI: 10.1016/j.pbb.2024.173790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Ketamine is a rapid-acting antidepressant associated with various cognitive side effects. To mitigate these side effects while enhancing efficacy, it can be co-administered with other antidepressants. In our study, we adopted a similar strategy by combining ketamine with environmental enrichment, a potent sensory-motor paradigm, in adult male Wistar rats. We divided the animals into four groups based on a combination of housing conditions and ketamine versus vehicle injections. The groups included those housed in standard cages or an enriched environment for 50 days, which encompassed a 13-day-long behavioral testing period. Each group received either two doses of ketamine (20 mg/kg, IP) or saline as a vehicle. We tested the animals in the novel object recognition test (NORT), forced swim test (FST), open field test (OFT), elevated plus maze (EPM), and Morris water maze (MWM), which was followed by ex vivo c-Fos immunohistochemistry. We observed that combining environmental enrichment with ketamine led to a synergistic antidepressant effect. Environmental enrichment also ameliorated the spatial memory deficits caused by ketamine in the MWM. There was enhanced neuronal activity in the habenula of the enrichment only group following the probe trial of the MWM. In contrast, no differential activity was observed in enriched animals that received ketamine injections. The present study showed how environmental enrichment can enhance the antidepressant properties of ketamine while reducing some of its side effects, highlighting the potential of combining pharmacological and sensory-motor manipulations in the treatment of mood disorders.
Collapse
Affiliation(s)
- Deren Aykan
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342 Istanbul, Turkey
| | - Mert Genc
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342 Istanbul, Turkey
| | - Gunes Unal
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342 Istanbul, Turkey.
| |
Collapse
|
8
|
Shen Z, Gao D, Lv X, Wang H, Yue W. A meta-analysis of the effects of ketamine on suicidal ideation in depression patients. Transl Psychiatry 2024; 14:248. [PMID: 38858391 PMCID: PMC11164699 DOI: 10.1038/s41398-024-02973-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/23/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024] Open
Abstract
The treatment of suicidal ideation in patients with depression has been a major problem faced by psychiatric and emergency departments, and reasonable drug selection is particularly important. Ketamine has been shown to reduce suicidal ideation rapidly, but the strength of the effect is unclear and there is little evidence-based medical evidence to support this. We systematically searched all articles published on PubMed, Cochrane Library, Web of Science, CNKI and EMBASE. Stata 15 and R 4.1.3 were used for meta-analysis, and odds ratios were calculated in fixed effects or random effects models based on the heterogeneity test results. Our search resulted in 505 articles; we analyzed 14 studies, which included 1,380 participants. The 14 studies included 10 randomized controlled trial (RCT) studies and 4 single-arm studies. Our study suggests that, ketamine has a significant therapeutic effect on suicidal ideation throughout the treatment cycle. We performed network meta-analyses(NMA) and pairwise meta-analyses to compare the efficacy of ketamine in the reduction of suicidal ideation. There was a significant reduction in suicidal ideation within the first day after treatment (NMA ketamine day1 RR = 10.02, 95%CI = 4.24 to 23.68). In repeated treatment, the degree of recovery of suicidal ideation after the last dose was significantly greater than that after the first dose (RR = 0.56, 95%CI = 0.51 to 0.62). Recovery of suicidal ideation was also significantly better in the treatment end point than in the placebo group at the same time point (NMA ketamine day26 RR = 4.29, 95%CI = 1.41 to 13.08). This is the first network meta-analysis to demonstrate the role of ketamine in the alleviation of suicidal ideation. Our network meta-analysis also compared the effects of different drugs at different time points, which was not done in previous studies. This is of great reference significance for future drug research andrational drug use.
Collapse
Affiliation(s)
- ZuoYao Shen
- The First Affiliated Hospital of Xinxiang Medical College Henan, Henan, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No.2018RU006), Beijing, China
| | - DaiQuan Gao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xue Lv
- The First Affiliated Hospital of Xinxiang Medical College Henan, Henan, China
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No.2018RU006), Beijing, China
| | - HongXing Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - WeiHua Yue
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China.
- NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
- Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences (No.2018RU006), Beijing, China.
| |
Collapse
|
9
|
Rizk MM, Murrough JW. Ketamine, Dissociation, and Depression: What Is "Special" About Ketamine? (Revisited). Int J Neuropsychopharmacol 2024; 27:pyae024. [PMID: 38864154 PMCID: PMC11193139 DOI: 10.1093/ijnp/pyae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/11/2024] [Indexed: 06/13/2024] Open
Affiliation(s)
- Mina M Rizk
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - James W Murrough
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
10
|
Beaglehole B, Porter R, Douglas K, Lacey CJ, de Bie A, Jordan J, Mentzel C, Thwaites B, Manuel J, Murray G, Frampton C, Glue P. Protocol for a randomised controlled trial of ketamine versus ketamine and behavioural activation therapy for adults with treatment-resistant depression in the community. BMJ Open 2024; 14:e084844. [PMID: 38692731 PMCID: PMC11086269 DOI: 10.1136/bmjopen-2024-084844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/03/2024] [Indexed: 05/03/2024] Open
Abstract
INTRODUCTION Although short-term benefits follow parenteral ketamine for treatment-resistant major depressive disorder (TR-MDD), there are challenges that prevent routine use of ketamine by clinicians. These include acute dissociative effects of parenteral ketamine, high relapse rates following ketamine dosing and the uncertain role of psychotherapy. This randomised controlled trial (RCT) seeks to establish the feasibility of evaluating repeated oral doses of ketamine and behavioural activation therapy (BAT), compared with ketamine treatment alone, for TR-MDD. We also aim to compare relapse rates between treatment arms to determine the effect size of adding BAT to oral ketamine. METHODS AND ANALYSIS This is a prospectively registered, two-centre, single-blind RCT. We aim to recruit 60 participants with TR-MDD aged between 18 and 65 years. Participants will be randomised to 8 weeks of oral ketamine and BAT, or 8 weeks of oral ketamine alone. Feasibility will be assessed by tracking attendance for ketamine and BAT, acceptability of treatment measures and retention to the study follow-up protocol. The primary efficacy outcome measure is the Montgomery-Asberg Depression Rating Scale (MADRS) measured weekly during treatment and fortnightly during 12 weeks of follow-up. Other outcome measures will assess the tolerability of ketamine and BAT, cognition and activity (using actigraphy). Participants will be categorised as non-responders, responders, remitters and relapsed during follow-up. MADRS scores will be analysed using a linear mixed model. For a definitive follow-up RCT study to be recommended, the recruitment expectations will be met and efficacy outcomes consistent with a >20% reduction in relapse rates favouring the BAT and ketamine arm will be achieved. ETHICS AND DISSEMINATION Ethics approval was granted by the New Zealand Central Health and Disability Ethics Committee (reference: 2023 FULL18176). Study findings will be reported to participants, stakeholder groups, conferences and peer-reviewed publications. TRIAL REGISTRATION NUMBER UTN: U1111-1294-9310, ACTRN12623000817640p.
Collapse
Affiliation(s)
- Ben Beaglehole
- Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Richard Porter
- Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Katie Douglas
- Psychological Medicine, University of Otago, Christchurch, New Zealand
| | | | - Aroha de Bie
- Te Whatu Ora-Health New Zealand Waitaha Canterbury, Christchurch, Canterbury, New Zealand
| | - Jennifer Jordan
- Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Charlie Mentzel
- Department of Psychological Medicine, University of Otago, Dunedin, New Zealand
| | | | - Jenni Manuel
- Psychological Medicine, University of Otago, Christchurch, New Zealand
| | - Greg Murray
- Centre for Mental Health, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | | | - Paul Glue
- Psychological Medicine, University of Otago, Dunedin School of Medicine, Dunedin, New Zealand
| |
Collapse
|
11
|
Liu H, Wang C, Lan X, Li W, Zhang F, Hu Z, Ye Y, Ning Y, Zhou Y. Functional connectivity of the amygdala subregions and the antidepressant effects of repeated ketamine infusions in major depressive disorder. Eur Psychiatry 2024; 67:e33. [PMID: 38572583 PMCID: PMC11059247 DOI: 10.1192/j.eurpsy.2024.1744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Amygdala subregion-based network dysfunction has been determined to be centrally implicated in major depressive disorder (MDD). Little is known about whether ketamine modulates amygdala subarea-related networks. We aimed to investigate the relationships between changes in the resting-state functional connectivity (RSFC) of amygdala subregions and ketamine treatment and to identify important neuroimaging predictors of treatment outcomes. METHODS Thirty-nine MDD patients received six doses of ketamine (0.5 mg/kg). Depressive symptoms were assessed, and magnetic resonance imaging (MRI) scans were performed before and after treatment. Forty-five healthy controls underwent one MRI scan. Seed-to-voxel RSFC analyses were performed on the amygdala subregions, including the centromedial amygdala (CMA), laterobasal amygdala (LBA), and superficial amygdala subregions. RESULTS Abnormal RSFC between the left LBA and the left precuneus in MDD patients is related to the therapeutic efficacy of ketamine. There were significant differences in changes in bilateral CMA RSFC with the left orbital part superior frontal gyrus and in changes in the left LBA with the right middle frontal gyrus between responders and nonresponders following ketamine treatment. Moreover, there was a difference in the RSFC of left LBA and the right superior temporal gyrus/middle temporal gyrus (STG/MTG) between responders and nonresponders at baseline, which could predict the antidepressant effect of ketamine on Day 13. CONCLUSIONS The mechanism by which ketamine improves depressive symptoms may be related to its regulation of RSFC in the amygdala subregion. The RSFC between the left LBA and right STG/MTG may predict the response to the antidepressant effect of ketamine.
Collapse
Affiliation(s)
- Haiyan Liu
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Chengyu Wang
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiaofeng Lan
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Weicheng Li
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- Department of Psychology, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Fan Zhang
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- Department of Psychology, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhibo Hu
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yanxiang Ye
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yuping Ning
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
- Department of Psychology, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yanling Zhou
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| |
Collapse
|
12
|
Brown KA, Gould TD. Targeting metaplasticity mechanisms to promote sustained antidepressant actions. Mol Psychiatry 2024; 29:1114-1127. [PMID: 38177353 PMCID: PMC11176041 DOI: 10.1038/s41380-023-02397-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The discovery that subanesthetic doses of (R, S)-ketamine (ketamine) and (S)-ketamine (esketamine) rapidly induce antidepressant effects and promote sustained actions following drug clearance in depressed patients who are treatment-resistant to other therapies has resulted in a paradigm shift in the conceptualization of how rapidly and effectively depression can be treated. Consequently, the mechanism(s) that next generation antidepressants may engage to improve pathophysiology and resultant symptomology are being reconceptualized. Impaired excitatory glutamatergic synapses in mood-regulating circuits are likely a substantial contributor to the pathophysiology of depression. Metaplasticity is the process of regulating future capacity for plasticity by priming neurons with a stimulation that alters later neuronal plasticity responses. Accordingly, the development of treatment modalities that specifically modulate the duration, direction, or magnitude of glutamatergic synaptic plasticity events such as long-term potentiation (LTP), defined here as metaplastogens, may be an effective approach to reverse the pathophysiology underlying depression and improve depression symptoms. We review evidence that the initiating mechanisms of pharmacologically diverse rapid-acting antidepressants (i.e., ketamine mimetics) converge on consistent downstream molecular mediators that facilitate the expression/maintenance of increased synaptic strength and resultant persisting antidepressant effects. Specifically, while the initiating mechanisms of these therapies may differ (e.g., cell type-specificity, N-methyl-D-aspartate receptor (NMDAR) subtype-selective inhibition vs activation, metabotropic glutamate receptor 2/3 antagonism, AMPA receptor potentiation, 5-HT receptor-activating psychedelics, etc.), the sustained therapeutic mechanisms of putative rapid-acting antidepressants will be mediated, in part, by metaplastic effects that converge on consistent molecular mediators to enhance excitatory neurotransmission and altered capacity for synaptic plasticity. We conclude that the convergence of these therapeutic mechanisms provides the opportunity for metaplasticity processes to be harnessed as a druggable plasticity mechanism by next-generation therapeutics. Further, targeting metaplastic mechanisms presents therapeutic advantages including decreased dosing frequency and associated diminished adverse responses by eliminating the requirement for the drug to be continuously present.
Collapse
Affiliation(s)
- Kyle A Brown
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA.
| |
Collapse
|
13
|
Carles A, Freyssin A, Perin-Dureau F, Rubinstenn G, Maurice T. Targeting N-Methyl-d-Aspartate Receptors in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:3733. [PMID: 38612544 PMCID: PMC11011887 DOI: 10.3390/ijms25073733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
N-methyl-d-aspartate receptors (NMDARs) are the main class of ionotropic receptors for the excitatory neurotransmitter glutamate. They play a crucial role in the permeability of Ca2+ ions and excitatory neurotransmission in the brain. Being heteromeric receptors, they are composed of several subunits, including two obligatory GluN1 subunits (eight splice variants) and regulatory GluN2 (GluN2A~D) or GluN3 (GluN3A~B) subunits. Widely distributed in the brain, they regulate other neurotransmission systems and are therefore involved in essential functions such as synaptic transmission, learning and memory, plasticity, and excitotoxicity. The present review will detail the structure, composition, and localization of NMDARs, their role and regulation at the glutamatergic synapse, and their impact on cognitive processes and in neurodegenerative diseases (Alzheimer's, Huntington's, and Parkinson's disease). The pharmacology of different NMDAR antagonists and their therapeutic potentialities will be presented. In particular, a focus will be given on fluoroethylnormemantine (FENM), an investigational drug with very promising development as a neuroprotective agent in Alzheimer's disease, in complement to its reported efficacy as a tomography radiotracer for NMDARs and an anxiolytic drug in post-traumatic stress disorder.
Collapse
Affiliation(s)
- Allison Carles
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France; (A.C.); (A.F.)
| | - Aline Freyssin
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France; (A.C.); (A.F.)
- ReST Therapeutics, 34095 Montpellier, France; (F.P.-D.); (G.R.)
| | | | | | - Tangui Maurice
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France; (A.C.); (A.F.)
| |
Collapse
|
14
|
Parikh SV, Vande Voort JL, Yocum AK, Achtyes E, Goes FS, Nykamp L, Singh B, Lopez-Vives D, Sera CE, Maixner D, Tarnal V, Severe J, Bartek S, Tye SJ, Rico J, Stoppel CJ, Becerra A, Smart L, Miller CR, Frye MA, Greden JF, Bobo WV. Clinical outcomes in the biomarkers of ketamine (Bio-K) study of open-label IV ketamine for refractory depression. J Affect Disord 2024; 348:143-151. [PMID: 38142892 DOI: 10.1016/j.jad.2023.12.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 11/28/2023] [Accepted: 12/13/2023] [Indexed: 12/26/2023]
Abstract
OBJECTIVE We conducted an open-label clinical trial ("Bio-K") using IV ketamine for treatment-resistant depression to identify biomarkers linked to remission. Here, we report the clinical efficacy and side effect outcomes of Bio-K. METHODS Across 4 US sites, 75 patients ages 18-65 with treatment-refractory unipolar or bipolar depression received 3 IV ketamine infusions over an 11-day period. Key exclusion criteria were psychotic symptoms, significant substance abuse, unstable medical conditions, and any use of cannabis. Pre-existing antidepressant medication was maintained. Primary outcome was remission as measured by Montgomery-Asberg Depression Rating Scale (MADRS), with secondary outcome of 50 % reduction in Beck Suicide Scale score. Safety monitoring and varying durations of infusions were also key parameters. RESULTS Using remission as MADRS score <10, after 3 infusions 52 % achieved remission, with 67 % achieving response. Of those achieving response after a single infusion, 66 % (22 of 33) reached remission after 3 infusions, while 40 % (16 of 40) non-responders after the first infusion went on to achieve remission after 3 infusions. Only 20 % of non-responders after 2 infusions achieved remission. Most (81 %) participants had significant suicidal ideation at baseline; of these, two-thirds (67 %) experienced at least a 50 % reduction in suicidality. Side effects were minimal. Uniquely, we had three different types of infusion categories, with individuals receiving: (1) slow (100-min) infusions only or (2) regular (40-min) infusions only or (3) a mix of infusion durations. These three infusion groups showed comparable safety and efficacy. Exploration of clinical factors revealed no link between BMI, age, or gender to remission. CONCLUSIONS The consistency of outcomes across 4 clinical sites and across multiple instruments, suggests high acute efficacy and safety of IV ketamine for serious depressive episodes. Duration of infusion did not alter outcomes. Meaningfully, 40 % of non-responders after a single infusion did reach remission subsequently, while only 20 % of non-responders after 2 infusions achieved remission, suggesting early response is suggestive for eventual remission. Our data on varying ketamine infusion duration adds novel insights into the clinical administration of this new treatment for refractory and severe patients. Our limitations included a lack of a control group, necessitating caution about conclusions of efficacy, balanced by the utility of reporting "real-world" outcomes across multiple clinical sites. We could also not separately analyze results for bipolar disorder due to small numbers. Together, the Bio-K clinical results are promising and provide significant sample sizes for forthcoming biological markers analyses.
Collapse
Affiliation(s)
| | | | | | - Eric Achtyes
- Pine Rest Christian Mental Health Services, Michigan State University, USA; Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | | | - Louis Nykamp
- Pine Rest Christian Mental Health Services, Michigan State University, USA
| | - Balwinder Singh
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | - Susannah J Tye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Jose Rico
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Cynthia J Stoppel
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | - LeAnn Smart
- Pine Rest Christian Mental Health Services, Michigan State University, USA
| | | | - Mark A Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | - William V Bobo
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
15
|
Liu Y, Hu Q, Xu S, Li W, Liu J, Han L, Mao H, Cai F, Liu Q, Zhu R, Fang C, Lou Y, Wang Z, Yang H, Wang W. Antidepressant effects of dexmedetomidine compared with ECT in patients with treatment-resistant depression. J Affect Disord 2024; 347:437-444. [PMID: 38000472 DOI: 10.1016/j.jad.2023.11.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/16/2023] [Accepted: 11/19/2023] [Indexed: 11/26/2023]
Abstract
OBJECTIVE This pilot study was designed to investigate the antidepressant effects of dexmedetomidine (DEX), a selective α2-adrenergic receptor agonist, in patients with treatment-resistant depression (TRD). The antidepressant effects of dexmedetomidine was compared with ECT, which is widely used in clinical practice for treatment of patients with TRD. METHODS Seventy six patients with TRD were randomly assigned to receive 10 sessions of DEX infusions or electroconvulsive therapy (ECT) treatment. The primary outcome was the changes of depression severity determined by the improvement of 24-item Hamilton Depression Rating Scale (HDRS-24). The second outcomes were the rates of therapeutic response (reduction in HDRS-24 ≥ 50 %) and remission (HDRS-24 ≤ 10 and reduction in HDRS-24 ≥ 60 %) at posttreatment and after 3 months of follow-up visits. RESULTS We found that 10 sessions of DEX infusions or ECT treatments significantly improved HDRS-24 scores at posttreatment and after 3 months of follow-up visits compared with the baseline. In addition, there was no significant difference between DEX infusions and ECT treatments regarding HDRS-24 at these evaluating points. Furthermore, the depression severity dropped to mild after 2 sessions of DEX infusion. In contrast, at least 6 sessions of ECT treatment were needed to achieve a same level. Finally, the rates of therapeutic response and remission were comparable between the two groups. No serious adverse events were observed. CONCLUSIONS Based on current published evidence, we conclude that DEX exhibits rapid and durable antidepressant properties similar to ECT but with fewer side effects.
Collapse
Affiliation(s)
- Yusi Liu
- Jinzhou Medical University, Jinzhou, Liaoning 121001, China
| | - Qiyun Hu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Sen Xu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Wanwen Li
- Department of Psychiatry, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310063, China
| | - Junyun Liu
- Department of Psychiatry, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310063, China
| | - Liang Han
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Hui Mao
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Fang Cai
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Qiaoyan Liu
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Renlai Zhu
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Caiyun Fang
- Department of Anesthesiology, Affiliated Mental Health Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310063, China
| | - Yifei Lou
- Department of Anesthesiology, Affiliated Hangzhou Xixi Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310023, China
| | - Zhenhua Wang
- Department of Anesthesiology, Jiaxing Hospital of T.C.M., Affiliated Hospital of Zhejiang Chinese Medical University, Jiaxing, Zhejiang 314015, China
| | - Huiling Yang
- Department of Anesthesiology, Affiliated Hangzhou Xixi Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310023, China
| | - Wenyuan Wang
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
16
|
Cherian KN, Keynan JN, Anker L, Faerman A, Brown RE, Shamma A, Keynan O, Coetzee JP, Batail JM, Phillips A, Bassano NJ, Sahlem GL, Inzunza J, Millar T, Dickinson J, Rolle CE, Keller J, Adamson M, Kratter IH, Williams NR. Magnesium-ibogaine therapy in veterans with traumatic brain injuries. Nat Med 2024; 30:373-381. [PMID: 38182784 PMCID: PMC10878970 DOI: 10.1038/s41591-023-02705-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/10/2023] [Indexed: 01/07/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of disability. Sequelae can include functional impairments and psychiatric syndromes such as post-traumatic stress disorder (PTSD), depression and anxiety. Special Operations Forces (SOF) veterans (SOVs) may be at an elevated risk for these complications, leading some to seek underexplored treatment alternatives such as the oneirogen ibogaine, a plant-derived compound known to interact with multiple neurotransmitter systems that has been studied primarily as a treatment for substance use disorders. Ibogaine has been associated with instances of fatal cardiac arrhythmia, but coadministration of magnesium may mitigate this concern. In the present study, we report a prospective observational study of the Magnesium-Ibogaine: the Stanford Traumatic Injury to the CNS protocol (MISTIC), provided together with complementary treatment modalities, in 30 male SOVs with predominantly mild TBI. We assessed changes in the World Health Organization Disability Assessment Schedule from baseline to immediately (primary outcome) and 1 month (secondary outcome) after treatment. Additional secondary outcomes included changes in PTSD (Clinician-Administered PTSD Scale for DSM-5), depression (Montgomery-Åsberg Depression Rating Scale) and anxiety (Hamilton Anxiety Rating Scale). MISTIC resulted in significant improvements in functioning both immediately (Pcorrected < 0.001, Cohen's d = 0.74) and 1 month (Pcorrected < 0.001, d = 2.20) after treatment and in PTSD (Pcorrected < 0.001, d = 2.54), depression (Pcorrected < 0.001, d = 2.80) and anxiety (Pcorrected < 0.001, d = 2.13) at 1 month after treatment. There were no unexpected or serious adverse events. Controlled clinical trials to assess safety and efficacy are needed to validate these initial open-label findings. ClinicalTrials.gov registration: NCT04313712 .
Collapse
Affiliation(s)
- Kirsten N Cherian
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Jackob N Keynan
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Lauren Anker
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Afik Faerman
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | | | - Ahmed Shamma
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Or Keynan
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - John P Coetzee
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
- Polytrauma Division, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Jean-Marie Batail
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Angela Phillips
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Nicholas J Bassano
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Gregory L Sahlem
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Jose Inzunza
- Ambio Life Sciences, Vancouver, British Columbia, Canada
| | - Trevor Millar
- Ambio Life Sciences, Vancouver, British Columbia, Canada
| | | | - C E Rolle
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Jennifer Keller
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Maheen Adamson
- WRIISC-WOMEN & Department of Rehabilitation, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Neurosurgery, Stanford School of Medicine, Stanford, CA, USA
| | - Ian H Kratter
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA
| | - Nolan R Williams
- Brain Stimulation Lab, Department of Psychiatry & Behavioral Sciences, Stanford School of Medicine, Stanford, CA, USA.
- Department of Radiology, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
17
|
Elmeseiny OSA, Müller HK. A molecular perspective on mGluR5 regulation in the antidepressant effect of ketamine. Pharmacol Res 2024; 200:107081. [PMID: 38278430 DOI: 10.1016/j.phrs.2024.107081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
Ketamine, a non-competitive N-methyl-D-aspartate receptor (NMDAR) antagonist, has received much attention for its rapid antidepressant effects. A single administration of ketamine elicits rapid and sustained antidepressant effects in both humans and animals. Current efforts are focused on uncovering molecular mechanisms responsible for ketamine's antidepressant activity. Ketamine primarily acts via the glutamatergic pathway, and increasing evidence suggests that ketamine induces synaptic and structural plasticity through increased translation and release of neurotrophic factors, activation of mammalian target of rapamycin (mTOR), and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR)-mediated synaptic potentiation. However, the initial events triggering activation of intracellular signaling cascades and the mechanisms responsible for the sustained antidepressant effects of ketamine remain poorly understood. Over the last few years, it has become apparent that in addition to the fast actions of the ligand-gated AMPARs and NMDARs, metabotropic glutamate receptors (mGluRs), and particularly mGluR5, may also play a role in the antidepressant action of ketamine. Although research on mGluR5 in relation to the beneficial actions of ketamine is still in its infancy, a careful evaluation of the existing literature can identify converging trends and provide new interpretations. Here, we review the current literature on mGluR5 regulation in response to ketamine from a molecular perspective and propose a possible mechanism linking NMDAR inhibition to mGluR5 modulation.
Collapse
Affiliation(s)
- Ola Sobhy A Elmeseiny
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Heidi Kaastrup Müller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
18
|
Abstract
Major depressive disorder (MDD) is a leading cause of suicide in the world. Monoamine-based antidepressant drugs are a primary line of treatment for this mental disorder, although the delayed response and incomplete efficacy in some patients highlight the need for improved therapeutic approaches. Over the past two decades, ketamine has shown rapid onset with sustained (up to several days) antidepressant effects in patients whose MDD has not responded to conventional antidepressant drugs. Recent preclinical studies have started to elucidate the underlying mechanisms of ketamine's antidepressant properties. Herein, we describe and compare recent clinical and preclinical findings to provide a broad perspective of the relevant mechanisms for the antidepressant action of ketamine.
Collapse
Affiliation(s)
- Ji-Woon Kim
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Institute of Regulatory Innovation through Science, Kyung Hee University, Seoul, Republic of Korea
| | - Kanzo Suzuki
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Ege T Kavalali
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
| | - Lisa M Monteggia
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
| |
Collapse
|
19
|
Hovda N, Gerrish W, Frizzell W, Shackelford R. A systematic review of the incidence of medical serious adverse events in sub-anesthetic ketamine treatment of psychiatric disorders. J Affect Disord 2024; 345:262-271. [PMID: 37875227 DOI: 10.1016/j.jad.2023.10.120] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/04/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND Limited published data exists that collates serious adverse outcomes involving ketamine as a psychiatric intervention. This systematic review assesses the reported incidence of medical serious adverse events (MSAEs), including but not limited to cardiovascular events, in patients receiving sub-anesthetic doses of ketamine for psychiatric disorders to guide practitioners during treatment planning, risk-benefit analyses, and the informed consent process. METHODS Pubmed database was searched for clinical trials of sub-anesthetic ketamine for psychiatric disorders in non-pregnant adult patients. Of the 2275 articles identified, 93 met inclusion criteria, over half of which were published in 2017 or later. Only studies that reported adverse events were included, and the incidence of MSAEs was calculated. RESULTS Of the 3756 participants who received at least one sub-anesthetic dose of ketamine, four participants experienced a MSAE, resulting in an incidence of approximately 0.1 % of individuals. The four MSAEs resolved without reported sequelae. Eighty-three percent of studies reported screening for medical illness and exclusion of high-risk patients. There were no serious cardiac adverse events or deaths observed in any participants; however, most trials' study designs excluded those with high cardiovascular complication risk. LIMITATIONS Most studies were small, underpowered for detecting rare MSAEs, at potential high-risk of bias of non-report of MSAEs, and limited mostly to intranasal and intravenous routes. CONCLUSIONS Findings suggest that with basic medical screening there is a very low incidence of MSAEs including adverse cardiac or cerebrovascular events in individuals receiving sub-anesthetic ketamine for psychiatric disorders.
Collapse
Affiliation(s)
- Nicholas Hovda
- Sojourn Psychotherapy, Boise, United States of America; University of Washington School of Medicine, Department of Psychiatry & Behavioral Sciences, United States of America; Boise VAMC, Psychiatry & Behavioral Sciences Department, United States of America.
| | - Winslow Gerrish
- University of Washington School of Medicine, Department of Psychiatry & Behavioral Sciences, United States of America; Family Medicine Residency of Idaho - Boise, Full Circle Health, United States of America.
| | - William Frizzell
- University of Washington School of Medicine, Department of Psychiatry & Behavioral Sciences, United States of America; Boise VAMC, Psychiatry & Behavioral Sciences Department, United States of America.
| | - Ryan Shackelford
- Sojourn Psychotherapy, Boise, United States of America; University of Washington School of Medicine, Department of Psychiatry & Behavioral Sciences, United States of America; Family Medicine Residency of Idaho - Boise, Full Circle Health, United States of America.
| |
Collapse
|
20
|
Aicher HD, Mueller MJ, Dornbierer DA, Suay D, Elsner C, Wicki I, Meling D, Caflisch L, Hempe A, Steinhart C, Mueller J, Von Rotz R, Kleim B, Scheidegger M. Potential therapeutic effects of an ayahuasca-inspired N,N-DMT and harmine formulation: a controlled trial in healthy subjects. Front Psychiatry 2024; 14:1302559. [PMID: 38264636 PMCID: PMC10804806 DOI: 10.3389/fpsyt.2023.1302559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024] Open
Abstract
Background There is growing scientific evidence for the therapeutic benefits of the Amazonian plant-based psychedelic "ayahuasca" for neuropsychiatric disorders such as depression and anxiety. However, there are certain challenges when incorporating botanical ayahuasca into biomedical research and clinical therapy environments. Formulations inspired by ayahuasca, which contain specific and standardized active components, are a potential remedy. Methods We investigated subjective acute and persisting effects of a novel formulation containing the reversible monoamine oxidase inhibitor harmine (orodispersible tablet containing 100 mg MAO-I) and N,N-dimethyltryptamine (incremental intranasal dosing of up to 100 mg DMT), compared with two other conditions, namely harmine alone and placebo, in a crossover RCT in 31 healthy male subjects. Results DMT + harmine, but not harmine alone, induced a psychedelic experience assessed with the 5D-ASC rating scale [global score: F(2,60) = 80.21, p < 0.001] and acute experience sampling items over time, characterized by psychological insights [PIQ, F(2,58.5) = 28.514, p < 0.001], emotional breakthroughs [EBI, F(2,60) = 26.509, p < 0.001], and low scores on the challenging experience questionnaire [CEQ, F(2,60) = 12.84, p < 0.001]. Participants attributed personal and spiritual significance to the experience (GSR) with mainly positive persisting effects (PEQ) at 1- and 4-months follow-up. Acute drug effects correlated positively with persisting effects. We found no changes in trait measures of personality, psychological flexibility, or general well-being, and no increases in psychopathology (SCL-90-R) were reported. Discussion and Conclusion Our results suggest that the experience induced by the standardized DMT + harmine formulation induces a phenomenologically rich psychedelic experience, demonstrates good psychological safety and tolerability, is well tolerated, and induces beneficial psychological processes that could possibly support psychotherapy. Further studies are required to investigate the psychotherapeutic potential in patients.
Collapse
Affiliation(s)
- Helena D. Aicher
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Psychology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Switzerland
| | - Michael J. Mueller
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Switzerland
- Department of Health Science and Technology, ETH, Zurich, Switzerland
| | - Dario A. Dornbierer
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Department of Forensic Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Dila Suay
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- MoMiLab, IMT School for Advanced Studies Luca, Lucca, Italy
| | - Claudius Elsner
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ilhui Wicki
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Daniel Meling
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Psychosomatic Medicine and Psychotherapy, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Luzia Caflisch
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alexandra Hempe
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Biopsychology, Department of Psychology, TUD Dresden University of Technology, Dresden, Germany
| | - Camilla Steinhart
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jovin Mueller
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Robin Von Rotz
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Birgit Kleim
- Department of Psychology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Switzerland
- Experimental Psychopathology and Psychotherapy, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Milan Scheidegger
- Psychedelic Research and Therapy Development, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH, Zurich, Switzerland
| |
Collapse
|
21
|
Schatzberg AF, Mathew SJ. The why, when, where, how, and so what of so-called rapidly acting antidepressants. Neuropsychopharmacology 2024; 49:189-196. [PMID: 37460770 PMCID: PMC10700639 DOI: 10.1038/s41386-023-01647-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/22/2023] [Accepted: 06/27/2023] [Indexed: 12/08/2023]
Abstract
Developing antidepressants that are not only more effective but are rapidly acting is the Holy Grail for psychiatry. We review multiple issues that arise in determining rapid responses in antidepressant trials. The current status of purportedly rapid acting agents is first reviewed. Then, a number of key questions/issues are addressed: Is there a unifying definition for rapid response across studies? Should rapid response criteria be based on required measurable effects on overall improvement? On specific symptoms such as psychomotor retardation, depressed mood, or anhedonia? In associated symptoms such as anxiety or insomnia? When should onset be considered rapid-by Day 3? Day7? Day 14? If there is a rapid response, for how long should the effects be maintained? Is maintenance of effect dependent on continuing the medication? Is rapid response associated with specific mechanisms of action? Do the mechanisms of action suggest possible risk for drug abuse? How important is rapid response really in an often chronic or recurrent depressive disorder? In which types of patients could rapid response be particularly important? What are the study design issues that need to be considered for assessing rapid response, including: selection of specific types of depressed patients, multiple doses of drug studied, designation of primary and secondary outcome measures, specific time points at which to determine efficacy, requirements for demonstrating durability, etc. A framework for approaching this complex area is developed for both researchers and clinicians.
Collapse
Affiliation(s)
- Alan F Schatzberg
- Kenneth T. Norris, Jr., Professor of Psychiatry and Behavioral Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - Sanjay J Mathew
- Marjorie Bintliff Johnson and Raleigh White Johnson, Jr. Chair for Research in Psychiatry, Menninger Department of Psychiatry and Behavioral Science, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
22
|
Garcia-Carachure I, Lira O, Themann A, Rodriguez M, Flores-Ramirez FJ, Lobo MK, Iñiguez SD. Sex-Specific Alterations in Spatial Memory and Hippocampal AKT-mTOR Signaling in Adult Mice Pre-exposed to Ketamine and/or Psychological Stress During Adolescence. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:240-251. [PMID: 38298791 PMCID: PMC10829642 DOI: 10.1016/j.bpsgos.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/24/2023] [Accepted: 07/29/2023] [Indexed: 02/02/2024] Open
Abstract
Background Ketamine (KET) is administered to manage major depression in adolescent patients. However, the long-term effects of juvenile KET exposure on memory-related tasks have not been thoroughly assessed. We examined whether exposure to KET, psychological stress, or both results in long-lasting alterations in spatial memory in C57BL/6 mice. Furthermore, we evaluated how KET and/or psychological stress history influenced hippocampal protein kinase B-mechanistic target of rapamycin (AKT-mTOR)-related signaling. Methods On postnatal day 35, male and female mice underwent vicarious defeat stress (VDS), a form of psychological stress that reduces sociability in both sexes, with or without KET exposure (20 mg/kg/day, postnatal days 35-44). In adulthood (postnatal day 70), mice were assessed for spatial memory performance on a water maze task or euthanized for hippocampal tissue collection. Results Juvenile pre-exposure to KET or VDS individually increased the latency (seconds) to locate the escape platform in adult male, but not female, mice. However, juvenile history of concomitant KET and VDS prevented memory impairment. Furthermore, individual KET or VDS pre-exposure, unlike their combined history, decreased hippocampal AKT-mTOR signaling in adult male mice. Conversely, KET pre-exposure alone increased AKT-mTOR in the hippocampus of adult female mice. Lastly, rapamycin-induced decreases of mTOR in naïve adult female mice induced spatial memory retrieval deficits, mimicking adult male mice with a history of exposure to VDS or KET. Conclusions Our preclinical model shows how KET treatment for the management of adolescent psychological stress-induced sequelae does not impair spatial memory later in life. However, juvenile recreational KET misuse, like psychological stress history, results in long-term spatial memory deficits and hippocampal AKT-mTOR signaling changes in a sex-specific manner.
Collapse
Affiliation(s)
| | - Omar Lira
- Department of Psychology, The University of Texas at El Paso, El Paso, Texas
| | - Anapaula Themann
- Department of Psychology, The University of Texas at El Paso, El Paso, Texas
| | - Minerva Rodriguez
- Department of Psychology, The University of Texas at El Paso, El Paso, Texas
| | | | - Mary Kay Lobo
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sergio D. Iñiguez
- Department of Psychology, The University of Texas at El Paso, El Paso, Texas
| |
Collapse
|
23
|
Shim I. Distinct functions of S-ketamine and R-ketamine in mediating biobehavioral processes of drug dependency: comments on Bonaventura et al. Mol Psychiatry 2023; 28:4939-4940. [PMID: 35595979 DOI: 10.1038/s41380-022-01629-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/06/2022] [Accepted: 05/12/2022] [Indexed: 01/28/2023]
Affiliation(s)
- Insop Shim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
24
|
Jelovac A, McCaffrey C, Terao M, Shanahan E, Mohamed E, Whooley E, McDonagh K, McDonogh S, Igoe A, Loughran O, Shackleton E, O'Neill C, McLoughlin DM. Study protocol for Ketamine as an adjunctive therapy for major depression (2): a randomised controlled trial (KARMA-Dep [2]). BMC Psychiatry 2023; 23:850. [PMID: 37974160 PMCID: PMC10655414 DOI: 10.1186/s12888-023-05365-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Depression is a common psychiatric disorder and a leading cause of disability worldwide. Conventional monoaminergic antidepressants have limited efficacy and take weeks to exert a therapeutic effect. Single infusions of subanaesthetic doses of ketamine exhibit rapid antidepressant action but effects are transient and relapse is common. One potential strategy for increasing ketamine's antidepressant efficacy and/or prolonging its therapeutic benefit may be serial infusions. There is limited evidence on the efficacy and safety of repeated ketamine infusions against an active comparator. METHODS This protocol describes an ongoing pragmatic, randomised, controlled, parallel-group, patient- and rater-blind, superiority trial. Eligible adult inpatients with a confirmed DSM-5 diagnosis of a major depressive episode (unipolar or bipolar) are randomly allocated in a 1:1 ratio to a course of up to eight infusions of ketamine or midazolam twice-weekly over four weeks. The primary objective is to assess the efficacy of serial adjunctive ketamine infusions versus active comparator midazolam by measuring Montgomery-Åsberg Depression Rating Scale score difference between arms from before the first infusion to 24 h after the final infusion, supplemented by a 95% confidence interval. To facilitate generalisability of results, the trial takes place under "real world" conditions with both groups continuing to receive regular inpatient care including treatment-as-usual pharmacotherapy, nursing care, and psychological and other therapies during the randomised treatment phase and regular outpatient care thereafter. Participants are monitored for relapse during a 24-week follow-up after the end of the randomised phase. Secondary objectives of the trial are to assess: response and remission rates at the end of randomised phase; relapse status during the 24-week follow-up after the end of the randomised phase; the safety and tolerability of repeated ketamine infusions regarding psychotomimetic and other psychiatric side effects, cognitive side effects, as well as withdrawal symptoms, haemodynamic stability, neurological, urological, and other physical side effects; and quality of life and cost-effectiveness. DISCUSSION There is an unmet clinical need for rapidly-acting novel antidepressants. This trial will provide efficacy, safety and health economic data on serial ketamine infusions and thus help inform clinical practice on the potential role of this treatment in the management of depression. TRIAL REGISTRATION EudraCT 2019-003109-92. Registered 2 October 2019. CLINICALTRIALS gov NCT04939649. Registered 25 June 2021.
Collapse
Affiliation(s)
- Ana Jelovac
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Cathal McCaffrey
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Masashi Terao
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Enda Shanahan
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Enas Mohamed
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Emma Whooley
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Kelly McDonagh
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Sarah McDonogh
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Anna Igoe
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Orlaith Loughran
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Ellie Shackleton
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland
| | - Ciaran O'Neill
- Centre for Public Health, Global Research Institute for Health Sciences, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Declan M McLoughlin
- Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, D08 K7YW, Ireland.
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
25
|
Dębowska W, Więdłocha M, Dębowska M, Kownacka Z, Marcinowicz P, Szulc A. Transcranial magnetic stimulation and ketamine: implications for combined treatment in depression. Front Neurosci 2023; 17:1267647. [PMID: 37954877 PMCID: PMC10637948 DOI: 10.3389/fnins.2023.1267647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Drug-resistant mental disorders, particularly treatment-resistant depression, pose a significant medical and social problem. To address this challenge, modern psychiatry is constantly exploring the use of novel treatment methods, including biological treatments, such as transcranial magnetic stimulation (TMS), and novel rapid-acting antidepressants, such as ketamine. While both TMS and ketamine demonstrate high effectiveness in reducing the severity of depressive symptoms, some patients still do not achieve the desired improvement. Recent literature suggests that combining these two methods may yield even stronger and longer-lasting results. This review aims to consolidate knowledge in this area and elucidate the potential mechanisms of action underlying the increased efficacy of combined treatment, which would provide a foundation for the development and optimization of future treatment protocols.
Collapse
Affiliation(s)
- Weronika Dębowska
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Więdłocha
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
- KeyClinic, Warsaw, Poland
| | - Marta Dębowska
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Zuzanna Kownacka
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Marcinowicz
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
- KeyClinic, Warsaw, Poland
| | - Agata Szulc
- Department of Psychiatry, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
- MindHealth, Warsaw, Poland
| |
Collapse
|
26
|
Yang SQ, Zhou YY, Yang ST, Mao XY, Chen L, Bai ZH, Ping AQ, Xu SY, Li QW, Gao K, Wang SY, Duan KM. Effects of different doses of esketamine intervention on postpartum depressive symptoms in cesarean section women: A randomized, double-blind, controlled clinical study. J Affect Disord 2023; 339:333-341. [PMID: 37442447 DOI: 10.1016/j.jad.2023.07.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 06/02/2023] [Accepted: 07/08/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND The optimal dosage and method of esketamine for postpartum depressive symptoms (PDS) are unclear. We conducted a randomized controlled trial (RCT) to investigate the effect of different doses of esketamine on PDS in women undergoing cesarean section, with evidence of prenatal depression. METHODS The three groups were high- (2 mg kg-1) and low-dose (1 mg kg-1) esketamine via patient controlled intravenous analgesia (PCIA), following an initial intravenous infusion of 0.25 mg kg-1 esketamine, compared to placebo (0.9 % saline infusion). All groups also received the sufentanil (2.2 μg kg-1). The primary outcome was the incidence of PDS at 7 and 42 days postpartum. The secondary outcomes were: the remission from depression and total EPDS scores at 7 days and 42 days postpartum; mean change from baseline in the EPDS score; postoperative analgesia. RESULTS i). 0.25 mg kg-1 of esketamine intravenous infusion combined with 1 mg kg-1 (n = 99) or 2 mg kg-1 (n = 99) esketamine PCIA reduces PDS incidence at 7 days postpartum (p < 0.05), with high-dose esketamine PCIA also reduces PDS incidence 42 days postpartum (p < 0.05), compared to placebo (n = 97). ii). Low- and high-dose esketamine PCIA lowers NRS scores at rest within 48 h postoperatively (p < 0.01), with high-dose esketamine also reducing the NRS score during movement at 48 h postoperatively (p = 0.018). iii). Neither high- nor low-dose esketamine PCIA increased postoperative adverse reactions (p > 0.05). CONCLUSIONS Esketamine (0.25 mg kg-1) intravenous infusion combined with 1 mg kg-1 or 2 mg kg-1 esketamine PCIA seems safe and with few adverse effects in the management of PDS and pain in women undergoing cesarean section. LIMITATIONS The tolerability and safety of esketamine requires further investigation based on more specific scales; the transient side effects of esketamine could have biased the staff and patients. TRIAL REGISTRATION ChiCTR-ROC-2000039069.
Collapse
Affiliation(s)
- Si Qi Yang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Ying Yong Zhou
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Shu Ting Yang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiao Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Liang Chen
- Department of Anesthesiology, The Maternal and Child Health Hospital of the Hu Nan Province, Changsha, China
| | - Zhi Hong Bai
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - An Qi Ping
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Shou Yu Xu
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Qiu Wen Li
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Kai Gao
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Sai Ying Wang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China.
| | - Kai Ming Duan
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
27
|
Beaglehole B, Glue P, Clarke M, Porter R. Multidisciplinary development of guidelines for ketamine treatment for treatment-resistant major depression disorder for use by adult specialist mental health services in New Zealand. BJPsych Open 2023; 9:e191. [PMID: 37828915 PMCID: PMC10594164 DOI: 10.1192/bjo.2023.577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/09/2023] [Accepted: 09/02/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND The evidence base for racemic ketamine treatment for treatment-resistant major depressive disorder (TRD) continues to expand, but there are major challenges translating this evidence base into routine clinical care. AIM To prepare guidelines for ketamine treatment of TRD that are suitable for routine use by publicly funded specialist mental health services. METHOD We consulted with senior leadership, clinical pharmacy, psychiatrists, nursing, service users and Māori mental health workers on issues relating to ketamine treatment. We prepared treatment guidelines taking the evidence base for ketamine treatment and the consultation into account. RESULTS Ketamine treatment guidance is reported. This offers two treatment pathways, including a test of ketamine responsiveness with intramuscular ketamine and the dominant use of oral ketamine for a 3-month course to maximise the opportunity for the short-term benefits of ketamine to accumulate. CONCLUSIONS We have responded to the challenges of translating the evidence base for ketamine treatment into a form suitable for routine care.
Collapse
Affiliation(s)
- Ben Beaglehole
- Department of Psychological Medicine, University of Otago, New Zealand
| | - Paul Glue
- Department of Psychological Medicine, University of Otago, New Zealand
| | - Mike Clarke
- Specialist Mental Health Services, Te Whatu Ora – Health New Zealand Waitaha Canterbury, New Zealand
| | - Richard Porter
- Department of Psychological Medicine, University of Otago, New Zealand
| |
Collapse
|
28
|
Fancy F, Rodrigues NB, Di Vincenzo JD, Chau EH, Sethi R, Husain MI, Gill H, Tabassum A, Mckenzie A, Phan L, McIntyre RS, Rosenblat JD. Real-World Effectiveness of Repeated Ketamine Infusions for Treatment-Resistant Bipolar Depression. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2023; 21:420-429. [PMID: 38694999 PMCID: PMC11058957 DOI: 10.1176/appi.focus.23021022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Background Clinical trials have demonstrated rapid antidepressant effects with intravenous (IV) ketamine for major depressive disorder, with relatively less research specifically for bipolar depression. Herein, we describe the real-world effectiveness of repeated ketamine infusions for treatment-resistant bipolar depression. Methods This study was conducted in a community clinic in Mississauga, Ontario (Canadian Rapid Treatment Centre of Excellence; Braxia Health). In this observational study (NCT04209296), patients with treatment-resistant bipolar I/II depression (n = 66) received four sub-anesthetic doses of IV ketamine (0.5-0.75 mg/kg) over a two-week period. Symptoms of depression, suicidality, anxiety, and functioning were assessed with validated self-report measures. Results Statistically and clinically significant antidepressant effects were observed in the overall sample, as measured by the Quick Inventory for Depression Symptomatology-Self Report-16 (QIDS-SR16) with further reductions in depressive symptoms observed after each subsequent infusion (n = 66; mean QIDS-SR16 reduction of 6.08+/-1.39; p < 0.0001). Significant reductions of suicidal thoughts (QIDS-SR16-Suicide Item) and anxiety (Generalized Anxiety Disorder-7) were also observed with functional improvements on the Sheehan Disability Scale (p < 0.0001 on all measures). Moreover, the response rate (QIDS-SR16 total score decrease ≥50% from baseline) was 35% and remission rate (QIDS-SR16 total score ≤5) was 20% after four infusions. Infusions were generally well tolerated with treatment-emergent hypomania observed in only three patients (4.5%) with zero cases of mania or psychosis. Conclusions Real-world effectiveness of IV ketamine for bipolar depression was observed. Repeated doses were associated with greater symptom reduction and adequate tolerability.Reprinted from Bipolar Disord 2023; 25:99-109, with permission from John Wiley and Sons. Copyright © 2023.
Collapse
Affiliation(s)
- Farhan Fancy
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Nelson B Rodrigues
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Joshua D Di Vincenzo
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Edmond H Chau
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Rickinder Sethi
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Muhammad I Husain
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Hartej Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Aniqa Tabassum
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Andrea Mckenzie
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Lee Phan
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada (Fancy, Rodrigues, Di Vicenzo, Sethi, Gill, Tabassum, Mckenzie, Phan, McIntyre, Rosenblat); Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada (Fancy, Gill, McIntyre, Rosenblat); Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada (Chau, McIntyre, Rosenblat); Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada (Sethi, Husain, McIntyre, Rosenblat); Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada (Husain); Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (McIntyre, Rosenblat); Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada (McIntyre)
| |
Collapse
|
29
|
Fancy F, Haikazian S, Johnson DE, Chen-Li DCJ, Levinta A, Husain MI, Mansur RB, Rosenblat JD. Ketamine for bipolar depression: an updated systematic review. Ther Adv Psychopharmacol 2023; 13:20451253231202723. [PMID: 37771417 PMCID: PMC10524067 DOI: 10.1177/20451253231202723] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/31/2023] [Indexed: 09/30/2023] Open
Abstract
Background The therapeutic potential of subanesthetic doses of ketamine appears promising in unipolar depression; however, its effectiveness in treating bipolar depression (BD) remains uncertain. Objective This systematic review aimed to summarize findings on the use of ketamine for the treatment of BD by assessing its efficacy, safety, and tolerability. Design Systematic review. Methods We conducted a systematic review of studies that investigated the use of ketamine for adults with BD. We searched PubMed and Embase for relevant randomized-controlled trials, open-label trials, and retrospective chart analyses published from inception to 13 March 2023. Results Eight studies were identified [pooled n = 235; mean (SD) age: 45.55 (5.54)]. All participants who received intravenous (IV) ketamine were administered a dose of 0.5-0.75 mg/kg as an adjunctive treatment to a mood-stabilizing agent, whereas participants who received esketamine were administered a dosage ranging from 28 to 84 mg. Flexible dosing was used in real-world analyses. A total of 48% of participants receiving ketamine achieved a response (defined as ⩾50% reduction in baseline depression severity), whereas only 5% achieved a response with a placebo. Real-world studies demonstrated lower rates of response (30%) compared to the average across clinical trials (63%). Reductions in suicidal ideation were noted in some studies, although not all findings were statistically significant. Ketamine and esketamine were well tolerated in most participants; however, six participants (2% of the overall sample pool, 5 receiving ketamine) developed hypomanic/manic symptoms after infusions. Significant dissociative symptoms were observed at the 40-min mark in some trials. Conclusion Preliminary evidence suggests IV ketamine as being safe and effective for the treatment of BD. Future studies should focus on investigating the effects of repeated acute and maintenance infusions using a randomized study design.
Collapse
Affiliation(s)
- Farhan Fancy
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Sipan Haikazian
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Danica E. Johnson
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - David C. J. Chen-Li
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Anastasia Levinta
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Muhammad I. Husain
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Rodrigo B. Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Joshua D. Rosenblat
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, Toronto, ON M5T 2S8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, ON, Canada
| |
Collapse
|
30
|
Hartelius G, Muscat SA, Bartova L. Editorial: Bridging the gap: an interdisciplinary perspective on ketamine in psychiatric disorders. Front Psychiatry 2023; 14:1246891. [PMID: 37645640 PMCID: PMC10461637 DOI: 10.3389/fpsyt.2023.1246891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023] Open
Affiliation(s)
| | | | - Lucie Bartova
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
31
|
Zheng W, Zhou YL, Wang CY, Lan XF, Ning YP. A comparative analysis of antidepressant and anti-suicidal effects of repeated ketamine infusions in elderly and younger adults with depression. J Affect Disord 2023; 334:145-151. [PMID: 37160235 DOI: 10.1016/j.jad.2023.04.120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/02/2023] [Accepted: 04/29/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVES This study aims to investigate the differences in safety and antidepressant effects of multi-infusion ketamine treatment between elderly and young adults with depression. METHODS The safety, antidepressant, and anti-suicidal effects of multi-infusion ketamine were compared between 19 elderly (≥50 years) and 116 younger (<50 years) adults with depression; all were treated with six ketamine infusions (0.5 mg/kg). Montgomery-Åsberg Depression Rating Scale (MADRS) was used to measure the depressive symptoms, and suicidal ideation was measured with Beck Scale for Suicide Ideation (SSI)-part 1, Hamilton Rating Scale for Depression (HAMD) item 3, and (MADRS) item 10. Dissociative and psychotomimetic symptoms were evaluated based on the Clinician-Administered Dissociative States Scale (CADSS) and the Brief Psychiatric Rating Scale (BPRS)-four items. RESULTS Multi-Ketamine infusions resulted in a lower (trend) antidepressant response (37.1 % versus 57.8 %) and antidepressant remission (15.8 % versus 47.4 %) in elderly patients with depression compared with younger patients with depression (all ps > 0.05). Interestingly, elderly patients with depression had a higher MADRS score after six ketamine infusions compared with younger patients (p = 0.04). No significant differences in SSI-part 1 scores, HAMD item 3 scores, MADRS item 10 scores, CADSS scores, and BPRS-four items scores were found between the two groups at any assessment point (all ps > 0.05). CONCLUSION Our study shows that repeated-dose infusions of ketamine may be a feasible treatment strategy in elderly Chinese patients with depression; however, elderly patients with depression may be less responsive to ketamine compared with younger adults with depression.
Collapse
Affiliation(s)
- Wei Zheng
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yan-Ling Zhou
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Cheng-Yu Wang
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao-Feng Lan
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu-Ping Ning
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China; The first School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
32
|
Yang L, Liu H, Wang E, Liu H, Liu H, Zhou L, Deng T, Pan X, Hu Z, Yang X. Design, synthesis and evaluation of novel 1-phenyl-pyrrolo[1,2-b]isoquinolin-3-one derivatives as antagonists for the glycine binding site of the NMDA receptor. Eur J Med Chem 2023; 258:115624. [PMID: 37423124 DOI: 10.1016/j.ejmech.2023.115624] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
A new series of 1-phenyl-pyrrolo[1,2-b]isoquinolin-3-one derivatives were designed, synthesized and demonstrated to act as antagonists for the glycine binding site of the NMDA receptor. These new derivatives protected PC12 cells against NMDA-induced injury and cell apoptosis in vitro, among which compound 13b exhibited excellent cytoneuroprotective potency and shown a dose-dependent prevention. The increased intracellular Ca2+ influx caused by NMDA in PC12 cells was reversed when pretreated with compound 13b. Furthermore, the interaction between compound 13b and the glycine binding site of the NMDA receptor was validated via MST assay. It was observed that the stereochemistry of compound 13b did not influence the binding affinity, which was consistent with the neuroprotective result. Molecular docking study confirmed the observed activity of compound 13b by virtue of their Pi-stacking, cation-Pi, H-bonding and Pi-electron interactions with the key amino acids in the glycine binding pocket. These results confirm the potential of 1-phenyl-pyrrolo[1,2-b]isoquinolin-3-one derivatives as neuroprotective agents targeting the glycine binding site of the NMDA receptor.
Collapse
Affiliation(s)
- Lishou Yang
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Huadan Liu
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Enhua Wang
- Department of Medicine and Food, Guizhou Vocational College of Agriculture, Guiyang, 550041, PR China
| | - Huanhuan Liu
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Hongshi Liu
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Lang Zhou
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Tingfei Deng
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Xiong Pan
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Zhanxing Hu
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China
| | - Xiaosheng Yang
- School of Basic Medical Sciences/State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, 550014, PR China.
| |
Collapse
|
33
|
Feder A, Costi S, Rutter SB, Collins AB, Govindarajulu U, Jha MK, Horn SR, Kautz M, Corniquel M, Collins KA, Bevilacqua L, Glasgow AM, Brallier J, Pietrzak RH, Murrough JW, Charney DS. A Randomized Controlled Trial of Repeated Ketamine Administration for Chronic Posttraumatic Stress Disorder. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2023; 21:296-305. [PMID: 37404970 PMCID: PMC10316213 DOI: 10.1176/appi.focus.23021014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Objective Posttraumatic stress disorder (PTSD) is a chronic and disabling disorder, for which available pharmacotherapies have limited efficacy. The authors' previous proof-of-concept randomized controlled trial of single-dose intravenous ketamine infusion in individuals with PTSD showed significant and rapid PTSD symptom reduction 24 hours postinfusion. The present study is the first randomized controlled trial to test the efficacy and safety of repeated intravenous ketamine infusions for the treatment of chronic PTSD. Methods Individuals with chronic PTSD (N=30) were randomly assigned (1:1) to receive six infusions of ketamine (0.5 mg/kg) or midazolam (0.045 mg/kg) (psychoactive placebo control) over 2 consecutive weeks. Clinician-rated and self-report assessments were administered 24 hours after the first infusion and at weekly visits. The primary outcome measure was change in PTSD symptom severity, as assessed with the Clinician-Administered PTSD Scale for DSM-5 (CAPS-5), from baseline to 2 weeks (after completion of all infusions). Secondary outcome measures included the Impact of Event Scale-Revised, the Montgomery-Åsberg Depression Rating Scale (MADRS), and side effect measures. Results The ketamine group showed a significantly greater improvement in CAPS-5 and MADRS total scores than the midazolam group from baseline to week 2. At week 2, the mean CAPS-5 total score was 11.88 points (SE=3.96) lower in the ketamine group than in the midazolam group (d=1.13, 95% CI=0.36, 1.91). Sixty-seven percent of participants in the ketamine group were treatment responders, compared with 20% in the midazolam group. Among ketamine responders, the median time to loss of response was 27.5 days following the 2-week course of infusions. Ketamine infusions were well tolerated overall, without serious adverse events. Conclusions This randomized controlled trial provides the first evidence of efficacy of repeated ketamine infusions in reducing symptom severity in individuals with chronic PTSD. Further studies are warranted to understand ketamine's full potential as a treatment for chronic PTSD.Reprinted from Am J Psychiatry 2021; 178:193-202, with permission from American Psychiatric Association Publishing. Copyright © 2021.
Collapse
|
34
|
de A Simoes Moreira D, Gauer LE, Teixeira G, Fonseca da Silva AC, Cavalcanti S, Quevedo J. Efficacy and adverse effects of ketamine versus electroconvulsive therapy for major depressive disorder: A systematic review and meta-analysis. J Affect Disord 2023; 330:227-238. [PMID: 36907464 PMCID: PMC10497186 DOI: 10.1016/j.jad.2023.02.152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/14/2023]
Abstract
BACKGROUND ECT is considered the fastest and most effective treatment for TRD. Ketamine seems to be an attractive alternative due to its rapid-onset antidepressant effects and impact on suicidal thoughts. This study aimed to compare efficacy and tolerability of ECT and ketamine for different depression outcomes (PROSPERO/CRD42022349220). METHODS We searched MEDLINE, Web of Science, Embase, PsycINFO, Google Scholar, Cochrane Library and trial registries, which were the ClinicalTrials.gov and the World Health Organization's International Clinical Trials Registry Platform, without restrictions on publication date. SELECTION CRITERIA randomized controlled trials or cohorts comparing ketamine versus ECT in patients with TRD. RESULTS Eight studies met the inclusion criteria (of 2875 retrieved). Random-effects models comparing ketamine and ECT regarding the following outcomes were conducted: a) reduction of depressive symptoms severity through scales, g = -0.12, p = 0.68; b) response to therapy, RR = 0.89, p = 0.51; c) reported side-effects: dissociative symptoms, RR = 5.41, p = 0.06; nausea, RR = 0.73, p = 0.47; muscle pain, RR = 0.25, p = 0.02; and headache, RR = 0.39, p = 0.08. Influential & subgroup analyses were performed. LIMITATIONS Methodological issues with high risk of bias in some of the source material, reduced number of eligible studies with high in-between heterogeneity and small sample sizes. CONCLUSION Our study showed no evidence to support the superiority of ketamine over ECT for severity of depressive symptoms and response to therapy. Regarding side effects, there was a statistically significant decreased risk of muscle pain in patients treated with ketamine compared to ECT.
Collapse
Affiliation(s)
| | | | | | | | - Stefanie Cavalcanti
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA; Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA
| | - João Quevedo
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA; Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth Houston), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA; Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
35
|
Rikhani K, Vas C, Jha MK. Approach to Diagnosis and Management of Treatment-Resistant Depression. Psychiatr Clin North Am 2023; 46:247-259. [PMID: 37149343 DOI: 10.1016/j.psc.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Major depressive disorder is a chronic and recurrent illness that affects 20% of adults during their lifetime and is one of the leading causes of suicide in the United States. A systematic measurement-based care approach is the essential first step in the diagnosis and management of treatment-resistant depression (TRD) by promptly identifying individuals with depression and avoiding delays in treatment initiation. As comorbidities may be associated with poorer outcomes to commonly used antidepressants and increase risk of drug-drug interactions, their recognition and treatment is an essential component of management of TRD.
Collapse
Affiliation(s)
- Karina Rikhani
- Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9119, USA
| | - Collin Vas
- Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9119, USA
| | - Manish Kumar Jha
- Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9119, USA; Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9119, USA.
| |
Collapse
|
36
|
Tsang VW, Tao B, Dames S, Walsh Z, Kryskow P. Safety and tolerability of intramuscular and sublingual ketamine for psychiatric treatment in the Roots To Thrive ketamine-assisted therapy program: a retrospective chart review. Ther Adv Psychopharmacol 2023; 13:20451253231171512. [PMID: 37256163 PMCID: PMC10225955 DOI: 10.1177/20451253231171512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 04/06/2023] [Indexed: 06/01/2023] Open
Abstract
Background In the last few years, ketamine is becoming increasingly common in the treatment of mental health conditions, but there is a lack of safety data informing intramuscular and sublingual dosing in a community-focused group psychotherapy setting. The Roots To Thrive ketamine-assisted therapy (RTT-KaT) program is a unique 12-week RTT-KaT program with 12 community of practice (a form of group therapy) sessions and three ketamine medicine sessions. Objectives This study reports on adverse effects of intramuscular and sublingual ketamine dosing in a community group psychotherapy setting among 128 participants across four cohorts. Design Retrospective chart review. Methods A chart review of the RTT-KaT Program was performed retrospectively on four cohorts (n = 128) that participated in 448 sessions running between September 2020 and December 2021. Baseline characteristics and adverse events were captured including medication administration before, during, and after RTT-KaT sessions. Analyses by session and by individual were conducted. Chi-square test with Yates' continuity correction was used to assess side effects in subgroups from ketamine administration. Results RTT-KaT was well tolerated with none of the 128 participants dropping out of the program. Primarily, of the 448 sessions, 49.16% had elevated blood pressures post-KaT session by session. In terms of other adverse effects, 12.05% of participant-sessions experienced nausea, 2.52% had an episode of vomiting, 3.35% had a headache, and seven participant-sessions experienced dizziness. Analysis by individual revealed congruent findings. Conclusion These findings suggest good safety and tolerability for RTT-KaT among individuals seeking treatment for mental health issues. The majority of participants did not experience adverse reactions and the adverse events that were recorded involved transient symptoms that were resolved with rest and/or medications. The group therapy model described provides a comprehensive approach and presents a promising model for operating a KaT program in a community setting.
Collapse
Affiliation(s)
| | - Brendan Tao
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Shannon Dames
- Health Sciences and Human Services, Vancouver Island University, Nanaimo, BC, Canada
| | - Zach Walsh
- Department of Psychology, The University of British Columbia, Kelowna, BC, Canada
| | - Pam Kryskow
- Department of Family Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
37
|
Yan S, Xu C, Yang M, Zhang H, Cheng Y, Xue Z, He Z, Wang T, Bai S, Wang G, Wu J, Tong Z, Cai X. The expression of agmatinase manipulates the affective state of rats subjected to chronic restraint stress. Neuropharmacology 2023; 229:109476. [PMID: 36849038 DOI: 10.1016/j.neuropharm.2023.109476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
Agmatine is an endogenous polyamine produced from l-arginine and degraded by agmatinase (AGMAT). Studies in humans and animals have shown that agmatine has neuroprotective, anxiolytic, and antidepressant-like actions. However, little is known about the role of AGMAT in the action of agmatine or in the pathophysiology of psychiatric disorders. Therefore, this study aimed to investigate the role of AGMAT in the pathophysiology of MDD. In this study, we observed that AGMAT expression increased in the ventral hippocampus rather than in the medial prefrontal cortex in the chronic restraint stress (CRS) animal model of depression. Furthermore, we found that AGMAT overexpression in the ventral hippocampus elicited depressive- and anxiety-like behaviors, whereas knockdown of AGMAT exhibited antidepressant and anxiolytic effects in CRS animals. Field and whole-cell recordings of hippocampal CA1 revealed that AGMAT blockage increased Schaffer collateral-CA1 excitatory synaptic transmission, which was expressed both pre- and post-synaptically and was probably due to the inhibition of AGMAT-expressing local interneurons. Therefore, our results suggest that dysregulation of AGMAT is involved in the pathophysiology of depression and is a potential target for designing more effective antidepressants with fewer adverse effects to offer a better therapy for depression.
Collapse
Affiliation(s)
- Shi Yan
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorder, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Chang Xu
- College of Life Science, Shaanxi Normal University, 620 West Chang'an Street, Xi'an, Shaanxi 710119, China
| | - Mengli Yang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorder, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Huiqiang Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorder, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Ye Cheng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorder, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Zeping Xue
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorder, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Zecong He
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorder, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Tiantian Wang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorder, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Shangying Bai
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorder, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Gang Wang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorder, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders Beijing Anding Hospital Capital Medical University, Beijing 100088, China
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei 430070, China; Advanced Innovation Center for Human Brain Protection, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Zhiqian Tong
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiang Cai
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Beijing Institute of Brain Disorders, Advanced Innovation Center for Human Brain Protection, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
38
|
Chronic oral ketamine prevents anhedonia and alters neuronal activation in the lateral habenula and nucleus accumbens in rats under chronic unpredictable mild stress. Neuropharmacology 2023; 228:109468. [PMID: 36813161 DOI: 10.1016/j.neuropharm.2023.109468] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023]
Abstract
Acute injections of ketamine lead to rapid but transient antidepressant effects. Chronic oral treatment at low doses, a promising non-invasive alternative, may prolong this therapeutic effect. Here, we examine the antidepressant effects of chronic oral ketamine in rats under chronic unpredictable mild stress (CUMS), and reveal their neuronal correlates. Male Wistar rats were divided into control, ketamine, CUMS, and CUMS-ketamine groups. The CUMS protocol was applied to the latter two groups for 9 weeks, and ketamine (0.013 mg/ml) was provided ad libitum to the ketamine and CUMS-ketamine groups for 5 weeks. The sucrose consumption test, forced swim test, open field test, elevated plus maze, and Morris water maze were respectively used to assess anhedonia, behavioral despair, general locomotor activity, anxiety-like behavior and spatial reference memory. CUMS caused a reduction of sucrose consumption and impaired spatial memory, accompanied by increased neuronal activation in the lateral habenula (LHb) and paraventricular thalamic nucleus (PVT). Oral ketamine prevented behavioral despair and CUMS-induced anhedonia. Reward-triggered c-Fos immunoreactivity was decreased in the LHb and increased in the nucleus accumbens shell (NAcSh) in the CUMS-ketamine group compared to the CUMS group. Ketamine did not produce a differential effect in the OFT, EPM and MWM. These results show that chronic oral ketamine at low doses prevents anhedonia without impairing spatial reference memory. The observed neuronal activation changes in the LHb and NAcSh may be involved in the preventive effects of ketamine on anhedonia. This article is part of the Special Issue on "Ketamine and its Metabolites".
Collapse
|
39
|
Kim JW, Suzuki K, Kavalali ET, Monteggia LM. Bridging rapid and sustained antidepressant effects of ketamine. Trends Mol Med 2023; 29:364-375. [PMID: 36907686 PMCID: PMC10101916 DOI: 10.1016/j.molmed.2023.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/05/2023] [Accepted: 02/15/2023] [Indexed: 03/12/2023]
Abstract
Acute administration of (R,S)-ketamine (ketamine) produces rapid antidepressant effects that in some patients can be sustained for several days to more than a week. Ketamine blocks N-methyl-d-asparate (NMDA) receptors (NMDARs) to elicit specific downstream signaling that induces a novel form of synaptic plasticity in the hippocampus that has been linked to the rapid antidepressant action. These signaling events lead to subsequent downstream transcriptional changes that are involved in the sustained antidepressant effects. Here we review how ketamine triggers this intracellular signaling pathway to mediate synaptic plasticity which underlies the rapid antidepressant effects and links it to downstream signaling and the sustained antidepressant effects.
Collapse
Affiliation(s)
- Ji-Woon Kim
- Department of Pharmacology and the Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA; College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea; Department of Regulatory Science, Gradaute School, Kyung Hee University, Seoul, Republic of Korea; Institute of Regulatory Innovation through Science, Kyung Hee University, Seoul, Republic of Korea
| | - Kanzo Suzuki
- Department of Pharmacology and the Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA; Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Japan
| | - Ege T Kavalali
- Department of Pharmacology and the Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA
| | - Lisa M Monteggia
- Department of Pharmacology and the Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA.
| |
Collapse
|
40
|
Fancy F, Rodrigues NB, Di Vincenzo JD, Chau EH, Sethi R, Husain MI, Gill H, Tabassum A, Mckenzie A, Phan L, McIntyre RS, Rosenblat JD. Real-world effectiveness of repeated ketamine infusions for treatment-resistant bipolar depression. Bipolar Disord 2023; 25:99-109. [PMID: 36516343 DOI: 10.1111/bdi.13284] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Clinical trials have demonstrated rapid antidepressant effects with intravenous (IV) ketamine for major depressive disorder, with relatively less research specifically for bipolar depression. Herein, we describe the real-world effectiveness of repeated ketamine infusions for treatment-resistant bipolar depression. METHODS This study was conducted in a community clinic in Mississauga, Ontario (Canadian Rapid Treatment Centre of Excellence; Braxia Health). In this observational study (NCT04209296), patients with treatment-resistant bipolar I/II depression (n = 66) received four sub-anesthetic doses of IV ketamine (0.5-0.75 mg/kg) over a two-week period. Symptoms of depression, suicidality, anxiety, and functioning were assessed with validated self-report measures. RESULTS Statistically and clinically significant antidepressant effects were observed in the overall sample, as measured by the Quick Inventory for Depression Symptomatology-Self Report-16 (QIDS-SR16 ) with further reductions in depressive symptoms observed after each subsequent infusion (n = 66; mean QIDS-SR16 reduction of 6.08+/-1.39; p < 0.0001). Significant reductions of suicidal thoughts (QIDS-SR16 -Suicide Item) and anxiety (Generalized Anxiety Disorder-7) were also observed with functional improvements on the Sheehan Disability Scale (p < 0.0001 on all measures). Moreover, the response rate (QIDS-SR16 total score decrease ≥50% from baseline) was 35% and remission rate (QIDS-SR16 total score ≤5) was 20% after four infusions. Infusions were generally well tolerated with treatment-emergent hypomania observed in only three patients (4.5%) with zero cases of mania or psychosis. CONCLUSIONS Real-world effectiveness of IV ketamine for bipolar depression was observed. Repeated doses were associated with greater symptom reduction and adequate tolerability.
Collapse
Affiliation(s)
- Farhan Fancy
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Nelson B Rodrigues
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Joshua D Di Vincenzo
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Edmond H Chau
- Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada
| | - Rickinder Sethi
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Muhammad I Husain
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Hartej Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Aniqa Tabassum
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Andrea Mckenzie
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Lee Phan
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Harris L, Regan MC, Myers SJ, Nocilla KA, Akins NS, Tahirovic YA, Wilson LJ, Dingledine R, Furukawa H, Traynelis SF, Liotta DC. Novel GluN2B-Selective NMDA Receptor Negative Allosteric Modulator Possesses Intrinsic Analgesic Properties and Enhances Analgesia of Morphine in a Rodent Tail Flick Pain Model. ACS Chem Neurosci 2023; 14:917-935. [PMID: 36779874 PMCID: PMC9983021 DOI: 10.1021/acschemneuro.2c00779] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/24/2023] [Indexed: 02/14/2023] Open
Abstract
Many cases of accidental death associated with drug overdose are due to chronic opioid use, tolerance, and addiction. Analgesic tolerance is characterized by a decreased response to the analgesic effects of opioids, requiring increasingly higher doses to maintain the desired level of pain relief. Overactivation of GluN2B-containing N-methyl-d-Aspartate receptors is thought to play a key role in mechanisms underlying cellular adaptation that takes place in the development of analgesic tolerance. Herein, we describe a novel GluN2B-selective negative allosteric modulator, EU93-108, that shows high potency and brain penetrance. We describe the structural basis for binding at atomic resolution. This compound possesses intrinsic analgesic properties in the rodent tail immersion test. EU93-108 has an acute and significant anodyne effect, whereby morphine when combined with EU93-108 produces a higher tail flick latency compared to that of morphine alone. These data suggest that engagement of GluN2B as a target has utility in the treatment of pain, and EU93-108 could serve as an appropriate tool compound to interrogate this hypothesis. Future structure-activity relationship work around this scaffold could give rise to compounds that can be co-administered with opioids to diminish the onset of tolerance due to chronic opioid use, thereby modifying their utility.
Collapse
Affiliation(s)
- Lynnea
D. Harris
- Department
of Chemistry, Emory University, Atlanta, Georgia30322, United States
| | - Michael C. Regan
- W.M.
Keck Structural Biology Laboratory, Cold
Spring Harbor Laboratory, New York, New York11724, United States
- RADD
Pharmaceuticals, Westport, Connecticut06880, United States
| | - Scott J. Myers
- Department
of Pharmacology and Chemical Biology, Emory
University, Atlanta, Georgia30322, United States
| | - Kelsey A. Nocilla
- Department
of Pharmacology and Chemical Biology, Emory
University, Atlanta, Georgia30322, United States
| | - Nicholas S. Akins
- Department
of Chemistry, Emory University, Atlanta, Georgia30322, United States
| | - Yesim A. Tahirovic
- Department
of Chemistry, Emory University, Atlanta, Georgia30322, United States
| | - Lawrence J. Wilson
- Department
of Chemistry, Emory University, Atlanta, Georgia30322, United States
| | - Ray Dingledine
- Department
of Pharmacology and Chemical Biology, Emory
University, Atlanta, Georgia30322, United States
| | - Hiro Furukawa
- W.M.
Keck Structural Biology Laboratory, Cold
Spring Harbor Laboratory, New York, New York11724, United States
| | - Stephen F. Traynelis
- Department
of Pharmacology and Chemical Biology, Emory
University, Atlanta, Georgia30322, United States
| | - Dennis C. Liotta
- Department
of Chemistry, Emory University, Atlanta, Georgia30322, United States
| |
Collapse
|
42
|
Jha MK, Mathew SJ. Pharmacotherapies for Treatment-Resistant Depression: How Antipsychotics Fit in the Rapidly Evolving Therapeutic Landscape. Am J Psychiatry 2023; 180:190-199. [PMID: 36855876 DOI: 10.1176/appi.ajp.20230025] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
One in three adults with major depressive disorder (MDD) do not experience clinically significant improvement after multiple sequential courses of antidepressants and have treatment-resistant depression (TRD). The presence of TRD contributes to the morbidity and excess mortality associated with MDD and has been linked to significantly increased health care expenses. In the absence of a consensus definition of TRD, this report takes a broad approach by considering inadequate response to one or more courses of antidepressants and focuses on atypical antipsychotics that are approved by the U.S. Food and Drug Administration for treatment of depression (aripiprazole, brexpiprazole, cariprazine, extended-release quetiapine, and olanzapine-fluoxetine combination). While multiple acute-phase studies have demonstrated the efficacy of these medications in improving depressive symptoms, clinically meaningful improvement (i.e., remission) remains limited, with significant concerns about side effects (including weight gain, metabolic dysfunction, extrapyramidal symptoms, and tardive dyskinesia), especially with long-term use. With the rapidly evolving landscape of antidepressant treatments over the past few years, which has witnessed approval of rapid-acting antidepressants (e.g., esketamine nasal spray and dextromethorphan-bupropion combination) and several more in the late-stage pipeline (e.g., zuranolone and psilocybin), it remains to be seen whether the use of atypical antipsychotics will go the way of the older and rarely prescribed antidepressants (such as tricyclics and monoamine oxidase inhibitors). Pragmatic clinical trials are needed to compare the effectiveness of atypical antipsychotics with TRD-specific pharmacotherapies and neuromodulation treatments and to identify the optimal sequencing of these varied approaches for patients with MDD. When using atypical antipsychotics, clinicians and patients are encouraged to use a shared decision-making approach by personalizing treatment selection based on anticipated side effects, tolerability, cost, and feasibility.
Collapse
Affiliation(s)
- Manish K Jha
- Center for Depression Research and Clinical Care, Department of Psychiatry, UT Southwestern Medical Center, and O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas (Jha); Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Mathew); Michael E. DeBakey VA Medical Center, Houston (Mathew); Menninger Clinic, Houston (Mathew)
| | - Sanjay J Mathew
- Center for Depression Research and Clinical Care, Department of Psychiatry, UT Southwestern Medical Center, and O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas (Jha); Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston (Mathew); Michael E. DeBakey VA Medical Center, Houston (Mathew); Menninger Clinic, Houston (Mathew)
| |
Collapse
|
43
|
Liu H, Wang C, Lan X, Li W, Zhang F, Fu L, Ye Y, Ning Y, Zhou Y. Functional connectivity of the amygdala and the antidepressant and antisuicidal effects of repeated ketamine infusions in major depressive disorder. Front Neurosci 2023; 17:1123797. [PMID: 36816116 PMCID: PMC9932998 DOI: 10.3389/fnins.2023.1123797] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023] Open
Abstract
Background Dysfunction of the amygdala is the core pathogenesis of major depressive disorder (MDD). However, it remains unclear whether ketamine treatment could modulate characteristics of amygdala-related networks. We aimed to explore the relationship between changes in the resting-state functional connectivity (RSFC) of the amygdala and the treatment of ketamine in MDD patients and to identify important neuroimaging predictors of treatment outcome. Methods Thirty-nine MDD patients received six subanesthetic dose infusions of ketamine. Depressive and suicidal symptoms were assessed and magnetic resonance imaging (MRI) scans were performed before and after six ketamine infusions. Forty-five healthy controls also underwent once MRI scans. Seed-based RSFC analyses were performed, focusing on the bilateral amygdala. Results After ketamine treatment, the RSFC between the left amygdala (LA) and the left medial superior frontal gyrus (mSFG) of MDD patients enhanced significantly, and this change was positively correlated with the reduction in depressive symptoms (r = 0.40, p = 0.012). The combination baseline RSFC of LA - right putamen and right amygdala (RA) - right putamen was related to the antidepressant and antisuicidal effects of ketamine. The combination baseline RSFC of LA - right putamen and RA - right putamen could predict the ineffective antidepressant (AUC = 0.739, p = 0.011) and antisuicidal effects of ketamine (AUC = 0.827, p = 0.001). Conclusion Ketamine can regulate the relevant circuits of amygdala and mSFG, and the baseline RSFC between bilateral amygdala and right putamen may be a predictor of the response of ketamine's antidepressant and antisuicidal treatment. Clinical trial registration https://www.chictr.org.cn/showproj.aspx?proj=20875, identifier ChiCTR-OOC-17012239.
Collapse
Affiliation(s)
- Haiyan Liu
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Chengyu Wang
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiaofeng Lan
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Weicheng Li
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China,Department of Psychology, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Fan Zhang
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China,Department of Psychology, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ling Fu
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China,Department of Psychology, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yanxiang Ye
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yuping Ning
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China,Department of Psychology, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China,*Correspondence: Yuping Ning,
| | - Yanling Zhou
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China,Yanling Zhou,
| |
Collapse
|
44
|
Ago Y, Yokoyama R, Asano S, Hashimoto H. Roles of the monoaminergic system in the antidepressant effects of ketamine and its metabolites. Neuropharmacology 2023; 223:109313. [PMID: 36328065 DOI: 10.1016/j.neuropharm.2022.109313] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
Abstract
While the molecular target of (R,S)-ketamine (ketamine) is thought to be the NMDA receptor, subanesthetic doses of ketamine have been known to modulate monoaminergic neurotransmission in the central nervous system. Although the involvement of the serotonergic system in the antidepressant effects of ketamine has been reported in most studies of this topic, some recent studies have reported that the dopaminergic system plays a key role in the effects of ketamine. Additionally, several lines of evidence suggest that the antidepressant-like effects of (R)-ketamine might be independent of the monoaminergic system. Ketamine metabolites also differ considerably in their ability to regulate monoamine neurotransmitters relative to (S)-ketamine and (R)-ketamine, while (2R,6R)-hydroxynorketamine might share common serotonergic signaling mechanisms with ketamine. In the current review, we summarize the effects of ketamine and its metabolites on monoamine neurotransmission in the brain and discuss the potential roles of the monoaminergic system in the mechanism of action of ketamine.
Collapse
Affiliation(s)
- Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, 734-8553, Japan.
| | - Rei Yokoyama
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Satoshi Asano
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, 734-8553, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan; Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Osaka, 565-0871, Japan; Division of Bioscience, Institute for Datability Science, Osaka University, Suita, Osaka, 565-0871, Japan; Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, 565-0871, Japan; Department of Molecular Pharmaceutical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
45
|
Huang MC, Chen CH, Liu TH, Chung AN, Liu YL, Quednow BB, Bavato F. Comorbidity of ketamine dependence with major depressive disorder increases the vulnerability to neuroaxonal pathology. J Psychiatr Res 2023; 158:360-364. [PMID: 36640660 DOI: 10.1016/j.jpsychires.2023.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/02/2023] [Accepted: 01/09/2023] [Indexed: 01/11/2023]
Abstract
We recently demonstrated that patients with ketamine dependence (KD) have increased serum levels of neurofilament light chain (NfL), a novel marker of active neuroaxonal pathology, with NfL levels being significantly higher in those KD patients comorbid with major depressive disorder (MDD). However, considering that NfL elevation has been associated with both ketamine-related brain pathology and MDD, we could not determine whether the observed elevation of NfL levels was driven by an interaction of KD with MDD or by MDD itself. Therefore, we compared serum NfL levels between 35 patients with MDD without ketamine use (MDD group), 23 with KD without MDD (KD without MDD group), 30 KD with MDD (KD with MDD group), and 86 healthy controls (HC group). Using a 2*2 (KD*MDD) generalized linear model controlling for age, sex, body mass index, and smoking status, we found that KD and KD*MDD interactions, but not MDD factor, significantly affected NfL levels. Posthoc tests showed that the KD with MDD group had significantly higher NfL levels than all other groups. The KD without MDD group also showed higher NfL levels than the MDD and, as shown before, HC groups. The levels in MDD group were not different from the HC group. These results suggest that the interaction of KD with MDD, but not MDD alone, results in increased vulnerability to neuroaxonal pathology.
Collapse
Affiliation(s)
- Ming-Chyi Huang
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan; Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chun-Hsin Chen
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Psychiatric Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Psychiatry, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Tung-Hsia Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - An-Nie Chung
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan; Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan.
| | - Boris B Quednow
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy, and Psychosomatics, Psychiatric Hospital of the University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Francesco Bavato
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy, and Psychosomatics, Psychiatric Hospital of the University of Zurich, Zurich, Switzerland
| |
Collapse
|
46
|
von Rotz R, Schindowski EM, Jungwirth J, Schuldt A, Rieser NM, Zahoranszky K, Seifritz E, Nowak A, Nowak P, Jäncke L, Preller KH, Vollenweider FX. Single-dose psilocybin-assisted therapy in major depressive disorder: A placebo-controlled, double-blind, randomised clinical trial. EClinicalMedicine 2023; 56:101809. [PMID: 36636296 PMCID: PMC9830149 DOI: 10.1016/j.eclinm.2022.101809] [Citation(s) in RCA: 71] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Psilocybin has been suggested as a novel, rapid-acting treatment for depression. Two consecutive doses have been shown to markedly decrease symptom severity in an open-label setting or when compared to a waiting list group. To date, to our knowledge, no other trial compared a single, moderate dose of psilocybin to a placebo condition. METHODS In this double-blind, randomised clinical trial, 52 participants diagnosed with major depressive disorder and no unstable somatic conditions were allocated to receive either a single, moderate dose (0.215 mg/kg body weight) of psilocybin or placebo in conjunction with psychological support. MADRS and BDI scores were assessed to estimate depression severity, while changes from baseline to 14 days after the intervention were defined as primary endpoints. The trial took place between April 11th, 2019 and October 12th, 2021 at the psychiatric university hospital in Zürich, Switzerland and was registered with clinicaltrials.gov (NCT03715127). FINDINGS The psilocybin condition showed an absolute decrease in symptom severity of -13.0 points compared to baseline and were significantly larger than those in the placebo condition (95% CI -15.0 to -1.3; Cohens' d = 0.97; P = 0.0011; MADRS) and -13.2 points (95% CI; -13.4 to -1.3; Cohens' d = 0.67; P = 0.019; BDI) 14 days after the intervention. 14/26 (54%) participants met the MADRS remission criteria in the psilocybin condition. INTERPRETATION These results suggest that a single, moderate dose of psilocybin significantly reduces depressive symptoms compared to a placebo condition for at least two weeks. No serious adverse events were recorded. Larger, multi-centric trials with longer follow-up periods are needed to inform further optimisation of this novel treatment paradigm. FUNDING The study was funded by the Swiss National Science Foundation, Crowdfunding, the Swiss Neuromatrix Foundation, and the Heffter Research Institute.
Collapse
Affiliation(s)
- Robin von Rotz
- Neurophenomenology of Consciousness Lab, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
- Corresponding author.
| | - Eva M. Schindowski
- Neurophenomenology of Consciousness Lab, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
| | - Johannes Jungwirth
- Neurophenomenology of Consciousness Lab, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
| | - Anna Schuldt
- Neurophenomenology of Consciousness Lab, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
| | - Nathalie M. Rieser
- Neurophenomenology of Consciousness Lab, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
| | - Katharina Zahoranszky
- Neurophenomenology of Consciousness Lab, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
| | - Albina Nowak
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
| | - Peter Nowak
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
| | - Lutz Jäncke
- Division Neuropsychology, Department of Psychology, University of Zürich, Zürich, Switzerland
| | - Katrin H. Preller
- Neurophenomenology of Consciousness Lab, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
| | - Franz X. Vollenweider
- Neurophenomenology of Consciousness Lab, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zürich, Switzerland
| |
Collapse
|
47
|
Channer B, Matt SM, Nickoloff-Bybel EA, Pappa V, Agarwal Y, Wickman J, Gaskill PJ. Dopamine, Immunity, and Disease. Pharmacol Rev 2023; 75:62-158. [PMID: 36757901 PMCID: PMC9832385 DOI: 10.1124/pharmrev.122.000618] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson's disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity. SIGNIFICANCE STATEMENT: Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
Collapse
Affiliation(s)
- Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Emily A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Vasiliki Pappa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Jason Wickman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| |
Collapse
|
48
|
Mu Opioid Receptor Activation Mediates (S)-ketamine Reinforcement in Rats: Implications for Abuse Liability. Biol Psychiatry 2022:S0006-3223(22)01854-6. [PMID: 36841701 DOI: 10.1016/j.biopsych.2022.12.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/06/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND (S)-ketamine is an NMDA receptor antagonist, but it also binds to and activates mu opioid receptors (MORs) and kappa opioid receptors in vitro. However, the extent to which these receptors contribute to (S)-ketamine's in vivo pharmacology is unknown. METHODS We investigated the extent to which (S)-ketamine interacts with opioid receptors in rats by combining in vitro and in vivo pharmacological approaches, in vivo molecular and functional imaging, and behavioral procedures relevant to human abuse liability. RESULTS We found that the preferential opioid receptor antagonist naltrexone decreased (S)-ketamine self-administration and (S)-ketamine-induced activation of the nucleus accumbens, a key brain reward region. A single reinforcing dose of (S)-ketamine occupied brain MORs in vivo, and repeated doses decreased MOR density and activity and decreased heroin reinforcement without producing changes in NMDA receptor or kappa opioid receptor density. CONCLUSIONS These results suggest that (S)-ketamine's abuse liability in humans is mediated in part by brain MORs.
Collapse
|
49
|
ERK/mTOR signaling may underlying the antidepressant actions of rapastinel in mice. Transl Psychiatry 2022; 12:522. [PMID: 36550125 PMCID: PMC9780240 DOI: 10.1038/s41398-022-02290-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/10/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Rapastinel as the allosteric modulator of N-methyl-D-aspartate receptor (NMDAR) produces rapid antidepressant-like effects dependent on brain-derived neurotrophic factor (BDNF) and VGF (nonacryonimic) release. Herein, we further explore the molecular mechanisms of the antidepressant effects of repeated administration with rapastinel in mice. Our results showed that continuous 3-day rapastinel (5 and 10 mg/kg, i.v.) produced antidepressant-like actions dependent on the increase in extracellular regulated protein kinase (ERK)/mammalian target of rapamycin (mTOR) signaling and downstream substrates p70S6 kinase (p70S6k) and the eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), which may induce the expression of VGF and BDNF in the hippocampus and prefrontal cortex of mice. Furthermore, compared with a single treatment, our data indicated that 3-day repeated rapastinel treatment produced antidepressant-like actions accompanied by potentiation of ERK/mTOR/VGF/BDNF/tropomyosin-related kinase receptor B (TrkB) signaling. Based on previous and our supplementary data that showed the pivotal role of on α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) in the rapid release of VGF and BDNF and activation of TrkB by a single dose of rapastinel, we postulate that the antidepressant-like effects of single or repeated administration of rapastinel may result in the rapid release of VGF and BDNF or ERK/mTOR signaling pathway-mediated VGF/BDNF/TrkB autoregulatory feedback loop respectively. Our current work adds new knowledge to the molecular mechanisms that underlie the antidepressant-like actions of rapastinel in mice.
Collapse
|
50
|
Chapman N, Browning M, Baghurst D, Hotopf M, Willis D, Haylock S, Zakaria S, Speechley J, Withey J, Brooks E, Chan F, Pappa S, Geddes J, Insole L, Mohammed Z, Kessler D, Jones PB, Mansoori P. Setting national research priorities for difficult-to-treat depression in the UK between 2021-2026. J Glob Health 2022; 12:09004. [PMID: 36472926 PMCID: PMC9727823 DOI: 10.7189/jogh.12.09004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Difficult-to-treat depression (DTD) presents a substantial health care challenge, with around one-third of people diagnosed with a depressive episode in the UK finding that their symptoms persist following treatment. This study aimed to identify priority research questions (RQs) that could inform the development of new and improved treatments, interventions, and support for people with DTD. Methods Using an adapted Child Health and Nutrition Research Initiative (CHNRI) method, this national prioritisation exercise engaged 60 leading researchers and health care professionals in the UK, as well as 25 wider stakeholders with relevant lived experience to produce a ranked list of priority RQs in DTD. The final list of 99 distinct RQs was independently scored by 42 individuals against a list of five criteria: answerability, effectiveness, impact on health, deliverability, and equity. Results Highly ranked RQs covered a range of novel and existing treatments. The three highest scoring RQs included evaluation of psychological and pharmacological therapies (eg, behavioural activation, and augmentation therapies), as well as social interventions to reduce loneliness or increase support for people with DTD. Conclusions This exercise identified and prioritised 99 RQs that could inform future research and funding decisions over the next five years. The results of this research could improve treatment and support for people affected by DTD. It also serves as an example of ways in which the CHNRI method can be adapted in a collaborative manner to provide a more active role for patients, carers, and health care professionals.
Collapse
Affiliation(s)
- Natalya Chapman
- National Institute for Health and Care Research Central Commissioning Facility, Twickenham, UK
| | - Michael Browning
- Department of Psychiatry, University of Oxford, Oxford, UK,Oxford Health NHS Foundation Trust, Oxford, UK
| | - David Baghurst
- National Institute for Health and Care Research Central Commissioning Facility, Twickenham, UK,National Institute for Health and Care Research Office for Clinical Research Infrastructure, Twickenham, UK
| | - Matthew Hotopf
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK,South London and the Maudsley NHS Foundation Trust, London, UK
| | - Debbie Willis
- National Institute for Health and Care Research Evaluation, Trials and Studies Coordinating Centre, Southampton, UK
| | - Stuart Haylock
- National Institute for Health and Care Research Central Commissioning Facility, Twickenham, UK
| | - Sana Zakaria
- National Institute for Health and Care Research Central Commissioning Facility, Twickenham, UK
| | - Jan Speechley
- National Institute for Health and Care Research Central Commissioning Facility, Twickenham, UK
| | - James Withey
- National Institute for Health and Care Research Central Commissioning Facility, Twickenham, UK
| | - Edmund Brooks
- National Institute for Health and Care Research Central Commissioning Facility, Twickenham, UK
| | - Fiona Chan
- National Institute for Health and Care Research Central Commissioning Facility, Twickenham, UK
| | - Sofia Pappa
- West London NHS Trust, London, UK,Department of Psychiatry, Imperial College London, London, UK
| | - John Geddes
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Lisa Insole
- Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust, Newcastle, UK
| | - Zeid Mohammed
- Cumbria, Northumberland, Tyne and Wear NHS Foundation Trust, Newcastle, UK
| | - David Kessler
- Centre for Academic Mental Health, Bristol Medical School, University of Bristol, Bristol, UK
| | - Peter B Jones
- Department of Psychiatry, University of Cambridge, Cambridge and NIHR ARC East of England, Cambridgeshire & Peterborough NHS Foundation Trust, UK
| | - Parisa Mansoori
- National Institute for Health and Care Research Office for Clinical Research Infrastructure, Twickenham, UK
| | | |
Collapse
|