1
|
Kheirkhah M, Hejazi NS, Nugent AC, Gilbert JR, Leistritz L, Walter M, Duncan WC, Goldman D, Zarate CA. Exploring the link between waking gamma and sleep delta power in healthy volunteers and individuals with treatment-resistant depression. J Affect Disord 2025; 385:119448. [PMID: 40398609 PMCID: PMC12172513 DOI: 10.1016/j.jad.2025.119448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/28/2025] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Sleep disruptions are a core feature of both major depressive disorder and treatment-resistant depression (TRD), which is defined by persistent symptoms despite multiple treatment efforts. In addition, disruptions in wakeful gamma power and sleep-related delta power have been observed in individuals with TRD. This study explored the association between gamma oscillations (30-100 Hz) occurring during wakefulness and delta power (0.5-4 Hz) in non-rapid eye movement (NREM) sleep-both of which have separately been implicated in plasticity-in healthy volunteers (HVs) and individuals with TRD. Specifically, the study explored whether a relationship exists between daytime wake gamma power and sleep NREM delta power in HVs and those with TRD. METHODS Brain activity was measured via electroencephalography (night-time) and magnetoencephalography (daytime resting state) in 23 HVs (9M/14F; 20-56 yrs) and 40 medication-free TRD participants (20 M/20F; 18-63 yrs). RESULTS In HVs, NREM episode (NREM1) sleep delta power correlated with daytime wake gamma power (r = 0.417; p = 0.04). This correlation was absent in TRD participants (r = 0.108; p = 0.50). LIMITATIONS The sample size of the HVs (n = 23) was smaller than the TRD participants (n = 40), and the measurement order for wake gamma power and sleep delta power varied. CONCLUSIONS These findings identify a possible link between daytime wake gamma power and NREM1 sleep delta power in HVs, supporting an association between gamma and delta power in sleep homeostasis. The lack of such a relationship in medication-free individuals with TRD suggests disrupted synaptic homeostasis that may contribute to impaired synaptic plasticity in TRD. Clinical Trials Identifiers: NCT00088699 and NCT01204918.
Collapse
Affiliation(s)
- Mina Kheirkhah
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA; Department of Psychiatry and Psychotherapy, Jena University Hospital, 07740 Jena, Germany
| | - Nadia S Hejazi
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Allison C Nugent
- Magnetoencephalography Core, National Institute of Mental Health, Bethesda, MD, USA.
| | - Jessica R Gilbert
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Lutz Leistritz
- Institute of Medical Statistics, Computer and Data Sciences, Jena University Hospital, 07740 Jena, Germany.
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, 07740 Jena, Germany.
| | - Wallace C Duncan
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - David Goldman
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Ohtani Y, Tani H, Honda S, Nomoto-Takahashi K, Yatomi T, Yonezawa K, Tomiyama S, Nagai N, Kusudo K, Moriyama S, Noda Y, Koike S, Edden RAE, Uchida H, Nakajima S. Glutamate plus glutamine to GABA ratio as a predictor of ketamine response in treatment-resistant depression: A double-blind, randomized, open-label extension study. J Affect Disord 2025; 383:354-362. [PMID: 40311814 DOI: 10.1016/j.jad.2025.04.165] [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: 01/22/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Approximately 30 % of patients with treatment-resistant depression (TRD) respond to ketamine; however, no replicable predictors of response have been reported. The imbalance between excitatory and inhibitory neurotransmissions may be implicated in the mechanism of action of ketamine. This study aimed to evaluate whether the ratio of glutamate and glutamine (Glx) to GABA levels at baseline in the dorsal anterior cingulate cortex (dACC) could predict ketamine response in patients with TRD. METHOD This exploratory study analyzed data from a double-blind randomized clinical trial with an open-label extension study (jRCTs031210124). Fifteen participants in the ketamine group and 15 of 16 participants in the placebo group received repeated intravenous ketamine during the double-blind and open-label extension periods, respectively. We measured Glx and GABA levels in the dACC before and after treatment during the double-blind period using proton magnetic resonance spectroscopy. The 17-item Hamilton Depression Rating Scale (HDRS-17) was measured for depressive symptomatology. General linear models were used to examine the relationship between baseline Glx/GABA ratio and HDRS-17 score changes. RESULT Changes in HDRS-17 scores (mean (±SD)) following ketamine treatment were -4.9 (6.5) and -4.9 (5.2) in the double-blind and open-label periods, respectively. A higher baseline dACC Glx/GABA ratio was correlated with greater improvement in HDRS-17 (β = -0.42, p = 0.040). In the ketamine group, a reduction in the dACC Glx/GABA ratio was correlated with greater HDRS-17 improvement (β = 0.74, p = 0.009) with no such association in the placebo group. CONCLUSION These results suggest that excitatory-inhibitory imbalance in the dACC may predict the efficacy of ketamine in TRD.
Collapse
Affiliation(s)
- Yohei Ohtani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan; Department of Psychiatry, Minami-Hanno Hospital, Saitama, Japan
| | - Hideaki Tani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan.
| | - Shiori Honda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan; Department of Psychiatry and Behavioral Health, Renaissance School of Medicine, Stony Brook University, USA
| | | | - 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
| | - Sotaro Moriyama
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yoshihiro Noda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shinsuke Koike
- Center for Evolutionary Cognitive Sciences, Graduate School of Art and Sciences, The University of Tokyo, Tokyo, Japan; University of Tokyo Institute for Diversity and Adaptation of Human Mind, The University of Tokyo, Tokyo, Japan; The International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo, Japan
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
3
|
Moore TM, Walker K, Tung E, Teed AR, Hell F, Kinreich S, Jung R, Abdel F, Hanson RW, Ahmed SS. Combined ketamine and psychotherapy provide no additional benefit beyond ketamine alone in treating depression or PTSD: Evidence from a help-seeking sample. J Affect Disord 2025; 381:233-241. [PMID: 40203964 DOI: 10.1016/j.jad.2025.04.041] [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: 12/09/2024] [Revised: 03/22/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Depression and PTSD are prevalent psychiatric conditions that often co-occur and significantly impact quality of life. Ketamine has emerged as a promising rapid-acting treatment for both conditions, while traditional treatments like psychotherapy typically require weeks to show effects. This study investigated whether combining ketamine with psychotherapy produces greater symptom improvement compared to ketamine alone. We analyzed overlapping samples of N = 202, N = 470, and N = 624 help-seeking individuals (all samples ∼60 % female, mean age ∼ 42 years) who received either ketamine alone (KET) or ketamine plus psychotherapy (KET+PSY) across 4-14 treatment sessions within a 30- or 180-day period. Depression symptoms were measured using the PHQ-9, and PTSD symptoms were assessed using the PCL-5. Trajectories of symptom change were analyzed using generalized additive mixed-effects models, controlling for baseline symptoms, demographics, and treatment intervals. Both treatment groups showed substantial improvement in depression and PTSD symptoms, with similar patterns of rapid initial decline followed by stabilization. Contrary to our hypothesis, we found no significant differences in symptom trajectories between the KET and KET+PSY groups. Exploratory analyses revealed non-significant but notable patterns where younger females showed better outcomes with combined treatment, while older males showed better outcomes with ketamine alone. These findings suggest that ketamine's therapeutic effects may be robust enough that additional psychotherapy during the acute treatment phase does not significantly enhance 30-day (and possibly 180-day) outcomes. However, longer-term benefits of combined treatment and potential demographic-specific treatment responses warrant further investigation. These results have important implications for treatment planning and resource allocation in clinical settings.
Collapse
Affiliation(s)
- Tyler M Moore
- Sama Therapeutics, Cambridge, MA 02141, USA; Department of Psychiatry, Brain Behavior Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | - Emma Tung
- University of Chicago, IL 60637, USA
| | - Adam R Teed
- Sama Therapeutics, Cambridge, MA 02141, USA; Laureate Institute for Brain Research, Tulsa, OK 74136, USA
| | - Franz Hell
- Sama Therapeutics, Cambridge, MA 02141, USA
| | - Sivan Kinreich
- State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Rex Jung
- Sama Therapeutics, Cambridge, MA 02141, USA; Brain and Behavioral Associates, Albuquerque, NM 87106, USA
| | - Fadi Abdel
- Alto Neuroscience, Mountain View, CA 94041, USA
| | | | | |
Collapse
|
4
|
Zheng F, Hu Z, Chen H, Cheng J, Hou Q, Zheng J, Gong X, Ji J, Zayniddin N, Abduahadi S, Mamateli O, Wang G, Li P, Hu T, Tian G, Xu Z, Zhu W, Aisa HA, Shen J, He Y. Heterocycle-fused phenylcyclohexylamines as novel multi-target antagonists of N-methyl-D-aspartate (NMDA) receptor and monoamine transporter for treating depression. Eur J Med Chem 2025; 291:117538. [PMID: 40188584 DOI: 10.1016/j.ejmech.2025.117538] [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: 12/31/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/08/2025]
Abstract
Simultaneously modulating the glutamatergic and monoaminergic systems represents a promising strategy for treating depression. In this study, a series of multi-target antagonists targeting both NMDAR and monoamine transporters (SERT, DAT, and NET) was designed and evaluated for their antidepressant potential in vitro and in vivo. Among these heterocycle-fused phenylcyclohexylamine derivatives, compound A16 demonstrated potent and relatively balanced multi-target activity (A16: IC50(NMDAR): IC50(SERT): IC50(DAT): IC50(NET) = 1.8:1.0:1.9:1.3) compared to the lead compound S1. Pharmacokinetic studies revealed that A16 exhibited moderate clearance in microsomes and favorable oral brain exposure in mice. In vivo assessments showed that A16 and its R-isomer A17 exhibited significant antidepressant-like effects in the forced swim test and tail suspension test in mice. Notably, A17 demonstrated significant antidepressant-like effects at doses as low as 1 mg/kg, with no indication of addiction risk at 20 mg/kg. Collectively, these findings identify A17, a heterocycle-fused phenylcyclohexylamine as a promising scaffold for developing non-addictive, rapid-acting antidepressants.
Collapse
Affiliation(s)
- Fuqiang Zheng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Lingang Laboratory, Shanghai, 200031, China
| | - Zhengtao Hu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hai Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiaxin Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Jiefang Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xudong Gong
- Vigonvita Shanghai Co., Ltd., Shanghai, 201210, China
| | - Jing Ji
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nuriddinov Zayniddin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Safomuddin Abduahadi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Obul Mamateli
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Guan Wang
- Vigonvita Shanghai Co., Ltd., Shanghai, 201210, China
| | - Pengcheng Li
- Vigonvita Shanghai Co., Ltd., Shanghai, 201210, China
| | - Tianwen Hu
- Vigonvita Shanghai Co., Ltd., Shanghai, 201210, China
| | - Guanghui Tian
- Vigonvita Shanghai Co., Ltd., Shanghai, 201210, China
| | - Zhijian Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weiliang Zhu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haji Akber Aisa
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, 830011, China; University of Chinese Academy of Sciences, Beijing, 100049, China; School of Pharmacy, Xinjiang Medical University, Urumqi, 830054, China.
| | - Jingshan Shen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yang He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
5
|
Quintanilla B, Greenstein D, Tripathi A, Bartosh A, Yuan P, Zarate CA, Pillai A. Assessment of complement cascade components in patients with major depressive disorder. Brain Behav Immun 2025; 127:229-237. [PMID: 40064429 DOI: 10.1016/j.bbi.2025.03.009] [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: 10/07/2024] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
Recent evidence suggests that the rapid-acting antidepressant ketamine has immune regulatory functions. The complement system is an important component of the innate immune response and plays a key role in synaptic plasticity. An increase in complement component 3 (C3) expression was previously found in the prefrontal cortex of individuals with depression. Given the complement system's role in depression and ketamine's potential anti-inflammatory properties, there is reason to suspect overlap between the complement system and ketamine's mechanism of action. This post-hoc study analyzed data from 39 individuals with major depressive disorder (MDD) and 25 healthy volunteers who previously participated in a randomized, double-blind trial comparing intravenous ketamine (0.5 mg/kg) to placebo. Blood was obtained at baseline, 230 min, Day 1, and Day 3. Plasma levels of C3a and C4a, two key complement proteins implicated in synaptic plasticity, were determined by ELISA. Linear mixed models were used to test baseline sex differences, whether differences varied by diagnosis, and ketamine's effects (versus placebo) on C3a and C4a levels in the MDD group only. A significant diagnosis-by-sex interaction was observed for C3a but not C4a levels. Drug effects on C3a and C4a levels did not vary over time. These results suggest that treatment strategies targeting the complement pathway may yield fruitful insights and/or advances in treatment options for MDD.
Collapse
Affiliation(s)
- Brandi Quintanilla
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Dede Greenstein
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ashutosh Tripathi
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Alona Bartosh
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Peixiong Yuan
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Anilkumar Pillai
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Research and Development, Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
6
|
Hu H, Tillman AN, Fujita M, Yoshikawa M, Ballard ED, Lee Y, Zarate CA. Can ketamine and other glutamate receptor modulators be considered entactogens? Psychiatry Res 2025; 349:116513. [PMID: 40328099 PMCID: PMC12117387 DOI: 10.1016/j.psychres.2025.116513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/08/2025]
Abstract
Subanesthetic-dose ketamine has recently been reported to improve hedonic pleasures associated with social interactions and altruism in individuals with treatment-resistant depression. However, whether ketamine-among other glutamate receptor modulators-also improves empathy and/or prosocial behavior in humans remains unknown. Under a framework grounded in neurobiology that proposes that prosocial behavior is preceded by empathy, this systematic review sought to: (1) explore the entactogenic effects of glutamate receptor modulators observed in clinical trials (as either primary or secondary outcomes), and (2) synthesize the findings regarding which glutamate receptor modulators produce entactogenic effects. Thirty studies that included self-reported ratings, neuroimaging, and/or behavioral task outcomes met inclusion criteria suggesting potential entactogenic effects associated with ketamine and, to less convincing extent, d-cycloserine (DCS). The findings suggest that ketamine and DCS may modulate self- and other-perception, involving changes in activity in brain regions involved in empathetic concerns and mentalizing, the ability to understand one's own and others' thoughts and feelings. These findings may guide potential therapeutic interventions for neuropsychiatric conditions associated with impaired empathy and prosocial behavior, including mood disorders, neurodevelopmental disorders, psychotic disorders, and personality disorders.
Collapse
Affiliation(s)
- Hiroe Hu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, NIMH-NIH, 10 Center Drive, Bldg. 10, Room 7-5545, Bethesda, MD 20892, USA.
| | - Alaina N Tillman
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, NIMH-NIH, 10 Center Drive, Bldg. 10, Room 7-5545, Bethesda, MD 20892, USA
| | - Miyu Fujita
- Georgetown University School of Medicine, Washington, DC, USA
| | | | - Elizabeth D Ballard
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, NIMH-NIH, 10 Center Drive, Bldg. 10, Room 7-5545, Bethesda, MD 20892, USA
| | - Yoojin Lee
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, NIMH-NIH, 10 Center Drive, Bldg. 10, Room 7-5545, Bethesda, MD 20892, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, NIMH-NIH, 10 Center Drive, Bldg. 10, Room 7-5545, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Agnorelli C, Cinti A, Barillà G, Lomi F, Scoccia A, Benelli A, Neri F, Smeralda CL, Cuomo A, Santarnecchi E, Tatti E, Godfrey K, Tarantino F, Fagiolini A, Rossi S. Neurophysiological correlates of ketamine-induced dissociative state in bipolar disorder: insights from real-world clinical settings. Mol Psychiatry 2025; 30:2848-2859. [PMID: 39809847 PMCID: PMC12185349 DOI: 10.1038/s41380-025-02889-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Ketamine, a dissociative compound, shows promise in treating mood disorders, including treatment-resistant depression (TRD) and bipolar disorder (BD). Despite its therapeutic potential, the neurophysiological mechanisms underlying ketamine's effects are not fully understood. This study explored acute neurophysiological changes induced by subanesthetic doses of ketamine in BD patients with depression using electroencephalography (EEG) biomarkers. A cohort of 30 BD (F = 12) inpatients with TRD undergoing ketamine treatment was included in the study. EEG recordings were performed during one of the ketamine infusions with doses ranging from 0.5 to 1 mg/kg, and subjective effects were evaluated using the Clinician-Administered Dissociative States Scale (CADSS). Both rhythmic and arrhythmic features were extrapolated from the EEG signal. Patients who exhibited a clinical response to ketamine treatment within one week were classified as early responders (ER), whereas those who responded later were categorized as late responders (LR). Ketamine reduced low-frequency spectral power density while increasing gamma oscillatory power. Additionally, ketamine flattened the slope of the power spectra, indicating altered scale-free dynamics. Ketamine also increased brain signal entropy, particularly in high-frequency bands. Notably, LR exhibited greater EEG changes compared to ER, suggesting endophenotypic differences in treatment sensitivity. These findings provide valuable insights into the neurophysiological effects of ketamine in BD depression, highlighting the utility of EEG biomarkers for assessing ketamine's therapeutic mechanisms in real-world clinical settings. Understanding the neural correlates of ketamine response may contribute to personalized treatment approaches and improved management of mood disorders.
Collapse
Affiliation(s)
- Claudio Agnorelli
- Department of Molecular Medicine, Division of Psychiatry, School of Medicine, University of Siena, Siena, Italy.
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, UK.
| | - Alessandra Cinti
- Siena Brain Investigation and Neuromodulation Lab, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Giovanni Barillà
- Department of Molecular Medicine, Division of Psychiatry, School of Medicine, University of Siena, Siena, Italy
| | - Francesco Lomi
- Siena Brain Investigation and Neuromodulation Lab, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Adriano Scoccia
- Siena Brain Investigation and Neuromodulation Lab, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Alberto Benelli
- Siena Brain Investigation and Neuromodulation Lab, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Francesco Neri
- Siena Brain Investigation and Neuromodulation Lab, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Carmelo Luca Smeralda
- Siena Brain Investigation and Neuromodulation Lab, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Alessandro Cuomo
- Department of Molecular Medicine, Division of Psychiatry, School of Medicine, University of Siena, Siena, Italy
| | - Emiliano Santarnecchi
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Neurology, Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elisa Tatti
- Department of Molecular, Cellular & Biomedical Sciences, CUNY, School of Medicine, New York, NY, USA
| | - Kate Godfrey
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, UK
| | - Francesca Tarantino
- Unit of Anesthesia and Neurological Intensive Care, Department of Neurological and Motor Sciences, University of Siena, Siena, Italy
| | - Andrea Fagiolini
- Department of Molecular Medicine, Division of Psychiatry, School of Medicine, University of Siena, Siena, Italy
| | - Simone Rossi
- Siena Brain Investigation and Neuromodulation Lab, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
8
|
Yuksel B, Sen Z, Unal G. Ketamine differentially affects implicit and explicit memory processes in rats. Psychopharmacology (Berl) 2025; 242:1245-1258. [PMID: 39589435 DOI: 10.1007/s00213-024-06720-8] [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: 07/28/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024]
Abstract
RATIONALE Ketamine, a non-competitive NMDA receptor antagonist, produces antidepressant effects at subanesthetic doses. The therapeutic effect, however, is often accompanied by cognitive side effects, including memory impairments. Yet, the specific effects of ketamine on different processes of implicit and explicit memory remain to be elucidated. OBJECTIVES We examined the effect of an antidepressant dose of ketamine (10 mg/kg, IP) on the encoding, retrieval, and modulation processes of fear memory and spatial memory in adult Wistar rats. METHODS Ketamine was administered before the fear acquisition, retrieval, or extinction procedures in a Pavlovian fear conditioning task. In another set of experiments, it was administered before the training, probe trial, or reversal training phases of the Morris Water Maze (MWM). RESULTS The antidepressant dose of ketamine partially impaired fear extinction when administered before the acquisition or retrieval. In contrast, it facilitated memory modulation and decreased the escape latency in the first day of reversal training in the MWM when administered before the training or reversal training sessions. Encoding or retrieval performance in either type of memory was not affected. CONCLUSIONS These findings show that ketamine does not impair the acquisition or retrieval processes of cued fear or spatial memory; but exerts differential effects on memory modulation of these implicit and explicit memory paradigms, by disrupting fear extinction and facilitating reversal spatial learning.
Collapse
Affiliation(s)
- Bahar Yuksel
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, Istanbul, 34342, Turkey
| | - Zeynep Sen
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, Istanbul, 34342, Turkey
| | - Gunes Unal
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, Istanbul, 34342, Turkey.
| |
Collapse
|
9
|
Pałucha-Poniewiera A, Bobula B, Rafało-Ulińska A, Kaczorowska K. Depression-like effects induced by chronic unpredictable mild stress in mice are rapidly reversed by a partial negative allosteric modulator of mGlu 5 receptor, M-5MPEP. Psychopharmacology (Berl) 2025; 242:1259-1273. [PMID: 39615019 DOI: 10.1007/s00213-024-06724-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/22/2024] [Indexed: 05/17/2025]
Abstract
RATIONALE Due to the numerous limitations of ketamine as a rapid-acting antidepressant drug (RAAD), research is still being conducted to find an effective and safe alternative to this drug. Recent studies indicate that the partial mGlu5 receptor negative allosteric modulator (NAM), 2-(2-(3-methoxyphenyl)ethynyl)-5-methylpyridine (M-5MPEP), has therapeutic potential as an antidepressant. OBJECTIVES The study aimed to investigate the potential rapid antidepressant-like effect of M-5MPEP in a mouse model of depression and to determine the mechanism of this action. METHODS Chronic unpredictable mild stress (CUMS) was used as an animal model of depression. The effects of single and four-day administration of M-5MPEP on CUMS-induced animal behaviors reflecting anhedonia, apathy, and helplessness were studied. Western blot was applied to measure the levels of proteins potentially involved in a rapid antidepressant effect, including mammalian target of rapamycin (mTOR), eukaryotic elongation factor 2 (eEF2), tropomyosin receptor kinase B (TrkB), and serotonin transporter (SERT), both in the hippocampus and the prefrontal cortex (PFC). Furthermore, excitatory synaptic transmission and long-term potentiation (LTP) were measured in the medial PFC (mPFC). RESULTS We showed that M-5MPEP administration for four consecutive days abolished CUMS-induced apathy- and anhedonia-like symptoms in a mouse model of depression. We also found that these effects were accompanied by changes in hippocampal TrkB levels and mTOR and eEF2 levels in the PFC. Using electrophysiological techniques, we showed that the four-day M-5MPEP treatment reversed chronic stress-induced increases in excitatory synaptic potential and CUMS-impaired LTP in the mPFC. CONCLUSIONS Partial mGlu5 receptor NAM, M-5MPEP, appears to be a potentially effective new RAAD and deserves further study.
Collapse
Affiliation(s)
- Agnieszka Pałucha-Poniewiera
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland.
| | - Bartosz Bobula
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
| | - Anna Rafało-Ulińska
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
| | - Katarzyna Kaczorowska
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
| |
Collapse
|
10
|
Chen M, Jin J, Bi H, Zhang Y, Sun M, Li X, Wang Y. Advances in the study of NMDA receptors in depression pathogenesis and the antidepressant efficacy of their antagonists. Asian J Psychiatr 2025; 108:104502. [PMID: 40300235 DOI: 10.1016/j.ajp.2025.104502] [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: 01/31/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025]
Abstract
N-methyl-D-aspartate receptors (NMDA receptors) play a crucial role as ionotropic glutamate receptors in regulating neuroplasticity, learning, memory, and a range of psychiatric disorders. Studies indicate that dysfunction of NMDA receptors is a key pathological mechanism in depression, where abnormal activation can result in neuronal excitotoxicity, excessive extracellular calcium ion accumulation, and disrupted neuroplasticity. As a non-competitive NMDA receptor antagonist, ketamine quickly relieves depressive symptoms by decreasing the activity of extracellular NMDA receptors and activating the mTOR signaling pathway. The treatment can improve severe depression and suicide thoughts within hours, but its potential for hallucinations, dissociative symptoms, and dependency restricts its broader application. Esketamine has demonstrated improvements in both side effects and efficacy and has received FDA approval, while other compounds with NMDA receptor modulating functions, such as memantine and rapastinel, are also showing potential in exploration. Future studies should concentrate on the molecular mechanisms of NMDA receptors, aiming to develop safer and more effective medications, and refine treatment strategies to offer personalized choices and longer-lasting efficacy for the treatment of depression.
Collapse
Affiliation(s)
- Mingrui Chen
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Jingyan Jin
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Hongsheng Bi
- Department of Psychiatry, The First Hospital of China Medical University, China; The third hospital of Daqing, Psychiatric Ward No. 9, China
| | - Yihan Zhang
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Mingyuan Sun
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Xiaobai Li
- Department of Psychiatry, The First Hospital of China Medical University, China.
| | - Yan Wang
- Center for Psychological Development, China Medical University, China.
| |
Collapse
|
11
|
Camargo A, Nilsson A, Shariatgorji R, Appleton E, Branzell N, Doyon D, Giovenzana M, Zhang X, Dautan D, Andren PE, Svenningsson P. Enduring modulation of dorsal raphe nuclei regulates (R,S)-ketamine-mediated resilient stress-coping behavior. Mol Psychiatry 2025; 30:2504-2516. [PMID: 39592824 DOI: 10.1038/s41380-024-02853-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024]
Abstract
Ketamine may be a novel pharmacologic approach to enhance resilience and protect against stress-related disorders, but the molecular targets underlying this response remain to be fully characterized. The multifunctional protein p11 is crucial in the pathophysiology of depression and antidepressant responses. However, it is still unclear whether p11 plays a role in the pro-resilience effects induced by ketamine. Here, we demonstrated that prophylactic administration of ketamine buffers passive stress-induced maladaptive phenotypes induced by chronic stress exposure. Spatial neurotransmitter and metabolite analysis revealed that prophylactic ketamine was also effective in blunting stress-induced disturbances of tryptophan metabolism in dorsal raphe nuclei (DRN). Additionally, we demonstrated that ketamine prevented chronic restraint stress-induced p11 reduction in DRN, a highly p11-enriched region. Furthermore, we provide novel evidence indicating that p11 deficiency regulates susceptibility to stress-induced depression-related phenotypes, and these behavioral maladaptations are dependent, at least in part, on p11 function in serotonergic neurons. Spatial neurotransmitter and metabolite analysis also showed a reduction of tryptophan and dopamine metabolism in DRN of serotonergic p11-deficient mice. Viral-mediated downregulation of p11 within DRN induced a stress-susceptible phenotype. Finally, our results also unveiled that the ability of ketamine to elicit a pro-resilience response against stress-induced maladaptive phenotypes was occluded when p11 was selectively deleted in serotonergic neurons. Altogether, we showed a previously unexplored role of the DRN circuit in regulating stress susceptibility and resilience-enhancing actions of ketamine.
Collapse
Affiliation(s)
- Anderson Camargo
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Anna Nilsson
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Reza Shariatgorji
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ellen Appleton
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Niclas Branzell
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Doyon
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mattia Giovenzana
- Department of Medicine and Surgery, University of Milano Bicocca Monza, Monza, Italy
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Dautan
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Per E Andren
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
12
|
Kharasch ED. The Nine Lives of Ketamine: From CI-581 to Present Day Clinical Application-Commentary on Domino et al. Clin Pharmacol Ther 2025; 117:1499-1515. [PMID: 40388065 DOI: 10.1002/cpt.3634] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/24/2025] [Indexed: 05/20/2025]
Abstract
The classic article by Domino et al. was the first clinical report and presented the first-in-human study of what would become the drug ketamine. Ketamine has become an analgesic, anesthetic, antidepressant, and drug of misuse. It remains today the only "dissociative anesthetic". It has challenged classical pharmacologic theory and drug development paradigms. The first report on CI-581 could never have envisioned the pharmacologic complexity or clinical utility of this molecule.
Collapse
Affiliation(s)
- Evan D Kharasch
- Department of Anesthesiology, Duke University, Durham, North Carolina, USA
- Bermaride LLC, Durham, North Carolina, USA
| |
Collapse
|
13
|
Riggs LM, Aronson S, Mou TCM, Pereira EFR, Thompson SM, Gould TD. Bioactive ketamine metabolite exerts in vivo neuroplastogenic effects to improve hippocampal function in a treatment-resistant depression model. Cell Rep 2025; 44:115743. [PMID: 40408248 DOI: 10.1016/j.celrep.2025.115743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/04/2025] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
An acute increase in excitatory synaptic transmission contributes to the rapid antidepressant actions of neuroplastogens, including ketamine and its bioactive metabolite, (2R,6R)-hydroxynorketamine (HNK). It is hypothesized that drug-induced metaplastic changes in synaptic strength account for therapeutically relevant behavioral adaptations in vivo. Using the plasticity-deficient Wistar Kyoto model of treatment-resistant depression, we demonstrate that (2R,6R)-HNK potentiates glutamatergic transmission, promotes synaptic strength, restores long-term potentiation (LTP), and reverses deficits in hippocampal-dependent synaptic activity and behavior. (2R,6R)-HNK selectively potentiated CA1 pyramidal neuron activity during novelty exploration and restored Schaffer collateral-dependent spatial recognition memory. Prior experience with spatial learning partially occluded LTP in control rats, an effect mimicked in LTP-impaired rats in which spatial learning deficits were reversed by (2R,6R)-HNK. These findings demonstrate that (2R,6R)-HNK exerts rapid neuroplastogenic effects in vivo, which improve cognitive function and promote adaptive changes in synaptic strength at functionally impaired synapses.
Collapse
Affiliation(s)
- Lace M Riggs
- Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Ta-Chung M Mou
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Edna F R Pereira
- Department of Pharmacology and Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Pharmacology and Physiology, 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 and Physiology, 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
|
14
|
Nagele P, Conway CR, Zorumski CF. Purposeful Drug Repurposing. JAMA Psychiatry 2025:2833556. [PMID: 40366672 DOI: 10.1001/jamapsychiatry.2025.0900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
This Viewpoint discusses a proposal to identify candidates for drug repurposing and allocate US Food and Drug Administration funds to cover the cost of evaluating and approving such drugs for their novel indications.
Collapse
Affiliation(s)
- Peter Nagele
- Department of Anesthesia & Critical Care, University of Chicago, Chicago, Illinois
| | - Charles R Conway
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, Missouri
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
15
|
Ammendolia I, Mannucci C, Esposito E, Calapai G, Currò M, Midiri P, Mondello C, Cardia L, Calapai F. Safety Profile and Suicidality Associated with the Use of Esketamine in the Treatment of Major Depressive Disorder in European Countries: An EudraVigilance Database Analysis. Pharmaceuticals (Basel) 2025; 18:702. [PMID: 40430521 PMCID: PMC12114692 DOI: 10.3390/ph18050702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/27/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objective: Major depressive disorder (MDD) is a common mental disorder, with a significant portion of patients developing treatment-resistant depression (TRD). Esketamine is an antagonist of the N-methyl-D-aspartate receptor indicated as a nasal spray in combination with other antidepressants for adults with TRD. Signals of suspected adverse reactions (SARs) to esketamine from the EudraVigilance database in European countries were analyzed for a more defined safety profile of this drug in the real world. Methods: SARs to esketamine reported in the data system EudraVigilance were analyzed, and disproportionality analysis for adverse reactions indicating suicidality for esketamine, in comparison to the antidepressants fluoxetine and venlafaxine, was performed. Results: Increases in blood pressure (15.4%) and dissociation (15.0%) were the most frequently reported SARs. The sex distribution indicates the prevalence of women, except for increased blood pressure and completed suicide, which were signaled in men, while adults (18-64 years) and elders (65-85 years) were the ages with the largest number of reported adverse reactions to esketamine. The results indicate the existence of a potential increase in the risk of suicide in depressed patients taking esketamine when compared with fluoxetine and venlafaxine. Conclusions: Apart from carefulness due to the known limitations of pharmacovigilance research conducted by using data systems of spontaneous signals for SARs, the analysis of data points toward the need for greater attention being paid to the potential risk of suicide following the prescription of esketamine in depressed subjects. In this regard, as regulatory agencies also recommend, patients with a history of suicide-related events or those exhibiting a significant degree of suicidal ideation prior to beginning treatment should receive more careful monitoring during treatment.
Collapse
Affiliation(s)
- Ilaria Ammendolia
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy; (I.A.); (E.E.)
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.C.); (P.M.)
| | - Carmen Mannucci
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, 98122 Messina, Italy; (C.M.); (C.M.); (F.C.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy; (I.A.); (E.E.)
| | - Gioacchino Calapai
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy; (I.A.); (E.E.)
| | - Mariaconcetta Currò
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.C.); (P.M.)
| | - Paola Midiri
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.C.); (P.M.)
| | - Cristina Mondello
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, 98122 Messina, Italy; (C.M.); (C.M.); (F.C.)
| | - Luigi Cardia
- Department of Human Pathology of Adult and Childhood “Gaetano Barresi”, University of Messina, 98122 Messina, Italy;
| | - Fabrizio Calapai
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, 98122 Messina, Italy; (C.M.); (C.M.); (F.C.)
| |
Collapse
|
16
|
Lucantonio F, Roeglin J, Li S, Lu J, Shi A, Czerpaniak K, Fiocchi FR, Bontempi L, Shields BC, Zarate CA, Tadross MR, Pignatelli M. Ketamine rescues anhedonia by cell-type- and input-specific adaptations in the nucleus accumbens. Neuron 2025; 113:1398-1412.e4. [PMID: 40112815 PMCID: PMC12064382 DOI: 10.1016/j.neuron.2025.02.021] [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: 08/08/2024] [Revised: 01/09/2025] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Ketamine is recognized as a rapid and sustained antidepressant, particularly for major depression unresponsive to conventional treatments. Anhedonia is a common symptom of depression for which ketamine is highly efficacious, but the underlying circuits and synaptic changes are not well understood. Here, we show that the nucleus accumbens (NAc) is essential for ketamine's effect in rescuing anhedonia in mice subjected to chronic stress. Specifically, a single exposure to ketamine rescues stress-induced decreased strength of excitatory synapses on NAc-D1 dopamine receptor-expressing medium spiny neurons (D1-MSNs). Using a cell-specific pharmacology method, we establish the necessity of this synaptic restoration for the sustained therapeutic effects of ketamine on anhedonia. Examining causal sufficiency, artificially increasing excitatory synaptic strength onto D1-MSNs recapitulates the behavioral amelioration induced by ketamine. Finally, we used opto- and chemogenetic approaches to determine the presynaptic origin of the relevant synapses, implicating monosynaptic inputs from the medial prefrontal cortex and ventral hippocampus.
Collapse
Affiliation(s)
- Federica Lucantonio
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jacob Roeglin
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Shuwen Li
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Jaden Lu
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Aleesha Shi
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Katherine Czerpaniak
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Francesca R Fiocchi
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | | | - Brenda C Shields
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Michael R Tadross
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Marco Pignatelli
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
17
|
Brown KA, Ajibola MI, Gould TD. Rapid hippocampal synaptic potentiation induced by ketamine metabolite (2R,6R)-hydroxynorketamine persistently primes synaptic plasticity. Neuropsychopharmacology 2025; 50:928-940. [PMID: 40097740 PMCID: PMC12032166 DOI: 10.1038/s41386-025-02085-4] [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: 10/24/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
The pharmacologically active (R,S)-ketamine (ketamine) metabolite (2 R,6 R)-hydroxynorketamine (HNK) maintains ketamine's preclinical antidepressant profile without adverse effects. While hypotheses have been proposed to explain how ketamine and its metabolites initiate their antidepressant-relevant effects, it remains unclear how sustained therapeutic actions arise following drug elimination. To distinguish the physiological mechanisms involved in the rapid from sustained actions of HNK, we utilized extracellular electrophysiology combined with pharmacology to develop an in vitro hippocampal slice incubation model that exhibited pharmacological fidelity to the 1) rapid synaptic potentiation induced by HNK at the Schaffer collateral-CA1 (SC-CA1) synapse during bath-application to slices collected from mice, and 2) maintenance of metaplastic (priming) activity that enhanced N-methyl-D-aspartate receptor (NMDAR) activation-dependent long-term potentiation (LTP) hours after in vivo dosing. We used this model to reveal novel mechanisms engaged in HNK's temporally-sensitive antidepressant-relevant synaptic actions, finding that the induction of synaptic potentiation by HNK did not require NMDAR activity, but NMDAR activity was necessary to maintain synaptic priming. HNK required protein kinase A (PKA) activity to rapidly potentiate SC-CA1 neurotransmission to facilitate synaptic priming that persistently promoted LTP formation. HNK's rapid actions were blocked by inhibitors of adenylyl cyclase 1 (AC1), but not an AC5 inhibitor. We conclude that HNK rapidly potentiates SC-CA1 synaptic efficacy, which then stimulates priming mechanisms that persistently favor plasticity. Targeting such priming mechanisms may be an effective antidepressant strategy, and our incubation model may aid in revealing novel pharmacological targets.
Collapse
Affiliation(s)
- Kyle A Brown
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Musa I Ajibola
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Todd D Gould
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
- Department of Pharmacology and Physiology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
- Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, MD, 21201, USA.
- Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA.
| |
Collapse
|
18
|
Kheirkhah M, Duncan WC, Yuan Q, Wang PR, Jamalabadi H, Leistritz L, Walter M, Goldman D, Zarate CA, Hejazi NS. REM density predicts rapid antidepressant response to ketamine in individuals with treatment-resistant depression. Neuropsychopharmacology 2025; 50:941-946. [PMID: 39955416 PMCID: PMC12032024 DOI: 10.1038/s41386-025-02066-7] [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: 10/10/2024] [Revised: 01/06/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Abnormalities during rapid eye movement (REM) sleep contribute to the pathophysiology of major depressive disorder (MDD), but few studies have explored the relationship between REM sleep and treatment-resistant depression (TRD). In MDD, REM sleep abnormalities often manifest as alterations in total night REM Density (RD), RD in the first REM period (RD1), and REM Latency (RL). Among these, RD1 is notably considered a potential endophenotype of depression. This study compared REM sleep markers between 63 drug-free individuals with TRD (39 F/24 M) and 41 healthy volunteers (25 F/16 M). It also investigated the effects of ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, on these REM sleep variables. Specifically, the study investigated whether RD1 could predict antidepressant response to ketamine. TRD participants showed higher RD1 and shorter RL at baseline compared to HVs, as assessed via non-parametric tests, but Total Night RD did not differ between the two groups. Ketamine treatment decreased RD1 in TRD participants but did not affect Total Night RD or RL. As assessed via the Support Vector Machine (SVM) algorithm, baseline RD1 level moderately predicted antidepressant response to ketamine versus non-response (area under the receiver operating characteristic (ROC) curve (AUC) = 0.73, with a median accuracy of 0.75), wherein TRD participants with higher baseline RD1 were more likely to respond to ketamine. These results underscore the utility of RD1 for identifying individuals most likely to benefit from ketamine treatment, enabling more targeted and effective therapeutic strategies. Clinical Trials Identifier: NCT00088699, NCT01204918.
Collapse
Affiliation(s)
- Mina Kheirkhah
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Wallace C Duncan
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Qiaoping Yuan
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Philip R Wang
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
| | - Hamidreza Jamalabadi
- Department of Psychiatry and Psychotherapy, Philipps University of Marburg, Marburg, Germany
| | - Lutz Leistritz
- Institute of Medical Statistics, Computer and Data Sciences, Jena University Hospital, Jena, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - David Goldman
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Nadia S Hejazi
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
19
|
Witkin JM, Smith JL, Radin DP. Is there a continued need for behavioral pharmacology in the discovery and development of medicines? J Pharmacol Exp Ther 2025; 392:103571. [PMID: 40305918 DOI: 10.1016/j.jpet.2025.103571] [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: 12/16/2024] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Since their introduction in the 1950s by Peter Dews, behavioral pharmacology laboratories have collected experimental data for discovery, approval, and regulation of many new and improved therapeutic agents across a host of therapeutic domains. The ethical use of animals in research requires evaluation of the value of animal data to justify their use. The present commentary provides a discussion of the risk-benefit analysis of the use of behavioral methods in drug discovery and drug development using antidepressants as an example. It is concluded that there are some misconceptions both within and outside drug discovery organizations as to the value of behavioral methods. We argue that there are no fully functional disease models of psychiatric disorders. The methods used in behavioral pharmacology are best viewed as whole-animal biological readouts of the actions of drugs. Behavioral pharmacology data, placed in the context of other pharmacological findings, enable the construction of confidence levels regarding the hypothesis that a new chemical entity will provide therapeutic benefit at tolerable and safe dose levels. The exclusion of behavioral pharmacology would diminish the ability of the drug discovery data set to provide needed levels of assurance that the efficacy and safety of the new chemical entity will translate into patients. New methodologies for the discovery of medicines are being developed (eg, tissue on chips and artificial intelligence paradigms). However, until nonbehavioral procedures are validated, the absence of behavioral pharmacology in drug discovery research shifts the risk of making new medicines onto the health of patients. SIGNIFICANCE STATEMENT: The pros and cons of using behavioral pharmacology in the discovery and development of new medicines are discussed.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, Indiana; Departments of Neuroscience and Trauma Research, Ascension St. Vincent Hospital, Indianapolis, Indiana; RespireRx Pharmaceuticals Inc, Glen Rock, New Jersey.
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent Hospital, Indianapolis, Indiana
| | | |
Collapse
|
20
|
Mastrodonato A, Jin M, Kee N, Lanio M, Tapia J, Quintana L, Muñoz Zamora A, Deng SX, Xu X, Landry DW, Denny CA. Prophylactic (R,S)-Ketamine and (2S,6S)-Hydroxynorketamine Decrease Fear Expression by Differentially Modulating Fear Neural Ensembles. Biol Psychiatry 2025; 97:887-899. [PMID: 39389408 PMCID: PMC11978926 DOI: 10.1016/j.biopsych.2024.09.024] [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: 12/21/2023] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND We previously reported that a single injection of (R,S)-ketamine or its metabolite (2S,6S)-hydroxynorketamine (HNK) prior to stress attenuated learned fear. However, whether these drugs attenuate learned fear through divergent or convergent effects on neural activity remains to be determined. METHODS 129S6/SvEv male mice were injected with saline, (R,S)-ketamine, or (2S,6S)-HNK 1 week before a 3-shock contextual fear conditioning paradigm. Five days later, mice were re-exposed to the aversive context and euthanized 1 hour later to quantify active cells. Brains were processed for c-fos immunoreactivity, and neural networks were built with a novel, wide-scale imaging pipeline. RESULTS We found that (R,S)-ketamine and (2S,6S)-HNK attenuated learned fear. Fear-related neural activity was altered in dorsal CA3 following (2S,6S)-HNK; ventral CA3 and CA1, infralimbic and prelimbic regions, insular cortex, retrosplenial cortex, piriform cortex, nucleus reuniens, and periaqueductal gray following both (R,S)-ketamine and (2S,6S)-HNK; and in the paraventricular nucleus of the thalamus (PVT) following (R,S)-ketamine. Dorsal CA3 and ventral hippocampus activation correlated with freezing in the (R,S)-ketamine group, and retrosplenial cortex activation correlated with freezing in both (R,S)-ketamine and (2S,6S)-HNK groups. (R,S)-ketamine increased connectivity between cortical and subcortical regions while (2S,6S)-HNK increased connectivity within these regions. CONCLUSIONS This work identifies novel nodes in fear networks that involve the nucleus reuniens, piriform cortex, insular cortex, periaqueductal gray, and retrosplenial cortex that can be targeted with neuromodulatory strategies or pharmaceutical compounds to treat fear-induced disorders. This approach could be used to optimize target engagement and dosing strategies of existing medications.
Collapse
Affiliation(s)
- Alessia Mastrodonato
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York; MIND Area, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York.
| | - Michelle Jin
- Neurobiology and Behavior Graduate Program, Columbia University, New York, New York; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, New York
| | - Noelle Kee
- Department of Neuroscience and Behavior, Barnard College, New York, New York
| | - Marcos Lanio
- Neurobiology and Behavior Graduate Program, Columbia University, New York, New York; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, New York
| | - Juliana Tapia
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York
| | - Liliette Quintana
- Department of Neuroscience and Behavior, Barnard College, New York, New York
| | - Andrea Muñoz Zamora
- Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York
| | - Shi-Xian Deng
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Organic Chemistry Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Xiaoming Xu
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Organic Chemistry Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Donald W Landry
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Organic Chemistry Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Christine A Denny
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York.
| |
Collapse
|
21
|
Zhang Y, Song Z, Zhang H, Lin H, Xu P, Li Z, He Q, Wei B. Advancing Antidepressive Agents: Drug Discovery and Polymer-Based Drug Delivery Systems for Improved Treatment Outcome. Biomedicines 2025; 13:1081. [PMID: 40426909 PMCID: PMC12109423 DOI: 10.3390/biomedicines13051081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/14/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Depressive disorder (a subclass of mental disorders) is characterized by persistent affective symptoms. Without timely therapeutic intervention, it leads to clinical deterioration manifested as reduced quality of life and may increase suicide risk in severe cases. Given its complex etiology, intertwined with intrinsic factors such as genetics and environment, and impacted by various issues such as first-pass effect and blood-brain barrier, the therapeutic efficacy of many antidepressant medications is limited for patients. Therefore, by delving into the exploration of novel antidepressant drugs and biomaterials, this review aims to offer fresh perspectives that may facilitate the discovery of innovative antidepressant medications and enhance their therapeutic outcomes. Notably, the review highlights polymers' crucial role in enhancing antidepressants' pharmacological efficacy and pharmacokinetic properties by optimizing their parameters, and they will undoubtedly become powerful tools in improving antidepressive outcomes in future research.
Collapse
Affiliation(s)
- Yufei Zhang
- Department of Pharmacy, Jinqiu Hospital of Liaoning Province, Shenyang 110122, China;
| | - Zengyi Song
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Z.S.); (H.Z.); (H.L.); (P.X.); (Z.L.); (Q.H.)
| | - Hongxi Zhang
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Z.S.); (H.Z.); (H.L.); (P.X.); (Z.L.); (Q.H.)
| | - Haijiao Lin
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Z.S.); (H.Z.); (H.L.); (P.X.); (Z.L.); (Q.H.)
| | - Pu Xu
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Z.S.); (H.Z.); (H.L.); (P.X.); (Z.L.); (Q.H.)
| | - Zijia Li
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Z.S.); (H.Z.); (H.L.); (P.X.); (Z.L.); (Q.H.)
| | - Qingyun He
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Z.S.); (H.Z.); (H.L.); (P.X.); (Z.L.); (Q.H.)
| | - Binbin Wei
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Z.S.); (H.Z.); (H.L.); (P.X.); (Z.L.); (Q.H.)
| |
Collapse
|
22
|
Veraart JK, Smith-Apeldoorn SY, van der Meij A, Spijker J, Schoevers RA, Kamphuis J. Oral esketamine for patients with severe treatment-resistant depression: Effectiveness, safety, and tolerability of a six-week open-label treatment program. J Psychopharmacol 2025:2698811251332831. [PMID: 40285334 DOI: 10.1177/02698811251332831] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
BACKGROUND Oral esketamine for patients with treatment-resistant depression (TRD) could offer certain advantages over other routes, such as intravenous or intranasal, but it has not been systematically studied in a real-world setting. AIMS Here we present results from a relatively large naturalistic study to evaluate the effectiveness, tolerability, and safety of oral esketamine in patients with TRD. METHODS One hundred eighty-five adults with severe TRD (average of 8.1 antidepressant trials plus electroconvulsive therapy in 63% without beneficial outcome) received oral esketamine treatment twice-weekly for 6 weeks with individually titrated doses ranging from 0.5 to 3 mg/kg. Outcome measures included change from baseline to week 6 on the Hamilton Depression Rating Scale (HDRS17), Minimal Clinically Important Difference (MCID), response, remission, self-reported symptom improvement, functioning, and side effects. RESULTS Oral esketamine treatment improved depressive symptom severity on the HDRS17 from 21.2 to 15.8 (p < 0.001). MCID, response, and remission rates were 47.1%, 26.8% and 15.6% respectively. In 45.9% of participants, treatment was continued after 6 weeks to maintain initial positive effects. Side effects were reported frequently but were overall well tolerated. The drop-out rate was 7.6%. We found no significant adverse effects associated with urinary tract or cognition. CONCLUSIONS Repeated treatment with oral esketamine is effective in improving depressive symptom severity in highly treatment-resistant depressive patients. It is safe, well tolerated, and patient-friendly. Considering the level of treatment resistance, outcomes were in the range of studies investigating other routes of (es)ketamine administration.
Collapse
Affiliation(s)
- Jolien Ke 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
| | - Sanne Y Smith-Apeldoorn
- 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
| | - Robert A Schoevers
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Behavioural Science Institute, University of Groningen, Groningen, The Netherlands
| | - Jeanine Kamphuis
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
23
|
Seo MK, Kim H, Choi AJ, Seog DH, Kho WG, Park SW, Lee JG. Effects of tianeptine on mTORC1-mediated neuronal autophagy in primary rat hippocampal neurons under nutrient deprivation. Sci Rep 2025; 15:14488. [PMID: 40280952 PMCID: PMC12032415 DOI: 10.1038/s41598-025-92988-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/04/2025] [Indexed: 04/29/2025] Open
Abstract
The aim of this study was to investigate the effects of the antidepressant tianeptine on the mechanistic target of rapamycin complex 1(mTORC1)-mediated autophagy pathway in primary hippocampal neurons exposed to B27-deprived conditions. When primary hippocampal neurons were treated with tianeptine at doses of 1, 10, 50, and 100 µM for 3 days under B27-deprived conditions, we observed that it activated autophagy and increased the formation of autophagosomes through the upregulation of autophagic proteins, including autophagy-activating kinase 1 (ULK1), Beclin 1, LC3B-II/I, and p62. And at a concentration of 100 µM tianeptine, the decrease in mTORC1 phosphorylation induced by B27 deprivation was significantly reversed. Changes in the expression of autophagic proteins induced by B27 deprivation were reversed by tianeptine treatment in a concentration-dependent manner, and tianeptine significantly reduced the increase in LC3B membrane number induced by B27 deprivation, an effect that was blocked by pretreatment with rapamycin. In conclusion, tianeptine attenuated the activity of mTORC1-mediated autophagy in primary rat hippocampal neurons under B27-deprived conditions. These results may suggest a novel mechanism by which tianeptine may affect autophagy in neurons.
Collapse
Affiliation(s)
- Mi Kyoung Seo
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Hyewon Kim
- Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Inje University, Busan, 48108, Republic of Korea
| | - Ah Jeong Choi
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
| | - Dae-Hyun Seog
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea
- Department of Biochemistry, College of Medicine, Inje University, Busan, 47392, Republic of Korea
- Dementia and Neurodegenerative Disease Research Center, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Weon-Gyu Kho
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
- Department of Parasitology, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Sung Woo Park
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea.
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea.
| | - Jung Goo Lee
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea.
- Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Inje University, Busan, 48108, Republic of Korea.
| |
Collapse
|
24
|
Nist AN, Walsh SJ, Shahan TA. Cognitive flexibility of male rats is increased by augmented punishment in a reversal learning task but ketamine has no detectable long-term effects. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06794-y. [PMID: 40263162 DOI: 10.1007/s00213-025-06794-y] [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: 08/01/2024] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
RATIONALE The probabilistic reversal learning task (PRL) is sometimes used in the context of major depressive disorder (MDD) to assess impairments in cognitive flexibility and feedback sensitivity because behavior in the task is sensitive to pharmacological interventions. Because traditional antidepressants are limited in their effectiveness, new drugs are needed to combat MDD. Ketamine has recently been investigated in the context of probabilistic reversal learning (PRL), but findings regarding its therapeutic efficacy have been mixed. One reason for this could be that almost all non-human versions of the PRL use signaled reward omission (i.e., timeout) as the punishing stimulus. It has long been known that timeout periods do not always function as punishers, and the inclusion of a known effective punishing stimulus could help to produce results of improved translational value. OBJECTIVE The present experiment sought to examine the effects of ketamine in the PRL when electric footshocks accompanied timeout periods or not. METHODS A baseline of PRL performance was established with 40 rats in which typical timeouts followed non-rewarded trials. In Phase 2, half the rats also received probabilistic footshock punishment for non-rewarded trials, while the other half continued under baseline conditions. Finally, a single dose of ketamine was administered to half of the rats (n = 10) in each condition (i.e., shock and no shock). RESULTS Shock punishment increased behavioral persistence and cognitive flexibility in the PRL, but ketamine had no effect beyond causing acute impairments. CONCLUSION These results suggest that the conditions of punishment during the PRL can have a significant impact on performance in the task and corroborate previous findings that ketamine may not impact cognitive flexibility or reward processing in healthy rats.
Collapse
Affiliation(s)
- Anthony N Nist
- Department of Psychology, Utah State University, 1600 Old Main Hill, Logan, UT, 84322, USA.
| | - Stephen J Walsh
- Department of Mathematics and Statistics, Utah State University, 1600 Old Main Hill, Logan, UT, 84322, USA
| | - Timothy A Shahan
- Department of Psychology, Utah State University, 1600 Old Main Hill, Logan, UT, 84322, USA
| |
Collapse
|
25
|
Woody ML, Rohac R, Cooper I, Griffo A, McDonald N, Spotts C, Fournier J, Jones N, Peciña M, Young K, Shivanekar S, Rengasamy M, Grafton B, Price RB. The Impact of Intravenous Ketamine on Attentional Bias: Probing Mechanisms of Rapid-Acting Antidepressant Effects in Two Clinical Studies. Biol Psychiatry 2025; 97:835-842. [PMID: 39581291 PMCID: PMC11954668 DOI: 10.1016/j.biopsych.2024.10.024] [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: 03/21/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Ketamine is known for its rapid antidepressant effect, but its impact on affective information processing (including attentional bias [AB], a putative cognitive mechanism of depression) remains largely unexplored. We leveraged a novel measurement of AB and sought to 1) establish adequate test-retest reliability and validity among participants with depression prior to ketamine treatment and 2) harness a single dose of ketamine to assess mechanistic shifts in AB and their relationship to antidepressant efficacy. METHODS A novel dual probe video task was used to index AB toward sad film clips. In study 1, treatment-seeking adults with moderate-to-severe depression (N = 40) completed the task at baseline, 1-week retest, and 1-month retest; a subset of participants (n = 15) also performed the task at 24 hours postketamine infusion (0.5 mg/kg over 40 minutes). In study 2, participants (N = 43) completed the task pre- and 24 hours postketamine. RESULTS Indices from the novel AB task were stable prior to ketamine, demonstrating good 1-week and 1-month test-retest reliability. Participants in both studies exhibited a robust reduction in AB from pre- to 24 hours postketamine infusion. In study 1, cross-sectional correlations were observed between AB and clinician-rated depressive symptoms at each pretreatment assessment. In study 2, changes in AB were correlated with improved symptoms from pre- to postinfusion. CONCLUSIONS Results provide evidence for the validity of a novel, psychometrically robust measure of AB among individuals with depression. Findings indicate that ketamine reliably and rapidly reduces AB, offering insight into a replicable, potential cognitive mechanism involved in its antidepressant action.
Collapse
Affiliation(s)
- Mary L Woody
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Rebecca Rohac
- School of Public Health, Boston University, Boston, Massachusetts
| | - Iya Cooper
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Angela Griffo
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nastasia McDonald
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Crystal Spotts
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jay Fournier
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, Ohio
| | - Neil Jones
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marta Peciña
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kymberly Young
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sharvari Shivanekar
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Manivel Rengasamy
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ben Grafton
- School of Psychological Science, The University of Western Australia, Perth, Australia
| | - Rebecca B Price
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
26
|
Bertholomey ML, Forbes C, McElroy BD, Torregrossa MM. Sex specific effects of ketamine, but not other glutamate receptor modulators, on ethanol self-administration and reinstatement of ethanol seeking in rats. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06782-2. [PMID: 40198329 DOI: 10.1007/s00213-025-06782-2] [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: 09/12/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025]
Abstract
RATIONALE Alcohol use and major depressive disorder are frequently comorbid, with individuals diagnosed with a substance use disorder being nearly three times as likely to have major depression. Poor treatment responses are found for both disorders and are further complicated when they co-occur, underscoring the need for better therapies. One potential candidate is ketamine, which has been shown to have rapid and long-lasting effects in individuals with treatment-resistant depression and, in some studies, reduces drinking in alcohol use disorder. However, though women are more likely to have this comorbidity, few studies have examined sex-specific effects of ketamine on alcohol drinking, nor have studies assessed the potential for ketamine to reduce reinstatement of alcohol seeking. OBJECTIVES The primary goal of the present studies was to determine the effects of ketamine on alcohol-motivated behaviors in male and female rats, including in a model of stress + cue-induced reinstatement of alcohol seeking using yohimbine (YOH). RESULTS We found a selective reduction in alcohol self-administration and YOH + cue-induced reinstatement in females, but not males at a dose of 10 mg/kg ketamine. However, the same dose of ketamine was effective in reducing YOH + cue-induced reinstatement of saccharin seeking in both sexes. In addition, a different NMDAR antagonist, memantine, was effective in reducing alcohol seeking in both sexes, while the ketamine metabolite hydroxynorketamine (HNK) had no effects. CONCLUSIONS In summary, these data suggest that antagonism of NMDARs may be effective in reducing stress-related alcohol seeking, but that ketamine has unique properties that lead to female-specific effects on alcohol seeking.
Collapse
Affiliation(s)
- Megan L Bertholomey
- Department of Psychiatry, Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Psychology and Neuroscience Program, Allegheny College, Meadville, PA, 16335, USA
| | - Camryn Forbes
- Department of Psychiatry, Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Bryan D McElroy
- Department of Psychiatry, Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Neural Sciences, Temple University, Philadelphia, PA, 19140, USA
| | - Mary M Torregrossa
- Department of Psychiatry, Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
27
|
Cichon J, Joseph TT, Lu X, Wasilczuk AZ, Kelz MB, Mennerick SJ, Zorumski CF, Nagele P. Nitrous oxide activates layer 5 prefrontal neurons via SK2 channel inhibition for antidepressant effect. Nat Commun 2025; 16:2999. [PMID: 40180931 PMCID: PMC11968965 DOI: 10.1038/s41467-025-57951-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Nitrous oxide (N2O) induces rapid and durable antidepressant effects. The cellular and circuit mechanisms mediating this process are not known. Here we find that a single dose of inhaled N2O induces rapid and specific activation of layer V (L5) pyramidal neurons in the cingulate cortex of rodents exposed to chronic stress conditions. N2O-induced L5 activation rescues a stress-associated hypoactivity state, persists following exposure, and is necessary for its antidepressant-like activity. Although NMDA-receptor antagonism is believed to be a primary mechanism of action for N2O, L5 neurons activate even when NMDA-receptor function is attenuated through both pharmacological and genetic approaches. By examining different molecular and circuit targets, we identify N2O-induced inhibition of calcium-sensitive potassium (SK2) channels as a key molecular interaction responsible for driving specific L5 activity along with ensuing antidepressant-like effects. These results suggest that N2O-induced L5 activation is crucial for its fast antidepressant action and this effect involves novel and specific molecular actions in distinct cortical cell types.
Collapse
Affiliation(s)
- Joseph Cichon
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Thomas T Joseph
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xinguo Lu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrzej Z Wasilczuk
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Max B Kelz
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven J Mennerick
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter Nagele
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, USA
| |
Collapse
|
28
|
Wei Q, Li M, Du Q, Zhang H, Liang Y, Cheng C, Mei B, Yang X, Fan Y, Zhu J, Zhang J, Yu Y, Shen Q, Liu X, Sessler DI. Effect of esketamine on postoperative depression in women with breast cancer and preoperative depressive symptoms: The EASE randomized trial. J Clin Anesth 2025; 103:111821. [PMID: 40153893 DOI: 10.1016/j.jclinane.2025.111821] [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: 12/24/2024] [Revised: 02/09/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
STUDY OBJECTIVE To determine whether intraoperative low-dose esketamine ameliorates depression in women having breast cancer surgery. DESIGN A prospective single-center double blind randomized placebo-controlled trial. SETTING Perioperative period, operating room, post anesthesia care unit and hospital ward. PATIENTS 108 women 18-65 years old who were scheduled for elective breast cancer surgery. All had preoperative depressive symptoms as defined by Montgomery-Åsberg depression scores ≥12 (range, 0-60; higher scores indicate more severe depression). INTERVENTIONS Eligible participants were randomized to esketamine 0.25 mg/kg or saline placebo. Blinded trial drugs were given intravenously over the initial 40 min of anesthesia. MEASUREMENTS Our primary outcome was the fraction of patients who had at least a 50 % reduction in the Montgomery-Åsberg depression score within 3 postoperative days. Secondary outcomes included the fraction of patients with depression remission defined as Montgomery-Åsberg scores ≤10, the numeric value of the Montgomery-Åsberg depression scores, postoperative severe pain, and anxiety as determined by the Generalized Anxiety Disorder 7-item score. Adverse events were monitored for 72 postoperative hours. MAIN RESULTS 54 women each were randomized to esketamine and saline, and 104 were available for our intent-to-treat analysis. The mean age was 50 years. Esketamine non-significantly doubled the fraction of patients who had a 50 % reduction in their depressions scores: 27 % vs 13 %, odds ratio 2.4, [95 % CI 0.9 to 6.6], P = 0.087. Montgomery-Åsberg depression scores were nearly a factor-of-two and significantly lower (better) on postoperative days 1 to 5 in patients given esketamine. Montgomery-Åsberg scores decreased significantly more from baseline in patients randomized to esketamine: mean difference - 2.5 [95 % CI -4.5 to -0.6], P = 0.010. Esketamine treatment had no significant effect on other secondary outcomes or on adverse events. CONCLUSIONS Intraoperative administration of 0.25 mg/kg esketamine did not significantly improve the fraction of depressed women having breast cancer patients who had a 50 % reduction in their depression scores at 3 days postoperatively. However, the observed factor-of-two treatment effect was clinically meaningful and esketamine significantly reduced short-term postoperative depression scores without provoking complications. Robust trials are warranted. Registration Trial registry:http://www.chictr.org.cn/; Identifier: ChiCTR2300071062.
Collapse
Affiliation(s)
- Qingfeng Wei
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mengmeng Li
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qiuling Du
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Huiwen Zhang
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, China
| | - Yongjie Liang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Cen Cheng
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Bin Mei
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, China
| | - Xiaowei Yang
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yinguang Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Jiajia Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jingjie Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Qiying Shen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, China.
| | - Xuesheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Hefei, Anhui, China; OUTCOMES RESEARCH Consortium®, Houston, TX, USA.
| | - Daniel I Sessler
- OUTCOMES RESEARCH Consortium®, Houston, TX, USA; Center for OUTCOMES RESEARCH and Department of Anesthesiology, UTHealth, Houston, TX, USA; Population Health Research Institute, McMaster University, ON, Canada
| |
Collapse
|
29
|
Cardona-Acosta AM, Parise LF, Bolaños-Guzmán CA, Parise EM. Prophylactic Ketamine: Current Knowledge and Future Directions. Biol Psychiatry 2025:S0006-3223(25)01103-5. [PMID: 40158609 DOI: 10.1016/j.biopsych.2025.03.015] [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: 09/30/2024] [Revised: 02/21/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
The prevalence of stress-induced disorders, including depression, anxiety, posttraumatic stress disorder, and postpartum depression, has been increasing, while current treatment approaches are limited. As a result, researchers are exploring alternative treatments that include ketamine as a prophylactic against these disorders. This review provides an overview of the current knowledge on the use of ketamine as a prophylactic for stress-induced disorders, including preclinical and clinical findings on (R,S)-ketamine, as well as (2R,6R)- and (2S,6S)-hydroxynorketamine. We also explore the potential underlying mechanisms involved in preventing these disorders, including the brain regions/circuits, as well as glutamatergic, dopaminergic, serotonergic, and inflammatory processes known to be involved, as evidenced by studies with ketamine and its metabolites. Additionally, we highlight the limitations and risks associated with ketamine use, such as age- and sex-specific efficacy, potential long-term and adverse effects, and legal and ethical considerations. Finally, we discuss future research directions, including the implications for clinical practice, integrating ketamine into current treatment approaches, and potential advancements in ketamine-based therapies. Overall, the literature emphasizes the importance of continuing research to better understand the potential benefits and risks of ketamine as a prophylactic for stress-induced disorders.
Collapse
Affiliation(s)
- Astrid M Cardona-Acosta
- Department of Psychological and Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, Texas
| | - Lyonna F Parise
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Carlos A Bolaños-Guzmán
- Department of Psychological and Brain Sciences, and Institute for Neuroscience, Texas A&M University, College Station, Texas
| | - Eric M Parise
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
30
|
Ivanović A, Petrović J, Stanić D, Nedeljković J, Ilić M, Jukić MM, Pejušković B, Pešić V. Single subanesthetic dose of ketamine exerts antioxidant and antidepressive-like effect in ACTH-induced preclinical model of depression. Mol Cell Neurosci 2025; 133:104006. [PMID: 40157469 DOI: 10.1016/j.mcn.2025.104006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
Hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis and oxidative stress represent important mechanisms that have been implicated in etiopathology of depression. Although first antidepressants were introduced in clinical practice more than six decades ago, approximately 30 % of patients with a diagnosis of depression show treatment resistance. A noncompetitive N-methyl-d-aspartate receptor antagonist ketamine has shown promising rapid antidepressant effects and has been approved for treatment-resistant depression (TRD). In the present study, we investigated antioxidant and antidepressant-like activity of a single subanesthetic dose of ketamine (10 mg/kg, ip) in a rodent model of TRD induced by adrenocorticotropic hormone (10 μg ACTH/day, sc, 21 days). Behavioral assessment was performed, and plasma biomarkers of oxidative stress and DNA damage in peripheral blood lymphocytes (PBLs) were determined. We observed that ACTH produced depressive-like behavior and significant increase in superoxide anion (O2·-), advanced oxidation protein products (AOPP), malondialdehyde (MDA) and total oxidant status (TOS) in male Wistar rats. This effect was accompanied by reduced activity of antioxidant enzymes - superoxide dismutase (SOD) and paraoxonase1 (PON1) in plasma and increase in DNA damage in PBLs. In the described model of TRD, we have demonstrated antidepressant effects of ketamine for the first time. Our results reveal that ketamine was effective in reducing O2.-, AOPP, MDA and TOS, while enhancing SOD and PON1 activity in ACTH-rats. Collectively, our study sheds light on molecular mechanisms implicated in antioxidant activity of ketamine, thus incentivizing further investigation of its effects on ROS metabolism and antioxidant defenses in clinical trials, particularly in depression.
Collapse
Affiliation(s)
- Ana Ivanović
- Department of Physiology, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11121 Belgrade, Serbia
| | - Jelena Petrović
- Department of Physiology, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11121 Belgrade, Serbia
| | - Dušanka Stanić
- Department of Physiology, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11121 Belgrade, Serbia.
| | - Jelena Nedeljković
- Department of Physiology, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11121 Belgrade, Serbia
| | - Miloš Ilić
- Department of Physiology, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11121 Belgrade, Serbia
| | - Marin M Jukić
- Department of Physiology, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11121 Belgrade, Serbia; Pharmacogenetics Section, Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
| | - Bojana Pejušković
- Institute of Mental Health, School of Medicine, University of Belgrade, Palmotićeva 37, 11000 Belgrade, Serbia
| | - Vesna Pešić
- Department of Physiology, University of Belgrade, Faculty of Pharmacy, Vojvode Stepe 450, 11121 Belgrade, Serbia
| |
Collapse
|
31
|
Gao K, Oruc EB, Koparal B. Pharmacological Monotherapy for Depressive Disorders: Current and Future-A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:558. [PMID: 40282849 PMCID: PMC12028769 DOI: 10.3390/medicina61040558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025]
Abstract
Objective: To narratively review currently available antidepressants and future potential antidepressants as monotherapy for the treatment of depressive disorders. Methods: Selective serotonin reuptake inhibitors (SSRIs), serotonin norepinephrine reuptake inhibitors (SNRIs), dopamine reuptake inhibitor (bupropion), tricyclic antidepressants (TCAs), and monoamine oxidase inhibitors (MAOIs) were reviewed according to the results from Sequenced Treatment Alternatives to Relieve Depression (STAR*D) Study and systematic reviews. For the rest of the antidepressants, a PubMed/Medline search was conducted with priority for systematic reviews. For drugs in development for depressive disorders, PubMed, Google, and Clinicaltrials.gov databases were used. Results: The STAR*D Study demonstrated that sertraline, venlafaxine, and bupropion monotherapy had similar efficacy in patients with major depressive disorder (MDD) who failed citalopram. A network meta-analyses of randomized, placebo-controlled trials found that SSRIs, SNRIs, bupropion, TCAs, mirtazapine, and agomelatine had similar relative efficacy compared to placebo, but had different acceptability. Gepirone had more failed/negative studies and smaller effect size relative to placebo compared to other antidepressants. The combination of dextromethorphan and bupropion, ketamine infusion, and intranasal esketamine had faster onset of action but similar effect size compared to monoamine-based antidepressants as monotherapy. Brexanolone and zuranolone are effective in postpartum depression (PPD), but the effect size of zuranolone in MDD as monotherapy or adjunctive therapy was very small. Psychedelics, glutamate receptor-related agents, kappa opioid receptor antagonists, orexin receptor antagonists, new anti-inflammatory agents, and biomarker-based antidepressant therapy have been under investigation for depressive disorders. Psychedelics showed faster onset of action, large effect size, and long durability. Conclusions: Monoamine-based antidepressants likely continue to be the mainstream antidepressants for depressive disorder. NMDA receptor antagonists and neurosteroid antidepressants will play a bigger role with the improvement of accessibility. Psychedelics may become a game changer if phase III studies validate their efficacy and safety in depressive disorders.
Collapse
Affiliation(s)
- Keming Gao
- Department of Mind and Body Medicine, Sichuan Lansheng Brian Hospital, Chengdu 610036, China
- Department of Psychiatry, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
- Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | - Buket Koparal
- Department of Psychiatry, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA;
- Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Department of Psychiatry, Gazi University School of Medicine, Ankara 06500, Turkey
| |
Collapse
|
32
|
Gyles T, Parise EM, Estill MS, Browne CJ, Shen L, Nestler EJ, Torres-Berrío A. Transcriptional Profiles in Nucleus Accumbens of Antidepressant Resistance in Chronically Stressed Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643727. [PMID: 40166343 PMCID: PMC11956914 DOI: 10.1101/2025.03.17.643727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Unsuccessful response to several courses of antidepressants is a core feature of treatment-resistant depression (TRD), a severe condition that affects a third of patients with depression treated with conventional pharmacotherapy. However, the molecular mechanisms underlying TRD remain poorly understood. Here, we assessed the successful vs. unsuccessful response to ketamine (KET) in chronically stressed mice that failed to respond to initial treatment with fluoxetine (FLX) as a rodent model of TRD and characterized the associated transcriptional profiles in the nucleus accumbens (NAc) using RNA-sequencing. We observed that failed treatment with FLX exerts a priming effect that promotes behavioral and transcriptional responses to subsequent ketamine treatment. We also identified specific gene networks that are linked to both susceptibility to stress and resistance to antidepressant response. Collectively, these findings offer valuable insights into the molecular mechanisms underlying antidepressant resistance and help address a critical gap in preclinical models of TRD.
Collapse
|
33
|
Georgiou P, Farmer CA, Medeiros GC, Yuan P, Johnston J, Kadriu B, Gould TD, Zarate CA. Associations between hypothalamic-pituitary-adrenal (HPA) axis hormone levels, major depression features and antidepressant effects of ketamine. J Affect Disord 2025; 373:126-132. [PMID: 39674325 DOI: 10.1016/j.jad.2024.12.036] [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: 07/19/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Subanesthetic doses of (R,S)-ketamine (ketamine) have demonstrated rapid and robust antidepressant effects in individuals with depression. However, individual variability in response to ketamine exists, and current biomarkers of ketamine treatment response are not entirely understood. Preclinical evidence suggests a link between hypothalamic-pituitary-adrenal (HPA) axis activation, a determinant of the stress response system, and ketamine's efficacy in stressed mice exhibiting enhanced antidepressant responses. Here, we assessed the relationship between HPA axis, major depression features, and antidepressant response to ketamine in humans. METHODS We investigated 42 participants following medication washout with treatment-resistant depression who participated in a randomized, placebo-controlled, crossover trial receiving intravenous ketamine. Plasma levels of corticotropin-releasing factor (CRF), adrenocorticotropic hormone (ACTH), and cortisol were measured at baseline. Ketamine's antidepressant effects were assessed using the Montgomery-Asberg Depression Rating Scale. RESULTS We found that baseline HPA axis hormone levels did not significantly moderate the antidepressant effects of ketamine. However, a negative association was observed between ACTH and CRF levels and the overall duration of depressive episodes, suggesting potential biomarker implications. Also, a negative correlation between baseline depressive scores and age of onset was observed, suggesting that the severity of depression might be greater if it develops at a younger age, indicating more enduring stress on the brain and body. DISCUSSION Although we did not find a moderation effect of the plasma HPA axis hormones on the antidepressant effects of ketamine, moderation effects of the brain HPA axis hormones cannot be precluded and warrants further investigation. Importantly, our results implicate HPA axis components as potential biomarkers for the duration of depressive episodes.
Collapse
Affiliation(s)
- Polymnia Georgiou
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA; Department of Psychology, University of Wisconsin, Milwaukee, WI, USA
| | - Cristan A Farmer
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Gustavo C Medeiros
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Peixiong Yuan
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Jenessa Johnston
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Todd D Gould
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD, USA; Veterans Affairs Maryland Health Care System, Baltimore, MD, USA; Departments of Pharmacology, Neurobiology School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
34
|
Repple J, Bayas M, Möser C, Kobayashi NF, Reif A. Current Evidence for the Role of Rapid-Acting Antidepressants in Bipolar Depression: A Perspective and Plan for Action. Biol Psychiatry 2025:S0006-3223(25)01019-4. [PMID: 40064389 DOI: 10.1016/j.biopsych.2025.02.903] [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: 10/15/2024] [Revised: 01/30/2025] [Accepted: 02/25/2025] [Indexed: 05/15/2025]
Abstract
After decades of limited progress in depression treatment, recent advancements have sparked renewed interest in developing novel antidepressants, particularly rapid-acting antidepressants (RAADs). Despite these promising developments, there remains a significant gap in research on bipolar depression. While several antipsychotics have been investigated for their efficacy in bipolar depression due to the reduced risk of mania induction, research on RAADs, such as (es)ketamine, remains scarce despite their demonstrated safety and effectiveness. In this review, we give an overview of current developments in RAADs in the context of bipolar disorder. Both published studies as well as phase II, III, and IV studies on bipolar depression (based on ClinicalTrials.gov) are reviewed in this work. The following RAAD substance classes have been or are currently being investigated as possible treatments for bipolar depression: NMDA antagonists and indirect AMPA agonists (ketamine, esketamine, riluzole, felbamate), GABAA (gamma-aminobutyric acid A) activators or positive allosteric modulators (zuranolone, pregnenolone, PEA), psychedelics (psilocybin, 5-MeO-DMT), muscarine receptor antagonists (scopolamine), and kappa opioid receptor antagonists (navacaprant). Other than the well-established efficacy and safety of (es)ketamine in treating bipolar depression, there has been little research effort in the treatment of bipolar depression. Recent research into RAADs demonstrates the growing field of novel mechanisms of action in the pharmacological treatment of bipolar depression. However, there is an urgent need for well-controlled clinical studies on RAADs in bipolar depression to expand treatment options and improve outcomes for millions of affected individuals worldwide.
Collapse
Affiliation(s)
- Jonathan Repple
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Frankfurt, Germany; Institute for Translational Psychiatry, University of Münster, Münster, Germany.
| | - Maximilian Bayas
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | - Chiara Möser
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | - Nene F Kobayashi
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Frankfurt, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology, Frankfurt am Main, Germany
| |
Collapse
|
35
|
Abbott JA, Wen H, Liu B, Gupta SS, Iacobucci GJ, Zheng W, Popescu GK. Allosteric inhibition of NMDA receptors by low dose ketamine. Mol Psychiatry 2025; 30:1009-1018. [PMID: 39237721 PMCID: PMC11948614 DOI: 10.1038/s41380-024-02729-9] [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: 08/28/2023] [Revised: 08/12/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
Ketamine, a general anesthetic, has rapid and sustained antidepressant effects when administered at lower doses. Anesthetic levels of ketamine reduce excitatory transmission by binding deep into the pore of NMDA receptors where it blocks current influx. In contrast, the molecular targets responsible for antidepressant levels of ketamine remain controversial. We used electrophysiology, structure-based mutagenesis, and molecular and kinetic modeling to investigate the effects of ketamine on NMDA receptors across an extended range of concentrations. We report functional and structural evidence that, at nanomolar concentrations, ketamine interacts with membrane-accessible hydrophobic sites on NMDA receptors, which are distinct from the established pore-blocking site. These interactions stabilize receptors in pre-open states and produce an incomplete, voltage- and pH-dependent reduction in receptor gating. Notably, this allosteric inhibitory mechanism spares brief synaptic-like receptor activations and preferentially reduces currents from receptors activated tonically by ambient levels of neurotransmitters. We propose that the hydrophobic sites we describe here account for clinical effects of ketamine not shared by other NMDA receptor open-channel blockers such as memantine and represent promising targets for developing safe and effective neuroactive therapeutics.
Collapse
Affiliation(s)
- Jamie A Abbott
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Han Wen
- Department of Physics, College of Biological Sciences, Buffalo, NY, 14260, USA
| | - Beiying Liu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Sheila S Gupta
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Gary J Iacobucci
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Wenjun Zheng
- Department of Physics, College of Biological Sciences, Buffalo, NY, 14260, USA
| | - Gabriela K Popescu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
36
|
Baldinger-Melich P, Spies M, Bozic I, Kasper S, Rujescu D, Frey R. Perspectives in treatment-resistant depression: esketamine and electroconvulsive therapy. Wien Klin Wochenschr 2025; 137:134-147. [PMID: 38662240 DOI: 10.1007/s00508-024-02358-w] [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: 12/29/2023] [Accepted: 02/23/2024] [Indexed: 04/26/2024]
Abstract
Modern electroconvulsive therapy (ECT) and the approval of nasal esketamine for clinical use have significantly improved the approach to treatment-resistant depression (TRD), which is defined as non-response to at least two different courses of antidepressants with verified adherence to treatment, adequate dosage, and duration of treatment. The goal of this literature review is to present the newest evidence regarding efficacy and safety. Furthermore, we aim to provide an overview of future perspectives in this field of research, for example, regarding structural and molecular effects. Both treatment methods will be critically evaluated for their individual advantages, disadvantages, and response rates. Firstly, we will discuss the well-established method of ECT and its different treatment modalities. Secondly, we will discuss the properties of ketamine, the discovery of its antidepressive effects and the route to clinical approval of the esketamine nasal spray. We will comment on research settings which have evaluated intravenous ketamine against ECT. The decision-making process between esketamine nasal spray or ECT should include the assessment of contraindications, age, severity of disease, presence of psychotic symptoms, patient preference and treatment accessibility. We conclude that both treatment options are highly effective in TRD. If both are indicated, pragmatically esketamine will be chosen before ECT; however, ECT studies in ketamine non-responders are missing.
Collapse
Affiliation(s)
- Pia Baldinger-Melich
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Vienna, Austria
| | - Marie Spies
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Vienna, Austria
| | - Ina Bozic
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Vienna, Austria
| | - Siegfried Kasper
- Department of Molecular Neurosciences, Center for Brain Research, Vienna, Austria
| | - Dan Rujescu
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Vienna, Austria
| | - Richard Frey
- Department of Psychiatry and Psychotherapy, Clinical Division of General Psychiatry, Medical University Vienna, Vienna, Austria.
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Vienna, Austria.
| |
Collapse
|
37
|
Ballard ED, Neely L, Waldman L, Greenstein D, Zarate CA. Clinical indicators of the suicide crisis and response to ketamine. J Affect Disord 2025; 372:126-133. [PMID: 39617360 PMCID: PMC11729457 DOI: 10.1016/j.jad.2024.11.080] [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: 09/03/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND This analysis sought to identify potential clinical targets for the suicide crisis. Characteristics of a useful clinical target include elevation at the time of suicide crisis and responsiveness to rapid-acting interventions. Suicidal ideation (SI), depression, and hopelessness were hypothesized to meet these criteria. METHODS Participants were 118 adults across the continuum of suicide risk, including 14 high-risk (HR) individuals who had attempted or seriously considered suicide within the last two weeks. Clinical characteristics were evaluated by: 1) comparing individuals with a recent crisis state to those whose suicide crises had resolved; 2) quantifying responses to a semi-structured interview about the time just before a suicide crisis; and 3) comparing symptomatology before and after an open-label ketamine infusion (0.5 mg/kg) in a subset of the HR group (n = 10). RESULTS As hypothesized, SI, depression, and hopelessness were elevated just after a suicide crisis and responded to ketamine, although findings were mixed depending on the assessment used. Psychological pain and traumatic stress symptoms were also associated with the suicide crisis and responded to ketamine. Participants reported high levels of SI, depression, and anxiety just before their suicide attempt. LIMITATIONS Limitations include the small sample size, inconsistent assessments across analyses, and that ketamine was the only intervention examined. CONCLUSIONS These results underscore the importance of SI, depression, hopelessness, psychological pain, and traumatic stress in this population, all of which were elevated during the suicide crisis and responded to ketamine. A multifactorial and longitudinal approach is indicated to assess and treat suicide risk.
Collapse
Affiliation(s)
- Elizabeth D Ballard
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Lucinda Neely
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Laura Waldman
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Dede Greenstein
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
38
|
Kundu D, Wang M, Paudel S, Wang S, Jang CG, Kim KM. Structure-Activity Relationship of NMDA Receptor Ligands and Their Activities on the ERK Activation through Metabotropic Signaling Pathway. Biomol Ther (Seoul) 2025; 33:278-285. [PMID: 39934970 PMCID: PMC11893489 DOI: 10.4062/biomolther.2024.216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 02/13/2025] Open
Abstract
The N-methyl-D-aspartate receptor (NMDA-R) subunit GluN2B is abundantly expressed in brain regions critical for synaptic plasticity and cognitive processes. This study investigated the structure-activity relationships (SAR) of NMDA-R ligands using GluN2B as a molecular target. Thirty potential NMDA-R antagonists were categorized into two structural classes: 1-(1-phenylcyclohexyl) amines (series A) and α-amino-2-phenylcyclohexanone derivatives (series B). In series A compounds, the phenyl ring and R1 substituents were positioned at the carbon center of the cyclohexyl ring, with R2 substituents at the para- or meta-positions of the phenyl ring. SAR analysis revealed optimal binding affinity when R1 was carbonyl (C=O) and R2 was 4-methoxy (4-OMe). Series B compounds featured a cyclohexanone scaffold with NH-R1 at the α-position and a phenyl ring bearing R2 substituents at ortho-, meta-, or para-positions. Maximum binding affinity was achieved with R1 as hydrogen (H) and R2 as hydroxyl (OH). Compounds were assessed for GluN2B-mediated ERK activation to evaluate potential metabotropic signaling properties. Approximately 50% of the compounds demonstrated ERK activation through a non-ionotropic signaling cascade involving Src, phosphatidylinositol 3-kinase, and protein kinase C. This study elucidated key structural determinants for NMDA-R binding and characterized a novel metabotropic signaling pathway. Notably, our findings suggest that compounds acting as antagonists at the ionotropic site may simultaneously function as agonists through non-ionotropic mechanisms.
Collapse
Affiliation(s)
- Dooti Kundu
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Mengling Wang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Suresh Paudel
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Shujie Wang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
39
|
Levinstein MR, Budinich RC, Bonaventura J, Schatzberg AF, Zarate CA, Michaelides M. Redefining Ketamine Pharmacology for Antidepressant Action: Synergistic NMDA and Opioid Receptor Interactions? Am J Psychiatry 2025; 182:247-258. [PMID: 39810555 PMCID: PMC11872000 DOI: 10.1176/appi.ajp.20240378] [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] [Indexed: 01/16/2025]
Abstract
Ketamine is a racemic compound and medication comprised of (S)-ketamine and (R)-ketamine enantiomers and its metabolites. It has been used for decades as a dissociative anesthetic, analgesic, and recreational drug. More recently, ketamine, its enantiomers, and its metabolites have been used or are being investigated for the treatment of refractory depression, as well as for comorbid disorders such as anxiety, obsessive-compulsive, and opioid use disorders. Despite its complex pharmacology, ketamine is referred to as an N-methyl-d-aspartate (NMDA) receptor antagonist. In this review, the authors argue that ketamine's pharmacology should be redefined to include opioid receptors and the endogenous opioid system. They also highlight a potential mechanism of action of ketamine for depression that is attributed to bifunctional, synergistic interactions involving NMDA and opioid receptors.
Collapse
Affiliation(s)
- Marjorie R. Levinstein
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Section, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Reece C. Budinich
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Section, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Jordi Bonaventura
- Departament de Patologia i Terapèutica Experimental, Institut de Neurociències, Universitat de Barcelona, L’Hospitalet de Llobregat
- Neuropharmacology and Pain Group, Neuroscience Program, IDIBELL-Institut d’Investigació Biomèdica de Bellvitge, L’Hospitalet de Llobregat
| | - Alan F. Schatzberg
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, California, USA
| | - Carlos A. Zarate
- Experimental Therapeutics & Pathophysiology Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Section, National Institute on Drug Abuse, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
40
|
Elazar D, Alvarez N, Drobeck S, Gunn TM. SLC1A4 and Serine Homeostasis: Implications for Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2025; 26:2104. [PMID: 40076728 PMCID: PMC11900201 DOI: 10.3390/ijms26052104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The solute carrier family 1 member 4 (SLC1A4) gene encodes a neutral amino acid transporter, also referred to as alanine-serine-cysteine transporter 1, ASCT1, that helps maintain amino acid balance in the brain and periphery. In the brain, SLC1A4 plays an important role in transporting levo (L) and dopa (D) isomers of serine. L-serine is required for many cellular processes, including protein and sphingolipid synthesis, while D-serine is a co-agonist required for normal neurotransmission through N-methyl-D-aspartate receptors. Through its roles transporting L-serine across the blood-brain barrier and regulating synaptic D-serine levels, SLC1A4 helps establish and maintain brain health across the lifespan. This review examines the role of SLC1A4 in neurodevelopment and neurodegeneration and assesses the therapeutic potential of serine supplementation to treat neurodevelopmental symptoms associated with mutations in SLC1A4, as well as schizophrenia, depression, traumatic brain injury, and Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Dana Elazar
- Touro College of Osteopathic Medicine, Touro University, Great Falls, MT 59405, USA; (D.E.); (N.A.); (S.D.)
| | - Natalie Alvarez
- Touro College of Osteopathic Medicine, Touro University, Great Falls, MT 59405, USA; (D.E.); (N.A.); (S.D.)
| | - Sabrina Drobeck
- Touro College of Osteopathic Medicine, Touro University, Great Falls, MT 59405, USA; (D.E.); (N.A.); (S.D.)
| | - Teresa M. Gunn
- Touro College of Osteopathic Medicine, Touro University, Great Falls, MT 59405, USA; (D.E.); (N.A.); (S.D.)
- McLaughlin Research Institute, Great Falls, MT 59405, USA
| |
Collapse
|
41
|
Wade BSC, Pindale R, Luccarelli J, Li S, Meisner RC, Seiner SJ, Camprodon JA, Henry ME. Prediction of individual treatment allocation between electroconvulsive therapy or ketamine using the Personalized Advantage Index. NPJ Digit Med 2025; 8:127. [PMID: 40016503 PMCID: PMC11868618 DOI: 10.1038/s41746-025-01523-3] [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: 03/15/2024] [Accepted: 02/17/2025] [Indexed: 03/01/2025] Open
Abstract
Electroconvulsive therapy (ECT) and ketamine are effective treatments for depression; however, evidence-based guidelines are needed to inform individual treatment selection. We adapted the Personalized Advantage Index (PAI) using machine learning to predict optimal treatment assignment to ECT or ketamine using EHR data on 2506 ECT and 196 ketamine patients. Depressive symptoms were evaluated using the Quick Inventory of Depressive Symptomatology (QIDS) before and during acute treatment. Propensity score matching across treatments was used to address confounding by indication, yielding a sample of 392 patients (n = 196 per treatment). Models predicted differential minimum QIDS scores (min-QIDS) over acute treatment using pretreatment EHR measures and SHAP values identified prescriptive predictors. Patients with large PAI scores who received a predicted optimal had significantly lower min-QIDS compared to the non-optimal treatment group (mean difference = 1.19 [95% CI: 0.32, ∞], t = 2.25, q < 0.05, d = 0.26). Our model identified candidate pretreatment factors to provide actionable, effective antidepressant treatment selection guidelines.
Collapse
Affiliation(s)
- Benjamin S C Wade
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Ryan Pindale
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - James Luccarelli
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuang Li
- Department of Psychiatry, McLean Hospital, Belmont, MA, USA
| | - Robert C Meisner
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, McLean Hospital, Belmont, MA, USA
| | | | - Joan A Camprodon
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael E Henry
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Ponce-Regalado MD, Becerril-Villanueva E, Maldonado-García JL, Moreno-Lafont MC, Martínez-Ramírez G, Jacinto-Gutiérrez S, Arreola R, Sánchez-Huerta K, Contis-Montes de Oca A, López-Martínez KM, Bautista-Rodríguez E, Chin-Chan JM, Pavón L, Pérez-Sánchez G. Comprehensive view of suicide: A neuro-immune-endocrine approach. World J Psychiatry 2025; 15:98484. [PMID: 39974471 PMCID: PMC11758041 DOI: 10.5498/wjp.v15.i2.98484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 11/26/2024] [Accepted: 12/23/2024] [Indexed: 01/14/2025] Open
Abstract
Suicide is defined as the act of a person attempting to take their own life by causing death. Suicide is a complex phenomenon that is influenced by a multitude of factors, including psychosocial, cultural, and religious aspects, as well as genetic, biochemical, and environmental factors. From a biochemical perspective, it is crucial to consider the communication between the endocrine, immune, and nervous systems when studying the etiology of suicide. Several pathologies involve the bidirectional communication between the peripheral activity and the central nervous system by the action of molecules such as cytokines, hormones, and neurotransmitters. These humoral signals, when present in optimal quantities, are responsible for maintaining physiological homeostasis, including mood states. Stress elevates the cortisol and proinflammatory cytokines levels and alter neurotransmitters balance, thereby increasing the risk of developing a psychiatric disorder and subsequently the risk of suicidal behavior. This review provides an integrative perspective about the neurochemical, immunological, and endocrinological disturbances associated with suicidal behavior, with a particular focus on those alterations that may serve as potential risk markers and/or indicators of the state preceding such a tragic act.
Collapse
Affiliation(s)
- María D Ponce-Regalado
- Departamento de Ciencias de la Salud, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos 47620, Jalisco, Mexico
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - José Luis Maldonado-García
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11350, Mexico
| | - Martha C Moreno-Lafont
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11350, Mexico
| | - Gabriela Martínez-Ramírez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
- Facultad de Medicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional autónoma de México, Tlalnepantla 54090, Mexico
| | - Salomón Jacinto-Gutiérrez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - Rodrigo Arreola
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - Karla Sánchez-Huerta
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico
| | - Arturo Contis-Montes de Oca
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | | | | | - José Miguel Chin-Chan
- Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Campeche, Campeche 24039, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México 11340, Mexico
| |
Collapse
|
43
|
Shi ZM, Lan XJ, Chen Q, Chen JJ, Su ZA, Huang XB, Ning YP, Yang XH, Wei X, Zheng W. Intravenous ketamine versus electroconvulsive therapy for major depressive disorder or bipolar depression: A meta-analysis of randomized controlled trials. J Affect Disord 2025; 371:45-53. [PMID: 39549887 DOI: 10.1016/j.jad.2024.11.034] [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: 09/26/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Intravenous (IV) ketamine has been evaluated alongside electroconvulsive therapy (ECT) in addressing major depressive disorder (MDD) or bipolar depression (BD), though the comparative outcomes remain inconclusive. This meta-analysis aimed to provide a systematic assessment of the efficacy, safety, and tolerability of IV ketamine relative to ECT for treating MDD or BD. METHODS Randomized controlled trials (RCTs) comparing IV ketamine and ECT in terms of efficacy, safety, and tolerability for MDD or BD were identified and reviewed. Three independent investigators extracted relevant data, which was synthesized using RevMan 5.3 software under a random effects model. RESULTS Five RCTs encompassing 664 patients diagnosed with MDD or BD were analyzed. At 24 h post-initial treatment, IV ketamine demonstrated a statistically greater reduction in depressive symptoms compared to ECT (standardized mean difference (SMD) = -0.49, 95 % CI = -0.96 to -0.03, I2 = 77 %; P = 0.04). However, no significant differences were observed between groups in terms of the study-defined response (risk ratio (RR) = 1.49, 95 % CI = 0.08 to 28.42, I2 = 0 %; P = 0.79) at 24 h. Similarly, no notable differences were found for depressive symptom improvement (SMD = -0.48, 95 % CI = -2.41 to 1.45, I2 = 99 %; P = 0.63), the study-defined response (RR = 0.96, 95 % CI: 0.70 to 1.31, I2 = 75 %; P = 0.79) or remission (RR = 0.91, 95 % CI: 0.45 to 1.82, I2 = 90 %; P = 0.78) at the end-of-treatment visit. Among the three RCTs (60 %) comparing the neurocognitive outcomes of IV ketamine and ECT through varying test batteries, results were inconsistent. IV ketamine was associated with marked increases in dissociation, blurred vision, dizziness, and diplopia, while ECT led to a significant rise in muscle pain (all Ps < 0.05). Discontinuation rates due to any cause were comparable between both groups (P > 0.05). CONCLUSION IV ketamine demonstrates a faster onset of antidepressant effects compared to ECT, despite both treatments yielding comparable outcomes at the end-of-treatment visit for patients with MDD or BD. However, large-scale RCTs are required to thoroughly evaluate the long-term efficacy and safety of IV ketamine relative to ECT in these populations.
Collapse
Affiliation(s)
- Zhan-Ming Shi
- Chongqing Jiangbei Mental Health Center, Chongqing, China
| | - Xian-Jun Lan
- The Brain Hospital of Guangxi Zhuang Autonomous Region, LiuZhou, China
| | - Qing Chen
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Jun-Jun Chen
- Chongqing Eleventh People's Hospital, Chongqing, China
| | - Zhi-Ang Su
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Xing-Bing Huang
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Yu-Ping Ning
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Xin-Hu Yang
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | - Xin Wei
- The Brain Hospital of Guangxi Zhuang Autonomous Region, LiuZhou, China.
| | - Wei Zheng
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
44
|
Aboharb F, Davoudian PA, Shao LX, Liao C, Rzepka GN, Wojtasiewicz C, Indajang J, Dibbs M, Rondeau J, Sherwood AM, Kaye AP, Kwan AC. Classification of psychedelics and psychoactive drugs based on brain-wide imaging of cellular c-Fos expression. Nat Commun 2025; 16:1590. [PMID: 39939591 PMCID: PMC11822132 DOI: 10.1038/s41467-025-56850-6] [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: 04/19/2024] [Accepted: 01/31/2025] [Indexed: 02/14/2025] Open
Abstract
Psilocybin, ketamine, and MDMA are psychoactive compounds that exert behavioral effects with distinguishable but also overlapping features. The growing interest in using these compounds as therapeutics necessitates preclinical assays that can accurately screen psychedelics and related analogs. We posit that a promising approach may be to measure drug action on markers of neural plasticity in native brain tissues. We therefore developed a pipeline for drug classification using light sheet fluorescence microscopy of immediate early gene expression at cellular resolution followed by machine learning. We tested male and female mice with a panel of drugs, including psilocybin, ketamine, 5-MeO-DMT, 6-fluoro-DET, MDMA, acute fluoxetine, chronic fluoxetine, and vehicle. In one-versus-rest classification, the exact drug was identified with 67% accuracy, significantly above the chance level of 12.5%. In one-versus-one classifications, psilocybin was discriminated from 5-MeO-DMT, ketamine, MDMA, or acute fluoxetine with >95% accuracy. We used Shapley additive explanation to pinpoint the brain regions driving the machine learning predictions. Our results suggest a unique approach for characterizing and validating psychoactive drugs with psychedelic properties.
Collapse
Affiliation(s)
- Farid Aboharb
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Weill Cornell Medicine/Rockefeller/Sloan-Kettering Tri-Institutional MD/PhD Program, New York, NY, USA
| | - Pasha A Davoudian
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT, USA
| | - Ling-Xiao Shao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Clara Liao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Gillian N Rzepka
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | | | - Jonathan Indajang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Mark Dibbs
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Jocelyne Rondeau
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | | | - Alfred P Kaye
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Clinical Neurosciences Division, VA National Center for PTSD, West Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Alex C Kwan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
45
|
Choquet D, Opazo P, Zhang H. AMPA receptor diffusional trapping machinery as an early therapeutic target in neurodegenerative and neuropsychiatric disorders. Transl Neurodegener 2025; 14:8. [PMID: 39934896 PMCID: PMC11817889 DOI: 10.1186/s40035-025-00470-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
Over the past two decades, there has been a growing recognition of the physiological importance and pathological implications surrounding the surface diffusion of AMPA receptors (AMPARs) and their diffusional trapping at synapses. AMPAR surface diffusion entails the thermally powered random Brownian lateral movement of these receptors within the plasma membrane, facilitating dynamic exchanges between synaptic and extrasynaptic compartments. This process also enables the activity-dependent diffusional trapping and accumulation of AMPARs at synapses through transient binding to synaptic anchoring slots. Recent research highlights the critical role of synaptic recruitment of AMPARs via diffusional trapping in fundamental neural processes such as the development of the early phases of long-term potentiation (LTP), contextual fear memory, memory consolidation, and sensory input-induced cortical remapping. Furthermore, studies underscore that regulation of AMPAR diffusional trapping is altered across various neurological disease models, including Huntington's disease (HD), Alzheimer's disease (AD), and stress-related disorders like depression. Notably, pharmacological interventions aimed at correcting deficits in AMPAR diffusional trapping have demonstrated efficacy in restoring synapse numbers, LTP, and memory functions in these diverse disease models, despite their distinct pathogenic mechanisms. This review provides current insights into the molecular mechanisms underlying the dysregulation of AMPAR diffusional trapping, emphasizing its role as a converging point for multiple pathological signaling pathways. We propose that targeting AMPAR diffusional trapping represents a promising early therapeutic strategy to mitigate synaptic plasticity and memory deficits in a spectrum of brain disorders, encompassing but not limited to HD, AD, and stress-related conditions. This approach underscores an integrated therapeutic target amidst the complexity of these neurodegenerative and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Daniel Choquet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000, Bordeaux, France
- Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, 33000, Bordeaux, France
| | - Patricio Opazo
- UK Dementia Research Institute, Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh, EH16 4SB, UK
| | - Hongyu Zhang
- Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.
- Mohn Research Center for the Brain, University of Bergen, 5009, Bergen, Norway.
- Department of Radiology, Haukeland University Hospital, 5021, Bergen, Norway.
| |
Collapse
|
46
|
Lucido MJ, Dunlop BW. Emerging Medications for Treatment-Resistant Depression: A Review with Perspective on Mechanisms and Challenges. Brain Sci 2025; 15:161. [PMID: 40002494 PMCID: PMC11853532 DOI: 10.3390/brainsci15020161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Non-response to initial treatment options for major depressive disorder (MDD) is a common clinical challenge with profound deleterious impacts for affected patients. Few treatments have received regulatory approval for treatment-resistant depression (TRD). Methods: A systematic search of United States and European Union clinical trials registries was conducted to identify Phase II, III, or IV clinical trials, with a last update posted on or after 1 January 2020, that were evaluating medications for TRD. For both the US and EU registries, the condition term "treatment resistant depression" and associated lower-level terms (per registry search protocol) were used. For the US registry, a secondary search using the condition term "depressive disorders" and the modifying term "inadequate" was also performed to capture registrations not tagged as TRD. Two additional searches were also conducted in the US registry for the terms "suicide" and "anhedonia" as transdiagnostic targets of investigational medications. Trials were categorized based on the primary mechanism of action of the trial's investigational medication. Results: Fifty clinical trials for TRD, 20 for anhedonia, and 25 for suicide were identified. Glutamate system modulation was the mechanism currently with the most compounds in development, including antagonists and allosteric modulators of NMDA receptors, AMPA receptors, metabotropic type 2/3 glutamate receptors, and intracellular effector molecules downstream of glutamate signaling. Psychedelics have seen the greatest surge among mechanistic targets in the past 5 years, however, with psilocybin in particular garnering significant attention. Other mechanisms included GABA modulators, monoamine modulators, anti-inflammatory/immune-modulating agents, and an orexin type 2 receptor antagonist. Conclusions: These investigations offer substantial promise for more efficacious and potentially personalized medication approaches for TRD. Challenges for detecting efficacy in TRD include the heterogeneity within the TRD population stemming from the presumed variety of biological dysfunctions underlying the disorder, comorbid disorders, chronic psychosocial stressors, and enduring effects of prior serotonergic antidepressant medication treatments.
Collapse
Affiliation(s)
| | - Boadie W. Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA;
| |
Collapse
|
47
|
Sajid S, Mann JJ, Grunebaum MF. Clinical trials since 2020 of rapid anti-suicidal ideation effects of ketamine and its enantiomers: a systematic review. Transl Psychiatry 2025; 15:44. [PMID: 39915491 PMCID: PMC11802767 DOI: 10.1038/s41398-025-03255-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 12/12/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Suicide is a global public health problem with few empirically supported treatments. METHODS We conducted a systematic review of clinical trials (CT) since 2020 of racemic ketamine or one of its enantiomers' (R/S) potential to reduce suicidal ideation or behavior (SIB). An initial PubMed search on April 15th, 2024 yielded 2483 results. 104 relevant CTs were identified. An additional search using other search engines on March 19th, 2024 yielded 52 sources. After screening, 14 RCTs met the inclusion criteria which required clinically significant SIB among participants, ketamine or one of its enantiomers as an anti-SIB treatment, and SIB as an outcome. We excluded neuroimaging studies, meta-analyses, reviews, and case reports. Open-label studies were also excluded except in the case of R-ketamine where we included 2 open trials due to limited published data for this enantiomer, yielding a total of 16 CTs. We used the Revised Cochrane risk-of-bias tool for the RCTs. CTs reviewed had suicidal ideation (SI) but none had suicidal behavior as an outcome. RESULTS The studies include ketamine augmentation of other treatments such as electroconvulsive therapy (ECT), various routes of administration - intravenous (IV), intramuscular (IM), and intranasal (IN) - and single versus multiple dose designs. Multiple doses of IV ketamine/S-ketamine produced reductions in SI for periods of several days to weeks, while single doses showed shorter, more variable effects. Multiple and single doses of IN ketamine/S-ketamine and single doses of IV ketamine produced less consistent anti-SI results. IN and IV ketamine/S-ketamine administration appears to be well tolerated. R-ketamine appears to produce fewer side effects, but additional clinical research is needed to clarify its antidepressant and anti-SI effects in humans. CONCLUSION This review affirms the time-limited, anti-SI effects of ketamine and the need for personalized treatment. Limitations include study heterogeneity, small samples, and paucity of data for suicidal behavior or R-ketamine.
Collapse
Affiliation(s)
- Sumra Sajid
- Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, NY, USA
| | - J John Mann
- Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, NY, USA
| | - Michael F Grunebaum
- Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
48
|
Darwish MY, Helal AA, Othman YA, Mabrouk MA, Alrawi A, Ashraf TAE, Abdelsattar NK, Sayed FM, Abd-ElGawad M. Efficacy and safety of ketamine and esketamine in reducing the incidence of postpartum depression: an updated systematic review and meta-analysis. BMC Pregnancy Childbirth 2025; 25:125. [PMID: 39915701 PMCID: PMC11800651 DOI: 10.1186/s12884-025-07186-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/16/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Postpartum depression (PPD) is categorized by the Disorders-Fifth Edition as depression that begins during pregnancy or within the first month after giving birth. Ketamine and esketamine have shown promising results in the treatment of several depressive disorders, which suggests that they may have a role in the prevention of PPD. This systematic review and meta-analysis aim to update evidence about the efficacy and safety of using ketamine and esketamine to reduce PPD incidence. METHODS We searched four databases, PubMed, Scopus, Web of Science, and Cochrane, to collect relevant studies. We included studies which investigated the preventive effect of ketamine or esketamine on PPD among women after giving birth through caesarean or vaginal delivery. We extracted PPD occurrence rate, PPD score, pain score and side effects. Finally, a meta-analysis was conducted using RevMan software. RESULTS Twenty-one eligible studies were incorporated in the current systematic review and meta-analysis involving 4,389 pregnant women. Esketamine was the intervention in 14 studies, and ketamine was used in 7 studies. In subgroup analysis, both ketamine and esketamine were significantly effective in reducing the incidence of short-term PPD (ketamine: RR = 0.72, 95% CI [0.56, 0.93], P = 0.01; esketamine: RR = 0.43, P < 0.0001). Esketamine only significantly reduced the incidence of long-term PPD (RR = 0.44, P < 0.00001). Low doses and high doses were effective in reducing the incidence of both short-term (high dose: RR = 0.48, P = 0.0005; low dose: RR = 0.46, P = 0.002) and long-term PPD (high dose: RR = 0.54, P < 0.0001; low dose: RR = 0.61, P = 0.009). Regarding the risk of side effects, patients in the Ketamine/esketamine group showed statistically significant higher rates of developing dizziness (P = 0.0007), blurred vision (P = 0.02), vomiting (P = 0.004) and hallucinations (P = 0,002) than women in the control group. CONCLUSION Both ketamine and esketamine are effective in lowering the incidence of short-term PPD. On the other hand, only esketamine is effective in reducing the incidence of long-term PPD. It is recommended to use smaller doses for a more tolerable treatment period since doses less than 0.5 mg are significantly effective. Temporary side effects such as dizziness, blurred vision, vomiting and hallucinations were reported.
Collapse
Affiliation(s)
| | | | | | | | - Aya Alrawi
- Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | | | | | - Fatma Mohammed Sayed
- Department of Obstetrics and Gynecology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | | |
Collapse
|
49
|
Doan LV, Burr J, Perez R, Martinez H, Cuevas R, Watt K, Wang J. The Use of Electronic Consent (eConsent) Within the Ketamine for Long-Lasting Pain Relief After Surgery (KALPAS) Multicenter Trial. J Pain Res 2025; 18:589-595. [PMID: 39931425 PMCID: PMC11808973 DOI: 10.2147/jpr.s488917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 01/04/2025] [Indexed: 02/13/2025] Open
Abstract
Background The informed consent process has traditionally taken place in person. The introduction of electronic consent (eConsent) has made remote consenting processes possible. Use of eConsent has increased since the COVID-19 pandemic. It has streamlined the process of consenting patients and has been shown to benefit the research study team and participants. eConsent in the Ketamine Analgesia for Long-Lasting Pain Relief After Surgery Kalpas Study The KALPAS study is a multicenter, double-blind, randomized controlled study investigating the effectiveness of ketamine in reducing chronic post-mastectomy pain in women undergoing mastectomy for oncologic indication. The study uses a two-part consent form consisting of a master consent with information applicable to all sites and site-specific information. All potential participants receive the full two-part consent form for review. When signing the eConsent, however, all potential participants are provided with a concise summary of the informed consent document, an approach not widely used by multicenter studies. eConsent has been noted to be beneficial to research staff when trying to gather informed consent from participants who live far away from the hospital, want to include their family and friends, and for researchers who can approach patients outside of their clinical appointments. Conclusion The ability to consent patients remotely has allowed for a flexible workflow within sites and a more patient-centric process that focuses on including loved ones in the discussion and scheduling time to speak to a principal investigator. Demand for eConsent will likely continue in the post-COVID era, and use of a concise summary can allow for a more efficient consenting process.
Collapse
Affiliation(s)
- Lisa V Doan
- Department of Anesthesiology, Perioperative Care, and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Interdisciplinary Pain Research Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Jeri Burr
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Raven Perez
- Department of Anesthesiology, Perioperative Care, and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Interdisciplinary Pain Research Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Hamleini Martinez
- Department of Anesthesiology, Perioperative Care, and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Interdisciplinary Pain Research Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Randy Cuevas
- Department of Anesthesiology, Perioperative Care, and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Interdisciplinary Pain Research Program, New York University Grossman School of Medicine, New York, NY, USA
| | - Kevin Watt
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Jing Wang
- Department of Anesthesiology, Perioperative Care, and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Interdisciplinary Pain Research Program, New York University Grossman School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
50
|
Dai Y, Harrison BJ, Davey CG, Steward T. Towards an expanded neurocognitive account of ketamine's rapid antidepressant effects. Int J Neuropsychopharmacol 2025; 28:pyaf010. [PMID: 39921611 PMCID: PMC11879094 DOI: 10.1093/ijnp/pyaf010] [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: 09/24/2024] [Accepted: 02/06/2025] [Indexed: 02/10/2025] Open
Abstract
Ketamine is an N-methyl-D-aspartate receptor antagonist that has shown effectiveness as a rapidly acting treatment for depression. Although advances have been made in understanding ketamine's antidepressant pharmacological and molecular mechanisms of action, the large-scale neurocognitive mechanisms driving its therapeutic effects are less clearly understood. To help provide such a framework, we provide a synthesis of current evidence linking ketamine treatment to the modulation of brain systems supporting reward processing, interoception, and self-related cognition. We suggest that ketamine's antidepressant effects are, at least in part, driven by dynamic multi-level influences across these key functional domains.
Collapse
Affiliation(s)
- Yingliang Dai
- Department of Psychiatry, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Ben J Harrison
- Department of Psychiatry, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Christopher G Davey
- Department of Psychiatry, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Trevor Steward
- Department of Psychiatry, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
- Melbourne School of Psychological Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|