1
|
Dantio CD, Fasoranti DO, Teng C, Li X. Seizures in brain tumors: pathogenesis, risk factors and management (Review). Int J Mol Med 2025; 55:82. [PMID: 40116082 PMCID: PMC11964414 DOI: 10.3892/ijmm.2025.5523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/05/2025] [Indexed: 03/23/2025] Open
Abstract
Seizures in the context of brain tumors are a relatively common symptom, with higher occurrence rates observed in glioneuronal tumors and gliomas. It is a serious burden that can have a significant impact on the quality of life (QoL) of patients and influence the disease's prognosis. Brain tumor‑related epilepsy (BTRE) is a challenging entity because the pathophysiological mechanisms are not fully understood yet. Nonetheless, neuroinflammation is considered to play a pivotal role. Next to neuroinflammation, findings on the pathogenesis of BTRE have established that certain genetic mutations are involved, of which the most known would be IDH mutations in gliomas. Others discussed more thoroughly in the present review include genes such as PTEN, TP53, IGSF3, and these findings all provide fresh and fascinating insights into the pathogenesis of BTRE. Treatment for BTRE presents unique challenges, mainly related to burdens of polytherapy, debated necessity of anti‑epileptic prophylaxis, and overall impact on the QoL. In fact, there are no established anti‑seizure medications (ASMs) of choice for BTRE, nor is there any protocol to guide the use of these medications at every step of disease progression. Treatment strategies aimed at the tumor, that is surgical procedures, radio‑ and chemotherapy appear to influence seizure control. Conversely, some ASMs have also shown antitumor properties. The present review summarizes and retrospectively analyzes the literature on the pathogenesis and management of BTRE to provide an updated comprehensive understanding. Furthermore, the challenges and opportunities for developing future therapies aimed at BTRE are discussed.
Collapse
Affiliation(s)
- Cyrille D. Dantio
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
- Hunan International Scientific and Technological Cooperation, Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Deborah Oluwatosin Fasoranti
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
- Hunan International Scientific and Technological Cooperation, Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Chubei Teng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
- Hunan International Scientific and Technological Cooperation, Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
- Hunan International Scientific and Technological Cooperation, Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
2
|
Albrecht J, Czuczwar SJ, Zielińska M, Miziak B. Methionine Sulfoximine as a Tool for Studying Temporal Lobe Epilepsy: Initiator, Developer, Attenuator. Neurochem Res 2025; 50:84. [PMID: 39843842 DOI: 10.1007/s11064-024-04329-z] [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: 10/15/2024] [Revised: 12/03/2024] [Accepted: 12/27/2024] [Indexed: 01/24/2025]
Abstract
Methionine sulfoximine (MSO) is a compound originally discovered as a byproduct of agene-based milled flour maturation. MSO irreversibly inhibits the astrocytic enzyme glutamine synthase (GS) but also interferes with the transport of glutamine (Gln) and of glutamate (Glu), and γ-aminobutyric acid (GABA) synthesized within the Glu/Gln-GABA cycle, in this way dysregulating neurotransmission balance in favor of excitation. No wonder that intraperitoneal administration of MSO has long been known to induce behavioral and/or electrographic seizures. Recently, a temporal lobe epilepsy (TLE) model based on local continuous infusion of MSO into the hippocampus has been developed reproducing the main features of human mesial TLE: induction of focal seizures, their spreading, increase in intensity over time, and development of spontaneous recurrent seizures. Fully developed TLE in this model is associated with hippocampal degeneration, hallmarked by reactive astrogliosis, and causally related to the concomitant loss of GS-containing astrocytes. By contrast, short-term pre-exposure of rats to relatively low MSO doses that only moderately inhibited GS, attenuated and delayed the initial seizures in the lithium-pilocarpine model of TLE and in other seizure-associated contexts: in the pentylenetetrazole kindling model in rat, and in spontaneously firing or electrically stimulated brain slices. The anti-initial seizure activity of MSO may partly bypass inhibition of GS: the postulated mechanisms include: (i) decreased release of excitatory neurotransmitter Glu, (ii) prevention or diminution of seizure-associated brain edema, (iii) stimulation of glycogenesis, an energy-sparing process; (iv) central or peripheral hypothermia. Further work is needed to verify either of the above mechanisms.
Collapse
Affiliation(s)
- Jan Albrecht
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| | - Stanisław J Czuczwar
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Magdalena Zielińska
- Department of Neurotoxicology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland
| | - Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, 20-090, Lublin, Poland
| |
Collapse
|
3
|
Hameed MQ, D'Ambrosio R, Eastman C, Hui B, Lin R, Vermudez SAD, Liebhardt A, Choe Y, Klein P, Rundfeldt C, Löscher W, Rotenberg A. A comparison of the antiepileptogenic efficacy of two rationally chosen multitargeted drug combinations in a rat model of posttraumatic epilepsy. Exp Neurol 2024; 382:114962. [PMID: 39288831 DOI: 10.1016/j.expneurol.2024.114962] [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: 06/19/2024] [Revised: 08/08/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Post-traumatic epilepsy (PTE) is a recurrent and often drug-refractory seizure disorder caused by traumatic brain injury (TBI). No single drug treatment prevents PTE, but preventive drug combinations that may prophylax against PTE have not been studied. Based on a systematic evaluation of rationally chosen drug combinations in the intrahippocampal kainate (IHK) mouse model of acquired epilepsy, we identified two multi-targeted drug cocktails that exert strong antiepileptogenic effects. The first, a combination of levetiracetam (LEV) and topiramate, only partially prevented spontaneous recurrent seizures in the model. We therefore added atorvastatin (ATV) to the therapeutic cocktail (TC) to increase efficacy, forming "TC-001". The second cocktail - a combination of LEV, ATV, and ceftriaxone, termed "TC-002" - completely prevented epilepsy in the mouse IHK model. In the present proof-of-concept study, we tested whether the two drug cocktails prevent epilepsy in a rat PTE model in which recurrent electrographic seizures develop after severe rostral parasagittal fluid percussion injury (FPI). Following FPI, rats were either treated over 3-4 weeks with vehicle or drug cocktails, starting either 1 or 4-6 h after the injury. Using mouse doses of TC-001 and TC-002, no significant antiepileptogenic effect was obtained in the rat PTE model. However, when using allometric scaling of drug doses to consider the differences in body surface area between mice and rats, PTE was prevented by TC-002. Furthermore, the latter drug cocktail partially prevented the loss of perilesional cortical parvalbumin-positive GABAergic interneurons. Plasma and brain drug analysis showed that these effects of TC-002 occurred at clinically relevant levels of the individual TC-002 drug components. In silico analysis of drug-drug brain protein interactions by the STITCH database indicated that TC-002 impacts a larger functional network of epilepsy-relevant brain proteins than each drug alone, providing a potential network pharmacology explanation for the observed antiepileptogenic and neuroprotective effects observed with this combination.
Collapse
Affiliation(s)
- Mustafa Q Hameed
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raimondo D'Ambrosio
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Cliff Eastman
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Benjamin Hui
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rui Lin
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sheryl Anne D Vermudez
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amanda Liebhardt
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yongho Choe
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pavel Klein
- PrevEp, Inc., Bethesda, MD, USA; Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| | | | - Wolfgang Löscher
- PrevEp, Inc., Bethesda, MD, USA; Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany.
| | - Alexander Rotenberg
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; PrevEp, Inc., Bethesda, MD, USA.
| |
Collapse
|
4
|
Moncion C, West PJ, Metcalf CS. Wnt Signaling: The Next Step on the Road to Anti-Epileptogenic Therapies? Epilepsy Curr 2024; 24:355-357. [PMID: 39508005 PMCID: PMC11536405 DOI: 10.1177/15357597241280467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Canonical Wnt activator Chir99021 prevents epileptogenesis in the intrahippocampal kainate mouse model of temporal lobe epilepsy Mardones MD Rostam KD Nickerson MC Gupta K. Experimental Neurology. 2024 Mar 21;376:114767. The Wnt signaling pathway mediates the development of dentate granule cell neurons in the hippocampus. These neurons are central to the development of temporal lobe epilepsy and undergo structural and physiological remodeling during epileptogenesis, which results in the formation of epileptic circuits. The pathways responsible for granule cell remodeling during epileptogenesis have yet to be well defined, and represent therapeutic targets for the prevention of epilepsy. The current study explores Wnt signaling during epileptogenesis and for the first time describes the effect of Wnt activation using Wnt activator Chir99021 as a novel anti-epileptogenic therapeutic approach. Focal mesial temporal lobe epilepsy was induced by intrahippocampal kainate (IHK) injection in wild-type and POMC-eGFP transgenic mice. Wnt activator Chir99021 was administered daily, beginning 3 h after seizure induction, and continued up to 21-days. Immature granule cell morphology was quantified in the ipsilateral epileptogenic zone and the contralateral peri-ictal zone 14 days after IHK, targeting the end of the latent period. Bilateral hippocampal electrocorticographic recordings were performed for 28-days, 7-days beyond treatment cessation. Hippocampal behavioral tests were performed after the completion of Chir99021 treatment. Consistent with previous studies, IHK resulted in the development of epilepsy after a 14 day latent period in this well-described mouse model. Activation of the canonical Wnt pathway with Chir99021 significantly reduced bilateral hippocampal seizure number and duration. Critically, this effect was retained after treatment cessation, suggesting a durable antiepileptogenic change in epileptic circuitry. Morphological analyses demonstrated that Wnt activation prevented pathological remodeling of the primary dendrite in both the epileptogenic zone and peri-ictal zone, changes in which may serve as a biomarker of epileptogenesis and anti-epileptogenic treatment response in pre-clinical studies. These findings were associated with improved object location memory with Chir99021 treatment after IHK. This study provides novel evidence that canonical Wnt activation prevents epileptogenesis in the IHK mouse model of mesial temporal lobe epilepsy, preventing pathological remodeling of dentate granule cells. Wnt signaling may therefore play a key role in mesial temporal lobe epileptogenesis, and Wnt modulation may represent a novel therapeutic strategy in the prevention of epilepsy.
Collapse
Affiliation(s)
| | - Peter J West
- Department of Pharmacology & Toxicology, University of Utah
| | | |
Collapse
|
5
|
Talevi A, Bellera C. An update on the novel methods for the discovery of antiseizure and antiepileptogenic medications: where are we in 2024? Expert Opin Drug Discov 2024; 19:975-990. [PMID: 38963148 DOI: 10.1080/17460441.2024.2373165] [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: 05/12/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Despite the availability of around 30 antiseizure medications, 1/3 of patients with epilepsy fail to become seizure-free upon pharmacological treatment. Available medications provide adequate symptomatic control in two-thirds of patients, but disease-modifying drugs are still scarce. Recently, though, new paradigms have been explored. AREAS COVERED Three areas are reviewed in which a high degree of innovation in the search for novel antiseizure and antiepileptogenic medications has been implemented: development of novel screening approaches, search for novel therapeutic targets, and adoption of new drug discovery paradigms aligned with a systems pharmacology perspective. EXPERT OPINION In the past, worldwide leaders in epilepsy have reiteratively stated that the lack of progress in the field may be explained by the recurrent use of the same molecular targets and screening procedures to identify novel medications. This landscape has changed recently, as reflected by the new Epilepsy Therapy Screening Program and the introduction of many in vitro and in vivo models that could possibly improve our chances of identifying first-in-class medications that may control drug-resistant epilepsy or modify the course of disease. Other milestones include the study of new molecular targets for disease-modifying drugs and exploration of a systems pharmacology perspective to design new drugs.
Collapse
Affiliation(s)
- Alan Talevi
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, University of La Plata (UNLP), La Plata, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CCT La Plata, La Plata, Argentina
| | - Carolina Bellera
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, University of La Plata (UNLP), La Plata, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CCT La Plata, La Plata, Argentina
| |
Collapse
|
6
|
Pérez-Pérez D, Monío-Baca C, von Rüden EL, Buchecker V, Wagner A, Schönhoff K, Zvejniece L, Klimpel D, Potschka H. Preclinical efficacy profiles of the sigma-1 modulator E1R and of fenfluramine in two chronic mouse epilepsy models. Epilepsia 2024; 65:2470-2482. [PMID: 39119787 DOI: 10.1111/epi.18037] [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: 01/11/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE Given its key homeostatic role affecting mitochondria, ionotropic and metabotropic receptors, and voltage-gated ion channels, sigma-1 receptor (Sig1R) represents an interesting target for epilepsy management. Antiseizure effects of the positive allosteric modulator E1R have already been reported in acute seizure models. Although modulation of serotonergic neurotransmission is considered the main mechanism of action of fenfluramine, its interaction with Sig1R may be of additional relevance. METHODS To further explore the potential of Sig1R as a target, we assessed the efficacy and tolerability of E1R and fenfluramine in two chronic mouse models, including an amygdala kindling paradigm and the intrahippocampal kainate model. The relative contribution of the interaction with Sig1R was analyzed using combination experiments with the Sig1R antagonist NE-100. RESULTS Whereas E1R exerted pronounced dose-dependent antiseizure effects at well-tolerated doses in fully kindled mice, only limited effects were observed in response to fenfluramine, without a clear dose dependency. In the intrahippocampal kainate model, E1R failed to influence electrographic seizure activity. In contrast, fenfluramine significantly reduced the frequency of electrographic seizure events and their cumulative duration. Pretreatment with NE-100 reduced the effects of E1R and fenfluramine in the kindling model. Surprisingly, pre-exposure to NE-100 in the intrahippocampal kainate model rather enhanced and prolonged fenfluramine's antiseizure effects. SIGNIFICANCE In conclusion, the kindling data further support Sig1R as an interesting target for novel antiseizure medications. However, it is necessary to further explore the preclinical profile of E1R in chronic epilepsy models with spontaneous seizures. Despite the rather limited effects in the kindling paradigm, the findings from the intrahippocampal kainate model suggest that it is of interest to further assess a possible broad-spectrum potential of fenfluramine.
Collapse
Affiliation(s)
- Daniel Pérez-Pérez
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Cristina Monío-Baca
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Eva-Lotta von Rüden
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Verena Buchecker
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Amelie Wagner
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katharina Schönhoff
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Liga Zvejniece
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Dennis Klimpel
- Department of Forensic and Clinical Toxicology, Medizinisches Versorgungszentrum Labor Krone, Bad Salzuflen, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
7
|
Liu X, Zhang Y, Zhao Y, Zhang Q, Han F. The Neurovascular Unit Dysfunction in the Molecular Mechanisms of Epileptogenesis and Targeted Therapy. Neurosci Bull 2024; 40:621-634. [PMID: 38564049 PMCID: PMC11127907 DOI: 10.1007/s12264-024-01193-3] [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: 08/04/2023] [Accepted: 12/09/2023] [Indexed: 04/04/2024] Open
Abstract
Epilepsy is a multifaceted neurological syndrome characterized by recurrent, spontaneous, and synchronous seizures. The pathogenesis of epilepsy, known as epileptogenesis, involves intricate changes in neurons, neuroglia, and endothelium, leading to structural and functional disorders within neurovascular units and culminating in the development of spontaneous epilepsy. Although current research on epilepsy treatments primarily centers around anti-seizure drugs, it is imperative to seek effective interventions capable of disrupting epileptogenesis. To this end, a comprehensive exploration of the changes and the molecular mechanisms underlying epileptogenesis holds the promise of identifying vital biomarkers for accurate diagnosis and potential therapeutic targets. Emphasizing early diagnosis and timely intervention is paramount, as it stands to significantly improve patient prognosis and alleviate the socioeconomic burden. In this review, we highlight the changes and molecular mechanisms of the neurovascular unit in epileptogenesis and provide a theoretical basis for identifying biomarkers and drug targets.
Collapse
Affiliation(s)
- Xiuxiu Liu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China.
- International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Ying Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yanming Zhao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Qian Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China.
- International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- Institute of Brain Science, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 211166, China.
- Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 210019, China.
| |
Collapse
|
8
|
Guarnieri L, Amodio N, Bosco F, Carpi S, Tallarico M, Gallelli L, Rania V, Citraro R, Leo A, De Sarro G. Circulating miRNAs as Novel Clinical Biomarkers in Temporal Lobe Epilepsy. Noncoding RNA 2024; 10:18. [PMID: 38525737 PMCID: PMC10961783 DOI: 10.3390/ncrna10020018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024] Open
Abstract
Temporal lobe epilepsy (TLE) represents the most common form of refractory focal epilepsy. The identification of innovative clinical biomarkers capable of categorizing patients with TLE, allowing for improved treatment and outcomes, still represents an unmet need. Circulating microRNAs (c-miRNAs) are short non-coding RNAs detectable in body fluids, which play crucial roles in the regulation of gene expression. Their characteristics, including extracellular stability, detectability through non-invasive methods, and responsiveness to pathological changes and/or therapeutic interventions, make them promising candidate biomarkers in various disease settings. Recent research has investigated c-miRNAs in various bodily fluids, including serum, plasma, and cerebrospinal fluid, of TLE patients. Despite some discrepancies in methodologies, cohort composition, and normalization strategies, a common dysregulated signature of c-miRNAs has emerged across different studies, providing the basis for using c-miRNAs as novel biomarkers for TLE patient management.
Collapse
Affiliation(s)
- Lorenza Guarnieri
- Section of Pharmacology, Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (L.G.); (F.B.); (S.C.); (M.T.); (L.G.); (V.R.); (A.L.); (G.D.S.)
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Francesca Bosco
- Section of Pharmacology, Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (L.G.); (F.B.); (S.C.); (M.T.); (L.G.); (V.R.); (A.L.); (G.D.S.)
| | - Sara Carpi
- Section of Pharmacology, Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (L.G.); (F.B.); (S.C.); (M.T.); (L.G.); (V.R.); (A.L.); (G.D.S.)
| | - Martina Tallarico
- Section of Pharmacology, Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (L.G.); (F.B.); (S.C.); (M.T.); (L.G.); (V.R.); (A.L.); (G.D.S.)
| | - Luca Gallelli
- Section of Pharmacology, Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (L.G.); (F.B.); (S.C.); (M.T.); (L.G.); (V.R.); (A.L.); (G.D.S.)
- Research Center FAS@UMG, Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Vincenzo Rania
- Section of Pharmacology, Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (L.G.); (F.B.); (S.C.); (M.T.); (L.G.); (V.R.); (A.L.); (G.D.S.)
| | - Rita Citraro
- Section of Pharmacology, Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (L.G.); (F.B.); (S.C.); (M.T.); (L.G.); (V.R.); (A.L.); (G.D.S.)
- Research Center FAS@UMG, Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Antonio Leo
- Section of Pharmacology, Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (L.G.); (F.B.); (S.C.); (M.T.); (L.G.); (V.R.); (A.L.); (G.D.S.)
- Research Center FAS@UMG, Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Giovambattista De Sarro
- Section of Pharmacology, Science of Health Department, School of Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (L.G.); (F.B.); (S.C.); (M.T.); (L.G.); (V.R.); (A.L.); (G.D.S.)
- Research Center FAS@UMG, Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
9
|
Talevi A. Computer-Aided Drug Discovery and Design: Recent Advances and Future Prospects. Methods Mol Biol 2024; 2714:1-20. [PMID: 37676590 DOI: 10.1007/978-1-0716-3441-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Computer-aided drug discovery and design involve the use of information technologies to identify and develop, on a rational ground, chemical compounds that align a set of desired physicochemical and biological properties. In its most common form, it involves the identification and/or modification of an active scaffold (or the combination of known active scaffolds), although de novo drug design from scratch is also possible. Traditionally, the drug discovery and design processes have focused on the molecular determinants of the interactions between drug candidates and their known or intended pharmacological target(s). Nevertheless, in modern times, drug discovery and design are conceived as a particularly complex multiparameter optimization task, due to the complicated, often conflicting, property requirements.This chapter provides an updated overview of in silico approaches for identifying active scaffolds and guiding the subsequent optimization process. Recent groundbreaking advances in the field have also analyzed the integration of state-of-the-art machine learning approaches in every step of the drug discovery process (from prediction of target structure to customized molecular docking scoring functions), integration of multilevel omics data, and the use of a diversity of computational approaches to assist target validation and assess plausible binding pockets.
Collapse
Affiliation(s)
- Alan Talevi
- Laboratory of Bioactive Compound Research and Development (LIDeB), Faculty of Exact Sciences, National University of La Plata (UNLP), La Plata, Argentina.
- Argentinean National Council of Scientific and Technical Research (CONICET), La Plata, Argentina.
| |
Collapse
|
10
|
Ots HD, Anderson T, Sherrerd-Smith W, DelBianco J, Rasic G, Chuprin A, Toor Z, Fitch E, Ahuja K, Reid F, Musto AE. Scoping review of disease-modifying effect of drugs in experimental epilepsy. Front Neurol 2023; 14:1097473. [PMID: 36908628 PMCID: PMC9997527 DOI: 10.3389/fneur.2023.1097473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Objective Epilepsy affects ~50 million people worldwide causing significant medical, financial, and sociologic concerns for affected patients and their families. To date, treatment of epilepsy is primarily symptomatic management because few effective preventative or disease-modifying interventions exist. However, recent research has identified neurobiological mechanisms of epileptogenesis, providing new pharmacologic targets to investigate. The current scientific evidence remains scattered across multiple studies using different model and experimental designs. The review compiles different models of anti-epileptogenic investigation and highlights specific compounds with potential epileptogenesis-modifying experimental drugs. It provides a platform for standardization of future epilepsy research to allow a more robust compound analysis of compounds with potential for epilepsy prevention. Methods PubMed, Ovid MEDLINE, and Web of Science were searched from 2007 to 2021. Studies with murine models of epileptogenesis and explicitly detailed experimental procedures were included in the scoping review. In total, 51 articles were selected from 14,983 and then grouped by five core variables: (1) seizure frequency, (2) seizure severity, (3) spontaneous recurrent seizures (SRS), (4) seizure duration, and (5) mossy fiber sprouting (MFS). The variables were differentiated based on experimental models including methods of seizure induction, treatment schedule and timeline of data collection. Data was categorized by the five core variables and analyzed by converting original treatment values to units of percent of its respective control. Results Discrepancies in current epileptogenesis models significantly complicate inter-study comparison of potential anti-epileptogenic interventions. With our analysis, many compounds showed a potential to reduce epileptogenic characteristics defined by the five core variables. WIN55,212-2, aspirin, rapamycin, 1400W, and LEV + BQ788 were identified compounds with the potential of effective anti-epileptic properties. Significance Our review highlights the need for consistent methodology in epilepsy research and provides a novel approach for future research. Inconsistent experimental designs hinder study comparison, slowing the progression of treatments for epilepsy. If the research community can optimize and standardize parameters such as methods of seizure induction, administration schedule, sampling time, and aniMal models, more robust meta-analysis and collaborative research would follow. Additionally, some compounds such as rapamycin, WIN 55,212-2, aspirin, 1400W, and LEV + BQ788 showed anti-epileptogenic modulation across multiple variables. We believe they warrant further study both individually and synergistically.
Collapse
Affiliation(s)
- Heather D. Ots
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Taylor Anderson
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | | | - John DelBianco
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Gordana Rasic
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Anthony Chuprin
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Zeeshan Toor
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Elizabeth Fitch
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Kripa Ahuja
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Faith Reid
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Alberto E. Musto
- Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, United States
- Department of Neurology, Eastern Virginia Medical School, Norfolk, VA, United States
| |
Collapse
|
11
|
Dong X, Fan J, Lin D, Wang X, Kuang H, Gong L, Chen C, Jiang J, Xia N, He D, Shen W, Jiang P, Kuang R, Zeng L, Xie Y. Captopril alleviates epilepsy and cognitive impairment by attenuation of C3-mediated inflammation and synaptic phagocytosis. J Neuroinflammation 2022; 19:226. [PMID: 36104755 PMCID: PMC9476304 DOI: 10.1186/s12974-022-02587-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/04/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractEvidence from experimental and clinical studies implicates immuno-inflammatory responses as playing an important role in epilepsy-induced brain injury. Captopril, an angiotensin-converting enzyme inhibitor (ACEi), has previously been shown to suppress immuno-inflammatory responses in a variety of neurological diseases. However, the therapeutic potential of captopril on epilepsy remains unclear. In the present study, Sprague Dawley (SD) rats were intraperitoneally subjected to kainic acid (KA) to establish a status epilepticus. Captopril (50 mg/kg, i.p.) was administered daily following the KA administration from day 3 to 49. We found that captopril efficiently suppressed the KA-induced epilepsy, as measured by electroencephalography. Moreover, captopril ameliorated the epilepsy-induced cognitive deficits, with improved performance in the Morris water maze, Y-maze and novel objective test. RNA sequencing (RNA-seq) analysis indicated that captopril reversed a wide range of epilepsy-related biological processes, particularly the glial activation, complement system-mediated phagocytosis and the production of inflammatory factors. Interestingly, captopril suppressed the epilepsy-induced activation and abnormal contact between astrocytes and microglia. Immunohistochemical experiments demonstrated that captopril attenuated microglia-dependent synaptic remodeling presumably through C3–C3ar-mediated phagocytosis in the hippocampus. Finally, the above effects of captopril were partially blocked by an intranasal application of recombinant C3a (1.3 μg/kg/day). Our findings demonstrated that captopril reduced the occurrence of epilepsy and cognitive impairment by attenuation of inflammation and C3-mediated synaptic phagocytosis. This approach can easily be adapted to long-term efficacy and safety in clinical practice.
Graphical Abstract
Collapse
|
12
|
Löscher W, Howe CL. Molecular Mechanisms in the Genesis of Seizures and Epilepsy Associated With Viral Infection. Front Mol Neurosci 2022; 15:870868. [PMID: 35615063 PMCID: PMC9125338 DOI: 10.3389/fnmol.2022.870868] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/05/2022] [Indexed: 12/16/2022] Open
Abstract
Seizures are a common presenting symptom during viral infections of the central nervous system (CNS) and can occur during the initial phase of infection ("early" or acute symptomatic seizures), after recovery ("late" or spontaneous seizures, indicating the development of acquired epilepsy), or both. The development of acute and delayed seizures may have shared as well as unique pathogenic mechanisms and prognostic implications. Based on an extensive review of the literature, we present an overview of viruses that are associated with early and late seizures in humans. We then describe potential pathophysiologic mechanisms underlying ictogenesis and epileptogenesis, including routes of neuroinvasion, viral control and clearance, systemic inflammation, alterations of the blood-brain barrier, neuroinflammation, and inflammation-induced molecular reorganization of synapses and neural circuits. We provide clinical and animal model findings to highlight commonalities and differences in these processes across various neurotropic or neuropathogenic viruses, including herpesviruses, SARS-CoV-2, flaviviruses, and picornaviruses. In addition, we extensively review the literature regarding Theiler's murine encephalomyelitis virus (TMEV). This picornavirus, although not pathogenic for humans, is possibly the best-characterized model for understanding the molecular mechanisms that drive seizures, epilepsy, and hippocampal damage during viral infection. An enhanced understanding of these mechanisms derived from the TMEV model may lead to novel therapeutic interventions that interfere with ictogenesis and epileptogenesis, even within non-infectious contexts.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Charles L. Howe
- Division of Experimental Neurology, Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
13
|
Abstract
The brain is a highly energy-demanding organ and requires bioenergetic adaptability to balance normal activity with pathophysiological fuelling of spontaneous recurrent seizures, the hallmark feature of the epilepsies. Recurrent or prolonged seizures have long been known to permanently alter neuronal circuitry and to cause excitotoxic injury and aberrant inflammation. Furthermore, pathological changes in bioenergetics and metabolism are considered downstream consequences of epileptic seizures that begin at the synaptic level. However, as we highlight in this Review, evidence is also emerging that primary derangements in cellular or mitochondrial metabolism can result in seizure genesis and lead to spontaneous recurrent seizures. Basic and translational research indicates that the relationships between brain metabolism and epileptic seizures are complex and bidirectional, producing a vicious cycle that compounds the deleterious consequences of seizures. Metabolism-based treatments such as the high-fat, antiseizure ketogenic diet have become mainstream, and metabolic substrates and enzymes have become attractive molecular targets for seizure prevention and recovery. Moreover, given that metabolism is crucial for epigenetic as well as inflammatory changes, the idea that epileptogenesis can be both negatively and positively influenced by metabolic changes is rapidly gaining ground. Here, we review evidence that supports both pathophysiological and therapeutic roles for brain metabolism in epilepsy.
Collapse
|
14
|
Alan T. Antiseizure medication discovery: Recent and future paradigm shifts. Epilepsia Open 2022; 7 Suppl 1:S133-S141. [PMID: 35090197 PMCID: PMC9340309 DOI: 10.1002/epi4.12581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 11/06/2022] Open
Abstract
Despite the ever-increasing number of available options for the treatment of epilepsies and the remarkable advances on the understanding of their pathophysiology, the proportion of refractory patients has remained approximately unmodified during the last 100 years. How efficient are we translating positive outcomes from basic research to clinical trials and/or the clinical scenario? It is possible that fresh thinking and exploration of new paradigms is required to arrive at truly novel therapeutic solutions, as seemingly proven by recently approved first-in-class antiseizure medications and drug candidates undergoing late clinical trials. Here, the author discusses some approximations in line with the network pharmacology philosophy, which may result in highly innovative (and, hopefully, safer and/or more efficacious) medications for the control of seizures, as embodied with some recent examples in the field, namely tailored multi-target agents and low-affinity ligands.
Collapse
Affiliation(s)
- Talevi Alan
- Laboratory of Bioactive Research and Development (LIDeB), Faculty of exact Sciences, University of La Plata (UNLP), 47 & 15, La Plata (B1900AJK), Buenos Aires, Argentina
| |
Collapse
|
15
|
Guo L, Zhen Q, Zhen X, Cui Z, Jiang C, Zhang Q, Gao K, Luan D, Zhou X. A network pharmacology approach to explore and validate the potential targets of ginsenoside on osteoporosis. Int J Immunopathol Pharmacol 2022; 36:3946320221107239. [PMID: 35791093 PMCID: PMC9272184 DOI: 10.1177/03946320221107239] [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] [Indexed: 11/15/2022] Open
Abstract
Background: Osteoporosis (OP) is determined as a chronic systemic bone disorder to increase the susceptibility to fracture. Ginsenosides have been found the anti-osteoporotic activity of in vivo and in vitro. However, its mechanism remains unknown.Methods: The potential mechanism of ginsenosides in anti-osteoporotic activity was identified by using network phamacology analysis. The active compounds of ginsenosides and their targets associated to OP were retrieved from Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform, Drug Bank, Pharmmapper, and Cytoscape. The Gene ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis target genes were performed in String, Phenopedia, DisGeNET database, and Metascape software. The protein to protein interaction were created by String database and Cytoscape software. The molecular docking was used to investigate the interactions between active coumpounds and potential targets by utilizing SwissDock tool, UCSF Chimera, and Pymol software. Results: A total of eight important active ingredients and 17 potential targets related to OP treatment were subjected to analyze. GO analysis showed the anti-osteoporosis targets of ginsenoside mainly play a role in the response to steroid hormone. KEGG enrichment analysis indicated that ginsenoside treats OP by osteoblast differentiation signal pathway. Lastly, the molecular docking outcomes indicated that ginsenoside rh2 had a good binding ability with four target proteins IL1B, TNF, IFNG, and NFKBIA. Conclusion: IL1B, TNF, IFNG, and NFKBIA are the most important targets and osteoblast differentiation is the most valuable signaling pathways in ginsenoside for the treatment of OP, which might be beneficial to elucidate the mechanism concerned to the action of ginsenoside and might supply a better understanding of its anti-OP effects.
Collapse
Affiliation(s)
- Ling Guo
- Department of Otorhinolaryngology Head and Neck Surgery, 34708Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Qingliu Zhen
- Department of Anesthesiology, 34708Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Xiaoyue Zhen
- Minimally Invasive Urology Center, 34708Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhaoyang Cui
- Department of Otorhinolaryngology Head and Neck Surgery, 34708Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Chao Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, 34708Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Qiang Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, 34708Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Kun Gao
- Department of Otorhinolaryngology Head and Neck Surgery, 34708Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Deheng Luan
- Department of Otorhinolaryngology Head and Neck Surgery, 34708Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Xuanchen Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, 34708Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
16
|
Löscher W. Single-Target Versus Multi-Target Drugs Versus Combinations of Drugs With Multiple Targets: Preclinical and Clinical Evidence for the Treatment or Prevention of Epilepsy. Front Pharmacol 2021; 12:730257. [PMID: 34776956 PMCID: PMC8580162 DOI: 10.3389/fphar.2021.730257] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/04/2021] [Indexed: 01/09/2023] Open
Abstract
Rationally designed multi-target drugs (also termed multimodal drugs, network therapeutics, or designed multiple ligands) have emerged as an attractive drug discovery paradigm in the last 10-20 years, as potential therapeutic solutions for diseases of complex etiology and diseases with significant drug-resistance problems. Such agents that modulate multiple targets simultaneously are developed with the aim of enhancing efficacy or improving safety relative to drugs that address only a single target or to combinations of single-target drugs. Although this strategy has been proposed for epilepsy therapy >25 years ago, to my knowledge, only one antiseizure medication (ASM), padsevonil, has been intentionally developed as a single molecular entity that could target two different mechanisms. This novel drug exhibited promising effects in numerous preclinical models of difficult-to-treat seizures. However, in a recent randomized placebo-controlled phase IIb add-on trial in treatment-resistant focal epilepsy patients, padsevonil did not separate from placebo in its primary endpoints. At about the same time, a novel ASM, cenobamate, exhibited efficacy in several randomized controlled trials in such patients that far surpassed the efficacy of any other of the newer ASMs. Yet, cenobamate was discovered purely by phenotype-based screening and its presumed dual mechanism of action was only described recently. In this review, I will survey the efficacy of single-target vs. multi-target drugs vs. combinations of drugs with multiple targets in the treatment and prevention of epilepsy. Most clinically approved ASMs already act at multiple targets, but it will be important to identify and validate new target combinations that are more effective in drug-resistant epilepsy and eventually may prevent the development or progression of epilepsy.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany, and Center for Systems Neuroscience Hannover, Hannover, Germany
| |
Collapse
|
17
|
Selected Molecular Targets for Antiepileptogenesis. Int J Mol Sci 2021; 22:ijms22189737. [PMID: 34575901 PMCID: PMC8466306 DOI: 10.3390/ijms22189737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 02/07/2023] Open
Abstract
The term epileptogenesis defines the usually durable process of converting normal brain into an epileptic one. The resistance of a significant proportion of patients with epilepsy to the available pharmacotherapy prompted the concept of a causative treatment option consisting in stopping or modifying the progress of epileptogenesis. Most antiepileptic drugs possess only a weak or no antiepileptogenic potential at all, but a few of them appear promising in this regard; these include, for example, eslicarbazepine (a sodium and T-type channel blocker), lamotrigine (a sodium channel blocker and glutamate antagonist) or levetiracetam (a ligand of synaptic vehicle protein SV2A). Among the approved non-antiepileptic drugs, antiepileptogenic potential seems to reside in losartan (a blocker of angiotensin II type 1 receptors), biperiden (an antiparkinsonian drug), nonsteroidal anti-inflammatory drugs, antioxidative drugs and minocycline (a second-generation tetracycline with anti-inflammatory and antioxidant properties). Among other possible antiepileptogenic compounds, antisense nucleotides have been considered, among these an antagomir targeting microRNA-134. The drugs and agents mentioned above have been evaluated in post-status epilepticus models of epileptogenesis, so their preventive efficacy must be verified. Limited clinical data indicate that biperiden in patients with brain injuries is well-tolerated and seems to reduce the incidence of post-traumatic epilepsy. Exceptionally, in this regard, our own original data presented here point to c-Fos as an early seizure duration, but not seizure intensity-related, marker of early epileptogenesis. Further research of reliable markers of early epileptogenesis is definitely needed to improve the process of designing adequate antiepileptogenic therapies.
Collapse
|
18
|
Łukawski K, Czuczwar SJ. Understanding mechanisms of drug resistance in epilepsy and strategies for overcoming it. Expert Opin Drug Metab Toxicol 2021; 17:1075-1090. [PMID: 34310255 DOI: 10.1080/17425255.2021.1959912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The present evidence indicates that approximately 70% of patients with epilepsy can be successfully treated with antiepileptic drugs (AEDs). A significant proportion of patients are not under sufficient control, and pharmacoresistant epilepsy is clearly associated with poor quality of life and increased morbidity and mortality. There is a great need for newer therapeutic options able to reduce the percentage of drug-resistant patients. AREAS COVERED A number of hypotheses trying to explain the development of pharmacoresistance have been put forward. These include: target hypothesis (altered AED targets), transporter (overexpression of brain efflux transporters), pharmacokinetic (overexpression of peripheral efflux transporters in the intestine or kidneys), intrinsic severity (initial high seizure frequency), neural network (aberrant networks), and gene variant hypothesis (genetic polymorphisms). EXPERT OPINION A continuous search for newer AEDs or among non-AEDs (blockers of efflux transporters, interleukin antagonists, cyclooxygenase inhibitors, mTOR inhibitors, angiotensin II receptor antagonists) may provide efficacious drugs for the management of drug-resistant epilepsy. Also, combinations of AEDs exerting synergy in preclinical and clinical studies (for instance, lamotrigine + valproate, levetiracetam + valproate, topiramate + carbamazepine) might be of importance in this respect. Preclinically antagonistic combinations must be avoided (lamotrigine + carbamazepine, lamotrigine + oxcarbazepine).
Collapse
Affiliation(s)
- Krzysztof Łukawski
- Department of Physiopathology, Institute of Rural Health, Lublin, Poland.,Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | | |
Collapse
|
19
|
Scopolamine prevents aberrant mossy fiber sprouting and facilitates remission of epilepsy after brain injury. Neurobiol Dis 2021; 158:105446. [PMID: 34280524 DOI: 10.1016/j.nbd.2021.105446] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/01/2021] [Accepted: 07/13/2021] [Indexed: 11/21/2022] Open
Abstract
Prevention or modification of acquired epilepsy in patients at risk is an urgent, yet unmet, clinical need. Following acute brain insults, there is an increased risk of mesial temporal lobe epilepsy (mTLE), which is often associated with debilitating comorbidities and reduced life expectancy. The latent period between brain injury and the onset of epilepsy may offer a therapeutic window for interfering with epileptogenesis. The pilocarpine model of mTLE is widely used in the search for novel antiepileptogenic treatments. Recent biochemical studies indicated that cholinergic mechanisms play a role in the epileptogenic alterations induced by status epilepticus (SE) in this and other models of mTLE, which prompted us to evaluate whether treatment with the muscarinic antagonist scopolamine during the latent period after SE is capable of preventing or modifying epilepsy and associated behavioral and cognitive alterations in female Sprague-Dawley rats. First, in silico pharmacokinetic modeling was used to select a dosing protocol by which M-receptor inhibitory brain levels of scopolamine are maintained during prolonged treatment. This protocol was verified by drug analysis in vivo. Rats were then treated twice daily with scopolamine over 17 days after SE, followed by drug wash-out and behavioral and video/EEG monitoring up to ~6 months after SE. Compared to vehicle controls, rats that were treated with scopolamine during the latent period exhibited a significantly lower incidence of spontaneous recurrent seizures during periods of intermittent recording in the chronic phase of epilepsy, less behavioral excitability, less cognitive impairment, and significantly reduced aberrant mossy fiber sprouting in the hippocampus. The present data may indicate that scopolamine exerts antiepileptogenic/disease-modifying activity in the lithium-pilocarpine rat model, possibly involving increased remission of epilepsy as a new mechanism of disease-modification. For evaluating the rigor of the present data, we envision a study that more thoroughly addresses the gender bias and video-EEG recording limitations of the present study.
Collapse
|
20
|
Löscher W, Klein P. New approaches for developing multi-targeted drug combinations for disease modification of complex brain disorders. Does epilepsy prevention become a realistic goal? Pharmacol Ther 2021; 229:107934. [PMID: 34216705 DOI: 10.1016/j.pharmthera.2021.107934] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022]
Abstract
Over decades, the prevailing standard in drug discovery was the concept of designing highly selective compounds that act on individual drug targets. However, more recently, multi-target and combinatorial drug therapies have become an important treatment modality in complex diseases, including neurodegenerative diseases such as Alzheimer's and Parkinson's disease. The development of such network-based approaches is facilitated by the significant advance in our understanding of the pathophysiological processes in these and other complex brain diseases and the adoption of modern computational approaches in drug discovery and repurposing. However, although drug combination therapy has become an effective means for the symptomatic treatment of many complex diseases, the holy grail of identifying clinically effective disease-modifying treatments for neurodegenerative and other brain diseases remains elusive. Thus, despite extensive research, there remains an urgent need for novel treatments that will modify the progression of the disease or prevent its development in patients at risk. Here we discuss recent approaches with a focus on multi-targeted drug combinations for prevention or modification of epilepsy. Over the last ~10 years, several novel promising multi-targeted therapeutic approaches have been identified in animal models. We envision that synergistic combinations of repurposed drugs as presented in this review will be demonstrated to prevent epilepsy in patients at risk within the next 5-10 years.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| |
Collapse
|
21
|
Dey S, Doddamani RS, Banerjee Dixit A, Tripathi M, Sharma MC, Chandra PS, Banerjee J. Altered Spontaneous Glutamatergic and GABAergic Activity in the Peritumoral Cortex of Low-Grade Gliomas Presenting With History of Seizures. Front Neurosci 2021; 15:689769. [PMID: 34262432 PMCID: PMC8273299 DOI: 10.3389/fnins.2021.689769] [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: 04/01/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
The peritumoral regions of WHO grade II gliomas, like astrocytoma and oligodendroglioma, have been reported to show epileptiform activities. An imbalance of glutamatergic and GABAergic mechanisms is primarily responsible for the generation of epileptiform activities. Here we have compared the electrophysiological properties of pyramidal neurons in intraoperative peritumoral specimens obtained from glioma patients with (GS) and without (GN) a history of seizures at presentation. Histology and immunohistochemistry were performed to assess the infiltration of proliferating cells at the peritumoral tissues. Whole-cell patch clamp technique was performed to measure the spontaneous glutamatergic and GABAergic activity onto pyramidal neurons in the peritumoral samples of GS (n = 11) and GN (n = 15) patients. The cytoarchitecture of the peritumoral tissues was devoid of Ki67 immuno-positive cells. We observed a higher frequency of spontaneous glutamatergic and GABAergic activities onto pyramidal neurons of the peritumoral samples of GS patients. Our findings suggest that, in spite of similar histopathological features, the pyramidal neurons in the peritumoral samples of GS and GN patients showed differences in spontaneous excitatory and inhibitory synaptic neurotransmission. An alteration in postsynaptic currents may contribute to the spontaneous epileptiform activity in GS patients.
Collapse
Affiliation(s)
- Soumil Dey
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | | | - Aparna Banerjee Dixit
- Dr. B. R. Ambedkar Centre for Biomedical Research, University of Delhi, New Delhi, India
| | - Manjari Tripathi
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Meher Chand Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - P Sarat Chandra
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Jyotirmoy Banerjee
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
22
|
Boison D. Specialty Grand Challenge for Brain Disease Mechanisms. Front Mol Neurosci 2021; 14:689903. [PMID: 34040504 PMCID: PMC8141592 DOI: 10.3389/fnmol.2021.689903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
23
|
Hampel P, Johne M, Gailus B, Vogel A, Schidlitzki A, Gericke B, Töllner K, Theilmann W, Käufer C, Römermann K, Kaila K, Löscher W. Deletion of the Na-K-2Cl cotransporter NKCC1 results in a more severe epileptic phenotype in the intrahippocampal kainate mouse model of temporal lobe epilepsy. Neurobiol Dis 2021; 152:105297. [PMID: 33581254 DOI: 10.1016/j.nbd.2021.105297] [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] [Received: 01/06/2021] [Revised: 01/29/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
Increased neuronal expression of the Na-K-2Cl cotransporter NKCC1 has been implicated in the generation of seizures and epilepsy. However, conclusions from studies on the NKCC1-specific inhibitor, bumetanide, are equivocal, which is a consequence of the multiple potential cellular targets and poor brain penetration of this drug. Here, we used Nkcc1 knockout (KO) and wildtype (WT) littermate control mice to study the ictogenic and epileptogenic effects of intrahippocampal injection of kainate. Kainate (0.23 μg in 50 nl) induced limbic status epilepticus (SE) in both KO and WT mice with similar incidence, latency to SE onset, and SE duration, but the number of intermittent generalized convulsive seizures during SE was significantly higher in Nkcc1 KO mice, indicating increased SE severity. Following SE, spontaneous recurrent seizures (SRS) were recorded by continuous (24/7) video/EEG monitoring at 0-1, 4-5, and 12-13 weeks after kainate, using depth electrodes in the ipsilateral hippocampus. Latency to onset of electrographic SRS and the incidence of electrographic SRS were similar in WT and KO mice. However, the frequency of electrographic seizures was lower whereas the frequency of electroclinical seizures was higher in Nkcc1 KO mice, indicating a facilitated progression from electrographic to electroclinical seizures during chronic epilepsy, and a more severe epileptic phenotype, in the absence of NKCC1. The present findings suggest that NKCC1 is dispensable for the induction, progression and manifestation of epilepsy, and they do not support the widely held notion that inhibition of NKCC1 in the brain is a useful strategy for preventing or modifying epilepsy.
Collapse
Affiliation(s)
- Philip Hampel
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Neurona Therapeutics, San Francisco, CA, USA
| | - Marie Johne
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Björn Gailus
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Alexandra Vogel
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Alina Schidlitzki
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Birthe Gericke
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Kathrin Töllner
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Wiebke Theilmann
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Christopher Käufer
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Kerstin Römermann
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Kai Kaila
- Molecular and Integrative Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Finland
| | - Wolfgang Löscher
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
24
|
Garcia-Rosa S, de Freitas Brenha B, Felipe da Rocha V, Goulart E, Araujo BHS. Personalized Medicine Using Cutting Edge Technologies for Genetic Epilepsies. Curr Neuropharmacol 2021; 19:813-831. [PMID: 32933463 PMCID: PMC8686309 DOI: 10.2174/1570159x18666200915151909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/08/2020] [Accepted: 08/28/2020] [Indexed: 11/22/2022] Open
Abstract
Epilepsy is the most common chronic neurologic disorder in the world, affecting 1-2% of the population. Besides, 30% of epilepsy patients are drug-resistant. Genomic mutations seem to play a key role in its etiology and knowledge of strong effect mutations in protein structures might improve prediction and the development of efficacious drugs to treat epilepsy. Several genetic association studies have been undertaken to examine the effect of a range of candidate genes for resistance. Although, few studies have explored the effect of the mutations into protein structure and biophysics in the epilepsy field. Much work remains to be done, but the plans made for exciting developments will hold therapeutic potential for patients with drug-resistance. In summary, we provide a critical review of the perspectives for the development of individualized medicine for epilepsy based on genetic polymorphisms/mutations in light of core elements such as transcriptomics, structural biology, disease model, pharmacogenomics and pharmacokinetics in a manner to improve the success of trial designs of antiepileptic drugs.
Collapse
Affiliation(s)
- Sheila Garcia-Rosa
- Brazilian Biosciences National Laboratory (LNBio), Center for Research in Energy and Material (CNPEM), Campinas, SP, Brazil
| | - Bianca de Freitas Brenha
- Laboratory of Embryonic Genetic Regulation, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | - Vinicius Felipe da Rocha
- Brazilian Biosciences National Laboratory (LNBio), Center for Research in Energy and Material (CNPEM), Campinas, SP, Brazil
| | - Ernesto Goulart
- Human Genome and Stem-Cell Research Center (HUG-CEL), Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, SP, Brazil
| | - Bruno Henrique Silva Araujo
- Brazilian Biosciences National Laboratory (LNBio), Center for Research in Energy and Material (CNPEM), Campinas, SP, Brazil
| |
Collapse
|
25
|
Löscher W, Klein P. The Pharmacology and Clinical Efficacy of Antiseizure Medications: From Bromide Salts to Cenobamate and Beyond. CNS Drugs 2021; 35:935-963. [PMID: 34145528 PMCID: PMC8408078 DOI: 10.1007/s40263-021-00827-8] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 12/16/2022]
Abstract
Epilepsy is one of the most common and disabling chronic neurological disorders. Antiseizure medications (ASMs), previously referred to as anticonvulsant or antiepileptic drugs, are the mainstay of symptomatic epilepsy treatment. Epilepsy is a multifaceted complex disease and so is its treatment. Currently, about 30 ASMs are available for epilepsy therapy. Furthermore, several ASMs are approved therapies in nonepileptic conditions, including neuropathic pain, migraine, bipolar disorder, and generalized anxiety disorder. Because of this wide spectrum of therapeutic activity, ASMs are among the most often prescribed centrally active agents. Most ASMs act by modulation of voltage-gated ion channels; by enhancement of gamma aminobutyric acid-mediated inhibition; through interactions with elements of the synaptic release machinery; by blockade of ionotropic glutamate receptors; or by combinations of these mechanisms. Because of differences in their mechanisms of action, most ASMs do not suppress all types of seizures, so appropriate treatment choices are important. The goal of epilepsy therapy is the complete elimination of seizures; however, this is not achievable in about one-third of patients. Both in vivo and in vitro models of seizures and epilepsy are used to discover ASMs that are more effective in patients with continued drug-resistant seizures. Furthermore, therapies that are specific to epilepsy etiology are being developed. Currently, ~ 30 new compounds with diverse antiseizure mechanisms are in the preclinical or clinical drug development pipeline. Moreover, therapies with potential antiepileptogenic or disease-modifying effects are in preclinical and clinical development. Overall, the world of epilepsy therapy development is changing and evolving in many exciting and important ways. However, while new epilepsy therapies are developed, knowledge of the pharmacokinetics, antiseizure efficacy and spectrum, and adverse effect profiles of currently used ASMs is an essential component of treating epilepsy successfully and maintaining a high quality of life for every patient, particularly those receiving polypharmacy for drug-resistant seizures.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Bünteweg 17, 30559, Hannover, Germany. .,Center for Systems Neuroscience, Hannover, Germany.
| | - Pavel Klein
- grid.429576.bMid-Atlantic Epilepsy and Sleep Center, Bethesda, MD USA
| |
Collapse
|
26
|
Welzel L, Bergin DH, Schidlitzki A, Twele F, Johne M, Klein P, Löscher W. Systematic evaluation of rationally chosen multitargeted drug combinations: a combination of low doses of levetiracetam, atorvastatin and ceftriaxone exerts antiepileptogenic effects in a mouse model of acquired epilepsy. Neurobiol Dis 2020; 149:105227. [PMID: 33347976 DOI: 10.1016/j.nbd.2020.105227] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/24/2020] [Accepted: 12/16/2020] [Indexed: 01/22/2023] Open
Abstract
Epileptogenesis, the gradual process that leads to epilepsy after brain injury or genetic mutations, is a complex network phenomenon, involving a variety of morphological, biochemical and functional brain alterations. Although risk factors for developing epilepsy are known, there is currently no treatment available to prevent epilepsy. We recently proposed a multitargeted, network-based approach to prevent epileptogenesis by rationally combining clinically available drugs and provided first proof-of-concept that this strategy is effective. Here we evaluated eight novel rationally chosen combinations of 14 drugs with mechanisms that target different epileptogenic processes. The combinations consisted of 2-4 different drugs per combination and were administered systemically over 5 days during the latent epileptogenic period in the intrahippocampal kainate mouse model of acquired temporal lobe epilepsy, starting 6 h after kainate. Doses and dosing intervals were based on previous pharmacokinetic and tolerability studies in mice. The incidence and frequency of spontaneous electrographic and electroclinical seizures were recorded by continuous (24/7) video linked EEG monitoring done for seven days at 4 and 12 weeks post-kainate, i.e., long after termination of drug treatment. Compared to vehicle controls, the most effective drug combination consisted of low doses of levetiracetam, atorvastatin and ceftriaxone, which markedly reduced the incidence of electrographic seizures (by 60%; p<0.05) and electroclinical seizures (by 100%; p<0.05) recorded at 12 weeks after kainate. This effect was lost when higher doses of the three drugs were administered, indicating a synergistic drug-drug interaction at the low doses. The potential mechanisms underlying this interaction are discussed. We have discovered a promising novel multitargeted combination treatment for modifying the development of acquired epilepsy.
Collapse
Affiliation(s)
- Lisa Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - David H Bergin
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Marie Johne
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
27
|
Miziak B, Konarzewska A, Ułamek-Kozioł M, Dudra-Jastrzębska M, Pluta R, Czuczwar SJ. Anti-Epileptogenic Effects of Antiepileptic Drugs. Int J Mol Sci 2020; 21:ijms21072340. [PMID: 32231010 PMCID: PMC7178140 DOI: 10.3390/ijms21072340] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022] Open
Abstract
Generally, the prevalence of epilepsy does not exceed 0.9% of the population and approximately 70% of epilepsy patients may be adequately controlled with antiepileptic drugs (AEDs). Moreover, status epilepticus (SE) or even a single seizure may produce neurodegeneration within the brain and SE has been recognized as one of acute brain insults leading to acquired epilepsy via the process of epileptogenesis. Two questions thus arise: (1) Are AEDs able to inhibit SE-induced neurodegeneration? and (2) if so, can a probable neuroprotective potential of particular AEDs stop epileptogenesis? An affirmative answer to the second question would practically point to the preventive potential of a given neuroprotective AED following acute brain insults. The available experimental data indicate that diazepam (at low and high doses), gabapentin, pregabalin, topiramate and valproate exhibited potent or moderate neuroprotective effects in diverse models of SE in rats. However, only diazepam (at high doses), gabapentin and pregabalin exerted some protective activity against acquired epilepsy (spontaneous seizures). As regards valproate, its effects on spontaneous seizures were equivocal. With isobolography, some supra-additive combinations of AEDs have been delineated against experimental seizures. One of such combinations, levetiracetam + topiramate proved highly synergistic in two models of seizures and this particular combination significantly inhibited epileptogenesis in rats following status SE. Importantly, no neuroprotection was evident. It may be strikingly concluded that there is no correlation between neuroprotection and antiepileptogenesis. Probably, preclinically verified combinations of AEDs may be considered for an anti-epileptogenic therapy.
Collapse
Affiliation(s)
- Barbara Miziak
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
| | - Agnieszka Konarzewska
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
| | - Marzena Ułamek-Kozioł
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Monika Dudra-Jastrzębska
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
| | - Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland;
- Correspondence: (R.P.); (S.J.C.); Tel.: +48-22-6086-540 (ext. 6086-469) (R.P.); +48-81-448-65-00 (S.J.C.); Fax: +48-81-448-65-01 (S.J.C.); +48-22-6086-627/668-55-32 (R.P.)
| | - Stanisław J. Czuczwar
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (B.M.); (A.K.); (M.D.-J.)
- Correspondence: (R.P.); (S.J.C.); Tel.: +48-22-6086-540 (ext. 6086-469) (R.P.); +48-81-448-65-00 (S.J.C.); Fax: +48-81-448-65-01 (S.J.C.); +48-22-6086-627/668-55-32 (R.P.)
| |
Collapse
|
28
|
Supuran CT. An update on drug interaction considerations in the therapeutic use of carbonic anhydrase inhibitors. Expert Opin Drug Metab Toxicol 2020; 16:297-307. [DOI: 10.1080/17425255.2020.1743679] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Claudiu T. Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
29
|
Klein P, Friedman A, Hameed MQ, Kaminski RM, Bar-Klein G, Klitgaard H, Koepp M, Jozwiak S, Prince DA, Rotenberg A, Twyman R, Vezzani A, Wong M, Löscher W. Repurposed molecules for antiepileptogenesis: Missing an opportunity to prevent epilepsy? Epilepsia 2020; 61:359-386. [PMID: 32196665 PMCID: PMC8317585 DOI: 10.1111/epi.16450] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/11/2022]
Abstract
Prevention of epilepsy is a great unmet need. Acute central nervous system (CNS) insults such as traumatic brain injury (TBI), cerebrovascular accidents (CVA), and CNS infections account for 15%-20% of all epilepsy. Following TBI and CVA, there is a latency of days to years before epilepsy develops. This allows treatment to prevent or modify postinjury epilepsy. No such treatment exists. In animal models of acquired epilepsy, a number of medications in clinical use for diverse indications have been shown to have antiepileptogenic or disease-modifying effects, including medications with excellent side effect profiles. These include atorvastatin, ceftriaxone, losartan, isoflurane, N-acetylcysteine, and the antiseizure medications levetiracetam, brivaracetam, topiramate, gabapentin, pregabalin, vigabatrin, and eslicarbazepine acetate. In addition, there are preclinical antiepileptogenic data for anakinra, rapamycin, fingolimod, and erythropoietin, although these medications have potential for more serious side effects. However, except for vigabatrin, there have been almost no translation studies to prevent or modify epilepsy using these potentially "repurposable" medications. We may be missing an opportunity to develop preventive treatment for epilepsy by not evaluating these medications clinically. One reason for the lack of translation studies is that the preclinical data for most of these medications are disparate in terms of types of injury, models within different injury type, dosing, injury-treatment initiation latencies, treatment duration, and epilepsy outcome evaluation mode and duration. This makes it difficult to compare the relative strength of antiepileptogenic evidence across the molecules, and difficult to determine which drug(s) would be the best to evaluate clinically. Furthermore, most preclinical antiepileptogenic studies lack information needed for translation, such as dose-blood level relationship, brain target engagement, and dose-response, and many use treatment parameters that cannot be applied clinically, for example, treatment initiation before or at the time of injury and dosing higher than tolerated human equivalent dosing. Here, we review animal and human antiepileptogenic evidence for these medications. We highlight the gaps in our knowledge for each molecule that need to be filled in order to consider clinical translation, and we suggest a platform of preclinical antiepileptogenesis evaluation of potentially repurposable molecules or their combinations going forward.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, Maryland
| | - Alon Friedman
- Departments of Physiology and Cell Biology, and Brain and Cognitive Science, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Departments of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, Canada
| | - Mustafa Q. Hameed
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rafal M. Kaminski
- Neurosymptomatic Domains Section, Roche Pharma Research & Early Development, Roche Innovation Center, Basel, Switzerland
| | - Guy Bar-Klein
- McKusick-Nathans Institute of Genetic Medicine, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Henrik Klitgaard
- Neurosciences Therapeutic Area, UCB Pharma, Braine-l’Alleud, Belgium
| | - Mathias Koepp
- Department of Clinical and Experimental Epilepsy, University College London Institute of Neurology, London, UK
| | - Sergiusz Jozwiak
- Department of Pediatric Neurology, Warsaw Medical University, Warsaw, Poland
| | - David A. Prince
- Neurology and the Neurological Sciences, Stanford University School of Medicine, Stanford, California
| | - Alexander Rotenberg
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Annamaria Vezzani
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Scientific Institute for Research and Health Care, Milan, Italy
| | - Michael Wong
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|