1
|
Klein P, Koepp M, Rotenberg A, Hameed M, Löscher W. Clinical trials of prevention of acquired epilepsy: New proof-of-concept approach to restart trials. Epilepsia 2025. [PMID: 40184261 DOI: 10.1111/epi.18394] [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: 09/28/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/06/2025]
Abstract
Approximately 20% of epilepsy is caused by acute central nervous system insults such as traumatic brain injury (TBI), stroke, and infection. There is a latent period of weeks to years between the insult and epilepsy onset, which offers an opportunity to prevent epilepsy. No preventive treatments exist. Their development is a major unmet need in neurology. For logistical reasons, epilepsy acquired after TBI, posttraumatic epilepsy (PTE), is most suitable for epilepsy prevention studies. In the past 20 years, preclinical PTE research has flourished, offering potential treatments to prevent PTE, but clinical development has been dormant. The major barrier in the development of PTE preventive treatment is the lack of a viable proof of concept (POC) trial design. PTE trials use the first late unprovoked posttraumatic seizure as an outcome measure, which necessitates a long (~2-year) follow-up and makes POC studies nonfeasible. A reliable biomarker of early PTE detection would allow shorter follow-up duration and facilitate POC studies, but such a biomarker is not yet available. Biomarker, POC, and randomized clinical trial studies have virtually identical designs in terms of patient inclusion and follow-up. Done sequentially, the studies would take a generation to complete. We propose a novel trial design for studies of PTE prevention that combines discovery of biomarker(s) of early PTE detection with POC study and uses an adaptive study POC-phase 3 continuation design approach to incorporate POC study into phase 3 study following an interim futility analysis after 6 months of treatment of the first 25% of the cohort, the POC population. This approach would establish a POC model for treatment of PTE prevention, shorten development of PTE prevention treatment, and reopen the door to clinical trials to prevent epilepsy.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, Maryland, USA
| | - Matthias Koepp
- Department of Clinical and Experimental Epilepsy, University College London Queen Square Institute of Neurology, London, UK
| | - Alexander Rotenberg
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mustafa Hameed
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wolfgang Löscher
- Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the Ear, Nose and Throat Clinics, Hannover Medical School, Hannover, Germany
| |
Collapse
|
2
|
Yardi R, Vasireddy RP, Galovic M, Punia V. Antiseizure medication use in acute symptomatic seizures: A narrative review. Epilepsia 2025. [PMID: 39841056 DOI: 10.1111/epi.18275] [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: 11/09/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
Acute symptomatic seizures, occurring shortly after a central nervous system insult, constitute nearly half of all seizure cases. However, there is a conspicuous absence of clear, comprehensive, and cohesive guidelines for the management of these seizures with antiseizure medications, especially their duration of use. This lack of consensus on the optimal duration of therapy leads to prolonged treatments that may carry adverse consequences. The primary objective of this narrative review is to present the existing evidence-based literature on the management of acute symptomatic seizures within the context of the underlying pathologies that trigger them. We explore the risk of developing epilepsy for each specific etiology and identify the factors that influence this risk. Finally, to facilitate decision-making regarding treatment duration, we categorize acute seizures based on the temporal characteristics of hyperexcitability as acute, subacute, and prolonged. Such a rubric may offer clarity in an area where consensus and guidelines are lacking.
Collapse
Affiliation(s)
- Ruta Yardi
- Department of Neurology, Geisinger Neuroscience Institute, Geisinger Health System, Danville, Pennsylvania, USA
- Kentucky Neuroscience Institute, University of Kentucky, Lexington, Kentucky, USA
| | | | - Marian Galovic
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Vineet Punia
- Epilepsy Center, Cleveland Clinic, Cleveland, Ohio, USA
| |
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
|
Klein P, Kaminski RM, Koepp M, Löscher W. New epilepsy therapies in development. Nat Rev Drug Discov 2024; 23:682-708. [PMID: 39039153 DOI: 10.1038/s41573-024-00981-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2024] [Indexed: 07/24/2024]
Abstract
Epilepsy is a common brain disorder, characterized by spontaneous recurrent seizures, with associated neuropsychiatric and cognitive comorbidities and increased mortality. Although people at risk can often be identified, interventions to prevent the development of the disorder are not available. Moreover, in at least 30% of patients, epilepsy cannot be controlled by current antiseizure medications (ASMs). As a result of considerable progress in epilepsy genetics and the development of novel disease models, drug screening technologies and innovative therapeutic modalities over the past 10 years, more than 200 novel epilepsy therapies are currently in the preclinical or clinical pipeline, including many treatments that act by new mechanisms. Assisted by diagnostic and predictive biomarkers, the treatment of epilepsy is undergoing paradigm shifts from symptom-only ASMs to disease prevention, and from broad trial-and-error treatments for seizures in general to mechanism-based treatments for specific epilepsy syndromes. In this Review, we assess recent progress in ASM development and outline future directions for the development of new therapies for the treatment and prevention of epilepsy.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA.
| | | | - Matthias Koepp
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Wolfgang Löscher
- Translational Neuropharmacology Lab., NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
5
|
Foresti ML, Garzon E, de Moraes MT, Valeriano RPS, Santiago JP, dos Santos GM, Longo NM, Baise C, Andrade JCQF, Susemihl MA, Leite CDC, Naffah Mazzacoratti MDG, Paiva WS, de Andrade AF, Teixeira MJ, Mello LE. Initial clinical evidence on biperiden as antiepileptogenic after traumatic brain injury-a randomized clinical trial. Front Neurol 2024; 15:1443982. [PMID: 39175759 PMCID: PMC11339878 DOI: 10.3389/fneur.2024.1443982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
There is currently no efficacious intervention for preventing post-traumatic epilepsy (PTE). Preclinical studies support the potential use of anticholinergics for this condition. The purpose of this study was to evaluate the effects of biperiden as an intervention for preventing PTE. A randomized, double-blinded clinical trial was conducted at HC/FMUSP between 2018-2022. Adults with acute traumatic brain injury (TBI) were randomly assigned to receive biperiden or placebo, for 10 days. The primary outcome was the incidence of PTE while the secondary outcomes included the frequency of seizures, the frequency of any adverse events and mortality after 24 months. The study was powered at a planned enrolment of 132 patients. The trial began in January 2018 and was halted by researchers on March 2020 (and terminated in December 2022) in the face of the global COVID-19 pandemic. Overall, 123 participants were randomized and 112 contributed with data for modified mITT analysis, being that 61 (49.5%) participants completed the 24-month follow-up consult. Data analysis indicated lack of evidence of biperiden for either, the incidence of post-traumatic epilepsy (2.6, 95%CI, 0.65-10.57; p = 0.170) or the mortality rate (1.57, 95%CI, 0.73-3.38; p = 0.248). The frequency of late post-traumatic seizures was higher for biperiden group (2.03, 95%CI = 0.912-3.1597; p <0.001). The present study suggests that there was insufficient evidence regarding the effect of biperiden in preventing PTE after TBI, which underpins the need for larger studies. Clinical trial registration: ClinicalTrials.gov, identifier: NCT01048138.
Collapse
Affiliation(s)
- Maira Licia Foresti
- Neurology Neuroscience Postgraduation Program, Physiology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Instituto D’Or de Pesquisa e Ensino, São Paulo, Brazil
| | - Eliana Garzon
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Sociedade Beneficente de Senhoras Hospital Sírio-Libanês, São Paulo, Brazil
| | - Mariana Teichner de Moraes
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Rafael P. S. Valeriano
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - João Paulo Santiago
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Gustavo Mercenas dos Santos
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Natália Mata Longo
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Carla Baise
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Joaquina C. Q. F. Andrade
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Claudia da Costa Leite
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Maria da Graça Naffah Mazzacoratti
- Neurology Neuroscience Postgraduation Program, Physiology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Wellingson Silva Paiva
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Almir Ferreira de Andrade
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Manuel Jacobsen Teixeira
- Department of Neurology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Luiz E. Mello
- Neurology Neuroscience Postgraduation Program, Physiology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Instituto D’Or de Pesquisa e Ensino, São Paulo, Brazil
| |
Collapse
|
6
|
Frontera JA, Gilmore EJ, Johnson EL, Olson D, Rayi A, Tesoro E, Ullman J, Yuan Y, Zafar SF, Rowe S. Guidelines for Seizure Prophylaxis in Adults Hospitalized with Moderate-Severe Traumatic Brain Injury: A Clinical Practice Guideline for Health Care Professionals from the Neurocritical Care Society. Neurocrit Care 2024; 40:819-844. [PMID: 38316735 DOI: 10.1007/s12028-023-01907-x] [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: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND There is practice heterogeneity in the use, type, and duration of prophylactic antiseizure medications (ASMs) in patients with moderate-severe traumatic brain injury (TBI). METHODS We conducted a systematic review and meta-analysis of articles assessing ASM prophylaxis in adults with moderate-severe TBI (acute radiographic findings and requiring hospitalization). The population, intervention, comparator, and outcome (PICO) questions were as follows: (1) Should ASM versus no ASM be used in patients with moderate-severe TBI and no history of clinical or electrographic seizures? (2) If an ASM is used, should levetiracetam (LEV) or phenytoin/fosphenytoin (PHT/fPHT) be preferentially used? (3) If an ASM is used, should a long versus short (> 7 vs. ≤ 7 days) duration of prophylaxis be used? The main outcomes were early seizure, late seizure, adverse events, mortality, and functional outcomes. We used Grading of Recommendations Assessment, Development, and Evaluation (GRADE) methodology to generate recommendations. RESULTS The initial literature search yielded 1998 articles, of which 33 formed the basis of the recommendations: PICO 1: We did not detect any significant positive or negative effect of ASM compared to no ASM on the outcomes of early seizure, late seizure, adverse events, or mortality. PICO 2: We did not detect any significant positive or negative effect of PHT/fPHT compared to LEV for early seizures or mortality, though point estimates suggest fewer late seizures and fewer adverse events with LEV. PICO 3: There were no significant differences in early or late seizures with longer versus shorter ASM use, though cognitive outcomes and adverse events appear worse with protracted use. CONCLUSIONS Based on GRADE criteria, we suggest that ASM or no ASM may be used in patients hospitalized with moderate-severe TBI (weak recommendation, low quality of evidence). If used, we suggest LEV over PHT/fPHT (weak recommendation, very low quality of evidence) for a short duration (≤ 7 days, weak recommendation, low quality of evidence).
Collapse
Affiliation(s)
- Jennifer A Frontera
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Neurology, NYU, 150 55th St., Brooklyn, NY, USA.
| | - Emily J Gilmore
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Emily L Johnson
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - DaiWai Olson
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Appaji Rayi
- Department of Neurology, Charleston Area Medical Center, Charleston, WV, USA
| | - Eljim Tesoro
- Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Jamie Ullman
- Department of Neurosurgery, Northwell Health, Great Neck, NY, USA
| | - Yuhong Yuan
- Division of Gastroenterology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sahar F Zafar
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Shaun Rowe
- Department of Clinical Pharmacology, University of Tennessee Health Science Center College of Pharmacy, Knoxville, TN, USA
| |
Collapse
|
7
|
Ménétré E, De Stefano P, Megevand P, Sarasin FP, Vargas MI, Kleinschmidt A, Vulliemoz S, Picard F, Seeck M. Antiseizure medication ≤48 hours portends better prognosis in new-onset epilepsy. Eur J Neurol 2024; 31:e16107. [PMID: 37889889 PMCID: PMC11236038 DOI: 10.1111/ene.16107] [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: 02/03/2023] [Revised: 07/28/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND Several studies found that patients with new-onset epilepsy (NOE) have higher seizure recurrence rates if they presented already prior seizures. These observations suggest that timing of antiseizure medication (ASM) is crucial and should be offered immediately after the first seizure. Here, we wanted to assess whether immediate ASM is associated with improved outcome. METHODS Single-center study of 1010 patients (≥16 years) who presented with a possible first seizure in the emergency department between 1 March 2010 and 1 March 2017. A comprehensive workup was launched upon arrival, including routine electroencephalography (EEG), brain computed tomography/magnetic resonance imaging, long-term overnight EEG and specialized consultations. We followed patients for 5 years comparing the relapse rate in patients treated within 48 h to those with treatment >48 h. RESULTS A total of 487 patients were diagnosed with NOE. Of the 416 patients (162 female, age: 54.6 ± 21.1 years) for whom the treatment start could be retrieved, 80% (333/416) were treated within 48 h. The recurrence rate after immediate treatment (32%; 107/333) was significantly lower than in patients treated later (56.6%; 47/83; p < 0.001). For patients for whom a complete 5-year-follow-up was available (N = 297, 123 female), those treated ≤48 h (N = 228; 76.8%) had a significantly higher chance of remaining seizure-free compared with patients treated later (N = 69; 23.2%; p < 0.001). CONCLUSIONS In this retrospective study, immediate ASM therapy (i.e., within 48 h) was associated with better prognosis up to 5 years after the index event. Prospective studies are required to determine the value of immediate workup and drug therapy in NOE patients.
Collapse
Affiliation(s)
- Eric Ménétré
- EEG & Epilepsy Unit, Department of Clinical NeurosciencesUniversity Hospitals of GenevaGenevaSwitzerland
| | - Pia De Stefano
- EEG & Epilepsy Unit, Department of Clinical NeurosciencesUniversity Hospitals of GenevaGenevaSwitzerland
- Neuro‐Critical Care Unit, Department of Intensive CareUniversity Hospitals of GenevaGenevaSwitzerland
| | - Pierre Megevand
- EEG & Epilepsy Unit, Department of Clinical NeurosciencesUniversity Hospitals of GenevaGenevaSwitzerland
| | - François P. Sarasin
- Division of Emergency Medicine, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency MedicineUniversity of Geneva Hospitals and Faculty of MedicineGenevaSwitzerland
| | - Maria I. Vargas
- Neuroradiology DepartmentUniversity Hospitals of GenevaGenevaSwitzerland
| | - Andreas Kleinschmidt
- EEG & Epilepsy Unit, Department of Clinical NeurosciencesUniversity Hospitals of GenevaGenevaSwitzerland
| | - Serge Vulliemoz
- EEG & Epilepsy Unit, Department of Clinical NeurosciencesUniversity Hospitals of GenevaGenevaSwitzerland
| | - Fabienne Picard
- EEG & Epilepsy Unit, Department of Clinical NeurosciencesUniversity Hospitals of GenevaGenevaSwitzerland
| | - Margitta Seeck
- EEG & Epilepsy Unit, Department of Clinical NeurosciencesUniversity Hospitals of GenevaGenevaSwitzerland
| |
Collapse
|
8
|
Coelho LMG, Blacker D, Hsu J, Newhouse JP, Westover MB, Zafar SF, Moura LMVR. Association of Early Seizure Prophylaxis With Posttraumatic Seizures and Mortality: A Meta-analysis With Evidence Quality Assessment. Neurol Clin Pract 2023; 13:e200145. [PMID: 37066107 PMCID: PMC10101717 DOI: 10.1212/cpj.0000000000200145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/11/2023] [Indexed: 04/18/2023]
Abstract
Purpose of the Review To evaluate the quality of evidence about the association of primary seizure prophylaxis with antiseizure medication (ASM) within 7 days postinjury and the 18- or 24-month epilepsy/late seizure risk or all-cause mortality in adults with new-onset traumatic brain injury (TBI), in addition to early seizure risk. Results Twenty-three studies met the inclusion criteria (7 randomized and 16 nonrandomized studies). We analyzed 9,202 patients, including 4,390 in the exposed group and 4,812 in the unexposed group (894 in placebo and 3,918 in no ASM groups). There was a moderate to serious bias risk based on our assessment. Within the limitations of existing studies, our data revealed a lower risk for early seizures in the ASM prophylaxis group compared with placebo or no ASM prophylaxis (risk ratio [RR] 0.43, 95% confidence interval [CI] 0.33-0.57, p < 0.00001, I 2 = 3%). We identified high-quality evidence in favor of acute, short-term primary ASM use to prevent early seizures. Early ASM prophylaxis was not associated with a substantial difference in the 18- or 24-month risk of epilepsy/late seizures (RR 1.01, 95% CI 0.61-1.68, p = 0.96, I 2 = 63%) or mortality (RR 1.16, 95% CI 0.89-1.51, p = 0.26, I 2 = 0%). There was no evidence of strong publication bias for each main outcome. The overall quality of evidence was low and moderate for post-TBI epilepsy risk and all-cause mortality, respectively. Summary Our data suggest that the evidence showing no association between early ASM use and 18- or 24-month epilepsy risk in adults with new-onset TBI was of low quality. The analysis indicated a moderate quality for the evidence showing no effect on all-cause mortality. Therefore, higher-quality evidence is needed as a supplement for stronger recommendations.
Collapse
Affiliation(s)
- Lilian Maria Godeiro Coelho
- Department of Neurology (LMGC, MBW, SFZ, LMVRM), Massachusetts General Hospital; Department of Neurology (MBW, SFZ, LMVRM), Harvard Medical School; Department of Epidemiology (DB), Harvard T.H. Chan School of Public Health; Department of Psychiatry (DB), Massachusetts General Hospital; Department of Psychiatry (DB), Harvard Medical School; Department of Health Care Policy (JH, JPN), Harvard Medical School; Mongan Institute (JH), Massachusetts General Hospital; Department of Medicine (JH), Harvard Medical School, Boston; National Bureau of Economic Research (JPN), Cambridge; Department of Health Policy and Management (JPN), Harvard T.H. Chan School of Public Health, Boston; and Harvard Kennedy School (JPN), Cambridge, MA
| | - Deborah Blacker
- Department of Neurology (LMGC, MBW, SFZ, LMVRM), Massachusetts General Hospital; Department of Neurology (MBW, SFZ, LMVRM), Harvard Medical School; Department of Epidemiology (DB), Harvard T.H. Chan School of Public Health; Department of Psychiatry (DB), Massachusetts General Hospital; Department of Psychiatry (DB), Harvard Medical School; Department of Health Care Policy (JH, JPN), Harvard Medical School; Mongan Institute (JH), Massachusetts General Hospital; Department of Medicine (JH), Harvard Medical School, Boston; National Bureau of Economic Research (JPN), Cambridge; Department of Health Policy and Management (JPN), Harvard T.H. Chan School of Public Health, Boston; and Harvard Kennedy School (JPN), Cambridge, MA
| | - John Hsu
- Department of Neurology (LMGC, MBW, SFZ, LMVRM), Massachusetts General Hospital; Department of Neurology (MBW, SFZ, LMVRM), Harvard Medical School; Department of Epidemiology (DB), Harvard T.H. Chan School of Public Health; Department of Psychiatry (DB), Massachusetts General Hospital; Department of Psychiatry (DB), Harvard Medical School; Department of Health Care Policy (JH, JPN), Harvard Medical School; Mongan Institute (JH), Massachusetts General Hospital; Department of Medicine (JH), Harvard Medical School, Boston; National Bureau of Economic Research (JPN), Cambridge; Department of Health Policy and Management (JPN), Harvard T.H. Chan School of Public Health, Boston; and Harvard Kennedy School (JPN), Cambridge, MA
| | - Joseph P Newhouse
- Department of Neurology (LMGC, MBW, SFZ, LMVRM), Massachusetts General Hospital; Department of Neurology (MBW, SFZ, LMVRM), Harvard Medical School; Department of Epidemiology (DB), Harvard T.H. Chan School of Public Health; Department of Psychiatry (DB), Massachusetts General Hospital; Department of Psychiatry (DB), Harvard Medical School; Department of Health Care Policy (JH, JPN), Harvard Medical School; Mongan Institute (JH), Massachusetts General Hospital; Department of Medicine (JH), Harvard Medical School, Boston; National Bureau of Economic Research (JPN), Cambridge; Department of Health Policy and Management (JPN), Harvard T.H. Chan School of Public Health, Boston; and Harvard Kennedy School (JPN), Cambridge, MA
| | - M Brandon Westover
- Department of Neurology (LMGC, MBW, SFZ, LMVRM), Massachusetts General Hospital; Department of Neurology (MBW, SFZ, LMVRM), Harvard Medical School; Department of Epidemiology (DB), Harvard T.H. Chan School of Public Health; Department of Psychiatry (DB), Massachusetts General Hospital; Department of Psychiatry (DB), Harvard Medical School; Department of Health Care Policy (JH, JPN), Harvard Medical School; Mongan Institute (JH), Massachusetts General Hospital; Department of Medicine (JH), Harvard Medical School, Boston; National Bureau of Economic Research (JPN), Cambridge; Department of Health Policy and Management (JPN), Harvard T.H. Chan School of Public Health, Boston; and Harvard Kennedy School (JPN), Cambridge, MA
| | - Sahar F Zafar
- Department of Neurology (LMGC, MBW, SFZ, LMVRM), Massachusetts General Hospital; Department of Neurology (MBW, SFZ, LMVRM), Harvard Medical School; Department of Epidemiology (DB), Harvard T.H. Chan School of Public Health; Department of Psychiatry (DB), Massachusetts General Hospital; Department of Psychiatry (DB), Harvard Medical School; Department of Health Care Policy (JH, JPN), Harvard Medical School; Mongan Institute (JH), Massachusetts General Hospital; Department of Medicine (JH), Harvard Medical School, Boston; National Bureau of Economic Research (JPN), Cambridge; Department of Health Policy and Management (JPN), Harvard T.H. Chan School of Public Health, Boston; and Harvard Kennedy School (JPN), Cambridge, MA
| | - Lidia M V R Moura
- Department of Neurology (LMGC, MBW, SFZ, LMVRM), Massachusetts General Hospital; Department of Neurology (MBW, SFZ, LMVRM), Harvard Medical School; Department of Epidemiology (DB), Harvard T.H. Chan School of Public Health; Department of Psychiatry (DB), Massachusetts General Hospital; Department of Psychiatry (DB), Harvard Medical School; Department of Health Care Policy (JH, JPN), Harvard Medical School; Mongan Institute (JH), Massachusetts General Hospital; Department of Medicine (JH), Harvard Medical School, Boston; National Bureau of Economic Research (JPN), Cambridge; Department of Health Policy and Management (JPN), Harvard T.H. Chan School of Public Health, Boston; and Harvard Kennedy School (JPN), Cambridge, MA
| |
Collapse
|
9
|
Samara QA, Ifraitekh AS, Al Jayyousi O, Sawan S, Hazaimeh E, Jbarah OF. Use of antiepileptic drugs as prophylaxis against posttraumatic seizures in the pediatric population: a systematic review and meta-analysis. Neurosurg Rev 2023; 46:49. [PMID: 36746821 DOI: 10.1007/s10143-023-01963-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023]
Abstract
We aim to assess the effect of anti-epileptic drug (AED) prophylaxis for early or late posttraumatic seizures, targeting the pediatric population with traumatic brain injury (TBI). We systematically searched for studies reporting the incidence of posttraumatic seizures in pediatric patients who suffered from TBI and received AEDs prophylactically following their TBI incident. Studies that included adult patients, adult and pediatric patients but did not report results for the pediatric population separately, and patients who did not suffer from a TBI were excluded. Studies that did not indicate the use of antiepileptic drugs prophylactically following TBI were excluded. A total of 10 studies were included involving 4621 posttraumatic brain injury patients of the pediatric age population (<18). Five studies assessed the effect of prophylaxis on early seizures, four on late seizures and one on any seizure. The mean incidence of posttraumatic seizures with AED prophylaxis was 8% for early seizures and 7.1% for late seizures. Moreover, one study revealed no benefit of AED prophylaxis for early posttraumatic seizures. Meta-analysis revealed a significant difference in the incidence of early posttraumatic seizures with antiepileptic prophylaxis. However, no significant difference for late posttraumatic seizures has been shown. In conclusion, AED prophylaxis seems to be effective against early posttraumatic seizures for the pediatric population, with levetiracetam possibly being more effective. Also, there is no observed benefit for late posttraumatic seizures.
Collapse
Affiliation(s)
- Qais A Samara
- Department of Neurosurgery, Faculty of Medicine, Al-Balqa Applied University, PO Box 206, Al Salt, 19117, Jordan.
| | - Ali S Ifraitekh
- Faculty of Medicine, Hashemite University, PO Box 330127, Zarqa, Jordan
| | - Obada Al Jayyousi
- Faculty of Medicine, Jordan University of Science & Technology, PO Box 3030, 22110, Irbid, Jordan
| | - Siya Sawan
- Faculty of Medicine, University of Jordan, PO Box 11942, Amman, Jordan
| | - Ethar Hazaimeh
- Neurology Department, Faculty of Medicine, Jordan University of Science & Technology, PO Box 3030, 22110, Irbid, Jordan
| | - Omar F Jbarah
- Neurosurgery Department, Faculty of Medicine, Jordan University of Science & Technology, PO Box 3030, 22110, Irbid, Jordan
| |
Collapse
|
10
|
Sødal HF, Storvig G, Tverdal C, Robinson H, Helseth E, Taubøll E. Early post-traumatic seizures in hospitalized patients with traumatic brain injury. Acta Neurol Scand 2022; 146:485-491. [PMID: 35833266 PMCID: PMC9796016 DOI: 10.1111/ane.13670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/06/2022] [Accepted: 06/30/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Early post-traumatic seizures (EPTS) are a well-known complication of traumatic brain injury (TBI). EPTS increase the risk of secondary brain injury and may cause significant challenges during the period of critical care. Routine use of prophylactic anti-seizure medication is controversial due to conflicting reports on efficacy and risk of adverse effects. The purpose of this study was to expand the understanding of EPTS by examining incidence and risk factors in hospitalized patients with TBI. MATERIAL & METHODS Adult patients with TBI and evidence of intracranial injury admitted to Oslo University Hospital between 2015 and 2019 were identified from the Oslo TBI Registry - Neurosurgery. Demographic and clinical data including occurrence of seizures were retrieved from the registry. The patients did not receive routine seizure prophylaxis. Univariate and multivariable logistic regression analyses were used to investigate risk factors associated with EPTS. RESULTS 103 of 1827 patients (5.6%) had new-onset seizures within the first week after TBI. The following factors were in multivariable analyses associated with EPTS; alcohol abuse (odds ratio [OR] 3.6, 95% CI 2.3-5.7, p < .001), moderate and severe brain injury (OR 2.2, 95% CI 1.3-3.8, p = .004 and OR 2.1, 95% CI 1.2-3.6, p = .012), brain contusion (OR 1.6, 95% CI 1.0-2.4, p = .046) and subdural hematoma (OR 1.6, 95% CI 1.0-2.6, p = .052). CONCLUSION In our material, EPTS occurred in 5.6% of hospital-admitted TBI-patients. Alcohol abuse was the most significant risk factor, followed by moderate and severe brain injury. The results of this study contribute to the discussion about preventive treatment of EPTS in certain risk groups.
Collapse
Affiliation(s)
- Hild Flatmark Sødal
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway,Department of NeurologyOslo University HospitalOsloNorway
| | - Gøril Storvig
- Institute of Health and Society, Faculty of MedicineUniversity of OsloOsloNorway,Department of Psychology, Faculty of Social and Educational SciencesNorwegian University of Science and TechnologyTrondheimNorway
| | - Cathrine Tverdal
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway,Department of NeurosurgeryOslo University HospitalOsloNorway
| | | | - Eirik Helseth
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway,Department of NeurosurgeryOslo University HospitalOsloNorway
| | - Erik Taubøll
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway,Department of NeurologyOslo University HospitalOsloNorway
| |
Collapse
|
11
|
Huo X, Xu X, Li M, Xiao L, Wang Y, Li W, Wang C, Sun T. Effectiveness of antiseizure medications therapy in preventing seizures in brain injury patients: A network meta-analysis. Front Pharmacol 2022; 13:1001363. [PMID: 36188582 PMCID: PMC9521202 DOI: 10.3389/fphar.2022.1001363] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/30/2022] [Indexed: 12/03/2022] Open
Abstract
Purpose: To explore the effectiveness of different anti-seizure medications in preventing early and late post-traumatic epilepsy (PTE). The efficacy, treatment-related side-effects, and mortality of the different treatments were compared using a ranking model to identify the optimal treatment. Methods: A comprehensive literature search was performed using Pubmed, Medline, Embase, and Cochrane library databases. All relevant published articles up to 10 March 2022 were evaluated. The quality of the extracted data was assessed using either the Cochrane risk of bias tool or the Newcastle-Ottawa scale. The primary outcome measures were early or late post-traumatic seizures. The secondary outcome measures were mortality, treatment-related adverse effects, length of hospital stay, and length of stay within the intensive care unit (ICU). Results: A total of seven randomized controlled trials and 18 non-randomized controlled trials were included in this network meta-analysis. The trials included six interventions: Phenytoin (PHT)+phenobarbital (PB), levetiracetam (LEV), PHT, PHT-LEV, lacosamide (LCM), and valproate (VPA). All interventions except VPA significantly reduced the rate of early PTE in TBI patients compared with the placebo. Seven studies reported the impact of four treatments (PHT + PB, LEV, PHT, VPA) on late seizures and showed a significant reduction in the incidence of late seizures in patients with TBI compared with placebo. The impact of PHT, LEV, and VPA on mortality was reported in nine studies. PHT had no impact on mortality, but patients treated with both LEV and VPA had higher mortality than those treated with placebo. The treatment-related adverse effects of LEV, PHT, and LCM were reported in five studies. LEV and PHT had higher treatment-related adverse effects incidence than placebo, while LCM had no effect on treatment related-adverse effects. Conclusion: LEV and PHT prevented early and late PTE. PHT also reduced the mortality rate in patients with TBI. Both LEV and PHT had higher treatment-related adverse effects compared with placebo. However, LEV had a slightly lower incidence of treatment-related adverse effects when compared with PHT. Compared with PHT, LEV did not reduce the length of hospital stay but shortened the length of ICU stays. Therefore, based on the findings of this meta-analysis, we speculate that LEV is the best treatment option for TBI patients. However, further high-quality randomized controlled trials are required to confirm these findings.
Collapse
Affiliation(s)
- Xianhao Huo
- Neurosurgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Xingguo Xu
- Neurosurgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Mei Li
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Lifei Xiao
- Neurosurgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Yangyang Wang
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Neurosurgery Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Wenchao Li
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Neurosurgery Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Chaofan Wang
- Neurosurgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Neurosurgery Department, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- *Correspondence: Tao Sun,
| |
Collapse
|
12
|
Rubinos C, Waters B, Hirsch LJ. Predicting and Treating Post-traumatic Epilepsy. Curr Treat Options Neurol 2022. [DOI: 10.1007/s11940-022-00727-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
13
|
Matsuo T, Komori R, Nakatani M, Ochi S, Yokota-Nakatsuma A, Matsumoto J, Takata F, Dohgu S, Ishihara Y, Itoh K. Levetiracetam Suppresses the Infiltration of Neutrophils and Monocytes and Downregulates Many Inflammatory Cytokines during Epileptogenesis in Pilocarpine-Induced Status Epilepticus Mice. Int J Mol Sci 2022; 23:7671. [PMID: 35887020 PMCID: PMC9319101 DOI: 10.3390/ijms23147671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/30/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
Acute brain inflammation after status epilepticus (SE) is involved in blood-brain barrier (BBB) dysfunction and brain edema, which cause the development of post-SE symptomatic epilepsy. Using pilocarpine-induced SE mice, we previously reported that treatment with levetiracetam (LEV) after SE suppresses increased expression levels of proinflammatory mediators during epileptogenesis and prevents the development of spontaneous recurrent seizures. However, it remains unclear how LEV suppresses neuroinflammation after SE. In this study, we demonstrated that LEV suppressed the infiltration of CD11b+CD45high cells into the brain after SE. CD11b+CD45high cells appeared in the hippocampus between 1 and 4 days after SE and contained Ly6G+Ly6C+ and Ly6G-Ly6C+ cells. Ly6G+Ly6C+ cells expressed higher levels of proinflammatory cytokines such as IL-1β and TNFα suggesting that these cells were inflammatory neutrophils. Depletion of peripheral Ly6G+Ly6C+ cells prior to SE by anti-Ly6G antibody (NIMP-R14) treatment completely suppressed the infiltration of Ly6G+Ly6C+ cells into the brain. Proteome analysis revealed the downregulation of a variety of inflammatory cytokines, which exhibited increased expression in the post-SE hippocampus. These results suggest that Ly6G+Ly6C+ neutrophils are involved in the induction of acute brain inflammation after SE. The proteome expression profile of the hippocampus treated with LEV after SE was similar to that after NIMP-R14 treatment. Therefore, LEV may prevent acute brain inflammation after SE by suppressing inflammatory neutrophil infiltration.
Collapse
Affiliation(s)
- Taira Matsuo
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan; (T.M.); (R.K.); (M.N.); (S.O.)
| | - Rie Komori
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan; (T.M.); (R.K.); (M.N.); (S.O.)
| | - Minami Nakatani
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan; (T.M.); (R.K.); (M.N.); (S.O.)
| | - Shiori Ochi
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan; (T.M.); (R.K.); (M.N.); (S.O.)
| | - Aya Yokota-Nakatsuma
- Laboratory of Immunology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan;
| | - Junichi Matsumoto
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (J.M.); (F.T.); (S.D.)
| | - Fuyuko Takata
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (J.M.); (F.T.); (S.D.)
| | - Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan; (J.M.); (F.T.); (S.D.)
| | - Yasuhiro Ishihara
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 739-8521, Japan;
| | - Kouichi Itoh
- Laboratory for Pharmacotherapy and Experimental Neurology, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki 769-2193, Japan; (T.M.); (R.K.); (M.N.); (S.O.)
| |
Collapse
|
14
|
Regulation of Inflammation-Related Genes through Fosl1 Suppression in a Levetiracetam-Treated Pilocarpine-Induced Status Epilepticus Mouse Model. Int J Mol Sci 2022; 23:ijms23147608. [PMID: 35886955 PMCID: PMC9317701 DOI: 10.3390/ijms23147608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/28/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Levetiracetam (LEV) suppresses the upregulation of proinflammatory molecules that occurs during epileptogenesis after status epilepticus (SE). Based on previous studies, LEV likely helps prevent the onset of epilepsy after insults to the brain, unlike other conventional anti-epileptic drugs. Recently, we discovered that the increase in Fosl1 expression that occurs after lipopolysaccharide (LPS) stimulation is suppressed by LEV and that Fosl1 inhibition suppresses inflammation in BV-2 microglial cells. These data indicate that Fosl1 is an important target of LEV and a key factor in preventing epilepsy onset. In this study, we examined the effects of LEV on Fosl1 expression and neuroinflammation in vivo. During epileptogenesis, the post-SE upregulation of hippocampal levels of Fosl1 and many inflammatory factors were suppressed by LEV. Fosl1 expression showed a characteristic pattern different from that of the expression of Fos, an immediate-early gene belonging to the same Fos family. At 2 days after SE, Fosl1 was predominantly expressed in astrocytes but was rarely detected in microglia, whereas Fos expression was distributed in various brain cell types. The expression of A2 astrocyte markers was similar to that of Fosl1 and was significantly suppressed by LEV. These results suggest that LEV may regulate astrocyte reactivity through regulation of Fosl1.
Collapse
|
15
|
Contreras-García IJ, Cárdenas-Rodríguez N, Romo-Mancillas A, Bandala C, Zamudio SR, Gómez-Manzo S, Hernández-Ochoa B, Mendoza-Torreblanca JG, Pichardo-Macías LA. Levetiracetam Mechanisms of Action: From Molecules to Systems. Pharmaceuticals (Basel) 2022; 15:ph15040475. [PMID: 35455472 PMCID: PMC9030752 DOI: 10.3390/ph15040475] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Epilepsy is a chronic disease that affects millions of people worldwide. Antiepileptic drugs (AEDs) are used to control seizures. Even though parts of their mechanisms of action are known, there are still components that need to be studied. Therefore, the search for novel drugs, new molecular targets, and a better understanding of the mechanisms of action of existing drugs is still crucial. Levetiracetam (LEV) is an AED that has been shown to be effective in seizure control and is well-tolerable, with a novel mechanism of action through an interaction with the synaptic vesicle protein 2A (SV2A). Moreover, LEV has other molecular targets that involve calcium homeostasis, the GABAergic system, and AMPA receptors among others, that might be integrated into a single mechanism of action that could explain the antiepileptogenic, anti-inflammatory, neuroprotective, and antioxidant properties of LEV. This puts it as a possible multitarget drug with clinical applications other than for epilepsy. According to the above, the objective of this work was to carry out a comprehensive and integrative review of LEV in relation to its clinical uses, structural properties, therapeutical targets, and different molecular, genetic, and systemic action mechanisms in order to consider LEV as a candidate for drug repurposing.
Collapse
Affiliation(s)
| | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Antonio Romo-Mancillas
- Laboratorio de Diseño Asistido por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico;
| | - Cindy Bandala
- Neurociencia Básica, Instituto Nacional de Rehabilitación LGII, Secretaría de Salud, Ciudad de México 14389, Mexico;
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Sergio R. Zamudio
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico;
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México 06720, Mexico;
| | - Julieta Griselda Mendoza-Torreblanca
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
- Correspondence: (J.G.M.-T.); (L.A.P.-M.); Tel.: +52-55-1084-0900 (ext. 1441) (J.G.M.-T.)
| | - Luz Adriana Pichardo-Macías
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
- Correspondence: (J.G.M.-T.); (L.A.P.-M.); Tel.: +52-55-1084-0900 (ext. 1441) (J.G.M.-T.)
| |
Collapse
|
16
|
The Pharmacokinetics of Levetiracetam in Critically Ill Adult Patients: An Intensive Care Unit Clinical Study. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The aim of this study was to investigate levetiracetam pharmacokinetics in critically ill adult intensive care patients and to identify pathophysiological factors affecting its kinetics. Fourteen critically ill patients in an intensive care unit were enrolled in the study and received intravenous levetiracetam. Blood samples were collected at specific time points to determine the levetiracetam pharmacokinetics. Patient characteristics such as renal function, demographics, disease severity, organ dysfunction, and biochemical laboratory tests were evaluated for their influence on the kinetics of levetiracetam. Estimated glomerular filtration rate (eGFR) had a statistically significant (p = 0.001) effect on levetiracetam clearance. None of the other patient characteristics had a statistically significant effect on the pharmacokinetics. Simulations of dosing regimens revealed that even typically administered doses of levetiracetam may result in significantly increased concentrations and risk of drug toxicity in patients with impaired renal function. The Acute Physiology and Chronic Health Evaluation II (APACHE II) score differed significantly among the three groups with different epileptic activity (p = 0.034). The same groups also differed in terms of renal function (p = 0.031). Renal dysfunction should be considered when designing levetiracetam dosage. Patients with a low APACHE II score had the lowest risk of experiencing epileptic seizures.
Collapse
|
17
|
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
|
18
|
Punia V, Honomichl R, Chandan P, Ellison L, Thompson N, Sivaraju A, Katzan I, George P, Newey C, Hantus S. Long-term continuation of anti-seizure medications after acute stroke. Ann Clin Transl Neurol 2021; 8:1857-1866. [PMID: 34355539 PMCID: PMC8419404 DOI: 10.1002/acn3.51440] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/01/2021] [Accepted: 07/26/2021] [Indexed: 11/24/2022] Open
Abstract
Objective To investigate the factors associated with the long‐term continuation of anti‐seizure medications (ASMs) in acute stroke patients. Methods We performed a retrospective cohort study of stroke patients with concern for acute symptomatic seizures (ASySs) during hospitalization who subsequently visited the poststroke clinic. All patients had continuous EEG (cEEG) monitoring. We generated a multivariable logistic regression model to analyze the factors associated with the primary outcome of continued ASM use after the first poststroke clinic visit. Results A total of 507 patients (43.4% ischemic stroke, 35.7% intracerebral hemorrhage, and 20.9% aneurysmal subarachnoid hemorrhage) were included. Among them, 99 (19.5%) suffered from ASySs, 110 (21.7%) had epileptiform abnormalities (EAs) on cEEG, and 339 (66.9%) had neither. Of the 294 (58%) patients started on ASMs, 171 (33.7%) were discharged on them, and 156 (30.3% of the study population; 53.1% of patients started on ASMs) continued ASMs beyond the first poststroke clinic visit [49.7 (±31.7) days after cEEG]. After adjusting for demographical, stroke‐ and hospitalization‐related variables, the only independent factors associated with the primary outcome were admission to the NICU [Odds ratio (OR) 0.37 (95% CI 0.15–0.9)], the presence of ASySs [OR 20.31(95% CI 9.45–48.43)], and EAs on cEEG [OR 2.26 (95% CI 1.14–4.58)]. Interpretation Almost a third of patients with poststroke ASySs concerns may continue ASMs for the long term, including more than half started on them acutely. Admission to the NICU may lower the odds, and ASySs (convulsive or electrographic) and EAs on cEEG significantly increase the odds of long‐term ASM use.
Collapse
Affiliation(s)
- Vineet Punia
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ryan Honomichl
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Center for Outcomes Research and Evaluation, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Pradeep Chandan
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Lisa Ellison
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicolas Thompson
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Center for Outcomes Research and Evaluation, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Adithya Sivaraju
- Comprehensive Epilepsy Center, Department of Neurology, Yale University, New Haven, Connecticut, USA
| | - Irene Katzan
- Center for Outcomes Research and Evaluation, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Cerebrovascular Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Pravin George
- Cerebrovascular Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Chris Newey
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Cerebrovascular Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Stephen Hantus
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
19
|
Levetiracetam for Seizure Prophylaxis in Neurocritical Care: A Systematic Review and Meta-analysis. Neurocrit Care 2021; 36:248-258. [PMID: 34286461 DOI: 10.1007/s12028-021-01296-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/09/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Levetiracetam is commonly used for seizure prophylaxis in patients with intracerebral hemorrhage (ICH), traumatic brain injury (TBI), supratentorial neurosurgery, and spontaneous subarachnoid hemorrhage (SAH). However, its efficacy, optimal dosing, and the adverse events associated with levetiracetam prophylaxis remain unclear. METHODS A systematic search of PubMed, Embase, and Cochrane central register of controlled trials (CENTRAL) database was conducted from January 1, 2000, to October 30, 2020, including articles addressing treatment with levetiracetam for seizure prophylaxis after SAH, ICH, TBI, and supratentorial neurosurgery. Non-English, pediatric (aged < 18 years), preclinical, reviews, case reports, and articles that included patients with a preexisting seizure condition or epilepsy were excluded. The coprimary meta-analyses examined first seizure events in (1) levetiracetam versus no antiseizure medication and (2) levetiracetam versus other antiseizure medications in all ICH, TBI, SAH, and supratentorial neurosurgery populations. Secondary meta-analyses evaluated the same comparator groups in individual disease populations. Risk of bias in non-randomised studies - of interventions (ROBINS-I) and risk-of-bias tool for randomized trials (RoB-2) tools were used to assess risk of bias. RESULTS A total of 30 studies (n = 6 randomized trials, n = 9 prospective studies, and n = 15 retrospective studies), including 7609 patients (n = 4737 with TBI, n = 701 with SAH, n = 261 with ICH, and n = 1910 with neurosurgical diseases) were included in analyses. Twenty-seven of 30 (90%) studies demonstrated moderate to severe risk of bias, and 11 of 30 (37%) studies used low-dosage levetiracetam (250-500 mg twice daily). In the primary meta-analyses, there were no differences in seizure events for levetiracetam prophylaxis (n = 906) versus no antiseizure medication (n = 2728; odds ratio [OR] 0.79, 95% confidence interval [CI] 0.53-1.16, P = 0.23, fixed-effect, I2 = 26%, P = 0.23 for heterogeneity) or levetiracetam (n = 1950) versus other antiseizure prophylaxis (n = 2289; OR 0.84, 95% CI 0.55-1.28, P = 0.41, random-effects, I2 = 49%, P = 0.005 for heterogeneity). Only patients with supratentorial neurosurgical diseases benefited from levetiracetam compared with other antiseizure medications (median 0.70 seizure events per-patient-year with levetiracetam versus 2.20 seizure events per-patient-year for other antiseizure medications, OR 0.34, 95% CI 0.20-0.58, P < 0.001, fixed-effects, I2 = 39%, P = 0.13 for heterogeneity). There were no significant differences in meta-analyses of patients with ICH, SAH, or TBI. Adverse events of any severity were reported in a median of 8% of patients given levetiracetam compared with 21% of patients in comparator groups. CONCLUSIONS Based on the current moderately to seriously biased heterogeneous data, which frequently used low and possibly subtherapeutic doses of levetiracetam, our meta-analyses did not demonstrate significant reductions in seizure incidence and neither supports nor refutes the use of levetiracetam prophylaxis in TBI, SAH, or ICH. Levetiracetam may be preferred post supratentorial neurosurgery. More high-quality randomized trials of prophylactic levetiracetam are warranted.
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
|
Qi H, Li K. Civilian gunshot wounds to the head: a case report, clinical management, and literature review. Chin Neurosurg J 2021; 7:12. [PMID: 33531086 PMCID: PMC7856761 DOI: 10.1186/s41016-020-00227-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/15/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Civilian gunshot wounds to the head refer to brain injury caused by projectiles such as gun projectiles and various fragments generated by explosives in a power launch or explosion. Gunshot wounds to the head are the deadliest of all gun injuries. According to literature statistics, the survival rate of patients with gunshot wounds to the head is only 9%. Due to the strict management of various types of firearms, they rarely occur, so the injury mechanism, injury and trauma analysis, clinical management, and surgical standards are almost entirely based on military experience, and there are few related reports, especially of the head, in which an individual suffered a fatal blow more than once in a short time. We report a case with a return to almost complete recovery despite the patient suffering two gunshot injuries to the head in a short period of time. CASE PRESENTATIONS We present a case of a 53-year-old man who suffered two gunshot injuries to the head under unknown circumstances. On initial presentation, the patient had a Glasgow Coma Scale score of 6, was unable to communicate, and had loss of consciousness. The first bullet penetrated the right frontal area and finally reached the right occipital lobe. When the patient reflexively shielded his head with his hand, the second bullet passed through the patient's right palm bone, entered the right frontotemporal area, and came to rest deep in the lateral sulcus. The patient had a cerebral hernia when he was admitted to the hospital and immediately entered the operating room for rescue after a computed tomography scan. After two foreign body removals and skull repair, the patient recovered completely. CONCLUSIONS Gunshot wounds to the head have a high mortality rate and usually require aggressive management. Evaluation of most gunshot injuries requires extremely fast imaging examination upon arrival at the hospital, followed by proactive treatment against infection, seizure, and increased intracranial pressure. Surgical intervention is usually necessary, and its key points include the timing, method, and scope of the operation.
Collapse
Affiliation(s)
- Haoyi Qi
- Qinghai University, No. 251 Ningda Road, Xining, 810016 Qinghai Province China
| | - Kunzheng Li
- The Affiliated Hospital of Qinghai University, No. 29 Tongren Road, Xining, 810000 Qinghai Province China
| |
Collapse
|
22
|
Yang L, Afroz S, Valsamis HA, Michelson HB, Goodman JH, Ling DSF. Early intervention with levetiracetam prevents the development of cortical hyperexcitability and spontaneous epileptiform activity in two models of neurotrauma in rats. Exp Neurol 2020; 337:113571. [PMID: 33340499 DOI: 10.1016/j.expneurol.2020.113571] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/09/2020] [Accepted: 12/13/2020] [Indexed: 10/22/2022]
Abstract
This study examined the antiepileptogenic potential of the antiseizure drug (ASD) levetiracetam (LEV) using the in vitro traumatized-slice and in vivo controlled cortical impact (CCI) models of traumatic brain injury (TBI) in rats when administered early after the injury. For the in vitro model, acute coronal slices (400-450 μm) of rat neocortex (P21-32) were injured via a surgical cut that separated the superficial layers from the deeper regions. Persistent stimulus-evoked epileptiform activity developed within 1-2 h after trauma. In randomly selected slices, LEV (500 μM) was bath-applied for 1 h starting immediately or delayed by 30-80 min after injury. Treated and untreated slices were examined for epileptiform activity via intracellular and extracellular recordings. For the in vivo model, rats (P24-32) were subjected to a non-penetrating, focal, CCI injury targeting the neocortex (5.0 mm diameter; 2.0 mm depth). Immediately after injury, rats were given either a single dose of LEV (60-150 mg/kg, i.p.) or the saline vehicle. At 2-3 weeks after the injury, ex vivo cortical slices were examined for epileptiform activity. The results from the traumatized-slice experiments showed that in vitro treatment with LEV within 60 min of injury significantly reduced (> 50%) the proportion of slices that exhibited stimulus-evoked epileptiform activity. LEV treatment also increased the stimulus intensity required to trigger epileptiform bursts in injured slices by 2-4 fold. Consistent with these findings, LEV treatment of CCI-injured rats (n = 15) significantly reduced the proportion of animals that exhibited spontaneous and stimulus-evoked epileptiform bursts in ex vivo cortical slices compared to saline-treated controls (n = 15 rats), and also significantly increased the stimulus intensity required to evoke epileptiform bursts. These results suggest that early administration of LEV has the potential to prevent or reduce posttraumatic epileptogenesis and that there may be a narrow therapeutic window for successful prophylactic intervention.
Collapse
Affiliation(s)
- Lie Yang
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA
| | - Sonia Afroz
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Program in Neural and Behavioral Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA
| | - Helen A Valsamis
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Department of Neurology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 1213, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Neurology Service, Kings County Hospital Center, Brooklyn, NY 11203, USA.
| | - Hillary B Michelson
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA.
| | - Jeffrey H Goodman
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Department of Neurology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 1213, Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Department of Developmental Neurobiology, The New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA.
| | - Douglas S F Ling
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA; Program in Neural and Behavioral Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Brooklyn, NY 11203, USA; The Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, 450 Clarkson Ave., Box 29, Brooklyn, NY 11203, USA.
| |
Collapse
|
23
|
Shakkour Z, Habashy KJ, Berro M, Takkoush S, Abdelhady S, Koleilat N, Eid AH, Zibara K, Obeid M, Shear D, Mondello S, Wang KK, Kobeissy F. Drug Repurposing in Neurological Disorders: Implications for Neurotherapy in Traumatic Brain Injury. Neuroscientist 2020; 27:620-649. [PMID: 33089741 DOI: 10.1177/1073858420961078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) remains a significant leading cause of death and disability among adults and children globally. To date, there are no Food and Drug Administration-approved drugs that can substantially attenuate the sequelae of TBI. The innumerable challenges faced by the conventional de novo discovery of new pharmacological agents led to the emergence of alternative paradigm, which is drug repurposing. Repurposing of existing drugs with well-characterized mechanisms of action and human safety profiles is believed to be a promising strategy for novel drug use. Compared to the conventional discovery pathways, drug repurposing is less costly, relatively rapid, and poses minimal risk of the adverse outcomes to study on participants. In recent years, drug repurposing has covered a wide range of neurodegenerative diseases and neurological disorders including brain injury. This review highlights the advances in drug repurposing and presents some of the promising candidate drugs for potential TBI treatment along with their possible mechanisms of neuroprotection. Edaravone, glyburide, ceftriaxone, levetiracetam, and progesterone have been selected due to their potential role as putative TBI neurotherapeutic agents. These drugs are Food and Drug Administration-approved for purposes other than brain injuries; however, preclinical and clinical studies have shown their efficacy in ameliorating the various detrimental outcomes of TBI.
Collapse
Affiliation(s)
- Zaynab Shakkour
- Department of Biochemistry & Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Moussa Berro
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samira Takkoush
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nadia Koleilat
- Division of Child Neurology, Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Makram Obeid
- Division of Child Neurology, Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon.,Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Deborah Shear
- Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Sicilia, Italy
| | - Kevin K Wang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, USA
| | - Firas Kobeissy
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Schidlitzki A, Bascuñana P, Srivastava PK, Welzel L, Twele F, Töllner K, Käufer C, Gericke B, Feleke R, Meier M, Polyak A, Ross TL, Gerhauser I, Bankstahl JP, Johnson MR, Bankstahl M, Löscher W. Proof-of-concept that network pharmacology is effective to modify development of acquired temporal lobe epilepsy. Neurobiol Dis 2019; 134:104664. [PMID: 31678583 DOI: 10.1016/j.nbd.2019.104664] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/08/2019] [Accepted: 10/30/2019] [Indexed: 10/25/2022] Open
Abstract
Epilepsy is a complex network phenomenon that, as yet, cannot be prevented or cured. We recently proposed network-based approaches to prevent epileptogenesis. For proof of concept we combined two drugs (levetiracetam and topiramate) for which in silico analysis of drug-protein interaction networks indicated a synergistic effect on a large functional network of epilepsy-relevant proteins. Using the intrahippocampal kainate mouse model of temporal lobe epilepsy, the drug combination was administered during the latent period before onset of spontaneous recurrent seizures (SRS). When SRS were periodically recorded by video-EEG monitoring after termination of treatment, a significant decrease in incidence and frequency of SRS was determined, indicating antiepileptogenic efficacy. Such efficacy was not observed following single drug treatment. Furthermore, a combination of levetiracetam and phenobarbital, for which in silico analysis of drug-protein interaction networks did not indicate any significant drug-drug interaction, was not effective to modify development of epilepsy. Surprisingly, the promising antiepileptogenic effect of the levetiracetam/topiramate combination was obtained in the absence of any significant neuroprotective or anti-inflammatory effects as indicated by multimodal brain imaging and histopathology. High throughput RNA-sequencing (RNA-seq) of the ipsilateral hippocampus of mice treated with the levetiracetam/topiramate combination showed that several genes that have been linked previously to epileptogenesis, were significantly differentially expressed, providing interesting entry points for future mechanistic studies. Overall, we have discovered a novel combination treatment with promise for prevention of epilepsy.
Collapse
Affiliation(s)
- Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Pablo Bascuñana
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | | | - Lisa Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Birthe Gericke
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Rahel Feleke
- Division of Brain Sciences, Imperial College London, London, UK
| | - Martin Meier
- Central Animal Facility & Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Andras Polyak
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Germany
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | | | - Marion Bankstahl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany; Central Animal Facility & Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
27
|
Klein P, Tyrlikova I. No prevention or cure of epilepsy as yet. Neuropharmacology 2019; 168:107762. [PMID: 31499048 DOI: 10.1016/j.neuropharm.2019.107762] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/02/2019] [Accepted: 09/05/2019] [Indexed: 12/29/2022]
Abstract
Approximately 20% of all epilepsy is caused by acute acquired injury such as traumatic brain injury, stroke and CNS infection. The known onset of the injury which triggers the epileptogenic process, early presentation to medical care, and a latency between the injury and the development of clinical epilepsy present an opportunity to intervene with treatment to prevent epilepsy. No such treatment exists and yet there has been remarkably little clinical research during the last 20 years to try to develop such treatment. We review possible reasons for this, possible ways to rectify the situations and note some of the ways currently under way to do so. Resective surgical treatment can achieve "cure" in some patients but is sparsely utilized. In certain "self-limiting" syndromes of childhood and adolescence epilepsy remits spontaneously. In a proportion of patients who become seizure free on medications or with dietary treatment, seizure freedom persists when treatment is discontinued. We discuss these situations which can be considered "cures"; and note that at present we have little understanding of mechanism of such cures, and cannot therefore translate them into a treatment paradigm targeting a "cure" of epilepsy. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA.
| | | |
Collapse
|
28
|
Carvill GL, Dulla CG, Lowenstein DH, Brooks-Kayal AR. The path from scientific discovery to cures for epilepsy. Neuropharmacology 2019; 167:107702. [PMID: 31301334 DOI: 10.1016/j.neuropharm.2019.107702] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 02/06/2023]
Abstract
The epilepsies are a complex group of disorders that can be caused by a myriad of genetic and acquired factors. As such, identifying interventions that will prevent development of epilepsy, as well as cure the disorder once established, will require a multifaceted approach. Here we discuss the progress in scientific discovery propelling us towards this goal, including identification of genetic risk factors and big data approaches that integrate clinical and molecular 'omics' datasets to identify common pathophysiological signatures and biomarkers. We discuss the many animal and cellular models of epilepsy, what they have taught us about pathophysiology, and the cutting edge cellular, optogenetic, chemogenetic and anti-seizure drug screening approaches that are being used to find new cures in these models. Finally, we reflect on the work that still needs to be done towards identify at-risk individuals early, targeting and stopping epileptogenesis, and optimizing promising treatment approaches. Ultimately, developing and implementing cures for epilepsy will require a coordinated and immense effort from clinicians and basic scientists, as well as industry, and should always be guided by the needs of individuals affected by epilepsy and their families. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Gemma L Carvill
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.
| | - Dan H Lowenstein
- Department of Neurology, University of California, San Francisco, CA, 94941, USA
| | - Amy R Brooks-Kayal
- Department of Pediatrics and Neurology, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA
| |
Collapse
|
29
|
McGuire JL, Ngwenya LB, McCullumsmith RE. Neurotransmitter changes after traumatic brain injury: an update for new treatment strategies. Mol Psychiatry 2019; 24:995-1012. [PMID: 30214042 DOI: 10.1038/s41380-018-0239-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 08/15/2018] [Accepted: 08/20/2018] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) is a pervasive problem in the United States and worldwide, as the number of diagnosed individuals is increasing yearly and there are no efficacious therapeutic interventions. A large number of patients suffer with cognitive disabilities and psychiatric conditions after TBI, especially anxiety and depression. The constellation of post-injury cognitive and behavioral symptoms suggest permanent effects of injury on neurotransmission. Guided in part by preclinical studies, clinical trials have focused on high-yield pathophysiologic mechanisms, including protein aggregation, inflammation, metabolic disruption, cell generation, physiology, and alterations in neurotransmitter signaling. Despite successful treatment of experimental TBI in animal models, clinical studies based on these findings have failed to translate to humans. The current international effort to reshape TBI research is focusing on redefining the taxonomy and characterization of TBI. In addition, as the next round of clinical trials is pending, there is a pressing need to consider what the field has learned over the past two decades of research, and how we can best capitalize on this knowledge to inform the hypotheses for future innovations. Thus, it is critically important to extend our understanding of the pathophysiology of TBI, particularly to mechanisms that are associated with recovery versus development of chronic symptoms. In this review, we focus on the pathology of neurotransmission after TBI, reflecting on what has been learned from both the preclinical and clinical studies, and we discuss new directions and opportunities for future work.
Collapse
Affiliation(s)
- Jennifer L McGuire
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, USA.
| | - Laura B Ngwenya
- Department of Neurosurgery, University of Cincinnati, Cincinnati, OH, USA.,Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA.,Neurotrauma Center, University of Cincinnati Gardner Neuroscience Institute, Cincinnati, OH, 45219, USA
| | - Robert E McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA.,Department of Psychiatry, Cincinnati Veterans Administration Medical Center, Cincinnati, OH, USA
| |
Collapse
|
30
|
Abstract
Introduction: Brivaracetam (BRV) is an analog of levetiracetam (LEV) with 15-30 times greater affinity to SV2A and greater brain permeability than LEV. These properties have stimulated interest in its clinical trial data and post-marketing experience. Areas covered: The authors provide a background on epilepsy and its treatment, discuss the racetam family of antiepileptic drugs to which BRV belongs, and then discuss BRV properties and its efficacy and tolerability in the treatment of epilepsy. Expert opinion: While preclinical data suggest a broad spectrum of efficacy, BRV is only approved for focal epilepsy. The recommended starting dose is 100 mg per day, but in the absence of urgency, it may be prudent to start at 50 mg per day, considered the lowest effective dose. There was no added benefit when BRV was used adjunctively with LEV in clinical trials. However, post-marketing data suggest that some patients may experience improved seizure control when switching from LEV. Behavioral adverse effects seemed less common than with LEV, and most patients switched to BRV after experiencing behavioral adverse effects on LEV reported improvement. Prior or anticipated intolerability to LEV is the strongest indication for BRV in clinical practice.
Collapse
Affiliation(s)
- Yamane Makke
- Department of Neurology, Vanderbilt University Medical Center , Nashville , TN , USA
| | - Bassel Abou-Khalil
- Department of Neurology, Vanderbilt University Medical Center , Nashville , TN , USA
| |
Collapse
|
31
|
Löscher W. The holy grail of epilepsy prevention: Preclinical approaches to antiepileptogenic treatments. Neuropharmacology 2019; 167:107605. [PMID: 30980836 DOI: 10.1016/j.neuropharm.2019.04.011] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
Abstract
A variety of acute brain insults can induce epileptogenesis, a complex process that results in acquired epilepsy. Despite advances in understanding mechanisms of epileptogenesis, there is currently no approved treatment that prevents the development or progression of epilepsy in patients at risk. The current concept of epileptogenesis assumes a window of opportunity following acute brain insults that allows intervention with preventive treatment. Recent results suggest that injury-induced epileptogenesis can be a much more rapid process than previously thought, suggesting that the 'therapeutic window' may only be open for a brief period, as in stroke therapy. However, experimental data also suggest a second, possibly delayed process ("secondary epileptogenesis") that influences the progression and refractoriness of the epileptic state over time, allowing interfering with this process even after onset of epilepsy. In this review, both methodological issues in preclinical drug development and novel targets for antiepileptogenesis will be discussed. Several promising drugs that either prevent epilepsy (antiepileptogenesis) or slow epilepsy progression and alleviate cognitive or behavioral comorbidities of epilepsy (disease modification) have been described in recent years, using diverse animal models of acquired epilepsy. Promising agents include TrkB inhibitors, losartan, statins, isoflurane, anti-inflammatory and anti-oxidative drugs, the SV2A modulator levetiracetam, and epigenetic interventions. Research on translational target validity and on prognostic biomarkers that can be used to stratify patients (or experimental animals) at high risk of developing epilepsy will hopefully soon lead to proof-of-concept clinical trials with the most promising drugs, which will be essential to make prevention of epilepsy a reality. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
32
|
Guidelines for the Management of Pediatric Severe Traumatic Brain Injury, Third Edition: Update of the Brain Trauma Foundation Guidelines. Pediatr Crit Care Med 2019; 20:S1-S82. [PMID: 30829890 DOI: 10.1097/pcc.0000000000001735] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
33
|
Welzel L, Twele F, Schidlitzki A, Töllner K, Klein P, Löscher W. Network pharmacology for antiepileptogenesis: Tolerability and neuroprotective effects of novel multitargeted combination treatments in nonepileptic vs. post-status epilepticus mice. Epilepsy Res 2019; 151:48-66. [PMID: 30831337 DOI: 10.1016/j.eplepsyres.2019.02.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/06/2019] [Accepted: 02/23/2019] [Indexed: 01/08/2023]
Abstract
Network-based approaches in drug discovery comprise both development of novel drugs interacting with multiple targets and repositioning of drugs with known targets to form novel drug combinations that interact with cellular or molecular networks whose function is disturbed in a disease. Epilepsy is a complex network phenomenon that, as yet, cannot be prevented or cured. We recently proposed multitargeted, network-based approaches to prevent epileptogenesis by combinations of clinically available drugs chosen to impact diverse epileptogenic processes. In order to test this strategy preclinically, we developed a multiphase sequential study design for evaluating such drug combinations in rodents, derived from human clinical drug development phases. Because pharmacokinetics of such drugs are known, only the tolerability of novel drug combinations needs to be evaluated in Phase I in öhealthy" controls. In Phase IIa, tolerability is assessed following an epileptogenic brain insult, followed by antiepileptogenic efficacy testing in Phase IIb. Here, we report Phase I and Phase IIa evaluation of 7 new drug combinations in mice, using 10 drugs (levetiracetam, topiramate, gabapentin, deferoxamine, fingolimod, ceftriaxone, α-tocopherol, melatonin, celecoxib, atorvastatin) with diverse mechanisms thought to be important in epileptogenesis. Six of the 7 drug combinations were well tolerated in mice during prolonged treatment at the selected doses in both controls and during the latent phase following status epilepticus induced by intrahippocampal kainate. However, none of the combinations prevented hippocampal damage in response to kainate, most likely because treatment started only 16-18 h after kainate. This suggests that antiepileptogenic or disease-modifying treatment may need to start earlier after the brain insult. The present data provide a rich collection of tolerable, network-based combinatorial therapies as a basis for antiepileptogenic or disease-modifying efficacy testing.
Collapse
Affiliation(s)
- Lisa Welzel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD 20817, USA
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
34
|
Kumar RG, Breslin KB, Ritter AC, Conley YP, Wagner AK. Variability with Astroglial Glutamate Transport Genetics Is Associated with Increased Risk for Post-Traumatic Seizures. J Neurotrauma 2019; 36:230-238. [PMID: 29999457 PMCID: PMC6338569 DOI: 10.1089/neu.2018.5632] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Excitotoxicity contributes to epileptogenesis after severe traumatic brain injury (sTBI). Demographic and clinical risk factors for post-traumatic seizures (PTS) have been identified, but genetic risk remains largely unknown. Thus, we investigated whether genetic variation in astroglial glutamate transporter genes is associated with accelerated epileptogenesis and PTS risk after sTBI. Adults (n = 267) 18-75 years old were assessed over a three-year period post-TBI. Single nucleotide polymorphisms (SNPs) throughout the SLC1A2 and SLC1A3 genes were assayed. Kaplan-Meier estimates and log-rank statistics were used to compare seizure frequencies by genotype. Multivariate Cox proportional hazards regression was used to estimate hazard ratios (HRs) for genotypes significant in Kaplan-Meier analyses. Thirty-nine tagging SNPs were examined (SLC1A2: n = 21, SLC1A3: n = 18). PTS developed in 57 (21.4%) individuals. Of those with PTS, n = 20 (35.7%) had an immediate/early seizure within the first seven days, and n = 36 (64.3%) had a late seizure occurring between eight days and three years post-TBI. When adjusting for multiple comparisons, rs4869682 genotypes (SLC1A3, GG vs. T-carriers) were associated with time to first seizure (p = 0.003). Median time until first seizure was 20.4 days for individuals with a GG genotype and 44.8 days for T-carriers. After adjusting for covariates, rs4869682 GG-homozygotes had a 2.05 times increased PTS risk versus T-carriers (aHR = 2.08, 95% confidence interval: 1.20, 3.62, p = 0.009). Variation within SLC1A3 is associated with accelerated epileptogenesis and clinical PTS development after sTBI. Future studies should validate these findings and examine how genetic variation at rs4869682 may be a target for PTS prevention and treatment.
Collapse
Affiliation(s)
- Raj G. Kumar
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kristen B. Breslin
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anne C. Ritter
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yvette P. Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Amy K. Wagner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neuroscience, and University of Pittsburgh, Pittsburgh, Pennsylvania
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
35
|
Hazama A, Ripa V, Kwon CS, Abouelleil M, Hall W, Chin L. Full Recovery After a Bihemispheric Gunshot Wound to the Head: Case Report, Clinical Management, and Literature Review. World Neurosurg 2018; 117:309-314. [PMID: 29959075 DOI: 10.1016/j.wneu.2018.06.132] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND Nearly 33,600 people die each year in the United States as a result of gunshot wounds (GSWs). Penetrating craniocerebral GSWs are often fatal with a nearly 70% death rate at the scene of the trauma. Overall combined mortality rate for patients who die at the scene or at the hospital is almost 91%. Poor outcome is associated with initial low Glasgow Coma Scale score and bihemispheric and transventricular gunshot trajectory. We summarize current understanding in management, prognostic factors, and survival outcomes in patients with a penetrating GSWs to the head. We report a patient with return to full function despite bihemispheric, multilobar involvement. Full function is defined here as ability to return to previous work and perform activities of daily living. CASE DESCRIPTION A 33-year-old man sustained a GSW to the head under unknown circumstances. On initial presentation, he had a Glasgow Coma Scale score of 15. He was verbalizing and communicating but was amnestic for the event. From a left frontal entry wound, the bullet traversed both frontal lobes of the brain reaching the right frontal-parietal junction. Physical examination and vital signs were normal. Appropriate surgical and medical management resulted in complete recovery. CONCLUSIONS Craniocerebral GSWs have a high mortality rate and usually require aggressive management. Evaluation of most GSWs requires appropriate imaging studies followed by proactive treatment against infection, seizure, and increased intracranial pressure. Surgical intervention is often necessary and ranges from local wound débridement to craniectomy, decompression, and wound exploration.
Collapse
Affiliation(s)
- Ali Hazama
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, New York, USA.
| | | | - Churl-Su Kwon
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Walter Hall
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Lawrence Chin
- Department of Neurosurgery, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
36
|
Bakr A, Belli A. A systematic review of levetiracetam versus phenytoin in the prevention of late post-traumatic seizures and survey of UK neurosurgical prescribing practice of antiepileptic medication in acute traumatic brain injury. Br J Neurosurg 2018; 32:237-244. [PMID: 29688078 DOI: 10.1080/02688697.2018.1464118] [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] [Indexed: 10/17/2022]
Abstract
BACKGROUND Guidelines recommend 1 week of prophylactic phenytoin for post-traumatic seizures (PTS). Levetiracetam is gaining popularity as an alternative with a superior side-effect profile and may be suitable for extended use. We performed a systematic review comparing the efficacy of levetiracetam and phenytoin in reducing the incidence of late PTS. The secondary objectives were to compare their effects on the Extended Glasgow Outcome Scale (GOS-E) and length of stay. We also aimed to survey current prophylaxis prescribing practices. METHODS A systematic review was performed using Medline, Pubmed, Embase and Cochrane. Trials and observational studies comparing the efficacy of phenytoin and levetiracetam in the prevention of late PTS were included. A survey assessing prescribing practices was e-mailed to all consultant members of the Society of British Neurological Surgeons (n = 249) in March 2013. RESULTS One randomised controlled trial (RCT) (52 patients) and a cohort study (19 patients) met our criteria. Neither found a significant difference in the incidence of late PTS or length of hospital stay, although the RCT showed an improvement in the GOS-E with levetiracetam. Of the 249 consultants included in the survey, 55 responded (22.1%). Prophylaxis was prescribed by 32 consultants (58%), of whom 21 (65.6%) chose phenytoin, 7 (21.9%) chose levetiracetam, 3 (9.4%) chose valproate and 1 (3%) chose 'other'. Half indicated they would prescribe prophylaxis for 1 week, the remainder opting for extended use. CONCLUSION While our review found no evidence of a difference in late seizure incidence, there is evidence of improved long-term outcomes with levetiracetam. Neither study used an extended course of levetiracetam or continuous electroencephalography. Further research which accounts for these factors is required for the development of guidelines which take levetiracetam into account. Our survey showed a lack of awareness of the potential harms of extended phenytoin use and a move towards levetiracetam.
Collapse
Affiliation(s)
- Ahmed Bakr
- a College of Medical and Dental Sciences , University of Birmingham , Birmingham , UK
| | - Antonio Belli
- a College of Medical and Dental Sciences , University of Birmingham , Birmingham , UK.,b Department of Neurosurgery , Queen Elizabeth Hospital Birmingham , Birmingham , UK
| |
Collapse
|
37
|
Seizure Prophylaxis Guidelines Following Traumatic Brain Injury: An Evaluation of Compliance. J Head Trauma Rehabil 2018; 32:E13-E17. [PMID: 27323218 DOI: 10.1097/htr.0000000000000243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To determine degree of adherence to guidelines for seizure prophylaxis following traumatic brain injury (TBI). SETTING Tertiary care level 1 trauma center and affiliated inpatient rehabilitation facility. PARTICIPANTS A total of 173 individuals with TBI who required inpatient rehabilitation from January 1, 2007, to December 31, 2013. DESIGN Retrospective medical record review. MAIN MEASURES Overutilization rate of prophylactic antiepileptic drugs (AEDs); failure to stop rate of AED utilization upon admission to and during inpatient rehabilitation; and duration of overutilization. RESULTS Of the 173 participants included, 77 were started on seizure prophylaxis at hospital presentation and 96 were not. Of the 77 participants, 11 had a posttraumatic seizure. Of the 66 remaining, 18 participants (10.4%) were continued on AEDs for more than 7 days after injury. Of these 18 participants, 12 were continued on AEDs without indication upon admission to inpatient rehabilitation. Finally, 8 of the 12 were continued on AEDs at discharge from rehabilitation, resulting in a failure to stop rate of 66.67%. CONCLUSION Despite existing guidelines for stopping seizure prophylaxis after TBI, some patients remain on AEDs and may be inappropriately exposed to possible medication side effects. These findings highlight the importance of communication at the time of rehabilitation transfer and the need for ongoing education about AED guidelines.
Collapse
|
38
|
Wilson CD, Burks JD, Rodgers RB, Evans RM, Bakare AA, Safavi-Abbasi S. Early and Late Posttraumatic Epilepsy in the Setting of Traumatic Brain Injury: A Meta-analysis and Review of Antiepileptic Management. World Neurosurg 2018; 110:e901-e906. [DOI: 10.1016/j.wneu.2017.11.116] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 11/17/2022]
|
39
|
Łukawski K, Andres-Mach M, Czuczwar M, Łuszczki JJ, Kruszyński K, Czuczwar SJ. Mechanisms of epileptogenesis and preclinical approach to antiepileptogenic therapies. Pharmacol Rep 2017; 70:284-293. [PMID: 29477036 DOI: 10.1016/j.pharep.2017.07.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/17/2017] [Accepted: 07/12/2017] [Indexed: 12/30/2022]
Abstract
The prevalence of epilepsy is estimated 5-10 per 1000 population and around 70% of patients with epilepsy can be sufficiently controlled by antiepileptic drugs (AEDs). Epileptogenesis is the process responsible for converting normal into an epileptic brain and mechanisms responsible include among others: inflammation, neurodegeneration, neurogenesis, neural reorganization and plasticity. Some AEDs may be antiepileptiogenic (diazepam, eslicarbazepine) but the correlation between neuroprotection and inhibition of epileptogenesis is not evident. Antiepileptogenic activity has been postulated for mTOR ligands, resveratrol and losartan. So far, clinical evidence gives some hope for levetiracetam as an AED inhibiting epileptogenesis in neurosurgical patients. Biomarkers for epileptogenesis are needed for the proper selection of patients for evaluation of potential antiepileptogenic compounds.
Collapse
Affiliation(s)
- Krzysztof Łukawski
- Department of Physiopathology, Institute of Rural Health, Lublin, Poland; Department of Pathophysiology, Medical University of Lublin, Lublin, Poland
| | - Marta Andres-Mach
- Isobolographic Analysis Laboratory, Institute of Rural Health, Lublin, Poland
| | - Mirosław Czuczwar
- 2nd Department of Anesthesiology and Intensive Care, Medical University of Lublin, Lublin, Poland
| | - Jarogniew J Łuszczki
- Department of Pathophysiology, Medical University of Lublin, Lublin, Poland; Isobolographic Analysis Laboratory, Institute of Rural Health, Lublin, Poland
| | | | - Stanisław J Czuczwar
- Department of Physiopathology, Institute of Rural Health, Lublin, Poland; Department of Pathophysiology, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
40
|
Abstract
Epilepsy prevention is one of the great unmet needs in epilepsy. Approximately 15% of all epilepsy is caused by an acute acquired CNS insult such as traumatic brain injury (TBI), stroke or encephalitis. There is a latent period between the insult and epilepsy onset that presents an opportunity to intervene with preventive treatment that is unique in neurology. Yet no phase 3 epilepsy prevention studies, and only 2 phase 2 studies have been initiated in the last 16years. Current prevailing opinion is that the research community is not ready for clinical preventive epilepsy studies, and that animal models should first be refined and biomarkers of epileptogenesis and of epilepsy discovered before clinical studies are embarked upon. We review data to suggest that there is basis to do epilepsy prevention studies now with the current knowledge and available drugs, and that those studies are feasible with currently available tools. We suggest that a different approach is needed from the past in order to maximize chances of success, minimize the cost, and set up platform for future preventive treatment development. That approach should include close coordination of preclinical and clinical development programs in a combined PTE prevention strategy, consideration of polytherapy, and simultaneous, combined clinical development of preventive treatment and of biomarker discovery. We argue that the currently favored approach of eschewing clinical studies until biomarkers are available will delay the discovery of epilepsy prevention treatment by at least 10 years and significantly increase the cost of such discovery.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD 20817, United States.
| | - Ivana Tyrlikova
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD 20817, United States.
| |
Collapse
|
41
|
Kaur N, Singh T, Kumar S, Goel RK. Neurochemical evidence based suggested therapy for safe management of epileptogenesis. Epilepsy Behav 2017; 72:8-16. [PMID: 28570965 DOI: 10.1016/j.yebeh.2017.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/18/2017] [Accepted: 04/03/2017] [Indexed: 01/05/2023]
Abstract
Most of the clinically available antiepileptic drugs have only antiseizure effects and are reported unable to prevent epileptogenesis. In the past decade, several drugs underwent clinical trials for management of epileptogenesis, but none of the drugs tested was found effective. One of the major lacunas is availability of appropriate preclinical approaches to delineate mechanisms of epileptogenesis. Thus, the present study attempts to suggest a neurochemistry based approach for safe management of epileptogenesis. The altered neurochemical milieu in amygdala, cortex and hippocampus areas of the mice brain in naïve, kindled and kindling resistant animals has been delineated. The endogenous natural antiepileptogenic neurochemical defense mechanism observed in kindling resistant animals may uncover neurochemical mechanisms of epileptogenesis and in turn suggest us novel interventions for safe management of epileptogenesis. The kindling epileptogenesis was carried out in two month old male Swiss albino mice by administering subconvulsive pentylenetetrazole (35mg/kg; i.p.) at an interval of 48±2h for 42days. 2h after the last pentylenetetrazole injection, the animals were subjected to behavioral evaluations. Four hours after behavioral evaluation, all animals were euthanized and discrete parts of brain (amygdala, cortex and hippocampus) were harvested for neurochemical analysis. Results revealed that 60% of animals responded to kindling as observed with decreased seizure threshold, while the rest were found resistant. The kindled animals were found to be associated with anxiety, depression and cognitive impairment; while in kindling resistant animals no such behavioral deficits were observed. The neurochemical analysis revealed that in kindled animals altered glutamate-GABA neurotransmission, and decreased taurine, glycine, d-serine, monoamine levels with elevated indoleamine 2,3-dioxygenase activity were observed, which may be convicted for progression of kindling epileptogenesis. However, in kindling resistant animals elevated GABA, taurine, tryptophan, serotonin, glycine, and d-serine levels with decreased indoleamine 2,3-dioxygenase activity were observed as natural endogenous antiepileptogenic mechanisms, which may be foreseen as safe pharmacological targets for management of epileptogenesis.
Collapse
Affiliation(s)
- Navjot Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Tanveer Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Sandeep Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Rajesh Kumar Goel
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India.
| |
Collapse
|
42
|
Piccenna L, Shears G, O'Brien TJ. Management of post-traumatic epilepsy: An evidence review over the last 5 years and future directions. Epilepsia Open 2017; 2:123-144. [PMID: 29588942 PMCID: PMC5719843 DOI: 10.1002/epi4.12049] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2017] [Indexed: 12/17/2022] Open
Abstract
Post‐traumatic epilepsy (PTE) is a relatively underappreciated condition that can develop as a secondary consequence following traumatic brain injury (TBI). The aim of this rapid evidence review is to provide a synthesis of existing evidence on the effectiveness of treatment interventions for the prevention of PTE in people who have suffered a moderate/severe TBI to increase awareness and understanding among consumers. Electronic medical databases (n = 5) and gray literature published between January 2010 and April 2015 were searched for studies on the management of PTE. Twenty‐two eligible studies were identified that met the inclusion criteria. No evidence was found for the effectiveness of any pharmacological treatments in the prevention or treatment of symptomatic seizures in adults with PTE. However, limited high‐level evidence for the effectiveness of the antiepileptic drug levetiracetam was identified for PTE in children. Low‐level evidence was identified for nonpharmacological interventions in significantly reducing seizures in patients with PTE, but only in a minority of cases, requiring further high‐level studies to confirm the results. This review provides an opportunity for researchers and health service professionals to better understand the underlying pathophysiology of PTE to develop novel, more effective therapeutic targets and to improve the quality of life of people with this condition.
Collapse
Affiliation(s)
- Loretta Piccenna
- The Epilepsy Foundation Melbourne Victoria Australia.,Department of Medicine The University of Melbourne Parkville Victoria Australia
| | - Graeme Shears
- The Epilepsy Foundation Melbourne Victoria Australia
| | - Terence J O'Brien
- James Stewart Professor of Medicine Department of Medicine The Royal Melbourne Hospital The University of Melbourne Parkville Victoria Australia
| |
Collapse
|
43
|
Löscher W, Gillard M, Sands ZA, Kaminski RM, Klitgaard H. Synaptic Vesicle Glycoprotein 2A Ligands in the Treatment of Epilepsy and Beyond. CNS Drugs 2016; 30:1055-1077. [PMID: 27752944 PMCID: PMC5078162 DOI: 10.1007/s40263-016-0384-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The synaptic vesicle glycoprotein SV2A belongs to the major facilitator superfamily (MFS) of transporters and is an integral constituent of synaptic vesicle membranes. SV2A has been demonstrated to be involved in vesicle trafficking and exocytosis, processes crucial for neurotransmission. The anti-seizure drug levetiracetam was the first ligand to target SV2A and displays a broad spectrum of anti-seizure activity in various preclinical models. Several lines of preclinical and clinical evidence, including genetics and protein expression changes, support an important role of SV2A in epilepsy pathophysiology. While the functional consequences of SV2A ligand binding are not fully elucidated, studies suggest that subsequent SV2A conformational changes may contribute to seizure protection. Conversely, the recently discovered negative SV2A modulators, such as UCB0255, counteract the anti-seizure effect of levetiracetam and display procognitive properties in preclinical models. More broadly, dysfunction of SV2A may also be involved in Alzheimer's disease and other types of cognitive impairment, suggesting potential novel therapies for levetiracetam and its congeners. Furthermore, emerging data indicate that there may be important roles for two other SV2 isoforms (SV2B and SV2C) in the pathogenesis of epilepsy, as well as other neurodegenerative diseases. Utilization of recently developed SV2A positron emission tomography ligands will strengthen and reinforce the pharmacological evidence that SV2A is a druggable target, and will provide a better understanding of its role in epilepsy and other neurological diseases, aiding in further defining the full therapeutic potential of SV2A modulation.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| | | | | | | | | |
Collapse
|
44
|
Yang Y, Zheng F, Xu X, Wang X. Levetiracetam Versus Phenytoin for Seizure Prophylaxis Following Traumatic Brain Injury: A Systematic Review and Meta-Analysis. CNS Drugs 2016; 30:677-88. [PMID: 27395404 DOI: 10.1007/s40263-016-0365-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Seizure following traumatic brain injury (TBI) constitutes a common complication that requires effective prevention to improve the outcome of TBI. Phenytoin has been the only recommended antiepileptic drug (AED) for seizure prophylaxis; however, several shortcomings have affected its use. Intravenous levetiracetam has been available since 2006 and has been increasingly accepted as a seizure prophylaxis for brain injury, mainly due to its favorable pharmacokinetic features and minimal adverse events profile. However, the efficacy and safety of levetiracetam versus phenytoin for seizure prophylaxis following TBI are not well clarified. OBJECTIVE The aim of this study was to assess the efficacy and safety of levetiracetam versus phenytoin for seizure prophylaxis following TBI. METHODS We conducted a search of the MEDLINE, EMBASE, and Cochrane library databases to March 2016, and screened original research that included patients with TBI who received levetiracetam. We included randomized controlled trials (RCTs) or controlled observational cohort studies that compared levetiracetam and phenytoin, as well as uncontrolled case series regarding prophylactic levetiracetam following TBI. The outcomes included early or late seizure prophylaxis and safety. The estimates of seizure prophylaxis were pooled using a meta-analysis, and the estimates for the case series were pooled using descriptive statistics. RESULTS A total of 1614 patients from 11 studies were included in this review, of whom 1285 patients from eight controlled studies (one RCT and seven cohort studies) were included in the meta-analysis. Levetiracetam was not superior to phenytoin with regard to early seizure prophylaxis (risk ratio [RR] 1.10, 95 % confidence interval [CI] 0.64-1.88); the estimate of early seizure incidence was 0.05 (95 % CI 0.02-0.08). Three studies that assessed late seizure did not indicate the superiority of levetiracetam to phenytoin. There were no differences in mortality during hospitalization or after 6 months, or in the number of patients with adverse reactions between levetiracetam and phenytoin. CONCLUSIONS Levetiracetam does not appear to be superior to phenytoin in efficacy or safety with regard to early or late seizure prophylaxis following TBI; however, no class I evidence was identified. Additional evidence from high-quality studies is required.
Collapse
Affiliation(s)
- Yong Yang
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Fangshuo Zheng
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Xin Xu
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China
| | - Xuefeng Wang
- Department of Neurology, Chongqing Key Laboratory of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Road, Chongqing, 400016, China. .,Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
45
|
Zou H, Hurwitz M, Fowler L, Wagner AK. Abbreviated levetiracetam treatment effects on behavioural and histological outcomes after experimental TBI. Brain Inj 2016; 29:78-85. [PMID: 25255156 DOI: 10.3109/02699052.2014.955528] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Long-term prophylactic treatment with levetiracetam (LEV) has multiple neuroprotective effects in a traumatic brain injury (TBI) rat model. Although a rational time-frame of seizure prophylactic treatment with LEV for after TBI is not well established, clinical prophylaxis with LEV often includes treatment duration similar to clinical treatment guidelines with Phenytoin. Thus, this study investigated the effects of abbreviated LEV treatment on behavioural function and histological evidence of neuroprotection. RESEARCH DESIGN Pre-clinical trial of abbreviated LEV dosing in an experimental model of TBI Methods: After either controlled cortical impact (CCI) injury or sham surgery, rats received three 50 mg kg(-1) doses over 24 hours or vehicle. After injury/sham surgery, beam performance, spatial learning, contusion volume size and hippocampal neuron survival were assessed. RESULTS Abbreviated LEV did not improve motor or cognitive performance after TBI. Further, abbreviated LEV did not improve hippocampal neuron sparing or contusion volumes compared with vehicle controls. CONCLUSIONS Together with previous work assessing daily LEV treatment, these results suggest that longer-term therapy may be required to confer beneficial effects within these domains. These findings may guide (1) future experimental studies assessing minimal effective dosing for neuroprotection and anti-epileptogenesis and (2) treatment guideline updates for seizure prophylaxis post-TBI.
Collapse
Affiliation(s)
- Huichao Zou
- a Department of Physical Medicine and Rehabilitation and
| | | | | | | |
Collapse
|
46
|
Łukawski K, Gryta P, Łuszczki J, Czuczwar SJ. Exploring the latest avenues for antiepileptic drug discovery and development. Expert Opin Drug Discov 2016; 11:369-82. [DOI: 10.1517/17460441.2016.1154840] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
47
|
Klee R, Töllner K, Rankovic V, Römermann K, Schidlitzki A, Bankstahl M, Löscher W. Network pharmacology for antiepileptogenesis: Tolerability of multitargeted drug combinations in nonepileptic vs. post-status epilepticus mice. Epilepsy Res 2015; 118:34-48. [PMID: 26600369 DOI: 10.1016/j.eplepsyres.2015.11.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/31/2015] [Accepted: 11/05/2015] [Indexed: 12/30/2022]
Abstract
Prevention of symptomatic epilepsy ("antiepileptogenesis") in patients at risk is a major unmet clinical need. Several drugs underwent clinical trials for epilepsy prevention, but none of the drugs tested was effective. Similarly, most previous preclinical attempts to develop antiepileptogenic strategies failed. In the majority of studies, drugs were given as monotherapy. However, epilepsy is a complex network phenomenon, so that it is unlikely that a single drug can halt epileptogenesis. We recently proposed multitargeted approaches ("network pharmacology") to interfere with epileptogenesis. One strategy, which, if effective, would allow a relatively rapid translation into the clinic, is developing novel combinations of clinically used drugs with diverse mechanisms that are potentially relevant for antiepileptogenesis. In order to test this strategy preclinically, we developed an algorithm for testing such drug combinations, which was inspired by the established drug development phases in humans. As a first step of this algorithm, tolerability of four rationally chosen, repeatedly administered drug combinations was evaluated by a large test battery in mice: A, levetiracetam and phenobarbital; B, valproate, losartan, and memantine; C, levetiracetam and topiramate; and D, levetiracetam, parecoxib, and anakinra. As in clinical trials, tolerability was separately evaluated before starting efficacy experiments to identify any adverse effects of the combinations that may critically limit the successful translation of preclinical findings to the clinic. Except combination B, all drug cocktails were relatively well tolerated. Based on previous studies, we expected that tolerability would be lower in the latent and chronic phases following status epilepticus in mice, but, except combinations C and D, no significant differences were determined between nonepileptic and post-status epilepticus animals. As a next step, the rationally chosen drug combinations will be evaluated for antiepileptogenic activity in mouse and rat models of symptomatic epilepsy.
Collapse
Affiliation(s)
- Rebecca Klee
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Vladan Rankovic
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
48
|
Thompson K, Pohlmann‐Eden B, Campbell LA, Abel H. Pharmacological treatments for preventing epilepsy following traumatic head injury. Cochrane Database Syst Rev 2015; 2015:CD009900. [PMID: 26259048 PMCID: PMC9237755 DOI: 10.1002/14651858.cd009900.pub2] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Head injury is a common event and can cause a spectrum of motor and cognition disabilities. A frequent complication is seizures. Antiepileptic drugs (AED) such as phenytoin are often used in clinical practice with the hopes of preventing post-traumatic epilepsy. Whether immediate medical intervention following head trauma with either AEDs or neuroprotective drugs can alter the process of epileptogenesis and lead to a more favorable outcome is currently unknown. This review attempted to address the effectiveness of these treatment interventions. This review updates and expands on the earlier Cochrane review. OBJECTIVES To compare the efficacy of antiepileptic drugs and neuroprotective agents with placebo, usual care or other pharmacologic agents for the prevention of post-traumatic epilepsy in people diagnosed with any severity of traumatic brain injury. SEARCH METHODS We searched The Cochrane Epilepsy Group's specialized register, CENTRAL, MEDLINE, ClinicalTrials.gov and World Health Organization International Clinical Trials Registry Platform (ICTRP) in January 2015. We searched EMBASE, Biological Abstracts and National Research Register in September 2014 and SCOPUS in December 2013. The Cochrane Epilepsy Group performed handsearches of relevant journals. SELECTION CRITERIA We included randomized controlled trials (RCTs) that include AEDs or neuroprotective agents compared with placebo, another pharmacologic agent or a usual care group. The outcomes measured included a seizure occurring within one week of trauma (early seizure), seizure occurring later than one week post-trauma (late seizure), mortality and any adverse events. DATA COLLECTION AND ANALYSIS Two review authors independently assessed study quality and extracted the data. We calculated risk ratios (RR) and 95% confidence intervals (CI) for each outcome. We used random-effects models in the meta-analyses and performed pre-defined subgroup and sensitivity analyses. MAIN RESULTS This review included 10 RCTs (reported in 12 articles) consisting of 2326 participants The methodological quality of the studies varied. The type of intervention was separated into three categories; AED versus placebo or standard care, alternative neuroprotective agent versus placebo or standard care and AED versus other AED. Treatment with an AED (phenytoin or carbamazepine) decreased the risk of early seizure compared with placebo or standard care (RR 0.42, 95% CI 0.23 to 0.73; very low quality evidence). There was no evidence of a difference in the risk of late seizure occurrence between AEDs and placebo or standard care (RR 0.91, 95% CI 0.57 to 1.46; very low quality evidence). There was no evidence of a significant difference in all-cause mortality between AEDs and placebo or standard care (RR 1.08 95% CI 0.79 to 1.46,very low quality of evidence). Only one study looked at other potentially neuroprotective agents (magnesium sulfate) compared with placebo. The risk ratios were: late seizure 1.07 (95% CI 0.53 to 2.17) and all-cause mortality 1.20 (95% CI 0.80 to 1.81). The risk ratio for occurrence of early seizure was not estimable.Two studies looked at comparison of two AEDs (levetiracetam, valproate) with phenytoin used as the main comparator in each study. The risk ratio for all-cause mortality was 0.53 (95% CI 0.30 to 0.94). There was no evidence of treatment benefit of phenytoin compared with another AED for early seizures (RR 0.66, 95% 0.20 to 2.12) or late seizures(RR 0.77, 95% CI 0.46 to 1.30).Only two studies reported adverse events. The RR of any adverse event with AED compared with placebo was 1.65 (95% CI 0.73 to 3.66; low quality evidence). There were insufficient data on adverse events in the other treatment comparisons. AUTHORS' CONCLUSIONS This review found low-quality evidence that early treatment with an AED compared with placebo or standard care reduced the risk of early post-traumatic seizures. There was no evidence to support a reduction in the risk of late seizures or mortality. There was insufficient evidence to make any conclusions regarding the effectiveness or safety of other neuroprotective agents compared with placebo or for the comparison of phenytoin, a traditional AED, with another AED.
Collapse
Affiliation(s)
- Kara Thompson
- Dalhousie UniversityDepartment of MedicineRm 204, Centre for Clinical Research5790 University AvenueHalifaxNSCanadaB3H 1V7
| | | | - Leslie A Campbell
- Dalhousie UniversityDepartment of Community Health and EpidemiologyRoom 415, 5790 University AvenueHalifaxNSCanadaB3K 1V7
| | - Hannah Abel
- Dalhousie UniversityCommunity Health and Epidemiologyc/o Center for Clinical Research4th Floors, 5790 University AvenueHalifaxNSCanadaB3H 1V7
| | | |
Collapse
|
49
|
Chong SL, Lee KP, Lee JH, Ong GYK, Ong MEH. Pediatric head injury: a pain for the emergency physician? Clin Exp Emerg Med 2015; 2:1-8. [PMID: 27752566 PMCID: PMC5052852 DOI: 10.15441/ceem.14.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 01/10/2015] [Accepted: 01/26/2015] [Indexed: 11/23/2022] Open
Abstract
The prompt diagnosis and initial management of pediatric traumatic brain injury poses many challenges to the emergency department (ED) physician. In this review, we aim to appraise the literature on specific management issues faced in the ED, specifically: indications for neuroimaging, choice of sedatives, applicability of hyperventilation, utility of hyperosmolar agents, prophylactic anti-epileptics, and effect of hypothermia in traumatic brain injury. A comprehensive literature search of PubMed and Embase was performed in each specific area of focus corresponding to the relevant questions. The majority of the head injured patients presenting to the ED are mild and can be observed. Clinical prediction rules assist the ED physician in deciding if neuroimaging is warranted. In cases of major head injury, prompt airway control and careful use of sedation are necessary to minimize the chance of hypoxia, while avoiding hyperventilation. Hyperosmolar agents should be started in these cases and normothermia maintained. The majority of the evidence is derived from adult studies, and most treatment modalities are still controversial. Recent multicenter trials have highlighted the need to establish common platforms for further collaboration.
Collapse
Affiliation(s)
- Shu-Ling Chong
- Department of Emergency Medicine, KK Women's and Children's Hospital, Singapore
| | - Khai Pin Lee
- Department of Emergency Medicine, KK Women's and Children's Hospital, Singapore
| | - Jan Hau Lee
- Children's Intensive Care Unit, KK Women's and Children's Hospital, Singapore
| | - Gene Yong-Kwang Ong
- Department of Emergency Medicine, KK Women's and Children's Hospital, Singapore
| | | |
Collapse
|
50
|
|