Skip to main content

REVIEW article

Front. Neurol., 04 November 2013
Sec. Epilepsy
This article is part of the Research Topic Role of Intravenous Levetiracetam in Acute Seizure Management View all 13 articles

Prospects of Levetiracetam as a Neuroprotective Drug Against Status Epilepticus, Traumatic Brain Injury, and Stroke

  • 1Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, TX, USA
  • 2Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System, Temple, TX, USA
  • 3Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, USA

Levetiracetam (LEV) is an anti-epileptic drug commonly used for the treatment of partial onset and generalized seizures. In addition to its neuromodulatory and neuroinhibitory effects via its binding to the synaptic vesicle protein SV2A, multiple studies have suggested neuroprotective properties for LEV in both epileptic and non-epileptic conditions. The purpose of this review is to discuss the extent of LEV-mediated protection seen in different neurological conditions, the potential of LEV for easing epileptogenesis, and the possible mechanisms that underlie the protective properties of LEV. LEV has been found to be particularly beneficial for restraining seizures in animal models of spontaneous epilepsy, acute seizures, and status epilepticus (SE). However, its ability for easing epileptogenesis and cognitive dysfunction following SE remains controversial with some studies implying favorable outcomes and others reporting no beneficial effects. Efficacy of LEV as a neuroprotective drug against traumatic brain injury (TBI) has received much attention. While animal studies in TBI models have showed significant neuroprotection and improvements in motor and memory performance with LEV treatment, clinical studies suggest that LEV has similar efficacy as phenytoin in terms of its ability to prevent post-traumatic epilepsy. LEV treatment for TBI is also reported to have fewer adverse effects and monitoring considerations but electroencephalographic recordings suggest the presence of increased seizure tendency. Studies on stroke imply that LEV is a useful alternative to carbamazepine for preventing post-stroke seizures in terms of efficacy and safety. Thus, LEV treatment has promise for restraining SE-, TBI-, or stroke-induced chronic epilepsy. Nevertheless, additional studies are needed to ascertain the most apt dose, timing of intervention, and duration of treatment after the initial precipitating injury and the mechanisms underlying LEV-mediated beneficial effects.

Introduction

Levetiracetam [LEV; 2S-(oxo-1-pyrrolidinyl) butanamide] is an anti-epileptic drug (AED) often utilized for the treatment of partial onset and generalized seizures (1, 2). LEV has both anti-seizure and anti-epileptogenic properties. It has been also proposed that LEV is an attractive AED for managing post-traumatic seizures (PTSs) owing to its beneficial pharmacokinetic attributes, including excellent bioavailability, linear kinetics, minimal plasma protein binding, and rapid achievement of steady state concentrations (24). The underlying mechanisms by which LEV facilitates anti-epileptic and anti-epileptogenic effects are different from classic AEDs. Studies insinuate that LEV bestows its effects mainly through the inhibition of the synaptic vesicle protein 2A (1). Additional investigations have also revealed that LEV can inhibit HVA-Ca2 channels (N-type), negate the inhibition of negative allosteric modulators such as zinc and β-carbolines of γ-aminobutyric acid (GABA)- and glycine-gated currents, and diminish the calcium release from intraneuronal stores (1, 5).

Moreover, a multitude of studies have proposed that LEV has considerable neuroprotective properties in both epileptic and non-epileptic disorders (2, 69). The capability of LEV to augment the manifestation of glial glutamate transporters EAAT1/GLAST and EAAT2/GLT-1 has been proposed as one of the foremost mechanisms through which LEV mediates its neuroprotective properties (2, 10). This hypothesis fits well with one of the conspicuous changes detected following most brain insults, which is increased concentration of glutamate in the extracellular areas causing enhanced activation of N-methyl-d-aspartate (NMDA) receptors and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors on neurons and culminating in significant neurodegeneration (2, 11, 12). The efficacy of LEV as a neuroprotective compound has been examined in several brain injury and neurodegenerative disease prototypes. These include brain damage resulting from status epilepticus (SE) or acute seizures, spontaneous epilepsy, closed head trauma, subarachnoid hemorrhage (SAH), hypoxic-ischemia, and stroke. The goal of this review is to confer the extent of LEV-mediated neuroprotection observed in different brain injury models, the potential of LEV for easing epileptogenesis, and the possible mechanisms that underlie neuroprotective properties of LEV in different neurological conditions.

Efficacy of LEV for Easing Seizures and Seizure-Mediated Neurodegeneration

Levetiracetam administration appears to be beneficial for restraining seizures, and acute seizure or SE induced neurodegeneration in animal models. A single dose of LEV administered 30 min after the onset of behavioral SE was adequate for transiently attenuating seizure activity in animals treated with LEV at 800 mg/kg or higher (13). Increased doses of LEV (1000 mg/kg or higher) dampened behavioral seizures for prolonged periods. When administered early (i.e., 10 min) after the onset of SE, 400 mg/kg of LEV transiently attenuated behavioral seizures and higher doses dampened seizures for relatively longer periods. Pretreatment with LEV prior to pilocarpine injection delayed the onset of seizures but did not significantly alter ictal discharge measured through electroencephalographic (EEG) recordings (13). Analyses with TUNEL staining however demonstrated reduced neuronal injury in the hippocampus and other limbic brain regions in animals that responded behaviorally to LEV.

Levetiracetam treatment appears to mediate anti-seizure effects through several mechanisms. A study using acute hippocampal slices from spontaneously epileptic rats (SERs) have suggested that LEV modulates Ca2+ currents in neurons, as application of 10 μM of LEV decreased the amplitude of the Ca2+ current in CA3 pyramidal neurons and application of 100 nM–1 mM of LEV reduced the Ca2+ current in a concentration-dependent manner (14). LEV also elevated the threshold potential level for activation of the Ca2+ current and reduced the L-type Ca2+ current in neurons. Furthermore, LEV can abolish the SE-induced rise in brain-derived neurotrophic factor, a neurotrophic factor believed to contribute to seizures at higher concentrations. Moreover, administration of LEV after SE can enhance levels of Y1- and Y5-like receptors of neuropeptide Y (NPY; an endogenous anticonvulsant) in all subfields of the hippocampus (15). Also, anti-epileptic effect of LEV is apparent from a study in an animal model of hypoxia-induced seizures (16). LEV pretreatment in postnatal day 10 rats significantly decreased the cumulative duration of hypoxia-induced behavioral and electrographic seizures at 25 and 50 mg/kg doses. Additionally, kainate-induced seizures and neuronal loss were significantly diminished in postnatal day 40 rats previously treated with LEV. Thus, LEV treatment can not only suppress acute seizures but also diminish later-life seizure susceptibility and seizure-induced neuronal injury. This suggests that LEV treatment after injury or acute seizures has potential for disease modification.

A recent study has examined the neuroprotective property of LEV against SE in greater detail (17) by administering the drug 2 h after the onset of SE at 50, 100, or 150 mg/kg. Analyses through staining for Fluoro-Jade B (a marker of degenerating neurons) suggested that low dose administration of LEV (50 mg/kg) can reduce SE-induced loss of CA1 pyramidal neurons and dentate hilar neurons but not CA3 pyramidal neurons. However, LEV treatment at a higher dose (100 mg/kg) reduced degenerating neurons in CA1 and CA3 pyramidal cell layers as well as the dentate hilus. Furthermore, a much higher dose of LEV (150 mg/kg) greatly reduced the numbers of degenerating neurons in the CA3 pyramidal cell layer. Interestingly, when different doses of LEV were combined with diazepam (10 mg/kg), neurodegeneration was exacerbated in the hippocampus. Collectively, this study demonstrated that LEV alone is more efficacious for preventing SE-induced neurodegeneration in the hippocampus than other AEDs such as diazepam or valproate (17). Furthermore, the finding that combined administration of LEV and diazepam actually increases neurodegeneration suggested that LEV negatively interacts with diazepam, implying that LEV may be more suitable as a first line drug to minimize SE-induced neurodegeneration rather than as an add-on drug with benzodiazepines (17). Thus, LEV administration after the onset of SE is beneficial for suppressing seizures as well as reducing neurodegeneration. Mechanisms of LEV-mediated neuroprotection likely include its anti-seizure effects as well as its purported ability to decrease the expression of pro-oxidant protein iNOS and increase the expression of the antioxidant protein cystine/glutamate exchanger in the hippocampus (18). A study in pilocarpine model of SE also showed that LEV pretreatment could counteract oxidative stress through maintenance of lipid peroxidation, nitrite-nitrate levels, catalase activity, and glutathione at normal levels in the hippocampus (19).

Usefulness of LEV for Easing Epileptogenesis

Prolonged LEV treatment after SE appears to delay or restrain the development of chronic epilepsy in animal models. A study examined the effects of chronic LEV treatment on hippocampal field responses in rats subjected to pilocarpine induced SE (20). Hippocampal field potentials were recorded in vivo in anesthetized animals after 3-day washout period that followed 21-day treatment with different doses of LEV (50, 150, or 300 mg/kg/day) administered via osmotic minipumps. Chronic treatment with LEV yielded clinically relevant plasma concentrations throughout the experiment with complete washout of the drug 3 days after treatment cessation. At this point of time post-SE rats chronically treated with vehicle developed clear signs of hippocampal hyperexcitability typified by increased amplitude of population spike (PS) recorded in the DG and reduced paired-pulse inhibition in the CA1 area. LEV treatment dose-dependently counteracted these long-term effects of SE. Furthermore, at the dose of 300 mg/kg/day, LEV restored these parameters back to control levels (20). Several other studies have also shown beneficial effects of LEV treatment in acute seizure models. For example, the development of kindling (a progressive increase in seizure severity induced by repeated brain stimulation at certain intervals), and kindling-related abnormal gene expression can be considerably modulated through daily application of LEV (15, 21, 22).

Furthermore, LEV administration at 40 mg/kg is efficacious not only for suppressing the development of kindling but also for dampening kindling-induced expression of multiple immediate early genes (IEGs) including many synaptic plasticity-related IEGs, and some late response genes encoding transcription factors, neurotrophic factors, and proteins that are known to regulate synaptic remodeling (23). An additional potential mechanism by which LEV suppresses the development of kindling is through significant inhibition of kindling-induced synaptic potentiation (24). LEV treatment after kindling can also prevent asymmetric accumulation of hippocampal 7S SNARE complexes [the secretory machinery responsible for neurotransmitter (NT) release] and accumulation of SV2 (25). Thus, LEV treatment can ease multiple abnormalities induced by kindling at cellular and molecular levels.

Moreover, a study in a kainate model of SE examining the long-term effects of LEV treatment, commencing a day after the onset of SE and continuing for 25 days (26) demonstrated that LEV treatment after SE can decrease the mean duration of spontaneous electrographic seizures in the chronic phase after SE. Interestingly, LEV administration also greatly eased SE-induced aberrant migration of newly born neurons into the dentate hilus, an abnormal process that is believed to contribute to the formation of aberrant hippocampal circuitry and epileptogenesis after SE (27, 28). LEV administration has also been found to be effective for easing inflammatory responses in the hippocampus and piriform cortex of epileptic rats (29). These brain regions in epileptic animals typically demonstrate reactive astrocytes and activated microglia displaying strong expression of IL-1β and interleukin-1 receptor subtype 1 (IL-1R1). Interestingly, LEV administration reduced reactive gliosis and expression levels of IL-1β in both of these brain regions. These findings suggested that LEV likely mediates its anti-epileptogenic effects at least partially through modulation of inflammation in epileptic brain regions. Studies in SERs have also suggested anti-epileptogenic effects of LEV (30). Administration of LEV (80 mg/kg/day) to SERs from postnatal weeks 5–8 significantly inhibited seizures at postnatal weeks 5–13. It is of interest to note that inhibition of seizure expression in SERs was still apparent 5 weeks after the termination of LEV treatment, reinforcing that LEV possesses anti-epileptogenic properties.

From the above studies, it is tempting to conclude that chronic treatment with LEV is efficacious for restraining the evolution of initial SE-induced brain insults into a state of hippocampal hyperexcitability and chronic epilepsy. However, currently, there is no clear consensus regarding anti-epileptogenic effects of LEV in SE models. For instance, a study in amygdala kindling model of SE showed that prophylactic treatment with LEV has no effect on epileptogenesis, neuronal damage, or behavioral alterations in rats (31). In one set of studies, LEV treatment was initiated 24 h after onset of electrical amygdala stimulation without termination of SE and continued for 8 weeks using osmotic minipumps. In another set of studies, LEV treatment commenced 4 h after the onset of SE with seizure termination through diazepam and continued for 5 weeks. Interestingly, with either treatment regimen, LEV did not exert anti-epileptogenic or neuroprotective activity. Furthermore, behavioral hyperexcitability and learning deficits were not affected by treatment with LEV after SE. Another study investigating the effects of LEV on visual-spatial memory following SE corroborated these findings (32). Adult rats subjected to SE were treated first with LEV or vehicle for 14 days, tested for visual-spatial memory in the Morris water-maze and then used for unit recording in the CA1 region of the hippocampus. Animals undergoing SE displayed impaired learning and memory function in the water-maze test and abnormalities in firing patterns of pyramidal neurons (place cells) in the CA1 cell layer. LEV treatment had no major effects on water-maze performance or place cell function. Histological analyses however revealed severe neurodegeneration in the CA1 pyramidal cell layer of rats receiving vehicle after SE and relatively reduced neurodegeneration in rats receiving LEV after SE.

Thus, the extent of neuroprotection mediated by LEV treatment was not adequate for preventing SE-induced cognitive dysfunction. However, discrepancy in results between studies may reflect differences in species and strains of animals examined, timing, and dose of LEV treatment after SE, and severity of SE at the time of commencement of LEV treatment. Timing of treatment after SE is particularly important because a study using a rat perforant pathway stimulation model has shown that administration of LEV 5 h after SE does not protect from mitochondrial dysfunction but LEV treatment during established SE prevents mitochondrial dysfunction (7, 33).

Promise of LEV for Mediating Neuroprotection Against Traumatic Brain Injury

Investigation of the effects of LEV in animal models of closed head injury (CHI) and SAH suggested that LEV is neuroprotective against traumatic brain injury (TBI) (6). In this study, a single intravenous dose of LEV has been shown to improve both functional and histological outcomes after CHI. Moreover, the beneficial effects seemed specific for LEV treatment as fosphenytoin administration did not result in such effects. Administration of LEV also improved functional outcomes and reduced vasospasm following SAH. This was the first study to suggest that LEV could be a therapeutic alternative to phenytoin for TBI in clinical situations where seizure prophylaxis drugs are indicated. Moreover, a recent study has examined the effects of LEV on motor and cognitive function in a rat prototype of TBI (2). Adult male rats were administered LEV (50 mg/kg, i.p) or vehicle daily for 20 days beginning 1 day following a controlled cortical impact (CCI) injury or sham surgery. Animals were assessed for various behavioral tests, which comprised assessment of motor function via beam walking test and spatial learning and memory function through Y-maze and Morris water-maze tests. The results showed that daily LEV treatment for 20 days improved motor function and enhanced novel arm exploration in the Y-maze. Furthermore, LEV treatment promoted greater sparing of hippocampal neurons, decreased contusion volumes, reversed TBI-induced decreases observed in glutamate transporters and markers that promote neuroplasticity, and reduced the expression of pro-inflammatory cytokine IL-1β. However, LEV treatment did not improve spatial learning ability in rats with TBI. Collectively, these animal studies imply that daily LEV treatment has favorable effects on structural, molecular, and some of the behavioral components of neurological improvements after TBI, likely through modulation of excitatory and neuroinflammatory pathways (2).

Additionally, several clinical studies have ascertained the efficacy of LEV for preventing PTS or post-traumatic epilepsy (PTE). Amongst ∼275,000 individuals who are typically hospitalized with TBI every year, ∼7% experience PTS (34). As per guidelines of the Brain Trauma Foundation and the American Academy of Neurology for the management of severe TBI, administration of AEDs to prevent PTS is recommended only through the initial 7 days after TBI (34). Amid AEDs, the efficiency of phenytoin treatment has been extensively examined for preventing PTS after TBI. However, several clinical studies suggest that LEV treatment after TBI is also effective for decreasing the predilection for developing PTS. Jones and colleagues analyzed EEG recordings from patients receiving phenytoin or LEV for seizure prevention following severe TBI (35). This comparative analysis revealed that LEV is as efficient as phenytoin in averting early PTS but is allied with an increased seizure predisposition based on evaluation of EEG recordings. Another open label, non-randomized phase 2 study assessed the safety, tolerability, and effectiveness of LEV therapy in patients with TBI exhibiting greater susceptibility for PTE (36). LEV treatment was initiated within 8 h after injury and continued for 30 days in this study. Two-year follow-up uncovered that occurrence of PTE in patients receiving LEV (11%) is less than that observed in untreated TBI patients (20%). However, several recent clinical studies in TBI patients report that LEV does not outperform phenytoin as a prophylaxis drug against PTS (37, 38). Another recent clinical study has reported that LEV treatment to children ages 6–17 years with risk factors for the development of PTE decreased the incidence of PTE, as only 1 in 40 patients receiving LEV displayed PTE (39). Collectively, from the above studies, it emerges that LEV has analogous ability as phenytoin for thwarting PTS after TBI. It is also reported that LEV treatment for TBI is linked with fewer adverse effects and monitoring considerations [for details, see the review by Ref. (34)]. Nonetheless, because LEV administration was accompanied by an increased seizure propensity (35), the Brain Trauma Foundation has recommended using phenytoin for early PTS prophylaxis (34).

LEV as a Neuroprotective Compound Against Stroke

Seizures following stroke is one of the causes of epilepsy in adults, particularly in elderly patients (40). Seizures typically occur in ∼10% of stroke patients, depending on risk factors, such as the type of stroke, location of stroke-induced damage in the brain, and severity of the stroke (40). However, stroke accounts for ∼50% of seizures in individuals above the age of 65 years (41). Classically, the use of AEDs to avert recurrent post-stroke seizures is recommended. LEV has been suggested as a first-choice drug against post-stroke seizures, based on safety and efficacy profiles in clinical studies (42). Kutlu and colleagues examined the suitability of LEV monotherapy in individuals aged 60 or older and exhibiting a minimum of two late-onset post-stroke seizures (43). At daily doses of 1000–2000 mg, they reported that 82.4% of the patients were seizure free but seven patients (20.6%) had side effects. These results suggested that LEV monotherapy is efficient and well tolerated in elderly patients with late-onset post-stroke seizures. Consoli and associates compared the efficacy of LEV treatment with carbamazepine (CBZ) in patients with post-stroke seizures in a multicenter randomized open label study (41). Evaluation of results in 106 patients (52 treated with LEV and 54 treated with CBZ) showed no noteworthy variance in the number of seizure free patients between LEV and CBZ. Yet, interval to the first recurrence tended to be longer in patients receiving LEV. The results also suggested that LEV treatment caused considerably less side effects than CBZ, as attention deficit, frontal executive functions, and functional scales (Activities of Daily Living and Instrumental Activities of Daily Living indices) were notably poorer in patients receiving CBZ (41).

Thus, studies conducted so far imply that LEV is a useful alternative to CBZ for post-stroke seizures, predominantly in terms of efficacy and decreased adverse effects. However, another recent study reported that LEV is not effective for the treatment of central post-stroke pain, a severe chronic neuropathic pain state called allodynia resulting from a vascular lesion (44). Considering these, further studies are needed to ascertain the efficacy of LEV as a suitable neuroprotective and seizure-preventing drug after stroke. Additionally, rigorous studies in animal models of stroke are needed to understand the potential anti-seizure, neuroprotective, and anti-epileptogenic effects of LEV following stroke.

Overall Conclusion

From the analysis of literature pertaining to LEV treatment mediated protection in neurological disorders, it emerges that LEV treatment has potential for restraining SE-, TBI-, and stroke-induced chronic epilepsy development. Particularly, LEV administration has been found to be advantageous for restraining seizures and/or seizure-induced neurodegeneration in animal models of spontaneous epilepsy, acute seizures, and SE. LEV treatment appears to mediate anti-seizure and neuroprotective effects via modulation of Ca2+ currents in neurons, inhibition of the up-regulation of brain-derived neurotrophic factor, increases in NPY receptors, and antioxidant proteins, and decreases in pro-oxidant proteins (14, 15, 17). Nonetheless, the capability of LEV for easing epileptogenesis and cognitive dysfunction following SE remains contentious. Several studies report promising outcomes such as delayed development of hippocampal hyperexcitability, restraint of electrographic seizures, diminishment in the abnormal migration of newly born neurons into the dentate hilus, and inhibition of inflammatory responses in SE models (20, 26, 27, 29). There are also reports of mitigation of synaptic potentiation and abnormal expression of IEGs, and prevention of abnormal accumulation of 7S SNARE complexes and SV2 in kindling models (15, 2224). However, some studies report no beneficial effects of LEV treatment after SE in terms of easing epileptogenesis or cognitive dysfunction (31, 32). Discrepancy in the findings between studies may reflect differences in the timing of intervention with LEV, doses of LEV employed and severity of SE at the time of initial intervention with LEV.

The efficiency of LEV as a neuroprotective drug against TBI has received much consideration. Animal studies in TBI models validate greater sparing of hippocampal neurons, and improved motor and memory function with LEV treatment (2, 6). On the other hand, results of several recent clinical trials convey that LEV has comparable efficacy as phenytoin in terms of its ability for preventing PTE (34, 3739). The other positive effects of LEV treatment for TBI comprise fewer adverse effects and monitoring issues. However, one caveat of LEV treatment for TBI is the presence of increased seizure propensity in long-term EEG recordings (34, 35). Studies on stroke imply that LEV is a useful alternative to CBZ for preventing post-stroke seizures including elderly patients, in terms of efficacy and safety (4043) but does not seem to be useful for easing central post-stroke pain (44). Taken together, studies conducted so far suggest that LEV treatment is useful for easing SE-, TBI-, and stroke-induced chronic epilepsy development. Nevertheless, rigorous additional studies in animal models are needed to ascertain the most beneficial dose, timing of intervention, and duration of treatment after the initial precipitating injury and mechanisms underlying LEV-mediated beneficial effects on epileptogenesis.

Conflict of Interest Statement

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

This work was supported by grants from the Texas A&M Health Science Center (Emerging Technology Funds from the State of Texas), National Institute of Neurological Disorders and Stroke (R01 NS054780), and the Department of Veterans Affairs (VA Merit Review Award).

References

1. Surges R, Volynski KE, Walker MC. Is levetiracetam different from other antiepileptic drugs? Levetiracetam and its cellular mechanism of action in epilepsy revisited. Ther Adv Neurol Disord (2008) 1:13–24. doi: 10.1177/1756285608094212

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

2. Zou H, Brayer SW, Hurwitz M, Niyonkuru C, Fowler LE, Wagner AK. Neuroprotective, neuroplastic, and neurobehavioral effects of daily treatment with levetiracetam in experimental traumatic brain injury. Neurorehabil Neural Repair (2013) 27(9):878–88. doi:10.1177/1545968313491007

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

3. De Smedt T, Raedt R, Vonck K, Boon P. Levetiracetam: the profile of a novel anticonvulsant drug-part I: preclinical data. CNS Drug Rev (2007) 13:43–56. doi:10.1111/j.1527-3458.2007.00005.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

4. De Smedt T, Raedt R, Vonck K, Boon P. Levetiracetam: part II, the clinical profile of a novel anticonvulsant drug. CNS Drug Rev (2007) 13:57–78. doi:10.1111/j.1527-3458.2007.00005.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

5. Crepeau AZ, Treiman DM. Levetiracetam: a comprehensive review. Expert Rev Neurother (2010) 10:159–71. doi:10.1586/ern.10.3

CrossRef Full Text

6. Wang H, Gao J, Lassiter TF, McDonagh DL, Sheng H, Warner DS, et al. Levetiracetam is neuroprotective in murine models of closed head injury and subarachnoid hemorrhage. Neurocrit Care (2006) 5(1):71–8. doi:10.1385/NCC:5:1:71

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

7. Gibbs JE, Cock HR. Administration of levetiracetam after prolonged status epilepticus does not protect from mitochondrial dysfunction in a rodent model. Epilepsy Res (2007) 73:208–12. doi:10.1016/j.eplepsyres.2006.09.005

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

8. Szaflarski JP, Sangha KS, Lindsell CJ, Shutter LA. Prospective, randomized, single-blinded comparative trial of intravenous levetiracetam versus phenytoin for seizure prophylaxis. Neurocrit Care (2010) 12:165–72. doi:10.1007/s12028-009-9304-y

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

9. Steinbaugh LA, Lindsell CJ, Shutter LA, Szaflarski JP. Initial EEG predicts outcomes in a trial of levetiracetam vs. fosphenytoin for seizure prevention. Epilepsy Behav (2012) 23:280–4. doi:10.1016/j.yebeh.2011.12.005

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

10. Ueda Y, Doi T, Tokumaru J, Yokoyama H, Nakajima A, Mitsuyama Y, et al. Collapse of extracellular glutamate regulation during epileptogenesis: down-regulation and functional failure of glutamate transporter function in rats with chronic seizures induced by kainic acid. J Neurochem (2001) 76(3):892–900. doi:10.1046/j.1471-4159.2001.00087.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

11. Vespa P, Prins M, Ronne-Engstrom E, Caron M, Shalmon E, Hovda DA, et al. Increase in extracellular glutamate caused by reduced cerebral perfusion pressure and seizures after human traumatic brain injury: a microdialysis study. J Neurosurg (1998) 89:971–82. doi:10.3171/jns.1998.89.6.0971

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

12. Takeuchi H, Jin S, Suzuki H, Doi Y, Liang J, Kawanokuchi J, et al. Blockade of microglial glutamate release protects against ischemic brain injury. Exp Neurol (2008) 214:144–6. doi:10.1016/j.expneurol.2008.08.001

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

13. Zheng Y, Moussally J, Cash SS, Karnam HB, Cole AJ. Intravenous levetiracetam in the rat pilocarpine-induced status epilepticus model: behavioral, physiological and histological studies. Neuropharmacology (2010) 58:793–8. doi:10.1016/j.neuropharm.2009.12.007

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

14. Yan HD, Ishihara K, Seki T, Hanaya R, Kurisu K, Arita K, et al. Inhibitory effects of levetiracetam on the high-voltage-activated L-type Ca2+ channels in hippocampal CA3 neurons of spontaneously epileptic rat (SER). Brain Res Bull (2013) 2013(90):142–8. doi:10.1016/j.brainresbull.2012.10.006

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

15. Husum H, Bolwig TG, Sánchez C, Mathé AA, Hansen SL. Levetiracetam prevents changes in levels of brain-derived neurotrophic factor and neuropeptide Y mRNA and of Y1- and Y5-like receptors in the hippocampus of rats undergoing amygdala kindling: implications for antiepileptogenic and mood-stabilizing properties. Epilepsy Behav (2004) 5(2):204–15. doi:10.1016/j.yebeh.2003.12.004

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

16. Talos DM, Chang M, Kosaras B, Fitzgerald E, Murphy A, Folkerth RD, et al. Antiepileptic effects of levetiracetam in a rodent neonatal seizure model. Pediatr Res (2012) 73:24–30. doi:10.1038/pr.2012.151

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

17. Lee DS, Ryu HJ, Kim JE, Choi HC, Kim YI, Song HK, et al. The effect of levetiracetam on status epilepticus-induced neuronal death in the rat hippocampus. Seizure (2013) 22:368–77. doi:10.1016/j.seizure.2013.02.005

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

18. Ueda Y, Doi T, Takaki M, Nagatomo K, Nakajima A, Willmore LJ. Levetiracetam enhances endogenous antioxidant in the hippocampus of rats: in vivo evaluation by brain microdialysis combined with ESR spectroscopy. Brain Res (2009) 1266:1–7. doi:10.1016/j.brainres.2009.02.040

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

19. Oliveira AA, Almeida JP, Freitas RM, Nascimento VS, Aguiar LM, Júnior HV, et al. Effects of levetiracetam in lipid peroxidation level, nitrite-nitrate formation and antioxidant enzymatic activity in mice brain after pilocarpine-induced seizures. Cell Mol Neurobiol (2007) 27:395–406. doi:10.1007/s10571-006-9132-y

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

20. Margineanu DG, Matagne A, Kaminski RM, Klitgaard H. Effects of chronic treatment with levetiracetam on hippocampal field responses after pilocarpine-induced status epilepticus in rats. Brain Res Bull (2008) 77:282–5. doi:10.1016/j.brainresbull.2008.07.006

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

21. Loscher W, Honack D, Rundfeldt C. Antiepileptogenic effects of the novel anticonvulsant levetiracetam (ucb L059) in the kindling model of temporal lobe epilepsy. J Pharmacol Exp Ther (1998) 284:474–9.

22. Ishimaru Y, Chiba S, Serikawa T, Sasa M, Inaba H, Tamura Y, et al. Effects of levetiracetam on hippocampal kindling in Noda epileptic rats. Brain Res (2010) 1309:104–9. doi:10.1016/j.brainres.2009.10.056

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

23. Christensen KV, Leffers H, Watson WP, Sánchez C, Kallunki P, Egebjerg J. Levetiracetam attenuates hippocampal expression of synaptic plasticity-related immediate early and late response genes in amygdala-kindled rats. BMC Neurosci (2010) 11:9. doi:10.1186/1471-2202-11-9

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

24. Sugaya Y, Maru E, Kudo K, Shibasaki T, Kato N. Levetiracetam suppresses development of spontaneous EEG seizures and aberrant neurogenesis following kainate-induced status epilepticus. Brain Res (2010) 1352:187–99. doi:10.1016/j.brainres.2010.06.061

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

25. Matveeva EA, Vanaman TC, Whiteheart SW, Slevin JT. Levetiracetam prevents kindling-induced asymmetric accumulation of hippocampal 7S SNARE complexes. Epilepsia (2008) 49:1749–58. doi:10.1111/j.1528-1167.2008.01687.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

26. Sugaya Y, Jinde S, Kato N, Maru E. Levetiracetam inhibits kindling-induced synaptic potentiation in the dentate gyrus of freely moving rats. Neurosci Res (2010) 66:228–31. doi:10.1016/j.neures.2009.10.014

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

27. Jung KH, Chu K, Kim M, Jeong SW, Song YM, Lee ST, et al. Continuous cytosine-b-D-arabinofuranoside infusion reduces ectopic granule cells in adult rat hippocampus with attenuation of spontaneous recurrent seizures following pilocarpine-induced status epilepticus. Eur J Neurosci (2004) 19:3219–26. doi:10.1111/j.0953-816X.2004.03412.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

28. Scharfman HE, McCloskey DP. Postnatal neurogenesis as a therapeutic target in temporal lobe epilepsy. Epilepsy Res (2009) 85:150–61. doi:10.1016/j.eplepsyres.2009.03.006

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

29. Kim JE, Choi HC, Song HK, Jo SM, Kim DS, Choi SY, et al. Levetiracetam inhibits interleukin-1 beta inflammatory responses in the hippocampus and piriform cortex of epileptic rats. Neurosci Lett (2010) 471:94–9. doi:10.1016/j.neulet.2010.01.018

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

30. Yan HD, Ji-Qun C, Ishihara K, Nagayama T, Serikawa T, Sasa M. Separation of antiepileptogenic and antiseizure effects of levetiracetam in the spontaneously epileptic rat (SER). Epilepsia (2005) 46:1170–7. doi:10.1111/j.1528-1167.2005.35204.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

31. Brandt C, Glien M, Gastens AM, Fedrowitz M, Bethmann K, Volk HA, et al. Prophylactic treatment with levetiracetam after status epilepticus: lack of effect on epileptogenesis, neuronal damage, and behavioral alterations in rats. Neuropharmacology (2007) 53:207–21. doi:10.1016/j.neuropharm.2007.05.001

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

32. Zhou JL, Zhao Q, Holmes GL. Effect of levetiracetam on visual-spatial memory following status epilepticus. Epilepsy Res (2007) 73:65–74. doi:10.1016/j.eplepsyres.2006.08.007

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

33. Gibbs JE, Walker MC, Cock HR. Levetiracetam: antiepileptic properties and protective effects on mitochondrial dysfunction in experimental status epilepticus. Epilepsia (2006) 47:469–78. doi:10.1111/j.1528-1167.2006.00454.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

34. Torbic H, Forni AA, Anger KE, Degrado JR, Greenwood BC. Use of antiepileptics for seizure prophylaxis after traumatic brain injury. Am J Health Syst Pharm (2013) 70:759–66. doi:10.2146/ajhp120203

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

35. Jones KE, Puccio AM, Harshman KJ, Falcione B, Benedict N, Jankowitz BT, et al. Levetiracetam versus phenytoin for seizure prophylaxis in severe traumatic brain injury. Neurosurg Focus (2008) 25:E3. doi:10.3171/FOC.2008.25.10.E3

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

36. Klein P, Herr D, Pearl PL, Natale J, Levine Z, Nogay C, et al. Results of phase 2 safety and feasibility study of treatment with levetiracetam for prevention of posttraumatic epilepsy. Arch Neurol (2012) 69:1290–5. doi:10.1001/archneurol.2012.445

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

37. Inaba K, Menaker J, Branco BC, Gooch J, Okoye OT, Herrold J, et al. A prospective multicenter comparison of levetiracetam versus phenytoin for early posttraumatic seizure prophylaxis. J Trauma Acute Care Surg (2013) 74:766–71. doi:10.1097/TA.0b013e3182826e84

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

38. Kruer RM, Harris LH, Goodwin H, Kornbluth J, Thomas KP, Slater LA, et al. Changing trends in the use of seizure prophylaxis after traumatic brain injury: a shift from phenytoin to levetiracetam. J Crit Care (2013) 28(5):e9–13. doi:10.1016/j.jcrc.2012.11.020

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

39. Pearl PL, McCarter R, McGavin CL, Yu Y, Sandoval F, Trzcinski S, et al. Results of phase II levetiracetam trial following acute head injury in children at risk for posttraumatic epilepsy. Epilepsia (2013) 54(9):e135–37. doi:10.1111/epi.12326

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

40. Gilad R. Management of seizures following a stroke: what are the options? Drugs Aging (2012) 29(7):533–8. doi:10.2165/11631540-000000000-00000

CrossRef Full Text

41. Consoli D, Bosco D, Postorino P, Galati F, Plastino M, Perticoni GF, et al. Levetiracetam versus carbamazepine in patients with late poststroke seizures: a multicenter prospective randomized open-label study (EpIC Project). Cerebrovasc Dis (2012) 34:282–9. doi:10.1159/000342669

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

42. Belcastro V, Costa C, Galletti F, Autuori A, Pierguidi L, Pisani F, et al. Levetiracetam in newly diagnosed late-onset post-stroke seizures: a prospective observational study. Epilepsy Res (2008) 82:223–6. doi:10.1016/j.eplepsyres.2008.08.008

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

43. Kutlu G, Gomceli YB, Unal Y, Inan LE. Levetiracetam monotherapy for late poststroke seizures in the elderly. Epilepsy Behav (2008) 13:542–4. doi:10.1016/j.yebeh.2008.04.025

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

44. Jungehulsing GJ, Israel H, Safar N, Taskin B, Nolte CH, Brunecker P, et al. Levetiracetam in patients with central neuropathic post-stroke pain – a randomized, double-blind, placebo-controlled trial. Eur J Neurol (2013) 20:331–7. doi:10.1111/j.1468-1331.2012.03857.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Keywords: traumatic brain injury, stroke, acute seizures, chronic epilepsy, post-traumatic seizures, post-traumatic epilepsy, neurodegeneration, epileptogenesis

Citation: Shetty AK (2013) Prospects of levetiracetam as a neuroprotective drug against status epilepticus, traumatic brain injury, and stroke. Front. Neurol. 4:172. doi: 10.3389/fneur.2013.00172

Received: 23 August 2013; Paper pending published: 15 October 2013;
Accepted: 21 October 2013; Published online: 04 November 2013.

Edited by:

Batool F. Kirmani, Texas A&M Health Science Center College of Medicine, USA

Reviewed by:

Batool F. Kirmani, Texas A&M Health Science Center College of Medicine, USA
Lee A. Shapiro, Texas A&M University System Health Science Center College of Medicine, USA
Robert Joseph Buchanan, Seton Brain and Spine Institute, USA; The University of Texas, USA

Copyright: © 2013 Shetty. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ashok K. Shetty, Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, 5701 Airport Road, Module C, Temple, TX 76501, USA e-mail: shetty@medicine.tamhsc.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.