Skip to main content

Anti-high mobility group box protein 1 monoclonal antibody downregulating P-glycoprotein as novel epilepsy therapeutics

Abstract

Epilepsy, a neurological illness, is characterized by recurrent uncontrolled seizures. There are many treatments of options that can be used as the therapy of epilepsy. However, anti-seizure medications as the primary treatment choice for epilepsy show many possible adverse effects and even pharmacoresistance to the therapy. High Mobility Group Box 1 (HMGB1) as an initiator and amplifier of the neuroinflammation is responsible for the onset and progression of epilepsy by overexpressing P-glycoprotein on the blood brain barrier. HMGB1 proteins then activate TLR4 in neurons and astrocytes, in which proinflammatory cytokines are produced. Anti-HMGB1 mAb works by blocking the HMGB1, reducing inflammatory activity in the brain that may affect epileptogenesis. Through the process, anti-HMGB1 mAb reduces the TLR4 activity and other receptors that may involve in promote signal of epilepsy such as RAGE. Several studies have shown that anti-HMGB1 has the potential to inhibit the increase in serum HMGB1 in plasma and brain tissue. Further research is needed to identify the mechanism of the inhibiting of overexpression of P-glycoprotein through anti-HMGB1 mAb.

Introduction

Excessive and hypersynchronous firing of neurons in the brain causes a seizure, which is a paroxysmal change of neurologic function [1]. The term "epileptic seizure" is used to differentiate a seizure produced by abnormal neuronal firing from a psychogenic seizure [2]. Recurring spontaneous uncontrolled seizures are referred to as epilepsy [3]. In 2015, epilepsy contributed around 125,000 fatalities worldwide and around 23 million prevalent cases [4]. In high-income nations, the condition has a lifetime prevalence of 2.3 to 15.9 per 1000 people, while in low-income countries it has a prevalence of 3.6 to 15.4 per 1000 people [5]. Epilepsy is more common in the first year of life and over 85 years with yearly estimates of 86 per 100,000 people and 180 per 100,000 people, respectively [6]. An estimated 75% of epilepsy cases begin in youth, owing to the increased sensitivity of seizures in developing brains [7]. Moreover, the most prevalent precipitating factors include fever, traumatic brain injury, cerebrovascular illness, substance withdrawal, inflammation, and hormonal disturbances [8]. A number of studies have suggested the role of P-glycoprotein (P-gp) in pathogenesis of epilepsy. P-gp is found to be responsible in limiting wide ranges of drugs in epilepsy therapy [9].

Although there are a variety of other medication strategies for epilepsy, such as vagus nerve stimulation [10], surgery [11], and a ketogenic diet [12], anti-seizure medications (ASMs) are typically the first option [13]. The majority of data for the efficacy and safety of ASMs comes from subjects under 65 years old, and there are few randomized controlled studies in older individuals [14]. Pharmacokinetics shift, comorbidities, physiological alterations, polypharmacy and accompanying impairment of capability may complicate the administration and effectivity of ASMs in older age groups [15, 16]. Additionally, drug interactions and possible adverse effects, such as nausea, sleepiness, mental retardation, rashes, liver toxicity and metabolic changes should be properly examined while using ASMs [17]. Furthermore, more than half of epilepsy patients using ASMs have been reported to acquire bone abnormalities [17, 18], and numerous studies have found a connection between extensive ASM usage with rickets, osteomalacia [19] and increased risks of fracture [20].

HMGB1 (High Mobility Group Box 1) is a non-histomic, chromosome-linked small protein with cytokinin function that has nuclear, cytosolic, and extracellular activities [21]. Being the dominant nonhistone nucleoprotein in the HMGB gene family, HMGB1 shuttles between nucleus and cytoplasm serving as both a nonhistone nucleoprotein as well as an inflammatory cytokine [22]. Through TLR4/RAGE/NF-kB pathway activation in brain microvascular endothelium, HMGB1 leads to P-gp overexpression in epileptic brain regions [23, 24]. Due to its overexpression during seizures, P-gp has been intensively researched for developing multidrug resistance in epilepsy [25]. Thus, through the hyperexcitability of the HMGB1/RAGE/TLR4/NF-kB signaling pathway, HMGB1 has a pro-epileptic impact that induces temporal lobe epilepsy development [26,27,28,29]. This finding is also congruent with research conducted by Paundel et.al that proposed inhibiting the HMGB1/RAGE/TLR4/NF-kB pathway might be a potential treatment approach against HMGB1-mediated epileptic disorders [30]. Inhibiting the signaling pathway downregulate P-gp overexpression and it might be studied as a potential therapeutic approach [24, 31]. Fortunately, recent studies show that anti-HMGB1 monoclonal antibodies (mAb) treatment might be effective in treating a wide range of central nervous system (CNS) and peripheral nervous system (PNS) disorders such as epilepsy [32, 33]. In acute stage of status epilepticus, intravenous therapy with neutralizing anti-HMGB1 monoclonal antibody exhibited neuroprotective effect on neuronal death, inhibition of HMGB1 release, blood brain barrier (BBB) protection and anti-inflammatory actions [34].

The goal of this review is to evaluate the prospects of anti-HMGB1 monoclonal antibodies in downregulating P-glycoprotein as a therapeutic approach in epilepsy management.

The pathophysiology of epilepsy

The brain is responsible to control the activity of neurons, nerve cells which specially designed to channel electrical impulses around the central and peripheral nervous systems [35]. A seizure developed when an abnormal synchronous firing of neurons in some parts of the brain occurs [36]. A patient with seizure experienced certain distraction of the normal balance involving excitation and inhibition which happened in the brain [7]. Normal membrane conductance, inhibitory synaptic current breakdown and excessive excitability spread locally resulting in a focal seizure or widely resulting in generalized seizure [37].

Many acquired brain pathologies, such as tumors, stroke, infection, traumatic brain injury, and mutation of a single gene contribute to the presentation of epilepsy [38]. Some evidence shows that neuroinflammation contributes to the onset and progression of epilepsy alongside HMGB1 as an initiator and amplifier of the process [39]. In the inflammatory process of epileptic brain, the activity and level of P-gp escalate on the BBB with overexpression as the result [23]. The inhibition and induction of P-gp is known as essential underlying components of drug interactions in humans [40]. There are different theories interpreting the nature of drug resistance with the pharmacokinetic hypothesis and the transport hypothesis of the drug, displaying the possible mechanism of the resistance in ASMs by the overexpression of P-gp in the BBB [41]. The P-gp distribution is arranged to narrow drug entry to the brain or letting efflux of drugs from brain to blood, which reveals that P-gp plays an essential part in protecting the brain against xenobiotics [42]. Thus, the crucial efflux protein of the BBB is P-gp that repeatedly transports enormous amounts of lipophilic drugs, as well as ASMs, out of the BBB membrane and back toward the bloodstream, ended up in pharmacoresistance to therapeutic medications intended to target the brain [43]. Individuals who fail to respond or only partially respond to ASMs will continue to experience paralyzing seizures that lead to psychiatric, neuropsychological, and social impairments by such means decreasing the quality of life and increasing morbidity and mortality [44].

HMGB1 in expressing P-glycoprotein

Seizures begin with a strong stimulation of vulnerable epileptic neurons, which leads to gradual induction of linked neurons [45]. Uncontrolled impulses finally impact a portion of the brain and cause clinical symptoms [46]. In a patients epileptic brain study, researchers discovered higher levels of HMGB1, TLR4, RAGE, NF-kB and nitric oxide synthase, as well as higher amounts of IL-1, IL-6, TNF- α, TGF- β, and IL-10 [47]. The ligand–receptor effects of HMGB1-RAGE/TLR4 in the generation of seizures have been clearly established [48]. A rise in extracellular HMGB1 paralleled seizure onset, confirming HMGB1 as a significant contributor to the beginning of the hyperexcitability [49]. The activation of neuron TLR4 by HMGB1 is a critical process in seizure onset [27]. Hence, HMGB1/RAGE/TLR4 pathway may initiate the development and persistence of seizures in people (Fig. 1) and can be addressed to achieve anti-seizure results in drug-resistant epilepsies [30].

Fig. 1
figure 1

HMGB1/RAGE/TLR4 pathway signaling. RAGE; receptor for advanced glycation end products, TLR4; toll like receptor 4, NF-kB; nuclear factor kappa B, NMDA; N-methyl-D-aspartic acid

RAGE aids TLR4 trafficking to the cell surface, while TLR4 controls RAGE translation and translocation to cell surface [50]. The TLR4 receptor complex stimulates the recruitment of different adaptor proteins and activates multiple signaling pathways, ultimately activating NF-kB [51]. HMGB1 promotes nuclear translocation of nuclear factor-B, resulting in the production of proinflammatory cytokines such as TNF-α, IL-6 and IL-1β [52]. Through the N-methyl-D-aspartate (NMDA)/cyclooxygenase-2 (COX-2) pathway by NF-kB, extracellular glutamate enhanced the amount of P-gp expression [23]. TNF-α, an inflammatory cytokine, has been shown to promote P-gp activity in the BBB [53].

Anti-HMGB1 monoclonal antibody in epilepsy

In a clinical study involving adult zebrafishes, anti-HMGB1 mAb reduces seizures, improves associated cognitive deficits, and reduces seizure-induced elevation of proinflammatory cytokines, potentially protecting from future seizures through neuroinflammatory network regulation [54]. Intravenous administration of anti-HMGB1 mAb protects rats against BBB rupture caused by infarction and hemorrhage, and this protective activity is coupled with the reduction of production of inflammatory mediators markers and glial cell stimulation [55]. In modeled animals, the anti-HMGB1 monoclonal antibody (mAb) has also been proven to be useful for the treatment of a variety of CNS illnesses, notably hemorrhage, brain trauma, Parkinson's disease and Alzheimer's disease [33]. Although still in the phase of animal model clinical trials, researchers support the potential use of the substance in humans to downregulate the elevated HMGB1 [39].

HMGB1 will continue to increase during seizures and may holds a role in upregulating P-gp [33]. Some theories point out involvement of P-gp overexpression in status epilepticus [24]. Several experiments showed ASM being pumped by P-gp, resulting in low concentration of ASM in brain tissue [33, 54]. Nishibori et al. reported that the epileptiform activity in the brain slices was suppressed by superfusion of anti-HMGB1 mAb on the surface of brain slices taken from patients who had repeated seizures. Anti-HMGB1 mAb can be useful in the treatment of various types of inflammatory diseases that are specific to BBB disruptions in animal models [33]. Disruption of BBB may be caused by the participation of extracellular HMGB1 [56]. One of the current therapeutic strategies for epilepsy is to focus on molecular targeting of HMGB1 with specific antibodies or antagonists that have the potential to minimize the occurrence of epileptic seizures characterized by HMGB1 expression [39]. HMGB1, IL-1β, and S-100B can be molecular markers in the incidence of epilepsy characterized by changes in serum concentrations so that they have clinical significance in the epileptic process and can predict outcomes [57]. Anti-HMGB1 mAb has good drug potency with high therapeutic index and sufficient specificity to be used as therapy. Administration of anti-HMGB1 mAb and diazepam was shown to reduce the number of generalized seizures [58]. Anti-HMGB1 can help to regulate the occurrence of epilepsy and the process of epileptogenesis (Fig. 2) [26, 30, 33]. Epileptogenesis is a process when the brain under normal conditions undergoes functional changes in the form of abnormal electrical activity, resulting in chronic seizures. In general, epileptogenesis consists of three stages, namely acute events or precipitating events, latent or clinically silent periods, and stages of chronic epilepsy or spontaneous seizures [59,60,61]. Trigger factors can be in the form of oxidation, inflammation, neurogenesis, apoptosis, and synaptic plasticity. These factors can lead to functional and structural changes in nerve cells. These changes result in abnormal hyperexcitability and spontaneous seizures [62].

Fig. 2
figure 2

Anti-HMGB1 mAb can inhibit status epilepticus and epileptogenesis. HMGB1; High Mobility Group Box 1, BBB; blood brain barrier

Convulsant activation also occurs due to IL-1β and HMGB1 which result in neuronal adjustment, leading to a decrease in the threshold for focal ictal events. [63]. There will be activation of astrocytes and microglia through an increase in the number and shape of specific cells that occur in the cerebral cortex and hippocampus during acute epilepsy [34]. Activation of glial cells initiates epileptogenesis after status epilepticus. IP3 R2 which mediates Ca2 + signaling in reactive astrocytes will be proconvulsive in the epileptic brain and contribute to epileptogenesis. Activation of microglia is the initial process of epileptogenesis which is indicated by the increase of proinflammatory cytokines of microglia before the activation of astrocytic [64].

Anti-HMGB1 mAb can inhibit the activation of these cells systemically. Anti-HMGB1 mAb can reduce the number of cells undergoing apoptosis after the occurrence of acute status epilepticus [34]. HMGB1 translocation of neurons and astrocytes can be inhibited by anti-HMGB1 which can decrease the duration of tonic–clonic seizures by increasing the threshold [65]. HMGB1 is a molecule that plays an important role in increasing BBB permeability in the acute phase of epilepsy. HMGB1 released into the CNS and peripheral blood stream after pilocarpine injection plays a role in inducing damage to the BBB. Anti-HMGB1 can significantly inhibit the permeability of the BBB [34]. In a study involving epilepsy by inducing upregulation of IL-1β and TNF-α mRNA was shown to be blocked by anti-HMGB1 through a potential anti-inflammatory mechanism that could inhibit microglia activation and proinflammatory cytokine synthesis [66]. Expression of proinflammatory cytokines also occurs as a result of an excess of HMGB1 and will subsequently have an impact on the continuous induction of HMGB1 release [67]. In addition to its anti-inflammatory effect, anti-HMGB1 mAb has a mechanism in inhibiting the increase of serum HMGB1 in brain tissue and plasma as well as interfering with the translocation of HMGB1 from the nucleus to astrocytes and neuronal cytoplasm, thus forming BBB stabilization [68]. Anti-HMGB1 also can interrupt TNF-α formation and expression by western blot analysis [69]. Complex cellular internalization of HMGB1 and HMGB1-LPS can be inhibited by anti-HMGB1 mAb, HMGB1 antagonist box A protein, nicotinic acetylcholine receptor subtype alpha 7 GTS-21 agonist, and acetylcholine [70]. Through TLR4/NF-κB and RAGE/NF-κB, HMGB1 may have an effect on increasing P-gp expression in seizure-induced rat brain endothelial cells [23]. Excessive activation of P-gp results in decreased focal (R)-[11C] uptake of verapamil in pharmacoresistance seen in pharmaco-resistant temporal lobe epilepsy in an in vivo study [25]. Inhibition of HMGB1 with anti-HMGB1 mAb has an important role in obtaining therapeutic benefit in the incidence of seizures and their epileptogenesis [26, 39, 57].

Many experiments have been done by using animal models to observe involvement of HMGB1 in status epilepticus [33]. Finding by Fu et al. in observing hippocampal neurons to see significant translocation of HMGB1 after the pilocarpine injection and systemic injection of anti-HMGB1 mAb that significantly inhibited HMGB1 translocation, thereby suppressing the status epilepticus. In this model, administration of anti-HMGB1 gives protection effects against neuronal apoptosis, avoiding the BBB disruption and resisting the inflammation effect by pilocarpine [34]. An experiment by Zhao et al. using 4 types of epileptic animal models points out the beneficial effects using systemic injection of anti-HMGB1 mAb as the treatment. These animal models include the kainite-induced epilepsy, chronic seizure-induced epilepsy, electroshock-induced epilepsy and picrotoxin-induced epilepsy [65].

Pharmacokinetics and pharmacodynamics of anti-HMGB1 monoclonal antibody

Monoclonal antibody already being developed as clinical therapeutics nowadays [71]. Anti-HMGB1 impacts several central nervous system pathological conditions, such as Parkinson disease, traumatic brain injury, stroke, and spinal cord injury [33, 68, 72,73,74]. In dose-dependent manner, anti-HMGB1 monoclonal antibody (mAb) extended the generalized seizure’s latency, decreased tonic convulsion, and reduced seizure stage [65]. According to Okuma et al., in treatment of brain injury, the anti-HMGB1 mAb dosage is 1 mg/kg was administrated intravenously at 5 min and 6 h after injury [72]. Anti-HMGB1 mAb was diluted by 2 mg/ml phosphate-buffered saline [65]. Anti-HMGB1 mAb injected intravenously in rat experiments shows the anti-HMGB1 accumulation in ischemic site of the brain in PET imaging 16 h after injection. Small amount of anti-HMGB1 is also present in brain parenchyma. Anti-HMGB1 possible to spread into blood brain barrier and luminal side of vascular endothelial cell [33]. Anti-HMGB1 only targeting HMGB1 (prevent translocation from nuclei) may be safer treatment to treat epilepsy [65]. Anti-HMGB1 mAb binds solidly with HMGB1. Thus, anti-HMGB1 estimated have long time elimination in the brain [75]. In general, monoclonal antibody is mostly eliminated by lysosomal degradation to amino acids. The molecule of monoclonal antibody is too large to eliminate by the kidney [76].

Major aim of epilepsy therapy is not only about reduction of seizure frequency, but also how to prevent epileptogenesis, the process of the brain develops epilepsy [77]. Anti-HMGB1 mAb blocking the HMGB1 resulting in reduced of inflammatory activity in the brain that may affect epileptogenesis [65]. Intraperitoneal injection of anti-HMGB1 mAb in previous study, effectively reduced the presence of HMGB1 in the nuclei cell of central nervous system. Anti-HMGB1 monoclonal antibody neutralized extracellular HMGB1 in the CNS resulted in persistent nuclear HMGB1 staining and alleviated inflammation [78]. Previous report found, anti-HMGB1 mAb effectively reduced brain ischemia by efficient clearance of HMGB1 and inhibiting edema, ameliorates spontaneous arthritis model, prevent joint destruction, and repair cardiac pathological changes in experimental autoimmune myocarditis by suppress Th-17 cell [75, 79, 80]. Anti-HMGB1 mAb have immunohistochemical and neurochemical benefit reducing HMGB1 level in the plasma and brain evaluating by ELISA in day 1,4,7, 14 after injection [68]. HMGB1 translocation from the nuclei to the cytosol is inhibited by anti-HMGB1 mAb. Blocking of HMGB1 by regulatory axis using anti-HMGB1 mAb, reduced the TLR4 and others receptor that may involve in promote signal of epilepsy such as RAGE [27]. HMGB1 is strictly involved in the inflammatory response related to epilepsy [81].

A study reported the therapeutic effect of anti-HMGB1 mAb on epilepsy start from 1 h and last to 24 h, indicating that there’s a potential clinical epilepsy treatment. In addition, the potential of anti-HMGB1 mAb on human epileptic brain slices has not been eliminated, suggesting that anti-HMGB1 mAb binds strongly to HMGB1 with potential long-term anti-epileptic activity [39, 65]. Anti-HMGB1 mAb only targets the translocated and activated HMGB1. Moreover, it is reported on previous study that anti-HMGB1 in a high dose (25 mg/kg in mice) did not affect body growth rate, basic physical functions and thermoregulation [65]. Although anti-HMGB1 mAb showed lots of potential for specificity therapy with wider therapeutic window, this strategy targeting HMGB1 is restricted by problems like the probability in the tertiary structure by conformational switches of the Ab-recognized zone [82].

The use of anti-hmgb 1 mAb as a modality of epilepsy therapy may focus on the cost component, which depends on the antibody protein used and the encapsulation of the antibody therapy so that it can enter the human body and reach its targets. From the antibodies used, as with mAb therapy in COVID-19, the costs incurred in mAb therapy in patients with epilepsy may be more expensive than previously available conventional modalities [83]. The potential for monoclonal antibody therapy in the future will be seen from the prevalence of disease incidence from the indications of therapy given. mAb therapy in oncological conditions with a smaller market certainly has a higher cost compared to immunological conditions with a larger market [84]. The second is from the formulation of therapeutic protein encapsulation. The therapeutic potential possessed by anti-hmgb1 mAb in the future has the opportunity for further development in terms of the formation of long-acting protein formulations so as to maintain protein stability and can provide appropriate doses for a longer duration of time. This needs to be developed in the future because of the limitations possessed by a mAb, namely from its short pharmacokinetic properties, protein stability, protein aggregation, and denaturation that may occur due to the storage and transportation process of the protein which will ultimately have an impact on its clinical use. The common way to stabilize protein-based drugs is through improving the molecular structure with protein engineering and administering stabilizers. Several formulations are being developed, such as through a controlled release system (hydrogel, micelles, liposomes, nanoparticles) [85, 86]. Polyester-based nanoparticles as one of the strategies developed in mAb encapsulation are said to have the potential to increase antibody stability, prolong the duration of the therapeutic effect, and recognize selective cellular targets [87]. Therefore, anti-HMGB1 mAb that significantly blockade the HMGB1 is potential treatment for epilepsy and assessing the efficacies with several experiments of epilepsy models needed to be the objectives for future research.

Conclusion

Anti-HMGB1 mAb may have the potential to downregulate P-glycoprotein expression which is overexpressed due to an increase in HMGB1 in epilepsy. Several previous studies have shown that anti-HMGB1 can potentially inhibit the increase in serum HMGB1 in plasma as well as brain tissue and can interfere with the translocation of HMGB1 from the nucleus to astrocytes and neuron cytoplasm. Inhibition of HMGB1 by anti-HMGB1 has a significant clinical impact. Therefore, inhibition of the overexpression of P-glycoprotein via anti-HMGB1 mAb may be able to become a therapeutic target for epilepsy. The mechanism that occurs from the inhibition of overexpression of P-glycoprotein through anti-HMGB1 mAb needs further research. Anti-HMGB1 mAb may contribute to the development of anti-seizure medications in the future through its mechanism of action against P-glycoprotein.

Availability of data and materials

Not applicable.

References

  1. Ong LT. Temporal lobe epilepsy – pathophysiology and mechanisms. Eur Neurol Rev. 2019;14:66.

    Article  Google Scholar 

  2. Fisher RS, Cross JH, French JA, Higurashi N, Hirsch E, Jansen FE, et al. Operational classification of seizure types by the International League Against Epilepsy: Position Paper of the ILAE Commission for Classification and Terminology. Eur J Paediatr Neurol. 2017;58:522–30.

    Google Scholar 

  3. Huff JS, Murr N. Seizure. Treasure Island (FL), 2021.

  4. Beghi E, Giussani G. Aging and the Epidemiology of Epilepsy. Neuroepidemiology. 2018;51:216–23.

    Article  PubMed  Google Scholar 

  5. Bell GS, Neligan A, Sander JW. An unknown quantity–the worldwide prevalence of epilepsy. Epilepsia. 2014;55:958–62.

    Article  PubMed  Google Scholar 

  6. Fiest KM, Sauro KM, Wiebe S, Patten SB, Kwon C-S, Dykeman J, et al. Prevalence and incidence of epilepsy: A systematic review and meta-analysis of international studies. Neurology. 2017;88:296–303.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Stafstrom CE, Carmant L. Seizures and epilepsy: an overview for neuroscientists. Cold Spring Harb Perspect Med. 2015;5: a022426.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Beghi E. The Epidemiology of Epilepsy. Neuroepidemiology. 2020;54:185–91.

    Article  PubMed  Google Scholar 

  9. Kambli L, Bhatt LK, Oza M, Prabhavalkar K. Novel therapeutic targets for epilepsy intervention. Seizure. 2017;51:27–34.

    Article  PubMed  Google Scholar 

  10. Von HT, Oliveira F, Francisco AN, Junior ZD, Stebel SL. The role of vagus nerve stimulation in refractory epilepsy. Arq Neuropsiquiatr. 2017;90:657–66.

    Google Scholar 

  11. Ram S, Deepak L. Antiepileptic drugs—a review. Pediatr Ann. 2015;44:e36–42.

    Google Scholar 

  12. Meira I, Romão TT, Pires HJ, Krüger LT, Pires MEP, da Conceição PO. Ketogenic Diet and Epilepsy: What We Know So Far. Front Neurosci. 2019;13:5.

    Article  Google Scholar 

  13. Abou-Khalil BW. Update on Antiepileptic Drugs 2019. Continuum (Minneap Minn). 2019;25:508–36.

    Google Scholar 

  14. Werhahn KJ, Trinka E, Dobesberger J, Unterberger I, Baum P, Deckert-Schmitz M, et al. A randomized, double-blind comparison of antiepileptic drug treatment in the elderly with new-onset focal epilepsy. Epilepsia. 2015;56:450–9.

    Article  CAS  PubMed  Google Scholar 

  15. Glauser T, Ben-Menachem E, Bourgeois B, Cnaan A, Guerreiro C, Kälviäinen R, et al. Updated ILAE evidence review of antiepileptic drug efficacy and effectiveness as initial monotherapy for epileptic seizures and syndromes. Epilepsia. 2013;54:551–63.

    Article  CAS  PubMed  Google Scholar 

  16. Bruun E, Virta LJ, Ker T, Lj V. Choice of the first anti-epileptic drug in elderly patients with newly diagnosed epilepsy: A Finnish retrospective study. Seizure. 2015;31:27–32.

    Article  PubMed  Google Scholar 

  17. Fan H-C, Lee H-S, Chang K-P, Lee Y-Y, Lai H-C, Hung P-L, et al. The impact of anti-epileptic drugs on growth and bone metabolism. Int J Mol Sci. 2016;17:1242.

    Article  PubMed Central  Google Scholar 

  18. Lazzari AA, Dussault PM, Thakore-James M, Gagnon D, Baker E, Davis SA, et al. Prevention of bone loss and vertebral fractures in patients with chronic epilepsy–antiepileptic drug and osteoporosis prevention trial. Epilepsia. 2013;54:1997–2004.

    Article  CAS  PubMed  Google Scholar 

  19. Hamed SA. Markers of bone turnover in patients with epilepsy and their relationship to management of bone diseases induced by antiepileptic drugs. Expert Rev Clin Pharmacol. 2016;9:267–86.

    Article  CAS  PubMed  Google Scholar 

  20. Shen C, Chen F, Zhang Y, Guo Y, Ding M. Association between use of antiepileptic drugs and fracture risk: a systematic review and meta-analysis. Bone. 2014;64:246–53.

    Article  CAS  PubMed  Google Scholar 

  21. Street ME. HMGB1: a possible crucial therapeutic target for COVID-19? Horm Res Paediatr. 2020;93:73–5.

    Article  CAS  PubMed  Google Scholar 

  22. Yuan S, Liu Z, Xu Z, Liu J, Zhang J. High mobility group box 1 (HMGB1): a pivotal regulator of hematopoietic malignancies. J Hematol Oncol. 2020;13:91.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Chen Y, Huang X-J, Yu N, Xie Y, Zhang K, Wen F, et al. HMGB1 contributes to the expression of p-glycoprotein in mouse epileptic brain through toll-like receptor 4 and receptor for advanced glycation end products. PLoS ONE. 2015;10: e0140918.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Xie Y, Yu N, Chen Y, Zhang K, Ma H, Di Q. HMGB1 regulates P-glycoprotein expression in status epilepticus rat brains via the RAGE/NF-κB signaling pathway. Mol Med Rep. 2017;16:1691–700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Feldmann M, Asselin M-C, Liu J, Wang S, McMahon A, Anton-Rodriguez J, et al. P-glycoprotein expression and function in patients with temporal lobe epilepsy: A case-control study. Lancet Neurol DOI: https://doi.org/10.1016/S1474-4422(13)70109-1.

  26. Ravizza T, Terrone G, Salamone A, Frigerio F, Balosso S, Antoine DJ, et al. High Mobility Group Box 1 is a novel pathogenic factor and a mechanistic biomarker for epilepsy. Brain Behav Immun. 2018;72:14–21.

    Article  CAS  PubMed  Google Scholar 

  27. Iori V, Maroso M, Rizzi M, Iyer AM, Vertemara R, Carli M, et al. Receptor for Advanced Glycation Endproducts is upregulated in temporal lobe epilepsy and contributes to experimental seizures. Neurobiol Dis. 2013;58:102–14.

    Article  CAS  PubMed  Google Scholar 

  28. Balosso S, Liu J, Bianchi ME, Vezzani A. Disulfide-containing high mobility group box-1 promotes N-methyl-D-aspartate receptor function and excitotoxicity by activating Toll-like receptor 4-dependent signaling in hippocampal neurons. Antioxid Redox Signal. 2014;21:1726–40.

    Article  CAS  PubMed  Google Scholar 

  29. Yang W, Li J, Shang Y, Zhao L, Wang M, Shi J, et al. HMGB1-TLR4 axis plays a regulatory role in the pathogenesis of mesial temporal lobe epilepsy in immature rat model and children via the p38MAPK Signaling Pathway. Neurochem Res. 2017;42:1179–90.

    Article  CAS  PubMed  Google Scholar 

  30. Paudel YN, Shaikh MF, Chakraborti A, Kumari Y, Aledo-Serrano Á, Aleksovska K, et al. HMGB1: A Common Biomarker and Potential Target for TBI, Neuroinflammation, Epilepsy, and Cognitive Dysfunction. Front Neurosci. 2018;12:628.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Gidal BE. P-glycoprotein expression and pharmacoresistant epilepsy: cause or consequence? Epilepsy Curr. 2014;14:136–8.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Zhou H, Zhang S, Le Y, Huang J, Tong J, Ouyang W. Anti-HMGB1 antibody decreases surgery-induced upregulation of necroptosis-associated proteins in aged rats. J Anesth Perioper Med. 2018;5:114–24.

    Article  CAS  Google Scholar 

  33. Nishibori M, Mori S, Takahashi HK. Anti-HMGB1 monoclonal antibody therapy for a wide range of CNS and PNS diseases. J Pharmacol Sci. 2019;140:94–101.

    Article  CAS  PubMed  Google Scholar 

  34. Fu L, Liu K, Wake H, Teshigawara K, Yoshino T, Takahashi H, et al. Therapeutic effects of anti-HMGB1 monoclonal antibody on pilocarpine-induced status epilepticus in mice. Sci Rep. 2017;7:1179.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Lawal M, Omobayo H, Lawal K. Epilepsy: pathophysiology, clinical manifestations and treatment options. Br J Neurosci Nurs. 2018;14:58–72.

    Article  Google Scholar 

  36. Falco-Walter J. Epilepsy-definition, classification, pathophysiology, and epidemiology. Semin Neurol. 2020;40:617–23.

    Article  PubMed  Google Scholar 

  37. Kumar S, Singh G. Pathophysiology of epilepsy : An updated review. Int J Med Heal Res Int. 2016;2:32–6.

    CAS  Google Scholar 

  38. Goldberg EM, Coulter DA. Mechanisms of epileptogenesis: A convergence on neural circuit dysfunction. Nat Rev Neurosci. 2013;14:337–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Paudel YN, Semple BD, Jones NC, Othman I, Shaikh MF. High mobility group box 1 (HMGB1) as a novel frontier in epileptogenesis: from pathogenesis to therapeutic approaches. J Neurochem. 2019;151:542–57.

    Article  CAS  PubMed  Google Scholar 

  40. Elkhayat HA, Aly RH, Elagouza IA, El-Kabarity RH, Galal YI. Role of P-glycoprotein inhibitors in children with drug-resistant epilepsy. Acta Neurol Scand. 2017;136:639–44.

    Article  CAS  PubMed  Google Scholar 

  41. Bazhanova ED, Kozlov AA, Litovchenko AV. Mechanisms of drug resistance in the pathogenesis of epilepsy: Role of neuroinflammation: a literature review. Brain Sci;11:9 https://doi.org/10.3390/brainsci11050663.

  42. Zhang C, Kwan P, Zuo Z, Baum L. The transport of antiepileptic drugs by P-glycoprotein. Adv Drug Deliv Rev. 2012;64:930–42.

    Article  CAS  PubMed  Google Scholar 

  43. Le A, Thomas M, Stallman B, Meadows K. Bhargava V. Refractory Epilepsy: Mechanisms of Pharmacoresistance. Georg Sci Res J; 2021. p. 99–110.

    Google Scholar 

  44. Alexopoulos AV. Pharmacoresistant epilepsy: Definition and explanation. Epileptology. 2013;1:38–42.

    Article  Google Scholar 

  45. Huff JS, Fountain NB. Pathophysiology and definitions of seizures and status epilepticus. Emerg Med Clin. 2011;29:1–13.

    Article  Google Scholar 

  46. Pitkänen A, Lukasiuk K, Dudek FE, Staley KJ. Epileptogenesis. Cold Spring Harb Perspect Med. 2015;5: a022822.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Shi Y, Zhang L, Teng J, Miao W. HMGB1 mediates microglia activation via the TLR4/NF-κB pathway in coriaria lactone induced epilepsy. Mol Med Rep. 2018;17:5125–31.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Zaben M, Haan N, Asharouf F, Di PV, Khan D, Ahmed A, et al. Role of proinflammatory cytokines in the inhibition of hippocampal neurogenesis in mesial temporal lobe epilepsy. Lancet. 2017;389:S105.

    Article  Google Scholar 

  49. Kaneko Y, Pappas C, Malapira T, Vale FĹ, Tajiri N, Borlongan CV. Extracellular HMGB1 Modulates Glutamate Metabolism Associated with Kainic Acid-Induced Epilepsy-Like Hyperactivity in Primary Rat Neural Cells. Cell Physiol Biochem. 2017;41:947–59.

    Article  CAS  PubMed  Google Scholar 

  50. Zhong H, Li X, Zhou S, Jiang P, Liu X, Ouyang M, et al. Interplay between RAGE and TLR4 Regulates HMGB1-Induced Inflammation by Promoting Cell Surface Expression of RAGE and TLR4. J Immunol. 2020;34:1900860.

    Google Scholar 

  51. Küper C, Beck F-X, Neuhofer W. Toll-like receptor 4 activates NF-κB and MAP kinase pathways to regulate expression of proinflammatory COX-2 in renal medullary collecting duct cells. Am J Physiol Physiol. 2011;302:F38–46.

    Article  Google Scholar 

  52. He M, Bianchi ME, Coleman TR, Tracey KJ, Al-Abed Y. Exploring the biological functional mechanism of the HMGB1/TLR4/MD-2 complex by surface plasmon resonance. Mol Med. 2018;24:21.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Iqbal M, Ho HL, Petropoulos S, Moisiadis VG, Gibb W, Matthews SG. Pro-inflammatory cytokine regulation of p-glycoprotein in the developing blood-brain barrier. PLoS ONE. 2012;7: e43022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Paudel YN, Othman I, Shaikh MF. Anti-high mobility group box-1 monoclonal antibody attenuates seizure-induced cognitive decline by suppressing neuroinflammation in an adult zebrafish model. Front Pharmacol. 2020;11: 613009.

    Article  CAS  PubMed  Google Scholar 

  55. Wang D, Liu K, Wake H, Teshigawara K, Mori S, Nishibori M. Anti-high mobility group box-1 (HMGB1) antibody inhibits hemorrhage-induced brain injury and improved neurological deficits in rats. Sci Rep. 2017;7:46243.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Daei S, Zheng Y, Wang Y, Chen Z. HMGB1, neuronal excitability and epilepsy. Acta Epileptol. 2021. https://doi.org/10.1186/s42494-021-00048-y.

    Article  Google Scholar 

  57. Zhu M, Chen J, Guo H, Ding L, Zhang Y, Xu Y. High Mobility Group Protein B1 (HMGB1) and Interleukin-1β as prognostic biomarkers of epilepsy in children. J Child Neurol. 2018;33:909–17.

    Article  PubMed  Google Scholar 

  58. Zhao J, Zheng Y, Liu K, Chen J, Lai N, Fei F, et al. HMGB1 is a therapeutic target and biomarker in diazepam-refractory status epilepticus with wide time window. Neurotherapeutics. 2020;17:710–21.

    Article  CAS  PubMed  Google Scholar 

  59. Avanzini G, Franceschetti S. Mechanisms of Epileptogenesis. Treat Epilepsy Third Ed. 2009;14:67–79.

    Article  Google Scholar 

  60. Maguire J. Epileptogenesis: more than just the latent period. Epilepsy Curr. 2016;16:31–3.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Thom M. Epilepsy pathology. Encycl. Neurol Sci. 2014;2:136–41.

    Google Scholar 

  62. Reddy DS. Role of hormones and neurosteroids in epileptogenesis. Front Cell Neurosci. 2013;7:1–20.

    Article  Google Scholar 

  63. Chiavegato A, Zurolo E, Losi G, Aronica E, Carmignoto G. The inflammatory molecules IL-1β and HMGB1 can rapidly enhance focal seizure generation in a brain slice model of temporal lobe epilepsy. Front Cell Neurosci. 2014;8:1–9.

    Article  Google Scholar 

  64. Sano F, Shigetomi E, Shinozaki Y, Tsuzukiyama H, Saito K, Mikoshiba K, et al. Reactive astrocyte-driven epileptogenesis is induced by microglia initially activated following status epilepticus. JCI Insight. 2021;6:1–15.

    Article  Google Scholar 

  65. Zhao J, Wang Y, Xu C, Liu K, Wang Y, Chen L, et al. Therapeutic potential of an anti-high mobility group box-1 monoclonal antibody in epilepsy. Brain Behav Immun. 2017;64:308–19.

    Article  CAS  PubMed  Google Scholar 

  66. Li Z, Li B, Zhu X, Yin P, Liu J, Huang S, et al. Neuroprotective effects of anti-high-mobility group box 1 antibody in juvenile rat hippocampus after kainic acid-induced status epilepticus. NeuroReport. 2013;24:785–90.

    Article  CAS  PubMed  Google Scholar 

  67. Kan M, Song L, Zhang X, Zhang J, Fang P. Circulating high mobility group box-1 and toll-like receptor 4 expressions increase the risk and severity of epilepsy. Brazilian J Med Biol Res. 2019;52:1–7.

    Article  Google Scholar 

  68. Sasaki T, Liu K, Agari T, Yasuhara T, Morimoto J, Okazaki M, et al. Anti-high mobility group box 1 antibody exerts neuroprotection in a rat model of Parkinson’s disease. Exp Neurol. 2016;275:220–31.

    Article  CAS  PubMed  Google Scholar 

  69. Zhang BF, Wang PF, Cong YX, Lei JL, Wang H, Huang H, et al. Anti-high mobility group box-1 (HMGB1) antibody attenuates kidney damage following experimental crush injury and the possible role of the tumor necrosis factor-α and c-Jun N-terminal kinase pathway. J Orthop Surg Res. 2017;12:4–10.

    Article  Google Scholar 

  70. Yang H, Liu H, Zeng Q, Imperato GH, Addorisio ME, Li J, et al. Inhibition of HMGB1/RAGE-mediated endocytosis by HMGB1 antagonist box A, anti-HMGB1 antibodies, and cholinergic agonists suppresses inflammation. Mol Med. 2019;25:1–13.

    Article  Google Scholar 

  71. Udpa N, Million RP. Monoclonal antibody biosimilars. Nat Rev Drug Discov. 2016;15:13–4.

    Article  CAS  PubMed  Google Scholar 

  72. Okuma Y, Liu K, Wake H, Zhang J, Maruo T, Date I, et al. Anti-high mobility group box-1 antibody therapy for traumatic brain injury. Ann Neurol. 2012;72:373–84.

    Article  CAS  PubMed  Google Scholar 

  73. Nakajo M, Uezono N, Nakashima H, Wake H, Komiya S, Nishibori M, et al. Therapeutic time window of anti-high mobility group box-1 antibody administration in mouse model of spinal cord injury. Neurosci Res. 2019;141:63–70.

    Article  CAS  PubMed  Google Scholar 

  74. Uezono N, Zhu Y, Fujimoto Y, Yasui T, Matsuda T, Nakajo M, et al. Prior treatment with anti-high mobility group box-1 antibody boosts human neural stem cell transplantation-mediated functional recovery after spinal cord injury. Stem Cells. 2018;36:737–50.

    Article  CAS  PubMed  Google Scholar 

  75. Zhang J, Takahashi HK, Liu K, Wake H, Liu R, Maruo T, et al. Anti-high mobility group box-1 monoclonal antibody protects the blood-brain barrier from ischemia-induced disruption in rats. Stroke. 2011;42:1420–8.

    Article  CAS  PubMed  Google Scholar 

  76. Ryman JT, Meibohm B. Pharmacokinetics of monoclonal antibodies. CPT Pharmacometrics Syst Pharmacol. 2017;6:576–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Elger CE. Epilepsy in 2015: Classic antiepileptic drugs under fire, and new options emerge. Nat Rev Neurol. 2016;12:2015–7.

    Article  Google Scholar 

  78. Uzawa A, Mori M, Taniguchi J, Masuda S, Muto M, Kuwabara S. Anti-high mobility group box 1 monoclonal antibody ameliorates experimental autoimmune encephalomyelitis. Clin Exp Immunol. 2013;172:37–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Schierbeck H, Lundbäck P, Palmblad K, Klevenvall L, Erlandsson-Harris H, Andersson U, et al. Monoclonal anti-HMGB1 (high mobility group box chromosomal protein 1) antibody protection in two experimental arthritis models. Mol Med. 2011;17:1039–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Su Z, Sun C, Zhou C, Liu Y, Zhu H, Sandoghchian S, et al. HMGB1 blockade attenuates experimental autoimmune myocarditis and suppresses Th17-cell expansion. Eur J Immunol. 2011;41:3586–95.

    Article  CAS  PubMed  Google Scholar 

  81. Orsini A, Foiadelli T, Costagliola G, Michev A, Consolini R, Vinci F, et al. The role of inflammatory mediators in epilepsy: Focus on developmental and epileptic encephalopathies and therapeutic implications. Epilepsy Res. 2021;172: 106588.

    Article  CAS  PubMed  Google Scholar 

  82. Musumeci D, Roviello GN, Montesarchio D. An overview on HMGB1 inhibitors as potential therapeutic agents in HMGB1-related pathologies. Pharmacol Ther. 2014;141:347–57.

    Article  CAS  PubMed  Google Scholar 

  83. Wang J, Levi J, Ellis L, Hill A. Minimum Manufacturing Costs, National Prices, and Estimated Global Availability of New Repurposed Therapies for Coronavirus Disease 2019. Open Forum Infect Dis. 2022;9:1–9.

    Article  Google Scholar 

  84. Samaranayake H, Wirth T, Schenkwein D, Räty JK, Ylä-Herttuala S. Challenges in monoclonal antibody-based therapies. Ann Med. 2009;41:322–31.

    Article  CAS  PubMed  Google Scholar 

  85. Awwad S, Angkawinitwong U. Overview of antibody drug delivery. Pharmaceutics. 2018;10:1–24.

    Article  Google Scholar 

  86. Dos Santos ML, Quintilio W, Manieri TM, Tsuruta LR, Moro AM. Advances and challenges in therapeutic monoclonal antibodies drug development. Brazilian J Pharm Sci. 2018;54:1–15.

    Google Scholar 

  87. Sousa F, Fonte P, Cruz A, Kennedy PJ, Pinto IM, Sarmento B. Polyester-based nanoparticles for the encapsulation of monoclonal antibodies. Methods Mol Biol. 2018;1674:239–53.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable

Funding

There is no funding.

Author information

Authors and Affiliations

Authors

Contributions

The initial concept for this literature review was hatched by BGL. The text was written by BGL, SGT, JEF, and KAP with guidance from DKIU. BGL, SGT, JEF, and KAP completed, copyedited and revised the manuscript. All of the authors actively participated in the writing or revision of the article knowledge-based material. The final text has been read by all of the writers and have all given their consent publication. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Bryan Gervais de Liyis.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All authors have given their consent for publication.

Competing interests

There is no competing interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

de Liyis, B.G., Tandy, S.G., Endira, J.F. et al. Anti-high mobility group box protein 1 monoclonal antibody downregulating P-glycoprotein as novel epilepsy therapeutics. Egypt J Neurol Psychiatry Neurosurg 58, 121 (2022). https://doi.org/10.1186/s41983-022-00557-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41983-022-00557-8

Keywords