1
|
Oraee S, Alinejadfard M, Golsorkh H, Sadeghian M, Fanaei M, Centis R, D'Ambrosio L, Sotgiu G, Goudarzi H, Migliori GB, Nasiri MJ. Intranasal esketamine for patients with major depressive disorder: A systematic review and meta-analysis. J Psychiatr Res 2024; 180:371-379. [PMID: 39522447 DOI: 10.1016/j.jpsychires.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/27/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
A large number of people worldwide grapple with Major Depressive Disorder (MDD), and a significant portion of them confront resistance to conventional antidepressant treatments. Esketamine, an innovative intervention, has garnered attention due to its rapid-acting antidepressant effects. This meta-analysis aimed to evaluate the efficacy and safety of esketamine in individuals with MDD and Treatment-Resistant Depression (TRD). We performed an extensive search of electronic databases, including PubMed, EMBASE, and Cochrane Library, covering the period from inception to January 15, 2024, to identify clinical trials investigating the efficacy, safety, and tolerability of esketamine in individuals with MDD and TRD. Meta-analyses were conducted using a random-effect model, with the risk ratio (RR) chosen as the effect size. After screening, nine studies met inclusion criteria comprising 1752 patients. Esketamine dosages varied from 28 to 84 mg, administered intranasally in conjunction with an oral antidepressant. Intranasal esketamine exhibited significantly higher remission rates compared to the placebo (RR = 1.371, 95% CI: 1.194 to 1.574, p < 0.0001). Subgroup analysis revealed that the 84 mg and flexible doses were particularly effective. Moreover, intranasal esketamine demonstrated higher response rates compared to the placebo (RR = 1.274, 95% CI: 1.108 to 1.465, p-value = 0.001). Although adverse events were common in the esketamine group, they were generally tolerable. Intranasal esketamine demonstrated effectiveness in reducing scores among patients with MDD and TRD. The drug exhibited both safety and tolerability. However, further research is warranted to investigate its long-term effects and efficacy in specific patient subgroups.
Collapse
Affiliation(s)
- Soroush Oraee
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hossein Golsorkh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Sadeghian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maryam Fanaei
- Clinical Psychology Department, Islamic Azad University, Qom, Iran
| | - Rosella Centis
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Lia D'Ambrosio
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | - Giovanni Sotgiu
- Clinical Epidemiology and Medical Statistics Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Italy
| | - Hossein Goudarzi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Giovanni Battista Migliori
- Servizio di Epidemiologia Clinica delle Malattie Respiratorie, Istituti Clinici Scientifici Maugeri IRCCS, Tradate, Italy
| | | |
Collapse
|
2
|
Raja SM, Guptill JT, Mack M, Peterson M, Byard S, Twieg R, Jordan L, Rich N, Castledine R, Bourne S, Wilmshurst M, Oxendine S, Avula SG, Zuleta H, Quigley P, Lawson S, McQuaker SJ, Ahmadkhaniha R, Appelbaum LG, Kowalski K, Barksdale CT, Gufford BT, Awan A, Sancho AR, Moore MC, Berrada K, Cogan GB, DeLaRosa J, Radcliffe J, Pao M, Kennedy M, Lawrence Q, Goldfeder L, Amanfo L, Zanos P, Gilbert JR, Morris PJ, Moaddel R, Gould TD, Zarate CA, Thomas CJ. A Phase 1 Assessment of the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of (2R,6R)-Hydroxynorketamine in Healthy Volunteers. Clin Pharmacol Ther 2024; 116:1314-1324. [PMID: 39054770 PMCID: PMC11479831 DOI: 10.1002/cpt.3391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
(R,S)-Ketamine (ketamine) is a dissociative anesthetic that also possesses analgesic and antidepressant activity. Undesirable dissociative side effects and misuse potential limit expanded use of ketamine in several mental health disorders despite promising clinical activity and intensifying medical need. (2R,6R)-Hydroxynorketamine (RR-HNK) is a metabolite of ketamine that lacks anesthetic and dissociative activity but maintains antidepressant and analgesic activity in multiple preclinical models. To enable future assessments in selected human indications, we report the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of RR-HNK in a Phase 1 study in healthy volunteers (NCT04711005). A six-level single-ascending dose (SAD) (0.1-4 mg/kg) and a two-level multiple ascending dose (MAD) (1 and 2 mg/kg) study was performed using a 40-minute IV administration emulating the common practice for ketamine administration for depression. Safety assessments showed RR-HNK possessed a minimal adverse event profile and no serious adverse events at all doses examined. Evaluations of dissociation and sedation demonstrated that RR-HNK did not possess anesthetic or dissociative characteristics in the doses examined. RR-HNK PK parameters were measured in both the SAD and MAD studies and exhibited dose-proportional increases in exposure. Quantitative electroencephalography (EEG) measurements collected as a PD parameter based on preclinical findings and ketamine's established effect on gamma-power oscillations demonstrated increases of gamma power in some participants at the lower/mid-range doses examined. Cerebrospinal fluid examination confirmed RR-HNK exposure within the central nervous system (CNS). Collectively, these data demonstrate RR-HNK is well tolerated with an acceptable PK profile and promising PD outcomes to support the progression into Phase 2.
Collapse
Affiliation(s)
- Shruti M. Raja
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jeffrey T. Guptill
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
- Argenx BV, 9052 Gent, Belgium
| | - Michelle Mack
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - Stephen Byard
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | - Robert Twieg
- Labcorp Bioanalytical Services, Indianapolis, IN, 46214, USA
| | - Lynn Jordan
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | | | - Samuel Bourne
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | | | - Sarah Oxendine
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - Helen Zuleta
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Paul Quigley
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | - Sheila Lawson
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | | | - Reza Ahmadkhaniha
- National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 22124, USA
| | - Lawrence G. Appelbaum
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kevin Kowalski
- Labcorp Bioanalytical Services, Indianapolis, IN, 46214, USA
| | | | - Brandon T. Gufford
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Asaad Awan
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alfredo R. Sancho
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Max C. Moore
- Drug Discovery and Development Program, Frederick National Laboratory, Fredrick, MD, 21701, USA
| | - Karim Berrada
- Drug Discovery and Development Program, Frederick National Laboratory, Fredrick, MD, 21701, USA
| | - Gregory B. Cogan
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jesse DeLaRosa
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jeanne Radcliffe
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Maryland Pao
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | - Lisa Goldfeder
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Leslie Amanfo
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Panos Zanos
- Department of Psychology, University of Cyprus, Nicosia, 2109, Cyprus
| | - Jessica R Gilbert
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Patrick J. Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, 20850, USA
| | - Ruin Moaddel
- National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 22124, USA
| | - Todd D. Gould
- Departments of Psychiatry, Pharmacology, and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Baltimore Veterans Affairs Medical Center, Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, 20850, USA
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
3
|
Bremshey S, Groß J, Renken K, Masseck OA. The role of serotonin in depression-A historical roundup and future directions. J Neurochem 2024; 168:1751-1779. [PMID: 38477031 DOI: 10.1111/jnc.16097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Depression is one of the most common psychiatric disorders worldwide, affecting approximately 280 million people, with probably much higher unrecorded cases. Depression is associated with symptoms such as anhedonia, feelings of hopelessness, sleep disturbances, and even suicidal thoughts. Tragically, more than 700 000 people commit suicide each year. Although depression has been studied for many decades, the exact mechanisms that lead to depression are still unknown, and available treatments only help a fraction of patients. In the late 1960s, the serotonin hypothesis was published, suggesting that serotonin is the key player in depressive disorders. However, this hypothesis is being increasingly doubted as there is evidence for the influence of other neurotransmitters, such as noradrenaline, glutamate, and dopamine, as well as larger systemic causes such as altered activity in the limbic network or inflammatory processes. In this narrative review, we aim to contribute to the ongoing debate on the involvement of serotonin in depression. We will review the evolution of antidepressant treatments, systemic research on depression over the years, and future research applications that will help to bridge the gap between systemic research and neurotransmitter dynamics using biosensors. These new tools in combination with systemic applications, will in the future provide a deeper understanding of the serotonergic dynamics in depression.
Collapse
Affiliation(s)
- Svenja Bremshey
- Synthetic Biology, University of Bremen, Bremen, Germany
- Neuropharmacology, University of Bremen, Bremen, Germany
| | - Juliana Groß
- Synthetic Biology, University of Bremen, Bremen, Germany
| | - Kim Renken
- Synthetic Biology, University of Bremen, Bremen, Germany
| | | |
Collapse
|
4
|
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
|
5
|
Khalifian C, Rashkovsky K, Mitchell E, Bismark A, Wagner AC, Knopp KC. A novel framework for ketamine-assisted couple therapy. Front Psychiatry 2024; 15:1376646. [PMID: 39193577 PMCID: PMC11347343 DOI: 10.3389/fpsyt.2024.1376646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/17/2024] [Indexed: 08/29/2024] Open
Abstract
Intimate relationship distress is prevalent and is associated with poorer health, mental health, and mortality outcomes. Evidence-based couple therapies target cognitive, behavioral, and emotional processes that underlie relationship dysfunction. Increasing research and clinical evidence supports the efficacy of ketamine-assisted psychotherapy (KAP) for addressing clinical mental health concerns, including depression, anxiety disorders, posttraumatic stress disorder, and more. The purported mechanisms of KAP are also likely to improve psychosocial and relational functioning for patients and may be useful for supporting change mechanisms in couple therapy. This paper reviews the current evidence for therapeutic ketamine and KAP and outlines how the mechanisms of ketamine therapy may also augment the cognitive, behavioral, and emotional interventions in the most commonly used evidence-based couple therapies. Key mechanisms include increased neuroplasticity, changes in functional connectivity, adaptive dissociation, decreased inhibition, and reduced avoidance. Given the reciprocal interaction between relationship dysfunction and mental health problems, ketamine may also help alleviate relationship distress by directly treating clinical mental health symptoms. We then outline a proposed framework for ketamine-assisted couple therapy, addressing the application of KAP preparation, dosing, and integration to a dyadic intervention framework in a way that can be applied to different couple therapy modalities. This clinical framework for couples' KAP may be useful for clinicians and researchers working to improve the efficacy of couple therapy, particularly when one or both partners has accompanying mental health concerns.
Collapse
Affiliation(s)
- C. Khalifian
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - K. Rashkovsky
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - E. Mitchell
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - A. Bismark
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - A. C. Wagner
- Remedy, Toronto, ON, Canada
- Department of Psychology, Toronto Metropolitan University, Toronto, ON, Canada
| | - K. C. Knopp
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
6
|
Algin S, Banik D, Rahman SA, Mahmud Tusher S, Tuj Johora F, Akter A, Ahmed T, Biswas MAM, Sinha S, Haque M. Ketamine Infusion in a Resistant Obsessive-Compulsive Disorder Patient in Bangladesh with Severe Suicidal Ideation: A Case Report. Cureus 2024; 16:e57877. [PMID: 38596207 PMCID: PMC11002706 DOI: 10.7759/cureus.57877] [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] [Accepted: 04/08/2024] [Indexed: 04/11/2024] Open
Abstract
Treatment of resistant obsessive-compulsive disorder (OCD) typically results in insufficient symptom alleviation, and even long-term medication often fails to have the intended effect. Ketamine is a potent non-competitive antagonist of the N-methyl-D-aspartate (NMDA) receptor. Studies have shown that low-dose ketamine infusion results in a considerable reduction in obsessive-compulsive symptoms and a rapid resolution of suicidal ideation. This is a case report on the effect of intravenous ketamine infusion on a patient with resistant OCD and severe suicidal ideation. Intravenous (IV) ketamine was given once a week over consecutive three weeks with necessary precautions. Psychometric tools such as the Yale-Brown Obsessive Compulsive Scale (Y-BOCS), the Clinical Global Impressions Scale (CGI-S), the Beck Scale for Suicidal Ideations (BSSI), and Depression Anxiety and Stress Scale 21 (DASS-21) were applied before and after infusions. Obsessive-compulsive symptoms and suicidal severity started to decrease rapidly after the first infusion. However, after a transient improvement, these symptoms again began to increase after a stressful incident on the second day of the first infusion. All the symptoms measured by validated rating scales showed continued improvement after the following two infusions. The improvement was sustained until discharge (one week after the last infusion) and subsequent follow-up in the sixth and 12th weeks. The role of ketamine in reducing suicidal thoughts and behavior is already established. Very few studies emphasized its effectiveness in improving severe/resistant obsessive-compulsive symptoms. This pioneering work may offer scope for similar research in the relevant field.
Collapse
Affiliation(s)
- Sultana Algin
- Psychiatry, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - Debasish Banik
- Anesthesia, Analgesia, and Intensive Care Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - Sm Atikur Rahman
- Psychiatry, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - Saiful Mahmud Tusher
- Anesthesia, Analgesia, and Intensive Care Medicine, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - Fatema Tuj Johora
- Child and Adolescent Psychiatry, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - Asha Akter
- Psychiatry, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - Tanbir Ahmed
- Child and Adolescent Psychiatry, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | | | - Susmita Sinha
- Physiology, Khulna City Medical College and Hospital, Khulna, BGD
| | - Mainul Haque
- Karnavati Scientific Research Center (KSRC), School of Dentistry, Karnavati University, Gandhinagar, IND
- Pharmacology and Therapeutics, National Defence University of Malaysia, Kuala Lumpur, MYS
| |
Collapse
|
7
|
Whitaker T, Farrand KF, Thase ME. A Phase 2 Open Label Study of Efficacy, Safety, and Tolerability of SLS-002 (Intranasal Racemic Ketamine) in Adults with MDD at Imminent Risk of Suicide. PSYCHOPHARMACOLOGY BULLETIN 2024; 54:8-17. [PMID: 38449472 PMCID: PMC10913863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Background Despite the prevalence of Major Depressive Disorder (MDD) and the propensity of affected individuals to eventually die by suicide, there is no therapeutic approved specifically for suicidal ideation and behavior (SI/B) in MDD. The NMDA receptor antagonist ketamine has been investigated for the treatment of depression and shown to have a rapid effect on symptoms. Spravato® (esketamine) is approved by the FDA for use in treatment-resistant depression and Major Depressive Episodes with Suicidal Ideation based on studies conducted in adults also taking standard antidepressants. While esketamine was associated with a large reduction in suicidality indicators, the effects did not significantly exceed those associated with placebo. Racemic ketamine, a mixture of both esketamine and arketamine, may hold greater potential for the rapid alleviation of SI/B. SLS-002 was developed as an investigational intranasal racemic ketamine for the treatment of SI/B in individuals with MDD. Methods In part one of a two-part clinical trial, the safety, tolerability, and potential effectiveness of SLS-002 were evaluated in an open label study of 17 patients with MDD hospitalized with acute SI/B. Results Treatment with SLS-002 was associated with a significant reduction in depression and suicidality indicators on four clinical scales: the Montgomery-Åsberg Depression Rating Scale, the Sheehan-Suicidality Tracking Scale, and the Clinical and Patient Global Impression Scales for SI/B. SLS-002 was well tolerated with an acceptable safety profile. Conclusions The results of this open label study support the continued development of SLS-002. The randomized double-blind placebo-controlled part two of this trial was recently completed.
Collapse
Affiliation(s)
- Timothy Whitaker
- Whitaker, MD, Chief Medical Officer, Head of Research & Development, Seelos Therapeutics, Inc., New York, NY
| | - Kimberly F Farrand
- Farrand, MPH, Senior Director, Clinical Development, Seelos Therapeutics, Inc., New York, NY
| | - Michael E Thase
- Thase, MD, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA and Corporal Michael J Crescenz Veterans Affairs Medical Center, Philadelphia, PA
| |
Collapse
|
8
|
Ferber SG, Weller A, Soreq H. Boltzmann's Theorem Revisited: Inaccurate Time-to-Action Clocks in Affective Disorders. Curr Neuropharmacol 2024; 22:1762-1777. [PMID: 38500272 PMCID: PMC11284727 DOI: 10.2174/1570159x22666240315100326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/14/2023] [Accepted: 12/17/2023] [Indexed: 03/20/2024] Open
Abstract
Timely goal-oriented behavior is essential for survival and is shaped by experience. In this paper, a multileveled approach was employed, ranging from the polymorphic level through thermodynamic molecular, cellular, intracellular, extracellular, non-neuronal organelles and electrophysiological waves, attesting for signal variability. By adopting Boltzmann's theorem as a thermodynamic conceptualization of brain work, we found deviations from excitation-inhibition balance and wave decoupling, leading to wider signal variability in affective disorders compared to healthy individuals. Recent evidence shows that the overriding on-off design of clock genes paces the accuracy of the multilevel parallel sequencing clocks and that the accuracy of the time-to-action is more crucial for healthy behavioral reactions than their rapidity or delays. In affective disorders, the multilevel clocks run free and lack accuracy of responsivity to environmentally triggered time-to-action as the clock genes are not able to rescue mitochondria organelles from oxidative stress to produce environmentally-triggered energy that is required for the accurate time-to-action and maintenance of the thermodynamic equilibrium. This maintenance, in turn, is dependent on clock gene transcription of electron transporters, leading to higher signal variability and less signal accuracy in affective disorders. From a Boltzmannian thermodynamic and energy-production perspective, the option of reversibility to a healthier time-toaction, reducing entropy is implied. We employed logic gates to show deviations from healthy levelwise communication and the reversed conditions through compensations implying the role of nonneural cells and the extracellular matrix in return to excitation-inhibition balance and accuracy in the time-to-action signaling.
Collapse
Affiliation(s)
- Sari Goldstein Ferber
- Psychology Department and The Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, USA
| | - Aron Weller
- Psychology Department and The Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Hermona Soreq
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
9
|
Kamenish K, Robinson ESJ. Neuropsychological Effects of Antidepressants: Translational Studies. Curr Top Behav Neurosci 2024; 66:101-130. [PMID: 37955824 DOI: 10.1007/7854_2023_446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Pharmacological treatments that improve mood were first identified serendipitously, but more than half a century later, how these drugs induce their antidepressant effects remains largely unknown. With the help of animal models, a detailed understanding of their pharmacological targets and acute and chronic effects on brain chemistry and neuronal function has been achieved, but it remains to be elucidated how these effects translate to clinical efficacy. Whilst the field has been dominated by the monoamine and neurotrophic hypotheses, the idea that the maladaptive cognitive process plays a critical role in the development and perpetuation of mood disorders has been discussed since the 1950s. Recently, studies using objective methods to quantify changes in emotional processing found acute effects with conventional antidepressants in both healthy volunteers and patients. These positive effects on emotional processing and cognition occur without a change in the subjective ratings of mood. Building from these studies, behavioural methods for animals that quantify similar cognitive affective processes have been developed. Integrating these behavioural approaches with pharmacology and targeted brain manipulations, a picture is beginning to emerge of the underlying mechanisms that may link the pharmacology of antidepressants, these neuropsychological constructs and clinical efficacy. In this chapter, we discuss findings from animal studies, experimental medicine and patients investigating the neuropsychological effects of antidepressant drugs. We discuss the possible neural circuits that contribute to these effects and discuss whether a neuropsychological model of antidepressant effects could explain the temporal differences in clinical benefits observed with conventional delayed-onset antidepressants versus rapid-acting antidepressants.
Collapse
Affiliation(s)
- Katie Kamenish
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, UK
| | - Emma S J Robinson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, UK.
| |
Collapse
|
10
|
Sperber PS, Gebert P, Broersen LH, Kufner A, Huo S, Piper SK, Teegen B, Heuschmann PU, Prüss H, Endres M, Liman TG, Siegerink B. Depressive symptoms and anti-N-methyl-D-aspartate-receptor GluN1 antibody seropositivity in the PROSpective cohort with incident stroke. Brain Behav Immun Health 2023; 34:100705. [PMID: 38033615 PMCID: PMC10684375 DOI: 10.1016/j.bbih.2023.100705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/05/2023] [Indexed: 12/02/2023] Open
Abstract
Background Anti-NMDA-receptor GluN1 antibodies (NMDAR1-abs) are present in an autoimmune encephalitis with severe neuropsychiatric symptoms. We aimed to estimate the impact of serum NMDAR1-abs on depressive symptoms years after first-ever ischemic stroke (IS). Methods Data were used from the PROSpective Cohort with Incident Stroke-Berlin (PROSCIS-B; NCT01363856). Serum NMDAR1-abs (IgM/IgA/IgG) were measured within 7 days after IS using cell-based assays. We defined seropositivity as titers ≥1:10, thereof low titers as ≤1:100 and high titers as >1:100. We used the Center for Epidemiological Studies-Depression (CES-D) scale to measure depressive symptoms at year one, two and three following IS. We calculated crude and confounder adjusted weighted generalized linear models to quantify the impact of NMDAR1-abs on CES-D assessed at three annual time-points. Results NMDAR1-abs were measured in 583 PROSCIS-B IS patients (mean age = 67 [SD = 13]; 42%female; median NIHSS = 2 [IQR = 1-4]) of whom 76 (13%; IgM: n = 49/IgA: n = 43/IgG: n = 2) were seropositive, 55 (9%) with low and 21 (4%) with high titers. CES-D regarded over all follow-up time-points was higher in seropositive patients (βcrude = 2.56 [95%CI = -0.34 to 5.45]; βadjusted = 2.26 [95%CI = -0.68 to 5.20]) and effects were highest in patients with high titer (low titers: βcrude = 1.42 [95%CI = -1.79 to 4.62], βadjusted = 0.53 [95%CI = -2.47 to 3.54]; high titers: βcrude = 5.85 [95%CI = 0.20 to 11.50]; βadjusted = 7.20 [95%CI = 0.98 to 13.43]). Conclusion Patients with serum NMDAR1-abs (predominantly IgM&IgA) suffer more severe depressive symptoms after mild-to-moderate IS compared to NMDAR1-abs seronegative patients.
Collapse
Affiliation(s)
- Pia S. Sperber
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Center for Stroke Research Berlin (CSB), Berlin, Germany
- German Centre for Cardiovascular Research DZHK, Partner Site, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
- Charité – Universitätsmedizin Berlin & Max Delbrück Center for Molecular Medicine, Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Pimrapat Gebert
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Institute of Biometry and Clinical Epidemiology, Berlin, Germany
- Berlin Institute of Health (BIH), Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Leonie H.A. Broersen
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Center for Stroke Research Berlin (CSB), Berlin, Germany
| | - Anna Kufner
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Center for Stroke Research Berlin (CSB), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
| | - Shufan Huo
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Center for Stroke Research Berlin (CSB), Berlin, Germany
- German Centre for Cardiovascular Research DZHK, Partner Site, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
- Charité – Universitätsmedizin Berlin & Max Delbrück Center for Molecular Medicine, Experimental and Clinical Research Center (ECRC), Berlin, Germany
| | - Sophie K. Piper
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Institute of Biometry and Clinical Epidemiology, Berlin, Germany
- Berlin Institute of Health (BIH), Charité – Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité – Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Institute of Medical Informatics, Germany
| | - Bianca Teegen
- Institute of Experimental Immunology, EUROIMMUN AG, Luebeck, Germany
| | - Peter U. Heuschmann
- University of Würzburg, Institute of Clinical Epidemiology and Biometry, Würzburg, Germany
- University Hospital Würzburg, Clinical Trial Center Würzburg, Würzburg, Germany
| | - Harald Prüss
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
- German Center for Neurodegenerative Disease DZNE, Partner Site, Berlin, Germany
| | - Matthias Endres
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Center for Stroke Research Berlin (CSB), Berlin, Germany
- German Centre for Cardiovascular Research DZHK, Partner Site, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
- Charité – Universitätsmedizin Berlin & Max Delbrück Center for Molecular Medicine, Experimental and Clinical Research Center (ECRC), Berlin, Germany
- German Center for Neurodegenerative Disease DZNE, Partner Site, Berlin, Germany
| | - Thomas G. Liman
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Center for Stroke Research Berlin (CSB), Berlin, Germany
- German Centre for Cardiovascular Research DZHK, Partner Site, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Department of Neurology with Experimental Neurology, Berlin, Germany
- Carl von Ossietzky-University, Evangelisches Krankenhaus Oldenburg, Department of Neurology, Oldenburg, Germany
| | - Bob Siegerink
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, And Berlin Institute of Health, Center for Stroke Research Berlin (CSB), Berlin, Germany
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| |
Collapse
|
11
|
Ladagu AD, Olopade FE, Adejare A, Olopade JO. GluN2A and GluN2B N-Methyl-D-Aspartate Receptor (NMDARs) Subunits: Their Roles and Therapeutic Antagonists in Neurological Diseases. Pharmaceuticals (Basel) 2023; 16:1535. [PMID: 38004401 PMCID: PMC10674917 DOI: 10.3390/ph16111535] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ion channels that respond to the neurotransmitter glutamate, playing a crucial role in the permeability of calcium ions and excitatory neurotransmission in the central nervous system (CNS). Composed of various subunits, NMDARs are predominantly formed by two obligatory GluN1 subunits (with eight splice variants) along with regulatory subunits GluN2 (GluN2A-2D) and GluN3 (GluN3A-B). They are widely distributed throughout the CNS and are involved in essential functions such as synaptic transmission, learning, memory, plasticity, and excitotoxicity. The presence of GluN2A and GluN2B subunits is particularly important for cognitive processes and has been strongly implicated in neurodegenerative diseases like Parkinson's disease and Alzheimer's disease. Understanding the roles of GluN2A and GluN2B NMDARs in neuropathologies provides valuable insights into the underlying causes and complexities of major nervous system disorders. This knowledge is vital for the development of selective antagonists targeting GluN2A and GluN2B subunits using pharmacological and molecular methods. Such antagonists represent a promising class of NMDA receptor inhibitors that have the potential to be developed into neuroprotective drugs with optimal therapeutic profiles.
Collapse
Affiliation(s)
- Amany Digal Ladagu
- Department of Veterinary Anatomy, University of Ibadan, Ibadan 200284, Nigeria; (A.D.L.); (J.O.O.)
| | - Funmilayo Eniola Olopade
- Developmental Neurobiology Laboratory, Department of Anatomy, College of Medicine, University of Ibadan, Ibadan 200284, Nigeria
| | - Adeboye Adejare
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph’s University, Philadelphia, PA 19131, USA
| | - James Olukayode Olopade
- Department of Veterinary Anatomy, University of Ibadan, Ibadan 200284, Nigeria; (A.D.L.); (J.O.O.)
| |
Collapse
|
12
|
Acero VP, Cribas ES, Browne KD, Rivellini O, Burrell JC, O’Donnell JC, Das S, Cullen DK. Bedside to bench: the outlook for psychedelic research. Front Pharmacol 2023; 14:1240295. [PMID: 37869749 PMCID: PMC10588653 DOI: 10.3389/fphar.2023.1240295] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/30/2023] [Indexed: 10/24/2023] Open
Abstract
There has recently been a resurgence of interest in psychedelic compounds based on studies demonstrating their potential therapeutic applications in treating post-traumatic stress disorder, substance abuse disorders, and treatment-resistant depression. Despite promising efficacy observed in some clinical trials, the full range of biological effects and mechanism(s) of action of these compounds have yet to be fully established. Indeed, most studies to date have focused on assessing the psychological mechanisms of psychedelics, often neglecting the non-psychological modes of action. However, it is important to understand that psychedelics may mediate their therapeutic effects through multi-faceted mechanisms, such as the modulation of brain network activity, neuronal plasticity, neuroendocrine function, glial cell regulation, epigenetic processes, and the gut-brain axis. This review provides a framework supporting the implementation of a multi-faceted approach, incorporating in silico, in vitro and in vivo modeling, to aid in the comprehensive understanding of the physiological effects of psychedelics and their potential for clinical application beyond the treatment of psychiatric disorders. We also provide an overview of the literature supporting the potential utility of psychedelics for the treatment of brain injury (e.g., stroke and traumatic brain injury), neurodegenerative diseases (e.g., Parkinson's and Alzheimer's diseases), and gut-brain axis dysfunction associated with psychiatric disorders (e.g., generalized anxiety disorder and major depressive disorder). To move the field forward, we outline advantageous experimental frameworks to explore these and other novel applications for psychedelics.
Collapse
Affiliation(s)
- Victor P. Acero
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
- Penn Psychedelics Collaborative, University of Pennsylvania, Philadelphia, PA, United States
| | - Emily S. Cribas
- Penn Psychedelics Collaborative, University of Pennsylvania, Philadelphia, PA, United States
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kevin D. Browne
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Olivia Rivellini
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Penn Psychedelics Collaborative, University of Pennsylvania, Philadelphia, PA, United States
| | - Justin C. Burrell
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - John C. O’Donnell
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Penn Psychedelics Collaborative, University of Pennsylvania, Philadelphia, PA, United States
| | - Suradip Das
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - D. Kacy Cullen
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
13
|
Kaur P, Khan H, Grewal AK, Dua K, Singh TG. Therapeutic potential of NOX inhibitors in neuropsychiatric disorders. Psychopharmacology (Berl) 2023; 240:1825-1840. [PMID: 37507462 DOI: 10.1007/s00213-023-06424-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
RATIONALE Neuropsychiatric disorders encompass a broad category of medical conditions that include both neurology as well as psychiatry such as major depressive disorder, autism spectrum disorder, bipolar disorder, schizophrenia as well as psychosis. OBJECTIVE NADPH-oxidase (NOX), which is the free radical generator, plays a substantial part in oxidative stress in neuropsychiatric disorders. It is thought that elevated oxidative stress as well as neuroinflammation plays a part in the emergence of neuropsychiatric disorders. Including two linked with membranes and four with subunits of cytosol, NOX is a complex of multiple subunits. NOX has been linked to a significant source of reactive oxygen species in the brain. NOX has been shown to control memory processing and neural signaling. However, excessive NOX production has been linked to cardiovascular disorders, CNS degeneration, and neurotoxicity. The increase in NOX leads to the progression of neuropsychiatric disorders. RESULT Our review mainly emphasized the characteristics of NOX and its various mechanisms, the modulation of NOX in various neuropsychiatric disorders, and various studies supporting the fact that NOX might be the potential therapeutic target for neuropsychiatric disorders. CONCLUSION Here, we summarizes various pharmacological studies involving NOX inhibitors in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Parneet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | | | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | | |
Collapse
|
14
|
Barrutieta-Arberas I, Ortuzar N, Vaquero-Rodríguez A, Picó-Gallardo M, Bengoetxea H, Guevara MA, Gargiulo PA, Lafuente JV. The role of ketamine in major depressive disorders: Effects on parvalbumin-positive interneurons in hippocampus. Exp Biol Med (Maywood) 2023; 248:588-595. [PMID: 37158084 PMCID: PMC10350797 DOI: 10.1177/15353702231170007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
Major depressive disorder (MDD) is a complex illness that is arising as a growing public health concern. Although several brain areas are related to this type of disorders, at the cellular level, the parvalbumin-positive cells of the hippocampus interplay a very relevant role. They control pyramidal cell bursts, neuronal networks, basic microcircuit functions, and other complex neuronal tasks involved in mood disorders. In resistant depressions, the efficacy of current antidepressant treatments drops dramatically, so the new rapid-acting antidepressants (RAADs) are being postulated as novel treatments. Ketamine at subanesthetic doses and its derivative metabolites have been proposed as RAADs due to their rapid and sustained action by blocking N-methyl-d-aspartate (NMDA) receptors, which in turn lead to the release of brain-derived neurotrophic factor (BDNF). This mechanism produces a rapid plasticity activation mediated by neurotransmitter homeostasis, synapse recovery, and increased dendritic spines and therefore, it is a promising therapeutic approach to improve cognitive symptoms in MDD.
Collapse
Affiliation(s)
- I Barrutieta-Arberas
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - N Ortuzar
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain
| | - A Vaquero-Rodríguez
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain
| | - M Picó-Gallardo
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - H Bengoetxea
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain
| | - MA Guevara
- Laboratory of Neurosciences and Experimental Psychology, Area of Pharmacology, Department of Pathology, Faculty of Medical Sciences, National Council of Scientific and Technical Research, National University of Cuyo, 5502 Mendoza, Argentina
| | - PA Gargiulo
- Laboratory of Neurosciences and Experimental Psychology, Area of Pharmacology, Department of Pathology, Faculty of Medical Sciences, National Council of Scientific and Technical Research, National University of Cuyo, 5502 Mendoza, Argentina
| | - JV Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Neurodegenerative Diseases Group, BioCruces Health Research Institute, 48903 Barakaldo, Spain
| |
Collapse
|
15
|
Lynham AJ, Knott S, Underwood JFG, Hubbard L, Agha SS, Bisson JI, van den Bree MBM, Chawner SJRA, Craddock N, O'Donovan M, Jones IR, Kirov G, Langley K, Martin J, Rice F, Roberts NP, Thapar A, Anney R, Owen MJ, Hall J, Pardiñas AF, Walters JTR. DRAGON-Data: a platform and protocol for integrating genomic and phenotypic data across large psychiatric cohorts. BJPsych Open 2023; 9:e32. [PMID: 36752340 PMCID: PMC9970169 DOI: 10.1192/bjo.2022.636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/02/2022] [Accepted: 12/16/2022] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Current psychiatric diagnoses, although heritable, have not been clearly mapped onto distinct underlying pathogenic processes. The same symptoms often occur in multiple disorders, and a substantial proportion of both genetic and environmental risk factors are shared across disorders. However, the relationship between shared symptoms and shared genetic liability is still poorly understood. AIMS Well-characterised, cross-disorder samples are needed to investigate this matter, but few currently exist. Our aim is to develop procedures to purposely curate and aggregate genotypic and phenotypic data in psychiatric research. METHOD As part of the Cardiff MRC Mental Health Data Pathfinder initiative, we have curated and harmonised phenotypic and genetic information from 15 studies to create a new data repository, DRAGON-Data. To date, DRAGON-Data includes over 45 000 individuals: adults and children with neurodevelopmental or psychiatric diagnoses, affected probands within collected families and individuals who carry a known neurodevelopmental risk copy number variant. RESULTS We have processed the available phenotype information to derive core variables that can be reliably analysed across groups. In addition, all data-sets with genotype information have undergone rigorous quality control, imputation, copy number variant calling and polygenic score generation. CONCLUSIONS DRAGON-Data combines genetic and non-genetic information, and is available as a resource for research across traditional psychiatric diagnostic categories. Algorithms and pipelines used for data harmonisation are currently publicly available for the scientific community, and an appropriate data-sharing protocol will be developed as part of ongoing projects (DATAMIND) in partnership with Health Data Research UK.
Collapse
Affiliation(s)
- Amy J. Lynham
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Sarah Knott
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Jack F. G. Underwood
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Leon Hubbard
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Sharifah S. Agha
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Jonathan I. Bisson
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Marianne B. M. van den Bree
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Samuel J. R. A. Chawner
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Nicholas Craddock
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Michael O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Ian R. Jones
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - George Kirov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Kate Langley
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK; and School of Psychology, Cardiff University, UK
| | - Joanna Martin
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Frances Rice
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Neil P. Roberts
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK; and Directorate of Psychology and Psychological Therapies, Cardiff & Vale University Health Board, UK
| | - Anita Thapar
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Richard Anney
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Michael J. Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Jeremy Hall
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - Antonio F. Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - James T. R. Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| |
Collapse
|
16
|
Marguilho M, Figueiredo I, Castro-Rodrigues P. A unified model of ketamine's dissociative and psychedelic properties. J Psychopharmacol 2023; 37:14-32. [PMID: 36527355 PMCID: PMC9834329 DOI: 10.1177/02698811221140011] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ketamine is an N-methyl-d-aspartate antagonist which is increasingly being researched and used as a treatment for depression. In low doses, it can cause a transitory modification in consciousness which was classically labelled as 'dissociation'. However, ketamine is also commonly classified as an atypical psychedelic and it has been recently reported that ego dissolution experiences during ketamine administration are associated with greater antidepressant response. Neuroimaging studies have highlighted several similarities between the effects of ketamine and those of serotonergic psychedelics in the brain; however, no unified account has been proposed for ketamine's multi-level effects - from molecular to network and psychological levels. Here, we propose that the fast, albeit transient, antidepressant effects observed after ketamine infusions are mainly driven by its acute modulation of reward circuits and sub-acute increase in neuroplasticity, while its dissociative and psychedelic properties are driven by dose- and context-dependent disruption of large-scale functional networks. Computationally, as nodes of the salience network (SN) represent high-level priors about the body ('minimal' self) and nodes of the default-mode network (DMN) represent the highest-level priors about narrative self-experience ('biographical' self), we propose that transitory SN desegregation and disintegration accounts for ketamine's 'dissociative' state, while transitory DMN desegregation and disintegration accounts for ketamine's 'psychedelic' state. In psychedelic-assisted psychotherapy, a relaxation of the highest-level beliefs with psychotherapeutic support may allow a revision of pathological self-representation models, for which neuroplasticity plays a permissive role. Our account provides a multi-level rationale for using the psychedelic properties of ketamine to increase its long-term benefits.
Collapse
Affiliation(s)
| | | | - Pedro Castro-Rodrigues
- Centro Hospitalar Psiquiátrico de Lisboa, Lisbon, Portugal,NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal,Pedro Castro-Rodrigues, Centro Hospitalar Psiquiátrico de Lisboa, Avenida do Brasil, 53, Lisbon, 1749-002, Portugal.
| |
Collapse
|
17
|
Asch RH, Hillmer AT, Baldassarri SR, Esterlis I. The metabotropic glutamate receptor 5 as a biomarker for psychiatric disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:265-310. [PMID: 36868631 DOI: 10.1016/bs.irn.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of glutamate system in the etiology and pathophysiology of psychiatric disorders has gained considerable attention in the past two decades, including dysregulation of the metabotropic glutamatergic receptor subtype 5 (mGlu5). Thus, mGlu5 may represent a promising therapeutic target for psychiatric conditions, particularly stress-related disorders. Here, we describe mGlu5 findings in mood disorders, anxiety, and trauma disorders, as well as substance use (specifically nicotine, cannabis, and alcohol use). We highlight insights gained from positron emission tomography (PET) studies, where possible, and discuss findings from treatment trials, when available, to explore the role of mGlu5 in these psychiatric disorders. Through the research evidence reviewed in this chapter, we make the argument that, not only is dysregulation of mGlu5 evident in numerous psychiatric disorders, potentially functioning as a disease "biomarker," the normalization of glutamate neurotransmission via changes in mGlu5 expression and/or modulation of mGlu5 signaling may be a needed component in treating some psychiatric disorders or symptoms. Finally, we hope to demonstrate the utility of PET as an important tool for investigating mGlu5 in disease mechanisms and treatment response.
Collapse
Affiliation(s)
- Ruth H Asch
- Department of Psychiatry, Yale University, New Haven, CT, United States.
| | - Ansel T Hillmer
- Department of Psychiatry, Yale University, New Haven, CT, United States; Department of Radiology and Biomedical Imaging, New Haven, CT, United States
| | - Stephen R Baldassarri
- Yale Program in Addiction Medicine, Yale University, New Haven, CT, United States; Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Irina Esterlis
- Department of Psychiatry, Yale University, New Haven, CT, United States; Department of Psychology, Yale University, New Haven, CT, United States; Clinical Neurosciences Division, U.S. Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
18
|
Yen YT, Tsai YS, Su WL, Huang DY, Wu HH, Tseng SH, Wang HH, Chiu CY, Wang CF, Liu CY, Chyueh SC. New ketamine analogue: 2-fluorodeschloro-N-ethyl-ketamine and its suggested metabolites. Forensic Sci Int 2022; 341:111501. [DOI: 10.1016/j.forsciint.2022.111501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/24/2022]
|
19
|
Hadizadeh H, Flores JM, Mayerson T, Worhunsky PD, Potenza MN, Angarita GA. Glutamatergic Agents for the Treatment of Cocaine Use Disorder. Curr Behav Neurosci Rep 2022. [DOI: 10.1007/s40473-022-00252-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
20
|
Antidepressant Effect of Ketamine on Inflammation-Mediated Cytokine Dysregulation in Adults with Treatment-Resistant Depression: Rapid Systematic Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1061274. [PMID: 36160713 PMCID: PMC9507757 DOI: 10.1155/2022/1061274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/25/2022] [Accepted: 08/23/2022] [Indexed: 11/28/2022]
Abstract
Background Major depressive disorder (MDD) and treatment-resistant depression (TRD) represent a global source of societal and health burden. To advise proper management of inflammation-related depression among TRD patients, it is important to identify therapeutic clinical treatments. A key factor is related to proinflammatory cytokines such as interleukin- (IL-) 1β, IL-6, and tumor necrosis factor- (TNF-) α which have been implicated in the pathogenesis of depressive symptoms in MDD patients. Ketamine may provide an anti-inflammatory therapeutic strategy by targeting proinflammatory pathways associated with depressive disorders, which may be exacerbated in the ageing population with TRD. Objective Despite a burgeoning body of literature demonstrating that inflammation is linked to TRD, there is still a lack of comprehensive research on the relationship between proinflammatory biomarkers and ketamine's antidepressant effect on TRD patients. Method The Cochrane Library and PubMed/MEDLINE databases were systematically searched from inception up to February 1, 2022, adopting broad inclusion criteria to assess clinical topics related to the impact of ketamine on inflammatory cytokines in TRD patients. The present work is in compliance with the World Health Organization Rapid Review Guide. Results Five out of the seven studies examined in this review show that ketamine infusion may reduce depressive symptoms with a quick start of effect on TRD patients. Based on the Montgomery-Åsberg Depression Rating Scale (MADRS) and Hamilton Depression Rating Scale (HAM-D) scores, the overall response rate for ketamine was 56%; that is, 56% of those treated with ketamine had MADRS/HAM-D scores decreased by at least 50%. Conclusions While the anti-inflammatory effects of ketamine modulate specific proinflammatory cytokines, its rapid antidepressant effect on TRD patients remains inconsistent. However, our study findings can provide a reliable basis for future research on how to improve systemic inflammatory immune disorders and mental health. We suggest that ketamine infusion may be part of a comprehensive treatment approach in TRD patients with elevated levels of depression-specific inflammatory biomarkers.
Collapse
|
21
|
Burgdorf JS, Zhang XL, Stanton PK, Moskal JR, Donello JE. Zelquistinel Is an Orally Bioavailable Novel NMDA Receptor Allosteric Modulator That Exhibits Rapid and Sustained Antidepressant-Like Effects. Int J Neuropsychopharmacol 2022; 25:979-991. [PMID: 35882204 PMCID: PMC9743962 DOI: 10.1093/ijnp/pyac043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 06/22/2022] [Accepted: 07/25/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The role of glutamatergic receptors in major depressive disorder continues to be of great interest for therapeutic development. Recent studies suggest that both negative and positive modulation of N-methyl-D-aspartate receptors (NMDAR) can produce rapid antidepressant effects. Here we report that zelquistinel, a novel NMDAR allosteric modulator, exhibits high oral bioavailability and dose-proportional exposures in plasma and the central nervous system and produces rapid and sustained antidepressant-like effects in rodents by enhancing activity-dependent, long-term synaptic plasticity. METHODS NMDAR-mediated functional activity was measured in cultured rat brain cortical neurons (calcium imaging), hNR2A or B subtype-expressing HEK cells, and synaptic plasticity in rat hippocampal and medial prefrontal cortex slices in vitro. Pharmacokinetics were evaluated in rats following oral administration. Antidepressant-like effects were assessed in the rat forced swim test and the chronic social deficit mouse model. Target engagement and the safety/tolerability profile was assessed using phencyclidine-induced hyperlocomotion and rotarod rodent models. RESULTS Following a single oral dose, zelquistinel (0.1-100 µg/kg) produced rapid and sustained antidepressant-like effects in the rodent depression models. Brain/ cerebrospinal fluid concentrations associated with zelquistinel antidepressant-like activity also increased NMDAR function and rapidly and persistently enhanced activity-dependent synaptic plasticity (long-term potentiation), suggesting that zelquistinel produces antidepressant-like effects by enhancing NMDAR function and synaptic plasticity. Furthermore, Zelquistinel inhibited phencyclidine (an NMDAR antagonist)-induced hyperlocomotion and did not impact rotarod performance. CONCLUSIONS Zelquistinel produces rapid and sustained antidepressant effects by positively modulating the NMDARs, thereby enhancing long-term potentiation of synaptic transmission.
Collapse
Affiliation(s)
- Jeffrey S Burgdorf
- Correspondence: Jeffrey Burgdorf, PhD, 1801 Maple Ave, Suite 4300, Evanston, IL, 60201, USA ()
| | - Xiao-Lei Zhang
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| | - Patric K Stanton
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| | - Joseph R Moskal
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, McCormick School of Engineering and Applied Sciences, Northwestern University, Evanston, Illinois, USA
| | | |
Collapse
|
22
|
Drozdz SJ, Goel A, McGarr MW, Katz J, Ritvo P, Mattina GF, Bhat V, Diep C, Ladha KS. Ketamine Assisted Psychotherapy: A Systematic Narrative Review of the Literature. J Pain Res 2022; 15:1691-1706. [PMID: 35734507 PMCID: PMC9207256 DOI: 10.2147/jpr.s360733] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
Currently, ketamine is used in treating multiple pain, mental health, and substance abuse disorders due to rapid-acting analgesic and antidepressant effects. Its limited short-term durability has motivated research into the potential synergistic actions between ketamine and psychotherapy to sustain benefits. This systematic review on ketamine-assisted psychotherapy (KAP) summarizes existing evidence regarding present-day practices. Through rigorous review, seventeen articles that included 603 participants were identified. From available KAP publications, it is apparent that combined treatments can, in specific circumstances, initiate and prolong clinically significant reductions in pain, anxiety, and depressive symptoms, while encouraging rapport and treatment engagement, and promoting abstinence in patients addicted to other substances. Despite much variance in how KAP is applied (route of ketamine administration, ketamine dosage/frequency, psychotherapy modality, overall treatment length), these findings suggest psychotherapy, provided before, during, and following ketamine sessions, can maximize and prolong benefits. Additional large-scale randomized control trials are warranted to understand better the mutually influential relationships between psychotherapy and ketamine in optimizing responsiveness and sustaining long-term benefits in patients with chronic pain. Such investigations will assist in developing standardized practices and maintenance programs.
Collapse
Affiliation(s)
- Sandra J Drozdz
- Department of Anesthesia, St. Michael's Hospital, Toronto, ON, Canada
| | - Akash Goel
- Department of Anesthesia, St. Michael's Hospital, Toronto, ON, Canada.,Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University, Stanford, CA, USA.,Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
| | - Matthew W McGarr
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Joel Katz
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada.,Department of Psychology, York University, Toronto, ON, Canada.,Department of Anesthesia and Pain Management, University Health Network, Toronto, ON, Canada.,School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Paul Ritvo
- Department of Psychology, York University, Toronto, ON, Canada.,School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | | | - Venkat Bhat
- Interventional Psychiatry Program, St. Michael's Hospital, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Calvin Diep
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
| | - Karim S Ladha
- Department of Anesthesia, St. Michael's Hospital, Toronto, ON, Canada.,Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada.,Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| |
Collapse
|
23
|
Effects of RhoA on depression-like behavior in prenatally stressed offspring rats. Behav Brain Res 2022; 432:113973. [PMID: 35728732 DOI: 10.1016/j.bbr.2022.113973] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/08/2022] [Accepted: 06/15/2022] [Indexed: 11/20/2022]
Abstract
Depression is a common mental disease that can lead to suicide when severe. Exposure to prenatal stress (PS) can lead to depression-like behavior in offspring, but the mechanism is unclear. RhoA (Ras homology family member A) plays an important role in stress-induced changes in synaptic plasticity, participating in the development of depression by activating the downstream effector ROCK (Rho-associated protein kinase). This study explored the influence in the expression of RhoA and downstream molecules ROCK1/2 in prenatally stressed rats, and the effect of RhoA inhibitor simvastatin on depression-like behavior induced by PS. Depression-like behavior in offspring was detected by sucrose preference test, forced swimming test, and open-field test. The mRNA and protein expression of RhoA and ROCK1/2 in the hippocampus and prefrontal cortex of offspring rats were detected by qRT-PCR and western blotting, respectively. Our results showed that PS causes depression-like behavior in offspring rats, associated with elevated expression of RhoA, ROCK1/2 in the hippocampus and prefrontal cortex. After administration of simvastatin to PS rats, the expression of RhoA and ROCK2 was significantly reduced, alleviating depression-like behavior. Our study demonstrated that RhoA participates in the depression-like behavior in prenatally stressed offspring rats, which may be a potential target for antidepressant therapy.
Collapse
|
24
|
Ren Q, Hua L, Zhou X, Cheng Y, Lu M, Zhang C, Guo J, Xu H. Effects of a Single Sub-Anesthetic Dose of Ketamine on Postoperative Emotional Responses and Inflammatory Factors in Colorectal Cancer Patients. Front Pharmacol 2022; 13:818822. [PMID: 35479322 PMCID: PMC9037238 DOI: 10.3389/fphar.2022.818822] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: To investigate the effect of a single sub-anesthetic dose of ketamine on postoperative anxiety, depression, and inflammatory factors in patients with colorectal cancer. Methods: A total of 104 patients undergoing selective colorectal surgery in our hospital from Jan 2015 to Oct 2017 were included and randomly assigned (1:1:1:1) into a 0.1 mg kg-1 ketamine group (K1 group), 0.2 mg kg-1 ketamine group (K2 group), 0.3 mg kg-1 ketamine group (K3 group), or control group (C group). Corresponding doses of ketamine were given intravenously in the K groups (K1, K2, and K3 groups) 5 min before operation, and the same amount of normal saline was given in the C group. The intravenous analgesia program was identical in the four groups. The patients' emotional reactions (anxiety and depression) were assessed by the Hospital Anxiety and Depression Scale (HAD), the quality of postoperative recovery was evaluated by the Quality of Recovery-40 (QoR-40) questionnaire, and the levels of IL-6, IL-8, and TNF-α in peripheral blood were detected by enzyme-linked immunosorbent assay (ELISA) on the day before operation and within 24, 48, and 72 h post-operation respectively. Pain was estimated by the visual analog scale (VAS), and sedation was assessed with Ramsay score 30 min after extubation. The time points of anesthetic end and extubation were recorded. The complications during anesthesia and recovery such as cough and agitation 30 min after extubation were recorded. Results: The anxiety score (HAD-A) and depression score (HAD-D) of the K3 group were significantly lower than those of the C group post-operation (p < 0.05). The QoR-40 score of the K3 group was significantly higher than that of the C group (p < 0.05). The serum levels of IL-6, IL-8, and TNF-α in the K3 group were significantly lower than those in the C group (p < 0.05 and p < 0.01). There were no significant differences in HAD-A, HAD-D, and QoR-40 scores or serum levels of IL-6, IL-8, and TNF-α between the K1 and K2 groups and the C group. There were no significant differences in VAS pain score or Ramsay sedation score among the four groups 30 min after extubation. There were no significant differences in extubation time, postoperative cough, emergence agitation, or delirium among the four groups. Dizziness, nausea, vomiting, diplopia, or other adverse reactions were not found 30 min after extubation. Conclusion: A single sub-anesthetic dose (0.3 mg kg-1) of ketamine can significantly improve the postoperative anxiety and depression of colorectal cancer patients and reduce the levels of IL-6, IL-8, and TNF-α.
Collapse
Affiliation(s)
- Qin Ren
- Department of Anesthesiology, Shanghai Pudong New Area Gongli Hospital, Navy Military Medical University, Shanghai, China
| | - Ling Hua
- Department of Laboratory Medicine, Shanghai Pudong New Area Gongli Hospital, Navy Military Medical University, Shanghai, China
| | - Xiaofang Zhou
- Department of Anesthesiology, Shanghai Pudong New Area Gongli Hospital, Navy Military Medical University, Shanghai, China
| | - Yong Cheng
- Department of Anesthesiology, Shanghai Pudong New Area Gongli Hospital, Navy Military Medical University, Shanghai, China
| | - Mingjun Lu
- Department of Anesthesiology, Shanghai Pudong New Area Gongli Hospital, Navy Military Medical University, Shanghai, China
| | - Chuanqing Zhang
- Department of Anesthesiology, Shanghai Pudong New Area Gongli Hospital, Navy Military Medical University, Shanghai, China
| | - Jianrong Guo
- Department of Anesthesiology, Shanghai Pudong New Area Gongli Hospital, Navy Military Medical University, Shanghai, China
| | - Hua Xu
- Department of Anesthesiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
25
|
Yen YT, Tseng SH, Huang DY, Tsai YS, Lee LW, Chen PL, Liu YL, Chyueh SC. Identification of a novel norketamine precursor from seized powders: 2-(2-chlorophenyl)-2-nitrocyclohexanone. Forensic Sci Int 2022; 333:111241. [DOI: 10.1016/j.forsciint.2022.111241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 11/04/2022]
|
26
|
Subramanian S, Haroutounian S, Palanca BJA, Lenze EJ. Ketamine as a therapeutic agent for depression and pain: mechanisms and evidence. J Neurol Sci 2022; 434:120152. [PMID: 35092901 DOI: 10.1016/j.jns.2022.120152] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/14/2022]
Abstract
Ketamine is an anesthetic drug which is now used to treat chronic pain conditions and psychiatric disorders, especially depression. It is an N-methyl-D-aspartate (NMDA) receptor antagonist with additional effects on α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, opioid receptors, and monoaminergic receptors. This article focuses on ketamine's role in treating depression and pain, two commonly comorbid challenging conditions with potentially shared neurobiologic circuitry. Many clinical trials have utilized intravenous or intranasal ketamine for treating depression and pain. Intravenous ketamine is more bioavailable than intranasal ketamine and both are effective for acute depressive episodes. Intravenous ketamine is advantageous for post-operative analgesia and is associated with a reduction in total opioid requirements. Few studies have treated chronic pain or concurrent depression and pain with ketamine. Larger, randomized control trials are needed to examine the safety and efficacy of intravenous vs. intranasal ketamine, ideal target populations, and optimal dosing to treat both depression and pain.
Collapse
Affiliation(s)
- Subha Subramanian
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| | - Simon Haroutounian
- Department of Anesthesiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ben Julian A Palanca
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Eric J Lenze
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
27
|
Li W, Zhou Y, Liu W, Wang C, Lan X, Zhang Z, Zhang F, Ye Y, Liu H, Wu K, McIntyre RS, Ning Y. Long-term outcomes of repeated ketamine infusions in patients with unipolar and bipolar depression: A naturalistic follow-up study. J Affect Disord 2022; 300:172-178. [PMID: 34952122 DOI: 10.1016/j.jad.2021.12.084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/14/2021] [Accepted: 12/19/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Ketamine was proven to have short-term antidepressant effects. There is a paucity of studies focused on the long-term outcomes of repeated infusions of ketamine. This study aimed to examine the long-term outcomes of repeated ketamine infusions in patients with unipolar and bipolar depression METHODS: One hundred and eight patients with unipolar and bipolar depression completed the repeated treatment phase (administered ketamine three times weekly over a 12-day) and entered a 9-month naturalistic follow-up phase. Assessments were obtained at week 2, month 6, and month 9 after the repeated treatment phase. The Patient Health Questionnaire-9 (PHQ-9) Scale and the Global Assessment of Functioning (GAF) Scale were used to assess depressive symptoms and global functional status, respectively. RESULTS Seventy-one (65.7%) of patients completed the 9-month follow-up. On month 9, the response and remission rate were 80.3% and 78.9%, respectively. Among 56 patients who achieved response after the repeated treatment phase, 26 (46.4%) of patients sustained response during 9-month follow-up and their GAF score remained over 70. Sixteen patients relapsed during the 9-month follow-up and 14 (85.7%) of the relapse occurred during the first 2-week follow-up. LIMITATION The major limitation of this study is the open-label design. CONCLUSIONS This small sample study suggested that patients with unipolar and bipolar depression who response to repeated treatment with continued oral antidepressant may be a viable treatment option, and their global functional status improved with a follow-up. Relapse of depression tended to occur during the 2 weeks follow-up.
Collapse
Affiliation(s)
- Weicheng Li
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Metal Disorders, Guangzhou, China
| | - Yanling Zhou
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Metal Disorders, Guangzhou, China
| | - Weijian Liu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Metal Disorders, Guangzhou, China; Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing, China
| | - Chengyu Wang
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Metal Disorders, Guangzhou, China
| | - Xiaofeng Lan
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Metal Disorders, Guangzhou, China
| | - Zhipei Zhang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Metal Disorders, Guangzhou, China
| | - Fan Zhang
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Metal Disorders, Guangzhou, China
| | - Yanxiang Ye
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Metal Disorders, Guangzhou, China
| | - Haiyan Liu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Metal Disorders, Guangzhou, China
| | - Kai Wu
- Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, China
| | - Roger S McIntyre
- Canadian Rapid Treatment Center of Excellence, Mississauga, ON, Canada; Mood Disorders Psychopharmacology Unit, Poul Hansen Depression Centre, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| | - Yuping Ning
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Metal Disorders, Guangzhou, China.
| |
Collapse
|
28
|
Oluboka OJ, Katzman MA, Habert J, Khullar A, Oakander MA, McIntosh D, McIntyre RS, Soares CN, Lam RW, Klassen LJ, Tanguay R. Early Optimized Pharmacological Treatment in Patients With Depression and Chronic Pain. CNS Spectr 2022; 28:1-40. [PMID: 35195060 DOI: 10.1017/s1092852922000128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractMajor depressive disorder (MDD) is the leading cause of disability worldwide. Patients with MDD have high rates of comorbidity with mental and physical conditions, one of which is chronic pain. Chronic pain conditions themselves are also associated with significant disability, and the large number of patients with MDD who have chronic pain drives high levels of disability and compounds healthcare burden. The management of depression in patients who also have chronic pain can be particularly challenging due to underlying mechanisms that are common to both conditions, and because many patients with these conditions are already taking multiple medications. For these reasons, healthcare providers may be reluctant to treat such patients. The Canadian Network for Mood and Anxiety Treatments (CANMAT) guidelines provide evidence-based recommendations for the management of MDD and comorbid psychiatric and medical conditions such as anxiety, substance use disorder, and cardiovascular disease; however, comorbid chronic pain is not addressed. In this article, we provide an overview of the pathophysiological and clinical overlap between depression and chronic pain and review evidence-based pharmacological recommendations in current treatment guidelines for MDD and for chronic pain. Based on clinical experience with MDD patients with comorbid pain, we recommend rapidly and aggressively treating depression according to CANMAT treatment guidelines, using antidepressant medications with analgesic properties, while addressing pain with first-line pharmacotherapy as treatment for depression is optimized. We review options for treating pain symptoms that remain after response to antidepressant treatment is achieved.
Collapse
|
29
|
Raggi A, Lanza G, Ferri R. Auditory mismatch negativity in bipolar disorder: a focused review. Rev Neurosci 2022; 33:17-30. [PMID: 33837681 DOI: 10.1515/revneuro-2021-0010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023]
Abstract
The auditory mismatch negativity, a component of the event-related potential elicited by an unexpected stimulus in a sequence of acoustic stimuli, provides an objective measure of the accuracy of the echoic information processing of the human brain in vivo. Auditory mismatch negativity is also a useful probe of cortical glutamatergic N-methyl-d-aspartate receptor activity and disturbance. Notably, auditory mismatch negativity is consistently impaired in schizophrenia. Because of the wide spectrum extending from bipolar affective illness and schizoaffective psychosis to typical schizophrenia, we examined the literature on auditory mismatch negativity in bipolar disorder with the aim to find any neurophysiological dysfunction concerning pre-attentive information processing shared by these clinical conditions. This focused review includes 26 original articles published in peer-reviewed journals and indexed in the National Institutes of Health National Library of Medicine (PubMed) search system. Overall, evidence is consistent with the finding that auditory mismatch negativity is impaired in bipolar disorder with psychotic features, even though to a lesser extent than in schizophrenia. It must be acknowledged that, in a few twin and family studies, mismatch negativity abnormalities were not specifically associated with bipolar disorder. In conclusion, auditory mismatch negativity research supports the involvement of the N-methyl-d-aspartate system in the pathophysiology of bipolar disorder, as previously assessed for schizophrenia, thus creating an intriguing trait d'union between these two mental illnesses and stimulating the development of novel therapeutic agents. With additional replication and validation, auditory mismatch negativity may be further considered as a correlate of a common psychopathology of schizophrenia and bipolar spectrum illnesses.
Collapse
Affiliation(s)
- Alberto Raggi
- Unit of Neurology, G.B. Morgagni - L. Pierantoni Hospital, Via Carlo Forlanini 34, 47121 Forlì, Italy
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
- Department of Neurology IC, Oasi Research Institute-IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy
| | - Raffaele Ferri
- Department of Neurology IC, Oasi Research Institute-IRCCS, Via Conte Ruggero 73, 94018 Troina, Italy
| |
Collapse
|
30
|
Elkrief L, Payette O, Foucault JN, Longpré-Poirier C, Richard M, Desbeaumes Jodoin V, Lespérance P, Miron JP. Transcranial magnetic stimulation and intravenous ketamine combination therapy for treatment-resistant bipolar depression: A case report. Front Psychiatry 2022; 13:986378. [PMID: 36213934 PMCID: PMC9532540 DOI: 10.3389/fpsyt.2022.986378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022] Open
Abstract
About a third of patients suffering from major depression develop treatment-resistant depression (TRD). Although repetitive transcranial magnetic stimulation (rTMS) and intravenous ketamine have proven effective for the management of TRD, many patients remain refractory to treatment. We present the case of a patient suffering from bipolar TRD. The patient was referred to us after failure to respond to first-and second-line pharmacotherapy and psychotherapy. After minimal response to both rTMS and ketamine alone, we attempted a combination rTMS and ketamine protocol, which led to complete and sustained remission. Various comparable and complimentary mechanisms of antidepressant action of ketamine and rTMS are discussed, which support further study of this combination therapy. Future research should focus on the feasibility, tolerability, and efficacy of this novel approach.
Collapse
Affiliation(s)
- Laurent Elkrief
- Centre Hospitalier de l'Université de Montréal et Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada.,Département de Psychiatrie et d'Addictologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Olivier Payette
- Centre Hospitalier de l'Université de Montréal et Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada
| | - Jean-Nicolas Foucault
- Centre Hospitalier de l'Université de Montréal et Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada
| | - Christophe Longpré-Poirier
- Centre Hospitalier de l'Université de Montréal et Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada.,Département de Psychiatrie et d'Addictologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Maxime Richard
- Centre Hospitalier de l'Université de Montréal et Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada.,Département de Psychiatrie et d'Addictologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Véronique Desbeaumes Jodoin
- Centre Hospitalier de l'Université de Montréal et Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada
| | - Paul Lespérance
- Centre Hospitalier de l'Université de Montréal et Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada.,Département de Psychiatrie et d'Addictologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Jean-Philippe Miron
- Centre Hospitalier de l'Université de Montréal et Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada.,Département de Psychiatrie et d'Addictologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
31
|
Su S, Kay G, Hochadel T, Rojo J, Christopher Stein J, Boinpally R, Periclou A. A randomized, multicenter trial assessing the effects of rapastinel compared to ketamine, alprazolam, and placebo on simulated driving performance. Clin Transl Sci 2022; 15:255-266. [PMID: 34423904 PMCID: PMC8742651 DOI: 10.1111/cts.13145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/20/2021] [Accepted: 08/03/2021] [Indexed: 11/29/2022] Open
Abstract
N-methyl-D-aspartate ionotropic glutamatergic receptor (NMDAR) modulators, including rapastinel and ketamine, elicit rapid and sustained antidepressant responses in patients with treatment-resistant major depressive disorder. This phase I, randomized, multicenter, placebo-controlled, five-period, crossover, single-dose study evaluated simulated driving performance of healthy participants (N = 107) after single doses of rapastinel slow intravenous (i.v.) bolus 900 and 1800 mg, alprazolam oral 0.75 mg (positive control), ketamine i.v. infusion 0.5 mg/kg (clinical comparator), and placebo ~ 45 min before driving. The primary end point was SD of lateral position (SDLP) during the 60-min 100-km simulated driving scenario. Additional measures of driving performance, sleepiness, and cognition were also evaluated. To assess effects over time, mean SDLP was calculated for each 10-min interval of driving. Sensitivity of the assays was confirmed with alprazolam (all placebo comparisons p < 0.02). Rapastinel 900 and 1800 mg did not significantly affect simulated driving performance compared to placebo (both p > 0.5). Both rapastinel doses resulted in significantly less impaired driving compared to alprazolam or ketamine (all p < 0.002); ketamine significantly impaired driving compared to placebo (p = 0.0001). Results for the additional measures were similar to the primary end point. No new safety signals were observed for any study interventions. This first study of rapastinel effects on simulated driving found that rapastinel 900 and 1800 mg did not impair driving performance, but ketamine 0.5 mg/kg resulted in significantly impaired driving performance. Ketamine's effects on driving were maintained for at least 105 min, indicating that clinicians should be vigilant to prevent or postpone driving in patients after ketamine treatment.
Collapse
Affiliation(s)
- Shengfang Su
- Allergan plc (now AbbVie, Inc.)MadisonNew JerseyUSA
| | - Gary Kay
- Cognitive Research CorporationSt. PetersburgFloridaUSA
| | | | | | | | | | | |
Collapse
|
32
|
[Ketamine and other N-methyl-D-aspartate receptor modulators in treatment of depression]. DER NERVENARZT 2021; 93:234-242. [PMID: 34735585 DOI: 10.1007/s00115-021-01221-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 12/23/2022]
Abstract
Since the first successful ketamine application in treatment-resistant depressive patients, the newly developed pharmaceutical class of rapid-acting antidepressants has been intensively investigated. The underlying mechanism of action by influencing the glutamatergic neurotransmission via a modulation of N‑methyl-D-aspartate (NMDA) receptors, represents a completely new and promising interventional strategy in the treatment of affective disorders. In this very dynamic field, Spravato® (esketamine) is so far the only approved drug; however, many other substances are currently in the development and evaluation processes. This narrative review provides a critical overview of the most important substances, target structures and developmental stages of NMDAR modulators.
Collapse
|
33
|
Loureiro JRA, Sahib AK, Vasavada M, Leaver A, Kubicki A, Wade B, Joshi S, Hellemann G, Congdon E, Woods RP, Espinoza R, Narr KL. Ketamine's modulation of cerebro-cerebellar circuitry during response inhibition in major depression. Neuroimage Clin 2021; 32:102792. [PMID: 34571429 PMCID: PMC8476854 DOI: 10.1016/j.nicl.2021.102792] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 12/01/2022]
Abstract
Ketamine modulates cerebellar connectivity during response inhibition in depression. Cerebellar–frontoparietal/sensory connectivity decreases in ketamine remitters. Cerebellar-frontoparietal/salience connectivity predicts treatment outcome. Cerebro-cerebellar loops serve as treatment biomarkers in major depression.
Patients with major depressive disorder (MDD) exhibit impaired control of cognitive and emotional systems, including deficient response selection and inhibition. Though these deficits are typically attributed to abnormal communication between macro-scale cortical networks, altered communication with the cerebellum also plays an important role. Yet, how the circuitry between the cerebellum and large-scale functional networks impact treatment outcome in MDD is not understood. We thus examined how ketamine, which elicits rapid therapeutic effects in MDD, modulates cerebro-cerebellar circuitry during response-inhibition using a functional imaging NoGo/Go task in MDD patients (N = 46, mean age: 39.2, 38.1% female) receiving four ketamine infusions, and healthy controls (N = 32, mean age:35.2, 71.4% female). We fitted psychophysiological-interaction (PPI) models for a functionally-derived cerebellar-seed and extracted average PPI in three target functional networks, frontoparietal (FPN), sensory-motor (SMN) and salience (SN) networks. Time and remission status were then evaluated for each of the networks and their network-nodes. Follow-up tests examined whether PPI-connectivity differed between patient remitter/non-remitters and controls. Results showed significant decreases in PPI-connectivity after ketamine between the cerebellum and FPN (p < 0.001) and SMN networks (p = 0.008) in remitters only (N = 20). However, ketamine-related changes in PPI-connectivity between the cerebellum and the SN (p = 0.003) did not vary with remitter status. Cerebellar-FPN, -SN PPI values at baseline were also associated with treatment outcome. Using novel methodology to quantify the functional coupling of cerebro-cerebellar circuitry during response-inhibition, our findings highlight that these loops play distinct roles in treatment response and could potentially serve as novel biomarkers for fast-acting antidepressant therapies in MDD.
Collapse
Affiliation(s)
- Joana R A Loureiro
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA.
| | - Ashish K Sahib
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Megha Vasavada
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Antoni Kubicki
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Benjamin Wade
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Shantanu Joshi
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Gerhard Hellemann
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Eliza Congdon
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Roger P Woods
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Randall Espinoza
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Katherine L Narr
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
34
|
Watanabe M, Marcy B, Hiroki A, Watase H, Kinoshita K, Iijima M, Marumo T, Zarate CA, Chaki S. Evaluation of the Safety, Tolerability, and Pharmacokinetic Profiles of TP0473292 (TS-161), A Prodrug of a Novel Orthosteric mGlu2/3 Receptor Antagonist TP0178894, in Healthy Subjects and Its Antidepressant-Like Effects in Rodents. Int J Neuropsychopharmacol 2021; 25:106-117. [PMID: 34534292 PMCID: PMC8832229 DOI: 10.1093/ijnp/pyab062] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND TP0473292 (the active ingredient of TS-161) is a prodrug of a novel metabotropic glutamate (mGlu) 2/3 receptor antagonist being developed for the treatment of patients with depression. This study evaluated the safety, tolerability, and pharmacokinetics of orally administered TS-161 in healthy subjects. METHODS This was a first-in-human, phase 1, randomized, double-blind, placebo-controlled, single-ascending dose (15-400 mg TS-161) and 10-day multiple-ascending dose (50-150 mg TS-161) study in healthy subjects, conducted from June 2019 through February 2020. Plasma and urine concentrations of the prodrug and its metabolites, and cerebrospinal fluid (CSF) concentrations of the active metabolite TP0178894 were measured to evaluate the pharmacokinetic profiles after oral administration of TS-161. RESULTS Following single and multiple doses, TP0473292 was extensively converted into its active metabolite TP0178894. Plasma concentrations of TP0178894 reached peak (Cmax) within 5 hours post dose and declined with a t1/2 <13 hours. Plasma exposures of TP0178894 increased with increasing dose. TP0178894 penetrated into CSF and reached a Cmax of 9.892 ng/mL at a single dose of 100 mg, which was comparable with IC50 values of antagonist activity at mGlu2/3 receptors. The most frequently observed adverse events that showed exposure-related incidence during the study were nausea, vomiting, and dizziness. CONCLUSIONS The mGlu2/3 receptor antagonist prodrug TP0473292 is safe and well-tolerated, is orally bioavailable in humans with extensive conversion into the active metabolite TP0178894 with sufficient CSF penetration to exert the anticipated pharmacological effects, and is a promising candidate for further clinical development in treatment of patients with depression.
Collapse
Affiliation(s)
- Mai Watanabe
- Taisho Pharmaceutical R&D Inc., Morristown, New Jersey, USA,Correspondence: Mai Watanabe, MS, Taisho Pharmaceutical R&D Inc., 350 Mt. Kemble Avenue, Morristown, NJ 07960, USA ()
| | - Brian Marcy
- Taisho Pharmaceutical R&D Inc., Morristown, New Jersey, USA
| | | | | | | | | | | | - Carlos A Zarate
- National Institute of Mental Health, National Institute of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
35
|
NMDARs Containing NR2B Subunit Do Not Contribute to the LTP Form of Hippocampal Plasticity: In Vivo Pharmacological Evidence in Rats. Int J Mol Sci 2021; 22:ijms22168672. [PMID: 34445376 PMCID: PMC8395520 DOI: 10.3390/ijms22168672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 11/17/2022] Open
Abstract
Synaptic plasticity is the key to synaptic health, and aberrant synaptic plasticity, which in turn impairs the functioning of large-scale brain networks, has been associated with neurodegenerative and psychiatric disorders. The best known and most studied form of activity-dependent synaptic plasticity remains long-term potentiation (LTP), which is controlled by glutamatergic N-methyl-d-aspartate) receptors (NMDAR) and considered to be a mechanism crucial for cellular learning and memory. Over the past two decades, discrepancies have arisen in the literature regarding the contribution of NMDAR subunit assemblies in the direction of NMDAR-dependent synaptic plasticity. Here, the nonspecific NMDAR antagonist ketamine (5 and 10 mg/kg), and the selective NR2B antagonists CP-101606 and Ro 25-6981 (6 and 10 mg/kg), were administered intraperitoneally in Sprague Dawley rats to disentangle the contribution of NR2B subunit in the LTP induced at the Schaffer Collateral-CA1 synapse using the theta burst stimulation protocol (TBS). Ketamine reduced, while CP-101606 and Ro 25-6981 did not alter the LTP response. The administration of CP-101606 before TBS did not influence the effects of ketamine when administered half an hour after tetanization, suggesting a limited contribution of the NR2B subunit in the action of ketamine. This work confirms the role of NMDAR in the LTP form of synaptic plasticity, whereas specific blockade of the NR2B subunit was not sufficient to modify hippocampal LTP. Pharmacokinetics at the doses used may have contributed to the lack of effects with specific antagonists. The findings refute the role of the NR2B subunit in the plasticity mechanism of ketamine in the model.
Collapse
|
36
|
Zhang F, Hillhouse TM, Anderson PM, Koppenhaver PO, Kegen TN, Manicka SG, Lane JT, Pottanat E, Van Fossen M, Rice R, Porter JH. Opioid receptor system contributes to the acute and sustained antidepressant-like effects, but not the hyperactivity motor effects of ketamine in mice. Pharmacol Biochem Behav 2021; 208:173228. [PMID: 34224734 DOI: 10.1016/j.pbb.2021.173228] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 11/24/2022]
Abstract
In 2000, a subanesthetic dose (0.5 mg/kg i.v.) of the dissociative anesthetic ketamine was reported to have both rapid and robust antidepressant effects in patients diagnosed with major depressive disorder and later, ketamine also was shown to be effective in treatment-resistant depressed patients. However, the mechanisms responsible for ketamine's antidepressant effects remain unclear. In 2018, a clinical study reported that pretreatment with the nonselective opioid antagonist naltrexone attenuated the rapid antidepressant effect of ketamine in depressed patients. The current study investigated the potential role of the opioid receptor system in the acute and sustained antidepressant-like and hyperactive effects of ketamine. Mice were tested in the tail suspension test (TST) and differential-reinforcement-of-low-rate responding (DRL) 72 s task which are behavioral screens for antidepressant-like properties. Additionally, open field locomotor activity also was measured. In all behavioral assays, mice were pretreated with the nonselective opioid receptor antagonist naltrexone or saline prior to ketamine administration. The current study found that ketamine (10 mg/kg) produced acute (30 min) and sustained (24 h) antidepressant-like effects in TST, which were attenuated by pretreatment of 2 mg/kg naltrexone. Ketamine (32 mg/kg) also produced an acute antidepressant-like effect in the DRL 72 s task that was attenuated by pretreatment of 2 mg/kg naltrexone. Finally, ketamine (10 and 32 mg/kg) produced hyperactivity in the open field; however, pretreatment with 2 mg/kg naltrexone failed to block the hyperactivity effects ketamine. These results, along with recent clinical findings, suggest that ketamine's antidepressant effects, but not its hyperactive effects, involve activation of the opioid system.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Todd M Hillhouse
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, WI, USA
| | - Paige M Anderson
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, WI, USA
| | | | - Taylor N Kegen
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, WI, USA
| | - Sofia G Manicka
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Jackson T Lane
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Elizabeth Pottanat
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Madeline Van Fossen
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Remington Rice
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| | - Joseph H Porter
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
37
|
Angarita GA, Hadizadeh H, Cerdena I, Potenza MN. Can pharmacotherapy improve treatment outcomes in people with co-occurring major depressive and cocaine use disorders? Expert Opin Pharmacother 2021; 22:1669-1683. [PMID: 34042556 DOI: 10.1080/14656566.2021.1931684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Major depressive disorder (MDD) and cocaine use disorder (CUD) are prevalent and frequently co-occur. When co-occurring, the presence of one disorder typically negatively impacts the prognosis for the other. Given the clinical relevance, we sought to examine pharmacotherapies for co-occurring CUD and MDD. While multiple treatment options have been examined in the treatment of each condition individually, studies exploring pharmacological options for their comorbidity are fewer and not conclusive.Areas Covered: For this review, the authors searched the literature in PubMed using clinical query options for therapies and keywords relating to each condition. Then, they described potentially promising pharmacologic therapeutic options based on shared mechanisms between the two conditions and/or results from individual clinical trials conducted to date.Expert opinion: Medications like stimulants, dopamine (D3) receptors partial agonists or antagonists, antagonists of kappa opioid receptors, topiramate, and ketamine could be promising as there is significant overlap relating to reward deficiency models, antireward pathways, and altered glutamatergic systems. However, the available clinical literature on any one of these types of agents is mixed. Additionally, for some agents there is possible concern related to abuse potential (e.g. ketamine and stimulants).
Collapse
Affiliation(s)
- Gustavo A Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.,Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA
| | - Hasti Hadizadeh
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.,Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, USA
| | - Ignacio Cerdena
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.,Connecticut Mental Health Center, New Haven, CT, USA
| | - Marc N Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.,Connecticut Mental Health Center, New Haven, CT, USA.,Child Study Center, Yale University School of Medicine, New Haven, CT, USA.,Department of Neuroscience, Yale University, New Haven, CT, USA.,Connecticut Council on Problem Gambling, Wethersfield, CT, USA
| |
Collapse
|
38
|
An D, Wei C, Wang J, Wu A. Intranasal Ketamine for Depression in Adults: A Systematic Review and Meta-Analysis of Randomized, Double-Blind, Placebo-Controlled Trials. Front Psychol 2021; 12:648691. [PMID: 34140915 PMCID: PMC8204747 DOI: 10.3389/fpsyg.2021.648691] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 04/08/2021] [Indexed: 12/30/2022] Open
Abstract
Background There is growing interest in glutamatergic agents as a treatment for depression, especially intranasal ketamine, which has become a hot topic in recent years. We aim to assess the efficacy and safety of intranasal ketamine in the treatment of major depressive disorder (MDD), especially treatment-resistant depression (TRD). Methods We searched Medline, EMBASE, and the Cochrane Library until April 1, 2020 to identify double-blind, randomized controlled trials with allocation concealment evaluating intranasal ketamine in major depressive episodes. Clinical remission, response, and depressive symptoms were extracted by two independent raters. The outcome measures were Montgomery–Asberg Depression Rating Scale (MADRS) score improved from baseline, clinical response and remission, dissociative symptoms, and common adverse events. The analyses employed a random-effects model. Results Data were synthesized from five randomized controlled trials (RCTs) employing an intranasal esketamine and one RCT employing intranasal ketamine, representing 840 subjects in parallel arms, and 18 subjects in cross-over designs (n = 858 with MDD, n = 792 with TRD). The weighted mean difference of MADRS score was observed to decrease by 6.16 (95% CI 4.44–7.88) in 2–4 h, 9.96 (95% CI 8.97–10.95) in 24 h, and 4.09 (95% CI 2.18–6.00) in 28 day. The pooled relative risk (RR) was 3.55 (95% CI 1.5–8.38, z = 2.89, and p < 0.001) for clinical remission and 3.22 (95% CI 1.85–5.61, z = 4.14, and p < 0.001) for clinical response at 24 h, while the pooled RR was 1.7 (95% CI 1.28–2.24, z = 3.72, and p < 0.001) for clinical remission and 1.48 (95% CI 1.17–1.86, z = 3.28, and p < 0.001) for clinical response at 28 day. Intranasal ketamine was associated with the occurrence of transient dissociative symptoms and common adverse events, but no persistent psychoses or affective switches. Conclusion Our meta-analysis suggests that repeated intranasal ketamine conducted a fast-onset antidepression effect in unipolar depression, while the mild and transient adverse effects were acceptable. Systematic Review Registration PROSPERO, CRD42020196856.
Collapse
Affiliation(s)
- Dongjiao An
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jing Wang
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
39
|
A key requirement for synaptic Reelin signaling in ketamine-mediated behavioral and synaptic action. Proc Natl Acad Sci U S A 2021; 118:2103079118. [PMID: 33975959 PMCID: PMC8157952 DOI: 10.1073/pnas.2103079118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ketamine is a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist that produces rapid antidepressant action in some patients with treatment-resistant depression. However, recent data suggest that ∼50% of patients with treatment-resistant depression do not respond to ketamine. The factors that contribute to the nonresponsiveness to ketamine's antidepressant action remain unclear. Recent studies have reported a role for secreted glycoprotein Reelin in regulating pre- and postsynaptic function, which suggests that Reelin may be involved in ketamine's antidepressant action, although the premise has not been tested. Here, we investigated whether the disruption of Reelin-mediated synaptic signaling alters ketamine-triggered synaptic plasticity and behavioral effects. To this end, we used mouse models with genetic deletion of Reelin or apolipoprotein E receptor 2 (Apoer2), as well as pharmacological inhibition of their downstream effectors, Src family kinases (SFKs) or phosphoinositide 3-kinase. We found that disruption of Reelin, Apoer2, or SFKs blocks ketamine-driven behavioral changes and synaptic plasticity in the hippocampal CA1 region. Although ketamine administration did not affect tyrosine phosphorylation of DAB1, an adaptor protein linked to downstream signaling of Reelin, disruption of Apoer2 or SFKs impaired baseline NMDA receptor-mediated neurotransmission. These results suggest that maintenance of baseline NMDA receptor function by Reelin signaling may be a key permissive factor required for ketamine's antidepressant effects. Taken together, our results suggest that impairments in Reelin-Apoer2-SFK pathway components may in part underlie nonresponsiveness to ketamine's antidepressant action.
Collapse
|
40
|
Freo U, Brugnatelli V, Turco F, Zanette G. Analgesic and Antidepressant Effects of the Clinical Glutamate Modulators Acetyl-L-Carnitine and Ketamine. Front Neurosci 2021; 15:584649. [PMID: 34045938 PMCID: PMC8144463 DOI: 10.3389/fnins.2021.584649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 03/04/2021] [Indexed: 12/26/2022] Open
Abstract
Pain and depression are leading causes of disability and of profound social and economic burden. Their impact is aggravated by their chronicity and comorbidity and the insufficient efficacy of current treatments. Morphological and functional metabolism studies link chronic pain and depressive disorders to dysfunctional neuroplastic changes in fronto-limbic brain regions that control emotional responses to painful injuries and stressful events. Glutamate modulators are emerging new therapies targeting dysfunctional brain areas implicated in the generation and maintenance of chronic pain and depression. Here, we report the effects of two clinically approved glutamate modulators: acetyl-L-carnitine (ALCAR) and S, R(±)ketamine (KET). ALCAR is a natural neurotrophic compound currently marketed for the treatment of neuropathies. KET is the prototypical non-competitive antagonist at N-methyl-D-aspartate glutamate receptors and a clinically approved anesthetic. Although they differ in pharmacological profiles, ALCAR and KET both modulate aminergic and glutamatergic neurotransmissions and pain and mood. We assessed in rats the effects of ALCAR and KET on cerebral metabolic rates for glucose (rCMRglc) and assessed clinically the effects of ALCAR in chronic pain and of KET in post-operative pain. ALCAR and KET increased rCMRglc at similar degrees in prefrontal, somatosensory, and cingulate cortices, and KET increased rCMRglc at a different, much larger, degree in limbic and dopaminergic areas. While rCMRglc increases in prefrontal cortical areas have been associated with analgesic and antidepressant effects of ALCAR and KET, the marked metabolic increases KET induces in limbic and dopaminergic areas have been related to its psychotomimetic and abuse properties. In patients with chronic neuropathic pain, ALCAR (1,000 mg/day) yielded to a fast (2 weeks) improvement of mood and then of pain and quality of life. In day-surgery patients, KET improved dischargeability and satisfaction. In obese patients undergoing bariatric surgery, a single, low dose of KET (0.5 mg/kg) at induction of anesthesia determined a very fast (hours) amelioration of post-operative depression and pain and an opioid-sparing effect. These findings indicate that ALCAR and KET, two non-selective glutamate modulators, still offer viable therapeutic options in comorbid pain and depression.
Collapse
Affiliation(s)
- Ulderico Freo
- Section of Anesthesiology and Intensive Care, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Viola Brugnatelli
- Section of Dentistry, Department of Neurosciences-DNS, University of Padua, Padua, Italy
| | - Fabio Turco
- Molecular Biology and Biochemistry Laboratory, Department of Neurogastroenterology, University of Naples Federico II, Naples, Italy
| | - Gastone Zanette
- Section of Dentistry, Department of Neurosciences-DNS, University of Padua, Padua, Italy
| |
Collapse
|
41
|
Abstract
The efficacy of standard antidepressants is limited for many patients with mood disorders such as major depressive disorder (MDD) and bipolar depression, underscoring the urgent need to develop novel therapeutics. Both clinical and preclinical studies have implicated glutamatergic system dysfunction in the pathophysiology of mood disorders. In particular, rapid reductions in depressive symptoms have been observed in response to subanesthetic doses of the glutamatergic modulator racemic (R,S)-ketamine in individuals with mood disorders. These results have prompted investigation into other glutamatergic modulators for depression, both as monotherapy and adjunctively. Several glutamate receptor-modulating agents have been tested in proof-of-concept studies for mood disorders. This manuscript gives a brief overview of the glutamate system and its relevance to rapid antidepressant response and discusses the existing clinical evidence for glutamate receptor-modulating agents, including (1) broad glutamatergic modulators ((R,S)-ketamine, esketamine, (R)-ketamine, (2R,6R)-hydroxynorketamine [HNK], dextromethorphan, Nuedexta [a combination of dextromethorphan and quinidine], deudextromethorphan [AVP-786], axsome [AXS-05], dextromethadone [REL-1017], nitrous oxide, AZD6765, CLE100, AGN-241751); (2) glycine site modulators (D-cycloserine [DCS], NRX-101, rapastinel [GLYX-13], apimostinel [NRX-1074], sarcosine, 4-chlorokynurenine [4-Cl-KYN/AV-101]); (3) subunit (NR2B)-specific N-methyl-D-aspartate (NMDA) receptor antagonists (eliprodil [EVT-101], traxoprodil [CP-101,606], rislenemdaz [MK-0657/CERC-301]); (4) metabotropic glutamate receptor (mGluR) modulators (basimglurant, AZD2066, RG1578, TS-161); and (5) mammalian target of rapamycin complex 1 (mTORC1) activators (NV-5138). Many of these agents are still in the preliminary stages of development. Furthermore, to date, most have demonstrated relatively modest effects compared with (R,S)-ketamine and esketamine, though some have shown more favorable characteristics. Of these novel agents, the most promising, and the ones for which the most evidence exists, appear to be those targeting ionotropic glutamate receptors.
Collapse
|
42
|
Witkin JM, Cerne R, Newman AH, Izenwasser S, Smith JL, Tortella FC. N-Substituted-3-alkoxy-derivatives of dextromethorphan are functional NMDA receptor antagonists in vivo: Evidence from an NMDA-induced seizure model in rats. Pharmacol Biochem Behav 2021; 203:173154. [PMID: 33609599 PMCID: PMC9659398 DOI: 10.1016/j.pbb.2021.173154] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 11/30/2022]
Abstract
Interest in developing NMDA receptor antagonists with reduced side-effects for neurological and psychiatric disorders has been re-energized by the recent introduction of esketamine into clinical practice for treatment-resistant depression. Structural analogs of dextromethorphan bind with low affinity to the NMDA receptor ion channel, have functional effects in vivo, and generally display a lower propensity for side-effects than that of ketamine and other higher affinity antagonists. As such, the aim of the present study was to determine whether a series of N-substituted-3-alkoxy-substituted dextromethorphan analogs produce their anticonvulsant effects through NMDA receptor blockade. Compounds were studied against NMDA-induced seizures in rats. Compounds were administered intracerebroventricularly in order to mitigate confounds of drug metabolism that arise from systemic administration. Comparison of the anticonvulsant potencies to their affinities for NMDA, σ1, and σ2 binding sites were made in order to evaluate the contribution of these receptors to anticonvulsant efficacy. The potencies to block convulsions were positively associated with their affinities to bind to the NMDA receptor ion channel ([3H]-TCP binding) (r = 0.71, p < 0.05) but not to σ1 receptors ([3H]-SKF 10047 binding) (r = -0.31, p = 0.46) or to σ2 receptors ([3H]-DTG binding) (p = -0.38, p = 0.36). This is the first report demonstrating that these dextromethorphan analogs are functional NMDA receptor antagonists in vivo. Given their potential therapeutic utility and favorable side-effect profiles, such low affinity NMDA receptor antagonists could be considered for further development in neurological (e.g., anticonvulsant) and psychiatric (e.g., antidepressant) disorders.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA; Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.
| | - Rok Cerne
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA; Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Amy H Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Sari Izenwasser
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA
| | - Frank C Tortella
- Department of Neuropharmacology and Molecular Biology, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
43
|
Kris-Etherton PM, Petersen KS, Hibbeln JR, Hurley D, Kolick V, Peoples S, Rodriguez N, Woodward-Lopez G. Nutrition and behavioral health disorders: depression and anxiety. Nutr Rev 2021; 79:247-260. [PMID: 32447382 PMCID: PMC8453603 DOI: 10.1093/nutrit/nuaa025] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Suboptimal nutrition has been implicated in the underlying pathology of behavioral health disorders and may impede treatment and recovery. Thus, optimizing nutritional status should be a treatment for these disorders and is likely important for prevention. The purpose of this narrative review is to describe the global burden and features of depression and anxiety, and summarize recent evidence regarding the role of diet and nutrition in the prevention and management of depression and anxiety. Current evidence suggests that healthy eating patterns that meet food-based dietary recommendations and nutrient requirements may assist in the prevention and treatment of depression and anxiety. Randomized controlled trials are needed to better understand how diet and nutrition-related biological mechanisms affect behavioral health disorders, to assist with the development of effective evidence-based nutrition interventions, to reduce the impact of these disorders, and promote well-being for affected individuals.
Collapse
Affiliation(s)
- Penny M Kris-Etherton
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Kristina S Petersen
- Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Joseph R Hibbeln
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland, USA
| | | | - Valerie Kolick
- Substance Abuse and Mental Health Services Administration, Rockville, Maryland, USA
| | - Sevetra Peoples
- Milken Institute School of Public Health, George Washington University, Washington, DC, USA
| | - Nancy Rodriguez
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Gail Woodward-Lopez
- Nutrition Policy Institute, University of California, Agriculture and Natural Resources, Berkeley, California, USA
| |
Collapse
|
44
|
Robinson B, Gu Q, Kanungo J. Antidepressant Actions of Ketamine: Potential Role of L-Type Calcium Channels. Chem Res Toxicol 2021; 34:1198-1207. [PMID: 33566591 DOI: 10.1021/acs.chemrestox.0c00411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recently, the United States Food and Drug Administration approved esketamine, the S-enantiomer of ketamine, as a fast-acting therapeutic drug for treatment-resistant depression. Although ketamine is known as an N-methyl-d-aspartate (NMDA) receptor antagonist, the underlying mechanisms of how it elicits an antidepressant effect, specifically at subanesthetic doses, are not clear and remain an advancing field of research interest. On the other hand, high-dose (more than the anesthetic dose) ketamine-induced neurotoxicity in animal models has been reported. There has been progress in understanding the potential pathways involved in ketamine-induced antidepressant effects, some of which include NMDA-receptor antagonism, modulation of voltage-gated calcium channels, and brain-derived neurotrophic factor (BDNF) signaling. Often these pathways have been shown to be linked. Voltage-gated L-type calcium channels have been shown to mediate the rapid-acting antidepressant effects of ketamine, especially involving induction of BDNF synthesis downstream, while BDNF deficiency decreases the expression of L-type calcium channels. This review focuses on the reported studies linking ketamine's rapid-acting antidepressant actions to L-type calcium channels with an objective to present a perspective on the importance of the modulation of intracellular calcium in mediating the effects of subanesthetic (antidepressant) versus high-dose ketamine (anesthetic and potential neurotoxicant), the latter having the ability to reduce intracellular calcium by blocking the calcium-permeable NMDA receptors, which is implicated in potential neurotoxicity.
Collapse
Affiliation(s)
- Bonnie Robinson
- Division of Neurotoxicology, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Qiang Gu
- Division of Neurotoxicology, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Jyotshna Kanungo
- Division of Neurotoxicology, United States Food and Drug Administration, 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| |
Collapse
|
45
|
Rothberg RL, Azhari N, Haug NA, Dakwar E. Mystical-type experiences occasioned by ketamine mediate its impact on at-risk drinking: Results from a randomized, controlled trial. J Psychopharmacol 2021; 35:150-158. [PMID: 33307947 DOI: 10.1177/0269881120970879] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Sub-anesthetic ketamine administration may be helpful for substance use disorders. Converging evidence suggests that the efficacy of ketamine for certain conditions may implicate a subset of its psychoactive effects. AIMS The aim of this analysis is to evaluate whether the mystical-type effects of ketamine are critical for clinical efficacy in alcohol-dependent individuals. In this secondary analysis, we determine if a subset of the psychoactive effects of ketamine, the so-called mystical-type experience, mediates the effect of ketamine, when combined with motivational enhancement therapy, on at-risk drinking behavior in alcohol-dependent individuals interested in treatment. METHODS Forty alcohol dependent adults were randomized to either a 52-minute infusion of ketamine or midazolam, which they received on a designated quit-day during the second week of a five-week motivational enhancement therapy regimen. Psychoactive effects were assessed following the infusion, and alcohol use was monitored for the subsequent 3 weeks at each twice-weekly visit. RESULTS We found that ketamine leads to significantly greater mystical-type effects (by Hood Mysticism Scale) and dissociation (by Clinician Administered Dissociative States Scale) compared to the active control. Ketamine also led to significant reduction in at-risk drinking. The Hood Mysticism Scale, but not Clinician Administered Dissociative States Scale score, was found to mediate the effect of ketamine on drinking behavior. CONCLUSIONS This trial adds evidence to the literature on the importance of mystical-type experiences in addiction treatment. Future research should continue to investigate the relationship between the psychoactive effects of psychedelic therapeutics and clinical outcomes for other substance use and mental health disorders.
Collapse
Affiliation(s)
| | - Nour Azhari
- New York State Psychiatric Institute, Columbia University Medical Center, New York, USA
| | - Nancy A Haug
- Department of Psychology, Palo Alto University, Palo Alto, USA
| | - Elias Dakwar
- New York State Psychiatric Institute, Columbia University Medical Center, New York, USA
| |
Collapse
|
46
|
Melnyk MI, Dryn DO, Al Kury LT, Dziuba DO, Zholos AV. Suppression of mI CAT in Mouse Small Intestinal Myocytes by General Anaesthetic Ketamine and its Recovery by TRPC4 Agonist (-)-englerin A. Front Pharmacol 2021; 11:594882. [PMID: 33390980 PMCID: PMC7775583 DOI: 10.3389/fphar.2020.594882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/02/2020] [Indexed: 11/19/2022] Open
Abstract
A better understanding of the negative impact of general anesthetics on gastrointestinal motility requires thorough knowledge of their molecular targets. In this respect the muscarinic cationic current (mICAT carried mainly via TRPC4 channels) that initiates cholinergic excitation-contraction coupling in the gut is of special interest. Here we aimed to characterize the effects of one of the most commonly used “dissociative anesthetics”, ketamine, on mICAT. Patch-clamp and tensiometry techniques were used to investigate the mechanisms of the inhibitory effects of ketamine on mICAT in single mouse ileal myocytes, as well as on intestinal motility. Ketamine (100 µM) strongly inhibited both carbachol- and GTPγS-induced mICAT. The inhibition was slow (time constant of about 1 min) and practically irreversible. It was associated with altered voltage dependence and kinetics of mICAT. In functional tests, ketamine suppressed both spontaneous and carbachol-induced contractions of small intestine. Importantly, inhibited by ketamine mICAT could be restored by direct TRPC4 agonist (-)-englerin A. We identified mICAT as a novel target for ketamine. Signal transduction leading to TRPC4 channel opening is disrupted by ketamine mainly downstream of muscarinic receptor activation, but does not involve TRPC4 per se. Direct TRPC4 agonists may be used for the correction of gastrointestinal disorders provoked by general anesthesia.
Collapse
Affiliation(s)
- Mariia I Melnyk
- A.A. Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine.,Institute of Pharmacology and Toxicology, National Academy of Medical Science of Ukraine, Kyiv, Ukraine.,ESC "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Dariia O Dryn
- A.A. Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine.,Institute of Pharmacology and Toxicology, National Academy of Medical Science of Ukraine, Kyiv, Ukraine.,ESC "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Lina T Al Kury
- Department of Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
| | - Dmytro O Dziuba
- Shupyk National Medical Academy of Postgraduate Education, Kyiv, Ukraine
| | - Alexander V Zholos
- ESC "Institute of Biology and Medicine", Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| |
Collapse
|
47
|
Acevedo-Diaz EE, Cavanaugh GW, Greenstein D, Kraus C, Kadriu B, Park L, Zarate CA. Can 'floating' predict treatment response to ketamine? Data from three randomized trials of individuals with treatment-resistant depression. J Psychiatr Res 2020; 130:280-285. [PMID: 32861983 PMCID: PMC8073211 DOI: 10.1016/j.jpsychires.2020.06.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/23/2020] [Accepted: 06/16/2020] [Indexed: 01/04/2023]
Abstract
Ketamine has rapid-acting antidepressant properties but also potentially concerning transient dissociative side effects (SEs). Recent studies noted a positive correlation between treatment response to ketamine and general dissociative SEs, as well as "floating", a depersonalization SE (a subtype of the dissociative SEs). This analysis sought to determine whether floating mediates treatment response to ketamine. Data were pooled from three double-blind, crossover, placebo-controlled ketamine clinical trials across which 82 participants with treatment-resistant depression (TRD) (44 with bipolar depression and 38 with major depressive disorder) received placebo and ketamine (0.5 mg/kg) infusions. SEs were actively solicited in a standardized fashion before and after ketamine infusion. The hypothesis that a post-infusion experience of floating would mediate antidepressant response to ketamine was assessed at 230 min post-infusion and at Day 1. Montgomery-Asberg Depression Rating Scale (MADRS) total score was the dependent variable in a linear mixed effects model. Ketamine significantly decreased MADRS scores (p < 0.0001), but no relationship was detected between floating and MADRS score at either 230 min or Day 1 post-infusion. The hypothesized mediation effect of floating was also not detected at either 230 min or Day 1 post-infusion. Taken together, the findings do not support the hypothesis that ketamine's antidepressant effects are mediated by the dissociative depersonalization subtype SE of floating.
Collapse
Affiliation(s)
- Elia E Acevedo-Diaz
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Grace W Cavanaugh
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Dede Greenstein
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Christoph Kraus
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Bashkim Kadriu
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Lawrence Park
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
48
|
Crapser JD, Ochaba J, Soni N, Reidling JC, Thompson LM, Green KN. Microglial depletion prevents extracellular matrix changes and striatal volume reduction in a model of Huntington's disease. Brain 2020; 143:266-288. [PMID: 31848580 DOI: 10.1093/brain/awz363] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/22/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease is associated with a reactive microglial response and consequent inflammation. To address the role of these cells in disease pathogenesis, we depleted microglia from R6/2 mice, a rapidly progressing model of Huntington's disease marked by behavioural impairment, mutant huntingtin (mHTT) accumulation, and early death, through colony-stimulating factor 1 receptor inhibition (CSF1Ri) with pexidartinib (PLX3397) for the duration of disease. Although we observed an interferon gene signature in addition to downregulated neuritogenic and synaptic gene pathways with disease, overt inflammation was not evident by microglial morphology or cytokine transcript levels in R6/2 mice. Nonetheless, CSF1Ri-induced microglial elimination reduced or prevented disease-related grip strength and object recognition deficits, mHTT accumulation, astrogliosis, and striatal volume loss, the latter of which was not associated with reductions in cell number but with the extracellular accumulation of chondroitin sulphate proteoglycans (CSPGs)-a primary component of glial scars. A concurrent loss of proteoglycan-containing perineuronal nets was also evident in R6/2 mice, and microglial elimination not only prevented this but also strikingly increased perineuronal nets in the brains of naïve littermates, suggesting a new role for microglia as homeostatic regulators of perineuronal net formation and integrity.
Collapse
Affiliation(s)
- Joshua D Crapser
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Joseph Ochaba
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Neelakshi Soni
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Jack C Reidling
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA.,Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| |
Collapse
|
49
|
Ermers NJ, Hagoort K, Scheepers FE. The Predictive Validity of Machine Learning Models in the Classification and Treatment of Major Depressive Disorder: State of the Art and Future Directions. Front Psychiatry 2020; 11:472. [PMID: 32523557 PMCID: PMC7261928 DOI: 10.3389/fpsyt.2020.00472] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 05/07/2020] [Indexed: 11/13/2022] Open
Abstract
Major depressive disorder imposes a substantial disease burden worldwide, ranking as the third leading contributor to global disability. In spite of its ubiquity, classifying and treating depression has proven troublesome. One argument put forward to explain this predicament is the heterogeneity of patients diagnosed with the disorder. Recently, many areas of daily life have witnessed the surge of machine learning techniques, computational approaches to elucidate complex patterns in large datasets, which can be employed to make predictions and detect relevant clusters. Due to the multidimensionality at play in the pathogenesis of depression, it is suggested that machine learning could contribute to improving classification and treatment. In this paper, we investigated literature focusing on the use of machine learning models on datasets with clinical variables of patients diagnosed with depression to predict treatment outcomes or find more homogeneous subgroups. Identified studies based on best practices in the field are evaluated. We found 16 studies predicting outcomes (such as remission) and identifying clusters in patients with depression. The identified studies are mostly still in proof-of-concept phase, with small datasets, lack of external validation, and providing single performance metrics. Larger datasets, and models with similar variables present across these datasets, are needed to develop accurate and generalizable models. We hypothesize that harnessing natural language processing to obtain data 'hidden' in clinical texts might prove useful in improving prediction models. Besides, researchers will need to focus on the conditions to feasibly implement these models to support psychiatrists and patients in their decision-making in practice. Only then we can enter the realm of precision psychiatry.
Collapse
Affiliation(s)
- Nick J. Ermers
- Department of Psychiatry, University Medical Center Utrecht, Utrecht, Netherlands
| | | | | |
Collapse
|
50
|
Ramakrishnan N, Murphy NRE, Walker CP, Cuellar Leal VA, Soares JC, Cho RYJ, Selvaraj S. Neurophysiological Effect of Ketamine on Prefrontal Cortex in Treatment-Resistant Depression: A Combined Transcranial Magnetic Stimulation-Electroencephalography Study. CHRONIC STRESS 2020; 3:2470547019861417. [PMID: 32440596 PMCID: PMC7219868 DOI: 10.1177/2470547019861417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Nithya Ramakrishnan
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Nicholas R E Murphy
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Christopher P Walker
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Valeria A Cuellar Leal
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jair C Soares
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Raymond Y J Cho
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Sudhakar Selvaraj
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|