1
|
Sun Q, Peng S, Xu Q, Weikop P, Hussain R, Song W, Nedergaard M, Ding F. Enhancing glymphatic fluid transport by pan-adrenergic inhibition suppresses epileptogenesis in male mice. Nat Commun 2024; 15:9600. [PMID: 39505840 DOI: 10.1038/s41467-024-53430-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Epileptogenesis is the process whereby the previously normally functioning brain begins to generate spontaneous, unprovoked seizures. Status epilepticus (SE), which entails a massive release of neuronal glutamate and other neuroactive substances, is one of the best-known triggers of epileptogenesis. We here asked whether pharmacologically promoting glymphatic clearance during or after SE is beneficial and able to attenuate the subsequent epileptogenesis. We induced SE in adult male mice by intrahippocampal kainic acid (KA) infusion. Acute administration of a cocktail of adrenergic receptor antagonists (propranolol, prazosin, and atipamezole: PPA), enhanced glymphatic flow and effectively reduced the severity of spontaneous seizures in the chronic phase. The PPA treatment also reduced reactive gliosis and inhibited the loss of polarized expression of AQP4 water channels in the vascular endfeet of astrocytes. Administration of PPA after cessation of SE (30 hours post KA) also effectively suppressed epileptogenesis and improved outcome. Conversely, mice with constitutively low glymphatic transport due to genetic deletion of the aquaporin 4 (AQP4) water channel showed exacerbation of KA-induced epileptogenesis. We conclude that the pharmacological modulation of glymphatic fluid transport may represent a potential strategy to dampen epileptogenesis and the occurrence of spontaneous seizures following KA-induced SE.
Collapse
Affiliation(s)
- Qian Sun
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY, USA
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sisi Peng
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY, USA
- Department of PET/MR, Shanghai Universal Medical Imaging Diagnostic Center, Shanghai, China
| | - Qiwu Xu
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY, USA
| | - Pia Weikop
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Rashad Hussain
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY, USA
| | - Wei Song
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY, USA.
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark.
| | - Fengfei Ding
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY, USA.
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Pease M, Gupta K, Moshé SL, Correa DJ, Galanopoulou AS, Okonkwo DO, Gonzalez-Martinez J, Shutter L, Diaz-Arrastia R, Castellano JF. Insights into epileptogenesis from post-traumatic epilepsy. Nat Rev Neurol 2024; 20:298-312. [PMID: 38570704 DOI: 10.1038/s41582-024-00954-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
Post-traumatic epilepsy (PTE) accounts for 5% of all epilepsies. The incidence of PTE after traumatic brain injury (TBI) depends on the severity of injury, approaching one in three in groups with the most severe injuries. The repeated seizures that characterize PTE impair neurological recovery and increase the risk of poor outcomes after TBI. Given this high risk of recurrent seizures and the relatively short latency period for their development after injury, PTE serves as a model disease to understand human epileptogenesis and trial novel anti-epileptogenic therapies. Epileptogenesis is the process whereby previously normal brain tissue becomes prone to recurrent abnormal electrical activity, ultimately resulting in seizures. In this Review, we describe the clinical course of PTE and highlight promising research into epileptogenesis and treatment using animal models of PTE. Clinical, imaging, EEG and fluid biomarkers are being developed to aid the identification of patients at high risk of PTE who might benefit from anti-epileptogenic therapies. Studies in preclinical models of PTE have identified tractable pathways and novel therapeutic strategies that can potentially prevent epilepsy, which remain to be validated in humans. In addition to improving outcomes after TBI, advances in PTE research are likely to provide therapeutic insights that are relevant to all epilepsies.
Collapse
Affiliation(s)
- Matthew Pease
- Department of Neurosurgery, Indiana University, Bloomington, IN, USA.
| | - Kunal Gupta
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Solomon L Moshé
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, New York, NY, USA
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
- Department of Paediatrics, Albert Einstein College of Medicine, New York, NY, USA
| | - Daniel J Correa
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, New York, NY, USA
| | - Aristea S Galanopoulou
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, New York, NY, USA
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | - David O Okonkwo
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Lori Shutter
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | |
Collapse
|
3
|
Kerr WT, Reddy AS, Seo SH, Kok N, Stacey WC, Stern JM, Pennell PB, French JA. Increasing challenges to trial recruitment and conduct over time. Epilepsia 2023; 64:2625-2634. [PMID: 37440282 PMCID: PMC10592378 DOI: 10.1111/epi.17716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/14/2023]
Abstract
OBJECTIVE This study was undertaken to evaluate how the challenges in the recruitment and retention of participants in clinical trials for focal onset epilepsy have changed over time. METHODS In this systematic analysis of randomized clinical trials of adjunct antiseizure medications for medication-resistant focal onset epilepsy, we evaluated how the numbers of participants, sites, and countries have changed since the first such trial in 1990. We also evaluated the proportion of participants who completed each trial phase and their reasons for early trial exit. We analyzed these trends using mixed effects generalized linear models accounting for the influence of the number of trial sites and trial-specific variability. RESULTS The number of participants per site has steadily decreased over decades, with recent trials recruiting fewer than five participants per site (reduction by .16 participants/site/year, p < .0001). Fewer participants also progressed from recruitment to randomization over time (odds ratio = .94/year, p = .014). Concurrently, there has been an increase in the placebo response over time (increase in median percent reduction of .4%/year, p = .02; odds ratio of increase in 50% responder rate of 1.03/year, p = .02), which was not directly associated with the number of sites per trial (p > .20). SIGNIFICANCE This historical analysis highlights the increasing challenges with participant recruitment and retention, as well as increasing placebo response. It serves as a call to action to change clinical trial design to address these challenges.
Collapse
Affiliation(s)
- Wesley T. Kerr
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Advith S. Reddy
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sung Hyun Seo
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Neo Kok
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - William C. Stacey
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - John M. Stern
- Department of Neurology, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Page B. Pennell
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
4
|
Schmidt T, Meller S, Meyerhoff N, Twele F, Zanghi B, Volk HA. A six-month prospective, randomised, double-blinded, placebo-controlled, crossover, dietary trial design to investigate the potential of psychobiotics on seizure semiology and comorbidities in canine epilepsy: study protocol. BMC Vet Res 2023; 19:57. [PMID: 36864510 PMCID: PMC9983181 DOI: 10.1186/s12917-023-03609-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/16/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Epilepsy is the most common chronic neurological disease in dogs. More than two-thirds of these patients suffer from associated behavioural comorbidities. The latter could have their origin in partially overlapping pathomechanisms, with the intestinal microbiome as a potential key link between them. The current arsenal of drugs for epilepsy management remains limited. Most canine patients continue to have seizures despite treatment and the occurrence of comorbidities is not sufficiently addressed, limiting quality of life of affected dogs and owners. Therefore, novel additional epilepsy management options are urgently needed. The microbiome-gut-brain axis may serve as a new target for the development of innovative multimodal therapeutic approaches to overcome current shortcomings in epilepsy management. METHODS A six-month prospective, randomised, double-blinded, placebo-controlled, crossover, dietary trial was designed to investigate the potential of the psychobiotic Bifidobacterium longum on behavioural comorbidities in canine epilepsy. Seizure semiology will be evaluated as a secondary outcome measure. Thirty-four privately owned dogs are planned to be included in the ongoing study meeting the following inclusion criteria: Dogs displaying increased anxiety/fear behaviour since the start of the idiopathic epilepsy. Tier II confidence level of the International Veterinary Epilepsy Task Force for the diagnosis of idiopathic epilepsy, with a maximum seizure interval of 3 month and a minimum of three generalised seizures within that period and chronically treated with at least one antiseizure drug without improvement in seizure frequency Each dog will receive the allocated supplement (probiotic vs. placebo) alongside its normal diet for a 3-month period. After a three-week wash out period, the second phase starts by administering the respective other supplement for another 3 months. DISCUSSION The current study considers modern high-quality standards for epilepsy medication trials. Common biasing effects should be limited to a possible minimum (regression-to-the mean effect, placebo effect, observer effect), ensuring a high validity and accuracy of the acquired results, thus enabling a representative nature of the efficacy of Bifidobacterium longum as add-on supplement for dogs suffering from epilepsy and its comorbidities. This publication should provide a description of the study procedure and data acquisition methods, including prognosed statistical analysis.
Collapse
Affiliation(s)
- Teresa Schmidt
- grid.412970.90000 0001 0126 6191Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sebastian Meller
- grid.412970.90000 0001 0126 6191Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nina Meyerhoff
- grid.412970.90000 0001 0126 6191Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Friederike Twele
- grid.412970.90000 0001 0126 6191Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Brian Zanghi
- Research and Development, Nestlé Purina PetCare, St. Louis, MO USA
| | - Holger Andreas Volk
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Hannover, Germany. .,Centre for Systems Neuroscience, University of Veterinary Medicine Hannover, Hannover, Germany.
| |
Collapse
|
5
|
Löscher W, Stafstrom CE. Epilepsy and its neurobehavioral comorbidities: Insights gained from animal models. Epilepsia 2023; 64:54-91. [PMID: 36197310 DOI: 10.1111/epi.17433] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 01/21/2023]
Abstract
It is well established that epilepsy is associated with numerous neurobehavioral comorbidities, with a bidirectional relationship; people with epilepsy have an increased incidence of depression, anxiety, learning and memory difficulties, and numerous other psychosocial challenges, and the occurrence of epilepsy is higher in individuals with those comorbidities. Although the cause-and-effect relationship is uncertain, a fuller understanding of the mechanisms of comorbidities within the epilepsies could lead to improved therapeutics. Here, we review recent data on epilepsy and its neurobehavioral comorbidities, discussing mainly rodent models, which have been studied most extensively, and emphasize that clinically relevant information can be gained from preclinical models. Furthermore, we explore the numerous potential factors that may confound the interpretation of emerging data from animal models, such as the specific seizure induction method (e.g., chemical, electrical, traumatic, genetic), the role of species and strain, environmental factors (e.g., laboratory environment, handling, epigenetics), and the behavioral assays that are chosen to evaluate the various aspects of neural behavior and cognition. Overall, the interplay between epilepsy and its neurobehavioral comorbidities is undoubtedly multifactorial, involving brain structural changes, network-level differences, molecular signaling abnormalities, and other factors. Animal models are well poised to help dissect the shared pathophysiological mechanisms, neurological sequelae, and biomarkers of epilepsy and its comorbidities.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Carl E Stafstrom
- Division of Pediatric Neurology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Ling DSF, Yang L, Goodman JH. Brivaracetam prevents the development of epileptiform activity when administered early after cortical neurotrauma in rats. Epilepsia 2022; 63:992-1002. [PMID: 35037242 DOI: 10.1111/epi.17167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/06/2021] [Accepted: 01/04/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES There is no effective therapy to prevent the development of posttraumatic epilepsy (PTE). Recently, we reported that administration of the antiseizure medication (ASM) levetiracetam (LEV) shortly after trauma prevented the development of epileptiform activity in two experimental models of neurotrauma. However, the time window for effective intervention with LEV may be too narrow for most clinical settings. Using the controlled cortical impact (CCI) injury model, the current study tested whether early administration of brivaracetam (BRV), an ASM with 20 times the affinity of LEV for the SV2A synaptic vesicle protein, could improve upon the antiepileptogenic action observed with LEV. METHODS Rats (postnatal day [P] 24-32) subjected to CCI injury were given a single dose of BRV (21 or 100 mg/kg, i.p.) at one of three post-injury time points: immediately (0-2 minutes), 30 minutes, or 60 minutes. Control animals received only vehicle (0.9% saline). Posttraumatic electrographic epileptiform activity was assayed ex vivo from coronal neocortical slices collected proximal to the injury (four per rat) 3-4 weeks after injury. In this model, ictal-like burst discharges occur spontaneously or can be evoked in an "all or none" manner with applied electrical stimulation within the first 2 weeks after injury. RESULTS A single dose of BRV administered to rats up to 60 minutes after traumatic brain injury (TBI) significantly reduced the development of posttraumatic epileptiform activity by (1) inhibiting the development of both evoked and spontaneous epileptiform activity, (2) raising the threshold for stimulus-evoked epileptiform discharges, and (3) reducing the intensity of epileptiform bursts that arise after cortical neurotrauma. SIGNIFICANCE Clinically there has been little success preventing the development of posttraumatic epilepsy. The results of this study support the hypothesis that early intervention with BRV has the potential to prevent or reduce posttraumatic epileptogenesis, and that there may be a limited time window for successful prophylactic intervention.
Collapse
Affiliation(s)
- Douglas S F Ling
- Departments of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA.,Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Lie Yang
- Departments of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA
| | - Jeffrey H Goodman
- Departments of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA.,Robert F. Furchgott Center for Behavioral and Neural Science, SUNY Downstate Health Sciences University, Brooklyn, New York, USA.,Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, New York, USA.,Department of Developmental Neurobiology, The New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| |
Collapse
|
7
|
Eastman CL, Fender JS, Klein P, D'Ambrosio R. Therapeutic Effects of Time-Limited Treatment with Brivaracetam on Posttraumatic Epilepsy after Fluid Percussion Injury in the Rat. J Pharmacol Exp Ther 2021; 379:310-323. [PMID: 34593559 DOI: 10.1124/jpet.121.000585] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 09/28/2021] [Indexed: 11/22/2022] Open
Abstract
Mounting evidence suggests the synaptic vesicle glycoprotein 2A (SV2A) targeted by levetiracetam may contribute to epileptogenesis. Levetiracetam has shown anti-inflammatory, antioxidant, neuroprotective, and possible antiepileptogenic effects in brain injury and seizure/epilepsy models, and a phase 2 study has signaled a possible clinical antiepileptogenic effect. Brivaracetam shows greater affinity and specificity for SV2A than levetiracetam and broader preclinical antiseizure effects. Thus, we assessed the antiepileptogenic/disease-modifying potential of brivaracetam in an etiologically realistic rat posttraumatic epilepsy model optimized for efficient drug testing. Brivaracetam delivery protocols were designed to maintain clinical moderate-to-high plasma levels in young (5-week-old) male Sprague-Dawley rats for 4 weeks. Treatment protocols were rapidly screened in 4-week experiments using small groups of animals to ensure against rigorous testing of futile treatment protocols. The antiepileptogenic effects of brivaracetam treatment initiated 30 minutes, 4 hours, and 8 hours after rostral parasagittal fluid percussion injury (rpFPI) were then compared with vehicle-treated controls in a fully powered blind and randomized 16-week validation. Seizures were evaluated by video-electrocorticography using a 5-electrode epidural montage. Endpoint measures included incidence, frequency, duration, and spread of seizures. Group sizes and recording durations were supported by published power analyses. Three months after treatment ended, rats treated with brivaracetam starting at 4 hours post-FPI (the best-performing protocol) experienced a 38% decrease in overall incidence of seizures, 59% decrease in seizure frequency, 67% decrease in time spent seizing, and a 45% decrease in the proportion of spreading seizures that was independent of duration-based seizure definition. Thus, brivaracetam shows both antiepileptogenic and disease-modifying properties after rpFPI. SIGNIFICANCE STATEMENT: The rpFPI model, which likely incorporates epileptogenic mechanisms operating after human head injury, can be used to efficiently screen investigational treatment protocols and assess antiepileptogenic/disease-modifying effects. Our studies 1) support a role for SV2A in epileptogenesis, 2) suggest that brivaracetam and other drugs targeting SV2A should be considered for human clinical trials of prevention of post-traumatic epilepsy after head injury, and 3) provide data to inform the design of treatment protocols for clinical trials.
Collapse
Affiliation(s)
- Clifford L Eastman
- Department of Neurological Surgery, University of Washington, Seattle, Washington (C.L.E., J.S.F., R.D.); and Mid-Atlantic Epilepsy and Sleep Center, Bethesda, Maryland (P.K.)
| | - Jason S Fender
- Department of Neurological Surgery, University of Washington, Seattle, Washington (C.L.E., J.S.F., R.D.); and Mid-Atlantic Epilepsy and Sleep Center, Bethesda, Maryland (P.K.)
| | - Pavel Klein
- Department of Neurological Surgery, University of Washington, Seattle, Washington (C.L.E., J.S.F., R.D.); and Mid-Atlantic Epilepsy and Sleep Center, Bethesda, Maryland (P.K.)
| | - Raimondo D'Ambrosio
- Department of Neurological Surgery, University of Washington, Seattle, Washington (C.L.E., J.S.F., R.D.); and Mid-Atlantic Epilepsy and Sleep Center, Bethesda, Maryland (P.K.)
| |
Collapse
|
8
|
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: 22] [Impact Index Per Article: 7.3] [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
|
9
|
Sharma S, Tiarks G, Haight J, Bassuk AG. Neuropathophysiological Mechanisms and Treatment Strategies for Post-traumatic Epilepsy. Front Mol Neurosci 2021; 14:612073. [PMID: 33708071 PMCID: PMC7940684 DOI: 10.3389/fnmol.2021.612073] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death in young adults and a risk factor for acquired epilepsy. Severe TBI, after a period of time, causes numerous neuropsychiatric and neurodegenerative problems with varying comorbidities; and brain homeostasis may never be restored. As a consequence of disrupted equilibrium, neuropathological changes such as circuit remodeling, reorganization of neural networks, changes in structural and functional plasticity, predisposition to synchronized activity, and post-translational modification of synaptic proteins may begin to dominate the brain. These pathological changes, over the course of time, contribute to conditions like Alzheimer disease, dementia, anxiety disorders, and post-traumatic epilepsy (PTE). PTE is one of the most common, devastating complications of TBI; and of those affected by a severe TBI, more than 50% develop PTE. The etiopathology and mechanisms of PTE are either unknown or poorly understood, which makes treatment challenging. Although anti-epileptic drugs (AEDs) are used as preventive strategies to manage TBI, control acute seizures and prevent development of PTE, their efficacy in PTE remains controversial. In this review, we discuss novel mechanisms and risk factors underlying PTE. We also discuss dysfunctions of neurovascular unit, cell-specific neuroinflammatory mediators and immune response factors that are vital for epileptogenesis after TBI. Finally, we describe current and novel treatments and management strategies for preventing PTE.
Collapse
Affiliation(s)
- Shaunik Sharma
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Grant Tiarks
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Joseph Haight
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Alexander G Bassuk
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
10
|
Chen LY, Liang J, Fei F, Ruan YP, Cheng HM, Wang Y, Chen Z, Xu CL. Pharmaco-genetic inhibition of pyramidal neurons retards hippocampal kindling-induced epileptogenesis. CNS Neurosci Ther 2020; 26:1111-1120. [PMID: 32596972 PMCID: PMC7564188 DOI: 10.1111/cns.13434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/16/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
AIMS Pharmaco-genetics emerges as a new promising approach for epileptic seizures. Whether it can modulate epileptogenesis is still unknown. METHODS Here, parvalbumin neurons and pyramidal neurons of the seizure focus were transfected with engineered excitatory Gq-coupled human muscarinic receptor hM3Dq and engineered inhibitory Gi-coupled human muscarinic receptor hM4Di, respectively. And their therapeutic value in mouse hippocampal kindling-induced epileptogenesis was tested. RESULTS Pharmaco-genetic activating parvalbumin neurons limitedly retarded the progression of behavioral seizure stage and afterdischarge duration (ADD) during epileptogenesis induced by kindling. Activating parvalbumin neurons delayed seizure development only in the early stage, but accelerated it in late stages. On the contrary, pharmaco-genetic inhibiting pyramidal neurons robustly retarded the progression of seizure stages and ADDs, which greatly delayed seizure development in both early and late stages. Although both pharmaco-genetic therapeutics efficiently alleviated the severity of acute kindling-induced seizures, pharmaco-genetic inhibiting pyramidal neurons were able to reverse the enhanced synaptic plasticity during epileptogenesis, compared with that of pharmaco-genetic activating parvalbumin neurons. CONCLUSION Our results demonstrated that pharmaco-genetic inhibiting pyramidal neurons retard hippocampal kindling-induced epileptogenesis and reverse the enhanced synaptic plasticity during epileptogenesis, compared with that of pharmaco-genetic activating parvalbumin neurons. It suggests that pharmaco-genetics targeting pyramidal neurons may be a promising treatment for epileptogenesis.
Collapse
Affiliation(s)
- Li-Ying Chen
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jiao Liang
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Department of Neurology, Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fan Fei
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ye-Ping Ruan
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - He-Ming Cheng
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Department of Neurology, Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ceng-Lin Xu
- Institute of Pharmacology & Toxicology, NHC and CAMS Key Laboratory of Medical Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Department of Neurology, Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Eslami V, Lola MC, Karceski SC, Cavazos JE, Szabó CÁ. Changing characteristics of epilepsy interventional clinical trials over the last decade: Clinicaltrials.Gov registry. Epilepsy Res 2020; 164:106350. [PMID: 32447238 DOI: 10.1016/j.eplepsyres.2020.106350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/11/2020] [Accepted: 05/01/2020] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Epilepsy affects about 1% of the world's population (over 50 million). Of these, one-third have refractory or medication-resistant epilepsy. This group of people drives the development and testing of new interventions for epilepsy. To better address the needs of people with epilepsy, the characteristics of clinical trials, as well as the gaps in the population of interest, need to be evaluated. METHODS We searched the www.ClinicalTrials.gov database using the keywords "seizure" or "epilepsy" between 9/1/2008-9/1/2018 and filtering for Interventional Clinical trials. The data were categorized by three equal time intervals (tertiles), and evaluated by type of intervention (behavioral, diet, device, drug, other), primary purpose (treatment, diagnosis, prevention, or basic science), gender, age, phase (Phase1 to Phase 4 trials), length and status of the study, enrollment/recruitment/randomization, location, blinding status, assignment group (single/parallel/crossover/factorial/sequential), and funding. We focused on drugs and devices and used a binary logistic regression model to analyze the role of time, length of study, funding, location, randomization, and age. RESULTS We found 359 epilepsy clinical trials; of these, 245 (68.2%) clinical trials involved drugs, and 55 (15.3%) were device trials. Over the three tertiles, the percentage of device trials increased while medication trials decreased. Device:drug trial odds ratios increased six times by the third tertile. Also, the results showed that clinical trials for drugs and devices occurred more in adults than children. Industry funding decreased 20% over time. The US contribution to clinical research was stable, but device trials were more likely to occur outside of the US. CONCLUSION Drugs constitute the substantial fields of interventional trials in epilepsy but decreased in proportion over the last decade, while the presence of the device trials steadily increased. Device trials focused on treatment and diagnosis of seizures and have been more invested in non-US countries.
Collapse
Affiliation(s)
- Vahid Eslami
- Department of Neurology, UT Health Science Center, 7703 Floyd Curl Drive, San Antonio 78229-7883, TX, USA.
| | - Morgan C Lola
- Department of Neurology, UT Health Science Center, 7703 Floyd Curl Drive, San Antonio 78229-7883, TX, USA; South Texas Comprehensive Epilepsy Center, UT Health Science Center, San Antonio, TX, USA
| | - Steven C Karceski
- Department of Neurology, Weill Cornell Medical College, New York, NY, USA
| | - Jose E Cavazos
- Department of Neurology, UT Health Science Center, 7703 Floyd Curl Drive, San Antonio 78229-7883, TX, USA; South Texas Comprehensive Epilepsy Center, UT Health Science Center, San Antonio, TX, USA
| | - Charles Ákos Szabó
- Department of Neurology, UT Health Science Center, 7703 Floyd Curl Drive, San Antonio 78229-7883, TX, USA; South Texas Comprehensive Epilepsy Center, UT Health Science Center, San Antonio, TX, USA
| |
Collapse
|
12
|
Binder DK, Boison D, Eid T, Frankel WN, Mingorance A, Smith BN, Dacks PA, Whittemore V, Poduri A. Epilepsy Benchmarks Area II: Prevent Epilepsy and Its Progression. Epilepsy Curr 2020; 20:14S-22S. [PMID: 31937124 PMCID: PMC7031802 DOI: 10.1177/1535759719895274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Area II of the 2014 Epilepsy Research Benchmarks aims to establish goals for preventing the development and progression of epilepsy. In this review, we will highlight key advances in Area II since the last summary of research progress and opportunities was published in 2016. We also highlight areas of investigation that began to develop before 2016 and in which additional progress has been made more recently.
Collapse
Affiliation(s)
- Devin K Binder
- Division of Biomedical Sciences, School of Medicine, Center for Glial-Neuronal Interactions, University of California, Riverside, CA, USA
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson and New Jersey Medical Schools, Rutgers University, Piscataway, NJ, USA
| | - Tore Eid
- Department of Laboratory Medicine, Neurosurgery and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Wayne N Frankel
- Department of Genetics & Development, Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Bret N Smith
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | | | - Vicky Whittemore
- Division of Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Annapurna Poduri
- Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
13
|
Design and synthesis of selective and blood-brain barrier-permeable hydroxamate-based gelatinase inhibitors. Bioorg Chem 2020; 94:103365. [DOI: 10.1016/j.bioorg.2019.103365] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/13/2019] [Indexed: 12/31/2022]
|
14
|
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: 2.4] [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
|
15
|
Feyissa AM. Brivaracetam in the treatment of epilepsy: a review of clinical trial data. Neuropsychiatr Dis Treat 2019; 15:2587-2600. [PMID: 31571877 PMCID: PMC6750854 DOI: 10.2147/ndt.s143548] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/15/2019] [Indexed: 01/03/2023] Open
Abstract
Brivaracetam (BRV), an analog of levetiracetam (LEV), was discovered during a target-based rational drug discovery program that aimed to identify potent synaptic vesicle protein 2A (SV2A) ligands. Among the 12,000 compounds screened in vitro, BRV was found to have 15-30 times greater affinity for SV2A and faster brain permeability than LEV. Although preclinical and post-marketing studies suggest broad spectrum of efficacy, BRV is currently only approved as monotherapy and adjunctive therapy of focal-onset seizures in patients age 4 years and older. This review examines the use of BRV as add-on (5-200 mg/day) therapy for epilepsy with a particular emphasis on the six regulatory randomized clinical trialsinvolving 2399 participants. Participants receiving BRV add-on at doses of 50-200 mg/day were more likely to experience a 50% or greater reduction in seizure frequency (pooled risk ratio [RR]) 1.79 with 95% CI of 1.51-2.12) than those receiving placebo. Participants receiving BRV were also more likely to attain seizure freedom (57 [3.3%] vs 4 [0.5%]; RR 4.74, 95% CI 2.00-11.25) than those receiving placebo. In addition, BRV demonstrated a favorable safety profile similar to placebo across all BRV doses. Treatment emergent adverse events significantly associated with BRV were irritability, fatigue, somnolence, and dizziness. Post-hoc analysis of regulatory trials, post-marketing studies, and indirect comparison meta-analyses demonstrated equivalent efficacy and better tolerability of BRV when compared to other antiseizure drugs. Further, these studies appear to suggest that behavioral adverse events are likely to be less frequent and less severe with BRV than LEV. Therefore, switching to BRV may be considered for patients who have seizure control with LEV, but who cannot tolerate its behavioral adverse effects. In this setting, immediate switch from LEV to BRV at a 10:1-15:1 ratio without titration is feasible. Further research is needed to examine the long-term tolerability and efficacy of BRV as well as its role in the treatment of other types of epilepsies, particularly dementia-related epilepsy and brain tumor-related epilepsy.
Collapse
|
16
|
Kim JA, Boyle E, Wu AC, Cole AJ, Staley KJ, Zafar S, Cash SS, Westover MB. Epileptiform activity in traumatic brain injury predicts post-traumatic epilepsy. Ann Neurol 2018; 83:858-862. [PMID: 29537656 PMCID: PMC5912971 DOI: 10.1002/ana.25211] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 03/02/2018] [Accepted: 03/09/2018] [Indexed: 02/02/2023]
Abstract
We hypothesize that epileptiform abnormalities (EAs) in the electroencephalogram (EEG) during the acute period following traumatic brain injury (TBI) independently predict first-year post-traumatic epilepsy (PTE1 ). We analyze PTE1 risk factors in two cohorts matched for TBI severity and age (n = 50). EAs independently predict risk for PTE1 (odds ratio [OR], 3.16 [0.99, 11.68]); subdural hematoma is another independent risk factor (OR, 4.13 [1.18, 39.33]). Differences in EA rates are apparent within 5 days following TBI. Our results suggest that increased EA prevalence identifies patients at increased risk for PTE1 , and that EAs acutely post-TBI can identify patients most likely to benefit from antiepileptogenesis drug trials. Ann Neurol 2018;83:858-862.
Collapse
Affiliation(s)
- Jennifer A. Kim
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Emergency Neurology and Neurocritical Care, Massachusetts General Hospital, Boston, MA, USA
| | | | - Alexander C. Wu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Andrew J. Cole
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Kevin J Staley
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Sahar Zafar
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Emergency Neurology and Neurocritical Care, Massachusetts General Hospital, Boston, MA, USA
| | - Sydney S. Cash
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
17
|
Klein P, Dingledine R, Aronica E, Bernard C, Blümcke I, Boison D, Brodie MJ, Brooks-Kayal AR, Engel J, Forcelli PA, Hirsch LJ, Kaminski RM, Klitgaard H, Kobow K, Lowenstein DH, Pearl PL, Pitkänen A, Puhakka N, Rogawski MA, Schmidt D, Sillanpää M, Sloviter RS, Steinhäuser C, Vezzani A, Walker MC, Löscher W. Commonalities in epileptogenic processes from different acute brain insults: Do they translate? Epilepsia 2018; 59:37-66. [PMID: 29247482 PMCID: PMC5993212 DOI: 10.1111/epi.13965] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2017] [Indexed: 12/12/2022]
Abstract
The most common forms of acquired epilepsies arise following acute brain insults such as traumatic brain injury, stroke, or central nervous system infections. Treatment is effective for only 60%-70% of patients and remains symptomatic despite decades of effort to develop epilepsy prevention therapies. Recent preclinical efforts are focused on likely primary drivers of epileptogenesis, namely inflammation, neuron loss, plasticity, and circuit reorganization. This review suggests a path to identify neuronal and molecular targets for clinical testing of specific hypotheses about epileptogenesis and its prevention or modification. Acquired human epilepsies with different etiologies share some features with animal models. We identify these commonalities and discuss their relevance to the development of successful epilepsy prevention or disease modification strategies. Risk factors for developing epilepsy that appear common to multiple acute injury etiologies include intracranial bleeding, disruption of the blood-brain barrier, more severe injury, and early seizures within 1 week of injury. In diverse human epilepsies and animal models, seizures appear to propagate within a limbic or thalamocortical/corticocortical network. Common histopathologic features of epilepsy of diverse and mostly focal origin are microglial activation and astrogliosis, heterotopic neurons in the white matter, loss of neurons, and the presence of inflammatory cellular infiltrates. Astrocytes exhibit smaller K+ conductances and lose gap junction coupling in many animal models as well as in sclerotic hippocampi from temporal lobe epilepsy patients. There is increasing evidence that epilepsy can be prevented or aborted in preclinical animal models of acquired epilepsy by interfering with processes that appear common to multiple acute injury etiologies, for example, in post-status epilepticus models of focal epilepsy by transient treatment with a trkB/PLCγ1 inhibitor, isoflurane, or HMGB1 antibodies and by topical administration of adenosine, in the cortical fluid percussion injury model by focal cooling, and in the albumin posttraumatic epilepsy model by losartan. Preclinical studies further highlight the roles of mTOR1 pathways, JAK-STAT3, IL-1R/TLR4 signaling, and other inflammatory pathways in the genesis or modulation of epilepsy after brain injury. The wealth of commonalities, diversity of molecular targets identified preclinically, and likely multidimensional nature of epileptogenesis argue for a combinatorial strategy in prevention therapy. Going forward, the identification of impending epilepsy biomarkers to allow better patient selection, together with better alignment with multisite preclinical trials in animal models, should guide the clinical testing of new hypotheses for epileptogenesis and its prevention.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| | | | - Eleonora Aronica
- Department of (Neuro) Pathology, Academic Medical Center and Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Christophe Bernard
- Aix Marseille Univ, Inserm, INS, Instit Neurosci Syst, Marseille, 13005, France
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Detlev Boison
- Robert Stone Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| | - Martin J Brodie
- Epilepsy Unit, West Glasgow Ambulatory Care Hospital-Yorkhill, Glasgow, UK
| | - Amy R Brooks-Kayal
- Division of Neurology, Departments of Pediatrics and Neurology, University of Colorado School of Medicine, Aurora, CO, USA
- Children's Hospital Colorado, Aurora, CO, USA
- Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jerome Engel
- Departments of Neurology, Neurobiology, and Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Brain Research Institute, University of California, Los Angeles, CA, USA
| | | | | | | | | | - Katja Kobow
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | | | - Phillip L Pearl
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Asla Pitkänen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Noora Puhakka
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Michael A Rogawski
- Department of Neurology, University of California, Davis, Sacramento, CA, USA
| | | | - Matti Sillanpää
- Departments of Child Neurology and General Practice, University of Turku and Turku University Hospital, Turku, Finland
| | - Robert S Sloviter
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Institute for Pharmacological Research, Milan,, Italy
| | - Matthew C Walker
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
18
|
Schidlitzki A, Twele F, Klee R, Waltl I, Römermann K, Bröer S, Meller S, Gerhauser I, Rankovic V, Li D, Brandt C, Bankstahl M, Töllner K, Löscher W. A combination of NMDA and AMPA receptor antagonists retards granule cell dispersion and epileptogenesis in a model of acquired epilepsy. Sci Rep 2017; 7:12191. [PMID: 28939854 PMCID: PMC5610327 DOI: 10.1038/s41598-017-12368-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/08/2017] [Indexed: 01/01/2023] Open
Abstract
Epilepsy may arise following acute brain insults, but no treatments exist that prevent epilepsy in patients at risk. Here we examined whether a combination of two glutamate receptor antagonists, NBQX and ifenprodil, acting at different receptor subtypes, exerts antiepileptogenic effects in the intrahippocampal kainate mouse model of epilepsy. These drugs were administered over 5 days following kainate. Spontaneous seizures were recorded by video/EEG at different intervals up to 3 months. Initial trials showed that drug treatment during the latent period led to higher mortality than treatment after onset of epilepsy, and further, that combined therapy with both drugs caused higher mortality at doses that appear safe when used singly. We therefore refined the combined-drug protocol, using lower doses. Two weeks after kainate, significantly less mice of the NBQX/ifenprodil group exhibited electroclinical seizures compared to vehicle controls, but this effect was lost at subsequent weeks. The disease modifying effect of the treatment was associated with a transient prevention of granule cell dispersion and less neuronal degeneration in the dentate hilus. These data substantiate the involvement of altered glutamatergic transmission in the early phase of epileptogenesis. Longer treatment with NBQX and ifenprodil may shed further light on the apparent temporal relationship between dentate gyrus reorganization and development of spontaneous seizures.
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
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Rebecca Klee
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Inken Waltl
- 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
| | - Sonja Bröer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Sebastian Meller
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Center for Systems Neuroscience, 30559, Hannover, Germany
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Vladan Rankovic
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Institute for Auditory Neuroscience at University Medical Center Göttingen & German Primate Center, Göttingen, Germany
| | - Dandan Li
- Center for Systems Neuroscience, 30559, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 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
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 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
|
19
|
Ferlazzo E, Sueri C, Gasparini S, Russo E, Cianci V, Ascoli M, De Sarro G, Aguglia U. Methodological issues associated with clinical trials in epilepsy. Expert Rev Clin Pharmacol 2017; 10:1103-1108. [PMID: 28715945 DOI: 10.1080/17512433.2017.1356720] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION despite methodological advances in epilepsy clinical trials, the proportion of patients reaching seizure-freedom has not substantially changed over the years. We review the main methodological limitations of current trials, the possible strategies to overcome these limits, and the issues that need to be addressed in next future. Area covered: references were identified by PubMed search until March 2017 and unpublished literature was searched on ClinicalTrials.gov. Add-on trials mainly involve refractory epilepsy subjects, reducing overall response to the investigational drug. The inclusion of subjects with earlier disease from less developed countries has partially allowed overcoming this limitation, but has introduced more random variability of results. Monotherapy trials rise methodological, economical, and ethical concerns with different regulatory requirements in European Union and in the United States of America. Newer trial designs, such as futility trials or 'time-to-event' design, have been implemented. Moreover, both add-on and monotherapy trials results might be affected by patient's ability to recognize and record seizures, and by randomness of seizures occurrence over time. Possible strategies to achieve more reliable outcomes are detailed. Expert commentary: clinical trial methodology needs to be optimized to better address regulatory agencies requirements and to encounter both patients' and clinicians' needs.
Collapse
Affiliation(s)
- Edoardo Ferlazzo
- a Regional Epilepsy Centre , Bianchi-Melacrino-Morelli Hospital , Reggio Calabria , Italy.,b Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Chiara Sueri
- a Regional Epilepsy Centre , Bianchi-Melacrino-Morelli Hospital , Reggio Calabria , Italy
| | - Sara Gasparini
- a Regional Epilepsy Centre , Bianchi-Melacrino-Morelli Hospital , Reggio Calabria , Italy.,b Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Emilio Russo
- c Department of Science of Health , Magna Graecia University , Catanzaro , Italy
| | - Vittoria Cianci
- a Regional Epilepsy Centre , Bianchi-Melacrino-Morelli Hospital , Reggio Calabria , Italy
| | - Michele Ascoli
- a Regional Epilepsy Centre , Bianchi-Melacrino-Morelli Hospital , Reggio Calabria , Italy.,b Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | | | - Umberto Aguglia
- a Regional Epilepsy Centre , Bianchi-Melacrino-Morelli Hospital , Reggio Calabria , Italy.,b Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| |
Collapse
|
20
|
Non-invasive PET imaging of brain inflammation at disease onset predicts spontaneous recurrent seizures and reflects comorbidities. Brain Behav Immun 2017; 61:69-79. [PMID: 28017648 DOI: 10.1016/j.bbi.2016.12.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/05/2016] [Accepted: 12/18/2016] [Indexed: 02/06/2023] Open
Abstract
Brain inflammation is an important factor in the conversion of a healthy brain into an epileptic one, a phenomenon known as epileptogenesis, offering a new entry point for prognostic tools. The development of anti-epileptogenic therapies to treat before or at disease onset is hampered by our inability to predict the severity of the disease outcome. In a rat model of temporal lobe epilepsy we aimed to assess whether in vivo non-invasive imaging of brain inflammation at disease onset was predictive of spontaneous recurrent seizures (SRS) frequency and severity of depression-like and sensorimotor-related comorbidities. To this end, translocator protein, a biomarker of inflammation, was imaged by means of positron emission tomography (PET) 2 and 4weeks post-status epilepticus using [18F]-PBR111. Translocator protein was highly upregulated 2weeks post-status epilepticus in limbic structures (up to 2.1-fold increase compared to controls in temporal lobe, P<0.001), whereas 4weeks post-status epilepticus, upregulation decreased (up to 1.6-fold increase compared to controls in temporal lobe, P<0.01) and was only apparent in a subset of these regions. Animals were monitored with video-electroencephalography during all stages of disease (acute, latent - first seizures appearing around 2weeks post-status epilepticus - and chronic phases), for a total of 12weeks, in order to determine SRS frequency for each subject (range 0.00-0.83SRS/day). We found that regional PET uptake at 2 and 4weeks post-status epilepticus correlated with the severity of depression-like and sensorimotor-related comorbidities during chronic epilepsy (P<0.05 for each test). Regional PET imaging did not correlate with SRS frequency, however, by applying a multivariate data-driven modeling approach based on translocator protein PET imaging at 2weeks post-status epilepticus, we accurately predicted the frequency of SRS (R=0.92; R2=0.86; P<0.0001) at the onset of epilepsy. This study not only demonstrates non-invasive imaging of translocator protein as a prognostic biomarker to ascertain SRS frequency, but also shows its capability to reflect the severity of depression-like and sensorimotor-related comorbidities. Our results are an encouraging step towards the development of anti-epileptogenic treatments by providing early quantitative assessment of SRS frequency and severity of comorbidities with high clinical relevance.
Collapse
|
21
|
Stopping epilepsy treatment in seizure remission: Good or bad or both? Seizure 2017; 44:157-161. [DOI: 10.1016/j.seizure.2016.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 09/01/2016] [Accepted: 09/04/2016] [Indexed: 11/19/2022] Open
|
22
|
|
23
|
|
24
|
|
25
|
Franco V, French JA, Perucca E. Challenges in the clinical development of new antiepileptic drugs. Pharmacol Res 2016; 103:95-104. [DOI: 10.1016/j.phrs.2015.11.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/13/2015] [Accepted: 11/18/2015] [Indexed: 12/26/2022]
|
26
|
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: 23] [Impact Index Per Article: 2.6] [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
|
27
|
Löscher W, Hirsch LJ, Schmidt D. The enigma of the latent period in the development of symptomatic acquired epilepsy - Traditional view versus new concepts. Epilepsy Behav 2015; 52:78-92. [PMID: 26409135 DOI: 10.1016/j.yebeh.2015.08.037] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 08/30/2015] [Indexed: 01/21/2023]
Abstract
A widely accepted hypothesis holds that there is a seizure-free, pre-epileptic state, termed the "latent period", between a brain insult, such as traumatic brain injury or stroke, and the onset of symptomatic epilepsy, during which a cascade of structural, molecular, and functional alterations gradually mediates the process of epileptogenesis. This review, based on recent data from both animal models and patients with different types of brain injury, proposes that epileptogenesis and often subclinical epilepsy can start immediately after brain injury without any appreciable latent period. Even though the latent period has traditionally been the cornerstone concept representing epileptogenesis, we suggest that the evidence for the existence of a latent period is spotty both for animal models and human epilepsy. Knowing whether a latent period exists or not is important for our understanding of epileptogenesis and for the discovery and the trial design of antiepileptogenic agents. The development of antiepileptogenic treatments to prevent epilepsy in patients at risk from a brain insult is a major unmet clinical need.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany.
| | | | | |
Collapse
|
28
|
Barker-Haliski ML, Friedman D, French JA, White HS. Disease Modification in Epilepsy: From Animal Models to Clinical Applications. Drugs 2015; 75:749-67. [DOI: 10.1007/s40265-015-0395-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Leo A, Citraro R, Constanti A, De Sarro G, Russo E. Are big potassium-type Ca2+-activated potassium channels a viable target for the treatment of epilepsy? Expert Opin Ther Targets 2015; 19:911-26. [DOI: 10.1517/14728222.2015.1026258] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
30
|
|
31
|
Affiliation(s)
- Lara E Jehi
- Cleveland Clinic Epilepsy Center, S51 9500 Euclid Avenue, Cleveland, OH, 44195, USA,
| | | |
Collapse
|
32
|
Kaminski RM, Rogawski MA, Klitgaard H. The potential of antiseizure drugs and agents that act on novel molecular targets as antiepileptogenic treatments. Neurotherapeutics 2014; 11:385-400. [PMID: 24671870 PMCID: PMC3996125 DOI: 10.1007/s13311-014-0266-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
A major goal of contemporary epilepsy research is the identification of therapies to prevent the development of recurrent seizures in individuals at risk, including those with brain injuries, infections, or neoplasms; status epilepticus; cortical dysplasias; or genetic epilepsy susceptibility. In this review we consider the evidence largely from preclinical models for the antiepileptogenic activity of a diverse range of potential therapies, including some marketed antiseizure drugs, as well as agents that act by immune and inflammatory mechanisms; reduction of oxidative stress; activation of the mammalian target of rapamycin or peroxisome proliferator-activated receptors γ pathways; effects on factors related to thrombolysis, hematopoesis, and angiogenesis; inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reducatase; brain-derived neurotrophic factor signaling; and blockade of α2 adrenergic and cannabinoid receptors. Antiepileptogenesis refers to a therapy of which the beneficial action is to reduce seizure frequency or severity outlasting the treatment period. To date, clinical trials have failed to demonstrate that antiseizure drugs have such disease-modifying activity. However, studies in animal models with levetiracetam and ethosuximide are encouraging, and clinical trials with these agents are warranted. Other promising strategies are inhibition of interleukin 1β signaling by drugs such as VX-765; modulation of sphingosine 1-phosphate signaling by drugs such as fingolimod; activation of the mammalian target of rapamycin by drugs such as rapamycin; the hormone erythropoietin; and, paradoxically, drugs such as the α2 adrenergic receptor antagonist atipamezole and the CB1 cannabinoid antagonist SR141716A (rimonabant) with proexcitatory activity. These approaches could lead to a new paradigm in epilepsy drug therapy where treatment for a limited period prevents the occurrence of spontaneous seizures, thus avoiding lifelong commitment to symptomatic treatment.
Collapse
Affiliation(s)
| | - Michael A. Rogawski
- />Department of Neurology, University of California, Davis School of Medicine, Sacramento, CA USA
| | | |
Collapse
|