1
|
McCullock TW, Couch T, Kammermeier PJ. Unique pharmacology of mGlu homo- and heterodimers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.623856. [PMID: 39605629 PMCID: PMC11601466 DOI: 10.1101/2024.11.18.623856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background and Purpose Metabotropic glutamate receptors (mGlus) are obligate dimer G protein coupled receptors that can all homodimerize and heterodimerize in select combinations. Responses of mGlu heterodimers to selective ligands, including orthosteric agonists and allosteric modulators, are largely unknown. Experimental Approach The pharmacological properties of each group II and III mGlu homodimer (except mGlu6) and several heterodimers were examined when stochastically assembled in HEK293T cells, or specifically measured using an improved G protein mediated BRET assay employing complimented fragments of NanoLuciferase. Results Stochastically assembled receptors adopted unique signaling characteristics. Some favored the potency, efficacy or signaling kinetics of a dominant subunit, while others exhibited blended profiles reflective of a combination of homo- and heterodimers at various ratios of expressed receptor. Finally, group II and III mGlu dimers were examined for responses to selective agonists and allosteric modulators. Effects of glutamate and selective group II and III orthosteric agonists were found to result in unique concentration response profiles when examining each combination of group II and II mGlu. Effects of select allosteric modulators were examined for each mGlu2 containing dimer as well as several group III dimer pairs. Likewise, allosteric modulator effects were often unique across dimers containing the targeted subunit of the ligand being tested. Conclusions Results demonstrate that mGlu dimers respond uniquely to selective ligands, and show that the mGlu family is not governed by generalizable rules dictating consequences of dimeric subunit interactions leading to signaling consequences.
Collapse
|
2
|
Czapińska-Ciepiela EK, Łuszczki J, Czapiński P, Czuczwar SJ, Lasoń W. Presynaptic antiseizure medications - basic mechanisms and clues for their rational combinations. Pharmacol Rep 2024; 76:623-643. [PMID: 38776036 PMCID: PMC11294404 DOI: 10.1007/s43440-024-00603-7] [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: 02/22/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 08/02/2024]
Abstract
Among clinically highly efficient antiseizure medications (ASMs) there are modifiers of the presynaptic release machinery. Of them, levetiracetam and brivaracetam show a high affinity to the synaptic vesicle protein type 2 A (SV2A), whereas pregabalin and gabapentin are selective ligands for the α2δ1 subunits of the voltage-gated calcium channels. In this paper, we present recent progress in understanding the significance of presynaptic release machinery in the neurochemical mechanisms of epilepsy and ASMs. Furthermore, we discuss whether the knowledge of the basic mechanisms of the presynaptically acting ASMs might help establish a rational polytherapy for drug-resistant epilepsy.
Collapse
Affiliation(s)
| | - Jarogniew Łuszczki
- Department of Occupational Medicine, Medical University of Lublin, 20-090, Lublin, Poland
| | - Piotr Czapiński
- Epilepsy and Migraine Treatment Center, 31-209, Kraków, Poland
| | - Stanisław J Czuczwar
- Department of Pathophysiology, Medical University of Lublin, 20-090, Lublin, Poland
| | - Władysław Lasoń
- Maj Institute of Pharmacology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, 31-343, Kraków, Poland.
| |
Collapse
|
3
|
Metcalf CS, Gagangras S, Bulaj G, White HS. Synergistic effects of the galanin analog 810-2 with the antiseizure medication levetiracetam in rodent seizure models. Epilepsia 2022; 63:3090-3099. [PMID: 36177529 DOI: 10.1111/epi.17420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The use of many antiseizure medications (ASMs) is limited due to pharmacoresistance and dose-limiting side effects, suggesting an unmet need for novel therapeutic approaches. The neuropeptide galanin reduces seizures in several preclinical seizure and epilepsy models, but its clinical utility is limited due to rapid metabolism and poor blood-brain barrier penetration. The lead galanin analog 810-2 is systemically bioavailable and reduces seizures when administered alone. Further development of this analog, with the potential for use as an add-on therapy in patients with epilepsy, requires a better understanding of the use of this analog in combination with approved ASMs. We sought to evaluate 810-2 in combination with commonly used ASMs in rodent models of seizures. METHODS The mouse 6-Hz seizure assay was used to test efficacy of 810-2 in combination with levetiracetam (LEV), valproic acid (VPA), or lacosamide (LCM) using a 1:1 dose ratio in isobolographic studies. Further characterization was performed for the combination of 810-2 and LEV in the mouse corneal kindling and rat 6-Hz assays. RESULTS Whereas the combination of 810-2 with VPA and LCM yielded additive interactions, the combination of 810-2 with LEV demonstrated a synergistic interaction in the mouse 6-Hz assay. Supra-additive effects were also observed in the mouse corneal kindling and rat 6-Hz assays for this combination. SIGNIFICANCE The combination of 810-2 with LEV suggests the potential for this galanin analog to be further developed as an add-on therapy for patients with epilepsy, particularly when coadministered with LEV.
Collapse
Affiliation(s)
- Cameron S Metcalf
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Saurabh Gagangras
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Grzegorz Bulaj
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - H Steve White
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
4
|
Callis TB, Garrett TR, Montgomery AP, Danon JJ, Kassiou M. Recent Scaffold Hopping Applications in Central Nervous System Drug Discovery. J Med Chem 2022; 65:13483-13504. [PMID: 36206553 DOI: 10.1021/acs.jmedchem.2c00969] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The concept of bioisosterism and the implementation of bioisosteric replacement is fundamental to medicinal chemistry. The exploration of bioisosteres is often used to probe key structural features of candidate pharmacophores and enhance pharmacokinetic properties. As the understanding of bioisosterism has evolved, capabilities to undertake more ambitious bioisosteric replacements have emerged. Scaffold hopping is a broadly used term in the literature referring to a variety of different bioisosteric replacement strategies, ranging from simple heterocyclic replacements to topological structural overhauls. In this work, we have highlighted recent applications of scaffold hopping in the central nervous system drug discovery space. While we have highlighted the benefits of using scaffold hopping approaches in central nervous system drug discovery, these are also widely applicable to other medicinal chemistry fields. We also recommend a shift toward the use of more refined and meaningful terminology within the realm of scaffold hopping.
Collapse
Affiliation(s)
- Timothy B Callis
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Taylor R Garrett
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Jonathan J Danon
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
5
|
Elkommos S, Mula M. Current and future pharmacotherapy options for drug-resistant epilepsy. Expert Opin Pharmacother 2022; 23:2023-2034. [PMID: 36154780 DOI: 10.1080/14656566.2022.2128670] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Epilepsy is one of the most common and serious neurological conditions, affecting over 70 million individuals worldwide and despite advances in treatment, the proportion of drug-resistant patients has remained largely unchanged. AREAS COVERED The present paper reviews current and future (under preclinical and clinical development) pharmacotherapy options for the treatment of drug-resistant focal and generalized epilepsies. EXPERT OPINION Current pharmacotherapy options for drug-resistant epilepsy include perampanel, brivaracetam and the newly approved cenobamate for focal epilepsies; cannabidiol (Epidiolex) for Lennox-Gastaut Syndrome (LGS), Dravet and Tuberous Sclerosis Complex (TSC); fenfluramine for Dravet syndrome and ganaxolone for seizures in Cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder. Many compounds are under clinical development and may hold promise for future pharmacotherapies. For adult focal epilepsies, padsevonil and carisbamate are at a more advanced Phase III stage of clinical development followed by compounds at Phase II like selurampanel, XEN1101 and JNJ-40411813. For specific epilepsy syndromes, XEN 496 is under Phase III development for potassium voltage-gated channel subfamily Q member 2 developmental and epileptic encephalopathy (KCNQ2-DEE), carisbamate is under Phase III development for LGS and Ganaxolone under Phase III development for TSC. Finally, in preclinical models several molecular targets including inhibition of glycolysis, neuroinflammation and sodium channel inhibition have been identified in animal models although further data in animal and later human studies are needed.
Collapse
Affiliation(s)
- Samia Elkommos
- School of Neuroscience, King's College London, United Kingdom.,Atkinson Morley Regional Neurosciences Centre, St George's University Hospitals, United Kingdom
| | - Marco Mula
- Atkinson Morley Regional Neurosciences Centre, St George's University Hospitals, United Kingdom.,Institute of Medical and Biomedical Education, St George's University London, United Kingdom
| |
Collapse
|
6
|
Pong AW, Ross J, Tyrlikova I, Giermek AJ, Kohli MP, Khan YA, Salgado RD, Klein P. Epilepsy: expert opinion on emerging drugs in phase 2/3 clinical trials. Expert Opin Emerg Drugs 2022; 27:75-90. [PMID: 35341431 DOI: 10.1080/14728214.2022.2059464] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/25/2022] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Despite the existence of over 30 anti-seizure medications (ASM), including 20 over the last 30 years, a third of patients with epilepsy remain refractory to treatment, with no disease-modifying or preventive therapies until very recently. The development of new ASMs with new mechanisms of action is therefore critical. Recent clinical trials of new treatments have shifted focus from traditional common epilepsies to rare, genetic epilepsies with known mechanistic targets for treatment and disease-specific animal models. AREAS COVERED ASMs in phase 2a/b-3 clinical trials target cholesterol, serotonin, sigma-1 receptors, potassium channels and metabotropic glutamate receptors. Neuroinflammation, protein misfolding, abnormal thalamocortical firing, and molecular deficiencies are among the targeted pathways. Clinically, the current phase 2a/b-3 agents hold promise for variety of epilepsy conditions, from developmental epileptic encephalopathies (Dravet Syndrome, Lennox-Gastaut syndrome, CDKL5 and PCDH19, Rett's Syndrome), infantile spasms, tuberous sclerosis as well as focal and idiopathic generalized epilepsies and acute rescue therapy for cluster seizures. EXPERT OPINION New delivery mechanisms increase potency and site-specificity of existing drugs. Novel mechanisms of action involve cholesterol degradation, mitochondrial pathways, anti-inflammation, and neuro-regeneration. Earlier identification of genetic conditions through genetic testing will allow for earlier use of disease specific and disease-modifying therapies.
Collapse
Affiliation(s)
- Amanda W Pong
- Mid-Atlantic Epilepsy and Sleep Center, Comprehensive Neurology Clinics of Bethesda, Bethesda, MD, USA
| | - Jonathan Ross
- Mid-Atlantic Epilepsy and Sleep Center, Comprehensive Neurology Clinics of Bethesda, Bethesda, MD, USA
| | - Ivana Tyrlikova
- Mid-Atlantic Epilepsy and Sleep Center, Comprehensive Neurology Clinics of Bethesda, Bethesda, MD, USA
| | - Alexander J Giermek
- Mid-Atlantic Epilepsy and Sleep Center, Comprehensive Neurology Clinics of Bethesda, Bethesda, MD, USA
| | - Maya P Kohli
- Mid-Atlantic Epilepsy and Sleep Center, Comprehensive Neurology Clinics of Bethesda, Bethesda, MD, USA
| | - Yousef A Khan
- Mid-Atlantic Epilepsy and Sleep Center, Comprehensive Neurology Clinics of Bethesda, Bethesda, MD, USA
| | - Roger D Salgado
- Mid-Atlantic Epilepsy and Sleep Center, Comprehensive Neurology Clinics of Bethesda, Bethesda, MD, USA
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Comprehensive Neurology Clinics of Bethesda, Bethesda, MD, USA
| |
Collapse
|
7
|
Metcalf CS, Vanegas F, Underwood T, Johnson K, West PJ, Smith MD, Wilcox KS. Screening of prototype antiseizure and anti-inflammatory compounds in the Theiler's murine encephalomyelitis virus model of epilepsy. Epilepsia Open 2021; 7:46-58. [PMID: 34668659 PMCID: PMC8886069 DOI: 10.1002/epi4.12550] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Infection with Theiler's murine encephalomyelitis virus (TMEV) in C57Bl/6J mice results in handling-induced seizures and is useful for evaluating compounds effective against infection-induced seizures. However, to date only a few compounds have been evaluated in this model, and a comprehensive study of antiseizure medications (ASMs) has not yet been performed. Furthermore, as the TMEV infection produces marked neuroinflammation, an evaluation of prototype anti-inflammatory compounds is needed as well. METHODS Male C57Bl/6J mice were inoculated with TMEV (day 0) followed by daily administrations of test compounds (day 3-7) and subsequent handling sessions (day 3-7). Doses of ASMs, comprising several mechanistic classes, were selected based on previously published data demonstrating the effect of these compounds in reducing seizures in the 6 Hz model of pharmacoresistant seizures. Doses of anti-inflammatory compounds, comprising several mechanistic classes, were selected based on published evidence of reduction of inflammation or inflammation-related endpoints. RESULTS Several prototype ASMs reduced acute seizures following TMEV infection: lacosamide, phenytoin, ezogabine, phenobarbital, tiagabine, gabapentin, levetiracetam, topiramate, and sodium valproate. Of these, phenobarbital and sodium valproate had the greatest effect (>95% seizure burden reduction). Prototype anti-inflammatory drugs celecoxib, dexamethasone, and prednisone also moderately reduced seizure burden. SIGNIFICANCE The TMEV model is utilized by the Epilepsy Therapy Screening Program (ETSP) as a tool for evaluation of novel compounds. Compounds reducing seizures in the TMEV comprise distinct mechanistic classes, some with mechanisms of action that extend beyond traditional ASMs.
Collapse
Affiliation(s)
- Cameron S Metcalf
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Fabiola Vanegas
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Tristan Underwood
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Kristina Johnson
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Peter J West
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Misty D Smith
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA.,School of Dentistry, University of Utah, Salt Lake City, Utah, USA
| | - Karen S Wilcox
- Epilepsy Therapy Screening Program Contract Site, Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
8
|
Lou S, Cui S. Drug treatment of epilepsy: From serendipitous discovery to evolutionary mechanisms. Curr Med Chem 2021; 29:3366-3391. [PMID: 34514980 DOI: 10.2174/0929867328666210910124727] [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] [Received: 05/07/2021] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 11/22/2022]
Abstract
Epilepsy is a chronic brain disorder caused by abnormal firing of neurons. Up to now, using antiepileptic drugs is the main method of epilepsy treatment. The development of antiepileptic drugs lasted for centuries. In general, most agents entering clinical practice act on the balance mechanisms of brain "excitability-inhibition". More specifically, they target voltage-gated ion channels, GABAergic transmission and glutamatergic transmission. In recent years, some novel drugs representing new mechanisms of action have been discovered. Although there are about 30 available drugs in the market, it is still in urgent need of discovering more effective and safer drugs. The development of new antiepileptic drugs is into a new era: from serendipitous discovery to evolutionary mechanism-based design. This article presents an overview of drug treatment of epilepsy, including a series of traditional and novel drugs.
Collapse
Affiliation(s)
- Shengying Lou
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou. China
| | - Sunliang Cui
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou. China
| |
Collapse
|
9
|
de Lucas AI, Vega JA, García Molina A, Linares ML, Tresadern G, Lavreysen H, Oehlrich D, Trabanco AA, Cid JM. Scaffold Hopping to Imidazo[1,2- a]pyrazin-8-one Positive Allosteric Modulators of Metabotropic Glutamate 2 Receptor. ACS OMEGA 2021; 6:22997-23006. [PMID: 34514269 PMCID: PMC8427794 DOI: 10.1021/acsomega.1c03739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/10/2021] [Indexed: 06/13/2023]
Abstract
Glutamate hyperfunction is implicated in multiple neurological and psychiatric diseases. Activation of the mGlu2 receptor results in reduced glutamate release and decreased excitability representing a promising novel therapeutic agent for the treatment of disorders such as epilepsy, schizophrenia, mood, anxiety, and other neuropsychiatric disorders. We have previously reported substantial efforts leading to potent and selective mGlu2 PAMs from different chemical series. Herein, the discovery and optimization of a novel series of imidazopyrazinone mGlu2 PAMs are reported. This new scaffold originated from computational searching of fragment databases and comparison with our previously explored scaffolds. Optimization guided by our robust understanding of SAR from former series led to potent, selective, and brain-penetrant compounds.
Collapse
Affiliation(s)
- Ana I. de Lucas
- Discovery
Chemistry, Discovery Sciences, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - Juan A. Vega
- Discovery
Chemistry, Discovery Sciences, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - Aránzazu García Molina
- Discovery
Chemistry, Discovery Sciences, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - María Lourdes Linares
- Discovery
Chemistry, Discovery Sciences, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - Gary Tresadern
- Computational
Chemistry, Discovery Sciences, Janssen Research & Development, Division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Hilde Lavreysen
- Clinical
Research and Development, Janssen Pharmaceutica
N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Daniel Oehlrich
- Discovery
Chemistry, Discovery Sciences, Janssen Research & Development, Division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Andrés A. Trabanco
- Discovery
Chemistry, Discovery Sciences, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - José M. Cid
- Discovery
Chemistry, Discovery Sciences, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| |
Collapse
|
10
|
Alcoreza OB, Patel DC, Tewari BP, Sontheimer H. Dysregulation of Ambient Glutamate and Glutamate Receptors in Epilepsy: An Astrocytic Perspective. Front Neurol 2021; 12:652159. [PMID: 33828523 PMCID: PMC8019783 DOI: 10.3389/fneur.2021.652159] [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] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
Given the important functions that glutamate serves in excitatory neurotransmission, understanding the regulation of glutamate in physiological and pathological states is critical to devising novel therapies to treat epilepsy. Exclusive expression of pyruvate carboxylase and glutamine synthetase in astrocytes positions astrocytes as essential regulators of glutamate in the central nervous system (CNS). Additionally, astrocytes can significantly alter the volume of the extracellular space (ECS) in the CNS due to their expression of the bi-directional water channel, aquaporin-4, which are enriched at perivascular endfeet. Rapid ECS shrinkage has been observed following epileptiform activity and can inherently concentrate ions and neurotransmitters including glutamate. This review highlights our emerging knowledge on the various potential contributions of astrocytes to epilepsy, particularly supporting the notion that astrocytes may be involved in seizure initiation via failure of homeostatic responses that lead to increased ambient glutamate. We also review the mechanisms whereby ambient glutamate can influence neuronal excitability, including via generation of the glutamate receptor subunit GluN2B-mediated slow inward currents, as well as indirectly affect neuronal excitability via actions on metabotropic glutamate receptors that can potentiate GluN2B currents and influence neuronal glutamate release probabilities. Additionally, we discuss evidence for upregulation of System x c - , a cystine/glutamate antiporter expressed on astrocytes, in epileptic tissue and changes in expression patterns of glutamate receptors.
Collapse
Affiliation(s)
- Oscar B Alcoreza
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States.,School of Medicine, Virginia Tech Carilion, Roanoke, VA, United States.,Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA, United States
| | - Dipan C Patel
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States
| | - Bhanu P Tewari
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States
| | - Harald Sontheimer
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, United States
| |
Collapse
|
11
|
Gregory KJ, Goudet C. International Union of Basic and Clinical Pharmacology. CXI. Pharmacology, Signaling, and Physiology of Metabotropic Glutamate Receptors. Pharmacol Rev 2020; 73:521-569. [PMID: 33361406 DOI: 10.1124/pr.119.019133] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors respond to glutamate, the major excitatory neurotransmitter in the mammalian brain, mediating a modulatory role that is critical for higher-order brain functions such as learning and memory. Since the first mGlu receptor was cloned in 1992, eight subtypes have been identified along with many isoforms and splice variants. The mGlu receptors are transmembrane-spanning proteins belonging to the class C G protein-coupled receptor family and represent attractive targets for a multitude of central nervous system disorders. Concerted drug discovery efforts over the past three decades have yielded a wealth of pharmacological tools including subtype-selective agents that competitively block or mimic the actions of glutamate or act allosterically via distinct sites to enhance or inhibit receptor activity. Herein, we review the physiologic and pathophysiological roles for individual mGlu receptor subtypes including the pleiotropic nature of intracellular signal transduction arising from each. We provide a comprehensive analysis of the in vitro and in vivo pharmacological properties of prototypical and commercially available orthosteric agonists and antagonists as well as allosteric modulators, including ligands that have entered clinical trials. Finally, we highlight emerging areas of research that hold promise to facilitate rational design of highly selective mGlu receptor-targeting therapeutics in the future. SIGNIFICANCE STATEMENT: The metabotropic glutamate receptors are attractive therapeutic targets for a range of psychiatric and neurological disorders. Over the past three decades, intense discovery efforts have yielded diverse pharmacological tools acting either competitively or allosterically, which have enabled dissection of fundamental biological process modulated by metabotropic glutamate receptors and established proof of concept for many therapeutic indications. We review metabotropic glutamate receptor molecular pharmacology and highlight emerging areas that are offering new avenues to selectively modulate neurotransmission.
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| | - Cyril Goudet
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| |
Collapse
|
12
|
Bialer M, Johannessen SI, Koepp MJ, Levy RH, Perucca E, Perucca P, Tomson T, White HS. Progress report on new antiepileptic drugs: A summary of the Fifteenth Eilat Conference on New Antiepileptic Drugs and Devices (EILAT XV). I. Drugs in preclinical and early clinical development. Epilepsia 2020; 61:2340-2364. [DOI: 10.1111/epi.16725] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Meir Bialer
- Faculty of Medicine School of Pharmacy and David R. Bloom Center for Pharmacy Institute for Drug Research Hebrew University of Jerusalem Jerusalem Israel
| | - Svein I. Johannessen
- National Center for Epilepsy Sandvika Norway
- Department of Pharmacology Oslo University Hospital Oslo Norway
| | - Matthias J. Koepp
- Department of Clinical and Experimental Epilepsy UCL Institute of Neurology London UK
| | - René H. Levy
- Department of Pharmaceutics and Neurological Surgery University of Washington Seattle WA USA
| | - Emilio Perucca
- Department of Internal Medicine and Therapeutics University of Pavia Pavia Italy
- IRCCS Mondino Foundation (member of the ERN EpiCARE) Pavia Italy
| | - Piero Perucca
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria Australia
- Departments of Medicine and Neurology Royal Melbourne Hospital University of Melbourne Melbourne Victoria Australia
- Department of Neurology Alfred Health Melbourne Victoria Australia
| | - Torbjörn Tomson
- Department of Clinical Neuroscience Karolinska Institute Stockholm Sweden
| | - H. Steve White
- Department of Pharmacy School of Pharmacy University of Washington Seattle WA USA
| |
Collapse
|
13
|
de Lucas AI, Vega JA, Matesanz E, Linares ML, García Molina A, Tresadern G, Lavreysen H, Trabanco AA, Cid JM. Spiro-oxindole Piperidines and 3-(Azetidin-3-yl)-1 H-benzimidazol-2-ones as mGlu 2 Receptor PAMs. ACS Med Chem Lett 2020; 11:303-308. [PMID: 32184961 DOI: 10.1021/acsmedchemlett.9b00350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/10/2019] [Indexed: 11/29/2022] Open
Abstract
Starting from two weak mGlu2 receptor positive allosteric modulator (PAM) HTS hits (4 and 5), a molecular hybridization strategy resulted in the identification of a novel spiro-oxindole piperidine series with improved activity and metabolic stability. Scaffold hopping around the spiro-oxindole core identified the 3-(azetidin-3-yl)-1H-benzimidazol-2-one as bioisoster. Medicinal chemistry optimization of these two novel chemotypes resulted in the identification of potent, selective, orally bioavailable, and brain penetrant mGluR2 PAMs.
Collapse
Affiliation(s)
- Ana Isabel de Lucas
- Discovery Chemistry, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - Juan Antonio Vega
- Discovery Chemistry, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - Encarnación Matesanz
- Discovery Chemistry, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - María Lourdes Linares
- Discovery Chemistry, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - Aránzazu García Molina
- Discovery Chemistry, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - Gary Tresadern
- Computational Chemistry, Discovery Neuroscience, Janssen Research & Development, Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Hilde Lavreysen
- Discovery Neuroscience, Janssen Research & Development, Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Andrés A. Trabanco
- Discovery Chemistry, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| | - José María Cid
- Discovery Chemistry, Janssen Research & Development, Division of Janssen-Cilag S.A., Jarama 75A, Toledo 45007, Spain
| |
Collapse
|
14
|
Molosh AI, Dustrude ET, Lukkes JL, Fitz SD, Caliman IF, Abreu ARR, Dietrich AD, Truitt WA, Ver Donck L, Ceusters M, Kent JM, Johnson PL, Shekhar A. Panic results in unique molecular and network changes in the amygdala that facilitate fear responses. Mol Psychiatry 2020; 25:442-460. [PMID: 30108314 PMCID: PMC6410355 DOI: 10.1038/s41380-018-0119-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 04/03/2018] [Accepted: 05/25/2018] [Indexed: 11/12/2022]
Abstract
Recurrent panic attacks (PAs) are a common feature of panic disorder (PD) and post-traumatic stress disorder (PTSD). Several distinct brain regions are involved in the regulation of panic responses, such as perifornical hypothalamus (PeF), periaqueductal gray, amygdala and frontal cortex. We have previously shown that inhibition of GABA synthesis in the PeF produces panic-vulnerable rats. Here, we investigate the mechanisms by which a panic-vulnerable state could lead to persistent fear. We first show that optogenetic activation of glutamatergic terminals from the PeF to the basolateral amygdala (BLA) enhanced the acquisition, delayed the extinction and induced the persistence of fear responses 3 weeks later, confirming a functional PeF-amygdala pathway involved in fear learning. Similar to optogenetic activation of PeF, panic-prone rats also exhibited delayed extinction. Next, we demonstrate that panic-prone rats had altered inhibitory and enhanced excitatory synaptic transmission of the principal neurons, and reduced protein levels of metabotropic glutamate type 2 receptor (mGluR2) in the BLA. Application of an mGluR2-positive allosteric modulator (PAM) reduced glutamate neurotransmission in the BLA slices from panic-prone rats. Treating panic-prone rats with mGluR2 PAM blocked sodium lactate (NaLac)-induced panic responses and normalized fear extinction deficits. Finally, in a subset of patients with comorbid PD, treatment with mGluR2 PAM resulted in complete remission of panic symptoms. These data demonstrate that a panic-prone state leads to specific reduction in mGluR2 function within the amygdala network and facilitates fear, and mGluR2 PAMs could be a targeted treatment for panic symptoms in PD and PTSD patients.
Collapse
Affiliation(s)
- A I Molosh
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Paul and Carol Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - E T Dustrude
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J L Lukkes
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S D Fitz
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - I F Caliman
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A R R Abreu
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A D Dietrich
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - W A Truitt
- Paul and Carol Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - L Ver Donck
- Janssen Research & Development, Beerse, Belgium
| | - M Ceusters
- Janssen Research & Development, Beerse, Belgium
| | - J M Kent
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | - P L Johnson
- Paul and Carol Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Shekhar
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
- Paul and Carol Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Clinical and Translational Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
15
|
Trabanco AA, Bartolomé JM, Cid JM. mGluR2 positive allosteric modulators: an updated patent review (2013-2018). Expert Opin Ther Pat 2019; 29:497-507. [PMID: 31242055 DOI: 10.1080/13543776.2019.1637421] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Positive allosteric modulation of mGlu2 has attracted much interest as an alternative approach to classical orthosteric receptor activation. Two mGlu2 PAMS have advanced into the clinic. The results obtained in schizophrenia and MDD phase 2 clinical trials have tempered the high expectations put on selective mGlu2 receptor activation for treating these conditions; nevertheless, the search for novel therapeutic indications and novel chemotypes continues to be an active field of research. AREAS COVERED 2013-2018 patent literature on mGlu2 receptor PAMs. EXPERT OPINION After a decade of intensive research, the mGlu2 PAM field has seen a deceleration in the last five years. Negative phase 2 schizophrenia clinical trials with JNJ-40411813 and AZD8529 seem to have tempered the high expectations of the scientific community on the utility of mGlu2 PAMs for the treatment of schizophrenia. Nevertheless, novel therapeutic indications continue to be explored and AZD8529 is currently in a phase 2 study for smoking cessation. The advances in medicinal chemistry and in pharmacology, with novel indications such as epilepsy, have set the stage in the field of mGlu2 receptor PAMs. Ongoing preclinical and clinical studies will contribute to define their optimal therapeutic indication and potential to become novel therapeutic agents.
Collapse
Affiliation(s)
- Andrés A Trabanco
- a Discovery Chemistry , Janssen Research and Development, a division of Janssen-Cilag S.A ., Toledo , Spain
| | - José Manuel Bartolomé
- a Discovery Chemistry , Janssen Research and Development, a division of Janssen-Cilag S.A ., Toledo , Spain
| | - José María Cid
- a Discovery Chemistry , Janssen Research and Development, a division of Janssen-Cilag S.A ., Toledo , Spain
| |
Collapse
|
16
|
Celli R, Santolini I, Van Luijtelaar G, Ngomba RT, Bruno V, Nicoletti F. Targeting metabotropic glutamate receptors in the treatment of epilepsy: rationale and current status. Expert Opin Ther Targets 2019; 23:341-351. [PMID: 30801204 DOI: 10.1080/14728222.2019.1586885] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Several drugs targeting the GABAergic system are used in the treatment of epilepsy, but only one drug targeting glutamate receptors is on the market. This is surprising because an imbalance between excitatory and inhibitory neurotransmission lies at the core of the pathophysiology of epilepsy. One possible explanation is that drug development has been directed towards the synthesis of molecules that inhibit the activity of ionotropic glutamate receptors. These receptors mediate fast excitatory synaptic transmission in the central nervous system (CNS) and their blockade may cause severe adverse effects such as sedation, cognitive impairment, and psychotomimetic effects. Metabotropic glutamate (mGlu) receptors are more promising drug targets because these receptors modulate synaptic transmission rather than mediate it. Areas covered: We review the current evidence that links mGlu receptor subtypes to the pathophysiology and experimental treatment of convulsive and absence seizures. Expert opinion: While mGlu5 receptor negative allosteric modulators have the potential to be protective against convulsive seizures and hyperactivity-induced neurodegeneration, drugs that enhance mGlu5 and mGlu7 receptor function may have beneficial effects in the treatment of absence epilepsy. Evidence related to the other mGlu receptor subtypes is more fragmentary; further investigations are required for an improved understanding of their role in the generation and propagation of seizures.
Collapse
Affiliation(s)
| | | | | | | | - Valeria Bruno
- a IRCCS NEUROMED , Pozzilli , Italy.,d Departments of Physiology and Pharmacology , University Sapienza , Rome , Italy
| | - Ferdinando Nicoletti
- a IRCCS NEUROMED , Pozzilli , Italy.,d Departments of Physiology and Pharmacology , University Sapienza , Rome , Italy
| |
Collapse
|
17
|
Bialer M, Johannessen SI, Koepp MJ, Levy RH, Perucca E, Tomson T, White HS. Progress report on new antiepileptic drugs: A summary of the Fourteenth Eilat Conference on New Antiepileptic Drugs and Devices (EILAT XIV). I. Drugs in preclinical and early clinical development. Epilepsia 2018; 59:1811-1841. [DOI: 10.1111/epi.14557] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/08/2018] [Accepted: 08/08/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Meir Bialer
- Faculty of Medicine; School of Pharmacy and David R. Bloom Center for Pharmacy; Institute for Drug Research; Hebrew University of Jerusalem; Jerusalem Israel
| | - Svein I. Johannessen
- National Center for Epilepsy; Sandvika Norway
- Department of Pharmacology; Oslo University Hospital; Oslo Norway
| | - Matthias J. Koepp
- Department of Clinical and Experimental Epilepsy; UCL Institute of Neurology; London UK
| | - René H. Levy
- Departments of Pharmaceutics and Neurological Surgery; University of Washington; Seattle Washington
| | - Emilio Perucca
- Department of Internal Medicine and Therapeutics; University of Pavia; Pavia Italy
- IRCCS Mondino Foundation; Pavia Italy
| | - Torbjörn Tomson
- Department of Clinical Neuroscience; Karolinska Institute; Stockholm Sweden
| | - H. Steve White
- Department of Pharmacy; School of Pharmacy; University of Washington; Seattle Washington
| |
Collapse
|
18
|
Metcalf CS, Klein BD, Smith MD, Ceusters M, Lavreysen H, Pype S, Van Osselaer N, Twyman R, White HS. Potent and selective pharmacodynamic synergy between the metabotropic glutamate receptor subtype 2-positive allosteric modulator JNJ-46356479 and levetiracetam in the mouse 6-Hz (44-mA) model. Epilepsia 2018; 59:724-735. [PMID: 29360159 DOI: 10.1111/epi.14005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2017] [Indexed: 12/25/2022]
Abstract
OBJECTIVE We previously demonstrated that positive allosteric modulators (PAMs) of metabotropic glutamate subtype 2 (mGlu2 ) receptors have potential synergistic interactions with the antiseizure drug levetiracetam (LEV). The present study utilizes isobolographic analysis to evaluate the combined administration of JNJ-46356479, a selective and potent mGlu2 PAM, with LEV as well as sodium valproate (VPA) and lamotrigine (LTG). METHODS The anticonvulsant efficacy of JNJ-46356479 was evaluated in the 6-Hz model of psychomotor seizures in mice. JNJ-46356479 was administered in combination with LEV using 3 fixed dose-ratio treatment groups in the mouse 6-Hz (44-mA) seizure test. The combination of JNJ-46356479 with LEV was also evaluated in the mouse corneal kindling model. The potential interactions of JNJ-46356479 with the antiseizure drugs VPA and LTG were also evaluated using fixed dose-ratio combinations. Plasma levels were obtained for analysis of potential pharmacokinetic interactions for each combination studied in the mouse 6-Hz model. RESULTS JNJ-46356479 was active in the 6-Hz model at both 32-mA and 44-mA stimulus intensities (median effective dose = 2.8 and 10.2 mg/kg, respectively). Using 1:1, 1:3, and 3:1 fixed dose-ratio combinations (LEV:JNJ-46356479), coadministration was significantly more potent than predicted for additive effects, and plasma levels suggest this synergism was not due to pharmacokinetic interactions. Studies in kindled mice further demonstrate the positive pharmacodynamic interaction of LEV with JNJ-46356479. Using 1:1 dose-ratio combinations of JNJ-46356479 with either VPA or LTG, there were no significant differences observed for coadministration. SIGNIFICANCE These studies demonstrate a synergistic interaction of JNJ-46356479 with LEV, whereas no such effect occurred for JNJ-46356479 with either VPA or LTG. The synergy seems therefore to be specific to LEV, and the combination LEV/mGlu2 PAM has the potential to result in a rational polypharmacy approach to treat patients with refractory epilepsy, once it has been confirmed in clinical studies.
Collapse
Affiliation(s)
- Cameron S Metcalf
- NeuroAdjuvants, Salt Lake City, UT, USA.,Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Brian D Klein
- NeuroAdjuvants, Salt Lake City, UT, USA.,Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Misty D Smith
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | | | | | | | - Nancy Van Osselaer
- Janssen Research and Development, Beerse, Belgium.,UCB, Brussels, Belgium
| | - Roy Twyman
- Janssen Research and Development, Titusville, FL, USA
| | - H Steve White
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA.,Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
19
|
Novel Targets for Developing Antiseizure and, Potentially, Antiepileptogenic Drugs. Epilepsy Curr 2017; 17:293-298. [PMID: 29225544 DOI: 10.5698/1535-7597.17.5.293] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Epilepsy is a chronic neurological disorder caused by abnormal changes in the functions of neuronal circuits and manifested by seizures. It affects patients of all age, substantially worsens the quality of life for the patients as well as their families, and imposes a huge economic burden on the healthcare system. Historically, efforts for discovering and developing antiseizure therapies have been focused on modulating the functions of receptors, transporters, and enzymes expressed by neurons. These drug development efforts have paid off, as we have over 25 antiseizure drugs available in the clinic. However, these drugs mainly provide symptomatic relief from seizures and often cause serious adverse effects. Importantly, almost one-third of patients with epilepsy do not have their seizures adequately controlled by available drugs. To address this problem, researchers are investigating cellular and molecular mechanisms fundamental to the optimal function of neuronal circuits. Evidence shows that disruptions in these mechanisms cause impairment in neuroglial interactions, uncontrolled inflammation, aberrant synaptogenesis, and neurodegeneration in genetic and acquired epilepsies. Many novel therapeutic targets have been identified to target these mechanisms for developing new antiseizure drugs. In addition, the field is exploring new drug targets which may impede the development of epilepsy. We have summarized some of these novel targets in this brief review.
Collapse
|