You are on page 1of 11

Epilepsy Research 172 (2021) 106588

Contents lists available at ScienceDirect

Epilepsy Research
journal homepage: www.elsevier.com/locate/epilepsyres

Review article

The role of inflammatory mediators in epilepsy: Focus on developmental


and epileptic encephalopathies and therapeutic implications
Alessandro Orsini a, 1, Thomas Foiadelli b, *, 1, Giorgio Costagliola a, Alexandre Michev b,
Rita Consolini a, Federica Vinci b, Diego Peroni a, Pasquale Striano c, d, Salvatore Savasta b
a
Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, Via Roma 67, 56126 Pisa, Italy
b
Pediatric Clinic, IRCCS Policlinico San Matteo Foundation, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy
c
Paediatric Neurology and Muscular Diseases Unit, “G. Gaslini” Institute, Via Gaslini 5, 16147 Genova, Italy
d
Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Via Gaslini 5, 16147 Genova, Italy

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

Keywords: In recent years, there has been an increasing interest in the potential involvement of neuroinflammation in the
Neuroinflammation pathogenesis of epilepsy. Specifically, the role of innate immunity (that includes cytokines and chemokines) has
Epilepsy been extensively investigated either in animal models of epilepsy and in clinical settings. Developmental and
Cytokine
epileptic encephalopathies (DEE) are a heterogeneous group of epileptic disorders, in which uncontrolled
Chemokine
West syndrome
epileptic activity results in cognitive, motor and behavioral impairment. By definition, epilepsy in DEE is poorly
Steroid controlled by common antiepileptic drugs but may respond to alternative treatments, including steroids and
immunomodulatory drugs. In this review, we will focus on how cytokines and chemokines play a role in the
pathogenesis of DEE and why expanding our knowledge about the role of neuroinflammation in DEE may be
crucial to develop new and effective targeted therapeutic strategies to prevent seizure recurrence and devel­
opmental regression.

1. Introduction degrees of response (Nariai et al., 2018; Shandra et al., 2017). During the
last decade, there has been an increasing interest in the potential
Developmental and epileptic encephalopathies (DEE) are a hetero­ involvement of inflammation in the pathogenesis of epilepsy, with
geneous group of epileptic disorders, in which the epileptic activity in­ particular attention to DEE. Inflammation is a non-specific biological
terferes with the normal neuronal development, resulting in cognitive, defense response against different mechanisms of injury and is part of
motor, and behavioral impairment, with a poor prognosis (Kalser and the innate immune response. Inflammation involves both cells (e.g.
Cross, 2018). Different epileptic syndromes with various etiologies are macrophages and dendritic cells) and soluble inflammatory mediators
comprised among DEE, including West syndrome (WS), Epilepsy with (e.g. cytokines and chemokines) and can induce activation of the
myoclonic-atonic seizures, Landau-Kleffner, Dravet, Lennox-Gastaut adaptive immunity (Skaper et al., 2018). In the central nervous system
syndromes, and others (Helbig and Tayoun, 2016). (CNS), neuroinflammation is mostly dependent on the activation of
Epilepsy in DEE is drug-resistant, and alternative treatment strate­ microglial cells and astrocytes, the main cellular effectors of the cere­
gies include adrenocorticotropic hormone (ACTH), high-dose cortico­ brospinal innate immune system (Shandra et al., 2017; Graeber et al.,
steroids, neurostimulation, ketogenic diet, and surgery, with various 2011).

Abbreviations: ACTH, adrenocorticotropic hormone; BBB, blood-brain barrier; CD, cluster of differentiation; CNS, central nervous system; COX-2, cyclooxygenase-
2; CRH, corticotropin-releasing hormone; CSF, cerebrospinal fluid; DEE, developmental and epileptic encephalopathies; ESES, encephalopathy with status epilepticus
during sleep; FIRES, febrile infection-related epilepsy syndrome; GABA, gamma-aminobutyric acid; GC, glucocorticoids; HMGB1, high mobility group box 1; IFN,
interferon; IL, interleukin; IL-1RA, interleukin-1 receptor antagonist; NF-κB, nuclear factor κB; NMDA, N-methyl-D-aspartate; non-WS-DEE, developmental and
epileptic encephalopathies other than West Syndrome; PCDH19, protocadherin-19; RE, Rasmussen encephalitis; STAT3, signal transducer and activator of tran­
scription 3; TGF, transforming growth factor; Th17, T-helper 17; TLR, toll-like receptors; TNF, tumor necrosis factor; WS, West syndrome.
* Corresponding author.
E-mail address: t.foiadelli@smatteo.pv.it (T. Foiadelli).
1
Shared co-first authors.

https://doi.org/10.1016/j.eplepsyres.2021.106588
Received 14 December 2020; Received in revised form 28 January 2021; Accepted 16 February 2021
Available online 18 February 2021
0920-1211/© 2021 Elsevier B.V. All rights reserved.
A. Orsini et al. Epilepsy Research 172 (2021) 106588

Specifically, the role of innate immunity (including cytokines, che­ the role of neuroinflammation in the pathogenesis of seizures, and the
mokines, and their molecular pathways) has been extensively studied in etiology of other epilepsies and DEE has been extensively studied in the
animal models of epilepsy and epileptic patients, with a focus on West last years (Vezzani et al., 2013; Ravizza et al., 2011; Vitaliti et al., 2019).
syndrome (WS). However, the molecular mechanisms of its involvement The association between inflammation and seizures is complex and not
in the process of epileptogenesis, as well as its role in other forms of DEE, unidirectional: seizures can cause neuroinflammation and, conversely,
are yet to be fully clarified. There is only a limited number of studies systemic or regional inflammation can contribute to the development
analyzing neuroinflammation in both DEE and other epilepsies, and and persistence of seizures (Vezzani et al., 2013, 2008; Xanthos and
studies directly comparing the degree of inflammation between Sandkuhler, 2014; Wilson, 2015) (Fig. 1).
childhood-onset epilepsies with different etiology are lacking (Kagita­ In the brain tissue of rodents with epilepsy, high levels of Interleukin
ni-Shimono et al., 2021; Choi et al., 2009b). However, the research on (IL)-1β, Tumor Necrosis Factor (TNF)-α, and IL-6 have been demon­
the role of inflammation in the pathogenesis of DEE has a peculiar strated by different authors (Vezzani et al., 2008; De Simoni et al., 2000;
clinical relevance. This priority derives from the previous scientific Vezzani et al., 1999; Minami et al., 1990; Balosso et al., 2008; Marcon
works, which have demonstrated a significant elevation of different et al., 2009). Moreover, murine models of epilepsy evidenced that
inflammatory markers in children with DEE, and by the therapeutic inflammation occurs before the development of seizures, suggesting a
response to ACTH and corticosteroids, which act also (but not exclu­ direct role of inflammation in the complex process of epileptogenesis
sively) through anti-inflammatory mechanisms, as showed by children (Vezzani et al., 2013; De Simoni et al., 2000; Ravizza et al., 2008). On
suffering from WS and other DEE (Shandra et al., 2017). As a conse­ the other hand, the electrical induction of seizures in rodents is followed
quence, there is an increasing interest in the identification of new by the release of pro-inflammatory cytokines, including IL-1β, TNF-α,
therapeutic approaches targeting inflammation for the treatment of IL-6, and other mediators of inflammation, such as the danger molecule
drug-resistant epilepsy and, potentially, for the improvement of the high mobility group box 1 (HMGB1) (Vezzani et al., 2013; Heida and
long-term neurodevelopmental outcome. Pittman, 2005). Among cytokines, IL-1β seems to have a major role in
We review the available evidence for the association between the pathogenesis of seizures and epileptogenesis. Intrathecal adminis­
inflammation and epileptogenesis in DEE. We will focus on the role of tration of IL-1β causes the exacerbation of kainate and
cytokines and chemokines and describe pathogenic mechanisms and bicuculline-induced seizures (Vezzani et al., 1999), and a seizure
potential therapeutic strategies targeting inflammation in these threshold reduction in experimental models of febrile seizures (Heida
conditions. and Pittman, 2005; Dube et al., 2005).
Neuroinflammation is extremely difficult to study in vivo. The use of
2. Inflammation and epilepsy imaging is increasing but, currently, its clinical application is not yet
defined. In a study by Chugani and Kumar, the use of translocator pro­
Our knowledge of the molecular and immunological mechanisms tein (TPSO)-PET, which evidences the microglial activation, demon­
responsible for immune-mediated epileptic encephalopathies (Greco strated a reduction in the uptake of the radioligand in children with WS
et al., 2016) is increasing, along with the evidence of the efficacy of after the treatment with corticosteroids, suggesting that this method
immunomodulatory treatments in these conditions. On the other hand, could be a promising tool to dynamically study neuroinflammation

Fig. 1. Inflammation and epileptogenesis: molecular mechanisms.


Neuroinflammation contributes to epileptogenesis through several mechanisms. HMGB1, released after neuronal or glial injury, is responsible for the activation of
TLR-4, which activates the inflammasome complex, thus causing the production of other pro-inflammatory mediators. Moreover, HMGB1 promotes chemotaxis and
participates in BBB dysfunction, causing an enhancement of the inflammatory response. IL-1β and TGF-β cause increased activity of glutamate, responsible for both
epileptogenesis and neuronal toxicity, while TNF-α, produced also after the interaction between CXCL12 and CXCL4, acts on both glutamatergic and GABA-ergic
response. Concerning chemokines, CCL2 has an important role in promoting the production of IL-1β, causing neuronal cell death and altering calcium signaling,
while CX3CL1 negatively influences GABA-ergic activity. Finally, the BB dysfunction and the epileptic activity itself are enhancers of neuroinflammation.
BBB: blood-brain barrier; HMGB1: high mobility group box 1; IL: interleukin, TNF: tumor necrosis factor; TGF: transforming growth factor.

2
A. Orsini et al. Epilepsy Research 172 (2021) 106588

(Chugani and Kumar, 2020). However, the analysis of serum or cere­ 3. Cytokines and chemokines: role in neuroinflammation and
brospinal fluid (CSF) cytokines represents the most widely used instru­ epileptogenesis
ment to analyze the presence of inflammation and its molecular features
in epileptic patients. Studies on patients with epilepsy showed high Cytokines represent a wide family of soluble mediators implicated in
serum and CSF levels of several pro-inflammatory cytokines (Liba et al., inflammatory and immune responses. They regulate cellular growth,
2019; Choi et al., 2009a; Mao et al., 2013; Aronica and Crino, 2011), differentiation, and activity (Vilcek and Feldmann, 2004), and play a
including IL-1β (with consequently altered IL-1β/IL-1 Receptor Antag­ critical role even in the immune homeostasis of the brain. As distinct
onist [RA] ratio), IL-6, IL-17, TNF-α, Transforming Growth Factor cytokines may be secreted by different cellular sources and have vari­
(TGF)-β and Interferon (IFN)-γ (Vitaliti et al., 2019; de Vries et al., 2016; able biological effects, they can induce epilepsy through multiple mo­
Gao et al., 2017). When compared to healthy controls, patients with lecular mechanisms.
focal epilepsy show increased activation of different inflammatory Toll-like receptors (TLR) are implicated in the production of a wide
pathways, including serum Nuclear Factor kB (NF-kB,) (Ulusoy et al., range of cytokines following the recognition of exogenous or endoge­
2020). Other in vivo studies, performed on brain tissue resected from nous ligands able to activate the immune response (Vidya et al., 2018).
patients with temporal lobe epilepsy, showed an up-regulation of genes IL-1β, one of the main cytokines involved in epileptogenesis, is released
associated with inflammation (Aronica and Gorter, 2007). The associ­ after the activation of TLR4, which is triggered by multiple typologies of
ation between inflammation and epileptogenesis is also supported by a ligands, including lipopolysaccharides (Kigerl et al., 2007; Rossol et al.,
recent study by Numis et al., which demonstrated that high levels of 2011). When TRL4 binds its ligands, the molecular complex of the
pro-inflammatory cytokines in newborns with neonatal encephalopathy inflammasome is activated and causes the release of the soluble form of
are associated with an increased risk of developing epilepsy later in life IL-1β and HMGB1 with a caspase-1-mediated mechanism (Diamond
(Numis et al., 2019). Recently, the association between serum levels of et al., 2015; Yang et al., 2015).
cytokines, in particular IL-6, and disease activity in patients with en­ IL-1β can participate in the development of seizures and epilepto­
cephalopathy with electrical status epilepticus during sleep (ESES) was genesis through the influence on the calcium influx across the N-methyl-
demonstrated (van den Munckhof et al., 2016). Inflammation has also D-aspartate (NMDA) receptor, the reduction of glutamate uptake by
been associated with drug-resistance. Patients with drug-resistant epi­ astrocytes, and the increase of glutamate release by glial cells (Vitaliti
lepsy have higher serum levels of IL-1β, IL-6, and reduced IL-1RA et al., 2019; Kigerl et al., 2007; Hu et al., 2000). These mechanisms
compared to controls (Hulkkonen et al., 2004). The molecular mecha­ cause an enhanced effect of glutamate that induces neuronal excitation
nism responsible for drug-resistance could be partly dependent on and potential excitotoxicity leading to a reduction of the seizure
altered expression of the P-glycoprotein, which mediates drug efflux threshold. Consequently, the TLR4-IL-1β axis appears to be central in the
through the blood-brain barrier (BBB) (Roberts and Goralski, 2008), in inflammation-mediated epileptogenesis, as confirmed by the reduced
addition to the neuronal excitability that is consequent to an uncon­ epileptic activity in TLR4 knockout animal models of epilepsy (Iori et al.,
trolled inflammation (Vitaliti et al., 2019). 2013).
Several additional markers of inflammation increase during the ictal Preclinical studies also highlighted the role of HMGB1 (Maroso et al.,
phase, including nitric oxide, prostaglandins, produced through the 2010; Ravizza et al., 2018), which is similarly involved in the TLR4-
activation of the inducible enzyme cyclooxygenase-2 (COX-2), classical IL-1β axis, and potentially implicated in the genesis and maintenance of
complement pathway, the plasminogen system, and metalloproteases seizures. HMGB1 binds TLR4, activating the pro-inflammatory NF-κB
(Vezzani et al., 2013; Sayyah et al., 2003; Riazi et al., 2010; Shimada and pathway, and can play a chemoattractant action on leukocytes by
Takemiya, 2014; Aronica et al., 2007; Iyer et al., 2010; Quirico-Santos generating a molecular complex with the chemokine CXCL12. HMGB1
et al., 2013). also interferes with the function of the NMDA receptors (Maroso et al.,
BBB alterations play an important role in the brain’s immune ho­ 2011a). These pieces of evidence suggest that HMGB1 may enhance
meostasis by affecting its function and permeability (Kovacs et al., neuroinflammation through TLR4-mediated and TLR4-independent
2012). Inflammatory mediators enhance BBB permeability, and che­ pathways, and directly modulate neurotransmitter activity. Despite
mokines can promote the adhesion and recruitment of leukocytes. BBB the absence of specificity for the CNS, HMGB1 is strictly involved in the
leakage can be the result of epileptic activity but also participates in its inflammatory events related to epilepsy, thus representing an intriguing
progression, as evidenced by several studies on human and animal brain target for further therapies (Vitaliti et al., 2019; Yang et al., 2015;
tissue (van Vliet et al., 2007; Oby and Janigro, 2006). The mechanism Maroso et al., 2011b). On the other hand, IL-10 inhibits IL-1β produc­
underlying the seizure-associated BBB dysfunction involves both the tion, suggesting a protective role against the progression of epilepto­
direct action of pro-inflammatory cytokines and the activation of mast genesis (Sun et al., 2019).
cells during inflammation (Vezzani et al., 2013; Oby and Janigro, 2006; Noteworthy, other cytokines can influence seizure development
Suemitsu et al., 2010). Among the consequences of an altered BBB, there through non-inflammatory mechanisms, such as the production and
is a rise in brain concentration of albumin, which activates the tran­ signaling of neurotransmitters. TNF-α reduces the expression of GABA-A
scription signaling related to TGF-β (Friedman et al., 2009) and con­ receptors on neuronal membranes (Viviani et al., 2007), lowering the
tributes to the maintenance of inflammation and epileptogenesis seizure threshold. Moreover, and similarly to TGF-β, it influences the
(Cacheaux et al., 2009). release and the response to glutamate (Bezzi et al., 2001; Diniz et al.,
Finally, it is important to consider that cytokines and the other me­ 2014; Bae et al., 2011).
diators of neuroinflammation play a physiological role in normal neu­ IL-6 has a complex role in the pathogenesis of several CNS diseases,
rodevelopment, being involved in neurogenesis, synaptic function, and its role in the brain inflammatory response is not clear. IL-6 is
plasticity, brain modeling, and other functions (Vezzani et al., 2013; secreted not only by immune cells but also by microglial cells, astro­
Boulanger, 2009). It is, therefore, possible that enhanced inflammation cytes, and occasionally even by neurons (Munoz-Fernandez and Fresno,
in the early stages of brain maturation could affect neuronal develop­ 1998). Additionally, it can be released by perivascular and brain endo­
ment. This intriguing hypothesis could partly explain the association thelial cells in response to inflammatory stimuli and neurotropic in­
between DEE and their numerous neurodevelopmental comorbidities. fections (Jirik et al., 1989; Fabry et al., 1993; Joseph et al., 1993). Some
However, further studies are needed to confirm this hypothesis and to studies reported raised IL-6 levels in the serum and CSF after tonic-clonic
clarify the role of inflammation in epileptogenesis and the influence of seizures (Liimatainen et al., 2009), although it is not known whether
inflammation control on the progression of cognitive, developmental IL-6 can directly be responsible for epileptogenesis. IL-6 can also in­
and non-epileptic comorbidities. crease the production of ACTH, thus highlighting the complex in­
teractions between this cytokine, the endocrine system, and CNS (Akira

3
A. Orsini et al. Epilepsy Research 172 (2021) 106588

et al., 1993). Finally, IL-6 represents a target of IL-17 and is involved in resected brain tissue of patients with epilepsy (mainly temporal lobe
the positive feedback that drives the activation of T-helper 17 (Th17) epilepsy) (Choi et al., 2009a; Lee et al., 2007; van Gassen et al., 2008;
cells (Miossec et al., 2009). It is well known that Th17 cells play a key Wu et al., 2008; Arisi et al., 2015).
role in the immune response against pathogens at mucosal and epithelial
surfaces (Munoz-Fernandez and Fresno, 1998). The IL-17/Th17 4. Understanding the link between neuroinflammation and
pathway is involved in the pathogenesis of several autoimmune condi­ developmental epileptic encephalopathies
tions including inflammatory and demyelinating diseases such as mul­
tiple sclerosis and Guillain-Barré syndrome (Matusevicius et al., 1999; Li 4.1. The paradigm of West syndrome
et al., 2012). IL-17 produces tissue inflammation and blood-brain barrier
disruption in in-vitro studies (Kebir et al., 2007). Elevated CSF and West syndrome is an infantile form of epileptic encephalopathy. WS
serum concentrations of IL-17 are detectable in patients with epilepsy of is clinically defined by a triad of a) electroencephalographic abnor­
unknown origin, and there may be a correlation between IL-17 levels malities, b) epileptic spasms, c) neurodevelopmental arrest and regres­
and seizure burden (Miossec et al., 2009) that could be linked to a BBB sion (Kato, 2006). Although clinically homogeneous, WS has different
dysfunction (Kebir et al., 2007). causing factors that are either hereditary or acquired (Baram, 1993;
Chemokines are a subclass of cytokines that are implicated in cellular Tekgul et al., 1999). Still, in the majority of patients, the exact etiology
homing and trafficking. These are small proteins (8− 12 kDa), classified remains unknown (Frost and Hrachovy, 2005) and the overall mecha­
into four subfamilies according to the characteristics of their N-terminal nisms underlying the progression to severe epilepsy and cognitive
region: CXC, C, CC, Cx3C (Zlotnik and Yoshie, 2012; Charo and Ran­ regression are not fully understood. The use of ACTH and glucocorti­
sohoff, 2006). The interaction between chemokines and their receptor coids (GC) has proven its efficacy in WS by reducing epileptic spasms
participates in a large number of processes of the immune and inflam­ and improving background EEG activity (Shumiloff et al., 2013; Tsuji
matory response, including leukocyte recruitment to the injured size, et al., 2007). Several authors have long speculated on the possible role of
integrin-mediated endothelial adhesion, tissue repair, and lymphocyte the hypothalamic-pituitary-adrenal axis in the pathogenesis of WS. As
migration to lymphoid stations (Moser and Loetscher, 2001). Chemo­ evidenced in infant animal models, the Corticotropin-Releasing Hor­
kines and their receptors are widely represented in the central nervous mone (CRH) may induce epileptogenic activity and cause brain damage
system, mainly expressed by glial, neuronal, and vascular endothelial leading to cognitive impairment (Baram and Schultz, 1991). This effect
cells (Le Thuc et al., 2015). Moreover, chemokine concentrations is thought to be partly dependent on a reduction of circulating IGF-1
significantly vary following several pathological conditions (Milenkovic levels, which causes synaptic dysfunction (Vitaliti et al., 2019). ACTH
et al., 2019; Jaerve and Muller, 2012; Biber et al., 2002). In the attempt is thought to influence epileptic activity by binding the melanocortin
to identify potential therapeutic targets, their role in neuroinflammation receptors in the amygdala and reducing the secretion of CRH (Brunson
and immunomodulation has been investigated in different neurological et al., 2001, 2002). Other proposed mechanisms for the action of ACTH
diseases, including ischemic and hemorrhagic stroke, demyelinating are represented by its influence on IGF-1 production (Vitaliti et al.,
disorders, and progressive neurodegenerative pathologies such as Alz­ 2019) and GABA homeostasis. Indeed, studies have demonstrated that
heimer’s disease and Parkinson’s diseases (Ramesh et al., 2013; Gual­ ACTH can positively influence GABAA receptor functioning, mainly
tierotti et al., 2017; De Luca et al., 2018; Minami et al., 2006; DiSano through the stimulation of the secretion of adrenal glucocorticoids and
et al., 2019; Mracsko and Veltkamp, 2014; Zuena et al., 2019; Guedes mineralocorticoids (Rogawski and Reddy, 2002). Of note, GABA-ergic
et al., 2018; Bagheri et al., 2018; Lee et al., 2019). Studies have shown function and hypothalamus-pituitary-adrenal axis are linked in a bidi­
that chemokines are not only involved in neuroinflammation but also rectional way, as GABA has been shown to inhibit the axis at both hy­
directly interact with neuropeptides and neurotransmitters, thus pothalamic and pituitary levels (Giordano et al., 2006). Also, since
modulating neuronal activity (Le Thuc et al., 2015; Rostene et al., 2011; ACTH induces the synthesis and release of GC, several studies have
Adler and Rogers, 2005; Cardona et al., 2008). focused on its steroid-related immunomodulatory properties, following
Different murine models have highlighted the molecular mecha­ the hypothesis that WS develops within an inflammatory process. It is
nisms in which chemokines contribute to epileptogenesis. CCL2 unclear whether inflammation in WS is the result of the same
expression is enhanced after seizure induction with pilocarpine or tissue-damaging events that determine the disease or may represent it­
kainite (Foresti et al., 2009; Manley et al., 2007). In mice with self as the key trigger of the condition. Therefore, several studies have
kainate-induced seizures, the activation of the CCL2-receptor (CCR2) attempted to verify whether the therapeutic effects of ACTH in WS are
resulted in an induction of the STAT3 signaling pathway, implicated in mainly determined by its anti-inflammatory activity.
neuronal cell death, and the production of IL-1β (Tian and Peng, 2017). ACTH exerts anti-inflammatory processes through its influence on
CCL2 also triggers seizures by altering intracellular calcium signaling cytokines and immune cell activity, especially on lymphocytes (Talaber
and enhancing postsynaptic currents in the hippocampus (Zhou et al., et al., 2015). Table 1 resumes the main clinical studies that investigated
2011; Banisadr et al., 2005). the effects of ACTH on the immune system homeostasis and activation in
Differently, the interaction between CXCL12 and CXCL4 can induce patients with WS, focusing on a) the alteration in the baseline cytokines
TNF-α secretion by microglial cells, influencing the synaptic function, homeostasis in WS patients compared to controls (Liu et al., 2001;
and enhancing glutamate release (Lee et al., 2007). In the brain of epi­ Montelli et al., 2003; Tekgul et al., 2006; Haginoya et al., 2009; Ture
lepsy animal models, high levels of CCR5 (which binds CCL3, CCL4, and et al., 2016); b) the variation of composition and concentration of
CCL5) have been reported, suggesting a potential pathogenic role in the circulating and CSF cytokines after treatment with ACTH (Ohya et al.,
complex history of epileptogenesis (Cerri et al., 2017; van Gassen et al., 2009; Shiihara et al., 2010; Yamanaka et al., 2010, 2018; Dias De Sousa
2008). Finally, the chemokine CX3CL1/fractalkine can influence and et al., 2012).
modulate GABAergic inhibitory functions in patients with mesial tem­ Liu et al. evidenced higher serum levels of IL-2, TNF-α, and IFN-α in
poral lobe epilepsy through its receptor CX3CR1, expressed by micro­ WS children when compared to controls. Likewise, Türe et al. demon­
glial cells (Roseti et al., 2013), suggesting that CX3CL1 may have strated that serum levels of IL-6 and IL-17 were higher than controls in a
anti-seizure activity and a protective role in epileptogenesis. small cohort of WS patients (Ture et al., 2016), while others evidenced
Studies on humans have shown promising results and further reduced CSF baseline levels of IL-6 in a group of WS patients compared
stressed the role of chemokines in epileptogenesis (Fabene et al., 2010). to controls (Tekgul et al., 2006). The role of IL-6, IL-17, and other cy­
It is now well known that CCL2, CCL3 and CXCR4 (the tokines in WS still needs to be clarified, but the fact that many circu­
CXCL12-receptor), which are fundamental in driving the leukocyte lating cytokines (including IL-17) decrease after the administration of
trafficking towards inflamed areas, are highly expressed in the surgically ACTH in patients with WS may prompt research toward new therapeutic

4
A. Orsini et al. Epilepsy Research 172 (2021) 106588

Table 1
Available studies on serum cytokines and lymphocyte subsets in West syndrome at baseline (a) and their variation before/after ACTH treatment (b).

N. of N. of Results
Authors Year Main parameters studied
patients controls Increased Decreased

Liu et al. 2001 23 15 Serum cytokines IL-2, TNF-α, IFN-α


Peripheral lymphocyte subsets; CD1+, CD3+ cells, CD4+ cells and
Montelli et al. 2003 33 20 CD8+ cells
CD4+/CD8+ ratio CD4+/CD8+ ratio
a) Tekgul et al. 2006 12 12 IL-6 in CSF IL-6
Haginoya et al. 2009 24 13 Serum IL-1β, IL-1RA, IL-6 and TNF-α IL-1RA
Serum IL-1β, IL-2, IL-6, IL-17a, IL-23 and
Türe et al.* 2016 20 20 IL-6, IL-17a
TNF-α
Peripheral lymphocyte subsets; CD4+/
Ohya et al. 2008 18 0 CD4+ cells, CD4+/CD8+ ratio
CD8+ ratio
Pre ACTH, WS group vs.
controls: CD3+ CD25+ cells,
Pre ACTH, WS group vs. controls:
Peripheral lymphocyte subsets; serum CD19+ CD95+ cells;
Shiihara et al. 2010 76 26 IL-1RA, IL-5, IL-6, IL-15,
cytokines, chemokines, growth factors. Post ACTH, WS group: CD3+
eotaxin, bFGF, IP-10
b) cells, CD4+ cells, CD4+/
CD8+ ratio
Yamanaka et al. 2011 13 0 IL-1RA, IL-1β IL-1RA
Dias de Sousa et al. 2012 4 0 IL-1β, IL-5, IL-10, TNF-β and INF-γ in CSF IL-1β, INF-γ
IL-5, IL-9, IL-17, PDGF, bFGF,
Serum cytokines, chemokines, growth
Yamanaka et al. 2018 5 0 INF-γ, IP-10, CCL2, CCL3 and
factors and TNF-α.
CCL4
*
= this study compared inflammatory features before and after ACTH treatment, but found no significant differences. Therefore, only baseline WS parameters,
compared to controls, were considered (a). ACTH: adrenocorticotropic hormone; CSF: cerebrospinal fluid; IL-1RA: interleukin-1 receptor antagonist; INF: interferon;
TNF: tumor necrosis factor; WS: West syndrome.

targets (Yamanaka et al., 2018). Some studies suggested that peripheral 1992), protocadherin-19 female epilepsy (PCDH19-FE) (Higurashi et al.,
lymphocytes can be a parameter for evaluating the perturbation in the 2015), have shown good clinical responses to corticosteroids, in terms of
immune asset of patients with WS (Montelli et al., 2003) and to identify reduction in seizure frequency and seizure freedom rate in different
a possible immunomodulatory role of ACTH (Ohya et al., 2009; Shiihara case-reports and case-series. On the other hand, the efficacy of cortico­
et al., 2010) (Table 1). In these studies, baseline levels of CD3+ CD25+ steroids was less clear in other forms of epilepsy, such as
cells and CD19+ CD95+ cells were reduced in patients with WS Myoclonic-Atonic Epilepsy (Doose Syndrome) (Oguni et al., 2002;
compared to controls. After ACTH administration, there was a signifi­ Bakker et al., 2015) and Dravet syndrome. In a cohort of 6 patients with
cant reduction of CD3+ cells, CD4+ cells, and CD4+/CD8+ ratio in WS Dravet syndrome treated with prednisone by Sinclair, only two patients
patients (Ohya et al., 2009). These data may suggest a decreased became seizure-free, two relapsed after an initial improvement (one
T-regulatory activity with reduced B-cells apoptosis in WS patients and a showing a 50 % decrease in seizure frequency after relapse), while the
T cell inactivation as a result of ACTH administration. other two showed no change in the course of their disease (Sinclair,
The hypothesis that inflammation could play a role in the patho­ 2003). O’Regan and colleagues treated one patient with ACTH, with no
genesis of WS is intriguing both for its neuropathological implications in significant changes in seizure frequency or in the EEG pattern (O’Regan
epileptogenesis, and for its obvious therapeutic prospective. Still, it is and DCH, 2008).
not to exclude that all the mentioned inflammatory alterations may be The variability in the response to the therapy, may be partially
concomitant, rather than causal, and be the expression of an underlying explained by a lack of a consensus on the medication regimens (Gupta
brain-damaging process. The role of ACTH would, therefore, be more and Appleton, 2005). Pulse intravenous methylprednisolone
likely related to its direct, steroid-independent neurotropic effects (10− 30 mg/kg/day for 3 days) or oral prednisone (1− 2 mg/kg/day for
(Rogawski and Reddy, 2002). Further researches will better clarify the 6 weeks) are often used for the induction regimen, followed by a
actual role of inflammation in the genesis of WS and the precise thera­ maintenance with oral prednisone (0.5− 2 mg/kg/day tapered over 2–6
peutic mechanism of ACTH and steroids in the treatment of DEE. weeks) (Bakker et al., 2015; Sinclair, 2003; Bast et al., 2014; Sevilla-­
Castillo et al., 2009; You et al., 2008). However, the variable therapeutic
outcomes experienced in non-WS-DEE patients may underlie the exis­
4.2. Neuroinflammation in other epileptic encephalopathies tence of different therapeutic effects of steroids, for which a
dose-dependence is been less investigated (Riikonen, 1987; Dontin et al.,
4.2.1. The response to antinflammatory therapies 2015). A recent Cochraine review stated that there is lack of evidence for
Although many efforts have been carried out to investigate the as­ the efficacy and safety of corticosteroids in non-WS-DEE (Mehta et al.,
sociation between neuroinflammation and WS or epilepsy in general, 2015).
little has been done to assess whether this could apply to other severe Among other immunomodulatory treatment options, intravenous
forms of DEE (non-WS-DEE). After the first reports on steroids and ACTH immunoglobulins (IVIG) were introduced in the treatment of epilepsy
employed as anti-epileptic drugs (Mcquarrie and Ziegler, 1942; Klein after the first empirical observations made by Pechadre and colleagues
and Livingston, 1950; Lacy and Penry, 1976), and following the clinical in 1977 (Péchadre et al., 1977). IVIG are widely used for the treatment
evidence of their anticonvulsant activity in drug-resistant epilepsies of several immune-mediated diseases of the CNS, in which they act as a
(Vezzani et al., 2011), the use of corticosteroids has been progressively powerful modulator of both humoral and cell-mediated responses
extended from WS to non-WS-DEE and other forms of severe, often (Lünemann et al., 2015). Nevertheless, there is still uncertainty about
drug-resistant epilepsy characterized by the involvement of the immune their role in DEE. Studies focusing on both WS and non-WS-DEE re­
and inflammatory response. ported a variable rate of therapeutic response (Billiau et al., 2007).
Patients with Landau-Kleffner Syndrome (Sinclair and Snyder, Particular interest grew on Landau-Kleffner syndrome, in which an
2005), encephalopathy related to ESES (Buzatu et al., 2009), immune pathogenesis was strengthened by the detection of
Lennox-Gastaut Syndrome (Kurokawa et al., 1980; Charuvanij et al.,

5
A. Orsini et al. Epilepsy Research 172 (2021) 106588

autoantibodies against myelin and brain endothelial cells in a propor­ and epilepsy has led to a growing clinical interest in new immuno­
tion of patients (Nevšímalová et al., 1992). However, even if several modulatory agents for the treatment of severe forms of epilepsy. The
patients showed a good response to IVIG, prospective studies failed to adoption of Anakinra, a monoclonal IL-1 receptor antagonist, for the
confirm clear efficacy (Fayad et al., 1997; Lagae et al., 1998; Mikati and treatment of Febrile infection-related epilepsy syndrome (FIRES) (Ken­
Saab, 2000; Arts et al., 2009; Geva-Dayan et al., 2012; Geng et al., 2019) ney-Jung et al., 2016) is an interesting example of how targeted
and further randomized trials are needed to assess whether IVIG may be anti-inflammatory drugs may be used in the future to treat drug-resistant
a valuable treatment option in DEE. epilepsies.

4.2.2. The role of cytokines 5. Implications for future treatments


The involvement of cytokines in non-WS-DEE has been less exten­
sively studied compared to WS. Some authors found elevated IL-6 serum 5.1. The anti-inflammatory role of ketogenic diet in drug-resistant
levels in patients with LGS, ESES, and Dravet syndrome, indicating that epilepsy
recurrent seizures may activate inflammatory pathways leading to po­
tential neurotoxic effects and aggravating intellectual disabilities (Leh­ The anti-inflammatory effects of well-known antiepileptic therapies
timäki et al., 2011). Patients with Dravet syndrome, in particular, also such as the ketogenic diet (KD) have been largely investigated. KD is a
show a proinflammatory vaccine-responsiveness profile, with increased high-fat, low-carbohydrate, normal protein diet known since the very
production of IL-1β, IL-6, TNF-α after in vitro stimulation of peripheral first years of the ninetieth century. In the last decade, several studies
blood mononuclear cells (Auvin et al., 2018). reported the high efficacy of KD for the treatment of various causes of
To conclude, the liaison between epilepsy and neuroinflammation drug-resistant epilepsy beyond GLUT-1, such as infantile spasms, in
has been validated through studies on animal models (Vezzani et al., which KD can reduce seizure burden and improve the intellectual
2013) and assumed in DEE (Auvin et al., 2016) but the prognostic role of outcome (Sakuma et al., 2020; Nosadini et al., 2017; Dupuis et al., 2015;
neuroinflammation have been poorly investigated in non-WS-DEE. Pardo, 2020; Dressler et al., 2019; Hussain, 2018; Caraballo et al., 2013;
There is experimental evidence that neuroinflammation could influ­ Goldberg et al., 2019; Wells et al., 2020; Yu et al., 2013; Arsyad et al.,
ence brain excitability and induce long-term neurological sequelae, 2020; Wang et al., 2020; Dressler and Trimmel-Schwahofer, 2020). The
affecting crucial areas of the immature brain in a particular moment of antiepileptic mechanisms of KD are still not completely clear and
the development, or creating immune priming that would, thereby, include multiple effects of reduced cortical hyperexcitability due to
exacerbate a second inflammatory response and affect processes rele­ changes in neuronal metabolisms, synaptic function, mitochondrial ac­
vant for the development of cognitive impairment (Auvin et al., 2016). tivity, neurotransmitters production, but also changes in the gut
Major limitations, compared to WS, are the substantial lack of adequate microbiota and neuroinflammation. Recent evidence has demonstrated
animal disease models and the difficulty to provide large cohorts of the high efficacy of KD in FIRES and in new-onset refractory status
patients due to their relative rarity and the heterogeneous pathogenetic epilepticus (NORSE) supporting the hypothesis that a major contribu­
background of non-WS-DEE. tion to the anti-epileptic properties of KD may be directly linked to its
immunomodulatory effects (Sakuma et al., 2020). Multiple hypotheses
4.3. Inflammation in other severe epilepsies regarding the effect of ketosis on inflammation have been formulated in
recent years. (Sakuma et al., 2020; Nosadini et al., 2017; Dupuis et al.,
The experience with non-DEE epilepsies featured by immune and 2015; Pardo, 2020; Dressler et al., 2019; Hussain, 2018; Caraballo et al.,
inflammatory involvement helped to better characterize the mecha­ 2013) KD can modulate fever and seems to decrease peripheral
nisms underlying neuroinflammation and contributed to the knowledge inflammation and brain IL-1β expression. In KD-fed rats, decreased
on the effects of immune agents in the treatment of epilepsy. circulating amino acids result in a reduced ability to synthesize
Rasmussen encephalitis (RE) is a progressive DEE with unilateral proinflammatory-6 derivatives, with the dual effect of impairing both
hemispherical involvement characterized by a chronic and progressive body temperature and IL-1β increase (Dupuis et al., 2015). Other au­
process with histopathological features of inflammation (Pardo et al., thors reported a direct effect of KD on T-cells in KD-fed mice, that was
2014; Orsini et al., 2020). A role of humoral immunity was initially not observed in non-ketogenic high caloric density diet-fed mice. In
supposed (Rogers et al., 1994), even if later reconsidered (Mantegazza particular, KD increased γδT cell proliferation via fatty acid oxidation
et al., 2002), and recent evidence suggests rather a cell-mediated im­ (Goldberg et al., 2019). Moreover, KD seems to inhibit
mune response pathogenesis, with a major role of CD8+ cytotoxic cells nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3)
(Bien et al., 2002) and a brain tissue cytokine and chemokine profile inflammasome function and activate the hydroxycarboxylic acid 2
skewed towards a Th1 response (Owens et al., 2013). Different immu­ (HCA2) receptor, which acts on monocytes and macrophages via
nomodulatory drugs, including steroids, IVIG, and immunosuppressive β-hydroxybutyrate (Wells et al., 2020). Finally, recent studies have
therapies, have been proposed for the treatment of RE (Orsini et al., shown that KD also protects against excitotoxicity-mediated neuronal
2020, 2019). Recently, the pathogenic role of the CXR3/CXCL10 axis cell death by decreasing the cellular oxidative stress in the Krebs cycle
has been demonstrated, suggesting it as a possible therapeutic target (Yu et al., 2013).
(Mirones et al., 2013).
The role of neuroinflammation and its potential therapeutic impli­ 5.2. Anti-cytokines and anti-chemokines treatments
cations have been studied also in PCDH19-FE. Higurashi and colleagues
reported a reduction of acute clusters in five patients with PCDH19-FE The evidence on the contribution of neuroinflammation in the gen­
after corticosteroid administration. Authors did not find significant esis and perpetuation of epilepsy in DEE has led to an increased interest
brain inflammation in their patients, suggesting that the action of cor­ in potential therapeutic targets, as summarized in Table 2. In particular,
ticosteroids would rather consist in amelioration of the BBB dysfunction, the emerging role of cytokines and chemokines in the pathogenesis of
resulting -in turn- from a systemic inflammation (Higurashi et al., 2015). epilepsy has been extensively studied as a target for therapy (Vitaliti
Furthermore, an emerging role of neurosteroids as neuromodulators in et al., 2014). Treatments targeting the IL-1β /TLR4 axis are of extreme
PCDH19-FE has been recently investigated, supporting the use of ACTH clinical interest, given the role of this molecular pathway in epilepto­
to restore steroidogenesis, improve adrenal response and raise levels of genesis and the increased levels of IL-1β found in patients with WS. As
allopregnanolone, a positive modulator of GABAA receptor (Tan et al., expected, agents interfering with the IL-1β synthesis (Ravizza et al.,
2015; Trivisano et al., 2017). 2008; Maroso et al., 2011b) (such as the caspase-1 inhibitors (Vezzani
Finally, the evidence of these links between inflammatory pathways et al., 2010)) and signaling (including the anti-IL-1 receptor monoclonal

6
A. Orsini et al. Epilepsy Research 172 (2021) 106588

Table 2 seizure frequency (Zhao et al., 2017). The TLR4 pathway is another
Potential therapeutic targets in drug-resistant epilepsy and DEE: clinical and promising target for the reduction of neuroinflammation (Rahimifard
preclinical experience. et al., 2017), although there are no drugs available directed against
Target Role in Targeted- Clinical/ TLR4. Moreover, the inhibition of TGF-beta signaling prevented epi­
epileptogenesis therapy preclinical leptogenesis in animal models of BBB disruption (Shandra et al., 2017;
experience Bar-Klein et al., 2014). Clinical studies on cytokine-directed therapies
Influence on NMDA Case reports - inhuman subjects with DEE are lacking. However, biologic drugs
receptor; reduction of FIRES ( directed against IL-1, IL-6, and TNF-α have been used in patients with
glutamate uptake; Anakinra (IL-1 Kenney-Jung
epilepsy with promising results. As previously discussed, the first reports
IL-1 β increase of glutamate receptor et al., 2016;
release; enhanced antagonist) Dilena et al., of patients affected by FIRES successfully treated with anakinra have
glutamatergic effect 2019; Sa et al., shed light on a new and promising therapeutic field (Dilena et al., 2019;
with excitotoxicity 2019) Sa et al., 2019). Likewise, the administration of the anti-IL-6 monoclonal
Case series - antibody tocilizumab was effective to treat patients with refractory
Pro-inflammatory
Tocilizumab refractory
a) cytokines release
(anti-IL-6 status
status epilepticus (Jun and Lee, 2018; Gaspard, 2019). In patients with
IL-6 promotion and RE, the administration of the anti-TNF-α antibody adalimumab resulted
monoclonal epilepticus (Jun
maintenance of
antibody) and Lee, 2018; in a clinical response on seizures and, in 3/11 patients, also in an arrest
neuroinflammation
Gaspard, 2019) of the disease progression. (Lagarde et al., 2016) Interestingly, a recent
Case series -
Reduced expression of Adalimumab trial investigating the efficacy of the anti-α4-integrin antibody natali­
Rasmussen
TNF-α
GABA-A; influence on (anti-TNF-α
Encephalitis ( zumab in patients with refractory epilepsy showed a reduction of the
glutamatergic monoclonal seizure frequency, although the study did not reach its primary endpoint
Lagarde et al.,
transmission antibody)
2016) (Rizzo et al., 2020).
microRNA- Targeted-therapies directed against chemokines are currently avail­
Activation of the Experimental
146a analogue
inflammasome, and model of able for the treatment of HIV infection (Gilliam et al., 2011) and the
TLR4 (interference
release of IL-1β and epilepsy (Iori mobilization of human hematopoietic stem cells from the bone marrow
with IL-1/
HMGB1 et al., 2017)
TLR4 axis) (Yang et al., 2019), while their use in epilepsy has not been considered
Activation of NF-ĸB,
Anti-HMGB1
Experimental yet. Animal studies showed that interference with CCL2 signaling or
chemoattractant model of
HMGB1 monoclonal transcription can abrogate lipopolysaccharide-induced seizures in mice
action; interference epilepsy (Zhao
with NMDA function
antibodies
et al., 2017) (Cerri et al., 2016; Wu et al., 2019; Xu et al., 2017). In rats with
Experimental kainate-induced epilepsy, lowering the expression of CCR5 through RNA
Component of the
inflammasome, Inhibitors of
model of interfering agents reduced neuroinflammation while improving the ef­
Caspase-1 epilepsy ( ficiency of neuronal proliferation and repair (Louboutin and Strayer,
central for the IL-1 caspase 1
Vezzani et al.,
synthesis 2013). CX3CL1/fractalkine and its receptor and the CXCL12/CXCL4
2010)
Maintenance of Experimental interaction represent other potential targets for future antiepileptic ap­
Losartan
neuroinflammation; model of proaches (Roseti et al., 2013; Fabene et al., 2010).
(interference
TGF-β influence on epilepsy (
with TGF
glutamatergic Bar-Klein et al.,
b) signaling) 6. Conclusion
transmission 2014)
Promotion of Experimental
leukocyte recruitment, CCR5-RNA model of The path to understanding the role of neuroinflammation in epilepsy
integrin-mediated interfering epilepsy ( is still long. In the last few years, there has been increasing evidence of
endothelial adhesion, agents Louboutin and the involvement of inflammatory processes in the pathophysiology of
and tissue repairing; Strayer, 2013)
seizures and different forms of epilepsy, including DEE. Notably, the role
alteration of calcium miR-206
Chemokines
signaling; activation of (interference
Experimental of the innate immune system has been investigated both in vitro and in
model of
pro-inflammatory with CCL2 vivo, mainly in animal models. Yet, there are some intrinsic difficulties
epilepsy (Cerri
pathways (STAT3), signaling);
et al., 2016; Wu in studying the neuroimmune activation in patients with epilepsy,
modulation of CXCR2
et al., 2021; Xu because, in the majority of the cases, scientists only get access to the
neurotransmitters selective
activity antagonists;
et al., 2017) cerebrospinal fluid, or need to relay over non-invasive investigations
Experimental that must be further implemented in the future. Furthermore, we still
Neuroinflammatory
model of need to explore the role played by the adaptive immune system in this
Complement C5ar1 inhibitor epilepsy ( complex scenario.
enhancement
Benson et al.,
Even considering their etiopathogenic heterogeneity, taken as a
2015)
whole, DEEs constitute a unique opportunity to study the role of
FIRES: Febrile infection-related epilepsy syndrome; GABA: Gamma- inflammation in epilepsy (and vice-versa). Clinical practice has taught
aminobutyric acid; HMGB1: High mobility group box 1; IL: Interleukin; IL- us that different DEEs may share a similar clinical evolution and
1RA: Interleukin-1 receptor antagonist; NF-κB: Nuclear factor κB; NMDA: N-
response to immunomodulatory treatments (including steroids), which
methyl-D-aspartate, STAT3: Signal transducer and activator of transcription 3;
can be as effective as antiepileptic drugs in reducing seizure frequency or
TGF: Transforming growth factor; TLR4: Toll-like receptor 4; TNF: Tumor ne­
crosis factor.
aborting seizure clusters. Although the underlying molecular mecha­
nisms remain largely unknown, common clinical features may suggest
the presence of a vicious circle involving inflammation and epilepto­
antibody anakinra) are effective in reducing seizures (Vezzani et al.,
genesis within different stages (from childhood to adulthood) of brain
2013, 2000; van Vliet et al., 2018) in animal models of epilepsy. The
maturation (Striano et al., 2011).
administration of a microRNA-146a analog, which down-regulates the
Future studies on DEE should finally clarify whether targeted ther­
IL-1β /TLR4 pathway, is effective in reducing seizures and preventing
apies against neuroinflammation may be helpful in the prevention or
the progression of epileptogenesis in mice (van Vliet et al., 2018; Iori
control of seizures and developmental regression. Finally, from bedside
et al., 2017). Other strategies targeting the IL-1β/TLR axis include the
to bench, this would provide indirect insight into the physiopathological
use of HMGB1 antagonists (Musumeci et al., 2014): in animal models of
processes that promote seizures and regression in DEEs.
epilepsy, anti-HMGB1 monoclonal antibodies can significantly reduce

7
A. Orsini et al. Epilepsy Research 172 (2021) 106588

Funding details Benson, M.J., Thomas, N.K., Talwar, S., et al., 2015. A novel anticonvulsant mechanism
via inhibition of complement receptor C5ar1 in murine epilepsy models. Neurobiol.
Dis. 76, 87–97.
This research did not receive any specific grant from funding Bezzi, P., Domercq, M., Brambilla, L., et al., 2001. CXCR4-activated astrocyte glutamate
agencies in the public, commercial, or not-for-profit sectors. release via TNFalpha: amplification by microglia triggers neurotoxicity. Nat.
Neurosci. 4 (7), 702–710.
Biber, K., Zuurman, M.W., Dijkstra, I.M., Boddeke, H.W., 2002. Chemokines in the brain:
Disclosure statement neuroimmunology and beyond. Curr. Opin. Pharmacol. 2 (1), 63–68.
Bien, C.G., Bauer, J., Deckwerth, T.L., Wiendl, H., Deckert, M., Wiestler, O.D., et al.,
The Authors declare no conflict of interest. P.S. received speaker fees 2002. Destruction of neurons by cytotoxic T cells: a new pathogenic mechanism in
Rasmussen’s encephalitis. Ann. Neurol. 51, 311–318. https://doi.org/10.1002/
and participated at advisory boards for Biomarin, Zogenyx, GW Phar­ ana.10100.
maceuticals, and has received research funding by ENECTA srl, GW Billiau, A.D., Witters, P., Ceulemans, B., Kasran, A., Wouters, C., Lagae, L., 2007.
Pharmaceuticals, Kolfarma srl., Eisai. Intravenous immunoglobulins in refractory childhood-onset epilepsy: effects on
seizure frequency, EEG activity, and cerebrospinal fluid cytokine profile. Epilepsia
48, 1739–1749. https://doi.org/10.1111/j.1528-1167.2007.01134.x.
Author’s contribution Boulanger, L.M., 2009. Immune proteins in brain development and synaptic plasticity.
Neuron 64 (1), 93–109.
Brunson, K.L., Khan, N., Eghbal-Ahmadi, M., Baram, T.Z., 2001. Corticotropin (ACTH)
AO and TF conceived and designed the original study; AM, GC and acts directly on amygdala neurons to down-regulate corticotropin-releasing hormone
FV performed a systematic literature review and implemented the text gene expression. Ann. Neurol. 49 (3), 304–312.
with the tables; RC gave substantial scientific contribution for the Brunson, K.L., Avishai-Eliner, S., Baram, T.Z., 2002. ACTH treatment of infantile spasms:
mechanisms of its effects in modulation of neuronal excitability. Int. Rev. Neurobiol.
immunological implications; DP, SS and PS critically revised the 49, 185–197.
manuscript for intellectual content. PS developed this work within the Buzatu, M., Bulteau, C., Altuzarra, C., Dulac, O., Van Bogaert, P., 2009. Corticosteroids as
framework of the DINOGMI Department of Excellence of MIUR treatment of epileptic syndromes with continuous spike-waves during slow-wave
sleep. Epilepsia 50 (Suppl 7), 68–72.
2018–2022 (legge 232 del 2016). Cacheaux, L.P., Ivens, S., David, Y., et al., 2009. Transcriptome profiling reveals TGF-
beta signaling involvement in epileptogenesis. J. Neurosci. 29 (28), 8927–8935.
References Caraballo, R.H., Reyes, G., Avaria, M.F., et al., 2013. Febrile infection-related epilepsy
syndrome: a study of 12 patients. Seizure 22 (7), 553–559. https://doi.org/10.1016/
j.seizure.2013.04.005.
Adler, M.W., Rogers, T.J., 2005. Are chemokines the third major system in the brain?
Cardona, A.E., Li, M., Liu, L., Savarin, C., Ransohoff, R.M., 2008. Chemokines in and out
J. Leukoc. Biol. 78 (6), 1204–1209.
of the central nervous system: much more than chemotaxis and inflammation.
Akira, S., Taga, T., Kishimoto, T., 1993. Interleukin-6 in biology and medicine. Adv.
J. Leukoc. Biol. 84 (3), 587–594.
Immunol. 54, 1–78.
Cerri, C., Genovesi, S., Allegra, M., et al., 2016. The chemokine CCL2 mediates the
Arisi, G.M., Foresti, M.L., Katki, K., Shapiro, L.A., 2015. Increased CCL2, CCL3, CCL5,
seizure-enhancing effects of systemic inflammation. J. Neurosci. 36 (13),
and IL-1beta cytokine concentration in piriform cortex, hippocampus, and neocortex
3777–3788.
after pilocarpine-induced seizures. J. Neuroinflammation 12, 129.
Cerri, C., Caleo, M., Bozzi, Y., 2017. Chemokines as new inflammatory players in the
Aronica, E., Crino, P.B., 2011. Inflammation in epilepsy: clinical observations. Epilepsia
pathogenesis of epilepsy. Epilepsy Res. 136, 77–83.
52 (Suppl 3), 26–32.
Charo, I.F., Ransohoff, R.M., 2006. The many roles of chemokines and chemokine
Aronica, E., Gorter, J.A., 2007. Gene expression profile in temporal lobe epilepsy.
receptors in inflammation. N. Engl. J. Med. 354 (6), 610–621.
Neuroscientist 13 (2), 100–108.
Charuvanij, A., Ouvrier, R.A., Procopis, P.G., Antony, J.H., Fagan, E.R., 1992. ACTH
Aronica, E., Boer, K., van Vliet, E.A., et al., 2007. Complement activation in experimental
treatment in intractable seizures of childhood. Brain Dev. 14 (2), 102–106.
and human temporal lobe epilepsy. Neurobiol. Dis. 26 (3), 497–511.
Choi, J., Nordli Jr., D.R., Alden, T.D., et al., 2009a. Cellular injury and
Arsyad, A., Idris, I., Rasyid, A.A., et al., 2020. Long-term ketogenic diet induces
neuroinflammation in children with chronic intractable epilepsy.
metabolic acidosis, anemia, and oxidative stress in healthy Wistar rats. J. Nutr.
J. Neuroinflammation 6, 38.
Metab. 2020, 3642035 https://doi.org/10.1155/2020/3642035. Published 2020
Choi, J., Nordli Jr, D.R., Alden, T.D., DiPatri Jr, A., Laux, L., Kelley, K., et al., 2009 Dec.
Jun 19.
Cellular injury and neuroinflammation in children with chronic intractable epilepsy.
Arts, W.F.M., Aarsen, F.K., Scheltens-De Boer, M., Catsman-Berrevoets, C.E., 2009.
J. Neuroinflammation 19 (6), 38.
Landau-Kleffner syndrome and CSWS syndrome: treatment with intravenous
Chugani, H.T., Kumar, A., 2020. Neuroinflammation in children with infantile spasms: a
immunoglobulins, in: epilepsia. Epilepsia 55–58. https://doi.org/10.1111/j.1528-
prospective study before and after treatment with acthar gel (Repository
1167.2009.02221.x.
corticotropin injection). J. Child Neurol. 35 (12), 808–812.
Auvin, S., Cilio, M.R., Vezzani, A., 2016. Current understanding and neurobiology of
De Luca, C., Colangelo, A.M., Alberghina, L., Papa, M., 2018. Neuro-immune hemostasis:
epileptic encephalopathies. Neurobiol. Dis. 92, 72–89. https://doi.org/10.1016/j.
homeostasis and diseases in the central nervous system. Front. Cell. Neurosci. 12,
nbd.2016.03.007.
459.
Auvin, S., Jeljeli, M., Desnous, B., Soussi-Yanicostas, N., Dournaud, P., Sterkers, G., 2018.
De Simoni, M.G., Perego, C., Ravizza, T., et al., 2000. Inflammatory cytokines and related
Altered vaccine-induced immunity in children with Dravet syndrome. Epilepsia 59,
genes are induced in the rat hippocampus by limbic status epilepticus. Eur. J.
e45–50. https://doi.org/10.1111/epi.14038.
Neurosci. 12 (7), 2623–2633.
Bae, J.J., Xiang, Y.Y., Martinez-Canabal, A., Frankland, P.W., Yang, B.B., Lu, W.Y., 2011.
de Vries, E.E., van den Munckhof, B., Braun, K.P., van Royen-Kerkhof, A., de Jager, W.,
Increased transforming growth factor-beta1 modulates glutamate receptor
Jansen, F.E., 2016. Inflammatory mediators in human epilepsy: a systematic review
expression in the hippocampus. Int. J. Physiol. Pathophysiol. Pharmacol. 3 (1), 9–20.
and meta-analysis. Neurosci. Biobehav. Rev. 63, 177–190.
Bagheri, V., Khorramdelazad, H., Hassanshahi, G., Moghadam-Ahmadi, A., Vakilian, A.,
Diamond, C.E., Khameneh, H.J., Brough, D., Mortellaro, A., 2015. Novel perspectives on
2018. CXCL12 and CXCR4 in the peripheral blood of patients with Parkinson’s
non-canonical inflammasome activation. Immunotargets Ther. 4, 131–141.
disease. Neuroimmunomodulation 25 (4), 201–205.
Dias De Sousa, M.A., Bonatti, R.C.F., Rodrigues Jr, V., Azevedo, D.S., Santos, M.H.A.,
Bakker, D.P., Catsman-Berrevoets, C.E., Neuteboom, R.F., 2015. Effectiveness of a hybrid
Pereira, R.O.L., et al., 2012. Cytokines in cerebrospinal fluid of children with West
corticosteroid treatment regimen on refractory childhood seizures and a review of
syndrome. J. Epilepsy Clin. Neurophysiol. 18 (2), 63–66. https://doi.org/10.1590/
other corticosteroid treatments. Eur. J. Paediatr. Neurol. 19, 553–560. https://doi.
S1676-26492012000200010.
org/10.1016/j.ejpn.2015.04.006.
Dilena, R., Mauri, E., Aronica, E., et al., 2019. Therapeutic effect of Anakinra in the
Balosso, S., Maroso, M., Sanchez-Alavez, M., et al., 2008. A novel non-transcriptional
relapsing chronic phase of febrile infection-related epilepsy syndrome. Epilepsia
pathway mediates the proconvulsive effects of interleukin-1beta. Brain 131 (Pt 12),
Open 4 (2), 344–350.
3256–3265.
Diniz, L.P., Tortelli, V., Garcia, M.N., et al., 2014. Astrocyte transforming growth factor
Banisadr, G., Gosselin, R.D., Mechighel, P., Rostene, W., Kitabgi, P., Melik
beta 1 promotes inhibitory synapse formation via CaM kinase II signaling. Glia 62
Parsadaniantz, S., 2005. Constitutive neuronal expression of CCR2 chemokine
(12), 1917–1931.
receptor and its colocalization with neurotransmitters in normal rat brain: functional
DiSano, K.D., Linzey, M.R., Royce, D.B., Pachner, A.R., Gilli, F., 2019. Differential neuro-
effect of MCP-1/CCL2 on calcium mobilization in primary cultured neurons.
immune patterns in two clinically relevant murine models of multiple sclerosis.
J. Comp. Neurol. 492 (2), 178–192.
J. Neuroinflammation 16 (1), 109.
Baram, T.Z., 1993. Pathophysiology of massive infantile spasms: perspective on the
Dontin, S.O.S., Teutonico, F., Altieri, N., Balottin, U., Veggiotti, P., 2015. Corticosteroid
putative role of the brain adrenal axis. Ann. Neurol. 33 (3), 231–236.
therapy for epileptic encephalopathies other than West syndrome. J. Pediatr.
Baram, T.Z., Schultz, L., 1991. Corticotropin-releasing hormone is a rapid and potent
Epilepsy 02 (02), 097–107. https://doi.org/10.3233/PEP-13048.147.
convulsant in the infant rat. Brain Res. Dev. Brain Res. 61 (1), 97–101.
Dressler, A., Trimmel-Schwahofer, P., 2020. The ketogenic diet for infants: How long can
Bar-Klein, G., Cacheaux, L.P., Kamintsky, L., et al., 2014. Losartan prevents acquired
you go? Epilepsy Res. 164, 106339 https://doi.org/10.1016/j.
epilepsy via TGF-beta signaling suppression. Ann. Neurol. 75 (6), 864–875.
eplepsyres.2020.106339.
Bast, T., Richter, S., Ebinger, F., Rating, D., Wiemer-Kruel, A., Schubert-Bast, S., 2014.
Dressler, A., Benninger, F., Trimmel-Schwahofer, P., et al., 2019. Efficacy and tolerability
Efficacy and tolerability of methylprednisolone pulse therapy in childhood epilepsies
of the ketogenic diet versus high-dose adrenocorticotropic hormone for infantile
other than infantile spasms. Neuropediatrics 45, 378–385. https://doi.org/10.1055/
s-0034-1387817.

8
A. Orsini et al. Epilepsy Research 172 (2021) 106588

spasms: a single-center parallel-cohort randomized controlled trial. Epilepsia 60 (3), Jirik, F.R., Podor, T.J., Hirano, T., et al., 1989. Bacterial lipopolysaccharide and
441–451. https://doi.org/10.1111/epi.14679. inflammatory mediators augment IL-6 secretion by human endothelial cells.
Dube, C., Vezzani, A., Behrens, M., Bartfai, T., Baram, T.Z., 2005. Interleukin-1beta J. Immunol. 142 (1), 144–147.
contributes to the generation of experimental febrile seizures. Ann. Neurol. 57 (1), Joseph, J., Grun, J.L., Lublin, F.D., Knobler, R.L., 1993. Interleukin-6 induction in vitro
152–155. in mouse brain endothelial cells and astrocytes by exposure to mouse hepatitis virus
Dupuis, N., Curatolo, N., Benoist, J.F., Auvin, S., 2015. Ketogenic diet exhibits anti- (MHV-4, JHM). J. Neuroimmunol. 42 (1), 47–52.
inflammatory properties. Epilepsia 56 (7), e95–e98. https://doi.org/10.1111/ Jun, J.S., Lee, S.T., 2018. Tocilizumab treatment for new onset refractory status
epi.13038. epilepticus. Ann. Neurol. 84 (6), 940–945.
Fabene, P.F., Bramanti, P., Constantin, G., 2010. The emerging role for chemokines in Kagitani-Shimono, K., Kato, H., Kuwayama, R., Tominaga, K., Nabatame, S., Kishima, H.,
epilepsy. J. Neuroimmunol. 224 (1-2), 22–27. et al., 2021. Clinical evaluation of neuroinflammation in child-onset focal epilepsy: a
Fabry, Z., Fitzsimmons, K.M., Herlein, J.A., Moninger, T.O., Dobbs, M.B., Hart, M.N., translocator protein PET study. J. Neuroinflammation 18 (1), 8.
1993. Production of the cytokines interleukin 1 and 6 by murine brain microvessel Kalser, J., Cross, J.H., 2018. The epileptic encephalopathy jungle - from Dr West to the
endothelium and smooth muscle pericytes. J. Neuroimmunol. 47 (1), 23–34. concepts of aetiology-related and developmental encephalopathies. Curr. Opin.
Fayad, M.N., Choueiri, R., Mikati, M., 1997. Landau-Kleffner syndrome: consistent Neurol. 31 (2), 216–222.
response to repeated intravenous gamma-globulin doses: a case report. Epilepsia 38, Kato, M., 2006. A new paradigm for West syndrome based on molecular and cell biology.
489–494. https://doi.org/10.1111/j.1528-1157.1997.tb01740.x. Epilepsy Res. 70 (Suppl 1), S87–95.
Foresti, M.L., Arisi, G.M., Katki, K., Montanez, A., Sanchez, R.M., Shapiro, L.A., 2009. Kebir, H., Kreymborg, K., Ifergan, I., et al., 2007. Human TH17 lymphocytes promote
Chemokine CCL2 and its receptor CCR2 are increased in the hippocampus following blood-brain barrier disruption and central nervous system inflammation. Nat. Med.
pilocarpine-induced status epilepticus. J. Neuroinflammation 6, 40. 13 (10), 1173–1175.
Friedman, A., Kaufer, D., Heinemann, U., 2009. Blood-brain barrier breakdown-inducing Kenney-Jung, D.L., Vezzani, A., Kahoud, R.J., et al., 2016. Febrile infection-related
astrocytic transformation: novel targets for the prevention of epilepsy. Epilepsy Res. epilepsy syndrome treated with anakinra. Ann. Neurol. 80 (6), 939–945.
85 (2-3), 142–149. Kigerl, K.A., Lai, W., Rivest, S., Hart, R.P., Satoskar, A.R., Popovich, P.G., 2007. Toll-like
Frost Jr., J.D., Hrachovy, R.A., 2005. Pathogenesis of infantile spasms: a model based on receptor (TLR)-2 and TLR-4 regulate inflammation, gliosis, and myelin sparing after
developmental desynchronization. J. Clin. Neurophysiol. 22 (1), 25–36. spinal cord injury. J. Neurochem. 102 (1), 37–50.
Gao, F., Gao, Y., Zhang, S.J., et al., 2017. Alteration of plasma cytokines in patients with Klein, R., Livingston, S., 1950. The effect of adrenocorticotropic hormone in epilepsy.
active epilepsy. Acta Neurol. Scand. 135 (6), 663–669. J. Pediatr. 37 (5), 733–742.
Gaspard, N., 2019. A new hose to extinguish the FIRES? Epilepsy Curr. 19 (2), 86–87. Kovacs, R., Heinemann, U., Steinhauser, C., 2012. Mechanisms underlying blood-brain
Geng, J.S., Dong, J.C., Li, Y., Ni, H.J., Jiang, K., Shi, L.L., Wang, G.H., 2019. Intravenous barrier dysfunction in brain pathology and epileptogenesis: role of astroglia.
immunoglobulins for epilepsy. Cochrane Database Syst. Rev. https://doi.org/ Epilepsia 53 (Suppl 6), 53–59.
10.1002/14651858.CD008557.pub4. Kurokawa, T., Goya, N., Fukuyama, Y., Suzuki, M., Seki, T., Ohtahara, S., 1980. West
Geva-Dayan, K., Shorer, Z., Menascu, S., Linder, I., Goldberg-Stern, H., Heyman, E., syndrome and Lennox-Gastaut syndrome: a survey of natural history. Pediatrics 65
Lerman-Sagie, T., Ben Zeev, B., Kramer, U., 2012. Immunoglobulin treatment for (1), 81–88.
severe childhood epilepsy. Pediatr. Neurol. 46, 375–381. https://doi.org/10.1016/j. Lacy, Penry, J.K., 1976. Infantile Spasms. Raven Press, New York, NY.
pediatrneurol.2012.03.015. Lagae, L.G., Silberstein, J., Gillis, P.L., Casaer, P.J., 1998. Successful use of intravenous
Gilliam, B.L., Riedel, D.J., Redfield, R.R., 2011. Clinical use of CCR5 inhibitors in HIV immunoglobulins in Landau-Kleffner syndrome. Pediatr. Neurol. 18, 165–168.
and beyond. J. Transl. Med. 9 (Suppl 1), S9. https://doi.org/10.1016/S0887-8994(97)00157-4.
Giordano, R., Pellegrino, M., Picu, A., Bonelli, L., Balbo, M., Berardelli, R., Lanfranco, F., Lagarde, S., Villeneuve, N., Trebuchon, A., et al., 2016. Anti-tumor necrosis factor alpha
Ghigo, E., Arvat, E., 2006. Neuroregulation of the hypothalamus-pituitary-adrenal therapy (adalimumab) in Rasmussen’s encephalitis: an open pilot study. Epilepsia 57
(HPA) axis in humans: effects of GABA-, mineralocorticoid-, and GH-Secretagogue- (6), 956–966.
receptor modulation. Sci. World J. 6, 1–11. Le Thuc, O., Blondeau, N., Nahon, J.L., Rovere, C., 2015. The complex contribution of
Goldberg, E.L., Molony, R.D., Kudo, E., et al., 2019. Ketogenic diet activates protective γδ chemokines to neuroinflammation: switching from beneficial to detrimental effects.
T cell responses against influenza virus infection. Sci. Immunol. 4 (41), eaav2026. Ann. N. Y. Acad. Sci. 1351, 127–140.
https://doi.org/10.1126/sciimmunol.aav2026. Lee, T.S., Mane, S., Eid, T., et al., 2007. Gene expression in temporal lobe epilepsy is
Graeber, M.B., Li, W., Rodriguez, M.L., 2011. Role of microglia in CNS inflammation. consistent with increased release of glutamate by astrocytes. Mol. Med. 13 (1-2),
FEBS Lett. 585 (23), 3798–3805. 1–13.
Greco, A., Rizzo, M.I., De Virgilio, A., et al., 2016. Autoimmune epilepsy. Autoimmun. Lee, Y., Lee, S., Chang, S.C., Lee, J., 2019. Significant roles of neuroinflammation in
Rev. 15 (3), 221–225. Parkinson’s disease: therapeutic targets for PD prevention. Arch. Pharm. Res. 42 (5),
Gualtierotti, R., Guarnaccia, L., Beretta, M., et al., 2017. Modulation of 416–425.
neuroinflammation in the central nervous system: role of chemokines and Lehtimäki, K.A., Liimatainen, S., Peltola, J., Arvio, M., 2011. The serum level of
sphingolipids. Adv. Ther. 34 (2), 396–420. interleukin-6 in patients with intellectual disability and refractory epilepsy. Epilepsy
Guedes, J.R., Lao, T., Cardoso, A.L., El Khoury, J., 2018. Roles of microglial and Res. 95, 184–187. https://doi.org/10.1016/j.eplepsyres.2011.03.004.
monocyte chemokines and their receptors in regulating Alzheimer’s disease- Li, S., Yu, M., Li, H., Zhang, H., Jiang, Y., 2012. IL-17 and IL-22 in cerebrospinal fluid and
associated amyloid-beta and tau pathologies. Front. Neurol. 9, 549. plasma are elevated in Guillain-Barre syndrome. Mediators Inflamm. 2012, 260473.
Gupta, R., Appleton, R., 2005. Corticosteroids in the management of the paediatric Liba, Z., Nohejlova, H., Capek, V., Krsek, P., Sediva, A., Kayserova, J., 2019. Utility of
epilepsies. Arch. Dis. Child. 90, 379–384. https://doi.org/10.1136/ chemokines CCL2, CXCL8, 10 and 13 and interleukin 6 in the pediatric cohort for the
adc.2004.051375. recognition of neuroinflammation and in the context of traditional cerebrospinal
Haginoya, K., Noguchi, R., Zhao, Y., et al., 2009. Reduced levels of interleukin-1 receptor fluid neuroinflammatory biomarkers. PLoS One 14 (7) e0219987.
antagonist in the cerebrospinal fluid in patients with West syndrome. Epilepsy Res. Liimatainen, S., Fallah, M., Kharazmi, E., Peltola, M., Peltola, J., 2009. Interleukin-6
85 (2-3), 314–317. levels are increased in temporal lobe epilepsy but not in extra-temporal lobe
Heida, J.G., Pittman, Q.J., 2005. Causal links between brain cytokines and experimental epilepsy. J. Neurol. 256 (5), 796–802.
febrile convulsions in the rat. Epilepsia 46 (12), 1906–1913. Liu, Z.S., Wang, Q.W., Wang, F.L., Yang, L.Z., 2001. Serum cytokine levels are altered in
Helbig, I., Tayoun, A.A., 2016. Understanding genotypes and phenotypes in epileptic patients with West syndrome. Brain Dev. 23 (7), 548–551.
encephalopathies. Mol. Syndromol. 7 (4), 172–181. Louboutin, J.P., Strayer, D.S., 2013. Relationship between the chemokine receptor CCR5
Higurashi, N., Takahashi, Y., Kashimada, A., Sugawara, Y., Sakuma, H., Tomonoh, Y., and microglia in neurological disorders: consequences of targeting CCR5 on
et al., 2015. Immediate suppression of seizure clusters by corticosteroids in PCDH19 neuroinflammation, neuronal death and regeneration in a model of epilepsy. CNS
female epilepsy. Seizure 27, 1–5. https://doi.org/10.1016/J.SEIZURE.2015.02.006. Neurol. Disord. Drug Targets 12 (6), 815–829.
Hu, S., Sheng, W.S., Ehrlich, L.C., Peterson, P.K., Chao, C.C., 2000. Cytokine effects on Lünemann, J.D., Nimmerjahn, F., Dalakas, M.C., 2015. Intravenous immunoglobulin in
glutamate uptake by human astrocytes. Neuroimmunomodulation 7 (3), 153–159. neurology-mode of action and clinical efficacy. Nat. Rev. Neurol. https://doi.org/
Hulkkonen, J., Koskikallio, E., Rainesalo, S., Keranen, T., Hurme, M., Peltola, J., 2004. 10.1038/nrneurol.2014.25.
The balance of inhibitory and excitatory cytokines is differently regulated in vivo Manley, N.C., Bertrand, A.A., Kinney, K.S., Hing, T.C., Sapolsky, R.M., 2007.
and in vitro among therapy resistant epilepsy patients. Epilepsy Res. 59 (2-3), Characterization of monocyte chemoattractant protein-1 expression following a
199–205. kainate model of status epilepticus. Brain Res. 1182, 138–143.
Hussain, S.A., 2018. Treatment of infantile spasms. Epilepsia Open 3 (Suppl Suppl 2), Mantegazza, R., Bernasconi, P., Baggi, F., Spreafico, R., Ragona, F., Antozzi, C., et al.,
143–154. https://doi.org/10.1002/epi4.12264. Published 2018 Oct 23. 2002. Antibodies against GluR3 peptides are not specific for Rasmussen’s
Iori, V., Maroso, M., Rizzi, M., et al., 2013. Receptor for Advanced Glycation Endproducts encephalitis but are also present in epilepsy patients with severe, early onset disease
is upregulated in temporal lobe epilepsy and contributes to experimental seizures. and intractable seizures. J. Neuroimmunol. 131, 179–185. https://doi.org/10.1016/
Neurobiol. Dis. 58, 102–114. S0165-5728(02)00261-8.
Iori, V., Iyer, A.M., Ravizza, T., et al., 2017. Blockade of the IL-1R1/TLR4 pathway Mao, L.Y., Ding, J., Peng, W.F., et al., 2013. Interictal interleukin-17A levels are elevated
mediates disease-modification therapeutic effects in a model of acquired epilepsy. and correlate with seizure severity of epilepsy patients. Epilepsia 54 (9), e142–5.
Neurobiol. Dis. 99, 12–23. Marcon, J., Gagliardi, B., Balosso, S., et al., 2009. Age-dependent vascular changes
Iyer, A.M., Zurolo, E., Boer, K., et al., 2010. Tissue plasminogen activator and urokinase induced by status epilepticus in rat forebrain: implications for epileptogenesis.
plasminogen activator in human epileptogenic pathologies. Neuroscience 167 (3), Neurobiol. Dis. 34 (1), 121–132.
929–945. Maroso, M., Balosso, S., Ravizza, T., et al., 2010. Toll-like receptor 4 and high-mobility
Jaerve, A., Muller, H.W., 2012. Chemokines in CNS injury and repair. Cell Tissue Res. group box-1 are involved in ictogenesis and can be targeted to reduce seizures. Nat.
349 (1), 229–248. Med. 16 (4), 413–419.

9
A. Orsini et al. Epilepsy Research 172 (2021) 106588

Maroso, M., Balosso, S., Ravizza, T., Liu, J., Bianchi, M.E., Vezzani, A., 2011a. Electroencephalogr. Neurophysiol. Clin. 7, 443–447. https://doi.org/10.1016/
Interleukin-1 type 1 receptor/Toll-like receptor signalling in epilepsy: the S0370-4475(77)80049-X.
importance of IL-1beta and high-mobility group box 1. J. Intern. Med. 270 (4), Quirico-Santos, T., Nascimento Mello, A., Casimiro Gomes, A., de Carvalho, L.P., de
319–326. Souza, J.M., Alves-Leon, S., 2013. Increased metalloprotease activity in the
Maroso, M., Balosso, S., Ravizza, T., et al., 2011b. Interleukin-1beta biosynthesis epileptogenic lesion–Lobectomy reduces metalloprotease activity and urokinase-
inhibition reduces acute seizures and drug resistant chronic epileptic activity in type uPAR circulating levels. Brain Res. 1538, 172–181.
mice. Neurotherapeutics 8 (2), 304–315. Rahimifard, M., Maqbool, F., Moeini-Nodeh, S., et al., 2017. Targeting the TLR4
Matusevicius, D., Kivisakk, P., He, B., et al., 1999. Interleukin-17 mRNA expression in signaling pathway by polyphenols: a novel therapeutic strategy for
blood and CSF mononuclear cells is augmented in multiple sclerosis. Mult. Scler. 5 neuroinflammation. Ageing Res. Rev. 36, 11–19.
(2), 101–104. Ramesh, G., MacLean, A.G., Philipp, M.T., 2013. Cytokines and chemokines at the
Mcquarrie, I.A.J., Ziegler, M.R., 1942. Observations on the antagonistic effects of crossroads of neuroinflammation, neurodegeneration, and neuropathic pain.
posterior pituitary and cortico-adrenal hormones in, the epileptic subject. J. Clin. Mediators Inflamm. 2013, 480739.
Endocrinol. Metab. 2 (6), 406–410. https://doi.org/10.1210/jcem-2-6-406. Ravizza, T., Noe, F., Zardoni, D., Vaghi, V., Sifringer, M., Vezzani, A., 2008. Interleukin
Mehta, V., Ferrie, C.D., Cross, J.H., Vadlamani, G., 2015. Corticosteroids including ACTH converting enzyme inhibition impairs kindling epileptogenesis in rats by blocking
for childhood epilepsy other than epileptic spasms. Cochrane Database Syst. Rev. astrocytic IL-1beta production. Neurobiol. Dis. 31 (3), 327–333.
2015 https://doi.org/10.1002/14651858.CD005222.pub3. Ravizza, T., Balosso, S., Vezzani, A., 2011. Inflammation and prevention of
Mikati, M.A., Saab, R., 2000. Successful use of intravenous immunoglobulin as initial epileptogenesis. Neurosci. Lett. 497 (3), 223–230.
monotherapy in Landau-Kleffner syndrome. Epilepsia 41, 880–886. https://doi.org/ Ravizza, T., Terrone, G., Salamone, A., et al., 2018. High mobility group box 1 is a novel
10.1111/j.1528-1157.2000.tb00257.x. pathogenic factor and a mechanistic biomarker for epilepsy. Brain Behav. Immun.
Milenkovic, V.M., Stanton, E.H., Nothdurfter, C., Rupprecht, R., Wetzel, C.H., 2019. The 72, 14–21.
role of chemokines in the pathophysiology of major depressive disorder. Int. J. Mol. Riazi, K., Galic, M.A., Pittman, Q.J., 2010. Contributions of peripheral inflammation to
Sci. 20 (9). seizure susceptibility: cytokines and brain excitability. Epilepsy Res. 89 (1), 34–42.
Minami, M., Kuraishi, Y., Yamaguchi, T., Nakai, S., Hirai, Y., Satoh, M., 1990. Riikonen, R., 1987. Advances in therapy of infantile spasms. Current knowledge of
Convulsants induce interleukin-1 beta messenger RNA in rat brain. Biochem. actions of ACTH and corticosteroids. Brain Dev. 9 (4), 409–414.
Biophys. Res. Commun. 171 (2), 832–837. Rizzo, M., Cole, A., Faught, E., Theodore, W., Vezzani, A., Hubbard, S., et al., 2020.
Minami, M., Katayama, T., Satoh, M., 2006. Brain cytokines and chemokines: roles in A Randomized, Double-blind, Placebo Controlled, Phase 2 Study of Natalizumab As
ischemic injury and pain. J. Pharmacol. Sci. 100 (5), 461–470. Adjunctive Therapy in Adult Participants With Drug-resistant Focal Epilepsy. Abst.
Miossec, P., Korn, T., Kuchroo, V.K., 2009. Interleukin-17 and type 17 helper T cells. 764. Available at:. American Epilepsy Society https://www.aesnet.org/meetings_eve
N. Engl. J. Med. 361 (9), 888–898. nts/annual_meeting_abstracts/view/2423102.
Mirones, I., De Prada, I., Gómez, A.M., Luque, A., Martín, R., Pérez-Jiménez, M.Á, et al., Roberts, D.J., Goralski, K.B., 2008. A critical overview of the influence of inflammation
2013. A role for the CXCR3/CXCL10 axis in rasmussen encephalitis. Pediatr. Neurol. and infection on P-glycoprotein expression and activity in the brain. Expert Opin.
49, 451–457.e1. https://doi.org/10.1016/j.pediatrneurol.2013.07.019. Drug Metab. Toxicol. 4 (10), 1245–1264.
Montelli, T.C., Soares, A.M., Peracoli, M.T., 2003. Immunologic aspects of West Rogawski, M.A., Reddy, D.S., 2002. Neurosteroids and infantile spasms: the
syndrome and evidence of plasma inhibitory effects on T cell function. Arq. deoxycorticosterone hypothesis. Int. Rev. Neurobiol. 49, 199–219.
Neuropsiquiatr. 61 (3b), 731–737. Rogers, S.W., Andrews, P.I., Gahring, L.C., Whisenand, T., Cauley, K., Crain, B., et al.,
Moser, B., Loetscher, P., 2001. Lymphocyte traffic control by chemokines. Nat. Immunol. 1994. Autoantibodies to glutamate receptor GluR3 in Rasmussen’s encephalitis.
2 (2), 123–128. Science (80-) 265, 648–651. https://doi.org/10.1126/science.8036512.
Mracsko, E., Veltkamp, R., 2014. Neuroinflammation after intracerebral hemorrhage. Roseti, C., Fucile, S., Lauro, C., et al., 2013. Fractalkine/CX3CL1 modulates GABAA
Front. Cell. Neurosci. 8, 388. currents in human temporal lobe epilepsy. Epilepsia 54 (10), 1834–1844.
Munoz-Fernandez, M.A., Fresno, M., 1998. The role of tumour necrosis factor, Rossol, M., Heine, H., Meusch, U., et al., 2011. LPS-induced cytokine production in
interleukin 6, interferon-gamma and inducible nitric oxide synthase in the human monocytes and macrophages. Crit. Rev. Immunol. 31 (5), 379–446.
development and pathology of the nervous system. Prog. Neurobiol. 56 (3), Rostene, W., Dansereau, M.A., Godefroy, D., et al., 2011. Neurochemokines: a menage a
307–340. trois providing new insights on the functions of chemokines in the central nervous
Musumeci, D., Roviello, G.N., Montesarchio, D., 2014. An overview on HMGB1 inhibitors system. J. Neurochem. 118 (5), 680–694.
as potential therapeutic agents in HMGB1-related pathologies. Pharmacol. Ther. 141 Sa, M., Singh, R., Pujar, S., et al., 2019. Centromedian thalamic nuclei deep brain
(3), 347–357. stimulation and Anakinra treatment for FIRES - two different outcomes. Eur. J.
Nariai, H., Duberstein, S., Shinnar, S., 2018. Treatment of epileptic encephalopathies: Paediatr. Neurol. 23 (5), 749–754.
current state of the art. J. Child Neurol. 33 (1), 41–54. Sakuma, H., Horino, A., Kuki, I., 2020. Neurocritical care and target immunotherapy for
Nevšímalová, S., Tauberová, A., Doutlík, S., Kučera, V., Dlouhá, O., 1992. A role of febrile infection-related epilepsy syndrome. Biomed. J. https://doi.org/10.1016/j.
autoimmunity in the etiopathogenesis of Landau-Kleffner syndrome? Brain Dev. 14, bj.2020.03.009. S2319-4170(20)30038-X.
342–345. https://doi.org/10.1016/S0387-7604(12)80157-4. Sayyah, M., Javad-Pour, M., Ghazi-Khansari, M., 2003. The bacterial endotoxin
Nosadini, M., Sartori, S., Sharma, S., Dale, R.C., 2017. Immunotherapeutics in pediatric lipopolysaccharide enhances seizure susceptibility in mice: involvement of
autoimmune central nervous system disease: agents and mechanisms. Semin. proinflammatory factors: nitric oxide and prostaglandins. Neuroscience 122 (4),
Pediatr. Neurol. 24 (3), 214–228. https://doi.org/10.1016/j.spen.2017.08.002. 1073–1080.
Numis, A.L., Foster-Barber, A., Deng, X., Rogers, E.E., Barkovich, A.J., Ferriero, D.M., Sevilla-Castillo, R.A., Palacios, G.C., Ramirez-Campos, J., Mora-Puga, M., Diaz-
Glass, H.C., 2019. Early changes in pro-inflammatory cytokine levels in neonates Bustos, R., 2009. Methylprednisolone for the treatment of children with refractory
with encephalopathy are associated with remote epilepsy. Pediatr. Res. 86 (5), epilepsy. Neuropediatrics 40, 265–268. https://doi.org/10.1055/s-0030-1249653.
616–621. Shandra, O., Moshe, S.L., Galanopoulou, A.S., 2017. Inflammation in epileptic
O’Regan, M., DCH, J.B.M.F., 2008. Is ACTH a key to understanding anticonvulsant encephalopathies. Adv. Protein Chem. Struct. Biol. 108, 59–84.
action? Dev. Med. Child Neurol. 40, 82–89. https://doi.org/10.1111/j.1469- Shiihara, T., Miyashita, M., Yoshizumi, M., Watanabe, M., Yamada, Y., Kato, M., 2010.
8749.1998.tb15366.x. Peripheral lymphocyte subset and serum cytokine profiles of patients with West
Oby, E., Janigro, D., 2006. The blood-brain barrier and epilepsy. Epilepsia 47 (11), syndrome. Brain Dev. 32 (9), 695–702.
1761–1774. Shimada, T., Takemiya, T., 2014. Role of inflammatory mediators in the pathogenesis of
Oguni, H., Tanaka, T., Hayashi, K., et al., 2002. Treatment and long-term prognosis of epilepsy. Mediators Inflamm. 2014, 901902.
myoclonic-astatic epilepsy of early childhood. Neuropediatrics 33 (3), 122–132. Shumiloff, N.A., Lam, W.M., Manasco, K.B., 2013. Adrenocorticotropic hormone for the
Ohya, T., Nagai, T., Araki, Y., et al., 2009. A pilot study on the changes in immunity after treatment of West Syndrome in children. Ann. Pharmacother. 47 (5), 744–754.
ACTH therapy in patients with West syndrome. Brain Dev. 31 (10), 739–743. Sinclair, D.B., 2003. Prednisone therapy in pediatric epilepsy. Pediatr. Neurol. 28,
Orsini, A., Costagliola, G., Perna, D., Esposito, M.G., Bonfiglio, L., Striano, P., et al., 2019. 194–198. https://doi.org/10.1016/S0887-8994(02)00513-1.
Efficacy and tolerability of mycophenolate mofetil in a pediatric Rasmussen Sinclair, D.B., Snyder, T.J., 2005. Corticosteroids for the treatment of Landau-kleffner
syndrome. Epilepsy Behav Rep. 13, 100334. syndrome and continuous spike-wave discharge during sleep. Pediatr. Neurol. 32
Orsini, A., Foiadelli, T., Carli, N., et al., 2020. Rasmussen’s encephalitis: from immune (5), 300–306.
pathogenesis towards targeted-therapy. Seizure 81, 76–83. https://doi.org/ Skaper, S.D., Facci, L., Zusso, M., Giusti, P., 2018. An inflammation-centric view of
10.1016/j.seizure.2020.07.023. neurological disease: beyond the neuron. Front. Cell. Neurosci. 12, 72.
Owens, G.C., Huynh, M.N., Chang, J.W., McArthur, D.L., Hickey, M.J., Vinters, H.V., Striano, P., Pezzella, M., Brigati, G., Minetti, C., 2011. Lumping encephalopathies with
et al., 2013. Differential expression of interferon-γ and chemokine genes inflammation mediated status epilepticus: is there enough evidence? Epilepsy Behav.
distinguishes Rasmussen encephalitis from cortical dysplasia and provides evidence Mar. 20 (3), 592.
for an early Th1 immune response. J. Neuroinflammation 10. https://doi.org/ Suemitsu, S., Watanabe, M., Yokobayashi, E., et al., 2010. Fcgamma receptors contribute
10.1186/1742-2094-10-56. to pyramidal cell death in the mouse hippocampus following local kainic acid
Pardo, A.C., 2020. Ketogenic diet: a role in immunity? Pediatr. Neurol. Briefs 34, 5. injection. Neuroscience 166 (3), 819–831.
https://doi.org/10.15844/pedneurbriefs-34-5. Published 2020 Mar 3. Sun, Y., Ma, J., Li, D., et al., 2019. Interleukin-10 inhibits interleukin-1beta production
Pardo, C.A., Nabbout, R., Galanopoulou, A.S., 2014. Mechanisms of epileptogenesis in and inflammasome activation of microglia in epileptic seizures.
pediatric epileptic syndromes: rasmussen encephalitis, infantile spasms, and febrile J. Neuroinflammation 16 (1), 66.
infection-related epilepsy syndrome (FIRES). Neurotherapeutics 11 (2), 297–310. Talaber, G., Tuckermann, J.P., Okret, S., 2015. ACTH controls thymocyte homeostasis
Péchadre, J.C., Sauvezie, B., Osier, C., Gibert, J., 1977. Traitement des encéphalopathies independent of glucocorticoids. FASEB J. 29 (6), 2526–2534.
épileptiques de l’enfant par les gamma-globulines. Résultats préliminaires. Rev.

10
A. Orsini et al. Epilepsy Research 172 (2021) 106588

Tan, C., Shard, C., Ranieri, E., et al., 2015. Mutations of protocadherin 19 in female Vitaliti, G., Pavone, P., Mahmood, F., Nunnari, G., Falsaperla, R., 2014. Targeting
epilepsy (PCDH19-FE) lead to allopregnanolone deficiency. Hum. Mol. Genet. 24 inflammation as a therapeutic strategy for drug-resistant epilepsies: an update of
(18), 5250–5259. new immune-modulating approaches. Hum. Vaccin. Immunother. 10 (4), 868–875.
Tekgul, H., Tutuncuoglu, S., Coker, M., Coker, C., 1999. Infantile spasm: the effect of Vitaliti, G., Pavone, P., Marino, S., Saporito, M.A.N., Corsello, G., Falsaperla, R., 2019.
corticotropin (ACTH) on the free amino acid profile in cerebrospinal fluid. Brain Molecular mechanism involved in the pathogenesis of early-onset epileptic
Dev. 21 (1), 20–23. encephalopathy. Front. Mol. Neurosci. 12, 118.
Tekgul, H., Polat, M., Tosun, A., Serdaroglu, G., Kutukculer, N., Gokben, S., 2006. Viviani, B., Gardoni, F., Marinovich, M., 2007. Cytokines and neuronal ion channels in
Cerebrospinal fluid interleukin-6 levels in patients with West syndrome. Brain Dev. health and disease. Int. Rev. Neurobiol. 82, 247–263.
28 (1), 19–23. Wang, Y.Q., Fang, Z.X., Zhang, Y.W., Xie, L.L., Jiang, L., 2020. Efficacy of the ketogenic
Tian, D.S., Peng, J., 2017. Chemokine CCL2-CCR2 signaling induces neuronal cell death diet in patients with Dravet syndrome: A meta-analysis [published online ahead of
via STAT3 activation and IL-1beta production after status epilepticus. J. Neurosci. 37 print, 2020 Jul 18]. Seizure 81, 36–42. https://doi.org/10.1016/j.
(33), 7878–7892. seizure.2020.07.011.
Trivisano, M., Lucchi, C., Rustichelli, C., et al., 2017. Reduced steroidogenesis in patients Wells, J., Swaminathan, A., Paseka, J., Hanson, C., 2020. Efficacy and safety of a
with PCDH19-female limited epilepsy. Epilepsia 58 (6), e91–e95. ketogenic diet in children and adolescents with refractory Epilepsy-A review.
Tsuji, T., Okumura, A., Ozawa, H., Ito, M., Watanabe, K., 2007. Current treatment of Nutrients 12 (6), 1809. https://doi.org/10.3390/nu12061809. Published 2020 Jun
West syndrome in Japan. J. Child Neurol. 22 (5), 560–564. 17.
Ture, E., Kamasak, T., Cora, M., et al., 2016. Comparison of the serum cytokine levels Wilson, S., 2015. In response: indications and expectations for neuropsychological
before and after adrenocorticotropic hormone (ACTH) therapy in patients with assessment in routine epilepsy care: report of the ILAE Neuropsychology Task Force,
infantile spasm. Seizure 41, 112–115. Diagnostic Methods Commission, 2013-2017. Epilepsia 56 (8), 1316–1317.
Ulusoy, C., Vanlı-Yavuz, E.N., Şanlı, E., Timirci-Kahraman, Ö, Yılmaz, V., Bebek, N., Wu, Y., Wang, X., Mo, X., et al., 2008. Expression of monocyte chemoattractant protein-1
et al., 2020. Peripheral blood expression levels of inflammasome complex in brain tissue of patients with intractable epilepsy. Clin. Neuropathol. 27 (2), 55–63.
components in two different focal epilepsy syndromes. J. Neuroimmunol. 347, Wu, Z., Liu, Y., Huang, J., Huang, Y., Fan, L., 2019. MiR-206 inhibits epilepsy and
577343. seizure-induced brain injury by targeting CCL2. Cytotechnology 809–818.
van den Munckhof, B., de Vries, E.E., Braun, K.P., et al., 2016. Serum inflammatory Xanthos, D.N., Sandkuhler, J., 2014. Neurogenic neuroinflammation: inflammatory CNS
mediators correlate with disease activity in electrical status epilepticus in sleep reactions in response to neuronal activity. Nat. Rev. Neurosci. 15 (1), 43–53.
(ESES) syndrome. Epilepsia 57 (2), e45–50. Xu, T., Yu, X., Wang, T., et al., 2017. The effect of CXCR2 inhibition on seizure activity in
van Gassen, K.L., de Wit, M., Koerkamp, M.J., et al., 2008. Possible role of the innate the pilocarpine epilepsy mouse model. Brain Res. Bull. 134, 91–98.
immunity in temporal lobe epilepsy. Epilepsia 49 (6), 1055–1065. Yamanaka, G., Kawashima, H., Oana, S., et al., 2010. Increased level of serum
van Vliet, E.A., da Costa Araujo, S., Redeker, S., van Schaik, R., Aronica, E., Gorter, J.A., interleukin-1 receptor antagonist subsequent to resolution of clinical symptoms in
2007. Blood-brain barrier leakage may lead to progression of temporal lobe epilepsy. patients with West syndrome. J. Neurol. Sci. 298 (1-2), 106–109.
Brain 130 (Pt 2), 521–534. Yamanaka, G., Morishita, N., Morichi, S., et al., 2018. Serial analysis of multiple serum
van Vliet, E.A., Aronica, E., Vezzani, A., Ravizza, T., 2018. Review: Neuroinflammatory cytokine responses to adrenocorticotropic hormone therapy in patients with west
pathways as treatment targets and biomarker candidates in epilepsy: emerging syndrome. J. Child Neurol. 33 (8), 528–533.
evidence from preclinical and clinical studies. Neuropathol. Appl. Neurobiol. 44 (1), Yang, H., Wang, H., Chavan, S.S., Andersson, U., 2015. High mobility group Box protein
91–111. 1 (HMGB1): the prototypical endogenous danger molecule. Mol Med. 21 (Suppl 1),
Vezzani, A., Conti, M., De Luigi, A., et al., 1999. Interleukin-1beta immunoreactivity and S6–s12.
microglia are enhanced in the rat hippocampus by focal kainate application: Yang, X., Wan, M., Yu, F., Wang, Z., 2019. Efficacy and safety of plerixafor for
functional evidence for enhancement of electrographic seizures. J. Neurosci. 19 (12), hematopoietic stem cell mobilization for autologous transplantation in patients with
5054–5065. non-Hodgkin lymphoma and multiple myeloma: a systematic review and meta-
Vezzani, A., Moneta, D., Conti, M., et al., 2000. Powerful anticonvulsant action of IL-1 analysis. Exp. Ther. Med. 18 (2), 1141–1148.
receptor antagonist on intracerebral injection and astrocytic overexpression in mice. You, S.J., Jung, D.E., Kim, H.D., Lee, H.S., Kang, H.C., 2008. Efficacy and prognosis of a
Proc. Natl. Acad. Sci. U. S. A. 97 (21), 11534–11539. short course of prednisolone therapy for pediatric epilepsy. Eur. J. Paediatr. Neurol.
Vezzani, A., Balosso, S., Ravizza, T., 2008. The role of cytokines in the pathophysiology 12, 314–320. https://doi.org/10.1016/j.ejpn.2007.09.003.
of epilepsy. Brain Behav. Immun. 22 (6), 797–803. Yu, N., Liu, H., Di, Q., 2013. Modulation of immunity and the inflammatory response: a
Vezzani, A., Balosso, S., Maroso, M., Zardoni, D., Noe, F., Ravizza, T., 2010. ICE/caspase new target for treating drug-resistant epilepsy. Curr. Neuropharmacol. 11 (1),
1 inhibitors and IL-1beta receptor antagonists as potential therapeutics in epilepsy. 114–127. https://doi.org/10.2174/157015913804999540.
Curr. Opin. Investig. Drugs 11 (1), 43–50. Zhao, J., Wang, Y., Xu, C., et al., 2017. Therapeutic potential of an anti-high mobility
Vezzani, A., French, J., Bartfai, T., Baram, T.Z., 2011. The role of inflammation in group box-1 monoclonal antibody in epilepsy. Brain Behav. Immun. 64, 308–319.
epilepsy. Nat. Rev. Neurol. 7, 31–40. https://doi.org/10.1038/nrneurol.2010.178. Zhou, Y., Tang, H., Liu, J., Dong, J., Xiong, H., 2011. Chemokine CCL2 modulation of
Vezzani, A., Aronica, E., Mazarati, A., Pittman, Q.J., 2013. Epilepsy and brain neuronal excitability and synaptic transmission in rat hippocampal slices.
inflammation. Exp. Neurol. 244, 11–21. J. Neurochem. 116 (3), 406–414.
Vidya, M.K., Kumar, V.G., Sejian, V., Bagath, M., Krishnan, G., Bhatta, R., 2018. Toll-like Zlotnik, A., Yoshie, O., 2012. The chemokine superfamily revisited. Immunity 36 (5),
receptors: significance, ligands, signaling pathways, and functions in mammals. Int. 705–716.
Rev. Immunol. 37 (1), 20–36. Zuena, A.R., Casolini, P., Lattanzi, R., Maftei, D., 2019. Chemokines in Alzheimer’s
Vilcek, J., Feldmann, M., 2004. Historical review: cytokines as therapeutics and targets disease: new insights into prokineticins, chemokine-like proteins. Front. Pharmacol.
of therapeutics. Trends Pharmacol. Sci. 25 (4), 201–209. 10, 622.

11

You might also like