Skip to main content

MINI REVIEW article

Front. Neurol., 12 February 2021
Sec. Epilepsy

The Putative Role of mTOR Inhibitors in Non-tuberous Sclerosis Complex-Related Epilepsy

  • 1Department of Neurological Surgery, University of Washington, Seattle, WA, United States
  • 2Department of Neurosurgery, Seattle Children's Hospital, Seattle, WA, United States

Epilepsy affects ~5 out of every 10,000 children per year. Up to one-third of these children have medically refractory epilepsy, with limited to no options for improved seizure control. mTOR, a ubiquitous 289 kDa serine/threonine kinase in the phosphatidylinositol 3-kinase (PI3K)-related kinases (PIKK) family, is dysregulated in a number of human diseases, including tuberous sclerosis complex (TSC) and epilepsy. In cell models of epilepsy and TSC, rapamycin, an mTOR inhibitor, has been shown to decrease seizure frequency and duration, and positively affect cell growth and morphology. Rapamycin has also been shown to prevent or improve epilepsy and prolong survival in animal models of TSC. To date, clinical studies looking at the effects of mTOR inhibitors on the reduction of seizures have mainly focused on patients with TSC. Everolimus (Novartis Pharmaceuticals), a chemically modified rapamycin derivative, has been shown to reduce seizure frequency with reasonable safety and tolerability. Mutations in mTOR or the mTOR pathway have been found in hemimegalencephaly (HME) and focal cortical dysplasias (FCDs), both of which are highly correlated with medically refractory epilepsy. Given the evidence to date, a logical next step is to investigate the role of mTOR inhibitors in the treatment of children with medically refractory non-TSC epilepsy, particularly those children who have also failed resective surgery.

Introduction

Epilepsy affects ~5 out of every 10,000 children per year (1, 2), with up to one-third of these children having medically refractory epilepsy. When looking only at children with metabolic or structural epilepsy, as opposed to genetic or idiopathic epilepsy, the number of children who continue to have seizures despite multiple antiepileptic drugs (AEDs) jumps to 50% (35). A fraction of these patients are referred for surgical evaluation, and if deemed surgical candidates, ~60–80% become seizure free after epilepsy neurosurgery (618). However, for children whose seizures persist despite medical therapy and epilepsy surgery, there are limited to no options for improved seizure control.

mTOR, a ubiquitous 289 kDa serine/threonine kinase in the phosphatidylinositol 3-kinase (PI3K)-related kinases (PIKK) family (19) is dysregulated in a number of human diseases, including tuberous sclerosis complex (TSC) and epilepsy. Inhibition of mTOR reduces cell proliferation, angiogenesis, and glucose uptake by cells in both in vivo and in vitro studies (2023). Widely used in the treatment of subependymal giant cell astrocytoma (SEGA) in patients with TSC, including children <2 years of age, mTOR inhibitors have been shown to be beneficial for tumor control and also seizure control in this patient population (2426). However, there are still limited data on the use of mTOR inhibitors for the treatment of non-TSC epilepsy.

mTOR as an Anti-epileptic Target

In models of pilocarpine-induced seizures, representative of acquired limbic epilepsy, it has been shown that after pilocarpine injection, levels of phosphorylated S6K in the hippocampus and cortex increase at about 30 min and peak at 1 h (27). This rise in phosphorylated S6K can be blocked by pre-treatment with systemic rapamycin, an mTOR inhibitor, at 5 mg/kg/day for 3 days prior to pilocarpine injection, though the pre-treatment does not affect the severity of the acute seizures. In contrast, pilocarpine-treated animals with recurrent spontaneous seizures who are treated with chronic systemic rapamycin (5 mg/kg/day for 3 days, then every other day for 3 weeks) demonstrate a reduction in seizure frequency and duration during treatment; both of which then gradually increase following withdrawal of rapamycin. In a model of pilocarpine-induced status epilepticus, continuous infusion of rapamycin into the dorsal hippocampus prevented mossy fiber sprouting in the molecular and granular layers that then emerged upon withdrawal of treatment (28). Interestingly, when rapamycin was administered after mossy fiber sprouting began (2 months after seizure onset), no effects were seen. This effect on mossy fiber sprouting has been confirmed by others (27).

In another model of temporal lobe epilepsy induced by kainate injection, elevation in phosphorylated S6K in the hippocampus and cortex was noted at 1 h after kainate injection with a peak at 3–6 h and a return to baseline at 24 h (29). An additional phase of rising phosphorylated S6K levels was noted in the hippocampus only, starting at 3 days after injection, peaking by 5 days, and returning to baseline by 5 weeks. Similar to the studies by Huang et al. (27), when rapamycin was administered systemically at a dose of 6 mg/kg/day for 3 days before injection, the biphasic rise in phosphorylated S6K was blocked, however the severity of the acute seizures was not affected. Furthermore, rapamycin pre-treatment reduced kainate seizure-induced hippocampal cell death, kainate-seizure induced dentate granule cell neurogenesis, supragranular mossy fiber sprouting, and chronic recurrent kainate-induced spontaneous epilepsy. When rapamycin treatment was changed from a pre-treatment to a post-treatment paradigm (6 mg/kg/day for 6 days starting 24 h after onset of kainate status epilepticus, then every other day from that point forward), late phase mTOR activation, mossy fiber sprouting, and chronic kainate-induced spontaneous seizures were all reduced. There was no effect on cell death or neurogenesis (27).

In WAG/Rij rats, a genetic model of absence epilepsy, Russo et al. found that early chronic treatment, sub-chronic treatment, or acute treatment with rapamycin all had anti-absence properties. In this model, bacterial lipopolysaccharide (LPS) endotoxin administration causes an increased inflammatory response which results in an increase in absence seizures. However, with co-administration of rapamycin and LPS, this seizure increase was blocked, suggesting an anti-inflammatory pathway (30).

As expected, in models of TSC where Tsc1 is conditionally deleted from most cortical neurons, both rapamycin and RAD-001 (another mTOR inhibitor) increase survival, improve the histological phenotype (cortical organization, soma size and polarity, and myelination), and reduce seizures (31). Additional work has shown that rapamycin completely reverses the elevated endoplasmic reticulum and oxidative stress that can lead to cell death in Tsc2-deficient hippocampal neurons and Tsc1 deficient brain lysates (32). In another model of cortical dysgenesis in which PTEN is conditionally deleted from cortical neurons, rapamycin administration also improved the histological abnormalities (enlarged, disorganized neurons), reduced abnormal EEG activity, and suppressed the frequency and duration of spontaneous seizures (33).

In an animal model of tuberous sclerosis in which Tsc1 is conditionally deleted primarily in glia, rapamycin had significant beneficial effects (34). When rapamycin was given systemically starting at P14 (before the onset of seizures), astrogliosis was prevented, epilepsy did not develop, and animals did not die prematurely. When rapamycin was begun after the onset of epilepsy (at 6 weeks), seizure frequency was decreased, interictal EEG was improved, and survival was prolonged.

mTOR Inhibitors in TSC Epilepsy

To date, clinical studies looking at the effects of mTOR inhibitors on the reduction of seizures have mainly focused on patients with a known diagnosis of TSC. In an open-label prospective study, 52 pediatric participants with TSC complicated with epilepsy received rapamycin treatment (1 mg/m2/d) for at least 24 weeks (35). In participants who received rapamycin treatment for 24, 48, 72, and 96 weeks, reported seizure free rates were 25% (13/52), 19% (6/31), 29% (5/17), and 25% (3/12), respectively. Importantly, though rapamycin therapy did not always result in complete seizure freedom, prior to rapamycin therapy, the average frequency of seizures was 70.27 times/day and the average number of antiepileptic drugs was 1.30. After 24, 48, 72, and 96 weeks' treatment, the average seizure frequency was reduced to 1.94–2.80 times/day and the mean number of concomitant antiepileptic drugs were reduced to 0.83–0.97 (35).

Everolimus (Novartis Pharmaceuticals) is a chemically modified rapamycin derivative that is currently approved for the treatment of pediatric and adult patients with TSC who have surgically inaccessible SEGAs. In 2010, Krueger et al. reported the first large study evaluating everolimus in TSC (36). Expanding upon their findings with the phase 3 EXIST-1 trial, followed by further analyses, Franz et al. reported sustained efficacy in reducing the size of the SEGAs, seizure reduction, and reasonable safety and tolerability, though the study was not sufficiently powered to prove a positive effect on epilepsy (3741).

The most relevant iteration of the everolimus trials for TSC was EXIST-3, a double-blind placebo-controlled study evaluating everolimus as an adjunctive therapy for treatment-resistant focal-onset seizures in TSC (42). Participants were assigned to placebo, low-dose everolimus (3–7 ng/mL), or high-dose everolimus (9–15 ng/mL). In the study, the median percentage reduction in seizure frequency was 29% in the low dose group and 40% in the high-dose group, with reasonable safety and tolerability results (42).

Similarly, in a single-center, open prospective study of 15 pediatric participants with TSC and epilepsy, Samueli et al. (43) reported that 80% of participants treated with everolimus responded with seizure reduction. In another study by researchers at Cincinnati Children's Hospital, 78% of children treated with everolimus reported ≥50% reduction in seizure frequency at 2 years (44). Additional smaller reports have described reduction in seizure frequency in children with SEGA treated with everolimus (4547).

mTOR Inhibitors in Non-TSC Epilepsy

A number of structural developmental brain malformations and tumors have been grouped together and named “TORopathies” due to their shared underlying disruptions in the mTOR pathway (48, 49). These include hemimegalencephaly (HME), polyhydramnios, megalencephaly, and symptomatic epilepsy (PMSE) syndrome, gangliogliomas, dysembryonplastic neuroepithelial tumors (DNETs), and focal cortical dysplasias (FCDs). While each of these is a histopathologic diagnosis, a common clinical feature is intractable epilepsy, suggesting that mTOR plays a role in epileptogenesis.

Hemimegalencephaly and focal cortical dysplasias are both structural abnormalities that are the result of malformations during cortical development. Both are highly correlated with medically refractory epilepsy, and have been associated with mutations in mTOR or the PI3K/AKT/mTOR pathway (5065). PMSE is known to be caused by mutations in the STRADα gene, an upstream inhibitor of mTORC1, resulting in a decrease in the LKB1/AMPK pathway, and ultimately dysregulated mTOR signaling (66, 67). Gangliogliomas and DNETs have also been shown to exhibit irregular mTOR signaling (48, 50, 6870). Additionally, variants in genes in the mTOR pathway have been seen in patients with sporadic focal epilepsies (71).

Much of the current evidence for the utility of mTOR inhibitors in the treatment of non-TSC epilepsy is still in the pre-clinical phase. Unlike conventional antiepileptic medications, which tend to modulate neurotransmitter receptors or ion channels, mTOR inhibitors seem to indirectly regulate protein synthesis, which in turn affects ion channels, neuronal signaling, synaptic structure and plasticity (49, 7276). Animal models of limbic epilepsy (27) and absence seizures (30), both discussed previously, have shown that treatment with rapamycin results in decreased seizure activity.

There are some small clinical trials looking at mTOR inhibitors in non-TSC epilepsy. One open-label prospective study enrolled 5 children with PMSE. These children were treated with daily rapamycin (dose adjusted for a target trough blood concentration between 5 and 15 ng/ml). This study reported 4 out of 5 children were seizure free in the preceding 12-month period, with the fifth child having a single seizure during this time (67). Xu et al. (77) reported >50% reduction in seizures in a single patient with hemimegalencephaly after 1 week of treatment with rapamycin. Two ongoing trials are looking at the role of everolimus in the treatment of patients with FCD and medically refractory epilepsy (78, 79).

Discussion

There is a strong suggestion in the preclinical literature of an association between mTOR activity and epilepsy/epileptogenesis, particularly in malformations of cortical development. This, coupled with the clinical success of mTOR inhibitors in seizure control in TSC epilepsy and the growing body of evidence supporting the dysregulation of PI3K/Akt/mTOR in a variety of pathologies encountered in pediatric epilepsy, such as focal cortical dysplasia (FCD), suggests that the next step is to investigate the role of mTOR inhibitors in the treatment of children with medically refractory epilepsy, particularly those children who have also failed resective surgery. To this end, our research group is currently studying ABI-009 (nab-rapamycin) in a prospective, phase 1 safety trial to investigate the safety, tolerability, seizure control, and quality of life in participants with medically refractory epilepsy who failed epilepsy surgery (80). Additionally, there are ongoing trials looking at the role of everolimus in the treatment of patients with FCD and medically refractory epilepsy (78, 79).

While there remains more to learn and understand about the role of the mTOR pathway in epilepsy, as well as the underlying cause of seizures in many children with sporadic epilepsy, current data suggest that intervention along this pathway may lead to a reduction in seizure frequency, if not complete seizure freedom. This is of great value for children with medically and surgically refractory epilepsy, for whom even a 50% reduction in seizures can result in considerable improvements in quality of life and overall cognitive development.

Author Contributions

All authors contributed to the design, writing, critical revision of this review, and approved the final version of the manuscript.

Conflict of Interest

JH is a consultant for Medtronic.

The remaining 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

The authors appreciate the assistance of Sharon Durfy, PhD, with manuscript preparation.

References

1. Camfield CS, Camfield PR, Gordon K, Wirrell E, Dooley JM. Incidence of epilepsy in childhood and adolescence: a population-based study in Nova Scotia from 1977 to 1985. Epilepsia. (1996) 37:19–23. doi: 10.1111/j.1528-1157.1996.tb00506.x

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Olafsson E, Ludvigsson P, Gudmundsson G, Hesdorffer D, Kjartansson O, Hauser WA. Incidence of unprovoked seizures and epilepsy in Iceland and assessment of the epilepsy syndrome classification: a prospective study. Lancet Neurol. (2005) 4:627–34. doi: 10.1016/S1474-4422(05)70172-1

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Berg AT, Shinnar S, Levy SR, Testa FM, Smith-Rapaport S, Beckerman B. Early development of intractable epilepsy in children: a prospective study. Neurology. (2001) 56:1445–52. doi: 10.1212/WNL.56.11.1445

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Berg AT, Shinnar S, Levy SR, Testa FM, Smith-Rapaport S, Beckerman B, et al. Two-year remission and subsequent relapse in children with newly diagnosed epilepsy. Epilepsia. (2001) 42:1553–62. doi: 10.1046/j.1528-1157.2001.21101.x

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Berg AT, Vickrey BG, Testa FM, Levy SR, Shinnar S, DiMario F, et al. How long does it take for epilepsy to become intractable? A prospective investigation. Ann Neurol. (2006) 60:73–9. doi: 10.1002/ana.20852

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Davidson S, Falconer MA. Outcome of surgery in 40 children with temporal-lobe epilepsy. Lancet. (1975) 1:1260–3. doi: 10.1016/S0140-6736(75)92549-0

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Gilliam F, Wyllie E, Kashden J, Faught E, Kotagal P, Bebin M, et al. Epilepsy surgery outcome: comprehensive assessment in children. Neurology. (1997) 48:1368–74. doi: 10.1212/WNL.48.5.1368

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Wyllie E, Comair YG, Kotagal P, Bulacio J, Bingaman W, Ruggieri P. Seizure outcome after epilepsy surgery in children and adolescents. Ann Neurol. (1998) 44:740–8. doi: 10.1002/ana.410440507

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Wiebe S Blume WT Girvin JP Eliasziw M Effectiveness and Efficiency of Surgery for Temporal Lobe Epilepsy Study Group. A randomized, controlled trial of surgery for temporal-lobe epilepsy. N Engl J Med. (2001) 345:311–8. doi: 10.1056/NEJM200108023450501

CrossRef Full Text | Google Scholar

10. Chapell R, Reston J, Snyder D, Treadwell J, Treager S, Turkelson C. Management of treatment-resistant epilepsy. Evid Rep Technol Assess. (2003) 77:1–8.

Google Scholar

11. Sinclair DB, Aronyk KE, Snyder TJ, Wheatley BM, McKean JD, Bhargava R, et al. Pediatric epilepsy surgery at the University of Alberta: 1988-2000. Pediatr Neurol. (2003) 29:302–11. doi: 10.1016/S0887-8994(03)00307-2

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Tellez-Zenteno JF, Dhar R, Wiebe S. Long-term seizure outcomes following epilepsy surgery: a systematic review and meta-analysis. Brain. (2005) 128 (Pt 5):1188–98. doi: 10.1093/brain/awh449

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Terra-Bustamante VC, Inuzuca LM, Fernandes RM, Funayama S, Escorsi-Rosset S, Wichert-Ana L, et al. Temporal lobe epilepsy surgery in children and adolescents: clinical characteristics and post-surgical outcome. Seizure. (2005) 14:274–81. doi: 10.1016/j.seizure.2005.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Gleissner U, Clusmann H, Sassen R, Elger CE, Helmstaedter C. Postsurgical outcome in pediatric patients with epilepsy: a comparison of patients with intellectual disabilities, subaverage intelligence, and average-range intelligence. Epilepsia. (2006) 47:406–14. doi: 10.1111/j.1528-1167.2006.00436.x

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Khan RB, Boop FA, Onar A, Sanford RA. Seizures in children with low-grade tumors: outcome after tumor resection and risk factors for uncontrolled seizures. J Neurosurg. (2006) 104:377–82. doi: 10.3171/ped.2006.104.6.377

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Van Oijen M, De Waal H, Van Rijen PC, Jennekens-Schinkel A, van Huffelen AC, Van Nieuwenhuizen O, et al. Resective epilepsy surgery in childhood: the Dutch experience 1992-2002. Eur J Paediatr Neurol. (2006) 10:114–23. doi: 10.1016/j.ejpn.2006.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Kan P, Van Orman C, Kestle JR. Outcomes after surgery for focal epilepsy in children. Childs Nerv Syst. (2008) 24:587–91. doi: 10.1007/s00381-007-0545-9

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Kim SK, Wang KC, Hwang YS, Kim KJ, Chae JH, Kim IO, et al. Epilepsy surgery in children: outcomes and complications. J Neurosurg Pediatr. (2008) 1:277–83. doi: 10.3171/PED/2008/1/4/277

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Bai X, Jiang Y. Key factors in mTOR regulation. Cell Mol Life Sci. (2010) 67:239–53. doi: 10.1007/s00018-009-0163-7

CrossRef Full Text | Google Scholar

20. Vinals F, Chambard JC, Pouyssegur J. p70 S6 kinase-mediated protein synthesis is a critical step for vascular endothelial cell proliferation. J Biol Chem. (1999) 274:26776–82. doi: 10.1074/jbc.274.38.26776

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Guba M, von Breitenbuch P, Steinbauer M, Koehl G, Flegel S, Hornung M, et al. Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nat Med. (2002) 8:128–35. doi: 10.1038/nm0202-128

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. (2006) 124:471–84. doi: 10.1016/j.cell.2006.01.016

CrossRef Full Text | Google Scholar

23. Mabuchi S, Altomare DA, Connolly DC, Klein-Szanto A, Litwin S, Hoelzle MK, et al. RAD001 (Everolimus) delays tumor onset and progression in a transgenic mouse model of ovarian cancer. Cancer Res. (2007) 67:2408–13. doi: 10.1158/0008-5472.CAN-06-4490

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Hasbani DM, Crino PB. Tuberous sclerosis complex. Handb Clin Neurol. (2018) 148:813–22. doi: 10.1016/B978-0-444-64076-5.00052-1

CrossRef Full Text | Google Scholar

25. Marques R, Belousova E, Benedik MP, Carter T, Cottin V, Curatolo P, et al. Treatment patterns and use of resources in patients with tuberous sclerosis complex: insights from the TOSCA registry. Front Neurol. (2019) 10:1144. doi: 10.3389/fneur.2019.01144

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Saffari A, Brosse I, Wiemer-Kruel A, Wilken B, Kreuzaler P, Hahn A, et al. Safety and efficacy of mTOR inhibitor treatment in patients with tuberous sclerosis complex under 2 years of age - a multicenter retrospective study. Orphanet J Rare Dis. (2019) 14:96. doi: 10.1186/s13023-019-1077-6

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Huang X, Zhang H, Yang J, Wu J, McMahon J, Lin Y, et al. Pharmacological inhibition of the mammalian target of rapamycin pathway suppresses acquired epilepsy. Neurobiol Dis. (2010) 40:193–9. doi: 10.1016/j.nbd.2010.05.024

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Buckmaster PS, Ingram EA, Wen X. Inhibition of the mammalian target of rapamycin signaling pathway suppresses dentate granule cell axon sprouting in a rodent model of temporal lobe epilepsy. J Neurosci. (2009) 29:8259–69. doi: 10.1523/JNEUROSCI.4179-08.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Zeng LH, Rensing NR, Wong M. The mammalian target of rapamycin signaling pathway mediates epileptogenesis in a model of temporal lobe epilepsy. J Neurosci. (2009) 29:6964–72. doi: 10.1523/JNEUROSCI.0066-09.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Russo E, Citraro R, Donato G, Camastra C, Iuliano R, Cuzzocrea S, et al. mTOR inhibition modulates epileptogenesis, seizures and depressive behavior in a genetic rat model of absence epilepsy. Neuropharmacology. (2013) 69:25–36. doi: 10.1016/j.neuropharm.2012.09.019

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Meikle L, Pollizzi K, Egnor A, Kramvis I, Lane H, Sahin M, et al. Response of a neuronal model of tuberous sclerosis to mammalian target of rapamycin (mTOR) inhibitors: effects on mTORC1 and Akt signaling lead to improved survival and function. J Neurosci. (2008) 28:5422–32. doi: 10.1523/JNEUROSCI.0955-08.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Di Nardo A, Kramvis I, Cho N, Sadowski A, Meikle L, Kwiatkowski DJ, et al. Tuberous sclerosis complex activity is required to control neuronal stress responses in an mTOR-dependent manner. J Neurosci. (2009) 29:5926–37. doi: 10.1523/JNEUROSCI.0778-09.2009

CrossRef Full Text | Google Scholar

33. Ljungberg MC, Sunnen CN, Lugo JN, Anderson AE, D'Arcangelo G. Rapamycin suppresses seizures and neuronal hypertrophy in a mouse model of cortical dysplasia. Dis Model Mech. (2009) 2:389–98. doi: 10.1242/dmm.002386

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Zeng LH, Xu L, Gutmann DH, Wong M. Rapamycin prevents epilepsy in a mouse model of tuberous sclerosis complex. Ann Neurol. (2008) 63:444–53. doi: 10.1002/ana.21331

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Zou L, Liu Y, Pang L, Ju J, Shi Z, Zhang J, et al. Efficacy and safety of rapamycin in treatment of children with epilepsy complicated with tuberous sclerosis. Zhonghua Er Ke Za Zhi. (2014) 52:812–6.

PubMed Abstract | Google Scholar

36. Krueger DA, Care MM, Holland K, Agricola K, Tudor C, Mangeshkar P, et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Med. (2010) 363:1801–11. doi: 10.1056/NEJMoa1001671

CrossRef Full Text | Google Scholar

37. Franz DN, Belousova E, Sparagana S, Bebin EM, Frost M, Kuperman R, et al. Everolimus for subependymal giant cell astrocytoma in patients with tuberous sclerosis complex: 2-year open-label extension of the randomised EXIST-1 study. Lancet Oncol. (2014) 15:1513–20. doi: 10.1016/S1470-2045(14)70489-9

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Franz DN, Agricola K, Mays M, Tudor C, Care MM, Holland-Bouley K, et al. Everolimus for subependymal giant cell astrocytoma: 5-year final analysis. Ann Neurol. (2015) 78:929–38. doi: 10.1002/ana.24523

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Franz DN, Belousova E, Sparagana S, Bebin EM, Frost MD, Kuperman R, et al. Long-term use of everolimus in patients with tuberous sclerosis complex: final results from the EXIST-1 study. PLoS ONE. (2016) 11:e0158476. doi: 10.1371/journal.pone.0158476

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Franz DN, Lawson JA, Yapici Z, Brandt C, Kohrman MH, Wong M, et al. Everolimus dosing recommendations for tuberous sclerosis complex-associated refractory seizures. Epilepsia. (2018) 59:1188–97. doi: 10.1111/epi.14085

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Franz DN, Lawson JA, Yapici Z, Ikeda H, Polster T, Nabbout R, et al. Everolimus for treatment-refractory seizures in TSC: extension of a randomized controlled trial. Neurol Clin Pract. (2018) 8:412–20. doi: 10.1212/CPJ.0000000000000514

PubMed Abstract | CrossRef Full Text | Google Scholar

42. French JA, Lawson JA, Yapici Z, Ikeda H, Polster T, Nabbout R, et al. Adjunctive everolimus therapy for treatment-resistant focal-onset seizures associated with tuberous sclerosis (EXIST-3): a phase 3, randomised, double-blind, placebo-controlled study. Lancet. (2016) 388:2153–63. doi: 10.1016/S0140-6736(16)31419-2

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Samueli S, Abraham K, Dressler A, Groppel G, Muhlebner-Fahrngruber A, Scholl T, et al. Efficacy and safety of Everolimus in children with TSC - associated epilepsy - Pilot data from an open single-center prospective study. Orphanet J Rare Dis. (2016) 11:145. doi: 10.1186/s13023-016-0530-z

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Krueger DA, Wilfong AA, Mays M, Talley CM, Agricola K, Tudor C, et al. Long-term treatment of epilepsy with everolimus in tuberous sclerosis. Neurology. (2016) 87:2408–15. doi: 10.1212/WNL.0000000000003400

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Cappellano AM, Senerchia AA, Adolfo F, Paiva PM, Pinho R, Covic A, et al. Successful everolimus therapy for SEGA in pediatric patients with tuberous sclerosis complex. Childs Nerv Syst. (2013) 29:2301–5. doi: 10.1007/s00381-013-2170-0

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Aguilera D, Flamini R, Mazewski C, Schniederjan M, Hayes L, Boydston W, et al. Response of subependymal giant cell astrocytoma with spinal cord metastasis to everolimus. J Pediatr Hematol Oncol. (2014) 36:e448–51. doi: 10.1097/MPH.0000000000000005

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Mingarelli A, Vignoli A, La Briola F, Peron A, Giordano L, Banderali G, et al. Dramatic relapse of seizures after everolimus withdrawal. Eur J Paediatr Neurol. (2018) 22:203–6. doi: 10.1016/j.ejpn.2017.07.018

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Crino PB. mTOR: A pathogenic signaling pathway in developmental brain malformations. Trends Mol Med. (2011) 17:734–42. doi: 10.1016/j.molmed.2011.07.008

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Wong M. A critical review of mTOR inhibitors and epilepsy: from basic science to clinical trials. Expert Rev Neurother. (2013) 13:657–69. doi: 10.1586/ern.13.48

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Ljungberg MC, Bhattacharjee MB, Lu Y, Armstrong DL, Yoshor D, Swann JW, et al. Activation of mammalian target of rapamycin in cytomegalic neurons of human cortical dysplasia. Ann Neurol. (2006) 60:420–9. doi: 10.1002/ana.20949

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Lee JH, Huynh M, Silhavy JL, Kim S, Dixon-Salazar T, Heiberg A, et al. De novo somatic mutations in components of the PI3K-AKT3-mTOR pathway cause hemimegalencephaly. Nat Genet. (2012) 44:941–5. doi: 10.1038/ng.2329

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Aronica E, Crino PB. Epilepsy related to developmental tumors and malformations of cortical development. Neurotherapeutics. (2014) 11:251–68. doi: 10.1007/s13311-013-0251-0

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Liu J, Reeves C, Michalak Z, Coppola A, Diehl B, Sisodiya SM, et al. Evidence for mTOR pathway activation in a spectrum of epilepsy-associated pathologies. Acta Neuropathol Commun. (2014) 2:71. doi: 10.1186/2051-5960-2-71

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Crino PB. mTOR signaling in epilepsy: insights from malformations of cortical development. Cold Spring Harb Perspect Med. (2015) 5:a022442. doi: 10.1101/cshperspect.a022442

PubMed Abstract | CrossRef Full Text | Google Scholar

55. D'Gama AM, Geng Y, Couto JA, Martin B, Boyle EA, LaCoursiere CM, et al. Mammalian target of rapamycin pathway mutations cause hemimegalencephaly and focal cortical dysplasia. Ann Neurol. (2015) 77:720–5. doi: 10.1002/ana.24357

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Jansen LA, Mirzaa GM, Ishak GE, O'Roak BJ, Hiatt JB, Roden WH, et al. PI3K/AKT pathway mutations cause a spectrum of brain malformations from megalencephaly to focal cortical dysplasia. Brain. (2015) 138 (Pt 6):1613–28. doi: 10.1093/brain/awv045

CrossRef Full Text | Google Scholar

57. Leventer RJ, Scerri T, Marsh AP, Pope K, Gillies G, Maixner W, et al. Hemispheric cortical dysplasia secondary to a mosaic somatic mutation in MTOR. Neurology. (2015) 84:2029–32. doi: 10.1212/WNL.0000000000001594

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Lim JS, Kim WI, Kang HC, Kim SH, Park AH, Park EK, et al. Brain somatic mutations in MTOR cause focal cortical dysplasia type II leading to intractable epilepsy. Nat Med. (2015) 21:395–400. doi: 10.1038/nm.3824

CrossRef Full Text | Google Scholar

59. Nakashima M, Saitsu H, Takei N, Tohyama J, Kato M, Kitaura H, et al. Somatic mutations in the MTOR gene cause focal cortical dysplasia type IIb. Ann Neurol. (2015) 78:375–86. doi: 10.1002/ana.24444

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Mirzaa GM, Campbell CD, Solovieff N, Goold C, Jansen LA, Menon S, et al. Association of MTOR mutations with developmental brain disorders, including megalencephaly, focal cortical dysplasia, and pigmentary mosaicism. JAMA Neurol. (2016) 73:836–45. doi: 10.1001/jamaneurol.2016.0363

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Moller RS, Weckhuysen S, Chipaux M, Marsan E, Taly V, Bebin EM, et al. Germline and somatic mutations in the MTOR gene in focal cortical dysplasia and epilepsy. Neurol Genet. (2016) 2:e118. doi: 10.1212/NXG.0000000000000118

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Alcantara D, Timms AE, Gripp K, Baker L, Park K, Collins S, et al. Mutations of AKT3 are associated with a wide spectrum of developmental disorders including extreme megalencephaly. Brain. (2017) 140:2610–22. doi: 10.1093/brain/awx203

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Baldassari S, Ribierre T, Marsan E, Adle-Biassette H, Ferrand-Sorbets S, Bulteau C, et al. Dissecting the genetic basis of focal cortical dysplasia: a large cohort study. Acta Neuropathol. (2019) 138:885–900. doi: 10.1007/s00401-019-02061-5

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Dobyns WB, Mirzaa GM. Megalencephaly syndromes associated with mutations of core components of the PI3K-AKT-MTOR pathway: PIK3CA, PIK3R2, AKT3, and MTOR. Am J Med Genet C Semin Med Genet. (2019) 181:582–90. doi: 10.1002/ajmg.c.31736

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Garcia CAB, Carvalho SCS, Yang X, Ball LL, George RD, James KN, et al. mTOR pathway somatic variants and the molecular pathogenesis of hemimegalencephaly. Epilepsia Open. (2020) 5:97–106. doi: 10.1002/epi4.12377

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Orlova KA, Parker WE, Heuer GG, Tsai V, Yoon J, Baybis M, et al. STRADalpha deficiency results in aberrant mTORC1 signaling during corticogenesis in humans and mice. J Clin Invest. (2010) 120:1591–602. doi: 10.1172/JCI41592

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Parker WE, Orlova KA, Parker WH, Birnbaum JF, Krymskaya VP, Goncharov DA, et al. Rapamycin prevents seizures after depletion of STRADA in a rare neurodevelopmental disorder. Sci Transl Med. (2013) 5:182ra153. doi: 10.1126/scitranslmed.3005271

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Samadani U, Judkins AR, Akpalu A, Aronica E, Crino PB. Differential cellular gene expression in ganglioglioma. Epilepsia. (2007) 48:646–53. doi: 10.1111/j.1528-1167.2007.00925.x

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Boer K, Troost D, Timmermans W, van Rijen PC, Spliet WG, Aronica E. Pi3K-mTOR signaling and AMOG expression in epilepsy-associated glioneuronal tumors. Brain Pathol. (2010) 20:234–44. doi: 10.1111/j.1750-3639.2009.00268.x

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Citraro R, Leo A, Constanti A, Russo E, De Sarro G. mTOR pathway inhibition as a new therapeutic strategy in epilepsy and epileptogenesis. Pharmacol Res. (2016) 107:333–43. doi: 10.1016/j.phrs.2016.03.039

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Pippucci T, Licchetta L, Baldassari S, Marconi C, De Luise M, Myers C, et al. Contribution of ultrarare variants in mTOR pathway genes to sporadic focal epilepsies. Ann Clin Transl Neurol. (2019) 6:475–85. doi: 10.1002/acn3.722

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Tang SJ, Reis G, Kang H, Gingras AC, Sonenberg N, Schuman EM. A rapamycin-sensitive signaling pathway contributes to long-term synaptic plasticity in the hippocampus. Proc Natl Acad Sci USA. (2002) 99:467–72. doi: 10.1073/pnas.012605299

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Jaworski J, Spangler S, Seeburg DP, Hoogenraad CC, Sheng M. Control of dendritic arborization by the phosphoinositide-3'-kinase-Akt-mammalian target of rapamycin pathway. J Neurosci. (2005) 25:11300–12. doi: 10.1523/JNEUROSCI.2270-05.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Kumar V, Zhang MX, Swank MW, Kunz J, Wu GY. Regulation of dendritic morphogenesis by Ras-PI3K-Akt-mTOR and Ras-MAPK signaling pathways. J Neurosci. (2005) 25:11288–99. doi: 10.1523/JNEUROSCI.2284-05.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Raab-Graham KF, Haddick PC, Jan YN, Jan LY. Activity- and mTOR-dependent suppression of Kv1.1 channel mRNA translation in dendrites. Science. (2006) 314:144–8. doi: 10.1126/science.1131693

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Wang Y, Barbaro MF, Baraban SC. A role for the mTOR pathway in surface expression of AMPA receptors. Neurosci Lett. (2006) 401:35–9. doi: 10.1016/j.neulet.2006.03.011

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Xu Q, Uliel-Sibony S, Dunham C, Sarnat H, Flores-Sarnat L, Brunga L, et al. mTOR Inhibitors as a new therapeutic strategy in treatment resistant epilepsy in hemimegalencephaly: a case report. J Child Neurol. (2019) 34:132–8. doi: 10.1177/0883073818813238

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Devinsky O. A Pilot Study to Evaluate the Effects of Everolimus on Brain mTOR Activity and Cortical Hyperexcitability in TSC and FCD. New York, NY: NYU Lagone Health (2019).

Google Scholar

79. Kim HD. A Study Investigating the Anti-epileptic Efficacy of Afinitor (Everolimus) in Patients with Refractory Seizures who have Focal Cortical Dysplasia Type II (FCD II). Seoul: Yonsei University Healthcare System (2019).

Google Scholar

80. Hauptman J. ABI-009 (Nab-rapamycin) for Surgically-Refractory Epilepsy (RaSuRE). Seattle, WA: Seattle Children's Hospital (2019).

Google Scholar

Keywords: mTOR, cortical dysplasia, hemimegalencephaly, pediatric epilepsy, non-tuberous sclerosis complex-related epilepsy

Citation: Goldstein HE and Hauptman JS (2021) The Putative Role of mTOR Inhibitors in Non-tuberous Sclerosis Complex-Related Epilepsy. Front. Neurol. 12:639319. doi: 10.3389/fneur.2021.639319

Received: 09 December 2020; Accepted: 25 January 2021;
Published: 12 February 2021.

Edited by:

Stefano Seri, Aston University, United Kingdom

Reviewed by:

Sergiusz Jozwiak, Medical University of Warsaw, Poland
Shakti Kumar Agrawal, Birmingham Women's and Children's Hospital, United Kingdom

Copyright © 2021 Goldstein and Hauptman. 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) and the copyright owner(s) 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: Jason S. Hauptman, Jason.hauptman@seattlechildrens.org; respub2@uw.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.