Novel Approaches for the Management of Depressive Disorders

Total Page:16

File Type:pdf, Size:1020Kb

Novel Approaches for the Management of Depressive Disorders European Journal of Pharmacology 771 (2016) 236–240 Contents lists available at ScienceDirect European Journal of Pharmacology journal homepage: www.elsevier.com/locate/ejphar Perspective Novel approaches for the management of depressive disorders Manuella P. Kaster a, Morgana Moretti b, Mauricio P. Cunha a, Ana Lúcia S. Rodrigues a,n a Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil b Department of Nutrition, Center of Health Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil article info abstract Article history: Major depressive disorder is a disabling psychiatric condition that causes a significant burden on in- Received 22 July 2015 dividuals and society. There is still a lack of a clear understanding of the neuropathological changes Received in revised form associated with this illness and the efficacy of antidepressants is still far from optimal. Research into 14 November 2015 antidepressant therapies has evolved from serendipitous observation in human trials, but more than 60 Accepted 14 December 2015 years after the first monoaminergic antidepressants emerged they remain the mainstay for treating Available online 15 December 2015 depression. However, glutamatergic modulators such as ketamine became the forefront of anti- Keywords: depressant exploration, especially for treatment-resistant depression and suicidal ideation. The gluta- Depression matergic hypothesis of depression is not new, however other NMDA receptor modulators do not seem to Antidepressants share the rapid and sustained effects of ketamine, suggesting that a unique combination of intracellular Glutamatergic system targets might be involved in its effect. Interestingly, inflammation can impact the glutamatergic system Inflammation enhancing excitotoxicity and decreasing neuroplasticity. The points of convergence between the in- flammatory and glutamatergic hypotheses of depression are not completely established, especially re- garding the effects of fast-acting antidepressants. In this review, we discuss the most recent research surrounding glutamatergic fast-acting antidepressants, capable of modulating cellular plasticity and synaptogenesis and the potential of anti-inflammatory compounds evaluated from a different perspec- tive. The combination of innovative ideas plus improvements on the discoveries made so far might lead to advances in antidepressant research with the promise of finding compounds that are both effective and fast-acting, even in patients who have tried other therapies with limited success. & 2015 Elsevier B.V. All rights reserved. 1. Major depressive disorder (MDD) – pathophysiology and and prefrontal cortex, leading to stimulation of intracellular sig- current antidepressant treatment naling pathways and consequent regulation of genes associated with neuroplasticity and cell survival (Castrén and Rantamaki, MDD causes profound socioeconomic burden and negative 2010). impact on functioning and quality of life of patients (Papakostas Inflammation has also emerged as an important etiologic fac- and Ionescu, 2015). Although there has been significant research tor, and thus a potential pharmacological target for MDD. Pro-in- directed towards understanding the biologic underpinnings of flammatory cytokines are increased in depressed patients (Dowlati MDD, there is not a complete consensus about the neurochemical et al., 2009) resulting in an imbalance in critical neuroactive alterations present in depressive individuals. The monoamine compounds and changes in brain structural and synaptic plasticity hypothesis of depression proposes that underactivity of mono- (Dantzer and Walker, 2014). Interestingly, antidepressant therapy may attenuate this inflammatory dysfunction in depressed in- amines may underlie the pathophysiology of this disorder dividuals (Lanquillon et al., 2000). Moreover, neuroinflammation is (Schildkraut, 1965). In addition, impairments on structural plasti- associated with increased extracellular glutamate levels, causing city and cellular resilience associated with reduced levels of brain- neurotoxicity that contributes to depressive symptoms (Sanacora derived neurotrophic factor (BDNF) have been implicated in the et al., 2012). pathophysiology of MDD (Sen et al., 2008). Chronic antidepressant Current antidepressant treatments are limited in both efficacy treatment increases BDNF levels, particularly in the hippocampus and tolerability. The drawbacks include delayed therapeutic onset and side effects that reduce adhesion to treatment (Papakostas n Corresponding author. and Ionescu, 2015). Approaches targeting new pathways have E-mail address: [email protected] (A.L.S. Rodrigues). been investigated and may represent a significant improvement in http://dx.doi.org/10.1016/j.ejphar.2015.12.029 0014-2999/& 2015 Elsevier B.V. All rights reserved. M.P. Kaster et al. / European Journal of Pharmacology 771 (2016) 236–240 237 the therapeutic armamentarium for treating MDD. number of subjects (Shopsin, 2013) and ascorbic acid was effective as adjuvant agent for MDD in pediatric patients (Amr et al., 2013). Antidepressant effects of creatine supplementation were also re- 2. The search for fast-acting antidepressants: targeting the ported in depressive patients (Allen, 2012). However, there are no glutamatergic system clinical studies evaluating the mood effects of guanosine. The antidepressant effects of other NMDA receptor antagonists Seminal work by Trullas and Skolnick (1990) was the first re- and glutamate-modulators such as MK-801, AP-7, RO25-6981, porting that NMDA receptor antagonists exhibit antidepressant amantadine, memantine, CP-101,606 (traxoprodil) and riluzole properties in preclinical models. After this observation, several were extensively studied in preclinical models (Dutta et al., 2015). laboratories demonstrated that functional antagonists at multiple Most of these agents fail to afford rapid and sustained anti- NMDA receptor sites including ligands at glutamate, polyamine, depressant effects (Dutta et al., 2015; Preskorn et al., 2008; Zarate glycine, bivalent cations and ionophore recognition sites are effi- et al., 2004). Some promising results were observed with the low- cacious in models of depression (Sanacora et al., 2012). trapping NMDA channel blocker AZD6765, which elicits rapid, but In the late 1990s reduction of NMDA receptor function was not sustained, antidepressant effects in treatment-resistant MDD recognized as a long-term adaptation associated with anti- patients (Zarate et al., 2013). However, the intracellular pathways depressant effects (Skolnick, 1999). A promising clinical study by involved in its effect are not established. Also, antidepressant-like Berman et al. (2000) showed that the non-competitive NMDA effect of the NMDA receptor glycine-site partial agonist GLYX-13 receptor antagonist ketamine induced a rapid and persistent an- requires AMPA receptor and mTOR activation, similar to ketamine tidepressant response in severe depressive patients. Evidence now (Lu et al., 2014). The antidepressant potential of GLYX-13 was indicates that ketamine inhibits NMDA receptors on GABAergic confirmed in a clinical trial, and a fast (2 h) and sustained remis- interneurons increasing glutamate release. The consequent acti- sion of depressive symptoms was observed without dissociative vation of AMPA receptors causes depolarization of postsynaptic effects (Moskal et al., 2014; Preskorn et al., 2015). CP-101,606, a neurons, leading to voltage-dependent calcium channel activation. selective NR2B receptor antagonist, produced symptoms’ remis- As a consequence, BDNF released from vesicles activates mTOR sion by day 5 in depressed patients unresponsive to paroxetine signaling. Finally, translation of synaptic proteins in prefrontal (Preskorn et al., 2008). However, the intracellular pathways asso- cortex, particularly those implicated in synaptogenesis, including ciated with this effect need to be elucidated in preclinical studies. PSD-95, synapsin and GluA1 is the final event associated with Metabotropic glutamate receptor modulators and AMPA re- ketamine’s rapid antidepressant effect (Li et al., 2010). ceptor potentiators also represent interesting targets in the search Although the discovery of ketamine for treating severe de- for novel fast-acting antidepressant compounds. The blockade of pressed patients has represented a significant advance in the field, presynaptic mGluR2/3 receptors regulates glutamate release and its abuse liability and potential neurotoxicity upon repeated use produces rapid behavioral responses that require activation of ei- has led to the interest in the development of safer fast-acting ther AMPA receptors or mTOR signaling (Dwyer et al., 2012). antidepressants (Abdallah et al., 2015). Considering that ketamine Specifically, LY341495, an mGlu2/3 receptor antagonist, reduced modulates the glutamatergic system, there has been increasing the time required for antidepressants to exert their effects by a interest in targeting this system for the development of novel fast- mechanism dependent on stimulation and expression of AMPA acting antidepressants. receptors in prefrontal cortex (Matrisciano et al., 2005). Moreover, Our research group has particular interest in antidepressant preclinical studies have shown that AMPA potentiators LY392098 effects afforded by some endogenous compounds that target the and LY451616 exhibit antidepressant effects associated with hip- glutamatergic system. Several compounds
Recommended publications
  • The Excitotoxin Quinolinic Acid Induces Tau Phosphorylation in Human Neurons
    The Excitotoxin Quinolinic Acid Induces Tau Phosphorylation in Human Neurons Abdur Rahman1,4., Kaka Ting2, Karen M. Cullen3, Nady Braidy4, Bruce J. Brew2,5, Gilles J. Guillemin2,4.* 1 Department of Family Sciences, College for Women, Kuwait University, Shuwaikh, Kuwait, 2 St Vincent’s Hospital, Centre for Applied Medical Research, Department of Neuroimmunology, Darlinghurst, New South Wales, Australia, 3 Disciplines of Anatomy and Histology, School of Medical Science, The University of Sydney, New South Wales, Australia, 4 Department of Pharmacology, University of New South Wales, School of Medical Science, Sydney, New South Wales, Australia, 5 Department of Neurology, St Vincent’s Hospital, Darlinghurst, New South Wales, Australia Abstract Some of the tryptophan catabolites produced through the kynurenine pathway (KP), and more particularly the excitotoxin quinolinic acid (QA), are likely to play a role in the pathogenesis of Alzheimer’s disease (AD). We have previously shown that the KP is over activated in AD brain and that QA accumulates in amyloid plaques and within dystrophic neurons. We hypothesized that QA in pathophysiological concentrations affects tau phosphorylation. Using immunohistochemistry, we found that QA is co-localized with hyperphosphorylated tau (HPT) within cortical neurons in AD brain. We then investigated in vitro the effects of QA at various pathophysiological concentrations on tau phosphorylation in primary cultures of human neurons. Using western blot, we found that QA treatment increased the phosphorylation of tau at serine 199/202, threonine 231 and serine 396/404 in a dose dependent manner. Increased accumulation of phosphorylated tau was also confirmed by immunocytochemistry. This increase in tau phosphorylation was paralleled by a substantial decrease in the total protein phosphatase activity.
    [Show full text]
  • Download Product Insert (PDF)
    Product Information Quinolinic Acid Item No. 14941 CAS Registry No.: 89-00-9 Formal Name: 2,3-pyridinedicarboxylic acid Synonyms: NSC 13127, NSC 18836, NSC 403247 O N MF: C7H5NO4 FW: HO 167.1 HO Purity: ≥98% Stability: ≥2 years at -20°C Supplied as: A crystalline solid O λ UV/Vis.: max: 216, 264 nm Laboratory Procedures For long term storage, we suggest that quinolinic acid be stored as supplied at -20°. It should be stable for at least two years. Quinolinic acid is supplied as a crystalline solid. A stock solution may be made by dissolving the quinolinic acid in the solvent of choice. Quinolinic acid is soluble in organic solvents such as DMSO and dimethyl formamide, which should be purged with an inert gas. The solubility of quinolinic acid in these solvents is approximately 16 mg/ml. Further dilutions of the stock solution into aqueous buffers or isotonic saline should be made prior to performing biological experiments. Ensure that the residual amount of organic solvent is insignificant, since organic solvents may have physiological effects at low concentrations. Organic solvent-free aqueous solutions of quinolinic acid can be prepared by directly dissolving the crystalline solid in aqueous buffers. The solubility of quinolinic acid in PBS, pH 7.2, is approximately 0.5 mg/ml. We do not recommend storing the aqueous solution for more than one day. Quinolinic acid is an endogenous agonist at NMDA receptors that is generated through the metabolism of tryptophan in the kynurenine pathway.1 By overactivating NMDA receptors, quinolinic acid produces neurotoxicity, which has been implicated in certain neurodegenerative disorders.2 Quinolinic acid can also generate reactive oxygen species, has immunomodulatory actions, and promotes the formation of hyperphosphorylated tau proteins.3-5 References 1.
    [Show full text]
  • Sciatica and Chronic Pain
    Sciatica and Chronic Pain Past, Present and Future Robert W. Baloh 123 Sciatica and Chronic Pain Robert W. Baloh Sciatica and Chronic Pain Past, Present and Future Robert W. Baloh, MD Department of Neurology University of California, Los Angeles Los Angeles, CA, USA ISBN 978-3-319-93903-2 ISBN 978-3-319-93904-9 (eBook) https://doi.org/10.1007/978-3-319-93904-9 Library of Congress Control Number: 2018952076 © Springer International Publishing AG, part of Springer Nature 2019 This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed. The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations and therefore free for general use. The publisher, the authors, and the editors are safe to assume that the advice and information in this book are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty, express or implied, with respect to the material contained herein or for any errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
    [Show full text]
  • Linking Phencyclidine Intoxication to the Tryptophan-Kynurenine Pathway Therapeutic Implications for Schizophrenia
    Neurochemistry International 125 (2019) 1–6 Contents lists available at ScienceDirect Neurochemistry International journal homepage: www.elsevier.com/locate/neuint Linking phencyclidine intoxication to the tryptophan-kynurenine pathway: Therapeutic implications for schizophrenia T ∗ Hidetsugu Fujigakia, ,1, Akihiro Mourib,c,1, Yasuko Yamamotoa, Toshitaka Nabeshimac,d, Kuniaki Saitoa,c,d,e a Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Sciences, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470- 1192, Japan b Department of Regulatory Science, Fujita Health University Graduate School of Health Sciences, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan c Japanese Drug Organization of Appropriate Use and Research, 3-1509 Omoteyama, Tenpaku-ku, Nagoya, Aichi 468-0069, Japan d Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Sciences, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan e Human Health Sciences, Graduate School of Medicine and Faculty of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan ARTICLE INFO ABSTRACT Keywords: Phencyclidine (PCP) is a dissociative anesthetic that induces psychotic symptoms and neurocognitive deficits in Phencyclidine rodents similar to those observed in schizophrenia patients. PCP administration in healthy human subjects in- Kynurenic acid duces schizophrenia-like symptoms such as positive and negative symptoms, and a range of cognitive deficits. It Quinolinic acid has been reported that PCP, ketamine, and related drugs such as N-methyl-D-aspartate-type (NMDA) glutamate Kynurenine pathway receptor antagonists, induce behavioral effects by blocking neurotransmission at NMDA receptors. Further, Schizophrenia NMDA receptor antagonists reproduce specific aspects of the symptoms of schizophrenia.
    [Show full text]
  • A Human Stem Cell-Derived Test System for Agents Modifying Neuronal N
    Archives of Toxicology (2021) 95:1703–1722 https://doi.org/10.1007/s00204-021-03024-0 IN VITRO SYSTEMS A human stem cell‑derived test system for agents modifying neuronal 2+ N‑methyl‑D‑aspartate‑type glutamate receptor Ca ‑signalling Stefanie Klima1,2 · Markus Brüll1 · Anna‑Sophie Spreng1,3 · Ilinca Suciu1,3 · Tjalda Falt1 · Jens C. Schwamborn4 · Tanja Waldmann1 · Christiaan Karreman1 · Marcel Leist1,5 Received: 28 October 2020 / Accepted: 4 March 2021 / Published online: 13 March 2021 © The Author(s) 2021 Abstract Methods to assess neuronal receptor functions are needed in toxicology and for drug development. Human-based test systems that allow studies on glutamate signalling are still scarce. To address this issue, we developed and characterized pluripotent stem cell (PSC)-based neural cultures capable of forming a functional network. Starting from a stably proliferating neu- roepithelial stem cell (NESC) population, we generate “mixed cortical cultures” (MCC) within 24 days. Characterization by immunocytochemistry, gene expression profling and functional tests (multi-electrode arrays) showed that MCC contain various functional neurotransmitter receptors, and in particular, the N-methyl-D-aspartate subtype of ionotropic glutamate receptors (NMDA-R). As this important receptor is found neither on conventional neural cell lines nor on most stem cell- derived neurons, we focused here on the characterization of rapid glutamate-triggered Ca2+ signalling. Changes of the intra- 2+ cellular free calcium ion concentration ([Ca ]i) were measured by fuorescent imaging as the main endpoint, and a method to evaluate and quantify signals in hundreds of cells at the same time was developed. We observed responses to glutamate in the low µM range.
    [Show full text]
  • Systemic Approaches to Modifying Quinolinic Acid Striatal Lesions in Rats
    The Journal of Neuroscience, October 1988, B(10): 3901-3908 Systemic Approaches to Modifying Quinolinic Acid Striatal Lesions in Rats M. Flint Beal, Neil W. Kowall, Kenton J. Swartz, Robert J. Ferrante, and Joseph B. Martin Neurology Service, Massachusetts General Hospital, and Department of Neurology, Harvard Medical School, Boston, Massachusetts 02114 Quinolinic acid (QA) is an endogenous excitotoxin present mammalian brain, is an excitotoxin which producesaxon-spar- in mammalian brain that reproduces many of the histologic ing striatal lesions. We found that this compound produced a and neurochemical features of Huntington’s disease (HD). more exact model of HD than kainic acid, sincethe lesionswere In the present study we have examined the ability of a variety accompaniedby a relative sparingof somatostatin-neuropeptide of systemically administered compounds to modify striatal Y neurons (Beal et al., 1986a). QA neurotoxicity. Lesions were assessed by measurements If an excitotoxin is involved in the pathogenesisof HD, then of the intrinsic striatal neurotransmitters substance P, so- agentsthat modify excitotoxin lesionsin vivo could potentially matostatin, neuropeptide Y, and GABA. Histologic exami- be efficacious as therapeutic agents in HD. The best form of nation was performed with Nissl stains. The antioxidants therapy from a practical standpoint would be a drug that could ascorbic acid, beta-carotene, and alpha-tocopherol admin- be administered systemically, preferably by an oral route. In the istered S.C. for 3 d prior to striatal QA lesions had no sig- presentstudy we have therefore examined the ability of a variety nificant effect. Other drugs were administered i.p. l/2 hr prior of systemically administered drugs to modify QA striatal neu- to QA striatal lesions.
    [Show full text]
  • Advances in Non-Dopaminergic Treatments for Parkinson's Disease
    REVIEW ARTICLE published: 22 May 2014 doi: 10.3389/fnins.2014.00113 Advances in non-dopaminergic treatments for Parkinson’s disease Sandy Stayte 1,2 and Bryce Vissel 1,2* 1 Neuroscience Department, Neurodegenerative Disorders Laboratory, Garvan Institute of Medical Research, Sydney, NSW, Australia 2 Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia Edited by: Since the 1960’s treatments for Parkinson’s disease (PD) have traditionally been directed Eero Vasar, University of Tartu, to restore or replace dopamine, with L-Dopa being the gold standard. However, chronic Estonia L-Dopa use is associated with debilitating dyskinesias, limiting its effectiveness. This has Reviewed by: resulted in extensive efforts to develop new therapies that work in ways other than Andrew Harkin, Trinity College Dublin, Ireland restoring or replacing dopamine. Here we describe newly emerging non-dopaminergic Sulev Kõks, University of Tartu, therapeutic strategies for PD, including drugs targeting adenosine, glutamate, adrenergic, Estonia and serotonin receptors, as well as GLP-1 agonists, calcium channel blockers, iron Pille Taba, Universoty of Tartu, chelators, anti-inflammatories, neurotrophic factors, and gene therapies. We provide a Estonia Pekka T. Männistö, University of detailed account of their success in animal models and their translation to human clinical Helsinki, Finland trials. We then consider how advances in understanding the mechanisms of PD, genetics, *Correspondence: the possibility that PD may consist of multiple disease states, understanding of the Bryce Vissel, Neuroscience etiology of PD in non-dopaminergic regions as well as advances in clinical trial design Department, Neurodegenerative will be essential for ongoing advances. We conclude that despite the challenges ahead, Disorders Laboratory, Garvan Institute of Medical Research, patients have much cause for optimism that novel therapeutics that offer better disease 384 Victoria Street, Darlinghurst, management and/or which slow disease progression are inevitable.
    [Show full text]
  • Therapy Focus – J&J Confirms Hope for New Mechanism in Depression
    May 09, 2018 Therapy focus – J&J confirms hope for new mechanism in depression Amy Brown The first look at late-stage data on Johnson & Johnson’s hotly-tipped novel antidepressant esketamine came at a medical conference last weekend, and the results were not quite as strong as many were hoping to see. Encouraging signals could be certainly be found, however, and J&J seems undeterred from trying to seek regulatory approval. Others pursing NMDA modulation in depression are also likely to draw comfort in the results, which provide the first phase III validation of this mechanism, and a clear bar to beat (see table below). Hopes for esketamine and other products like it lie in their similarity to ketamine, which at low doses displays antidepressant effects that kick in very quickly. Traditional antidepressants which act via the serotonergic system – selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs) – can take weeks to have an effect; a large proportion of patients fail to respond at all. Hence the need for both faster acting agents – for example in suicidal patients – and novel mechanisms to treat those who need different options. A new target NMDA receptor modulation is a novel mechanism in depression. Over the last decade researchers have become increasingly convinced that dysregulation of glutamate, a neurotransmitter which signals through NMDA, plays an important role in the condition. This system is already known to be a factor in cognitive function and neurodegeneration and Namenda, the NMDA antagonist mematine, has been available as a treatment for Alzheimer’s dementia for several years.
    [Show full text]
  • Changes in Glutamate Levels Measured by Glutamate
    Changes in glutamate levels measured by glutamate voltammetry in the rat medial prefrontal cortex after treatment with N-methyl-D-aspartate receptor antagonists (P.1.g.023) Irina Ionescu, Kelly Allers, Roberto Arban, Cornelia Dorner-Ciossek and Lothar Kussmaul Boehringer Ingelheim, CNS Diseases Research Background Hypothesized antidepressant MoA of NMDA-R antagonists: Disinhibition of glutamatergic neurons in the PFC N-methyl-D-aspartate receptor (NMDA-R) have been described as potential novel treatment options in the therapy of treatment-reistant depression (TRD). A single sub-anesthetic dose of the noncompetitive NMDA-R antagonist GABA ketamine has been shown to exert rapid-onset and long-lasting antidepressant Glutamate Glutamate NMDA-R effects in TRD patients; these effects were accompanied by brief psychotic Increased glutamate release episodes. Fast-onset antidepressant effects without psychotic side effects have also been described in the clinic for the NR2B-selective negative allosteric modulator (NAM) traxoprodil. However, other unspecific NMDA-R blockers, Synaptic plasticity such as lanicemine, failed to show fast-onset long-lasting antidepressant NMDA-R antagonists (ketamine, traxoprodil) effects in the clinic after single application. Here, we used glutamate AMPA-R AMPA-R voltammetry in rats in order to identify a preclinical predictor of clinical efficacy The increase in glutamate release as proof of circuit by circuit engagement in the prefrontal cortex (PFC). engagement and antidepressant activity can be measured by glutamate voltammetry. Using glutamate voltammetry to show circuit engagement by clinically active NMDA-R antagonists in the PFC Glutamate biosensor (Pinnacle Technology Inc., USA) Advantages of glutamate voltammetry Experimental design - fast response time Experiments were performed in Wistar Han rats implanted with Recording site: PFC - fast sampling time (1 Hz) guide cannulae at the specified coordinates.
    [Show full text]
  • Ketamine: a Novel Antidepressant with a Fast Onset of Action?† COMMENTARY ON… COCHRANE CORNER Katharine Smith & Mary Jane Attenburrow
    BJPsych Advances (2016), vol. 22, 216–221 doi: 10.1192/apt.22.4.216 ROUND THE CORNER Ketamine: a novel antidepressant with a fast onset of action?† COMMENTARY ON… COCHRANE CORNER Katharine Smith & Mary Jane Attenburrow Katharine Smith is an honorary For bipolar depression, the treatment challenge SUMMARY consultant psychiatrist at the is even greater. There is scarce evidence that National Institute for Health Glutamate receptor modulators, including antidepressants are effective as a first-line Research (NIHR) Oxford cognitive ketamine, are possible candidates for new health Clinical Research Facility treatment (Sidor 2012), and mood instability is an antidepressants with a novel mode of action. and Oxford University Department additional challenge. Current recommendations The pair of reviews discussed in this month’s of Psychiatry. Mary Jane (Kendall 2014) focus on the use of quetiapine; Attenburrow is Clinical Lead at Round the Corner considered their use in treating the NIHR Oxford cognitive health unipolar major depression and bipolar depression. lamotrigine, fluoxetine plus olanzapine and Clinical Research Facility, a Senior Promising results were seen for ketamine, but lithium are also recommended. A recent trial Research Fellow at Oxford University further studies are needed, in particular to (CEQUEL) demonst ra ted that there may be benefit Department of Psychiatry and an investigate whether the benefits are sustained in the combination of lamotrigine with quetiapine honorary consultant psychiatrist. Correspondence Dr Katharine or can be extended by repeated or adjunctive (Geddes 2016). Smith, NIHR Oxford cognitive health treatment, whether ketamine is effective in Although the differences in treatment response Clinical Research Facility, Warneford treatment resistance, whether other modes of between unipolar and bipolar depression Hospital, Oxford OX3 7JX, UK.
    [Show full text]
  • Traxoprodil, a Selective Antagonist of the NR2B Subunit of the NMDA
    Metab Brain Dis (2016) 31:803–814 DOI 10.1007/s11011-016-9810-5 ORIGINAL ARTICLE Traxoprodil, a selective antagonist of the NR2B subunit of the NMDA receptor, potentiates the antidepressant-like effects of certain antidepressant drugs in the forced swim test in mice Ewa Poleszak1 & Weronika Stasiuk 2 & Aleksandra Szopa1 & Elżbieta Wyska3 & Anna Serefko1 & Anna Oniszczuk4 & Sylwia Wośko 1 & Katarzyna Świąder1 & Piotr Wlaź5 Received: 25 November 2015 /Accepted: 17 February 2016 /Published online: 29 February 2016 # The Author(s) 2016. This article is published with open access at Springerlink.com Abstract One of the newest substances, whose antidepres- these changes were not due to the severity of locomotor ac- sant activity was shown is traxoprodil, which is a selective tivity. The observed effect of traxoprodil is only partially as- antagonist of the NR2B subunit of the NMDA receptor. The sociated with serotonergic system and is independent of the main goal of the present study was to evaluate the effect of effect on the 5-HT1A and 5-HT2 serotonin receptors. The re- traxoprodil on animals’ behavior using the forced swim test sults of an attempt to assess the nature of the interaction be- (FST), as well as the effect of traxoprodil (10 mg/kg) on the tween traxoprodil and the tested drugs show that in the case of activity of antidepressants, such as imipramine (15 mg/kg), joint administration of traxoprodil and fluoxetine, imip- fluoxetine (5 mg/kg), escitalopram (2 mg/kg) and reboxetine ramine or escitalopram, there were interactions in the (2.5 mg/kg). Serotonergic lesion and experiment using the pharmacokinetic phase.
    [Show full text]
  • NMDA) Receptor Antagonist
    White Paper The Use of Robust Biomarkers in the Early Clinical Development of N-methyl-D-aspartate (NMDA) Receptor Antagonist Biotrial has extensive early clinical development experience with a high output of over 20 CNS studies per year. The experience and capabilities of Biotrial Neuroscience (now also incorporating the know-how of the legacy CNS CRO Forenap), enables the in-depth cognitive and neurophysiological assessment of NMDA antagonists. The aim of the present document is twofold. Firstly, to characterize the pharmacological effects of non-competitive NMDA antagonists in psychiatry and pain indications and to validate the use of ketamine as a positive control in the development of these compounds with this mechanism of action. Secondly, to present Biotrial’s strategy and rationale for the integration of surrogate endpoints, biomarkers and PD models into a Phase I program, in order to establish early proof of concept of new NMDA antagonist candidates. Pharmacological Effects of Non-Competitive NMDA Antagonists: The Example of Ketamine Glutamatergic antagonists, such as ketamine, acting at the NMDA open channel have a long history of use in neuropsychiatry – independently of their anesthetic or pain attenuating properties. The mechanism of action and pharmacological effects of ketamine have been extensively studied over the last 20 years. Ketamine was initially profiled as a reversible model of psychosis1, producing positive and negative symptoms of schizophrenia as well as cognitive impairment. As of today there are approximately one hundred publications on the symptoms, brain connectivity, imaging and cognitive function alterations induced by ketamine. The main hypothesis to explain the effect of ketamine was the reduction of the glutamatergic burst in the prefrontal cortex, secondarily enhancing AMPA α-(amino- 3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor) signaling.
    [Show full text]