1
|
Asch RH, Abdallah CG, Carson RE, Esterlis I. Challenges and rewards of in vivo synaptic density imaging, and its application to the study of depression. Neuropsychopharmacology 2024; 50:153-163. [PMID: 39039139 PMCID: PMC11525584 DOI: 10.1038/s41386-024-01913-3] [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: 03/20/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/24/2024]
Abstract
The development of novel radiotracers for Positron Emission Tomography (PET) imaging agents targeting the synaptic vesicle glycoprotein 2 A (SV2A), an integral glycoprotein present in the membrane of all synaptic vesicles throughout the central nervous system, provides a method for the in vivo quantification of synaptic density. This is of particular interest in neuropsychiatric disorders given that synaptic alterations appear to underlie disease progression and symptom severity. In this review, we briefly describe the development of these SV2A tracers and the evaluation of quantification methods. Next, we discuss application of SV2A PET imaging to the study of depression, including a review of our findings demonstrating lower SV2A synaptic density in people with significant depressive symptoms and the use of a ketamine drug challenge to examine synaptogenesis in vivo. We then highlight the importance of performing translational PET imaging in animal models in conjunction with clinical imaging. We consider the ongoing challenges, possible solutions, and present preliminary findings from our lab demonstrating the translational benefit and potential of in vivo SV2A imaging in animal models of chronic stress. Finally, we discuss methodological improvements and future directions for SV2A imaging, potentially in conjunction with other neural markers.
Collapse
Affiliation(s)
- Ruth H Asch
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Chadi G Abdallah
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale School of Engineering, New Haven, CT, USA
| | - Irina Esterlis
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Department of Radiology and Biomedical Imaging, Yale Positron Emission Tomography Center, Yale School of Medicine, New Haven, CT, USA.
- U.S. Department of Veteran Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
2
|
Das K, Sen J, Borode AS. Ketamine and α-Amino-3-Hydroxy-5-Methyl-4-Isoxazolepropionic Acid (AMPA) Receptor Potentiation in the Somatosensory Cortex: A Comprehensive Review. Cureus 2024; 16:e69261. [PMID: 39398836 PMCID: PMC11470829 DOI: 10.7759/cureus.69261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Ketamine, a dissociative anesthetic primarily recognized for its antagonism of N-methyl-D-aspartate (NMDA) receptors, has gained significant attention for its rapid antidepressant effects and potential in treating mood disorders. However, recent research indicates that ketamine's influence extends beyond NMDA receptor inhibition, affecting α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors and sensory processing. This review delves into ketamine's role in enhancing AMPA receptor function and its implications for sensory processing within the somatosensory cortex. AMPA receptors, essential for fast excitatory neurotransmission and synaptic plasticity, play a key role in sensory perception and integration. By examining preclinical and clinical studies, this review sheds light on how ketamine's modulation of AMPA receptors may improve sensory processing and contribute to its therapeutic effects. Additionally, the review explores the potential for ketamine-based therapies to treat sensory processing disorders and refine current treatment strategies. A deeper understanding of ketamine's complex effects on AMPA receptors and sensory processing could provide valuable insights for developing targeted interventions and advancing clinical applications.
Collapse
Affiliation(s)
- Kaustuv Das
- Anaesthesiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Jayshree Sen
- Anaesthesiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Aishwarya S Borode
- Anaesthesiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
3
|
Ohtani Y, Tani H, Nomoto-Takahashi K, Yatomi T, Yonezawa K, Tomiyama S, Nagai N, Kusudo K, Honda S, Moriyama S, Nakajima S, Yamada T, Morisaki H, Iwabuchi Y, Jinzaki M, Yoshimura K, Eiro T, Tsugawa S, Ichijo S, Fujimoto Y, Miyazaki T, Takahashi T, Uchida H. Efficacy and safety of intravenous ketamine treatment in Japanese patients with treatment-resistant depression: A double-blind, randomized, placebo-controlled trial. Psychiatry Clin Neurosci 2024. [PMID: 39210712 DOI: 10.1111/pcn.13734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/17/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024]
Abstract
AIM Although the antidepressant effect of ketamine on treatment-resistant depression (TRD) has been frequently reported in North American and European countries, evidence is scarce among the Asian population. We aimed to evaluate the efficacy and safety of intravenous ketamine in Japanese patients with TRD. METHODS In this double-blind randomized placebo-controlled trial, 34 Japanese patients with TRD were randomized to receive either intravenous ketamine (0.5 mg/kg) or placebo, administered over 40 min, twice a week, for 2 weeks. The primary outcome was the change in the Montgomery Åsberg Depression Rating Scale (MADRS) total score from baseline to post-treatment. Secondary outcomes included changes in other depressive symptomatology scores and remission, response, and partial response rates. We also examined the association between baseline clinical demographic characteristics and changes in the MADRS total score. RESULTS Intention-to-treat analysis indicated no significant difference in the decrease in MADRS total score between the groups (-8.1 ± 10.0 vs -2.5 ± 5.2, t[32] = 2.02, P = 0.052), whereas per-protocol analysis showed a significant reduction in the ketamine group compared to the placebo group (-9.1 ± 10.2 vs -2.7 ± 5.3, t[29] = 2.22, P = 0.034). No significant group differences were observed in other outcomes. Adverse events were more frequent in the ketamine group than in the placebo group, and no serious adverse events were reported. A higher baseline MADRS total score and body mass index were associated with a greater reduction in the MADRS total score. CONCLUSION Intravenous ketamine outperformed placebo in Japanese patients with TRD who completed the study, suggesting that ketamine could alleviate depressive symptoms of TRD across diverse ethnic populations.
Collapse
Affiliation(s)
- Yohei Ohtani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Tani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | | | - Taisuke Yatomi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kengo Yonezawa
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Sota Tomiyama
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhiro Nagai
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
- Department of Psychiatry, Minami-Hanno Hospital, Saitama, Japan
| | - Keisuke Kusudo
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shiori Honda
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Sotaro Moriyama
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Takashige Yamada
- Department of Anesthesiology, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Morisaki
- Department of Anesthesiology, Keio University School of Medicine, Tokyo, Japan
| | - Yu Iwabuchi
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Jinzaki
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Kimio Yoshimura
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Eiro
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sakiko Tsugawa
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sadamitsu Ichijo
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yu Fujimoto
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoyuki Miyazaki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takuya Takahashi
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
4
|
Kwaśna J, Cubała WJ, Kwaśny A, Wilkowska A. The quest for optimal ketamine dosing formula in treatment-resistant major depressive disorder. Pharmacol Rep 2024:10.1007/s43440-024-00637-x. [PMID: 39222174 DOI: 10.1007/s43440-024-00637-x] [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: 06/21/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Emerging evidence indicates that intravenous ketamine is effective in managing treatment-resistant unipolar and bipolar depression. Clinical studies highlight its favorable efficacy, safety, and tolerability profile within a dosage range of 0.5-1.0 mg/kg based on actual body weight. However, data on alternative dosage calculation methods, particularly in relation to body mass index (BMI) and therapeutic outcomes, remain limited. METHODS This retrospective analysis of an open-label study aims to evaluate dose calculation strategies and their impact on treatment response among inpatients with treatment-resistant major depressive disorder (MDD) (n = 28). The study employed the Boer and Devine formulas to determine lean body mass (LBM) and ideal body weight (IBW), and the Mosteller formula to estimate body surface area (BSA). The calculated doses were then compared with the actual doses administered or converted to a dosage per square meter for both responders and non-responders. RESULTS Regardless of treatment response, defined as a reduction of 50% in the Montgomery-Åsberg Depression Rating Scale, the use of alternative ketamine dosing formulas resulted in underdosing compared to the standardized dose of 0.5 mg/kg. Only two participants received higher doses (102.7% and 113.0%) when the Devine formula was applied. CONCLUSIONS This study suggests that ketamine dosing formulas, alternative to the standardized 0.5 mg/kg based on body weight, may lead to underdosing and potentially impact outcome interpretation. To enhance dosing accuracy, future studies should consider incorporating body impedance analysis and waist-to-hip ratio measurements, as this study did not account for body composition.
Collapse
Affiliation(s)
- Julia Kwaśna
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, 80-214, Poland.
| | - Wiesław Jerzy Cubała
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, 80-214, Poland
| | - Aleksander Kwaśny
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, 80-214, Poland
| | - Alina Wilkowska
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, 80-214, Poland
| |
Collapse
|
5
|
Miller EA, Afshar HT, Mishra J, McIntyre RS, Ramanathan D. Predicting non-response to ketamine for depression: An exploratory symptom-level analysis of real-world data among military veterans. Psychiatry Res 2024; 335:115858. [PMID: 38547599 DOI: 10.1016/j.psychres.2024.115858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/04/2024] [Accepted: 03/09/2024] [Indexed: 04/14/2024]
Abstract
Ketamine helps some patients with treatment resistant depression (TRD), but reliable methods for predicting which patients will, or will not, respond to treatment are lacking. Herein, we aim to inform prediction models of non-response to ketamine/esketamine in adults with TRD. This is a retrospective analysis of PHQ-9 item response data from 120 patients with TRD who received repeated doses of intravenous racemic ketamine or intranasal eskatamine in a real-world clinic. Regression models were fit to patients' symptom trajectories, showing that all symptoms improved on average, but depressed mood improved relatively faster than low energy. Principal component analysis revealed a first principal component (PC) representing overall treatment response, and a second PC that reflects variance across affective versus somatic symptom subdomains. We then trained logistic regression classifiers to predict overall response (improvement on PC1) better than chance using patients' baseline symptoms alone. Finally, by parametrically adjusting the classifier decision thresholds, we identified optimal models for predicting non-response with a negative predictive value of over 96 %, while retaining a specificity of 22 %. Thus, we could identify 22 % of patients who would not respond based purely on their baseline symptoms. This approach could inform rational treatment recommendations to avoid additional treatment failures.
Collapse
Affiliation(s)
- Eric A Miller
- Department of Mental Health, VA San Diego Medical Center, San Diego, CA 92161, USA; Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA
| | - Houtan Totonchi Afshar
- Department of Mental Health, VA San Diego Medical Center, San Diego, CA 92161, USA; Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA
| | - Jyoti Mishra
- Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA; Center of Excellence for Stress and Mental Health, VA San Diego Medical Center, USA
| | - Roger S McIntyre
- Department of Psychiatry, University of Toronto, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada; Brain and Cognition Discovery Foundation, Toronto, Canada
| | - Dhakshin Ramanathan
- Department of Mental Health, VA San Diego Medical Center, San Diego, CA 92161, USA; Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA; Center of Excellence for Stress and Mental Health, VA San Diego Medical Center, USA.
| |
Collapse
|
6
|
Lineham A, Avila-Quintero VJ, Bloch MH, Dwyer J. Exploring Predictors of Ketamine Response in Adolescent Treatment-Resistant Depression. J Child Adolesc Psychopharmacol 2024; 34:73-79. [PMID: 38170185 PMCID: PMC11262580 DOI: 10.1089/cap.2023.0047] [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: 01/05/2024]
Abstract
Objective: Ketamine has proved effective as a rapid-acting antidepressant agent, but treatment is not effective for everyone (approximately a quarter to a half of patients). Some adult studies have begun to investigate predictors of ketamine's antidepressant response, but no studies have examined this in adolescents with depression. Methods: We conducted a secondary data analysis of adolescents who participated in a randomized, single-dose, midazolam-controlled crossover trial of ketamine for adolescents with treatment-resistant depression. We examined the relationship between 19 exploratory demographic and clinical variables and depression symptom improvement (using the Montgomery-Åsberg Depression Rating Scale [MADRS]) at 1 and 7 days postinfusion. Results: Subjects who had fewer medication trials of both antidepressant medications and augmentation treatments were more likely to experience depression symptom improvement with ketamine. Subjects with shorter duration of their current depressive episode were more likely to experience depression symptom improvement with ketamine. Subjects currently being treated with selective serotonin reuptake inhibitor medications, and not being treated with serotonin-norepinephrine reuptake inhibitor medications, also experienced greater symptom improvement with ketamine. When receiving the midazolam control, less severe depressive symptoms, as measured by the Children's Depression Rating Scale (CDRS) (but not MADRS), and a comorbid attention-deficit/hyperactivity disorder diagnosis were associated with increased response. Conclusions: Findings should be viewed as preliminary and exploratory given the small sample size and multiple secondary analyses. Identifying meaningful predictors of ketamine response is important to inform future therapeutic use of this compound, however, considerably more research is warranted before such clinical guidance is established. The trial was registered in clinicaltrials.gov with the identifier NCT02579928.
Collapse
Affiliation(s)
- Alice Lineham
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Michael H. Bloch
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry and Yale School of Medicine, New Haven, Connecticut, USA
| | - Jennifer Dwyer
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
7
|
Yonezawa K, Uchida H, Yatomi T, Ohtani Y, Nomoto-Takahashi K, Nakajima S, Mimura M, Tani H. Factors Associated with Antidepressant Effects of Ketamine: A Reanalysis of Double-Blind Randomized Placebo-Controlled Trial of Intravenous Ketamine for Treatment-Resistant Depression. PHARMACOPSYCHIATRY 2024; 57:35-40. [PMID: 37846462 DOI: 10.1055/a-2179-8884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
INTRODUCTION Predictors of treatment response to intravenous ketamine remain unclear in patients with treatment-resistant depression (TRD); therefore, this study aimed to clarify these predictors using the US National Institutes of Health database of clinical trials. METHODS Data from a placebo-controlled, double-blind, randomized controlled trial were used to assess the efficacy of intravenous ketamine in adult patients with TRD (NCT01920555). For the analysis, data were used from the participants who had received therapeutic doses of intravenous ketamine (i. e., 0.5 and 1.0 mg/kg). Logistic and multivariable regression analyses were conducted to explore the demographic and clinical factors associated with response to treatment or changes in the Hamilton Depression Rating Scale 6 items (HAM-D-6) total score. RESULTS This study included 31 patients with TRD (13 women; mean±standard deviation age, 48.4±10.9 years). Logistic regression analysis showed that the age of onset was positively correlated with treatment response after three days of ketamine administration (β=0.08, p=0.037); however, no association was observed between treatment response and age, sex, baseline HAM-D-6 total score, or dissociative score assessed with the Clinician-Administered Dissociative States Scale 40 min after ketamine infusion. Multiple regression analysis showed that no factors were correlated significantly with the percentage change in the HAM-D-6 total score three days after ketamine administration. DISCUSSION Later disease onset correlates with a better treatment response three days after ketamine infusion in patients with TRD. Glutamatergic signal transmission may be impaired in patients with an earlier onset of depression, resulting in decreased neuroplasticity, which diminishes ketamine response.
Collapse
Affiliation(s)
- Kengo Yonezawa
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Taisuke Yatomi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Ohtani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | | | - Shinichiro Nakajima
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Tani
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
O'Brien B, Lee J, Kim S, Nandra GS, Pannu P, Tamman A, Amarneh D, Swann AC, Murphy N, Averill L, Jha M, Mathew SJ. Anti-suicidal effects of IV ketamine in a real-world setting. Psychiatry Res 2024; 331:115604. [PMID: 38064911 DOI: 10.1016/j.psychres.2023.115604] [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: 07/07/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 01/02/2024]
Abstract
The current study evaluated the effectiveness of intravenous ketamine treatment for suicidality in a community-based clinical sample of 295 outpatients (mean age= 40.37; 58.6 % male). We conducted growth mixture modeling to estimate latent classes of changes in symptoms of suicidality measured by the Concise Health Risk Tracking - Self-Report (CHRT-SR) across five infusions in a two-week course of treatment. Best-fit indices indicated three trajectory groups demonstrating non-linear, quadratic changes in CHRT-SR scores during ketamine treatment. The largest group of patients (n= 170, 57.6 %) had moderate CHRT-SR scores at baseline and showed gradual improvement during treatment. The other two groups of patients had severe CHRT-SR scores at baseline and diverged into one group with no improvement throughout treatment (n = 63, 21 %) and one group with rapid improvement (n = 62, 21 %). Of the clinical and demographic variables available and tested, only higher scores pertaining to active thoughts of death and/or plan were found to predict which of the patients with severe CHRT-SR scores at baseline would not benefit from treatment. The present study provides an important contribution to the knowledge of ketamine's effects on symptoms related to suicide over time. providing support for the possible effectiveness of ketamine in a proportion of patients.
Collapse
Affiliation(s)
- Brittany O'Brien
- Michael E. DeBakey VA Medical Center, 2002 Holcombe Boulevard, Houston, TX 77030, USA; Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, 1977 Butler Boulevard, Houston, TX 77030, USA. Brittany.O'
| | - Jaehoon Lee
- Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, 1977 Butler Boulevard, Houston, TX 77030, USA; Texas Tech University, Department of Educational Psychology and Leadership, 2500 Broadway, Lubbock, TX 79409, USA; The Menninger Clinic, 12301 S Main Street, Houston, TX 77035, USA
| | - Seungman Kim
- Texas Tech University, Department of Educational Psychology and Leadership, 2500 Broadway, Lubbock, TX 79409, USA
| | - Guriqbal S Nandra
- IV Solution and Ketamine Centers of Chicago and Kansas City, 712 North Dearborn Street, Chicago, IL 60654, USA
| | - Prabhneet Pannu
- IV Solution and Ketamine Centers of Chicago and Kansas City, 712 North Dearborn Street, Chicago, IL 60654, USA
| | - Amanda Tamman
- Michael E. DeBakey VA Medical Center, 2002 Holcombe Boulevard, Houston, TX 77030, USA; Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, 1977 Butler Boulevard, Houston, TX 77030, USA
| | - Dania Amarneh
- Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, 1977 Butler Boulevard, Houston, TX 77030, USA
| | - Alan C Swann
- Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, 1977 Butler Boulevard, Houston, TX 77030, USA
| | - Nicholas Murphy
- Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, 1977 Butler Boulevard, Houston, TX 77030, USA
| | - Lynnette Averill
- Michael E. DeBakey VA Medical Center, 2002 Holcombe Boulevard, Houston, TX 77030, USA; Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, 1977 Butler Boulevard, Houston, TX 77030, USA
| | - Manish Jha
- UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Sanjay J Mathew
- Michael E. DeBakey VA Medical Center, 2002 Holcombe Boulevard, Houston, TX 77030, USA; Baylor College of Medicine, Menninger Department of Psychiatry and Behavioral Sciences, 1977 Butler Boulevard, Houston, TX 77030, USA; The Menninger Clinic, 12301 S Main Street, Houston, TX 77035, USA
| |
Collapse
|
9
|
Costi S. Ketamine for Major Depressive Disorder. Curr Top Behav Neurosci 2024; 66:131-147. [PMID: 37922100 DOI: 10.1007/7854_2023_453] [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] [Indexed: 11/05/2023]
Abstract
Major Depressive Disorder (MDD) is a leading cause of disability worldwide. Conventional antidepressant treatment is characterised by a significant time to onset of therapeutic action (approximately 2 weeks) and fails to achieve a stable remission of symptoms in one-third of subjects with MDD. In the last 20 years the discovery of antidepressant effects of the N-methyl-d-aspartate (NMDA) receptor antagonist ketamine as a rapid acting (within hours) and sustained (up to 7 days) antidepressant has represented a major paradigm shift in the field.The present chapter reviews the pharmacology, safety, and efficacy of ketamine as a novel therapeutic agent for MDD and specifically for subjects who did not respond to conventional antidepressant (treatment resistant depression). The impact of ketamine on suicidal ideation, the availability of brain biomarkers of ketamine treatment response and the association of ketamine and psychotherapy are considered.
Collapse
Affiliation(s)
- Sara Costi
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK.
- Oxford Health Foundation Trust, Warneford Hospital, Oxford, UK.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Wade B, Pindale R, Camprodon J, Luccarelli J, Li S, Meisner R, Seiner S, Henry M. Individual Prediction of Optimal Treatment Allocation Between Electroconvulsive Therapy or Ketamine using the Personalized Advantage Index. RESEARCH SQUARE 2023:rs.3.rs-3682009. [PMID: 38077094 PMCID: PMC10705694 DOI: 10.21203/rs.3.rs-3682009/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Introduction Electroconvulsive therapy (ECT) and ketamine are two effective treatments for depression with similar efficacy; however, individual patient outcomes may be improved by models that predict optimal treatment assignment. Here, we adapt the Personalized Advantage Index (PAI) algorithm using machine learning to predict optimal treatment assignment between ECT and ketamine using medical record data from a large, naturalistic patient cohort. We hypothesized that patients who received a treatment predicted to be optimal would have significantly better outcomes following treatment compared to those who received a non-optimal treatment. Methods Data on 2526 ECT and 235 mixed IV ketamine and esketamine patients from McLean Hospital was aggregated. Depressive symptoms were measured using the Quick Inventory of Depressive Symptomatology (QIDS) before and during acute treatment. Patients were matched between treatments on pretreatment QIDS, age, inpatient status, and psychotic symptoms using a 1:1 ratio yielding a sample of 470 patients (n=235 per treatment). Random forest models were trained and predicted differential patientwise minimum QIDS scores achieved during acute treatment (min-QIDS) scores for ECT and ketamine using pretreatment patient measures. Analysis of Shapley Additive exPlanations (SHAP) values identified predictors of differential outcomes between treatments. Results Twenty-seven percent of patients with the largest PAI scores who received a treatment predicted optimal had significantly lower min-QIDS scores compared to those who received a non-optimal treatment (mean difference=1.6, t=2.38, q<0.05, Cohen's D=0.36). Analysis of SHAP values identified prescriptive pretreatment measures. Conclusions Patients assigned to a treatment predicted to be optimal had significantly better treatment outcomes. Our model identified pretreatment patient factors captured in medical records that can provide interpretable and actionable guidelines treatment selection.
Collapse
Affiliation(s)
- Benjamin 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
| | - Joan Camprodon
- 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 Meisner
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, McLean Hospital, Belmont, MA, USA
| | - Stephen Seiner
- Department of Psychiatry, McLean Hospital, Belmont, MA, USA
| | - Michael Henry
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Medeiros GC, Matheson M, Demo I, Reid MJ, Matheson S, Twose C, Smith GS, Gould TD, Zarate CA, Barrett FS, Goes FS. Brain-based correlates of antidepressant response to ketamine: a comprehensive systematic review of neuroimaging studies. Lancet Psychiatry 2023; 10:790-800. [PMID: 37625426 PMCID: PMC11534374 DOI: 10.1016/s2215-0366(23)00183-9] [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: 01/26/2023] [Revised: 04/28/2023] [Accepted: 05/12/2023] [Indexed: 08/27/2023]
Abstract
Ketamine is an effective antidepressant, but there is substantial variability in patient response and the precise mechanism of action is unclear. Neuroimaging can provide predictive and mechanistic insights, but findings are limited by small sample sizes. This systematic review covers neuroimaging studies investigating baseline (pre-treatment) and longitudinal (post-treatment) biomarkers of responses to ketamine. All modalities were included. We performed searches of five electronic databases (from inception to April 26, 2022). 69 studies were included (with 1751 participants). There was substantial methodological heterogeneity and no well replicated biomarker. However, we found convergence across some significant results, particularly in longitudinal biomarkers. Response to ketamine was associated with post-treatment increases in gamma power in frontoparietal regions in electrophysiological studies, post-treatment increases in functional connectivity within the prefrontal cortex, and post-treatment increases in the functional activation of the striatum. Although a well replicated neuroimaging biomarker of ketamine response was not identified, there are biomarkers that warrant further investigation.
Collapse
Affiliation(s)
- Gustavo C Medeiros
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Malcolm Matheson
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Isabella Demo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew J Reid
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Claire Twose
- Welch Medical Library, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gwenn S Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA; Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, NIMH-NIH, Bethesda, MD, USA
| | - Frederick S Barrett
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Department of Psychological and Brain Sciences, and Center for Psychedelic and Consciousness Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fernando S Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Fancy F, Haikazian S, Johnson DE, Chen-Li DCJ, Levinta A, Husain MI, Mansur RB, Rosenblat JD. Ketamine for bipolar depression: an updated systematic review. Ther Adv Psychopharmacol 2023; 13:20451253231202723. [PMID: 37771417 PMCID: PMC10524067 DOI: 10.1177/20451253231202723] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/31/2023] [Indexed: 09/30/2023] Open
Abstract
Background The therapeutic potential of subanesthetic doses of ketamine appears promising in unipolar depression; however, its effectiveness in treating bipolar depression (BD) remains uncertain. Objective This systematic review aimed to summarize findings on the use of ketamine for the treatment of BD by assessing its efficacy, safety, and tolerability. Design Systematic review. Methods We conducted a systematic review of studies that investigated the use of ketamine for adults with BD. We searched PubMed and Embase for relevant randomized-controlled trials, open-label trials, and retrospective chart analyses published from inception to 13 March 2023. Results Eight studies were identified [pooled n = 235; mean (SD) age: 45.55 (5.54)]. All participants who received intravenous (IV) ketamine were administered a dose of 0.5-0.75 mg/kg as an adjunctive treatment to a mood-stabilizing agent, whereas participants who received esketamine were administered a dosage ranging from 28 to 84 mg. Flexible dosing was used in real-world analyses. A total of 48% of participants receiving ketamine achieved a response (defined as ⩾50% reduction in baseline depression severity), whereas only 5% achieved a response with a placebo. Real-world studies demonstrated lower rates of response (30%) compared to the average across clinical trials (63%). Reductions in suicidal ideation were noted in some studies, although not all findings were statistically significant. Ketamine and esketamine were well tolerated in most participants; however, six participants (2% of the overall sample pool, 5 receiving ketamine) developed hypomanic/manic symptoms after infusions. Significant dissociative symptoms were observed at the 40-min mark in some trials. Conclusion Preliminary evidence suggests IV ketamine as being safe and effective for the treatment of BD. Future studies should focus on investigating the effects of repeated acute and maintenance infusions using a randomized study design.
Collapse
Affiliation(s)
- Farhan Fancy
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Sipan Haikazian
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Danica E. Johnson
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - David C. J. Chen-Li
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Anastasia Levinta
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Muhammad I. Husain
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Rodrigo B. Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Joshua D. Rosenblat
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, 399 Bathurst Street, Toronto, ON M5T 2S8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Braxia Scientific, Braxia Health, Canadian Rapid Treatment Centre of Excellence, Mississauga, ON, Canada
| |
Collapse
|
13
|
Charlton CE, Karvelis P, McIntyre RS, Diaconescu AO. Suicide prevention and ketamine: insights from computational modeling. Front Psychiatry 2023; 14:1214018. [PMID: 37457775 PMCID: PMC10342546 DOI: 10.3389/fpsyt.2023.1214018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Suicide is a pressing public health issue, with over 700,000 individuals dying each year. Ketamine has emerged as a promising treatment for suicidal thoughts and behaviors (STBs), yet the complex mechanisms underlying ketamine's anti-suicidal effect are not fully understood. Computational psychiatry provides a promising framework for exploring the dynamic interactions underlying suicidality and ketamine's therapeutic action, offering insight into potential biomarkers, treatment targets, and the underlying mechanisms of both. This paper provides an overview of current computational theories of suicidality and ketamine's mechanism of action, and discusses various computational modeling approaches that attempt to explain ketamine's anti-suicidal effect. More specifically, the therapeutic potential of ketamine is explored in the context of the mismatch negativity and the predictive coding framework, by considering neurocircuits involved in learning and decision-making, and investigating altered connectivity strengths and receptor densities targeted by ketamine. Theory-driven computational models offer a promising approach to integrate existing knowledge of suicidality and ketamine, and for the extraction of model-derived mechanistic parameters that can be used to identify patient subgroups and personalized treatment approaches. Future computational studies on ketamine's mechanism of action should optimize task design and modeling approaches to ensure parameter reliability, and external factors such as set and setting, as well as psychedelic-assisted therapy should be evaluated for their additional therapeutic value.
Collapse
Affiliation(s)
- Colleen E. Charlton
- Krembil Center for Neuroinformatics, Center for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Povilas Karvelis
- Krembil Center for Neuroinformatics, Center for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Roger S. McIntyre
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Andreea O. Diaconescu
- Krembil Center for Neuroinformatics, Center for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Johnston JN, Greenwald MS, Henter ID, Kraus C, Mkrtchian A, Clark NG, Park LT, Gold P, Zarate CA, Kadriu B. Inflammation, stress and depression: An exploration of ketamine's therapeutic profile. Drug Discov Today 2023; 28:103518. [PMID: 36758932 PMCID: PMC10050119 DOI: 10.1016/j.drudis.2023.103518] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/13/2022] [Accepted: 01/31/2023] [Indexed: 02/09/2023]
Abstract
Well-established animal models of depression have described a proximal relationship between stress and central nervous system (CNS) inflammation - a relationship mirrored in the peripheral inflammatory biomarkers of individuals with depression. Evidence also suggests that stress-induced proinflammatory states can contribute to the neurobiology of treatment-resistant depression. Interestingly, ketamine, a rapid-acting antidepressant, can partially exert its therapeutic effects via anti-inflammatory actions on the hypothalamic-pituitary adrenal (HPA) axis, the kynurenine pathway or by cytokine suppression. Further investigations into the relationship between ketamine, inflammation and stress could provide insight into ketamine's unique therapeutic mechanisms and stimulate efforts to develop rapid-acting, anti-inflammatory-based antidepressants.
Collapse
Affiliation(s)
- Jenessa N Johnston
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Maximillian S Greenwald
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ioline D Henter
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Christoph Kraus
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Anahit Mkrtchian
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Neil G Clark
- US School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Lawrence T Park
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Philip Gold
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
International pooled patient-level meta-analysis of ketamine infusion for depression: In search of clinical moderators. Mol Psychiatry 2022; 27:5096-5112. [PMID: 36071111 PMCID: PMC9763119 DOI: 10.1038/s41380-022-01757-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 01/14/2023]
Abstract
Depression is disabling and highly prevalent. Intravenous (IV) ketamine displays rapid-onset antidepressant properties, but little is known regarding which patients are most likely to benefit, limiting personalized prescriptions. We identified randomized controlled trials of IV ketamine that recruited individuals with a relevant psychiatric diagnosis (e.g., unipolar or bipolar depression; post-traumatic stress disorder), included one or more control arms, did not provide any other study-administered treatment in conjunction with ketamine (although clinically prescribed concurrent treatments were allowable), and assessed outcome using either the Montgomery-Åsberg Depression Rating Scale or the Hamilton Rating Scale for Depression (HRSD-17). Individual patient-level data for at least one outcome was obtained from 17 of 25 eligible trials [pooled n = 809]. Rates of participant-level data availability across 33 moderators that were solicited from these 17 studies ranged from 10.8% to 100% (median = 55.6%). After data harmonization, moderators available in at least 40% of the dataset were tested sequentially, as well as with a data-driven, combined moderator approach. Robust main effects of ketamine on acute [~24-hours; β*(95% CI) = 0.58 (0.44, 0.72); p < 0.0001] and post-acute [~7 days; β*(95% CI) = 0.38 (0.23, 0.54); p < 0.0001] depression severity were observed. Two study-level moderators emerged as significant: ketamine effects (relative to placebo) were larger in studies that required a higher degree of previous treatment resistance to federal regulatory agency-approved antidepressant medications (≥2 failed trials) for study entry; and in studies that used a crossover design. A comprehensive data-driven search for combined moderators identified statistically significant, but modest and clinically uninformative, effects (effect size r ≤ 0.29, a small-medium effect). Ketamine robustly reduces depressive symptoms in a heterogeneous range of patients, with benefit relative to placebo even greater in patients more resistant to prior medications. In this largest effort to date to apply precision medicine approaches to ketamine treatment, no clinical or demographic patient-level features were detected that could be used to guide ketamine treatment decisions.Review Registration: PROSPERO Identifier: CRD42021235630.
Collapse
|
16
|
Kawazoe K, McGlynn R, Felix W, Sevilla R, Liao S, Kulkarni P, Ferris CF. Dose-dependent effects of esketamine on brain activity in awake mice: A BOLD phMRI study. Pharmacol Res Perspect 2022; 10:e01035. [PMID: 36504448 PMCID: PMC9743060 DOI: 10.1002/prp2.1035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 12/14/2022] Open
Abstract
Pharmacological magnetic resonance imaging (phMRI) is a noninvasive method used to evaluate neural circuitry involved in the behavioral effects of drugs like ketamine, independent of their specific biochemical mechanism. The study was designed to evaluate the immediate effect of esketamine, the S-isomer of (±) ketamine on brain activity in awake mice using blood oxygenation level dependent (BOLD) imaging. It was hypothesized the prefrontal cortex, hippocampus, and brain areas associated with reward and motivation would show a dose-dependent increase in brain activity. Mice were given vehicle, 1.0, 3.3, or 10 mg/kg esketamine I.P. and imaged for 10 min post-treatment. Data for each treatment were registered to a 3D MRI mouse brain atlas providing site-specific information on 134 different brain areas. There was a global change in brain activity for both positive and negative BOLD signal affecting over 50 brain areas. Many areas showed a dose-dependent decrease in positive BOLD signal, for example, cortex, hippocampus, and thalamus. The most common profile when comparing the three doses was a U-shape with the 3.3 dose having the lowest change in signal. At 1.0 mg/kg there was a significant increase in positive BOLD in forebrain areas and hippocampus. The anticipated dose-dependent increase in BOLD was not realized; instead, the lowest dose of 1.0 mg/kg had the greatest effect on brain activity. The prefrontal cortex and hippocampus were significantly activated corroborating previous imaging studies in humans and animals. The unexpected sensitivity to the 1.0 mg/kg dose of esketamine could be explained by imaging in fully awake mice without the confound of anesthesia and/or its greater affinity for the N-methyl-d-aspartate receptor (NMDAR) receptor than (±) ketamine.
Collapse
Affiliation(s)
- Kyrsten Kawazoe
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Ryan McGlynn
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Wilder Felix
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Raquel Sevilla
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Siyang Liao
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Praveen Kulkarni
- Center for Translational NeuroimagingNortheastern UniversityMassachusettsBostonUSA
| | - Craig F. Ferris
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
- Center for Translational NeuroimagingNortheastern UniversityMassachusettsBostonUSA
- Department of PsychologyNortheastern UniversityBostonMassachusettsUSA
| |
Collapse
|
17
|
At-home, sublingual ketamine telehealth is a safe and effective treatment for moderate to severe anxiety and depression: Findings from a large, prospective, open-label effectiveness trial. J Affect Disord 2022; 314:59-67. [PMID: 35809678 DOI: 10.1016/j.jad.2022.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND At-home Ketamine-assisted therapy (KAT) with psychosocial support and remote monitoring through telehealth platforms addresses access barriers, including the COVID-19 pandemic. Large-scale evaluation of this approach is needed for questions regarding safety and effectiveness for depression and anxiety. METHODS In this prospective study, a large outpatient sample received KAT over four weeks through a telehealth provider. Symptoms were assessed using the Patient Health Questionnaire (PHQ-9) for depression, and the Generalized Anxiety Disorder scale (GAD-7) for anxiety. Demographics, adverse events, and patient-reported dissociation were also analyzed. Symptom trajectories were identified using Growth Mixture Modeling, along with outcome predictors. RESULTS A sample of 1247 completed treatment with sufficient data, 62.8 % reported a 50 % or greater improvement on the PHQ-9, d = 1.61, and 62.9 % on the GAD-7, d = 1.56. Remission rates were 32.6 % for PHQ-9 and 31.3 % for GAD-7, with 0.9 % deteriorating on the PHQ-9, and 0.6 % on the GAD-7. Four patients left treatment early due to side effects or clinician disqualification, and two more due to adverse events. Three patient subpopulations emerged, characterized by Improvement (79.3 %), Chronic (11.4 %), and Delayed Improvement (9.3 %) for PHQ-9 and GAD-7. Endorsing side effects at Session 2 was associated with delayed symptom improvement, and Chronic patients were more likely than the other two groups to report dissociation at Session 4. CONCLUSION At-home KAT response and remission rates indicated rapid and significant antidepressant and anxiolytic effects. Rates were consistent with laboratory- and clinic-administered ketamine treatment. Patient screening and remote monitoring maintained low levels of adverse events. Future research should assess durability of effects.
Collapse
|
18
|
Chen MH, Cheng CM, Li CT, Tsai SJ, Lin WC, Bai YM, Su TP. Comparative study of low-dose ketamine infusion and repetitive transcranial magnetic stimulation in treatment-resistant depression: A posthoc pooled analysis of two randomized, double-blind, placebo-controlled studies. Psychiatry Res 2022; 316:114749. [PMID: 35940087 DOI: 10.1016/j.psychres.2022.114749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND This posthoc analysis compared the antidepressant and antisuicidal effects of low-dose ketamine infusion with those of repetitive transcranial magnetic stimulation (rTMS) on treatment-resistant depression (TRD). METHODS In the ketamine infusion trial, 48 patients with TRD were randomized to receive a single infusion of 0.5 mg/kg ketamine or normal saline. In the rTMS trial, 105 patients were randomly assigned to intermittent theta-burst stimulation (iTBS), 10-Hz rTMS, or sham stimulation. The 17-item Hamilton Rating Scale for Depression (HDRS) was administered. RESULTS The antidepressant effect was prominent at Day 7 postinfusion in the ketamine group but steadily accumulated with the treatment duration from Day 7 to 14 in the iTBS and 10-Hz rTMS groups, regardless of the level of treatment resistance (all p < .01). Low-dose ketamine infusion and iTBS exerted superior effects on suicidal symptoms (HDRS item 3) than the other three groups (p < .001). The antidepressant effect of iTBS/10-Hz rTMS may persist for up to 3 months; however, the antidepressant effect of a single low-dose ketamine infusion did not persist over a month. DISCUSSION Both low-dose ketamine infusion and rTMS/TBS must be included in TRD treatment but may be applied in different clinical situations.
Collapse
Affiliation(s)
- Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, No. 201, Sec.2, Shih-Pai Road, Beitou district, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Chih-Ming Cheng
- Department of Psychiatry, Taipei Veterans General Hospital, No. 201, Sec.2, Shih-Pai Road, Beitou district, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Cheng-Ta Li
- Department of Psychiatry, Taipei Veterans General Hospital, No. 201, Sec.2, Shih-Pai Road, Beitou district, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, No. 201, Sec.2, Shih-Pai Road, Beitou district, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Chen Lin
- Department of Psychiatry, Taipei Veterans General Hospital, No. 201, Sec.2, Shih-Pai Road, Beitou district, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ya-Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, No. 201, Sec.2, Shih-Pai Road, Beitou district, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tung-Ping Su
- Department of Psychiatry, Taipei Veterans General Hospital, No. 201, Sec.2, Shih-Pai Road, Beitou district, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan.
| |
Collapse
|
19
|
Medeiros GC, Gould TD, Prueitt WL, Nanavati J, Grunebaum MF, Farber NB, Singh B, Selvaraj S, Machado-Vieira R, Achtyes ED, Parikh SV, Frye MA, Zarate CA, Goes FS. Blood-based biomarkers of antidepressant response to ketamine and esketamine: A systematic review and meta-analysis. Mol Psychiatry 2022; 27:3658-3669. [PMID: 35760879 PMCID: PMC9933928 DOI: 10.1038/s41380-022-01652-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/17/2022] [Accepted: 05/31/2022] [Indexed: 02/08/2023]
Abstract
(R,S)-ketamine (ketamine) and its enantiomer (S)-ketamine (esketamine) can produce rapid and substantial antidepressant effects. However, individual response to ketamine/esketamine is variable, and there are no well-accepted methods to differentiate persons who are more likely to benefit. Numerous potential peripheral biomarkers have been reported, but their current utility is unclear. We conducted a systematic review/meta-analysis examining the association between baseline levels and longitudinal changes in blood-based biomarkers, and response to ketamine/esketamine. Of the 5611 citations identified, 56 manuscripts were included (N = 2801 participants), and 26 were compatible with meta-analytical calculations. Random-effect models were used, and effect sizes were reported as standardized mean differences (SMD). Our assessments revealed that more than 460 individual biomarkers were examined. Frequently studied groups included neurotrophic factors (n = 15), levels of ketamine and ketamine metabolites (n = 13), and inflammatory markers (n = 12). There were no consistent associations between baseline levels of blood-based biomarkers, and response to ketamine. However, in a longitudinal analysis, ketamine responders had statistically significant increases in brain-derived neurotrophic factor (BDNF) when compared to pre-treatment levels (SMD [95% CI] = 0.26 [0.03, 0.48], p = 0.02), whereas non-responders showed no significant changes in BDNF levels (SMD [95% CI] = 0.05 [-0.19, 0.28], p = 0.70). There was no consistent evidence to support any additional longitudinal biomarkers. Findings were inconclusive for esketamine due to the small number of studies (n = 2). Despite a diverse and substantial literature, there is limited evidence that blood-based biomarkers are associated with response to ketamine, and no current evidence of clinical utility.
Collapse
Affiliation(s)
- Gustavo C. Medeiros
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.,Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Todd D. Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Departments of Pharmacology and Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | | | - Julie Nanavati
- Welch Medical Library, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael F. Grunebaum
- Columbia University Irving Medical Center and New York State Psychiatric Institute, New York City, NY, USA
| | - Nuri B. Farber
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Balwinder Singh
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Sudhakar Selvaraj
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Rodrigo Machado-Vieira
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Eric D. Achtyes
- Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA.,Pine Rest Christian Mental Health Services, Grand Rapids, MI, USA
| | - Sagar V. Parikh
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Mark A. Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Carlos A. Zarate
- Experimental Therapeutics & Pathophysiology Branch, NIMH-NIH, Bethesda, MD, USA
| | - Fernando S. Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.,Correspondence and requests for materials should be addressed to Fernando S. Goes.,
| |
Collapse
|
20
|
Jesus-Nunes AP, Leal GC, Correia-Melo FS, Vieira F, Mello RP, Caliman-Fontes AT, Echegaray MVF, Marback RF, Guerreiro-Costa LNF, Souza-Marques B, Santos-Lima C, Souza LS, Bandeira ID, Kapczinski F, Lacerda ALT, Quarantini LC. Clinical predictors of depressive symptom remission and response after racemic ketamine and esketamine infusion in treatment-resistant depression. Hum Psychopharmacol 2022; 37:e2836. [PMID: 35179810 DOI: 10.1002/hup.2836] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 01/11/2022] [Accepted: 02/07/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a leading cause of disability worldwide and most people do not achieve symptom remission. Treatment-resistant depression (TRD) is characterized by the failure of at least one adequate trial of a major class of antidepressant, with adequate time and dosage. We aimed to identify clinical predictors of depressive symptom remission and response 24 h and 7 days after racemic ketamine and esketamine infusions. METHODS A randomized, double-blind, active-controlled, non-inferiority trial using ketamine and esketamine in TRD. Individuals diagnosed with MDD according to Diagnostic and Statistical Manual of Mental Disorders version IV and fulfilling TRD criteria were recruited from March 2017 to June 2018. Participants received a single subanesthetic dose of ketamine (0.5 mg/kg) or esketamine (0.25 mg/kg) for 40 min. Depressive symptoms were assessed using the Montgomery-Åsberg Depression Rating Scale (MADRS) and symptom remission was defined as a MADRS score ≤7 and response defined as ≥50% reduction in depressive symptom severity, 24 h and 7 days after the infusion. Clinical variables were selected based on previous clinical trials. Stepwise backward logistic regression was used, considering a confidence level of 95%. RESULTS 61 subjects were included: 39 (63.9%) were females with a mean age of 47.2 ± 14.9. Higher number of therapeutic failures (Odds Ratio (OR) = 0.677; 95% confidence interval (CI): 0.47-0.97) and higher severity of illness (OR = 0.912; 95% CI: 0.83-0.99) were associated with fewer remissions of depressive symptoms 7 days after intervention, and with fewer response in 24 h (OR = 0.583; 95% CI: 0,40; 0,84 and OR = 0.909; 95% CI: 0,83; 0,99, respectively). CONCLUSION Number of treatment failures and severity of illness were predictors of fewer remissions and responses of depressive symptoms in this TRD population. Study of predictors of remission may contribute to better selection patients that may benefit from receiving ketamine.
Collapse
Affiliation(s)
- Ana Paula Jesus-Nunes
- Programa de Pós-graduação Em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Gustavo C Leal
- Programa de Pós-graduação Em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Fernanda S Correia-Melo
- Programa de Pós-graduação Em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Flávia Vieira
- Programa de Pós-graduação Em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Rodrigo P Mello
- Programa de Pós-graduação Em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Ana Teresa Caliman-Fontes
- Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Mariana V F Echegaray
- Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Roberta F Marback
- Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Lívia N F Guerreiro-Costa
- Programa de Pós-graduação Em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Breno Souza-Marques
- Programa de Pós-graduação Em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Cassio Santos-Lima
- Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Lucca S Souza
- Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Igor D Bandeira
- Programa de Pós-graduação Em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Flavio Kapczinski
- INCT-TM, and Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Acioly L T Lacerda
- Laboratório Interdisciplinar de Neurociências Clínicas, Universidade Federal de São Paulo, São Paulo, Brazil.,Programa de Distúrbios Afetivos, Universidade Federal de São Paulo, São Paulo, Brazil.,BR Trials - Clinical Research, São Paulo, Brazil.,National Institute of Science and Technology in Translational Medicine, CNPq/FAPESP/CAPES, São Paulo, Brazil
| | - Lucas C Quarantini
- Programa de Pós-graduação Em Medicina e Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Neuropsicofarmacologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
21
|
Benitah K, Siegel AN, Lipsitz O, Rodrigues NB, Meshkat S, Lee Y, Mansur RB, Nasri F, Lui LMW, McIntyre RS, Rosenblat JD. Sex differences in ketamine's therapeutic effects for mood disorders: A systematic review. Psychiatry Res 2022; 312:114579. [PMID: 35504148 DOI: 10.1016/j.psychres.2022.114579] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/17/2022] [Accepted: 04/24/2022] [Indexed: 12/28/2022]
Abstract
Replicated clinical trials have demonstrated rapid and robust antidepressant effects with ketamine in treatment resistant mood disorders. Sex (biological) and gender differences in therapeutic effects for any new intervention is an important consideration, however, the differential efficacy, safety and tolerability of ketamine in males versus females remains underexplored. The objective of the present systematic review is to identify and qualitatively synthesize all published clinical studies relevant to the sex differential effects of ketamine for mood disorders. A systematic search of PubMed, Medline, and PsycInfo from inception until January 20, 2021, yielded 27 reports including 1715 patients (742 males and 973 females) that met inclusion criteria. Results from the vast majority of studies (88.8%) do not support significant sex differences in antidepressant response, tolerability or safety of ketamine. Nine (33.3%) of the reports included a bioanalytical component in the analysis and only one reported on sex differences. Evidence from the present review does not support clinically or statistically significant sex differences in therapeutic effects with ketamine. Nevertheless, future studies should continue to consider sex and biological sex differences in study design and data analytic plans.
Collapse
Affiliation(s)
- Katie Benitah
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Ashley N Siegel
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Orly Lipsitz
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada; Canadian Rapid Treatment Center of Excellence, Mississauga, ON, Canada
| | - Nelson B Rodrigues
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada; Canadian Rapid Treatment Center of Excellence, Mississauga, ON, Canada
| | - Shakila Meshkat
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Yena Lee
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Rodrigo B Mansur
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Flora Nasri
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Leanna M W Lui
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Canadian Rapid Treatment Center of Excellence, Mississauga, ON, Canada; Brain and Cognition Discovery Foundation, Canada; University of Toronto, Toronto, ON, Canada
| | - Joshua D Rosenblat
- Mood Disorder Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Canadian Rapid Treatment Center of Excellence, Mississauga, ON, Canada.
| |
Collapse
|
22
|
Does body mass index predict response to intravenous ketamine treatment in adults with major depressive and bipolar disorder? Results from the Canadian Rapid Treatment Center of Excellence. CNS Spectr 2022; 27:322-330. [PMID: 33267928 DOI: 10.1017/s1092852920002102] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Higher body mass index (BMI) has been found to predict greater antidepressant response to intravenous (IV) ketamine treatment. We evaluated the association between BMI and response to repeat-dose IV ketamine in patients with treatment-resistant depression (TRD). METHODS Adults (N = 230) with TRD received four infusions of IV ketamine at a community-based clinic. Changes in symptoms of depression (ie, Quick Inventory for Depressive Symptomatology-Self-Report 16; QIDS-SR16), suicidal ideation (SI; ie, QIDS-SR16 SI item), anxiety (ie, Generalized Anxiety Disorder-7 Scale), anhedonic severity (ie, Snaith-Hamilton Pleasure Scale), and functioning (ie, Sheehan Disability Scale) following infusions were evaluated. Participants were stratified by BMI as normal (18.0-24.9 kg/m2; n = 72), overweight (25-29.9 kg/m2; n = 76), obese I (30-34.9 kg/m2; n = 47), or obese II (≥35.0 kg/m2; n = 35). RESULTS Similar antidepressant effects with repeat-dose ketamine were reported between BMI groups (P = .261). In addition, categorical partial response (P = .149), response (P = .526), and remission (P = .232) rates were similar between the four BMI groups. CONCLUSIONS The findings are limited by the observational, open-label design of this retrospective analysis. Pretreatment BMI did not predict response to IV ketamine, which was effective regardless of BMI.
Collapse
|
23
|
Lengvenyte A, Strumila R, Olié E, Courtet P. Ketamine and esketamine for crisis management in patients with depression: Why, whom, and how? Eur Neuropsychopharmacol 2022; 57:88-104. [PMID: 35219097 DOI: 10.1016/j.euroneuro.2022.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022]
Abstract
Currently, only a limited number of interventions can rapidly relieve depressive symptomatology in patients with major depressive disorder or bipolar disorder experiencing extreme distress. Such crises, especially when suicide attempt or ideation is involved, are a major risk factor of suicide. Ketamine, a N-methyl-d-aspartate glutamate receptor antagonist, and its enantiomer esketamine rapidly reduce depressive symptoms in depressed patients with current suicidal ideation. Recently, esketamine has been approved for use in patients with depression at risk of suicide and for psychiatric emergency by major medical agencies in the United States and Europe, whereas ketamine is increasingly used off-label. However, there is currently limited guidance on why, when, and how to use these drugs in patients with depression to treat a crisis. In this review article, we provide a succinct overview of the cellular and molecular mechanisms of action of ketamine and esketamine, and of the functional brain changes following their administration. We also summarize the major clinical studies on ketamine and esketamine efficacy in patients experiencing a crisis (generally, suicidal ideation), and propose a profile of patients who can benefit most from such drugs, on the basis of neurobiological and clinical observations. Finally, we describe the administration mode, the efficacy and tolerability profiles, the side effect management, possible concomitant treatments and the issue of deprescribing.
Collapse
Affiliation(s)
- Aiste Lengvenyte
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, France; IGF, University of Montpellier, CNRS, INSERM, Montpellier, France; Psychiatric Clinic, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania.
| | - Robertas Strumila
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, France; IGF, University of Montpellier, CNRS, INSERM, Montpellier, France; Psychiatric Clinic, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Emilie Olié
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, France; IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Philippe Courtet
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, France; IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
24
|
Highland JN, Farmer CA, Zanos P, Lovett J, Zarate CA, Moaddel R, Gould TD. Sex-dependent metabolism of ketamine and ( 2R,6R)-hydroxynorketamine in mice and humans. J Psychopharmacol 2022; 36:170-182. [PMID: 34971525 PMCID: PMC9904319 DOI: 10.1177/02698811211064922] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Ketamine is rapidly metabolized to norketamine and hydroxynorketamine (HNK) metabolites. In female mice, when compared to males, higher levels of (2R,6R;2S,6S)-HNK have been observed following ketamine treatment, and higher levels of (2R,6R)-HNK following the direct administration of (2R,6R)-HNK. AIM The objective of this study was to evaluate the impact of sex in humans and mice, and gonadal hormones in mice on the metabolism of ketamine to form norketamine and HNKs and in the metabolism/elimination of (2R,6R)-HNK. METHODS In CD-1 mice, we utilized gonadectomy to evaluate the role of circulating gonadal hormones in mediating sex-dependent differences in ketamine and (2R,6R)-HNK metabolism. In humans (34 with treatment-resistant depression and 23 healthy controls) receiving an antidepressant dose of ketamine (0.5 mg/kg i.v. infusion over 40 min), we evaluated plasma levels of ketamine, norketamine, and HNKs. RESULTS In humans, plasma levels of ketamine and norketamine were higher in males than females, while (2R,6R;2S,6S)-HNK levels were not different. Following ketamine administration to mice (10 mg/kg i.p.), Cmax and total plasma concentrations of ketamine and norketamine were higher, and those of (2R,6R;2S,6S)-HNK were lower, in intact males compared to females. Direct (2R,6R)-HNK administration (10 mg/kg i.p.) resulted in higher levels of (2R,6R)-HNK in female mice. Ovariectomy did not alter ketamine metabolism in female mice, whereas orchidectomy recapitulated female pharmacokinetic differences in male mice, which was reversed with testosterone replacement. CONCLUSION Sex is an important biological variable that influences the metabolism of ketamine and the HNKs, which may contribute to sex differences in therapeutic antidepressant efficacy or side effects.
Collapse
Affiliation(s)
- Jaclyn N. Highland
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore MD, USA.,Program in Toxicology, University of Maryland School of Medicine, Baltimore MD, USA
| | - Cristan A. Farmer
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda MD, USA
| | - Panos Zanos
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore MD, USA.,Pharmacology, University of Maryland School of Medicine, Baltimore MD, USA.,Physiology, University of Maryland School of Medicine, Baltimore MD, USA
| | - Jacqueline Lovett
- Biomedical Research Center, National Institute on Aging, Intramural Research Program, National Institutes of Health, 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
| | - Ruin Moaddel
- Biomedical Research Center, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore MD, USA
| | - Todd D. Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore MD, USA.,Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda MD, USA.,Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore MD, USA.,Veterans Affairs Maryland Health Care System, Baltimore MD, USA.,Reprint requests: Todd D. Gould, Rm. 936 MSTF 685 W. Baltimore St., Baltimore, MD, 21201, USA.
| |
Collapse
|
25
|
Seney ML, Glausier J, Sibille E. Large-Scale Transcriptomics Studies Provide Insight Into Sex Differences in Depression. Biol Psychiatry 2022; 91:14-24. [PMID: 33648716 PMCID: PMC8263802 DOI: 10.1016/j.biopsych.2020.12.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is a leading cause of disability, affecting more than 300 million people worldwide. We first review the well-known sex difference in incidence of MDD, with women being twice as likely to be diagnosed as men, and briefly summarize how the impact of MDD varies between men and women, with sex differences in symptoms, severity, and antidepressant drug response. We then attempt to deconstruct the biological bases for MDD and discuss implications for sex differences research. Next, we review findings from human postmortem studies, both from selected candidate gene studies and from well-powered, unbiased transcriptomics studies, which suggest distinct, and possibly opposite, molecular changes in the brains of depressed men and women. We then discuss inherent challenges of research on the human postmortem brain and suggest paths forward that rely on thoughtful cohort design. Although studies indicate that circulating gonadal hormones might underlie the observed sex differences in MDD, we discuss how additional sex-specific factors, such as genetic sex and developmental exposure to gonadal hormones, may also contribute to altered vulnerability, and we highlight various nuances that we believe should be considered when determining mechanisms underlying observed sex differences. Altogether, this review highlights not only how various sex-specific factors might influence susceptibility or resilience to depression, but also how those sex-specific factors might result in divergent pathology in men and women.
Collapse
Affiliation(s)
- Marianne L Seney
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania.
| | - Jill Glausier
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute at the Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
26
|
da Costa JPDOM, Bisol LW, Souza FGDME. Which are the demographic and clinical characteristics of patients who respond to subcutaneous esketamine? J Psychopharmacol 2021; 35:1542-1544. [PMID: 34344207 DOI: 10.1177/02698811211035392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
| | - Luísa Weber Bisol
- Walter Cantídio Teaching Hospital, Federal University of Ceará, Fortaleza, Brazil
| | | |
Collapse
|
27
|
Yavi M, Henter ID, Park LT, Zarate C. Key considerations in the pharmacological management of treatment-resistant depression. Expert Opin Pharmacother 2021; 22:2405-2415. [PMID: 34252320 PMCID: PMC8648908 DOI: 10.1080/14656566.2021.1951225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
Introduction: Treatment-resistant depression (TRD) is a complex, multifactorial, and biologically heterogeneous disorder with debilitating outcomes. Understanding individual reasons why patients do not respond to treatment is necessary for improving clinical recommendations regarding medication regimens, augmentation strategies, and alternative treatments.Areas covered: This manuscript reviews evidence-based treatment strategies for the clinical management of TRD. Current developments in the field and potential future recommendations for personalized treatment of TRD are also discussed.Expert opinion: Treatment guidelines for TRD are limited by the heterogeneous nature of the disorder. Furthermore, current strategies reflect this heterogeneity by emphasizing disease characteristics as well as drug trial response or failure. Developing robust biomarkers that could one day be integrated into clinical practice has the potential to advance specific treatment targets and ultimately improve treatment and remission outcomes.
Collapse
Affiliation(s)
- Mani Yavi
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental HealthNational Institutes of Health Bethesda, MD, USA
| | | | | | | |
Collapse
|
28
|
Beaudequin D, Can AT, Jones M, Yang C, Scherman JK, Dutton M, Schwenn P, Forsyth CGG, Jensen E, Hermens DF, Lagopoulos J. Relationships between reduction in symptoms and restoration of function and wellbeing: Outcomes of the Oral Ketamine Trial on Suicidality (OKTOS). Psychiatry Res 2021; 305:114212. [PMID: 34563973 DOI: 10.1016/j.psychres.2021.114212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/17/2022]
Abstract
Recovery of functioning is integral to successful treatment outcomes in depressive illness. Optimal antidepressant treatment results in both symptomatic remission and functional recovery. Oral ketamine rapidly reduces suicidality and depression; however, reports of functional and wellbeing outcomes are lacking. This study examines participants' social and occupational functioning and wellbeing outcomes in the Oral Ketamine Trial on Suicidality (OKTOS). Thirty adults with chronic suicidality participated in the trial over 10 weeks. Functional recovery and wellbeing were assessed using the Social and Occupational Functioning Scale (SOFAS) and World Health Organization Well-Being Index (WHO-5). Suicidality and depressive symptoms were assessed using the Beck Scale for Suicidal ideation (BSS) and Montgomery-Asberg Depression Rating Scale (MADRS). Relationships between the four treatment outcomes were analysed. Forty-three percent of participants achieved healthy function (SOFAS ≥ 80) and 27% reported healthy wellbeing (WHO-5 > 60%) at the four-week post-treatment follow-up. Wellbeing was revealed as the data-derived treatment endpoint for the sample. Effect sizes for functioning and wellbeing outcomes were smaller than for suicidality and depression outcomes. Results suggest that reduction in depressive symptoms and suicidal ideation may be necessary but not sufficient for full restoration of function and wellbeing in antisuicidal and antidepressant therapy, including clinical trials.
Collapse
Affiliation(s)
- Denise Beaudequin
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia.
| | - Adem T Can
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | - Monique Jones
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | - Cian Yang
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | | | - Megan Dutton
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | - Paul Schwenn
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | | | - Emma Jensen
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | - Daniel F Hermens
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| | - Jim Lagopoulos
- Thompson Institute, University of the Sunshine Coast, Queensland, Australia
| |
Collapse
|
29
|
Fitzgerald PJ, Kounelis-Wuillaume SK, Gheidi A, Morrow JD, Spencer-Segal JL, Watson BO. Sex- and stress-dependent effects of a single injection of ketamine on open field and forced swim behavior. Stress 2021; 24:857-865. [PMID: 33517825 PMCID: PMC8325703 DOI: 10.1080/10253890.2021.1871600] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ketamine has emerged as a novel treatment for common psychiatric conditions such as Major Depressive Disorder (MDD) and anxiety disorders, many of which can be initiated and exacerbated by psychological stress. Sex differences in the frequency of both anxiety and depressive disorders are well known and could be due to sex differences in neuroendocrine responses to stress. Ketamine is known to modulate the hormonal response to stress, specifically corticosterone. It is not clear if the acute effect of ketamine on corticosterone differs by sex, or what role this could play in subsequent behavior. Here we test whether a single injection of (R,S)-ketamine (30 mg/kg, i.p.), administered either with or without unpredictable chronic stress (UCS), has different sustained effects on open field test (OFT), elevated zero maze (EZM) or forced swim test (FST) behavior in female versus male C57BL/6J mice. In the OFT (24 h post-injection), ketamine increased center square exploration in males but not females. In contrast, in the FST (72 h post-injection), females showed a trend toward a decrease in immobility after ketamine whereas males were not strongly modulated. These behavioral effects of ketamine were stronger in the presence of UCS than in unstressed animals. UCS animals also showed lower corticosterone after injection than unstressed animals, and in the presence of UCS ketamine increased corticosterone; these effects were similar in both sexes. Corticosterone post-injection did not predict subsequent behavior. These findings complement a growing preclinical literature suggesting both stress-dependency and sex differences in OFT and FST behavioral responses to ketamine.LAY SUMMARYIn humans, it is known that major depression and anxiety disorders, which can be caused or made worse by exposure to psychological stress, occur roughly twice as frequently in women than in men, but the underpinnings of these effects are not well characterized. In the current study, we explored how sex interacts with stress and ketamine (a rapidly acting antidepressant) by assessing both open field and forced swim behavior in mice after chronic mild stress. We report the novel finding that male mice exhibit greater exploration of the aversive center square in the open field after ketamine, whereas females trended toward lower immobility (often interpreted as an antidepressant-like effect) in the forced swim test after this drug, and these effects were amplified by prior stress exposure.
Collapse
Affiliation(s)
- Paul J. Fitzgerald
- Department of Psychiatry, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109
| | | | - Ali Gheidi
- Department of Psychiatry, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Jonathan D. Morrow
- Department of Psychiatry, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Joanna L. Spencer-Segal
- Michigan Neuroscience Institute, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109
- Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109
- For correspondence: ,
| | - Brendon O. Watson
- Department of Psychiatry, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109
- For correspondence: ,
| |
Collapse
|
30
|
Age affects temporal response, but not durability, to serial ketamine infusions for treatment refractory depression. Psychopharmacology (Berl) 2021; 238:3229-3237. [PMID: 34363507 DOI: 10.1007/s00213-021-05939-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 07/14/2021] [Indexed: 12/30/2022]
Abstract
RATIONALE Ketamine is a novel, rapid-acting antidepressant for treatment refractory depression (TRD); however, clinical durability is poor and treatment response trajectories vary. Little is known about which patient characteristics predict faster or more durable ketamine responses. Ketamine's antidepressant mechanism may involve modulation of glutamatergic signaling and long-term potentiation (LTP); these neuroplasticity pathways are also attenuated with older age. OBJECTIVE A retrospective analysis examining the impact of patient age on the speed and durability of ketamine's antidepressant effects in 49 veterans receiving serial intravenous ketamine infusions for TRD. METHOD The relationship between age and percent change in Beck Depression Inventory (BDI-II) scores was compared across six serial ketamine infusions (twice-weekly for 3 weeks) using a linear-mixed model. RESULTS A significant Age-X-Infusion number interaction (F = 3.01, p = .0274) indicated that the relationship between age and treatment response depended on infusion number. Follow-up tests showed that younger age significantly predicted greater clinical improvement at infusion #4 (t = 3.02, p = .004); this relationship was attenuated at infusion #5 (t = 1.95, p = .057) and was absent at infusion #6. Age was not a significant predictor of treatment durability, defined as percent change in BDI-II 3 weeks following infusion #6. CONCLUSIONS These data preliminarily suggest that younger age is associated with a faster response over six serial ketamine infusions; by infusion #6 and subsequent weeks of clinical follow-up, age no longer predicts ketamine's antidepressant activity. Age may mediate the speed but not the durability or total efficacy of ketamine treatment, suggesting that dissociable mechanisms may underlie differing aspects of ketamine's antidepressant activity.
Collapse
|
31
|
Khadrawy YA, Hosny EN, Magdy M, Mohammed HS. Antidepressant effects of curcumin-coated iron oxide nanoparticles in a rat model of depression. Eur J Pharmacol 2021; 908:174384. [PMID: 34324858 DOI: 10.1016/j.ejphar.2021.174384] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/15/2021] [Accepted: 07/23/2021] [Indexed: 12/21/2022]
Abstract
The antidepressant effect of curcumin-coated iron oxide nanoparticles (Cur-IONPs) was investigated in the current study using depression rat model induced by reserpine. IONPs were synthesized by curcumin as a reducing agent producing Cur-IONPs. Rats were divided into control, depression rat model, and depressed rats treated with Cur-IONPs. After treatment rat behavior was evaluated using forced swimming test (FST). Serotonin (5-HT), norepinephrine (NE), dopamine (DA), monoamine oxidase (MAO), acetylcholinesterase (AchE), Na+, K+, ATPase, lipid peroxidation (MDA), reduced glutathione (GSH), glutathione-s-transferase (GST) and nitric oxide (NO) were measured in the cortex and hippocampus. In depressed rats, FST showed increased immobilization time and reduced swimming time. This was associated with a significant decrease in 5-HT, NE, DA and GSH and a significant increase in MDA and NO levels and GST, MAO, AchE and Na+, K+, ATPase activities in the cortex and hippocampus. Treatment with Cur-NONPs for two weeks increased the swimming time reduced the immobility time, and elevated 5-HT, NE and DA levels. Cur-IONPs attenuated the oxidative stress induced by reserpine and restored the MAO, AchE and Na+, K+, ATPase. The present green method used curcumin in the IONPs synthesis and has several merits; obtaining nanoform of iron oxide, increasing the bioavailability of curcumin and reducing the oxidative stress induced by iron. The present antidepressant effect of Cur-IONPs could be attributed to the ability of Cur-IONPs to restore monoamine neurotransmitter levels by increasing their synthesis and reducing their metabolism. In addition, the antioxidant activity of curcumin prevented oxidative stress in the depressed rats.
Collapse
Affiliation(s)
- Yasser A Khadrawy
- Medical Physiology Department, Medical Division, National Research Centre, Giza, Egypt.
| | - Eman N Hosny
- Medical Physiology Department, Medical Division, National Research Centre, Giza, Egypt
| | - Merna Magdy
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Haitham S Mohammed
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
32
|
Structural connectivity and subcellular changes after antidepressant doses of ketamine and Ro 25-6981 in the rat: an MRI and immuno-labeling study. Brain Struct Funct 2021; 226:2603-2616. [PMID: 34363521 PMCID: PMC8448713 DOI: 10.1007/s00429-021-02354-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 07/26/2021] [Indexed: 12/28/2022]
Abstract
Ketamine has rapid and robust antidepressant effects. However, unwanted psychotomimetic effects limit its widespread use. Hence, several studies examined whether GluN2B-subunit selective NMDA antagonists would exhibit a better therapeutic profile. Although preclinical work has revealed some of the mechanisms of action of ketamine at cellular and molecular levels, the impact on brain circuitry is poorly understood. Several neuroimaging studies have examined the functional changes in the brain induced by acute administration of ketamine and Ro 25-6981 (a GluN2B-subunit selective antagonist), but the changes in the microstructure of gray and white matter have received less attention. Here, the effects of ketamine and Ro 25-6981 on gray and white matter integrity in male Sprague-Dawley rats were determined using diffusion-weighted magnetic resonance imaging (DWI). In addition, DWI-based structural brain networks were estimated and connectivity metrics were computed at the regional level. Immunohistochemical analyses were also performed to determine whether changes in myelin basic protein (MBP) and neurofilament heavy-chain protein (NF200) may underlie connectivity changes. In general, ketamine and Ro 25-6981 showed some opposite structural alterations, but both compounds coincided only in increasing the fractional anisotropy in infralimbic prefrontal cortex and dorsal raphe nucleus. These changes were associated with increments of NF200 in deep layers of the infralimbic cortex (together with increased MBP) and the dorsal raphe nucleus. Our results suggest that the synthesis of NF200 and MBP may contribute to the formation of new dendritic spines and myelination, respectively. We also suggest that the increase of fractional anisotropy of the infralimbic and dorsal raphe nucleus areas could represent a biomarker of a rapid antidepressant response.
Collapse
|
33
|
Sumner RL, Chacko E, McMillan R, Spriggs MJ, Anderson C, Chen J, French A, Jung S, Rajan A, Malpas G, Hay J, Ponton R, Muthukumaraswamy SD, Sundram F. A qualitative and quantitative account of patient's experiences of ketamine and its antidepressant properties. J Psychopharmacol 2021; 35:946-961. [PMID: 33781107 DOI: 10.1177/0269881121998321] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Ketamine is central to one of the most rapidly growing areas of neuroscientific research into novel treatments for depression. Limited research has indicated that the psychedelic properties of ketamine may play a role in its antidepressant effects. AIM The aim of the current study was to explore the psychedelic experiences and sustained impact of ketamine in major depressive disorder. METHODS In the current study, ketamine (0.44 mg/kg) was administered to 32 volunteers with major depressive disorder in a crossover design with the active-placebo remifentanil, in a magnetic resonance imaging (MRI) environment. The 11-dimension altered states of consciousness questionnaire and individual qualitative interviews were used to capture the acute psychedelic experience. The Montgomery-Asberg Depression Rating Scale and further interviewing explored lasting effects. The second qualitative interview took place ⩾3 weeks post-ketamine. RESULTS Greater antidepressant response (reduction in Montgomery-Asberg Depression Rating Scale at 24 h) correlated with the 11-dimension altered states of consciousness dimensions: spirituality, experience of unity, and insight. The first qualitative interview revealed that all participants experienced perceptual changes. Additional themes emerged including loss of control and emotional and mood changes. The final interview showed evidence of a psychedelic afterglow, and changes to perspective on life, people, and problems, as well as changes to how participants felt about their depression and treatments. CONCLUSIONS The current study provides preliminary evidence for a role of the psychedelic experience and afterglow in ketamine's antidepressant properties. Reflexive thematic analysis provided a wealth of information on participants' experience of the study and demonstrated the psychedelic properties of ketamine are not fully captured by commonly used questionnaires.
Collapse
Affiliation(s)
- Rachael L Sumner
- School of Pharmacy, University of Auckland, Auckland, New Zealand
| | - Emme Chacko
- Department of Psychological Medicine, University of Auckland, Auckland, New Zealand
| | - Rebecca McMillan
- School of Pharmacy, University of Auckland, Auckland, New Zealand
| | - Meg J Spriggs
- Centre for Psychedelic Research, Imperial College London, London, UK
| | | | - James Chen
- School of Pharmacy, University of Auckland, Auckland, New Zealand
| | - Amelia French
- School of Pharmacy, University of Auckland, Auckland, New Zealand
| | - SungHun Jung
- School of Pharmacy, University of Auckland, Auckland, New Zealand
| | - Akshaya Rajan
- School of Pharmacy, University of Auckland, Auckland, New Zealand
| | - Gemma Malpas
- Department of Anaesthesia and Perioperative Medicine, Auckland District Health Board, Auckland, New Zealand
| | - John Hay
- Department of Anaesthesia and Perioperative Medicine, Auckland District Health Board, Auckland, New Zealand
| | - Rhys Ponton
- School of Pharmacy, University of Auckland, Auckland, New Zealand
| | | | - Frederick Sundram
- Department of Psychological Medicine, University of Auckland, Auckland, New Zealand
| |
Collapse
|
34
|
Abstract
BACKGROUND A rapid antidepressant effect of ketamine has repeatedly been documented in the literature, and identifying clinical features associated with a better response to this treatment is currently an essential question. Considering the relationship between rumination and depression and the need to identify potential predictors of response to ketamine, we analyzed the effect of a single injection of ketamine 0.5 mg/kg on rumination in treatment-resistant depressive (TRD) patients and explored whether baseline ruminative style and early improvements of rumination would predict a greater antidepressant effect of ketamine. METHODS Ten TRD outpatients who participated in a 4-week open study on the antidepressant effect of ketamine also completed the Ruminative Response Scale the day before, the day after, and a week after ketamine administration. RESULTS We found that in our patients, a single rapid 1-minute intravenous injection of ketamine 0.5 mg/kg was efficacious in reducing rumination, but neither severity of rumination at baseline nor early improvements of rumination after ketamine injection predicted antidepressant response. CONCLUSIONS Our preliminary data suggest that a single injection of ketamine 0.5 mg/kg can be efficacious in reducing rumination in TRD patients but rumination does not seem to be a useful clinical predictor of response to ketamine. Larger studies are necessary to confirm these results.
Collapse
|
35
|
Sanders B, Brula AQ. Intranasal esketamine: From origins to future implications in treatment-resistant depression. J Psychiatr Res 2021; 137:29-35. [PMID: 33647726 DOI: 10.1016/j.jpsychires.2021.02.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022]
Abstract
The approval of intranasal esketamine for treatment-resistant depression marks the next step in our understanding of and ability to treat treatment-resistant depression. The origin of ketamine is rooted in the search for a phencyclidine analog that could be used as a pre-surgical anesthetic with less emergence delirium. Following its inception, ketamine has been used in a variety of contexts. However, it was the seminal Berman et al., 2000 study, which published positive findings from the first human trial using subanesthetic intravenous ketamine for depression. Since then, a large body of research has investigated ketamine's various proposed antidepressant mechanisms of action, and the role its pharmacokinetic properties and route of administration play in producing its antidepressant effects. The results of this research were the eventual approval of intranasal esketamine for treatment-resistant depression by the U.S. Food and Drug Administration (FDA) in March 2019. By identifying and utilizing predictors of response, we can continue to refine our approach to treating treatment-resistant depression and optimize patient response to intranasal esketamine. In this article, we look at the history, pharmacology, landmark studies, and future implications of intranasal esketamine for treatment-resistant depression.
Collapse
Affiliation(s)
- Benjamin Sanders
- University of Kentucky- Bowling Green, 250 Park St. Attn: GME, Bowling Green, KY, 42101, USA.
| | - Abdul Q Brula
- University of Kentucky- Bowling Green, 250 Park St. Attn: GME, Bowling Green, KY, 42101, USA
| |
Collapse
|
36
|
Abstract
Over the last two decades, the dissociative anaesthetic agent ketamine, an uncompetitive N-Methyl-D-Aspartate (NMDA) receptor antagonist, has emerged as a novel therapy for treatment-resistant depression (TRD), demonstrating rapid and robust antidepressant effects within hours of administration. Ketamine is a racemic mixture composed of equal amounts of (S)-ketamine and (R)-ketamine. Although ketamine currently remains an off-label treatment for TRD, an (S)-ketamine nasal spray has been approved for use in TRD (in conjunction with an oral antidepressant) in the United States and Europe. Despite the promise of ketamine, key challenges including how to maintain response, concerns regarding short and long-term side-effects and the potential for abuse remain. This review provides an overview of the history of ketamine, its use in psychiatry and its basic pharmacology. The clinical evidence for the use of ketamine in depression and potential adverse effects associated with treatment are summarized. A synopsis of some of the putative neurobiological mechanisms underlying ketamine's rapid-acting antidepressant effects is provided before finally outlining future research directions, including the need to identify biomarkers for predicting response and treatment targets that may be used in the development of next-generation rapid-acting antidepressants that may lack ketamine's side-effects or abuse potential.
Collapse
Affiliation(s)
- Luke A Jelen
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - James M Stone
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,South London and Maudsley NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
37
|
McIntyre RS, Rosenblat JD, Nemeroff CB, Sanacora G, Murrough JW, Berk M, Brietzke E, Dodd S, Gorwood P, Ho R, Iosifescu DV, Jaramillo CL, Kasper S, Kratiuk K, Lee JG, Lee Y, Lui LM, Mansur RB, Papakostas GI, Subramaniapillai M, Thase M, Vieta E, Young AH, Zarate CA, Stahl S. Synthesizing the Evidence for Ketamine and Esketamine in Treatment-Resistant Depression: An International Expert Opinion on the Available Evidence and Implementation. Am J Psychiatry 2021; 178:383-399. [PMID: 33726522 PMCID: PMC9635017 DOI: 10.1176/appi.ajp.2020.20081251] [Citation(s) in RCA: 315] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Replicated international studies have underscored the human and societal costs associated with major depressive disorder. Despite the proven efficacy of monoamine-based antidepressants in major depression, the majority of treated individuals fail to achieve full syndromal and functional recovery with the index and subsequent pharmacological treatments. Ketamine and esketamine represent pharmacologically novel treatment avenues for adults with treatment-resistant depression. In addition to providing hope to affected persons, these agents represent the first non-monoaminergic agents with proven rapid-onset efficacy in major depressive disorder. Nevertheless, concerns remain about the safety and tolerability of ketamine and esketamine in mood disorders. Moreover, there is uncertainty about the appropriate position of these agents in treatment algorithms, their comparative effectiveness, and the appropriate setting, infrastructure, and personnel required for their competent and safe implementation. In this article, an international group of mood disorder experts provides a synthesis of the literature with respect to the efficacy, safety, and tolerability of ketamine and esketamine in adults with treatment-resistant depression. The authors also provide guidance for the implementation of these agents in clinical practice, with particular attention to practice parameters at point of care. Areas of consensus and future research vistas are discussed.
Collapse
Affiliation(s)
- Roger S. McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto; Department of Psychiatry, University of Toronto, Toronto; Department of Pharmacology, University of Toronto, Toronto; Brain and Cognition Discovery Foundation, Toronto
| | - Joshua D. Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto; Department of Psychiatry, University of Toronto, Toronto; Canadian Rapid Treatment Center of Excellence, Mississauga, Ontario
| | - Charles B. Nemeroff
- Department of Psychiatry and Behavioral Sciences, Austin Dell Medical School, University of Texas, Austin
| | - Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, New Haven, Conn
| | - James W. Murrough
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, and Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York
| | - Michael Berk
- Deakin University, Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, University of Melbourne, Melbourne, Australia
| | - Elisa Brietzke
- Department of Psychiatry, Queen’s University School of Medicine, and Centre for Neuroscience Studies, Queen’s University, Kingston, Ontario
| | - Seetal Dodd
- Deakin University, Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Centre for Youth Mental Health and Department of Psychiatry, University of Melbourne, Melbourne, Australia
| | - Philip Gorwood
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, and GHU Paris Psychiatrie et Neurosciences, CMME, Hôpital Sainte-Anne, Paris
| | - Roger Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, and Institute of Health Innovation and Technology, National University of Singapore, Singapore
| | - Dan V. Iosifescu
- Department of Psychiatry, NYU School of Medicine, and Clinical Research Division, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | | | | | - Kevin Kratiuk
- Canadian Rapid Treatment Center of Excellence, Mississauga, Ontario; Department of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jung Goo Lee
- Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Paik Institute for Clinical Research, and Department of Health Science and Technology, Graduate School, Inje University, Busan, Republic of Korea
| | - Yena Lee
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto; Institute of Medical Science, University of Toronto, Toronto
| | - Leanna M.W. Lui
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto
| | - Rodrigo B. Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto; Department of Psychiatry, University of Toronto, Toronto
| | | | | | - Michael Thase
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, and Corporal Michael J. Crescenz VA Medical Center, Philadelphia
| | - Eduard Vieta
- Hospital Clinic, Institute of Neuroscience, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona
| | - Allan H. Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London and South London, and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Beckenham, Kent
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch and Section on the Neurobiology and Treatment of Mood Disorders, Division of Intramural Research Program, NIMH, Bethesda, Md
| | - Stephen Stahl
- Department of Psychiatry and Neuroscience, University of California, Riverside, and University of California, San Diego
| |
Collapse
|
38
|
"Novel Psychopharmacology for Depressive Disorders". ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1305:449-461. [PMID: 33834412 DOI: 10.1007/978-981-33-6044-0_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Major depressive disorder carries a significant burden and a high risk for suicide. The need for more effective, safer, and faster-acting drugs is, therefore, compelling. The present chapter briefly assesses the most promising agents, focusing on non-monoamine-targeting compounds, namely, the glutamate antagonist ketamine and its enantiomer esketamine. A critical overview of the evidence and the pitfalls associated with current antidepressant drug development is likewise provided in the following text.
Collapse
|
39
|
Meyer T, Brunovsky M, Horacek J, Novak T, Andrashko V, Seifritz E, Olbrich S. Predictive value of heart rate in treatment of major depression with ketamine in two controlled trials. Clin Neurophysiol 2021; 132:1339-1346. [PMID: 33888426 DOI: 10.1016/j.clinph.2021.01.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 12/30/2020] [Accepted: 01/09/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Ketamine has been shown to be effective in treatment of episodes of major depressive disorder (MDD). This controlled study aimed to analyse the predictive and discriminative power of heart rate (HR) and heart rate variability (HRV) for ketamine treatment in MDD. METHODS In 51 patients, HR and HRV were assessed at baseline before and during ketamine infusion and 24 hours post ketamine infusion. Montgomery-Åsberg Depression Rating Scale (MADRS) was used to assess changes of depressive symptoms. A 30% or 50% reduction of symptoms after 24 hours or within 7 days was defined as response. A linear mixed model was used for analysis. RESULTS Ketamine infusion increased HR and HRV power during and after infusion. Responders to ketamine showed a higher HR during the whole course of investigation, including at baseline with medium effect sizes (Cohen's d = 0.47-0.67). Furthermore, HR and HRV power discriminated between responders and non-responders, while normalized low and high frequencies did not. CONCLUSION The findings show a predictive value of HR and HRV power for ketamine treatment. This further underlines the importance of the autonomous nervous system (ANS) and its possible malfunctions in MDD. SIGNIFICANCE The predictive power of HR and HRV markers should be studied in prospective studies. Neurophysiological markers could improve treatment for MDD via optimizing the choice of treatments.
Collapse
Affiliation(s)
- Torsten Meyer
- Department for Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zurich, Switzerland
| | - Martin Brunovsky
- National Institute of Mental Health, Klecany, Czech Republic; Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | - Jiri Horacek
- National Institute of Mental Health, Klecany, Czech Republic; Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | - Tomas Novak
- National Institute of Mental Health, Klecany, Czech Republic; Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | - Veronika Andrashko
- National Institute of Mental Health, Klecany, Czech Republic; Charles University, Third Faculty of Medicine, Prague, Czech Republic
| | - Erich Seifritz
- Department for Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zurich, Switzerland
| | - Sebastian Olbrich
- Department for Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zurich, Switzerland.
| |
Collapse
|
40
|
Nakamura T, Tomita M, Horikawa N, Ishibashi M, Uematsu K, Hiraki T, Abe T, Uchimura N. Functional connectivity between the amygdala and subgenual cingulate gyrus predicts the antidepressant effects of ketamine in patients with treatment-resistant depression. Neuropsychopharmacol Rep 2021; 41:168-178. [PMID: 33615749 PMCID: PMC8340826 DOI: 10.1002/npr2.12165] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 12/28/2022] Open
Abstract
Aim Approximately one‐third of patients with major depressive disorder develop treatment‐resistant depression. One‐third of patients with treatment‐resistant depression demonstrate resistance to ketamine, which is a novel antidepressant effective for this disorder. The objective of this study was to examine the utility of resting‐state functional magnetic resonance imaging for the prediction of treatment response to ketamine in treatment‐resistant depression. Methods An exploratory seed‐based resting‐state functional magnetic resonance imaging analysis was performed to examine baseline resting‐state functional connectivity differences between ketamine responders and nonresponders before treatment with multiple intravenous ketamine infusions. Results Fifteen patients with treatment‐resistant depression received multiple intravenous subanesthetic (0.5 mg/kg/40 minutes) ketamine infusions, and nine were identified as responders. The exploratory resting‐state functional magnetic resonance imaging analysis identified a cluster of significant baseline resting‐state functional connectivity differences associating ketamine response between the amygdala and subgenual anterior cingulate gyrus in the right hemisphere. Using anatomical region of interest analysis of the resting‐state functional connectivity, ketamine response was predicted with 88.9% sensitivity and 100% specificity. The resting‐state functional connectivity of significant group differences between responders and nonresponders retained throughout the treatment were considered a trait‐like feature of heterogeneity in treatment‐resistant depression. Conclusion This study suggests the possible clinical utility of resting‐state functional magnetic resonance imaging for predicting the antidepressant effects of ketamine in treatment‐resistant depression patients and implicated resting‐state functional connectivity alterations to determine the trait‐like pathophysiology underlying treatment response heterogeneity in treatment‐resistant depression. This study illustrates that the alteration in the RSFC within the right AN in TRD patients reflects the antidepressant response to ketamine at baseline. The alteration remained throughout the 2‐week treatment with multiple ketamine infusions and seemed to reflect the trait‐like features underlying treatment heterogeneity in TRD. By employing an anatomical ROI of the sc/sgACC, the present study also suggests the possible clinical utility of the rsfMRI to predict the treatment response to ketamine in TRD patients.
![]()
Collapse
Affiliation(s)
- Tomoyuki Nakamura
- Department of Neuropsychiatry, Kurume University School of Medicine, Kurume City, Japan
| | - Masaru Tomita
- Department of Neuropsychiatry, Kurume University School of Medicine, Kurume City, Japan.,Elm-tree Mental Clinic, Ogori City, Japan
| | - Naoki Horikawa
- Department of Neuropsychiatry, Kurume University School of Medicine, Kurume City, Japan.,Nozoe Hills Hospital, Kurume City, Japan
| | - Masatoshi Ishibashi
- Department of Neuropsychiatry, Kurume University School of Medicine, Kurume City, Japan
| | - Ken Uematsu
- Uematsu Mental Clinic, Chikugo City, Japan.,Department of Pharmacology, Kurume University School of Medicine, Kurume City, Japan
| | - Teruyuki Hiraki
- Department of Anaesthesiology, Kurume University School of Medicine, Kurume City, Japan
| | - Toshi Abe
- Department of Radiology, Kurume University School of Medicine, Kurume City, Japan
| | - Naohisa Uchimura
- Department of Neuropsychiatry, Kurume University School of Medicine, Kurume City, Japan
| |
Collapse
|
41
|
Chen MH, Wu HJ, Li CT, Lin WC, Bai YM, Tsai SJ, Hong CJ, Tu PC, Cheng CM, Su TP. Using classification and regression tree modelling to investigate treatment response to a single low-dose ketamine infusion: Post hoc pooled analyses of randomized placebo-controlled and open-label trials. J Affect Disord 2021; 281:865-871. [PMID: 33239245 DOI: 10.1016/j.jad.2020.11.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 09/08/2020] [Accepted: 11/07/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Evidence suggests that clinical markers, such as comorbid anxiety, body weight, and others can assist in predicting response to low-dose ketamine infusion in treatment resistant depression patients. However, whether a composite of clinical markers may improve the predicted probability of response is uncertain. METHODS The current study investigated the results of our previous randomized placebo-controlled and open-label trials in which 73 patients with treatment-resistant depression (TRD) received a single ketamine infusion of 0.5 mg/kg. Clinical characteristics at baseline, including depression severity, duration of the current episode, obesity, comorbidity of anxiety disorder, and current suicide risk, were assessed as potential predictors in a classification and regression tree model for treatment response to ketamine infusion. RESULTS The predicted probability of a composite of age at disease onset, depression severity, duration of current episode, and obesity/overweight was significantly greater (area under curve = .736, p = .001) than that of any one marker (all p > .05). The most powerful predictors of treatment response to ketamine infusion were younger age at disease onset and obesity/overweight. The strongest predictors of treatment nonresponse were longer duration of the current episode and greater depression severity at baseline. DISCUSSION Depression severity, duration of the current episode, obesity, and age at disease onset may predict treatment response versus nonresponse to low-dose ketamine infusion. However, whether our predicted probability for a single infusion may be applied to repeated infusions would require further investigation. CLINICAL TRIAL REGISTRATION UMIN Clinical Trials Registry (UMIN000023581 and UMIN000016985).
Collapse
Affiliation(s)
- Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Hui-Ju Wu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Cheng-Ta Li
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan.
| | - Wei-Chen Lin
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Ya-Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Chen-Jee Hong
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Chi Tu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Ming Cheng
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Tung-Ping Su
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan.
| |
Collapse
|
42
|
Lucchese AC, Sarin LM, Magalhães EJM, Del Sant LC, B Puertas C, Tuena MA, Nakahira C, Fava VA, Delfino R, Surjan J, Steiglich MS, Barbosa M, Abdo G, Cohrs FM, Liberatori A, Del Porto JA, Lacerda AL, B Andreoli S. Repeated subcutaneous esketamine for treatment-resistant depression: Impact of the degree of treatment resistance and anxiety comorbidity. J Psychopharmacol 2021; 35:142-149. [PMID: 33427015 DOI: 10.1177/0269881120978398] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND A large number of studies indicate that subanesthetic doses of ketamine induce a fast antidepressant effect. Limited studies have investigated the subcutaneous (SC) route, and it remains unclear for whom this treatment is most suitable. AIMS The aim of this study was to examine the effect on depressive symptoms of repeated subanesthetic doses of SC esketamine in unipolar and bipolar treatment-resistant depression (TRD) and clinical predictors of response. METHODS A retrospective analysis of 70 patients who received six SC esketamine doses weekly as an adjunctive treatment was carried out. Doses started at 0.5 mg/kg and it could be titrated up to 1 mg/kg, according to response. The primary outcome was reduction in depressive symptoms. Statistical analysis to investigate clinical predictors of effectiveness included logistic regression analysis using a dependent variable of a 50% reduction in rating scale scores at the end of treatment. Comparisons between groups were made through analysis of variance and treatment effects. RESULTS At baseline, our sample presented with severe treatment resistance in 65.7%, as assessed by the Maudsley Staging Method (MSM), and 47.1% had anxiety disorder comorbidity. The response rate was 50%. A better outcome was predicted by mild and moderate MSM scores (OR = 3.162, p = 0.041) and anxiety disorder comorbidity (OR = 3.149, p = 0.028). CONCLUSIONS Our results suggest that higher levels of treatment resistance may be associated with a poor response to SC esketamine. Unlike traditional pharmacotherapies, it might benefit those with poor prognosis such as patients with depression and comorbid anxiety. Therefore, future research could investigate whether esketamine should receive a more prominent place in the treatment algorithm for TRD.
Collapse
Affiliation(s)
- Ana C Lucchese
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Luciana M Sarin
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
| | | | - Lorena C Del Sant
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Camila B Puertas
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Marco A Tuena
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Carolina Nakahira
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Victor Ar Fava
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Rodrigo Delfino
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
- Department of Psychiatry, PRODAF - Programa de Transtornos Afetivos, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Juliana Surjan
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
- Department of Psychiatry, PRODAF - Programa de Transtornos Afetivos, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Matheus S Steiglich
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
- Department of Psychiatry, PRODAF - Programa de Transtornos Afetivos, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Matheus Barbosa
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Guilherme Abdo
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Frederico M Cohrs
- Escola Paulista de Medicina, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Aroldo Liberatori
- Escola Paulista de Medicina, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - José A Del Porto
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
- Department of Psychiatry, PRODAF - Programa de Transtornos Afetivos, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Acioly Lt Lacerda
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
- Department of Psychiatry, PRODAF - Programa de Transtornos Afetivos, Federal University of Sao Paulo, Sao Paulo, Brazil
- Department of Psychiatry, LiNC - Laboratory of Integrative Neuroscience, Federal University of Sao Paulo, Sao Paulo, Brazil
- Center for Interventional Psychiatry, Hospital Sao Marcos, Jaboticabal, Brazil
| | - Sergio B Andreoli
- Department of Psychiatry, Federal University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
43
|
Freeman MP, Hock RS, Papakostas GI, Judge H, Cusin C, Mathew SJ, Sanacora G, Iosifescu DV, DeBattista C, Trivedi MH, Fava M. Body Mass Index as a Moderator of Treatment Response to Ketamine for Major Depressive Disorder. J Clin Psychopharmacol 2021; 40:287-292. [PMID: 32332464 PMCID: PMC7185034 DOI: 10.1097/jcp.0000000000001209] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE/BACKGROUND Major depressive disorder (MDD) and obesity commonly co-occur. We sought to assess the impact of body mass index (BMI) on the acute antidepressant effects of ketamine in patients with treatment-resistant depression. METHODS/PROCEDURES Post hoc analyses were conducted from a multisite, randomized, double-blind, placebo-controlled trial designed to assess the rapid-onset effects of intravenous ketamine. Patients (n = 99) were randomized to a single dose administration of ketamine 0.1 mg/kg (n = 18), ketamine 0.2 mg/kg (n = 20), ketamine 0.5 mg/kg (n = 22), ketamine 1.0 mg/kg (n = 20), or active placebo, midazolam 0.045 mg/kg (n = 19). Patients were stratified for BMI. For patients randomized to ketamine (n = 80), BMI was assessed as a continuous variable and also categorically (obese, overweight, not obese/overweight [reference]). The primary outcome measure was the change on the 6-item Hamilton Depression Rating Scale 24 hours after treatment. Outcomes at day 3 were also assessed. FINDINGS/RESULTS The 6-item Hamilton Depression Rating Scale change scores at 24 hours were inversely associated with BMI (-0.28 ± 0.12, P = 0.02). With BMI operationalized categorically, both obese (-4.15 ± 1.41, P = 0.004) and overweight (-1.99 ± 1.14, P = 0.08) categories were inversely related to the 6-item Hamilton Depression Rating Scale change score at 24 hours, statistically significant for the obese category, as compared with the reference group. Similar but weaker findings were observed at 72 hours after infusion. IMPLICATIONS/CONCLUSIONS Higher BMI and obesity were associated with a more robust acute antidepressant response to ketamine. This may have clinical relevance for a great number of patients who have both MDD and obesity. CLINICAL TRIAL REGISTRATION NCT01920555.
Collapse
Affiliation(s)
| | - Rebecca S. Hock
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | | | - Heidi Judge
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Cristina Cusin
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Sanjay J. Mathew
- Department of Psychiatry, Baylor College of Medicine/Michael E. Debakey VA Medical Center, Houston, TX
| | | | - Dan V. Iosifescu
- Department of Psychiatry, NYU School of Medicine and Nathan Kline Institute, New York, NY
| | - Charles DeBattista
- Department of Psychiatry, Stanford University School of Medicine, Palo Alto, CA
| | | | - Maurizio Fava
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
44
|
Barbosa MG, Sarin LM, Tuena MA, Del Sant LC, Nakahira C, Magalhães EJM, Fava VAR, Delfino RS, Steglich MS, Puertas C, Lucchese AC, Surjan JC, Abdo GL, Cogo-Moreira H, Del Porto JA, Lacerda ALTD, Jackowski AP. Heterogeneous trajectory of depressive symptoms after repeated subcutaneous esketamine injections. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2021. [DOI: 10.1016/j.jadr.2020.100053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
45
|
Chen MH, Kao CF, Tsai SJ, Li CT, Lin WC, Hong CJ, Bai YM, Tu PC, Su TP. Treatment response to low-dose ketamine infusion for treatment-resistant depression: A gene-based genome-wide association study. Genomics 2020; 113:507-514. [PMID: 33370585 DOI: 10.1016/j.ygeno.2020.12.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUNDS Evidence suggested the crucial roles of brain-derived neurotrophic factor (BDNF) and glutamate system functioning in the antidepressant mechanisms of low-dose ketamine infusion in treatment-resistant depression (TRD). METHODS 65 patients with TRD were genotyped for 684,616 single nucleotide polymorphisms (SNPs). Twelve ketamine-related genes were selected for the gene-based genome-wide association study on the antidepressant effect of ketamine infusion and the resulting serum ketamine and norketamine levels. RESULTS Specific SNPs and whole genes involved in BDNF-TrkB signaling (i.e., rs2049048 in BDNF and rs10217777 in NTRK2) and the glutamatergic and GABAergic systems (i.e., rs16966731 in GRIN2A) were associated with the rapid (within 240 min) and persistent (up to 2 weeks) antidepressant effect of low-dose ketamine infusion and with serum ketamine and norketamine levels. DISCUSSION Our findings confirmed the predictive roles of BDNF-TrkB signaling and glutamatergic and GABAergic systems in the underlying mechanisms of low-dose ketamine infusion for TRD treatment.
Collapse
Affiliation(s)
- Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Chung-Feng Kao
- Department of Agronomy, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung, Taiwan; Advanced Plant Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan.
| | - Cheng-Ta Li
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Chen Lin
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Chen-Jee Hong
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Ya-Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Chi Tu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tung-Ping Su
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan; Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan.
| |
Collapse
|
46
|
Involvement of NMDA receptors containing the GluN2C subunit in the psychotomimetic and antidepressant-like effects of ketamine. Transl Psychiatry 2020; 10:427. [PMID: 33303736 PMCID: PMC7729946 DOI: 10.1038/s41398-020-01110-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 11/05/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
Acute ketamine administration evokes rapid and sustained antidepressant effects in treatment-resistant patients. However, ketamine also produces transient perceptual disturbances similarly to those evoked by other non-competitive NMDA-R antagonists like phencyclidine (PCP). Although the brain networks involved in both ketamine actions are not fully understood, PCP and ketamine activate thalamo-cortical networks after NMDA-R blockade in GABAergic neurons of the reticular thalamic nucleus (RtN). Given the involvement of thalamo-cortical networks in processing sensory information, these networks may underlie psychotomimetic action. Since the GluN2C subunit is densely expressed in the thalamus, including the RtN, we examined the dependence of psychotomimetic and antidepressant-like actions of ketamine on the presence of GluN2C subunits, using wild-type and GluN2C knockout (GluN2CKO) mice. Likewise, since few studies have investigated ketamine's effects in females, we used mice of both sexes. GluN2C deletion dramatically reduced stereotyped (circling) behavior induced by ketamine in male and female mice, while the antidepressant-like effect was fully preserved in both genotypes and sexes. Despite ketamine appeared to induce similar effects in both sexes, some neurobiological differences were observed between male and female mice regarding c-fos expression in thalamic nuclei and cerebellum, and glutamate surge in prefrontal cortex. In conclusion, the GluN2C subunit may discriminate between antidepressant-like and psychotomimetic actions of ketamine. Further, the abundant presence of GluN2C subunits in the cerebellum and the improved motor coordination of GluN2CKO mice after ketamine treatment suggest the involvement of cerebellar NMDA-Rs in some behavioral actions of ketamine.
Collapse
|
47
|
Gill H, Gill B, El-Halabi S, Chen-Li D, Lipsitz O, Rosenblat JD, Van Rheenen TE, Rodrigues NB, Mansur RB, Majeed A, Lui LMW, Nasri F, Lee Y, Mcintyre RS. Antidepressant Medications and Weight Change: A Narrative Review. Obesity (Silver Spring) 2020; 28:2064-2072. [PMID: 33022115 DOI: 10.1002/oby.22969] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/25/2020] [Accepted: 06/24/2020] [Indexed: 12/22/2022]
Abstract
Antidepressant medications are the first-line treatment option for moderate to severe major depressive disorder. However, most antidepressants have numerous documented adverse events, including cardiometabolic effects and weight gain, which are major public health concerns. Antidepressant agents provide varying risk of associated weight gain, including significant within-class differences. Some agents, such as mirtazapine, show significant levels of weight gain, while others, such as bupropion, demonstrate weight-loss effects. Current findings suggest the role of histamine and serotonin off-target appetite-promoting pathways in adverse weight-gain effects. Therefore, controlling for undesired weight effects is an important consideration for the selection of antidepressants.
Collapse
Affiliation(s)
- Hartej Gill
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Barjot Gill
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
| | - Sabine El-Halabi
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
| | - David Chen-Li
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
| | - Orly Lipsitz
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
| | - Joshua Daniel Rosenblat
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
| | - Tamsyn E Van Rheenen
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Mental Health, Faculty of Health, Arts and Design, School of Health Sciences, Swinburne University, Melbourne, Victoria, Australia
| | - Nelson B Rodrigues
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Amna Majeed
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
| | - Leanna M W Lui
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
| | - Flora Nasri
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
| | - Yena Lee
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Roger S Mcintyre
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, Ontario, Canada
- Department of Pharmacology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
48
|
Beaudequin D, Can AT, Dutton M, Jones M, Gallay C, Schwenn P, Yang C, Forsyth G, Simcock G, Hermens DF, Lagopoulos J. Predicting therapeutic response to oral ketamine for chronic suicidal ideation: a Bayesian network for clinical decision support. BMC Psychiatry 2020; 20:519. [PMID: 33115424 PMCID: PMC7594449 DOI: 10.1186/s12888-020-02925-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/19/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The glutamatergic modulator ketamine has been shown to result in rapid reductions in both suicidal ideation (SI) and depressive symptoms in clinical trials. There is a practical need for identification of pre-treatment predictors of ketamine response. Previous studies indicate links between treatment response and body mass index (BMI), depression symptoms and previous suicide attempts. Our aim was to explore the use of clinical and demographic factors to predict response to serial doses of oral ketamine for chronic suicidal ideation. METHODS Thirty-two participants completed the Oral Ketamine Trial on Suicidality (OKTOS). Data for the current study were drawn from pre-treatment and follow-up time-points of OKTOS. Only clinical and sociodemographic variables were included in this analysis. Data were used to create a proof of concept Bayesian network (BN) model of variables predicting prolonged response to oral ketamine, as defined by the Beck Scale for Suicide Ideation (BSS). RESULTS The network of potential predictors of response was evaluated using receiver operating characteristic (ROC) curve analyses. A combination of nine demographic and clinical variables predicted prolonged ketamine response, with strong contributions from BMI, Social and Occupational Functioning Assessment Scale (SOFAS), Montgomery-Asberg Depression Rating Scale (MADRS), number of suicide attempts, employment status and age. We evaluated and optimised the proposed network to increase the area under the ROC curve (AUC). The performance evaluation demonstrated that the BN predicted prolonged ketamine response with 97% accuracy, and AUC = 0.87. CONCLUSIONS At present, validated tools to facilitate risk assessment are infrequently used in psychiatric practice. Pre-treatment assessment of individuals' likelihood of response to oral ketamine for chronic suicidal ideation could be beneficial in making more informed decisions about likelihood of success for this treatment course. Clinical trials registration number ACTRN12618001412224, retrospectively registered 23/8/2018.
Collapse
Affiliation(s)
- Denise Beaudequin
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia.
| | - Adem T Can
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia
| | - Megan Dutton
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia
| | - Monique Jones
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia
| | - Cyrana Gallay
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia
| | - Paul Schwenn
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia
| | - Cian Yang
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia
| | - Grace Forsyth
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia
| | - Gabrielle Simcock
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia
| | - Daniel F Hermens
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia
| | - Jim Lagopoulos
- Thompson Institute, University of the Sunshine Coast, Locked Bag 4 (ML59), Maroochydore DC, QLD, 4558, Australia
| |
Collapse
|
49
|
Brunelin J, Iceta S, Plaze M, Gaillard R, Simon L, Suaud-Chagny MF, Galvao F, Poulet E. The Combination of Propofol and Ketamine Does Not Enhance Clinical Responses to Electroconvulsive Therapy in Major Depression-The Results From the KEOpS Study. Front Pharmacol 2020; 11:562137. [PMID: 33041803 PMCID: PMC7522396 DOI: 10.3389/fphar.2020.562137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/24/2020] [Indexed: 11/13/2022] Open
Abstract
Objective We investigated the clinical effects of the combination of ketamine and propofol as anesthetic agents during electroconvulsive therapy (ECT) in patients with uni- or bipolar major depressive episodes. We hypothesized that ketamine may confer short- and long- term advantages in improving depressive symptoms at the early stages of ECT. Methods In a randomized placebo-controlled trial, remission rates after 4 and 8 weeks of ECT were compared between patients who were randomly allocated to receive either the combination of ketamine (0.5 mg/kg) + propofol (n= 11) or placebo + propofol (n = 16). Depressive symptoms were assessed weekly using the Montgomery-Åsberg Depression Rating Scale (MADRS); ECT sessions were administered twice per week for a maximum of 8 weeks (16 sessions). Results After 4 weeks, we observed significantly fewer remitters (MADRS score < 10) in the ketamine + propofol group (0/11; 0%) than in the placebo + propofol group (5/16; 31%; χ2 = 4.22; p = 0.040). No significant difference was observed between the two groups regarding the number of patients who achieved remission weekly throughout the study period (Chi² = 3.588; p = 0.058). The mean duration of seizures was significantly shorter in the ketamine + propofol group than in the placebo + propofol group. Conclusions The results from the current study corroborated results from previously published studies and did not support the use of the combination of ketamine + propofol as an anesthetic agent for ECT in patients with major depressive episodes in clinical settings.
Collapse
Affiliation(s)
- Jerome Brunelin
- CH Le Vinatier, Bron, France.,INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, PSYR2 Team, Université de Lyon, Lyon, France
| | - Sylvain Iceta
- CH Le Vinatier, Bron, France.,INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, PSYR2 Team, Université de Lyon, Lyon, France.,Obesity Research Center, Quebec Heart and Lung Institute (IUCPQ), Québec, QC, Canada.,School of Nutrition, Laval University, Québec, QC, Canada
| | - Marion Plaze
- GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| | - Raphaël Gaillard
- GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| | - Louis Simon
- CH Le Vinatier, Bron, France.,INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, PSYR2 Team, Université de Lyon, Lyon, France
| | - Marie-Françoise Suaud-Chagny
- CH Le Vinatier, Bron, France.,INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, PSYR2 Team, Université de Lyon, Lyon, France
| | | | - Emmanuel Poulet
- CH Le Vinatier, Bron, France.,INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, PSYR2 Team, Université de Lyon, Lyon, France.,Service de psychiatrie des urgences, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
50
|
Nuñez NA, Joseph B, Pahwa M, Seshadri A, Prokop LJ, Kung S, Schak KM, Vande Voort JL, Frye MA, Singh B. An Update on the Efficacy and Tolerability of Oral Ketamine for Major Depression: A Systematic Review and Meta-Analysis. PSYCHOPHARMACOLOGY BULLETIN 2020; 50:137-163. [PMID: 33012876 PMCID: PMC7511150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
BACKGROUND Intravenous Ketamine has shown robust antidepressant efficacy although other routes of administration are currently needed. We conducted a systematic review and meta-analysis of studies evaluating the efficacy and tolerability of oral ketamine for depression. METHODS A comprehensive search of major electronic databases from inception to April 2020 was performed. Studies of oral ketamine for depression, from case series to randomized clinical trials, were eligible. Randomized controlled trials were included in a meta-analysis, focusing on response, remission, time to effect, and side effects. RESULTS A total of 917 articles were identified with 890 studies screened, yielding a total of 10 studies included in our systematic review.Three randomized controlled trials (RCTs) (N = 161, mean age 37.9 ± 9.5 years, 58.6% females) were included in the meta-analysis. Pooled analysis suggested a significant antidepressant effect of oral ketamine (SMD: -0.75; 95% CI: -1.08, -0.43; p<0.0001; I2 = 0%) although remission rates (RR:2.77; 95% CI:0.96, 8.00; p = 0.06) and response rates (RR:2.58; 95% CI:0.94,7.08; p = 0.07) were marginal compared to placebo at the endpoint. Oral ketamine antidepressant effects seemed to take effect at the 2nd week (SMD: -0.71; 95% CI: -1.08, -0.35; p = 0.001; I2 = 0%). There were no significant differences in the overall side-effects between oral ketamine and the placebo group (RR 1.28, 95% CI: 0.89-1.83; p = 0.19). CONCLUSION This focused meta-analysis of oral ketamine suggests a marginal efficacy for major depressive disorder without increased risk of adverse events. Further larger sample studies are needed to confirm these preliminary findings, analyzing differential response/remission rates by affective disorder, optimal dosing strategies, and its long-term effects.
Collapse
Affiliation(s)
- Nicolas A Nuñez
- Nuñez, MD, MS, Pahwa, MD, Seshadri, MD, Kung, MD, Schak, MD, VandeVoort, MD, Frye, MD,Singh MD, MS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA. Joseph, MBBS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. Prokop, MLS, Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Boney Joseph
- Nuñez, MD, MS, Pahwa, MD, Seshadri, MD, Kung, MD, Schak, MD, VandeVoort, MD, Frye, MD,Singh MD, MS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA. Joseph, MBBS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. Prokop, MLS, Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Mehak Pahwa
- Nuñez, MD, MS, Pahwa, MD, Seshadri, MD, Kung, MD, Schak, MD, VandeVoort, MD, Frye, MD,Singh MD, MS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA. Joseph, MBBS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. Prokop, MLS, Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Ashok Seshadri
- Nuñez, MD, MS, Pahwa, MD, Seshadri, MD, Kung, MD, Schak, MD, VandeVoort, MD, Frye, MD,Singh MD, MS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA. Joseph, MBBS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. Prokop, MLS, Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Larry J Prokop
- Nuñez, MD, MS, Pahwa, MD, Seshadri, MD, Kung, MD, Schak, MD, VandeVoort, MD, Frye, MD,Singh MD, MS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA. Joseph, MBBS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. Prokop, MLS, Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Simon Kung
- Nuñez, MD, MS, Pahwa, MD, Seshadri, MD, Kung, MD, Schak, MD, VandeVoort, MD, Frye, MD,Singh MD, MS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA. Joseph, MBBS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. Prokop, MLS, Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Kathryn M Schak
- Nuñez, MD, MS, Pahwa, MD, Seshadri, MD, Kung, MD, Schak, MD, VandeVoort, MD, Frye, MD,Singh MD, MS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA. Joseph, MBBS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. Prokop, MLS, Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jennifer L Vande Voort
- Nuñez, MD, MS, Pahwa, MD, Seshadri, MD, Kung, MD, Schak, MD, VandeVoort, MD, Frye, MD,Singh MD, MS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA. Joseph, MBBS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. Prokop, MLS, Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Mark A Frye
- Nuñez, MD, MS, Pahwa, MD, Seshadri, MD, Kung, MD, Schak, MD, VandeVoort, MD, Frye, MD,Singh MD, MS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA. Joseph, MBBS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. Prokop, MLS, Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Balwinder Singh
- Nuñez, MD, MS, Pahwa, MD, Seshadri, MD, Kung, MD, Schak, MD, VandeVoort, MD, Frye, MD,Singh MD, MS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA. Joseph, MBBS, Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA. Prokop, MLS, Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|