You are on page 1of 10

European Journal of Paediatric Neurology 47 (2023) 25–34

Contents lists available at ScienceDirect

European Journal of Paediatric Neurology


journal homepage: www.journals.elsevier.com/european-journal-of-paediatric-neurology

Updated clinical recommendations for the management of tuberous


sclerosis complex associated epilepsy
Nicola Specchio a, *, Rima Nabbout b, Eleonora Aronica c, d, Stephane Auvin e, f, g,
Arianna Benvenuto h, Luca de Palma a, Martha Feucht i, 1, Floor Jansen j, 2, Katarzyna Kotulska k, 3,
Harvey Sarnat l, Lieven Lagae m, Sergiusz Jozwiak n, 3, Paolo Curatolo o
a
Clinical and Experimental Neurology, Bambino Gesu’ Children’s Hospital IRCCS, Full Member of European Reference Network on Rare and Complex Epilepsies
EpiCARE, Rome, Italy
b
Department of Pediatric Neurology, Necker Enfants Malades Hospital, Université Paris Cité, Member of the European Reference Network on Rare and Complex
Epilepsies EpiCARE, INSERM U1163, Institut Imagine, Paris, France
c
Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Department of (Neuro)Pathology, Amsterdam, Netherlands
d
Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
e
APHP, Service de Neurologie Pédiatrique, Centre Epilepsies Rares, Member of the European Reference Network on Rare and Complex Epilepsies EpiCARE, Hôpital
Robert Debré, Paris, France
f
Université Paris-Cité, INSERM NeuroDiderot, Paris, France
g
Institut Universitaire de France (IUF), Paris, France
h
Libera Università Maria Santissima Assunta (LUMSA), Rome, Italy
i
Epilepsy Center, Department of Pediatrics, Medical University Vienna, Austria
j
Department of Pediatric Neurology, Brain Center UMC Utrecht, the Netherlands
k
Department of Neurology and Epileptology, The Children’s Memorial Health Institute, Warsaw, Poland
l
Department of Paediatrics (Neurology), Pathology and Laboratory Medicine (Neuropathology) and Clinical Neurosciences, University of Calgary Cumming School of
Medicine and Alberta Children’s Hospital Research Institute (Owerko Centre), Calgary, AB, Canada
m
Department of Paediatric Neurology, University of Leuven, Leuven, Belgium
n
Research Department, The Children’s Memorial Health Institute, ERN EPICARE, Warsaw, Poland
o
Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University, Rome, Italy

A R T I C L E I N F O A B S T R A C T

Keywords: Children with tuberous sclerosis complex (TSC), may experience a variety of seizure types in the first year of life,
Everolimus most often focal seizure sand epileptic spasms. Drug resistance is seen early in many patients, and the man­
Epileptogenesis agement of TSC associated epilepsy remain a major challenge for clinicians. In 2018 clinical recommendations
Developmental and epileptic encephalopathies
for the management of TSC associated epilepsy were published by a panel of European experts. In the last five
Neurodevelopmental disorders
Pre-symptomatic treatment
years considerable progress has been made in understanding the neurobiology of epileptogenesis and three
Drug resistant epilepsy interventional randomized controlled trials have changed the therapeutic approach for the management of TSC
associated epilepsy. Pre-symptomatic treatment with vigabatrin may delay seizure onset, may reduce seizure
severity and reduce the risk of epileptic encephalopathy. The efficacy of mTOR inhibition with adjunctive
everolimus was documented in patients with TSC associated refractory seizures and cannabidiol could be another
therapeutic option. Epilepsy surgery has significantly improved seizure outcome in selected patients and should
be considered early in all patients with drug resistant epilepsy.
There is a need to identify patients who may have a higher risk of developing epilepsy and autism spectrum
disorder (ASD).
In the recent years significant progress has been made owing to the early identification of risk factors for the
development of drug-resistant epilepsy.
Better understanding of the mechanism underlying epileptogenesis may improve the management for TSC-
related epilepsy. Developmental neurobiology and neuropathology give opportunities for the implementation

* Corresponding author. Clinical and Experimental Neurology, Bambino Gesu’ Children’s Hospital, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy.
E-mail address: nicola.specchio@opbg.net (N. Specchio).
1
Full Member of European Reference Network EpiCARE.
2
Full member of the European Reference Network on Rare and Complex Epilepsies EpiCARE.
3
Member of ERN EPICARE.

https://doi.org/10.1016/j.ejpn.2023.08.005
Received 8 March 2023; Received in revised form 27 August 2023; Accepted 28 August 2023
Available online 30 August 2023
1090-3798/© 2023 The Authors. Published by Elsevier Ltd on behalf of European Paediatric Neurology Society. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
N. Specchio et al. European Journal of Paediatric Neurology 47 (2023) 25–34

of concepts related to clinical findings, and an early genetic diagnosis and use of EEG and MRI biomarkers may
improve the development of pre-symptomatic and disease-modifying strategies.

1. Introduction meeting. The panel addressed the mechanisms of epileptogenesis, the


predictive value of clinical EEG, brain MRI and genetic biomarkers, and
TSC is a multisystem genetic disorder characterized by age-related evaluated the potential benefits of pre-symptomatic treatment with
development of benign tumors across several organs including the vigabatrin and targeted treatment with Everolimus, and other new
brain, skin, kidneys, heart, and eyes. TSC is caused by inactivating treatment possibilities, including CBD and indications for presurgical
mutations in either TSC-1 or TSC-2 genes [1–3]. The gene mutation evaluation.
results in an overactivation of the PI3K-Akt-mechanistic (formerly This paper outlines the issues discussed at the consensus meeting
mammalian) target of the rapamycin (mTOR) signaling pathway, which workshop and sets out updated clinical recommendations for current
plays a crucial role from brain development processes to aging [4]. treatment of TSC-associated epilepsy, considering new concepts and
Overall, more than 2000 pathogenic TSC1/2 variants have been changes since the last publication of the 2018 clinical recommendations.
described in the Leiden Open Variation Database (www.lovd.nl/TSC1
and www.lovd.nl/TSC2), making the genotype – phenotype correla­ 2. Progress in understanding mTOR dependent epileptogenesis
tion challenging. Identification of a pathogenic variant or deletion in and co-occurring neurodevelopmental disorders
TSC1/2 is sufficient for the diagnosis of TSC, regardless of clinical
findings (Northrup 2021) [5]. However, about 5–8% of TSC individuals TSC is genetically complex because it involves both germline genes
meeting definite clinical diagnostic criteria have no pathogenic TSC and somatic second-hit events. Second-hit loss of either TSC1 or TSC2,
gene variant or deletion identified by direct sequencing of both genes or inducing mTOR hyperactivation, has been identified in TSC neoplastic
by whole exome sequencing analysis [5–8]. lesions, but is less frequently detected in cortical tubers, suggesting a
Epilepsy is the most common (80–90% of patients) [9] of a large role for additional molecular mechanisms [23,24].
spectrum of neurological and psychiatric manifestation in patients with Extensive research over the past decade has provided strong evi­
TSC. In addition neurodevelopmental disorders with intellectual dence of the critical role of the mTOR pathway during brain develop­
disability and autism spectrum disorder (ASD), and behavioral abnor­ ment, controlling many basic cellular functions, such as energy
malities are highly prevalent [10,11]. metabolism, protein synthesis and autophagy, to regulate cell growth,
Patients with TSC may experience a variety of types of seizures: proliferation, migration, dendritic formation and axon elongation [25,
epileptic spasms, focal, tonic, atonic, and tonic-clonic seizures in the 26]. The mTOR signaling pathway has a crucial function in the process
context of an evolution toward Lennox-Gastaut syndrome. The majority of neuronal and astrocytes differentiation, as well as in oligodendrocyte
of patients experience seizures in the first year of life [12,13]. About 5% lineage development. Balanced spatiotemporal mTOR signaling is
of children develop epilepsy as newborns, often due to malformations of crucial for the proper development of the human cortex [25,27,28].
cortical development meeting the criteria of focal cortical dysplasia Accordingly dysregulation of the mTOR signaling represents a common
(FCD) [14]. The recognition of subtle focal seizures in infants by care­ pathogenic mechanism of a spectrum of malformations of cortical
givers may be difficult [15]. development, sharing histopathological and clinical features, including
In 2017, a panel of European specialists met in Rome to define epilepsy and co-occurring neurodevelopmental disorders [25,29].
clinical recommendations for the management of epilepsy associated Timing of mTOR activation plays a crucial role, and subsequently de­
with TSC [16]. In the last five years, considerable progress has been termines the neuropathological and clinical phenotype [29–31]. Inter­
made in understanding various aspects of this disorder, including the estingly, in experimental models, the level of mTOR-hyperactivity
neurobiology of (mTOR dependent) epileptogenesis. correlates with the severity of epilepsy and associated neuropathology
First of all the International ‘TuberOus SClerosis registry to increase [32].
disease Awareness’ (TOSCA) [17] has provided baseline data on more Cortical tubers, subependymal nodules and subependymal giant cell
than 2000 patients and new insights into the clinical characteristics and astrocytoma are the main pathologies in the brain of TSC patients.
the natural history of TSC. Moreover, three international randomized Cortical tubers represent the neuropathological basis of epilepsy and
controlled trials (RCTs) have changed clinical practices in the treatment consist of areas of cortical dyslamination containing different cell types,
of epilepsy associated with TSC. The EPISTOP (NCT02098759) trial including dysmorphic neurons, giant cells and reactive astrocytes [10].
showed that pre-symptomatic treatment with vigabatrin may modify the Cortical tuber formation is a complex and dynamic process starting in
natural history of seizures with a delay in seizure onset and a reduction fetal brain but continuing as the brain matures [10,33]. Visual detection
in seizure severity and the risk of epileptic encephalopathy (EE) [18]. of cortical tubers on neuroimaging is limited in pre-natal and early
The EXIST3 trial demonstrated the efficacy of mTOR inhibition with post-natal period, and it improves with the myelination process during
adjunctive Everolimus in reducing seizure frequency in patients with the first three years of life [34]. Prenatal brain MRI revealed cortical
TSC-associated refractory seizures [19]. Finally, the GWPCARE6 trial tubers in 20 out of 30 (66.7%) patients in the study of Slowinska et al.
documented efficacy and safety of Cannabidiol (CBD) in reducing [35]. Predefined advanced imaging protocols with 3D T1 and T2 se­
TSC-associated seizures compared with placebo [20]. CBD is now quences are required to improve lesion detection in the new-born with
approved by Food and Drug Administration (FDA) and European Med­ TSC. Some MRI sequences as arterial spin labelling (ASL) have been
icine Agency (EMA) for the treatment of seizures in patients with TSC. reported of help in early childhood to identify tubers in relation to EEG
Experts are now moving away from the traditional therapeutic slow waves [36], but this application needs further validation.
approach with antiseizure medication (ASM) applied after seizure onset, Insights from a variety of in vivo and in vitro genetic models and from
to a more proactive approach focused on pre-symptomatic treatment human pathology have shown that epileptogenesis in TSC is a chronic
with vigabatrin or molecular targeted treatment with mTOR inhibition and dynamic process leading to early onset and difficult to treat seizures,
[21]. Advances in surgery improves access to this treatment for patients co-occurring neurodevelopmental disorders and mTOR-related neuro­
with multiple tubers [22]. degeneration (i.e. increases risk for tauopathy) [10,37]. This process
To review new insights from the last years, a new panel of experts may start prenatally and can continue long after the epilepsy diagnosis.
met in Rome in November 2022 to address specific questions. Each panel Knowledge about the complex biological mechanisms contributing to
expert was assigned a topic to review beforehand and present at the network dysfunction during brain development, even beyond the early

26
N. Specchio et al. European Journal of Paediatric Neurology 47 (2023) 25–34

period of epileptogenesis, in individuals with TSC, is rapidly increasing Table 1


[10,38]. Experimental and human studies provide evidence of early and Predictors of drug resistance.
sustained oxidative stress and inflammation, accompanied by blood Predictors of drug resistance
brain barrier (BBB) dysfunction and extracellular matrix remodelling
Clinical
[10,38–40]. seizures
Dysregulation of mTOR signaling itself may result in developmental - ES + Nabbout R et al., 2019 [17], Jeong A
alterations of the balance between excitation and inhibition. Converging et al., 2017 [57]
evidence suggest that the pathological network underlying early - Focal seizures + ES +++ Nabbout R et al., 2019 [17], Jeong A
et al., 2017 [57]
appearance of seizures is characterized by persistence of immature - Younger age at onset of +++ Capal JK et al., 2017 [49]
patterns of neurotransmitter receptor expression and function (i.e. clinical seizures
GABAA-receptor [41]) and/or disrupted GABAergic interneuron EEG
network development and organization [10,38,42]. Moreover, the Interictal abnormalities
- Focal De Ridder J et al., 2020 [65]
direct influence of mTOR pathway dysregulation on oligodendrocyte +++
- Multi-focal +++ De Ridder J et al., 2020 [65], De Ridder
differentiation and myelin formation could further contribute to J et al., 2021 [62]
abnormal cell-signaling and network dysfunction [10,38,43]. Other - Younger age at onset of +++ De Ridder J et al., 2021 [62], Wu JY
pathways, that might be related to the mTOR pathway, have also been recording of IEDs et al., 2019 [59]
suggested as interesting targets in therapy of TSC-related seizures during Genetic
- TSC 2 gene variant Ogórek B et al., 2020 [60]
brain development such as the inflammatory pathway (IL-1-CXCL10)
+++

[44] and the GluN2C subunit of the NDMA receptor [45,46]. Combi­
nation of multiple targeted interventions might be needed to further MRI
increase the efficacy of mTOR inhibitor treatment in TSC-related sei­ - Higher tuber load +++ Hulshof HM et al., 2022 [66]
zures in humans. - Tuber location + Cohen AL et al., 2021 [67]
There is a complex relationship between epilepsy and autism, and ES: epileptic spasms, IEDs: interictal epileptiform discharges, R: right.
TSC is among the major causes of syndromic ASD [11,47]. TSC2 muta­
tions, tuber volumes and location, infantile spasms and early onset sei­ cannot confirm the predictive value of IEDs. The EPISTOP study has
zures have been reported as the most important risk factors for ASD recently identified useful pre-symptomatic genetic, EEG and MRI bio­
[48]. A developmental disruption of GABAergic cortical interneurons markers that can be used in clinical practice to identify infants at risk for
linked to abnormal generation, migration routes as well as their mo­ developing DRE at the age of two years [18,60–63].
lecular and functional diversifications can explain the concomitant TSC2 pathogenic variants were associated with a higher tuber load
occurrence of epilepsy and autism [10,38,42]. Although there is a strong on MRI and were predictive of more severe epilepsies with higher risk of
association between the presence of epilepsy and TSC-associated ASD, drug resistance [64]. Interictal focal or multifocal discharges on serial
ASD should not be considered solely as a co-morbidity of early onset sleep-wake EEG in pre-symptomatic infants with TSC are predictive of
epilepsy and the infantile epileptic spasms syndrome (IESS). an ongoing severe epilepsy [59]. Ninety-five per cent of infants with TSC
Ever since the last century, clinicians have always observed that enrolled in the EPISTOP trial developed IED at a medium age of 77 days,
seizures appear in the first months of life, while ASD risk is recognized and earlier recording of IED was associated with an increased risk of
much later and is typically not diagnosed before 2–3 years of age. DRE [62,65]. Although fetal MRI brain lesions did not predict the risk of
Therefore, their understanding was that early epilepsy onset may lead to clinical seizures or DRE at the age of two years [34], higher fetal lesion
autism even if there is morphological substrate of autism (brain lesions) load, was significantly associated with lower developmental outcome.
and some autism features are seen in patients without epilepsy [11,47]. Furthermore, higher tuber volume as a proportion of total brain volume
Recent evidence showed that early risk of autism can be detected on early postnatal MRI was associated with clinical seizures and DRE at
already in the first year of life [49,50]. Complex cellular and molecular the age of two years [66].
events play a significant role in determining a high risk for both epilepsy In a prospective observational cohort of 123 infants, tuber location
and neurodevelopmental deficits in infants and children with TSC was heterogeneous, however tubers functionally connected to the globi
[51–53]. TSC can be considered a model of genetic-determined devel­ pallidi and cerebellar vermis were stronger predictor of spasms [67].
opmental and epileptic encephalopathy, a group of severe epilepsies that Increased EEG connectivity was strongly associated with epileptic
are characterized by - often drug resistant - seizures and encephalopathy spasms in another cohort [68].
related to the dual impact of the seizures and of the etiology [54,55]. In summary early recognition of pathogenic variants and the use of
reliable EEG and MRI biomarkers could help to identify pathogenic
3. Predictors of drug resistance mechanisms for epileptogenesis and provide opportunities for pre-
symptomatic and disease modifying approaches. Clinical, EEG, MRI
TSC-associated epilepsy management is still a challenge because of and genetic predictors of DRE are summarized in Table 1. Development
high percentage of patients developing drug resistant epilepsy (DRE). of a prediction model that incorporates a variety of risk factors, as well
Data from the TOSCA registry revealed that patients presenting with as machine learning models with multimodality features could aid in
focal seizures (whether or not combined with IESS) were more re­ guidance and treatment of TSC patients with high risk of DRE or neu­
fractory than patients presenting with epileptic spasms only, but the rodevelopmental disorders [69,70].
prediction of DRE is still challenging [17].
In a prospective study, infants with an early seizure onset continued 4. Pre-symptomatic treatment
to perform worse at 24 months of age, in comparison with children with
a later seizure onset [56]. Data from the TSC Natural History Database, Pre-symptomatic treatment with VGB was already suggested by the
including 1965 subjects with TSC, showed that early focal-onset seizures previous expert panel in 2018, but its efficacy is now strongly confirmed
and treatment-resistant infantile spasms were clinical predictors for DRE by the EPISTOP trial [18]. The EPISTOP trial was carried out from 2014
[57]. Infantile spasms were also among strong indicators of DRE as re­ to 2018 in 9 European centers and one Australian site. The aim of the
ported in a systematic TSC review [58]. In a longitudinal observational trial was to evaluate the safety and efficacy of conventional versus
study of 40 infants interictal epileptiform discharges (IEDs) correctly “preventive” treatment of epilepsy with vigabatrin at the dosage of at
identified future epilepsy in 77% of seizure naïve infants [59]. However, least 100 mg/kg/day. Eligible patients were newborns and infants under
due to the high prevalence of epilepsy in TSC patients (80–90%) we

27
N. Specchio et al. European Journal of Paediatric Neurology 47 (2023) 25–34

Fig. 1. This is a 10-month-old boy with a TSC2 genetic variant. The diagnosis was suspected prenatally due to the presence of cardiac rhabdomyomas and confirmed
at 1 months of age, when brain MRI showed several tubers. At the age of 3 months, EEG monitoring was started (A) showing during sleep a normal background
activity with rare slow waves over the right temporal region. (B) 2 months later (age 5 months) spike and spikes and wave complexes appeared during sleep over the
left temporal region. Pre-symptomatic treatment with vigabatrin at the dose of 75 mg/kg day was started. (C) Another 2 and 4 months later (age 7 and 9 months
respectively) sleep EEG was and remained (D) normal. He did not experience clinical seizures, and the development is normal.

Table 2
Ongoing trials on prevention of epilepsy in TSC patients.
Ongoing trials

Trial Phase Intervention Study Population Primary Outcome(s) Completion

PREVeNT IIb Vigabatrin vs Placebo Infants with TSC aged 1 day to N= Cognitive assessment scores at 24 months May 2023
6 months 84
TSC- II Sirolimus vs Placebo N= % Reporting severe or serious AEs; time to seizure onset June 2026
STEPS 64 at 12 months of age
ViRap II/III Sirolimus vs Vigabatrin (masked Infants with TSC aged 4–16 N= Occurrence of clinical seizures; volume of TSC-associated Mar 2026
with placebo) weeks 60 tumors at 730 days

the age of 4 months, before the onset of seizures. All participants were seizure, or immediately after first clinical seizure) treatment resulted in
monitored with serial EEG and vigabatrin was introduced either when better neurodevelopmental outcome than in any other historical cohort
IED were detected or after the onset of seizures. Preventive treatment of TSC patients (median IQ within the normal range in the whole
with vigabatrin in 25 children resulted in delay of clinical seizures, cohort), however, longer follow up is warranted before firm conclusions
reduced risk of developing drug resistant epilepsy, and absence of in­ can be drawn with respect to neurodevelopmental outcomes [18].
fantile spasms, when compared with 25 patients taking treatment after Autistic features were detected irrespectively of treatment strategy,
seizure onset [18] (Fig. 1). suggesting either independent mechanisms or the impact of earlier
In the EPISTOP study treatment with vigabatrin in TSC infants was epileptogenesis-related events since intrauterine life [50,53].
safe and modified the natural history of seizures, reducing the risk and Ongoing RCTs, with the aim to prevent epilepsy, are summarized in
severity of epilepsy. Since epilepsy was not prevented in the majority of Table 2.
infants, the term pre-symptomatic treatment is to be preferred and is
now considered an established approach in several European country in 5. Targeted treatment with mTOR inhibitors
infants with TSC [71]. Supporting this more proactive use of Vigabatrin
for TSC-epilepsy, new data published from the US Vigabatrin registry mTOR inhibition can reduce structural abnormalities in the brain:
that followed >9000 patients on Vigabatrin for up to 16 years did not mTOR inhibition reduces neuronal morphology abnormalities, cell size
find that any patient experienced clinical vision loss, few (2%) patients and inflammatory mediators in the cortex, and improves neuronal
experienced new vision findings over time and none were determined to myelination and synaptic plasticity.
be caused by Vigabatrin. Thus, the risk to vision appears lower than Initially developed as anti-fungal agents and later discovered to have
previously thought [72]. Furthermore, early and frequent EEG moni­ potent immunosuppressive and antiproliferative properties, rapamycin
toring and early (before any type of seizures, after electrographic (Sirolimus) has been found to be a mechanistic target of mTOR

28
N. Specchio et al. European Journal of Paediatric Neurology 47 (2023) 25–34

overactivation [73]. Rapamycin is a macrolide compound, isolated for What is still not known is the optimal timing for mTOR inhibition,
the first time in 1972, from samples of Streptomyces hygroscopicus and its applicability in fetal life. Everolimus was used in a pregnant
found on Easter Island. A mechanistic target of rapamycin kinase woman to treat her fetus with TSC and regression and subsequent sta­
(mTOR) inhibitor acts on T and B cells by reducing their sensitivity to bilization of the cardiac and brain lesions were seen; postnatally, the
interleukin-2 (IL-2). The compound was approved by the FDA in 1999 as child did not develop seizures or autism and presented good neuro­
immunosuppressant indicated for the prophylaxis of organ rejection in developmental outcome [88,89].
patients aged ≥13 years receiving renal transplants and patients with Since mTOR inhibitors may have effects on various systemic mani­
lymphangioleiomyomatosis. Rapamycin/Sirolimus changes microglial festation of TSC, the panel suggest that everolimus should be considered
polarity in TCS mice [74]. It has also been shown to decrease seizures early when growing subependymal giant cell astrocytoma (SEGA) or
and neuropsychiatric symptoms, except for ASD, (and this effect seems angiomyolipoma (AML) are seen in combination with DRE [90]. Sup­
to be related to the regulation of microglia polarity) in TSC mouse porting evidence for the efficacy of combination treatment with siroli­
models [75–77]. Recent uncontrolled studies demonstrated a favorable mus and everolimus in treating the same/different TSC manifestations
safety and efficacy profile of sirolimus for treating manifestations of TSC should be available, as there are currently no head-to-head published
(including epilepsy); the study reported on efficacy and safety of rapa­ studies. It is important to demonstrate if dual targeting of TORC1/­
mycin in DRE in 32 children aged 11 months to 14 years, at least 50% TORC2 provides superior efficacy, above sirolimus or everolimus as
reduction of seizures was seen in 56% of patients [78]. However, monotherapy, without significant increases in toxicity. In addition, and
long-term efficacy/safety data from prospective controlled studies in in contrast to oncology, sirolimus in TSC patients is used in monotherapy
TSC are only available for everolimus. in young patients and therefore might have fewer side effects.
Everolimus (the 40-O-(2-hydroxyethyl) derivative of Sirolimus), is a
second-generation rapamycin derivative and works similarly to siroli­ 6. Surgery: relevance and limitations
mus as an inhibitor of mTOR pathway.
Although having a similar structure everolimus significant differs In 2017’s workshop, the panel was already suggesting that surgery
regarding its pharmacokinetic, pharmacodynamic, and toxic properties can be a good therapeutic option for TSC in a well selected population of
resulting in distinct clinical profile. Everolimus is more readily absor­ patients with recommendation of early presurgical evaluation to
bed, exhibits a greater oral bioavailability and is a selective inhibitor of attempt to improve neurodevelopmental outcome as well. New evidence
mTORC1 with little impact on mTORC2. Therefore, it could achieve emphasizes and strengthens this recommendation. Overall post-
many of the benefits of sirolimus on cell growth and proliferation operative seizure freedom is achieved in 50–60% of TSC patients, and
without glucose intolerance [79]. >50% seizure reduction is seen in another 15%, even in patients with
There are still ongoing efforts to limit the non-neurologic effects of multiple tubers. In selected series it has been observed that duration of
mTOR inhibitors. Indeed, experimental study showed that brain-specific follow up matters as there is a substantial decrease of seizure-free pa­
analogs of rapamycin (termed rapalogs) have recently been developed by tients with a longer follow-up [91].
inhibiting brain mTOR while sparing mTOR activity in other tissues Epilepsy surgery in TSC is still underused [92]. Indication have long
using the brain-permeable mTOR inhibitor RapaLink-1 and the brain- been limited to clear unifocal epilepsy, but the great majority of patients
impermeable FKBP12 ligand RapaBlock [80]. have multifocal EEG abnormalities and numerous tubers. Thus, indica­
Everolimus, has been considered as an add-on treatment option for tion may need to be widened taking the high burden of predominant
individuals with TSC associated drug resistant epilepsy. Treatment ef­ seizures into account, especially when semiology is consistent over time.
ficacy and safety were reported in the EXIST-3 trial where adjunctive A recent nationwide multicenter retrospective study in 364 patients
everolimus achieved a clinically meaningful reduction of seizure fre­ who underwent surgery at the median age of 8.5 years, reports sustained
quency with a favorable risk-benefit ratio [19,81]. Furthermore, seizure freedom in 51% at 10 years, particularly after complete removal
response to everolimus tends to improve with ongoing treatment and the of a large lesion on MRI or after tuberectomy plus cortectomy, meaning
post authorization safety study (PASS) confirmed the long-term safety of the epileptogenic zone beyond tuber borders [93]. A systematic review
everolimus in the real-world clinical practice, and a better response for of 46 studies regarding the outcome of 1157 patients showed an excel­
the <6 years subgroup was documented [82,83]. The target drug con­ lent seizure reduction in 59% of the individuals who underwent epilepsy
centration of 5–9 ng/ml was recommended [81]. surgery [91]. Very early onset epilepsy, the absence of a tuber which is
Everolimus has been also studied as the treatment of TSC-associated expected to predominate in generating epileptiform abnormalities and
neuropsychiatric disorders in a RCT study in patients aged 6–21 years seizures, and continuing spasms are probably related to worse outcome.
comparing oral everolimus (n = 32) vs placebo (n = 15) taken once daily Developmental progress, but very limited improvement in the stan­
for 6 months. Comprehensive neurocognitive and behavioral evaluation dard scales, was reported in a subset of individuals who showed a
battery was administered at baseline, 3 months, and 6 months. At the worthwhile reduction of seizures frequency [94]. Altogether, more in­
end of the study everolimus did not significantly improve neuro­ formation on causes of surgical failures and predictors of efficacy is
development or behavior in children with TSC, possibly related to small needed [22,95,96] but importantly, patient satisfaction even in patients
sample size, study design and outcomes chosen [84]. However, the panel who did not achieve seizure freedom, was high [97]. Laser ablation was
believe that definitive conclusion could not be provided for the efficacy performed in three patients with promising results, but more data is
of everolimus on cognition, considered that the drug was used in in­ needed to recommend its use in TSC patients [98].
dividuals with older than 2 years of age with already significant neu­ The current relevance and limitation of surgery for TSC-associated
rodevelopmental deficits. Recent evidence showed that everolimus is drug resistant epilepsy was widely discussed by the panel. Presurgical
safe also in young children under the age of 2 years [85]. evaluation should be considered in all TSC patient with DRE and
Since functional connectivity is important for neurodevelopment, consistent semiology, concordant with EEG and MRI findings. On the
the longitudinal effects of everolimus were studied on callosal white other hand, children with multiple seizure types, multiple cortical tu­
matter diffusion tensor imaging (DTI) in patients with TSC. mTOR bers, and multiple EEG abnormalities remain a challenge for presurgical
overactivity on white matter microstructural integrity in TSC were evaluation and likely postsurgical seizure freedom will not be achieved.
modified through pharmacologic inhibition of mTOR [86]. However, The panel proposed that the identification of ideal candidates for sur­
how this effected neurodevelopment was not studied. Everolimus was gery should be performed at expert centers specialized in both TSC
used in a preclinical model of TSC induced by Tsc2+/− , and it led to a management and epilepsy surgery and emphasized that it should be
persistent improvement of the social deficit while deficits related to tailored (with ancillary investigations as high-resolution source imag­
developmental status epilepticus did not respond to treatment [87]. ing, single photon emission computed tomography (SPECT), or invasive

29
N. Specchio et al. European Journal of Paediatric Neurology 47 (2023) 25–34

Table 3 as an emerging treatment for TSC associated seizures.


Unanswered questions as insights for future studies. In a recent RCT, metformin reduced seizure frequency compared to
Questions placebo in 21 patients. Ideally, these results would need further
confirmation with adequate statistical power [111].
- What is the rationale for selecting the optimal add-on therapy or combination
therapy if the seizures persist after the use of two appropriately chosen anti-seizure The relatively small number of patients included in this RCT hamper
medication? May we talk about first line and beyond? firm conclusions, and at present this panel cannot recommend the use of
- What is the optimal timing for pre-symptomatic treatment and for how many years metformin.
do we have to maintain pre-symptomatic treatment?
- Which is the optimal first add-on treatment option after vigabatrin failure in focal
seizures?
8. Challenges for the future and unmet needs
- When should mTOR inhibitors be introduced?
- What is the efficacy and safety (short and long term) of mTOR inhibitors as pre- Several questions still remain unanswered, providing hypotheses for
symptomatic therapy commenced in children below 2 years of age? future studies (see Table 3).
- Why is mTOR inhibition ineffective for seizure control in about 50% of patients? Are
there other mTOR-independent mechanisms involved or is this mainly related to the
already ongoing process of epileptogenesis, and what are the interactions with other 9. Conclusions
signaling pathways?
- Can we determine whether earlier use of mTOR inhibitors benefits Epileptogenesis in TSC is a chronic and dynamic process. Different
neurodevelopment in TSC?
convergent and divergent mechanisms are involved in the establishment
- Which are the patients specific presurgical characteristics related to better or worse
surgical outcome, and does early surgery improve neurodevelopmental outcome?
of a pathological “network” capable of generating seizures and co-
- What are the main barriers related to the underuse of epilepsy surgery? Is this occurring neurodevelopmental disorders. mTORC1-dependent and -in­
related to the complexity of epileptogenic networks? dependent mechanisms may occur in parallel and interact throughout,
therefore combinatorial treatment strategies to engage different targets
involved in the epileptogenic network have been hypothesized and
recording), to the individual patient.
referred to as “Network therapy” [112]. In TSC this is still a hypothesis,
and a careful selection of patients for any combination treatment is
7. Other treatment options
important. More data on its modifying effect is needed before combi­
nation therapy can be recommended in daily clinical care. In addition,
Two recent studies confirmed that both ketogenic diet therapies (KD)
there are important observations of spontaneous remission of epilepsy
and vagal nerve stimulation (VNS) could be of benefit in reducing the
with age that need to be acknowledged [113].
seizure frequency in individuals with the refractory epilepsy, who are
To identify the contribution of mTORC1 dependent and independent
not suitable for epilepsy surgery [99–102]. In a meta-analysis on the role
mechanisms to the clinical phenotype and to explain failures with tar­
of VNS in genetic epilepsies, VNS showed a significant effect on the
geted treatment with mTOR inhibitors, exploring combination treat­
proportion of patients with ≥50% seizure reduction (Z = 5.75, p <
ment strategies may be of interest. There is recent experimental
0.001). Heterogeneity (I2 = 40.96%, p = 0.06) and publication bias (p
evidence showing that ASD phenotypes in TSC may result from a com­
= 0.772) were non-significant. In the same meta-analysis the pooled
bination of TSC1/TSC2 molecular deficits and additional social im­
(95% CI) seizure freedom rate was 40% (12%–71%), ≥90% seizure
pairments caused by seizures, but through different mechanisms that
reduction rate was 31% (8%–56%), and ≥50% reduction rate was 68%
may not all respond to mTOR inhibitors [87].
(48%–91%) [100]. In a cost analysis of competing therapeutic options
Compared to the recommendations compiled 5 years ago, the panel
for drug resistant seizures associated with TSC, a probability of positive
believes that substantial progress has been made thanks to the early
outcome (Engel class 1) was 0.55 for resective surgery, 0.12 for VNS,
identification of risk factors, which can predict both the impending
0.00 for KD, and 0.20 for mTOR inhibition, respectively [103]. KD can
epilepsy and the possibility of a severe drug-resistant epilepsy.
be an effective therapeutic strategy for seizures particularly in infancy
Furthermore, pre-symptomatic assessment and treatment with vigaba­
and early childhood [101,102].
trin are now more and more adopted in very high-risk infants. The panel
In an international placebo controlled randomized clinical trial to
concluded that the clinical approach for TSC-related epilepsy has vastly
evaluate the efficacy and safety of cannabidiol (CBD) in 224 patients
improved over the last few years, based on the progress in predicting and
with TSC associated seizures, CBD at dosages of 25 or 50 mg/kg/day
modifying strategies and on the advent of molecular targeted therapy.
significantly reduced seizure frequency if compared with placebo, with
Progress in the knowledge of the mechanism underlying epileptogenesis
no significant difference in efficacy between 25 and 50 mg/kg/day. The
has paved the way for new concepts in the management for TSC-related
most common adverse events were diarrhea, somnolence and elevated
epilepsy. Developmental neurobiology and neuropathology offer a
transaminase, more often seen at higher dosages [20]. A good safety and
practical basis for approaching such concepts, and early genetic diag­
efficacy profile was reported in 199 patients who entered the open label
nosis and use of reliable EEG and MRI biomarkers may allow the
study to evaluate long-term safety and efficacy with add-on CBD [104].
development of pre-symptomatic and disease-modifying strategies. The
Importantly, CBD is known to significantly increase serum levels of
panel acknowledged the need to develop and validate a scoring method
mTOR inhibitors and such combined therapy requires dosing modifi­
to calculate the relative risk of impending epilepsy and timing of pre­
cation and close monitoring. In patients taking both CBD and ever­
ventive treatment. The goal is to develop a more robust and accurate
olimus, the everolimus dosage should be reduced by nearly a half, and
prediction model that importantly guides counseling and treatment.
through levels should be monitored regularly [105,106].
Based on the results of the EPISTOP trial, the panel emphasized the
CBD is hypothesized to act at a synaptic level by reduction in
importance of a close developmental follow-up of infants with TSC at a
excitatory neurotransmission through at least three possible mecha­
sensitive developmental timepoints (6, 9, 12 months) for an early
nisms: block of GPR55 receptors, desensitization of TRPV1 channels, or
detection of possible delay in motor, language or cognitive develop­
inhibition of ENT1 adenosine reuptake pumps [107,108]. Interestingly,
mental trajectories. Early intervention strategies are crucial to improve
CBD is also shown to be a selective modulator of the mTOR downstream
the neurodevelopmental outcome. All infants with a definite diagnosis
molecule rpS6 in the brain in a preclinical-model of TSC [109].
of TSC should be followed up with early and frequent EEG monitoring
Ganaxolone - a positive allosteric modulator of GABA a receptor –
(every 4 weeks in first 6–8 months and every 4–8 weeks thereafter until
was approved within the USA for the treatment of seizures associated
12 months and every 12 weeks between 12 and 24 months) to detect the
with CDKL5 deficiency disorder (CDD) in March 2022 [110], its use is
appearance of interictal epileptiform discharges and abnormal back­
under evaluation in a phase III study (TRUSTTSC) and could play a role
ground patterns. Pre-symptomatic treatment with vigabatrin as soon as

30
N. Specchio et al. European Journal of Paediatric Neurology 47 (2023) 25–34

Table 4 Table 4 (continued )


Summary of the therapeutic recommendations for TSC associated seizures Treatment Limitations
(modified and updated from Curatolo P, Nabbout R, Lagae L et al. Management
of epilepsy associated with tuberous sclerosis complex: Updated clinical rec­ • Decreased mTOR activation in animal
models may provide biological basis
ommendations. Eur J Paediatr Neurol. 2018; 22 (5):738–748).
for mechanism of action in TSC-
Treatment Limitations associated seizures
Vagus nerve stimulation
Antiseizure medications
• May be a first option in drug resistant • Almost no patients become seizure-
Vigabatrin
epilepsy when ketogenic diet is not free
• Recommended as first-line mono­ • Retinal toxicity (lack of data in infants
acceptable or may be used in
therapy in TSC related spasms or focal and young children)
combination with ketogenic diet
seizures in children <1 year of age • Retinal toxicity concerns, newer data
• Limited data indicate many patients
• Pre-symptomatic treatment with VGB suggest that this may be a lower risk
experience reduction of seizure
(see text for recommended time points than previously thought [72]
frequency
for clinical and EEG screening and
Cannabidiol
EEG indicators for initiating
• CBD at the dosage of 25 up to 50 mg/ • Important drug-drug interaction with
treatment) is now strongly
kg/day significantly reduces seizure everolimus should be considered.
recommended and may improve long-
frequency if compared with placebo
term epilepsy and neurodevelopment
• Dosages of 50 mg/kg/day are
outcomes [18]
associated with more adverse events
Other ASMs
• ACTH and prednisolone are effective • Careful selection for maximal synergy TSC: tuberos sclerosis complex, ACTH: Adrenocorticotropic hormone, mTOR:
for TSC-related infantile spasms and and minimal unfavorable reactions/ mammalian target of rapamycin, ASM: anti-seizure medication.
are used as second-line therapy toxicity
ASMs combination therapy
• Appropriate when Vigabatrin and focal spikes occur, but when multifocal spikes, pre-hypsarrhythmia/
steroid therapy has failed hypsarrhythmia, subclinical seizures are recorded on EEG, delays sei­
mTOR inhibitors: everolimus and sirolimus zures and DRE risk, and should be used in all patients. The panel
• Everolimus is effective as an • Potential drug-drug interactions
adjunctive treatment in patients aged • Seizures may recur when discontinued
emphasized the need to identify the subpopulation of infants that have
2 years and older with refractory focal • Glucose intolerance and an even higher risk to develop epilepsy and ASD. Moreover, parents
seizures, with or without evolution to immunosuppression. should be trained to recognize the appearance of subtle focal seizures or
focal to bilateral tonic-clonic seizures atypical behavior or development early. All infants with a definite
• Everolimus should be considered
diagnosis of TSC should be evaluated with standard developmental
before the age of 6 years as in real-
world clinical practice better response scales at 6, 12, 18 and 24 months to detect early deviations in motor,
was seen in this age group language, and cognitive trajectories. Initial neurodevelopmental atypias
• Everolimus adjunctive therapy can be in children with TSC may represent a risk of later ASD diagnosis. A
considered after failure of 2 summary of the therapeutic recommendations for TSC associated sei­
appropriate ASMs
• Sirolimus has a favorable safety and
zures is given in Table 4.
efficacy profile for treating The panel is pleased to acknowledge the great progress made in the
manifestations of TSC (including management of TSC- associated epilepsy and believes that a combina­
epilepsy) tion of different therapeutic options could be considered in a well
Surgery
selected population of children, even if a personalized medicine
• Currently underutilized and under • Children with multiple seizure types,
assessed in TSC-associated refractory multiple cortical tubers, and multiple approach remains a challenge for future years.
seizures EEG abnormalities remain a challenge
• Presurgical evaluation should be and likely postsurgical seizure Declaration of competing interest
considered in all TSC patient with freedom will not be achieved but
DRE palliative approach can yield clinical
• Early surgery significantly increases improvement We confirm that we have read the Journal’s position on issues
probability that patients will be involved in ethical publication and affirm that this report is consistent
seizure-free with those guidelines.
• Resection beyond tuber margins is
N Specchio has served on scientific advisory boards for GW Pharma,
associated with greater probability of
seizure freedom BioMarin, Arvelle, Marinus and Takeda; has received speaker honoraria
• Multifocal and bilateral lesions do not from Eisai, Biomarin, Livanova, Sanofi; has served as an investigator for
preclude presurgical assessment or Zogenix, Marinus, Biomarin, UCB, Roche.
subsequent resective surgery R Nabbout has served as principal investigators in clinical trials for
• Overall post-operative seizure
freedom is achieved in 50–60% of TSC
Novartis, Nutricia, Eisai, UCB, GW Pharma, Livanova. She received
patients, and >50% seizure reduction consulting and lecturer honoraria from Biogene, BioMarin, Praxis, GW
is seen in another 15%, even in pa­ Pharma, Zogenix, Novartis, Nutricia, Stoke, Ionis, Targeon, Neu­
tients with multiple tubers raxpharma, Takeda, Nutricia, Biocodex, Advicennes and Eisai. She
Ketogenic diet
received unrestricted research grants from Eisai, UCB, Livanova and GW
• Ketogenic dietary therapies can be an • Compliance may be a problem in
effective therapeutic strategy for children past infancy Pharma and academic research grants from EJP-RD (horizons 2020).
seizures particularly in infancy and E Aronica has nothing to disclose.
early childhood S Auvin is a deputy editor at Epilepsia and has received support from,
• Consider for patients who are not and/or has served as a paid consultant for Biocodex, Eisai, Encoded, Jazz
surgical candidates, who have failed
surgery, or with multifocal seizure
Pharmaceuticals, GRIN Therapeutics, Neuraxpharm, Nutricia, Orion
onset Pharma, Supernus Pharmaceuticals, Takeda Pharmaceutical Company,
• Use instead of other ASMs after UCB Pharma, Vitaflo, Xenon Pharmaceuticals, and Zogenix.
vigabatrin failure remains M Feucht received speaker and advisory board honoraria as well as
controversial
scientific grants from Biocodex, Eisai, Gerot, Livanova, Novartis,
Nutricia, Shire, UCB and GW Pharma.
F Jansen has served as a consultant for Jazz Pharmaceuticals,

31
N. Specchio et al. European Journal of Paediatric Neurology 47 (2023) 25–34

Nutricia, Novartis and UCB. She was an investigator of the GWEPCARE [13] C.J. Chu-Shore, P. Major, S. Camposano, D. Muzykewicz, E.A. Thiele, The natural
history of epilepsy in tuberous sclerosis complex, Epilepsia 51 (7) (2009)
1512 trial.
1236–1241, https://doi.org/10.1111/j.1528-1167.2009.02474.x.
L Lagae received grants and is a consultant and/or speaker for [14] K. Kotulska, E. Jurkiewicz, D. Domańska-Pakieła, et al., Epilepsy in newborns
Zogenix; LivaNova, UCB, Shire, Eisai, Novartis, Takeda/Ovid, NEL, with tuberous sclerosis complex, Eur. J. Paediatr. Neurol. 18 (6) (2014) 714–721,
Epihunter. https://doi.org/10.1016/j.ejpn.2014.06.009.
[15] M. Lynch, K. Smith, K. Riney, Clinical seizure semiology is subtle and
A Benvenuto, K Kotulska, S Jozwiak, P Curatolo report no conflicts of identification of seizures by parents is unreliable in infants with tuberous sclerosis
interest. complex, Epilepsia (November 2022), https://doi.org/10.1111/epi.17454.
[16] P. Curatolo, R. Nabbout, L. Lagae, et al., Management of epilepsy associated with
tuberous sclerosis complex: updated clinical recommendations, Eur. J. Paediatr.
Acknowledgments Neurol. 22 (5) (2018) 738–748, https://doi.org/10.1016/j.ejpn.2018.05.006.
[17] R. Nabbout, E. Belousova, M.P. Benedik, et al., Epilepsy in tuberous sclerosis
The contribution of Sergiusz Jozwiak and Katarzyna Kotulska has complex: findings from the TOSCA Study, Epilepsia Open 4 (1) (2019) 73–84,
https://doi.org/10.1002/epi4.12286.
been partly supported by grant EPIMARKER of the Polish National [18] K. Kotulska, D.J. Kwiatkowski, P. Curatolo, et al., Prevention of epilepsy in
Center for Research and Development no. STRATEGMED3/306306/4/ infants with tuberous sclerosis complex in the <scp>EPISTOP</scp> trial, Ann.
2016, grant VIRAP (Project number 2019/ABM/01/00034) and grant Neurol. 89 (2) (2021) 304–314, https://doi.org/10.1002/ana.25956.
[19] J.A. French, J.A. Lawson, Z. Yapici, et al., Adjunctive everolimus therapy for
RaRE-TS (Project number 2020/ABM/01/00054) of the Medical treatment-resistant focal-onset seizures associated with tuberous sclerosis (EXIST-
Research Agency, Poland, and a statutory grant of the Children’s Me­ 3): a phase 3, randomised, double-blind, placebo-controlled study, Lancet 388
morial Health Institute financed by the Polish Ministry of Science and (10056) (2016) 2153–2163, https://doi.org/10.1016/S0140-6736(16)31419-2.
[20] E.A. Thiele, E.M. Bebin, H. Bhathal, et al., Add-on cannabidiol treatment for drug-
Higher Education.
resistant seizures in tuberous sclerosis complex, JAMA Neurol. 78 (3) (2021) 285,
Eleonora Aronica acknowledges support by The Netherlands Orga­ https://doi.org/10.1001/jamaneurol.2020.4607.
nisation for Health Research and Development (ZonMw). [21] M. Słowińska, K. Kotulska, S. Szymańska, S.L. Roberds, C. Fladrowski, S. Jóźwiak,
Rima Nabbout, Eleonora Aronica, Arianna Benvenuto, Martha Approach to preventive epilepsy treatment in tuberous sclerosis complex and
current clinical practice in 23 countries, Pediatr. Neurol. 115 (2021) 21–27,
Feucht, Floor Jansen, Katarzyna Kotulska, Lieven Lagae, Sergiusz Joz­ https://doi.org/10.1016/j.pediatrneurol.2020.11.003.
wiak, and Paolo Curatolo were supported by the 7th Framework Pro­ [22] N. Specchio, C. Pepi, L. de Palma, et al., Surgery for drug-resistant tuberous
gram of the European Commission within the Large-Scale Integrating sclerosis complex-associated epilepsy: who, when, and what, Epileptic Disord. 23
(1) (2021) 53–73, https://doi.org/10.1684/epd.2021.1253.
Project EPISTOP (Proposal No. 602391–2; www.epistop.eu). [23] A. Bongaarts, K. Giannikou, R.J. Reinten, et al., Subependymal giant cell
Nicola Specchio is supported by #NEXTGENERATIONEU (NGEU) astrocytomas in Tuberous Sclerosis Complex have consistent TSC1/TSC2 biallelic
and funded by the Ministry of University and Research (MUR), National inactivation, and no BRAF mutations, Oncotarget 8 (56) (2017) 95516–95529,
https://doi.org/10.18632/oncotarget.20764.
Recovery and Resilience Plan (NRRP), project MNESYS (PE0000006) – [24] K.R. Martin, W. Zhou, M.J. Bowman, et al., The genomic landscape of tuberous
A Multiscale integrated approach to the study of the nervous system in sclerosis complex, Nat. Commun. 8 (1) (2017), 15816, https://doi.org/10.1038/
health and disease (DN. 1553 October 11, 2022). ncomms15816.
[25] P. Curatolo, R. Moavero, J. van Scheppingen, E. Aronica, mTOR dysregulation
and tuberous sclerosis-related epilepsy, Expert Rev. Neurother. 18 (3) (2018)
References 185–201, https://doi.org/10.1080/14737175.2018.1428562.
[26] D.Y. Lee, Roles of mTOR signaling in brain development, Exp Neurobiol 24 (3)
[1] P. Curatolo, R. Moavero, P.J. de Vries, Neurological and neuropsychiatric aspects (2015) 177–185, https://doi.org/10.5607/en.2015.24.3.177.
of tuberous sclerosis complex, Lancet Neurol. 14 (7) (2015) 733–745, https://doi. [27] M.G. Andrews, L. Subramanian, A.R. Kriegstein, mTOR signaling regulates the
org/10.1016/S1474-4422(15)00069-1. morphology and migration of outer radial glia in developing human cortex, Elife
[2] D. Ebrahimi-Fakhari, L.L. Mann, M. Poryo, et al., Incidence of tuberous sclerosis 9 (2020), https://doi.org/10.7554/eLife.58737.
and age at first diagnosis: new data and emerging trends from a national, [28] A.S. Musah, T.L. Brown, M.A. Jeffries, et al., Mechanistic target of rapamycin
prospective surveillance study, Orphanet J. Rare Dis. 13 (1) (2018) 117, https:// regulates the oligodendrocyte cytoskeleton during myelination, J. Neurosci. 40
doi.org/10.1186/s13023-018-0870-y. (15) (2020) 2993–3007, https://doi.org/10.1523/JNEUROSCI.1434-18.2020.
[3] S. Amin, A. Lux, N. Calder, M. Laugharne, J. Osborne, F. O’callaghan, Causes of [29] A. Mühlebner, A. Bongaarts, H.B. Sarnat, T. Scholl, E. Aronica, New insights into a
mortality in individuals with tuberous sclerosis complex, Dev. Med. Child Neurol. spectrum of developmental malformations related to mTOR dysregulations:
59 (6) (2017) 612–617, https://doi.org/10.1111/dmcn.13352. challenges and perspectives, J. Anat. 235 (3) (2019) 521–542, https://doi.org/
[4] P. Curatolo, Mechanistic target of rapamycin (mTOR) in tuberous sclerosis 10.1111/joa.12956.
complex-associated epilepsy, Pediatr. Neurol. 52 (3) (2015) 281–289, https:// [30] R. Guerrini, M. Cavallin, T. Pippucci, et al., Is focal cortical dysplasia/epilepsy
doi.org/10.1016/j.pediatrneurol.2014.10.028. caused by somatic MTOR mutations always a unilateral disorder? Neurol Genet 7
[5] H. Northrup, M.E. Aronow, E.M. Bebin, et al., Updated international tuberous (1) (2021), e540 https://doi.org/10.1212/NXG.0000000000000540.
sclerosis complex diagnostic criteria and surveillance and management [31] L.A. Jansen, The space-time continuum of cortical dysplasia, Epilepsy Curr. 19 (1)
recommendations, Pediatr. Neurol. 123 (2021) 50–66, https://doi.org/10.1016/ (2019) 54–56, https://doi.org/10.1177/1535759718822039.
j.pediatrneurol.2021.07.011. [32] L.H. Nguyen, T. Mahadeo, A. Bordey, mTOR hyperactivity levels influence the
[6] Z. Ye, S. Lin, X. Zhao, et al., Mosaicism in tuberous sclerosis complex: lowering severity of epilepsy and associated neuropathology in an experimental model of
the threshold for clinical reporting, Hum. Mutat. 43 (12) (2022) 1956–1969, tuberous sclerosis complex and focal cortical dysplasia, J. Neurosci. 39 (14)
https://doi.org/10.1002/humu.24454. (2019) 2762–2773, https://doi.org/10.1523/JNEUROSCI.2260-18.2019.
[7] M.E. Tyburczy, K.A. Dies, J. Glass, et al., Mosaic and intronic mutations in TSC1/ [33] A.B. Gelot, A. Represa, Progression of fetal brain lesions in tuberous sclerosis
TSC2 explain the majority of TSC patients with No mutation identified by complex, Front. Neurosci. 14 (2020) 899, https://doi.org/10.3389/
conventional testing, PLoS Genet. 11 (11) (2015), e1005637, https://doi.org/ fnins.2020.00899.
10.1371/journal.pgen.1005637. [34] H.M. Hulshof, E.M.H. Slot, M. Lequin, et al., Fetal brain magnetic resonance
[8] K. Bąbol-Pokora, M. Bielska, K. Bobeff, et al., A multistep approach to the imaging findings predict neurodevelopment in children with tuberous sclerosis
genotype-phenotype analysis of Polish patients with tuberous sclerosis complex, complex, J. Pediatr. 233 (2021) 156–162, https://doi.org/10.1016/j.
Eur. J. Med. Genet. 64 (10) (2021), 104309, https://doi.org/10.1016/j. jpeds.2021.02.060, e2.
ejmg.2021.104309. [35] M. Słowińska, S. Jóźwiak, A. Peron, et al., Early diagnosis of tuberous sclerosis
[9] H. Northrup, D.A. Krueger, H. Northrup, et al., Tuberous sclerosis complex complex: a race against time. How to make the diagnosis before seizures?
diagnostic criteria update: recommendations of the 2012 international tuberous Orphanet J. Rare Dis. 13 (1) (2018) 25, https://doi.org/10.1186/s13023-018-
sclerosis complex consensus conference, Pediatr. Neurol. 49 (4) (2013) 243–254, 0764-z.
https://doi.org/10.1016/j.pediatrneurol.2013.08.001. [36] C. Rutten, L. Fillon, M. Kuchenbuch, et al., The longitudinal evolution of cerebral
[10] P. Curatolo, N. Specchio, E. Aronica, Advances in the genetics and blood flow in children with tuberous sclerosis assessed by arterial spin labeling
neuropathology of tuberous sclerosis complex: edging closer to targeted therapy, magnetic resonance imaging may be related to cognitive performance, Eur.
Lancet Neurol. 21 (9) (2022) 843–856, https://doi.org/10.1016/S1474-4422(22) Radiol. 33 (1) (2023) 196–206, https://doi.org/10.1007/s00330-022-09036-3.
00213-7. [37] J.-H.L. Hwang, O.S. Perloff, S.E. Gaus, et al., Tuberous sclerosis complex is
[11] N. Specchio, N. Pietrafusa, M. Trivisano, et al., Autism and epilepsy in patients associated with a novel human tauopathy, Acta Neuropathol. (December 2022),
with tuberous sclerosis complex, Front. Neurol. 11 (2020) 639, https://doi.org/ https://doi.org/10.1007/s00401-022-02521-5.
10.3389/fneur.2020.00639. [38] E. Aronica, N. Specchio, M.J. Luinenburg, P. Curatolo, Epileptogenesis in
[12] P.E. Davis, R. Filip-Dhima, G. Sideridis, et al., Presentation and diagnosis of tuberous sclerosis complex-related developmental and epileptic encephalopathy,
tuberous sclerosis complex in infants, Pediatrics 140 (6) (2017), e20164040, Brain (February 2023), https://doi.org/10.1093/brain/awad048.
https://doi.org/10.1542/peds.2016-4040. [39] J.D. Mills, A.M. Iyer, J. van Scheppingen, et al., Coding and small non-coding
transcriptional landscape of tuberous sclerosis complex cortical tubers:

32
N. Specchio et al. European Journal of Paediatric Neurology 47 (2023) 25–34

implications for pathophysiology and treatment, Sci. Rep. 7 (1) (2017) 8089, study, Genet. Med. 22 (9) (2020) 1489–1497, https://doi.org/10.1038/s41436-
https://doi.org/10.1038/s41598-017-06145-8. 020-0823-4.
[40] T.S. Zimmer, D.W.M. Broekaart, V.-E. Gruber, E.A. van Vliet, A. Mühlebner, [65] J. De Ridder, M. Lavanga, B. Verhelle, et al., Prediction of neurodevelopment in
E. Aronica, Tuberous sclerosis complex as disease model for investigating mTOR- infants with tuberous sclerosis complex using early EEG characteristics, Front.
related gliopathy during epileptogenesis, Front. Neurol. 11 (2020) 1028, https:// Neurol. 11 (2020), 582891, https://doi.org/10.3389/fneur.2020.582891.
doi.org/10.3389/fneur.2020.01028. [66] H.M. Hulshof, H.J. Kuijf, K. Kotulska, et al., Association of early MRI
[41] G. Ruffolo, A. Iyer, P. Cifelli, et al., Functional aspects of early brain development characteristics with subsequent epilepsy and neurodevelopmental outcomes in
are preserved in tuberous sclerosis complex (TSC) epileptogenic lesions, children with tuberous sclerosis complex, Neurology 98 (12) (2022)
Neurobiol. Dis. 95 (2016) 93–101, https://doi.org/10.1016/j.nbd.2016.07.014. e1216–e1225, https://doi.org/10.1212/WNL.0000000000200027.
[42] E.M. Powell, Interneuron development and epilepsy: early genetic defects cause [67] A.L. Cohen, B.P.F. Mulder, A.K. Prohl, et al., Tuber locations associated with
long-term consequences in seizures and susceptibility, Epilepsy Curr. 13 (4) infantile spasms map to a common brain network, Ann. Neurol. 89 (4) (2021)
(2013) 172–176, https://doi.org/10.5698/1535-7597-13.4.172. 726–739, https://doi.org/10.1002/ana.26015.
[43] V.-E. Gruber, J. Lang, V. Endmayr, et al., Impaired myelin production due to an [68] A. Neal, R. Bouet, S. Lagarde, et al., Epileptic spasms are associated with
intrinsic failure of oligodendrocytes in mTORpathies, Neuropathol. Appl. increased stereo-electroencephalography derived functional connectivity in
Neurobiol. 47 (6) (2021) 812–825, https://doi.org/10.1111/nan.12744. tuberous sclerosis complex, Epilepsia (July 2022), https://doi.org/10.1111/
[44] B. Zhang, J. Zou, N.R. Rensing, M. Yang, M. Wong, Inflammatory mechanisms epi.17353.
contribute to the neurological manifestations of tuberous sclerosis complex, [69] L.S. Farach, M.A. Richard, P.J. Lupo, et al., Epilepsy risk prediction model for
Neurobiol. Dis. 80 (2015) 70–79, https://doi.org/10.1016/j.nbd.2015.04.016. patients with tuberous sclerosis complex, Pediatr. Neurol. 113 (2020) 46–50,
[45] N. Lozovaya, S. Gataullina, T. Tsintsadze, et al., Selective suppression of excessive https://doi.org/10.1016/j.pediatrneurol.2020.07.015.
GluN2C expression rescues early epilepsy in a tuberous sclerosis murine model, [70] X. Zhao, D. Jiang, Z. Hu, et al., Machine learning and statistic analysis to predict
Nat. Commun. 5 (2014) 4563, https://doi.org/10.1038/ncomms5563. drug treatment outcome in pediatric epilepsy patients with tuberous sclerosis
[46] S. Gataullina, G. Galvani, S. Touchet, et al., GluN2C selective inhibition is a target complex, Epilepsy Res. 188 (2022), 107040, https://doi.org/10.1016/j.
to develop new antiepileptic compounds, Epilepsia 63 (11) (2022) 2911–2924, eplepsyres.2022.107040.
https://doi.org/10.1111/epi.17396. [71] M. Słowińska, K. Kotulska, S. Szymańska, S.L. Roberds, C. Fladrowski, S. Jóźwiak,
[47] P.J. de Vries, L. Wilde, M.C. de Vries, R. Moavero, D.A. Pearson, P. Curatolo, A Approach to preventive epilepsy treatment in tuberous sclerosis complex and
clinical update on tuberous sclerosis complex-associated neuropsychiatric current clinical practice in 23 countries, Pediatr. Neurol. 115 (2021) 21–27,
disorders (TAND), Am J Med Genet C Semin Med Genet 178 (3) (2018) 309–320, https://doi.org/10.1016/j.pediatrneurol.2020.11.003.
https://doi.org/10.1002/ajmg.c.31637. [72] R. Foroozan, Vigabatrin: lessons learned from the United States experience, J.
[48] R.A. Mitchell, M. Mitchell, K. Williams, The autism spectrum disorder phenotype Neuro Ophthalmol. 38 (4) (2018) 442–450, https://doi.org/10.1097/
in children with tuberous sclerosis complex: a systematic review and meta- WNO.0000000000000609.
analysis, Dev. Med. Child Neurol. 64 (10) (2022) 1214–1229, https://doi.org/ [73] S.N. Sehgal, H. Baker, C. Vézina, Rapamycin (AY-22,989), a new antifungal
10.1111/dmcn.15307. antibiotic. II. Fermentation, isolation and characterization, J. Antibiot. (Tokyo)
[49] J.K. Capal, B. Bernardino-Cuesta, P.S. Horn, et al., Influence of seizures on early 28 (10) (1975) 727–732, https://doi.org/10.7164/antibiotics.28.727.
development in tuberous sclerosis complex, Epilepsy Behav. 70 (Pt A) (2017) [74] M. Koike-Kumagai, M. Fujimoto, M. Wataya-Kaneda, Sirolimus relieves seizures
245–252, https://doi.org/10.1016/j.yebeh.2017.02.007. and neuropsychiatric symptoms via changes of microglial polarity in tuberous
[50] R. Moavero, A. Benvenuto, L. Emberti Gialloreti, et al., Early clinical predictors of sclerosis complex model mice, Neuropharmacology 218 (2022), 109203, https://
autism spectrum disorder in infants with tuberous sclerosis complex: results from doi.org/10.1016/j.neuropharm.2022.109203.
the EPISTOP study, J. Clin. Med. 8 (6) (2019) 788, https://doi.org/10.3390/ [75] A. Sato, S. Kasai, T. Kobayashi, et al., Rapamycin reverses impaired social
jcm8060788. interaction in mouse models of tuberous sclerosis complex, Nat. Commun. 3 (1)
[51] D.M. Talos, H. Sun, B. Kosaras, et al., Altered inhibition in tuberous sclerosis and (2012) 1292, https://doi.org/10.1038/ncomms2295.
type IIb cortical dysplasia, Ann. Neurol. 71 (4) (2012) 539–551, https://doi.org/ [76] S.W. Way, N.S. Rozas, H.C. Wu, et al., The differential effects of prenatal and/or
10.1002/ana.22696. postnatal rapamycin on neurodevelopmental defects and cognition in a neuroglial
[52] P. Curatolo, E. Aronica, A. Jansen, et al., Early onset epileptic encephalopathy or mouse model of tuberous sclerosis complex, Hum. Mol. Genet. 21 (14) (2012)
genetically determined encephalopathy with early onset epilepsy? Lessons 3226–3236, https://doi.org/10.1093/hmg/dds156.
learned from TSC, Eur. J. Paediatr. Neurol. (2016), https://doi.org/10.1016/j. [77] C.A. Amegandjin, M. Choudhury, V. Jadhav, et al., Sensitive period for rescuing
ejpn.2015.12.005. parvalbumin interneurons connectivity and social behavior deficits caused by
[53] R. Moavero, K. Kotulska, L. Lagae, et al., Is autism driven by epilepsy in infants TSC1 loss, Nat. Commun. 12 (1) (2021) 3653, https://doi.org/10.1038/s41467-
with Tuberous Sclerosis Complex? Ann Clin Transl Neurol 7 (8) (2020) 021-23939-7.
1371–1381, https://doi.org/10.1002/acn3.51128. [78] K. Sadowski, K. Sijko, D. Domańska-Pakieła, et al., Antiepileptic effect and safety
[54] I.E. Scheffer, S. Berkovic, G. Capovilla, et al., ILAE classification of the epilepsies: profile of rapamycin in pediatric patients with tuberous sclerosis complex, Front.
position paper of the ILAE commission for classification and terminology, Neurol. 13 (2022), 704978, https://doi.org/10.3389/fneur.2022.704978.
Epilepsia 58 (4) (2017) 512–521, https://doi.org/10.1111/epi.13709. [79] R. Nabbout, M. Kuchenbuch, C. Chiron, P. Curatolo, Pharmacotherapy for
[55] N. Specchio, P. Curatolo, Developmental and epileptic encephalopathies: what we seizures in tuberous sclerosis complex, CNS Drugs 35 (9) (2021) 965–983,
do and do not know, Brain 144 (1) (2021) 32–43, https://doi.org/10.1093/brain/ https://doi.org/10.1007/s40263-021-00835-8.
awaa371. [80] Z. Zhang, Q. Fan, X. Luo, K. Lou, W.A. Weiss, K.M. Shokat, Brain-restricted mTOR
[56] J.K. Capal, B. Bernardino-Cuesta, P.S. Horn, et al., Influence of seizures on early inhibition with binary pharmacology, Nature 609 (7928) (2022) 822–828,
development in tuberous sclerosis complex, Epilepsy Behav. 70 (2017) 245–252, https://doi.org/10.1038/s41586-022-05213-y.
https://doi.org/10.1016/j.yebeh.2017.02.007. [81] D.A. Krueger, A.A. Wilfong, M. Mays, et al., Long-term treatment of epilepsy with
[57] A. Jeong, J.A. Nakagawa, M. Wong, Predictors of drug-resistant epilepsy in everolimus in tuberous sclerosis, Neurology 87 (23) (2016) 2408–2415, https://
tuberous sclerosis complex, J. Child Neurol. 32 (14) (2017) 1092–1098, https:// doi.org/10.1212/WNL.0000000000003400.
doi.org/10.1177/0883073817737446. [82] P. Curatolo, D.N. Franz, J.A. Lawson, et al., Adjunctive everolimus for children
[58] D. Miszewska, M. Sugalska, S. Jóźwiak, Risk factors associated with refractory and adolescents with treatment-refractory seizures associated with tuberous
epilepsy in patients with tuberous sclerosis complex: a systematic review, J. Clin. sclerosis complex: post-hoc analysis of the phase 3 EXIST-3 trial, Lancet Child
Med. (23) (2021) 10, https://doi.org/10.3390/jcm10235495. Adolesc Heal 2 (7) (2018) 495–504, https://doi.org/10.1016/S2352-4642(18)
[59] J.Y. Wu, M. Goyal, J.M. Peters, et al., Scalp EEG spikes predict impending 30099-3.
epilepsy in TSC infants: a longitudinal observational study, Epilepsia 60 (12) [83] J.C. Kingswood, E. Belousova, M.P. Benedik, et al., TuberOus SClerosis registry to
(2019) 2428–2436, https://doi.org/10.1111/epi.16379. increAse disease awareness (TOSCA) post-authorisation safety study of
[60] B. Ogórek, L. Hamieh, H.M. Hulshof, et al., TSC2 pathogenic variants are everolimus in patients with tuberous sclerosis complex, Front. Neurol. 12 (2021),
predictive of severe clinical manifestations in TSC infants: results of the EPISTOP 630378, https://doi.org/10.3389/fneur.2021.630378.
study, Genet. Med. 22 (9) (2020) 1489–1497, https://doi.org/10.1038/s41436- [84] I.E. Overwater, A.B. Rietman, S.E. Mous, et al., A randomized controlled trial with
020-0823-4. everolimus for IQ and autism in tuberous sclerosis complex, Neurology 93 (2)
[61] J. De Ridder, K. Kotulska, P. Curatolo, et al., Evolution of electroencephalogram (2019) e200–e209, https://doi.org/10.1212/WNL.0000000000007749.
in infants with tuberous sclerosis complex and neurodevelopmental outcome: a [85] D.A. Krueger, J.K. Capal, P. Curatolo, et al., Short-term safety of mTOR inhibitors
prospective cohort study, Dev. Med. Child Neurol. 64 (4) (2022) 495–501, in infants and very young children with tuberous sclerosis complex (TSC):
https://doi.org/10.1111/dmcn.15073. multicentre clinical experience, Eur. J. Paediatr. Neurol. 22 (6) (2018)
[62] J. De Ridder, B. Verhelle, J. Vervisch, et al., Early epileptiform EEG activity in 1066–1073, https://doi.org/10.1016/j.ejpn.2018.06.007.
infants with tuberous sclerosis complex predicts epilepsy and [86] J.M. Peters, A. Prohl, K. Kapur, et al., Longitudinal effects of everolimus on white
neurodevelopmental outcomes, Epilepsia 62 (5) (2021) 1208–1219, https://doi. matter diffusion in tuberous sclerosis complex, Pediatr. Neurol. 90 (2019) 24–30,
org/10.1111/epi.16892. https://doi.org/10.1016/j.pediatrneurol.2018.10.005.
[63] H.M. Hulshof, H.J. Kuijf, K. Kotulska, et al., Association of early MRI [87] T. Petrasek, I. Vojtechova, O. Klovrza, et al., mTOR inhibitor improves autistic-
characteristics with subsequent epilepsy and neurodevelopmental outcomes in like behaviors related to Tsc2 haploinsufficiency but not following developmental
children with tuberous sclerosis complex, Neurology 98 (12) (2022) status epilepticus, J. Neurodev. Disord. 13 (1) (2021) 14, https://doi.org/
e1216–e1225, https://doi.org/10.1212/WNL.0000000000200027. 10.1186/s11689-021-09357-2.
[64] B. Ogórek, L. Hamieh, H.M. Hulshof, et al., TSC2 pathogenic variants are [88] S. Cavalheiro, M.D.S. da Costa, R. Richtmann, Everolimus as a possible prenatal
predictive of severe clinical manifestations in TSC infants: results of the EPISTOP treatment of in utero diagnosed subependymal lesions in tuberous sclerosis

33
N. Specchio et al. European Journal of Paediatric Neurology 47 (2023) 25–34

complex: a case report, Childs Nerv Syst 37 (12) (2021) 3897–3899, https://doi. analysis, J. Neurosurg. Pediatr. (March 2022) 1–14, https://doi.org/10.3171/
org/10.1007/s00381-021-05218-4. 2022.1.PEDS222.
[89] D.N. Franz, J.A. Lawson, Z. Yapici, et al., Everolimus for treatment-refractory [101] Y. Fang, D. Li, M. Wang, et al., Ketogenic diet therapy for drug-resistant epilepsy
seizures in TSC, Neurol Clin Pract 8 (5) (2018) 412–420, https://doi.org/ and cognitive impairment in children with tuberous sclerosis complex, Front.
10.1212/CPJ.0000000000000514. Neurol. 13 (2022), 863826, https://doi.org/10.3389/fneur.2022.863826.
[90] P. Curatolo, M. Bjørnvold, P.E. Dill, et al., The role of mTOR inhibitors in the [102] S. Park, E.J. Lee, S. Eom, H.-C. Kang, J.S. Lee, H.D. Kim, Ketogenic diet for the
treatment of patients with tuberous sclerosis complex: evidence-based and expert management of epilepsy associated with tuberous sclerosis complex in children, J
opinions, Drugs 76 (5) (2016) 551–565, https://doi.org/10.1007/s40265-016- Epilepsy Res 7 (1) (2017) 45–49, https://doi.org/10.14581/jer.17008.
0552-9. [103] A. Fallah, A.G. Weil, S. Wang, E. Lewis, C.B. Baca, G.W. Mathern, Cost-utility
[91] J.Z. Nie, C.L. Karras, S.J. Trybula, P. Texakalidis, T.D. Alden, The role of analysis of competing treatment strategies for drug-resistant epilepsy in children
neurosurgery in the management of tuberous sclerosis complex-associated with Tuberous Sclerosis Complex, Epilepsy Behav. 63 (2016) 79–88, https://doi.
epilepsy: a systematic review, Neurosurg. Focus 52 (5) (2022) E6, https://doi. org/10.1016/j.yebeh.2016.07.034.
org/10.3171/2022.2.FOCUS21789. [104] E.A. Thiele, E.M. Bebin, F. Filloux, et al., Long-term cannabidiol treatment for
[92] R. Nabbout, E. Belousova, M.P. Benedik, et al., Historical patterns of diagnosis, seizures in patients with tuberous sclerosis complex: an open-label extension trial,
treatments, and outcome of epilepsy associated with tuberous sclerosis complex: Epilepsia 63 (2) (2022) 426–439, https://doi.org/10.1111/epi.17150.
results from TOSCA registry, Front. Neurol. 12 (2021), 697467, https://doi.org/ [105] A. Wiemer-Kruel, B. Stiller, T. Bast, Cannabidiol interacts significantly with
10.3389/fneur.2021.697467. everolimus-report of a patient with tuberous sclerosis complex, Neuropediatrics
[93] S. Liu, T. Yu, Y. Guan, et al., Resective epilepsy surgery in tuberous sclerosis 50 (6) (2019) 400–403, https://doi.org/10.1055/s-0039-1695786.
complex: a nationwide multicentre retrospective study from China, Brain 143 (2) [106] D. Ebrahimi-Fakhari, K.D. Agricola, C. Tudor, D. Krueger, D.N. Franz, Cannabidiol
(2020) 570–581, https://doi.org/10.1093/brain/awz411. elevates mechanistic target of rapamycin inhibitor levels in patients with
[94] C. Vannicola, L. Tassi, C. Barba, et al., Seizure outcome after epilepsy surgery in tuberous sclerosis complex, Pediatr. Neurol. 105 (2020) 59–61, https://doi.org/
tuberous sclerosis complex: results and analysis of predictors from a multicenter 10.1016/j.pediatrneurol.2019.11.017.
study, J. Neurol. Sci. 427 (2021), 117506, https://doi.org/10.1016/j. [107] R.A. Gray, B.J. Whalley, The proposed mechanisms of action of CBD in epilepsy,
jns.2021.117506. Epileptic Disord. 22 (S1) (2020) 10–15, https://doi.org/10.1684/epd.2020.1135.
[95] S. Chivukula, O. Modiri, A. Kashanian, et al., Effect of gene mutation on seizures [108] C. Ibeas Bih, T. Chen, A.V.W. Nunn, M. Bazelot, M. Dallas, B.J. Whalley,
in surgery for tuberous sclerosis complex, Can. J. Neurol. Sci. 48 (3) (2021) Molecular targets of cannabidiol in neurological disorders, Neurotherapeutics 12
327–334, https://doi.org/10.1017/cjn.2020.185. (4) (2015) 699–730, https://doi.org/10.1007/s13311-015-0377-3.
[96] V.M. Ravindra, P.J. Karas, T.T. Lazaro, et al., Epilepsy surgery in young children [109] G. Ruffolo, A. Gaeta, B. Cannata, et al., GABAergic neurotransmission in human
with tuberous sclerosis complex: a novel hybrid multimodal surgical approach, tissues is modulated by cannabidiol, Life 12 (12) (2022), https://doi.org/
Neurosurgery 92 (2) (2023) 398–406, https://doi.org/10.1227/ 10.3390/life12122042.
neu.0000000000002214. [110] Y.N. Lamb, Ganaxolone: first approval, Drugs 82 (8) (2022) 933–940, https://doi.
[97] K. Pearsson, M. Compagno-Strandberg, E.A. Eklund, O. Rask, K. Källén, org/10.1007/s40265-022-01724-0.
Satisfaction and seizure outcomes of epilepsy surgery in tuberous sclerosis: a [111] S. Amin, A.A. Mallick, H. Edwards, et al., The metformin in tuberous sclerosis
Swedish population-based long-term follow-up study, Seizure 103 (2022) 39–45, (MiTS) study: a randomised double-blind placebo-controlled trial,
https://doi.org/10.1016/j.seizure.2022.10.011. EClinicalMedicine 32 (2021), 100715, https://doi.org/10.1016/j.
[98] M.A. Stellon, K. Cobourn, M.T. Whitehead, N. Elling, W. McClintock, C. eclinm.2020.100715.
O. Oluigbo, “Laser and the Tuber”: thermal dynamic and volumetric factors [112] W. Löscher, P. Klein, New approaches for developing multi-targeted drug
influencing seizure outcomes in pediatric subjects with tuberous sclerosis combinations for disease modification of complex brain disorders. Does epilepsy
undergoing stereoencephalography-directed laser ablation of tubers, Child’s prevention become a realistic goal? Pharmacol. Ther. 229 (2022), 107934
Nerv. Syst. 35 (8) (2019) 1333–1340, https://doi.org/10.1007/s00381-019- https://doi.org/10.1016/j.pharmthera.2021.107934.
04255-4. [113] C.-C. Wei, J.-N. Sheu, J.-T. Liu, S.-H. Yang, I.-C. Chou, J.-D. Tsai, Trend of seizure
[99] X. Tong, X. Wang, L. Qin, et al., Vagus nerve stimulation for drug-resistant remission in patients with tuberous sclerosis complex: a retrospective medical
epilepsy induced by tuberous sclerosis complex, Epilepsy Behav. 126 (2022), review, J. Chin. Med. Assoc. 81 (8) (2018) 724–728, https://doi.org/10.1016/j.
108431, https://doi.org/10.1016/j.yebeh.2021.108431. jcma.2018.02.001.
[100] S. Hajtovic, M.A. LoPresti, L. Zhang, K.A. Katlowitz, D.J. Kizek, S. Lam, The role
of vagus nerve stimulation in genetic etiologies of drug-resistant epilepsy: a meta-

34

You might also like