GET THE APP

A Toll-Like Receptor 3-Agonist as Promising Candidate in Multiple
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2015) Volume 6, Issue 4

A Toll-Like Receptor 3-Agonist as Promising Candidate in Multiple Sclerosis Treatment

Maria Elsa Gambuzza1*, Vincenza Sofo2, Francesca Maria Salmeri2, Luca Soraci2, Silvia Marino3 and Placido Bramanti3
1Ministry of Health, Territorial Office of Messina, Italy
2Department of Environmental Protection, School of Medicine, University of Messina, Italy
3IRCCS Centro Neurolesi “Bonino Pulejo”, Messina, Italy
*Corresponding Author: Maria Elsa Gambuzza, Ministry of Health, Territorial Office of Messina, Italy, Tel: +39 090 2213348, Fax: +39 090 2212664 Email:

Abstract

Perturbations in immune processes play an important role in multiple sclerosis (MS), an autoimmune disorder where specific innate immune pattern-recognition receptors (PRRs), such as Toll-like receptors (TLRs) have recently been shown to play a major role in the initiation disease, the triggering of relapses, and regulation of CNS damage. The abnormal immune response in MS has been shown to be dependent on genetic background, despite environmental factors, including pathogens capable of overstimulating innate immune response through TLRs, appear to contribute to the development of autoreactive T cells that in turn cause myelin damage. However, whereas the upregulation of most TLRs plays a detrimental role in MS pathogenesis, recent studies suggest an ameliorative role of TLR3 in the onset and progression of MS and experimental autoimmune encephalomyelitis (EAE), a murine model of MS. TLR3 activation appears to protect from the disease, mainly through induction of interferon (IFN)β. Therefore, TLR3 stimulation with synthetic immunomodulators could represent a potential alternative approach in MS therapy. Among the investigational compounds TLR3-targeting, the mismatched double-stranded RNA molecule Ampligen®, can offer promise in the treatment of relapsing MS patients. Ampligen® is currently in phase III clinical trial in the treatment of chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME), an illness that shows remarkable levels of similarity with MS. The aim of this paper is to provide a brief overview about Ampligen® historical development, clinical pharmacology, clinical trials, and safety data, and to discuss about its potential role in MS treatment in the context of existing therapeutic options.

Keywords: Multiple sclerosis; Innate immunity; Toll-like receptors; Toll-like receptor 3-agonist; Interferon-beta.

Abbreviations

CFS/ME: Chronic Fatigue Syndrome/Myalgic Encephalomyelitis; CNS: Central Nervous System; DMAs: Disease-Modifying Agents; dsRNA: double-stranded Ribo Nucleic Acid; EAE: Experimental Autoimmune Encephalomyelitis; FDA: Food and Drug Administration; IFN: Inferferon; IKKε: IκB Kinase Epsilon; IRF: Interferon Regulatory Factor; JNK: c-Jun Amino-Terminal Kinase; MAP: Mitogen-Activated Protein; MS: Multiple Sclerosis; PAMPs: Pathogen-Associated Molecular Patterns; PRRs: Pattern-Recognition Receptors; TBK: TANK-Binding Kinase; TIR: Toll/IL-1 Receptor; TLRs: Toll-Like Receptors; TRAF: TNF Receptor Associated Factor; TRIF: TIR-Domain-Containing Adapter-Inducing IFNβ

Introduction

Multiple sclerosis (MS) is an autoimmune disorder characterized by demyelination, chronic inflammation and neuronal damage, mainly induced by activation of auto-reactive immune cells directed against myelin [1]. This abnormal immune activation appears to be triggered by a combination of genetic and environmental factors, including pathogens [2]. Several pathogens, that are likely involved in MS development, have shown to be capable of overstimulating innate immune response, leading to the development of autoreactive T cells, which play important roles in mediating demyelination and axonal damage [3]. More specifically, immune cells, including monocytes, dendritic cells, NK cells, CD4+ and CD8+ T cells, and B cells, are activated and migrate into the central nervous system (CNS), where mediate myelin destruction, axon damage and neuronal apoptosis [4]. These immunological phenomena have been recently shown to be the result of an initial activation of the innate immune response mediated by specific pattern-recognition receptors (PRRs), among which the Toll-like receptor (TLR) family represents a major component [2,4]. Pathogens able to induce innate immune responses TLR-mediated in the CNS seem to be responsible for the development of autoreactive T cells due to antigen spread, in a process known as bystander activation [2,5]. TLRs can be stimulated both by conserved structures of microbial agents, also known as pathogen-associated molecular patterns (PAMPs) [6-9], and/or by host-derived ligands, also known as damage-associated molecular patterns (DAMPs) [10]. Both PAMPs and DAMPs include various compounds, such as proteins, glycosaminoglycans, glycoproteins, RNA, and DNA. TLRs play then a crucial role in danger recognition and immune response induction, and the activation of TLR-mediated pathway modulates inflammatory responses and primes antigen-specific adaptive immunity [4]. In the CNS, TLRs play a key role in the development and regulation of inflammation, neuronal degeneration, and brain trauma, but they also provide to neurogenesis and neurite outgrowth [11]. In addition, they contribute to defence against hamful pathogens, and neurotoxic compounds, such as the “altered self” molecules and apoptotic cells [11]. This important and dual role of TLRs has been also outlined in several neurodegenerative disorders, including MS [4]. Results from recent studies showed that in this disabling disease the activation of specific TLRs, including TLR2 and TLR4, induces the differentiation of autoreactive T cells and the production of proinflammatory cytokines, so contributing to CNS damage [12]. In contrast, the upregulation of TLR3 has been shown to play a beneficial role through the production of endogenous IFNβ, that is capable of inhibiting the production of harmful cytokines, therefore improving MS severity [13-16]. The evidence for a neuroprotective role played by this receptor is further supported by studies showing that TLR3 activation and the downstream processes are capable of inducing a functional inhibition of neuronal cell death in human cell cultures, and this evidence also suggests additional roles for TLR3-mediated signaling in the CNS [17-18]. The possibility to modulate immune response with specific TLR agonists or antagonists and by inhibiting intracellular proteins involved in the signaling cascade has sparked great interest as alternative approach to treat immunological disorders, including MS [13]. Among the immunotherapeutic compounds TLR-targeting that are in preclinical or clinical studies for other diseases, specific TLR3 agonists could have beneficial effects in MS treatment [16]. According to recent studies the TLR3 agonist Rintatolimod (tradename Ampligen®, also known as poly(I):poly(C12U) (Hemispherx, Biopharma Inc., PA, USA), has shown promising results in the treatment of cancer, HIV, influenza, hepatitis B and C infection, and chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME), an illness that shows remarkable levels of similarity in phenomenology and neuroimmune characteristics with MS [19-22]. Therefore, this drug could represent a safe and effective treatment for MS patients, and could be considered a valuable adjunct to multiple different immunotherapies currently in use or in development [13]. This article reviews the potential therapeutic role of Ampligen® in the context of current and emerging options for the treatment of MS, by mainly focusing on the relevant findings that could promote its use in MS therapy, and discussing about the development, pharmacological properties, safety and tolerability of the new molecular formula.

TLR Signaling Pathways and the Role of Tlr3 in MS

TLRs are classically defined as a large family of PRRs that represent an important component of the innate immune response and form a primary danger signal response to the presence of microbial pathogens detected both internally and externally in the tissues and cells of higher organisms. TLRs are expressed on many types of innate immune cells [23,24], as well as non-immune cells, such as epithelial and endothelial cells [25-30]. In the brain TLRs are constitutively expressed by both infiltrating [4] and resident immune cells [31], including T cells, microglia [32] and astrocytes [33], that represent the key sentinels of innate CNS immunity [34]. TLRs are also expressed by other brain cells not strictly involved in the CNS immunity, such as oligodendrocytes [35], neurons and neuronal progenitor cells [36]. These receptors are characterized by a leucine-rich repeat (LRR) ectodomain, which mediates the ligand binding, a transmembrane region, and a Toll/IL-1 receptor (TIR) endodomain, with a highly conserved structure, required for downstream signal transduction [37]. Among the 10 human TLRs described in the literature, TLR1, TLR2, TLR4, TLR5, TLR6, and TLR10 are expressed on the cell surface, where they can be stimulated by molecular components, including proteic and lipidic PAMPs and DAMPs. In contrast, TLR3, TLR7/8, and TLR9, that are located in endosomal/lysosomal compartments and endoplasmic reticulum, have evolved to detect dsRNA, ssRNA, and ssDNA respectively [38-42]. The TLR pathways have been extensively reviewed elsewhere [43]. TLR stimulation in the CNS results in the production of cytokines, such as IL-6, IL-1β, and TNF-α, that promote the destruction of blood brain barrier and the recruitment of lymphocytes into sites of inflammation, where they can further promote inflammatory response and induce adaptive autoimmunity [44]. However, there is an increasing body of evidence that TLR signaling can also mediate beneficial effects, since they are also capable of influencing multiple dynamic processes in the developing and adult CNS, including neurogenesis, axonal growth and structural plasticity [45]. Altogether, the functional outcome of TLR-induced activation in the CNS is related to a correct balance between protective and harmful effects [45]. The subcellular distribution of TLRs in CNS is almost similar to others cell types [46], although there are some tissue-specific differences [47]. All TLRs signal through MyD88 downstream adaptor, except TLR3 and a subset of TLR4 signaling events, which are mediated by the exclusive use of the TIR-domain-containing adapter-inducing IFNβ (TRIF) [39]. Analysis of TLR gene expression in the CNS detected high expression of TLR3. More specifically, high levels of TLR3 were shown to be expressed on the endoplasm of astrocytes and a broad range of antigen-processing cells, tissue dendritic cells, mast cells, monocytes, natural killer, and other immune cells [48], in addition to cerebral endotelial cells, neurons, microglia, astrocytes and oligodendrocytes [49-51]. The MyD88-independent and TRIF-dependent signaling pathway TLR3-mediated leads to the production of type I IFNα/β as result of NF-κB, interferon-regulatory factor (IRF)-3, and mitogen-activated protein (MAP) kinase signaling, that is in turn mediated by p38 and c-Jun amino-terminal kinase (JNK) [52]. Type I IFNα/β are best known for their effects in viral infections, but they are also capable of inhibiting inflammatory responses [53,54], and this supports the hypothesis that specific TLR3 agonists could play a beneficial role, IFNβ-mediated, in several inflammatory diseases, as well as MS [4,13,55,56]. In addition, TLR3/TRIF-dependent signaling pathway has been shown to play a further positive roles, being capable both of triggering neuroprotective responses in astrocytes and controlling the growth of axons and neuronal progenitor cells [18]. TLR3 is triggered by dsRNA with a minimum size of at least 50 base pairs and by specific endogenous ligands, as well as the endogenous microtubule regulator stathmin, taken up into the endosome [57]. The dsRNA molecules can originate from the genome of specific viruses, can be also intermediates generated during the viral RNA replication, or dsRNA produced intracellularly by stem-loop forming or with siRNA-aligned mRNAs [58]. The size requirement or discrimination of dsRNA by TLR3 prevent responses to non-microbial sources of dsRNA, including micro or transfer RNA. Above 50 base pairs, binding affinity is a function of size with a progressive enhancement in binding affinity directly correlated to increased lenght in linear non-branched dsRNA [59]. The extracellular domain of TLR3 dimerizes when it binds dsRNA, and this dimeric complex appears to be composed of two dsRNA binding domains located near the N-terminus and the C-terminus domain, and when combined with dsRNA, a sole dsRNA molecule associates two TLR3 molecules through four dsRNA binding sites, to form an “m” shaped dimer” [60-62]. The TIR domain of TLR3 activated associates directly with the TIR domain of TRIF protein, that in turn activates the TNF receptor associated factor (TRAF)3. TRAF3 induces the activation of TANK-binding kinase (TBK)1, which then phosphorylates IRF3, leading to its dimerization and translocation to the nucleus, where it stimulates the transcription of the gene encoding IFNβ [61,63]. Peli1, a member of Pellino family, plays also an important role in the TLR3-TRIF-TBK1 signaling pathway, driving the positive feedback loop [64]. More specifically, Peli1, activated by TBK1 and IκB kinase epsilon (IKKε) by induction of its covalent modification, presumably phosphorylation, induces the interaction of IRF3 with the IFNβ promoter. Peli 1 not only stimulates the initial phase of IFNβ formation, but also drives the positive feedback loop by which the small amounts of IFNβ formed initially amplify IFNβ production. In this pathway, IFNβ stimulates the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway, leading to STAT1 and STAT2 phosphorylation [65,66], which is followed by the formation of a complex between STAT1/2 heterodimer and IRF9, to form the IFN-stimulated transcription factor (ISGF)3. This multimeric regulator of transcription stimulates IRF7, that in turn can activate the IFNβ promoter either directly or as heterodimeric complex with IRF3, or by stimulating the transcription of other proteins of this positive feedback loop, such as IFNα. Altogether, dsRNA, acting as agonist of TLR3, is capable of inducing the stabilization of IFNβ mRNA by a TRIF-dependent mechanism, so leading to ehnanced IFNβ levels [67] (Figure 1).

cellular-immunology-multimeric-regulator

Figure 1: TLR3-TRIF mediated pathway. TLR3 is the only TLR that exclusively signals through the TRIF-dependent pathway. The association of TLR3/TRIF TIR domains activates TRAF3 and the downstream signaling events, represented by IKK/NF-κB pathway, that in turn is regulated by Peli 1. Peli 1, activated by TBK1 and IKKε, induces the IRF3 phosphorylation, dimerization and translocation to the nucleus, so leading to IFNβ gene transcription. IFNβ stimulates the JAK/STAT pathway, that induces the STAT1 and STAT2 phosphorylation, and the association of STAT1/2 with IRF9. This multimeric regulator stimulates IRF7, that in turn activates the IFNβ promoter either directly, or as IRF3/IRF7 heterodimer [67].

Recent evidence also supports the positive role of TLR3/TRIF-mediated pathway in experimental autoimmune encephalomyelitis (EAE) mouse models, that represents the most commonly studied animal of MS [68]. In EAE mice TLR3 stimulation leads to the release of IL-27, that in turn mediates the inhibition of Th17 cells, that are known to play a critical role in the disease [69-71]. Other studies supporting this evidence showed that EAE mice, inoculated intraperitoneally with the synthetic TLR3 agonist poly(I):poly(C) acid, produced high levels of IFNβ, that significantly reduced disease severity [16]. Normal adult human astrocytes have shown to increase the production of anti-inflammatory cytokines such IL-10, and to downregulate proinflammatory cytokines such as IL-12 (p40) and IL-23, in response to TLR3 ligation [48]. Recent studies have also shown that the endogenous TLR3 ligand stathmin, identified in astrocytes, microglia, and neurons from human MS-affected brains, is capable of stimulating the same set of neuroprotective factors induced by poly(I):poly(C) [18].

Development of Ampligen

The invention of Ampligen® poly(I):poly(C12U) [72], relates to a novel synthetic mismatched dsRNA with an high specificity of binding to TLR3, that results in a selective activation of TLR3-mediated immune responses maybe depending on transient expression of multiple genes and subsequent activation of a signal transduction cascade. The genes induced by TLR3 activation include IFNs, 2-5’ adenylate synthetase, and protein kinase (p68) [73,74]. As reported by historical overviews, “this synthetic dsRNA molecule, first developed in the 1960’s by Merck and Co. to reduce tumor formation, resulting effective but too toxic for use, languished, until Dr. William Carter, working with other Johns Hopkins University researchers in the 1970’s, was able to reduce its toxicity” by adding uridylic acid molecules at specific intervals along the dsRNA molecule, so obtaining a particular dsRNA, denoted poly(I):poly(C12U), wherein one of the two polyribonucleotides is polyriboinosinic acid and the other is polyribocytidyl C12, uridylic acid. The new compound, called Ampligen® (for AMPLIfied GENetic activity) was capable of stimulating IFN production like poly (I): poly(C), but resulted less toxic [75]. Its smaller and rugged molecular structure, as measured by physico-chemical techniques, appeared more resistant to molecular unfolding, including denaturation and branching, and this led to an increase in bioactivity, due to higher ability to bind TLR3. Ampligen® was synthesized acting on the hypothesis that nucleotide sequence requirements for beneficial and adverse effects are different. More specifically, it was obtained by preserving the RNA double helical structure, that is required for TLR3 activation and type I IFN production [76], and by modifying the molecular folding with the occasional introduction of uridylate into the poly(C) strand, in order to produce supplexes free of adverse effects and containing specifically-configured regions which are not base paired (i.e. mismatched) at the position of the modifications [72] (Figure 2). This modified dsRNA is capable of activating TLR3-TRIF mediated pathway selectively, without activating other TLRs like TLR4 or RNA helicases like RIG-I or MDA-5, and without inducing proinflammatory responses and a TNFα/TNFR1 dependent toxicity, as seen with the non-selective TLR3 agonist poly (I):poly(C) in the phenomenon known as “cytokine storm” [19,56,77-79].

cellular-immunology-hybridized-bases

Figure 2: Partial view of poly(I):poly(C12U) partially hybridized strands and the interaction of bases of individual poly(I) and poly(C12U) strands [72].

The mismatched regions accelerate dsRNA hydrolysis, further reducing the toxicity [59,80], while retaining the ability of the nucleotide sequence to induce IFN synthesis, as well as its stability [81]. The hydrolysis is highly dependent on nucleic acid structure, as well as on the presence of nuclease and divalent cations, pH, and temperature. RNA is more susceptible to degradation than DNA, due to the ability of its 2’-OH groups to act as nucleophiles so facilitating hydrolysis [82]. Moreover, poly(I):poly(C12U) was designed to be degraded more rapidly than other dsRNA in a nuclease-containing environment, such as blood and other tissue fluids. In contrast, it is stable in physiological salt buffers at room temperature, but overtime it is degraded in a time- and temperature-dependent pattern [72]. To improve the biological activity of Ampligen®, that resulted lower than expected, a new version of dsRNA with a superior biological and therapeutic profile, called “rugged” dsRNA, was recently obtained by purification (by HPLC or other cromatographic methods) from the Ampligen® mixture, by subjecting the partially hybridized strands of a population of dsRNAs to conditions that denature most dsRNAs, and then selecting dsRNA molecules that remained partially hybridized. Chemically, Ampligen® is a poly(I):poly(C30-35U), wherein C30-35U indicates of 1 U for 30-35 C. The minimal lenght of this rugged dsRNA is about 50 base pairs requiring about 4 to 5 helical turns. This “rugged” dsRNA is resistant to unfolding (i.e. denaturation) of its helical structure and shows a reduced tendency to form branched dsRNA molecules. In addition, it has an improved dsRNA activity as potent and highly selective TLR3 agonist. Immune cells may be susceptible to specific cytokine response patterns activated by rugged dsRNA. Ampligen® can be administered by intravenous infusion, intradermal, subcutaneous, or intramuscular injection, intranasal or intratracheal inhalation, oropharyngeal or sublingual application, or transocularly [72].

Clinical Pharmacology of Ampligen® and Mechanism of Action in CFS/ME

In 2009 Ampligen® was undergoing clinical trials to treat specific immune-mediated diseases, including CFS/ME, multidrug resistant HIV/AIDS, and hepatitis C. Most of preclinical data available so far were recently discussed in the Arthritis Advisory Commitee Meeting: Ampligen® - Treatment of chronic fatigue syndrome (2012). The safety profile and immunomodulatory effects of Ampligen®, were recently shown in the treatment of CFS/ME, a debilitating disorder characterized by disabling fatigue and a combination of flu-like symptoms [21,83-86], and considered by the Centers for Disease Control and Prevention (CDC) as an economically and emotionally devastating illness, whose functional impairment can be equivalent to MS [86]. Since the main hallmark of CFS/ME is represented by the fatigue, that appears not improved by bed rest and may be worsened by physical activity, cardiopulmonary exercise tolerance testing was used as an objective measurement of treatment efficacy and it was accepted as a regulatory standard for drugs ameliorating exertional fatigue [21]. Recent studies have shown that Ampligen® is able to induce objective improvement in exercise tolerance in CFS/ME patients receiving the drug for 40 weeks, related to concomitant medication usage as well as other secondary outcomes, including the drugs commonly used by patients in an attempt to alleviate the symptoms of the disease [21]. The mechanism of action of Ampligen®, that has reached Phase III clinical trials in CFS/ME [21], consists mainly of selectively modulating the activity of an important part of the innate immune defense, represented by RNase L enzymatic pathway that some studies suggest is disrupted in CFS/ME patients [87]. RNase L is an highly regulated latent endoribonuclease induced by INFs and activated by dsRNA. Regulated turnover and processing of ssRNA, such as RNA produced during the viral replication cycle, by RNase is essential for a complete IFN response. This pathway has recently been shown to contribute to innate immunity and appears to protect the CNS against viral-induced demyelination [88]. In the patent titled “Methods of treatment of chronic immune diseases”, the inventors, who detailed their findings concerning the treatment of the two chronic immune diseases, represented by CFS/ME and MS, stated also that the CFS/ME diagnosis is accomplished by detecting the presence of RNase L fragments [89]. In addition to ability of Ampligen® to modulate RNase L enzymatic pathway, its agonistic activity towards TLR3, that is involved in the early recognition of pathogens, could also contribute to reduce the rates of opportunistic infections, that are increased in CFS/ME patients [90]. In CFS/ME patients, the beneficial effects of Ampligen® have shown to be related to its binding to the active sites of the TLR3 homodimer, represented by the N-terminals of each TLR3, with the subsequent IFN/RNase L enzyme pathway stimulation, IFN production, and oligoadenylate synthase-RNase L pathway activation [74,91]. RNase L plays a protective role in CNS demyelination viral-induced, since it is capable of both inhibiting the viral genome translation, and inducing apoptosis of infected cells, allowing then the propagation of IFNα/β pathways enhanced by RNA degradation products [92].

There are various local or systemic routes of administration available for Ampligen®, including enteral, topical, and parenteral. Consequently, permutations for administration, including acqueous solutions, powders, granules, containing different eccipients like binding agents, lubrificants, preservatives, and others are related to its use. In CFS/ME patients, Ampligen® is usually given intravenously twice weekly for an hour at the standard dose of 400 mg, even if the correct dosage is variable, often being too high and the appropriate dose being as low in some patients as 25 mg [93]. While 6 months may be enough to produce a significant response, 12-18 months are recommended depending on the patient’s condition. Since the “rugged” dsRNA obtained from Ampligen® mixture represents a purified compound with rugged physico-chemical structure and highly specific biologic activity, its recommended dosage should be lower, being comprised from about 10 mg to about 40 mg, albeit the dose amount and/or frequency may be varied in response to the subject’s symptoms, and with the age, conditions, gender, or health status. Recently, an oral form of Ampligen® (Oragen) is being evaluated [93]. No significant side effects are been reported, except for temporary and moderate flu-like symptoms, chills, vasodilatation and shortness of breath, that appeared increased in some patients, although these side effects appeared lower compared with other immunomodulating drugs [21]. Nevertheless, the high cost for the “infusion terapy”, that ranges from $1-2,000/month; $15-25,000 a year, depending on dosage, could greatly reduce the probability of using this compound in day-to-day practice. At present, data from clinical trials are not yet considered sufficient by the US Food and Drug Administration (FDA), in order to obtain the approval of Ampligen in CFS/ME therapy. In particular, the FDA claims that CFS/ME does not have clear biomarkers such as blood tests, although cardiopulmonary exercise tolerance testing could be used to define which patients are most likely to respond to the drug [94]. However, studies performed in CFS/ME patients receiving Ampligen®, reported positive and significant effects on increasing exercise tolerance, and these data, together with the safety profile and high specific immunostimulatory properties of this molecule, may represent a strong motivation to assess its potential role in MS treatment [95]. The only question about Ampligen®, concerning the study showing that TLR3 can mediate West Nilus virus entry into the brain, causing lethal encephalitis [96], has been overcome in more recent studies that, in contrast, showed that TLR3 can play a protective role also against West Nile virus, by partially restricting its replication in neurons [97]. Further studies concerning the manipulation of innate immune response as antiviral therapy showed that the use of Ampligen prevents Venezuelan encephalitis equine, despite it has not yet been evaluated in CNS infections [98]. In addition, independent researchers have demonstrated the antiviral activity of Ampligen against flaviviruses, including West Nile, Dengue fever virus and Japanese encephalitis virus as well as virus classes associated with bioterrorism [99].

Ampligen® in MS Treatment

MS is considered a form of encephalomyelitis disseminata showing remarkable levels of similarity with CFS/ME. Indeed, both disorders, classified as diseases of the CNS by the World Health Organization, show remarkable phenomenological and neuroimmune overlaps [100] (Table 1). Other remarkable levels of similarity concern “the findings produced by neuroimaging techniques that appear quite similar in both illnesses and show decreased cerebral blood flow, atrophy, gray matter reduction, whit matter hyperintensities, increased cerebral lactate and choline signaling, and lowered acetyl-aspartate levels” [101]. The neuroimmune similarities between MS and CFS/ME, are mainly based on shared immunoinflammatory oxidative and nitrosative stress, autoimmune and mitochondrial pathways. However, the two diseases show also remarkable differences, since MS is considered an immunologically mediated disease resulting in the demyelination of brain and spinal cord white matter disease, characterized by multifocal lesions, the MS plaques, which consist of a well-demarcated hypocellular area characterized by the loss of myelin, the formation of astrocytic scars, and the mononuclear cell infiltrates concentrated in perivascular spaces, where activated mononuclear cells, including lymphocytes, microglia, and macrophages destroy myelin [101]. Biochemical analysis of the spinal fluid provides evidence of the inflammatory response in the CNS. The two well-established spinal fluid markers used for the diagnosis of MS are represented by “oligoclonal bands and IgG index” something that is not found in CFS. Altogether, both MS and CFS/ME remain unsolved disorders with multiple symptoms and no single causative factor, with a potential to be described as inflammatory diseases of the CNS disorder yet to be identified [102,103].

Significant overlaps in MS and CFS/ME
Symptomatic similarities Disabling fatigue with chronic course, and relapsing-remitting nature
Muscle symptoms: exercise intolerance, pain/myalgia, fasciculations
Gastrointestinal dysfunction
Brain and CNS symptoms: mental fatigue, cognitive dysfunction (problems with short-term memory, concentration and mainaining attention), disequilibrium, word finding disabilities, headaches and migraines
Abnormalities in immune system function: sore throats, joint pains, intermittent flu-like feelings
Problems related with control of the autonomic nervous system: palpitations, postural hypotension
Disease exacerbated by stress and psychological stress
Similarities in immunoinflammatory, oxidative and nitrosative pathways Oxidative and nitrosative stress (O+NS)
Depressed levels of antioxidants and antioxidant enzymes (Vitamin E, ubiquinone, gluthatione peroxidase)
Chronic activation of immunoinflammatory pathways
Increased levels of proinflammatory cytokines (TNF-α,IL-1, IL-6)
COX2 and NFkB upregulation
T regulatory (Treg) dysfunction.
Coesistence of a T helper (Th)1 and Th2 response
Similarities in autoimmune responses Oxidative and nitrosative stress (O+NS)
Depressed levels of antioxidants and antioxidant enzymes (Vitamin E, ubiquinone, gluthatione peroxidase)
Chronic activation of immunoinflammatory pathways
Increased levels of proinflammatory cytokines (TNF-α, IL-1, IL-6).
Detection of autoantibodies (anti-nuclear, anti-cardiolipin, anti-phospholipid, anti-neuronal, anti-muscle)
IgM responses against palmitic and oleic acids
Antibodies against byproducts of lipid peroxidation
Response to B-cell depression treatment (e.g. Rituximab)

Table 1: Main similarities between MS and CFS/ME.

It has been recently reported that innate immune phaenomena concur in activation of autoimmune and inflammatory responses, as also shown by the increased expression of TLRs observed whitin the CNS during MS [13,104,105].

TLRs have shown to play a critical role in modulating cytokine and chemochine secretion in MS and its animal models and, more specifically, whereas MyD88-dependent pathways have shown to contribute to MS and EAE pathology [106], MyD88-independent pathways appear to mitigate disease severity. In particular, TLR3 agonists might favour the inhibition of signaling responsible for autoimmune and inflammatory responses in MS and EAE [107]. In addition, TLR3 stimulation leads to endogenous induction of IFNβ, that has shown to prevent inflammation and demyelination and, unlike the exogenous IFN used in MS therapy, does not induce antibodies anti-IFN [13,105]. IFNβ has also shown to increase the expression of CD73 on endothelial cells. CD73 ectoenzyme on CNS produces adenosine from AMP and adenosine possess both anti-inflammatory and neuroprotective activity [108].

At light of this, Ampligen® could offer promising results for this autoimmune disease that at present has no cure. Despite the pharmacological armamentarium for MS has been significantly expanded in the last years and new effective therapies have been proposed to modify the disease course, the most common treatments usually focus on strategies to treat MS attaks, manage symptoms and reduce the progress of the disease. As inflammation is the main factor contributing to axonal pathology, aggressive anti-inflammatory treatment is used to contribute to reducing the lesions and preventing axonal injury. A preliminary trial of poly ICLC in chronic progressive MS was conducted in 1985 [109,110]. In these studies, performed before the discovery of TLR-mediated mechanisms, MS patients were treated in an open preliminary trial of poly ICLC to induce endogenous type I IFN; such studies were limited by poly-IC toxicity. Another more recent study, that evidenced the beneficial role of endogenous production of IFNβ TLR3-mediated, showed that myeloid heme oxygenase-1 was required for the regulation of IFNβ production after TLR3 stimulation [111]. This study confirmed the previous observations of Touil et al. in SJL/J mice with relapsing EAE and C57BL/6 mice with chronic EAE [16]. IFNβ1b (Betaseron) was the first disease-modifying therapy approved by US FDA and also recognized as Betaferon by European Medicines Agency (EMA), in 1995. To date, a number of approved disease-modifying treatments have been shown to slow the progression of MS. These drugs may be included into two main categories: treatments that allow to improve symptom management, and treatments capable of slowing the progression of the disease. Currently, there are at least 8 different products approved by the FDA as disease modifying treatments for MS [112]. These include IFNβ1a compounds, such as Avonex® and Rebif®, IFNβ1b, such as Betaferon®/Extavia, the mimetic compound glatiramer acetate (Copaxone®), containing a group of aminoacids that look like myelin, the chemotherapic agent mitoxantrone (Novantrone®), acting by suppressing the activity of T and B cells, and the humanized monoclonal antiboby natalizumab (Tysabri), able to bind to specific receptors on immune cells that allow it to enter the brain and the spinal cord. Nevertheless, none of these compounds have shown to be effective against this autoimmune disease. In addition, the most commonly used first-line agent, represented by IFNβ, in a range of treated people comprised between 2 and 45%, can induce neutralizing antibodies that significantly reduce its biological activity, so leading to loss of clinical effects [113]. Many different innovative compounds are been approved for the safety and efficacy profiles in MS therapy, including specific immune-targeting humanized monoclonal antibodies such as alemtuzumab, immunomodulators, such as dimethyl fumarate, immunosoppressive compounds such as teriflunomide, but we are still far from finding a cure for MS. In addition, more drugs require long-term and regular administration via parenteral or subcutaneous, and this results unconfortable and inconvenient for the patients affected by this chronic disorder. Therefore, the most important factor in the future development of MS drugs is represented by the achievement of effective medications, possibly orally administrable, together with additional therapies for halting neurodegeneration, promoting remyelination and neuronal repair [114]. At light of this, Ampligen®, and more in particular, the new version of “rugged” molecule, can show promise as a relatively safe and efficacious drug in the treatment of MS, both as monotherapy or as an add-on agent to first-line disease-modifying agents (DMAs). In addition, the possibility of oral administration makes it an attractive option in addition to the available therapeutic armamentarium to manage relapsing MS. Among the adverse drug events that could affect MS patients taking Ampligen®, already reported in some clinical trials, in addition to flu-like syndrome, chills, vasodilatation and dyspnea, that altogether are side effects not particularly severe, a systemic inflammatory response dose-dependent TLR3-induced could contribute to the disease progression. However, despite toxicological analysis demonstrated the occurrence of systemic dose-dependent inflammatory cytokine responses in rats, more recent studies showed that primates are resilient to inflammatory cytokine toxicity induced by non-MyD88-dependent, TRIF-mediated activation of TLR3 [115,116], and these data appear consistent with differential TLR3 nuclear transcriptional activities, with the NF-κB pathway in inflammatory cytokine production in primates playing a relatively minor role compared with the IRF-3/IRF-7 nuclear IFN inducers [115].

Conclusions and Future Directions

Although there are many effective ways of managing MS, at present there is no cure for this disease. The modest effects in stabilizing disease, due to induction of anti-drug neutralizing antibodies against the first-line immunotherapeutic compound, represented by IFNβ, have reduced the efficacy of this important drug, similarly to many other biopharmaceuticals used in other chronic diseases (e.g. insulin, factor VIII). Nevertheless, the efficacy of IFNβ in order to counteract inflammatory processes, always reduced by host response, should encourage implementation of alternative approaches capable of triggering endogenous IFNβ production. In this case, Ampligen® could be effectiveness if used as an add-on agent to DMAs, being potentially able to induce similar or greater effects, when compared to exogenous IFNβ administration, togheter with the added convenience of oral administration. More specifically, Ampligen® might be considered for use in MS patients with suboptimal responses to IFNβ and who are reluctant or unable to use other approved DMAs. Future results from ongoing large-scale phase III clinical trials performed on CFS/ME patients will provide additional information on its effectiveness and tolerability, although its high cost could reduce the probability that this treatment can be incorporated into daily practice. Overall, the future appears bright for MS patients, since the modulation of innate immune response with this small molecule TLR3-targeting might represent a promising approach. The future will likely see other specific immunomodulators being utilized, perhaps along with other drugs able to prevent axonal degeneration and stimulate repair of damaged axon. Another winning point of TLR-targeting drugs is that they have fewer side effects and lower or no toxicity, compared with drugs commonly used in MS treatment, and this represents an important feature, since MS is a chronic disease that requires long-term treatments. Ongoing studies of innate immune pathways involved in autoimmunity and neurodegeneration could reveal new biological insights. In view of the likely overall complexity of TLR functions, additional advances will arise through the use of genomic, proteomic and other systems biology approaches. These approaches will allow to identify processes and pathways amenable to therapeutic manipulation, in order to ehnance innate immune responses and counteract autoimmune and neuroinflammatory processes, improving overall clinical outcomes in MS patients.

References

  1. Friese MA, Fugger L (2009) Pathogenic CD8(+) T cells in multiple sclerosis. Ann Neurol 66: 132-141.
  2. Racke MK, Drew PD (2009) Toll-like receptors in multiple sclerosis. Curr Top Microbiol Immunol 336: 155-168.
  3. Libbey JE, Cusick MF, Fujinami RS (2014) Role of pathogens in multiple sclerosis. Int Rev Immunol 33: 266-283.
  4. Miranda-Hernandez S, Baxter AG (2013) Role of toll-like receptors in multiple sclerosis. Am J Clin Exp Immunol 2: 75-93.
  5. McMahon EJ, Bailey SL, Castenada CV, Waldner H, Miller SD (2005) Epitope spreading initiates in the CNS in two mouse models of multiple sclerosis. Nat Med 11: 335-339.
  6. Akira S (2009) Innate immunity to pathogens: diversity in receptors for microbial recognition. Immunol Rev 227: 5-8.
  7. Galdiero M, Galdiero M, Finamore E, Rossano F, Gambuzza M, et al. (2004) Haemophilus influenzae porin induces Toll-like receptor 2-mediated cytokine production in human monocytes and mouse macrophages. Infect Immun 72: 1204-1209.
  8. Mancuso G, Midiri A, Biondo C, Beninati C, Gambuzza M, et al. (2005) Bacteroides fragilis-derived lipopolysaccharide produces cell activation and lethal toxicity via toll-like receptor 4. Infect Immun 73: 5620-5627.
  9. Mancuso G, Gambuzza M, Midiri A, Biondo C, Papasergi S, et al. (2009) Bacterial recognition by TLR7 in the lysosomes of conventional dendritic cells. Nat Immunol 10: 587-594.
  10. Ousman SS, Kubes P (2012) Immune surveillance in the central nervous system. Nat Neurosci 15: 1096-1101.
  11. van Noort JM, Bsibsi M (2009) Toll-like receptors in the CNS: implications for neurodegeneration and repair. Prog Brain Res 175: 139-148.
  12. Xu J, Wagoner G, Douglas JC, Drew PD (2013) ß-Lapachone ameliorization of experimental autoimmune encephalomyelitis. J Neuroimmunol 254: 46-54.
  13. Gambuzza M, Licata N, Palella E, Celi D, Foti Cuzzola V, et al. (2011) Targeting Toll-like receptors: emerging therapeutics for multiple sclerosis management. J Neuroimmunol 239: 1-12.
  14. Johnson TP, Tyagi R, Patel K, Schiess N, Calabresi PA, et al. (2013) Impaired toll-like receptor 8 signaling in multiple sclerosis. J Neuroinflammation 10: 74.
  15. Tao Y, Zhang X, Chopra M, Kim MJ, Buch KR, et al. (2014) The role of endogenous IFN-ß in the regulation of Th17 responses in patients with relapsing-remitting multiple sclerosis. J Immunol 192: 5610-5617.
  16. Touil T, Fitzgerald D, Zhang GX, Rostami A, Gran B (2006) Cutting Edge: TLR3 stimulation suppresses experimental autoimmune encephalomyelitis by inducing endogenous IFN-beta. J Immunol 177: 7505-7509.
  17. Bsibsi M, Persoon-Deen C, Verwer RW, Meeuwsen S, Ravid R, et al. (2006) Toll-like receptor 3 on adult human astrocytes triggers production of neuroprotective mediators. Glia 53: 688-695.
  18. Bsibsi M, Bajramovic JJ, Vogt MH, van Duijvenvoorden E, Baghat A, et al. (2010) The microtubule regulator stathmin is an endogenous protein agonist for TLR3. J Immunol 184: 6929-6937.
  19. Jasani B, Navabi H, Adams M (2009) Ampligen: a potential toll-like 3 receptor adjuvant for immunotherapy of cancer. Vaccine 27: 3401-3404.
  20. Nicodemus CF, Wang L, Lucas J, Varghese B, Berek JS (2010) Toll-like receptor-3 as a target to enhance bioactivity of cancer immunotherapy. Am J Obstet Gynecol 202: 608.
  21. Strayer DR, Carter WA, Stouch BC, Stevens SR, Bateman L, et al. (2012) A double-blind placebo-controlled randomized clinical trial of the TLR3 agonist rintatolimod in severe cases of chronic fatigue syndrome. PLos One 7: e31334.
  22. Navabi H, Jasani B, Reece A, Clayton A, Tabi Z, et al. (2009) A clinical grade poly I:C-analogue (Ampligen) promotes optimal DC maturation and Th1-type T cell responses of healthy donors and cancer patients in vitro. Vaccine 27: 107-115.
  23. Eriksson M, Meadows SK, Basu S, Mselle TF, Wira CR, et al. (2006) TLRs mediate IFN-gamma production by human uterine NK cells in endometrium. J Immunol 176: 6219-6224.
  24. Kaisho T, Akira S (2006) Toll-like receptor function and signaling. J Allergy Clin Immunol 117: 979-987.
  25. Gerondakis S, Grumont RJ, Banerjee A (2007) Regulating B-cell activation and survival in response to TLR signals. Immunol Cell Biol 85: 471-475.
  26. Gibson FC 3rd, Ukai T, Genco CA (2008) Engagement of specific innate immune signaling pathways during Porphyromonas gingivalis induced chronic inflammation and atherosclerosis. Front Biosci 13: 2041-2059.
  27. Iwamura C, Nakayama T (2008) Toll-like receptors in the respiratory system: their roles in inflammation. Curr Allergy Asthma Rep 8: 7-13.
  28. Sabroe I, Whyte MK (2007) Toll-like receptor (TLR)-based networks regulate neutrophilic inflammation in respiratory disease. Biochem Soc Trans 35: 1492-1495.
  29. Sutmuller R, Garritsen A, Adema GJ (2007) Regulatory T cells and toll-like receptors: regulating the regulators. Ann Rheum Dis 66 Suppl 3: iii91-95.
  30. Yoshimoto T, Nakanishi K (2006) Roles of IL-18 in basophils and mast cells. Allergol Int 55: 105-113.
  31. Carty M, Bowie AG (2011) Evaluating the role of Toll-like receptors in diseases of the central nervous system. Biochem Pharmacol 81: 825-837.
  32. Olson JK, Miller SD (2004) Microglia initiate central nervous system innate and adaptive immune responses through multiple TLRs. J Immunol 173: 3916-3924.
  33. Bowman CC, Rasley A, Tranguch SL, Marriott I (2003) Cultured astrocytes express toll-like receptors for bacterial products. Glia 43: 281-291.
  34. Nayak D, Zinselmeyer BH, Corps KN, McGavern DB (2012) In vivo dynamics of innate immune sentinels in the CNS. Intravital 1: 95-106.
  35. Aravalli RN, Peterson PK, Lokensgard JR (2007) Toll-like receptors in defense and damage of the central nervous system. J Neuroimmune Pharmacol 2: 297-312.
  36. Tang SC, Arumugam TV, Xu X, Cheng A, Mughal MR, et al. (2007) Pivotal role for neuronal Toll-like receptors in ischemic brain injury and functional deficits. Proc Natl Acad Sci U S A 104: 13798-13803.
  37. Rock FL, Hardiman G, Timans JC, Kastelein RA, Bazan JF (1998) A family of human receptors structurally related to Drosophila Toll. Proc Natl Acad Sci U S A 95: 588-593.
  38. Chen K, Huang J, Gong W, Iribarren P, Dunlop NM, et al. (2007) Toll-like receptors in inflammation, infection and cancer. Int Immunopharmacol 7: 1271-1285.
  39. Kawai T, Akira S (2010) The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol 11: 373-384.
  40. Rasmussen SB, Reinert LS, Paludan SR (2009) Innate recognition of intracellular pathogens: detection and activation of the first line of defense. APMIS 117: 323-337.
  41. Romagne F (2007) Current and future drugs targeting one class of innate immunity receptors: the Toll-like receptors. Drug Discov Today 12: 80-87.
  42. Wang RF, Miyahara Y, Wang HY (2008) Toll-like receptors and immune regulation: implications for cancer therapy. Oncogene 27: 181-189.
  43. Iwasaki A, Medzhitov R (2004) Toll-like receptor control of the adaptive immune responses. Nat Immunol 5: 987-995.
  44. Carpentier PA, Duncan DS, Miller SD (2008) Glial toll-like receptor signaling in central nervous system infection and autoimmunity. Brain Behav Immun 22: 140-147.
  45. Lehnardt S (2010) Innate immunity and neuroinflammation in the CNS: the role of microglia in Toll-like receptor-mediated neuronal injury. Glia 58: 253-263.
  46. Jack CS, Arbour N, Manusow J, Montgrain V, Blain M, et al. (2005) TLR signaling tailors innate immune responses in human microglia and astrocytes. J Immunol 175: 4320-4330.
  47. Gambuzza ME, Sofo V, Salmeri FM, Soraci L, Marino S, et al. (2014) Toll-like receptors in Alzheimer's disease: a therapeutic perspective. CNS Neurol Disord Drug Targets 13: 1542-1558.
  48. Bsibsi M, Ravid R, Gveric D, van Noort JM (2002) Broad expression of Toll-like receptors in the human central nervous system. J Neuropathol Exp Neurol 61: 1013-1021.
  49. Farina C, Krumbholz M, Glese T, Hartmann G, Aloisi F, et al. (2005) Preferential expression and function of TLR3 in human astrocytes. J Immunol. 159: 12-19.
  50. Lafon M, Megret F, Lafage M, Prehaud C (2006) The innate immune facet of brain: human neurons express TLR-3 and sense viral dsRNA. J Mol Neurosci 29: 185-194.
  51. Nagyoszi P, Wilhelm I, Farkas AE, Fazakas C, Dung NT, et al. (2010) Expression and regulation of toll-like receptors in cerebral endothelial cells. Neurochem Int 57: 556-564.
  52. Oshiumi H, Matsumoto M, Funami K, Akazawa T, Seya T (2003) TICAM-1, an adaptor molecule that participates in Toll-like receptor 3-mediated interferon-beta induction. Nat Immunol 4: 161-167.
  53. Taniguchi T, Ogasawara K, Takaoka A, Tanaka N (2001) IRF family of transcription factors as regulators of host defense. Annu Rev Immunol 19: 623-655.
  54. Yamamoto M, Sato S, Hemmi H, Hoshino K, Kaisho T, et al. (2003) Role of adaptor TRIF in the MyD88-independent toll-like receptor signaling pathway. Science 301: 640-643.
  55. Black KE, Collins SL, Hagan RS, Hamblin MJ, Chan-Li Y, et al. (2013) Hyaluronan fragments induce IFNß via a novel TLR4-TRIF-TBK1-IRF3-dependent pathway. J Inflamm (Lond) 10: 23.
  56. Liu L, Botos I, Wang Y, Leonard JN, Shiloach J, et al. (2008) Structural basis of toll-like receptor 3 signaling with double-stranded RNA. Science 320: 379-381.
  57. Nicodemus CF, Berek JS (2010) TLR3 agonists as immunotherapeutic agents. Immunotherapy 2: 137-140.
  58. Leonard JN, Ghirlando R, Askins J, Bell JK, Margulies DH, et al. (2008) The TLR3 signaling complex forms by cooperative receptor dimerization. Proc Natl Acad Sci U S A 105: 258-263.
  59. Botos I, Liu L, Wang Y, Segal DM, Davies DR (2009) The toll-like receptor 3:dsRNA signaling complex. Biochim Biophys Acta 1789: 667-674.
  60. Jin B, Sun T, Yu XH, Yang YX, Yeo AE (2012) The effects of TLR activation on T-cell development and differentiation. Clin Dev Immunol 2012: 836485.
  61. Manavalan B, Basith S, Choi S (2011) Similar Structures but Different Roles - An Updated Perspective on TLR Structures. Front Physiol 2: 41.
  62. Honda K, Takaoka A, Taniguchi T (2006) Type I interferon [corrected] gene induction by the interferon regulatory factor family of transcription factors. Immunity 25: 349-360.
  63. Enesa K, Ordureau A, Smith H, Barford D, Cheung PC, et al. (2012) Pellino1 is required for interferon production by viral double-stranded RNA. J Biol Chem 287: 34825-34835.
  64. Marié I, Durbin JE, Levy DE (1998) Differential viral induction of distinct interferon-alpha genes by positive feedback through interferon regulatory factor-7. EMBO J 17: 6660-6669.
  65. Sato M, Hata N, Asagiri M, Nakaya T, Taniguchi T, et al. (1998) Positive feedback regulation of type I IFN genes by the IFN-inducible transcription factor IRF-7. FEBS Lett 441: 106-110.
  66. Johnsen IB, Nguyen TT, Bergstrøm B, Lien E, Anthonsen MW (2012) Toll-like receptor 3-elicited MAPK activation induces stabilization of interferon-ß mRNA. Cytokine 57: 337-346.
  67. Constantinescu CS, Farooqi N, O'Brien K, Gran B (2011) Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 164: 1079-1106.
  68. Fitzgerald DC, Ciric B, Touil T, Harle H, Grammatikopolou J, et al. (2007) Suppressive effect of IL-27 on encephalitogenic Th17 cells and the effector phase of experimental autoimmune encephalomyelitis. J Immunol 179: 3268-3275.
  69. Aranami T, Yamamura T (2008) Th17 Cells and autoimmune encephalomyelitis (EAE/MS). Allergol Int 57: 115-120.
  70. Guo B, Chang EY, Cheng G (2008) The type I IFN induction pathway constrains Th17-mediated autoimmune inflammation in mice. J Clin Invest 118: 1680-1690.
  71. Carter WA (2014) Double-stranded ribonucleic acids with rugged physico-chemical sructure and highly specific biologic activity. Pat. Applic. N. US2014/0235841 A1.
  72. Geiss G, Jin G, Guo J, Bumgarner R, Katze MG, et al. (2001) A comprehensive view of regulation of gene expression by double-stranded RNA-mediated cell signaling. J Biol Chem 276: 30178-30182.
  73. Suhadolnik RJ, Reichenbach NL, Hitzges P, Sobol RW, Peterson DL, et al. (1994) Upregulation of the 2-5 synthetase/RNase L antiviral pathway in a controlled clinical trial with poly(I)-poly(C12U) in chronic fatigue syndrome. In Vivo 8:599-604.
  74. Mindy K (1999) A history of Ampligen: the AIDS drug no one can have. Philadelphia Magazine; 1994. p. 94-105.
  75. Schröder M, Bowie AG (2005) TLR3 in antiviral immunity: key player or bystander? Trends Immunol 26: 462-468.
  76. Chaitman BR, Pepine CJ, Parker JO, Skopal J, Chumakova G, et al. (2004) Effects of ranolazine with atenolol, amlodipine, or diltiazem on exercise tolerance and angina frequency in patients with severe chronic angina: a randomized controlled trial. JAMA 291: 309-316.
  77. Steelman AJ, Li J (2011) Poly(I:C) promotes TNFa TNFR1-dependent oligodendrocyte death in mixed glial cultures. J Neuroinflammation 8: 89.
  78. Hemispherx Biopharma (2014) New publication enlarges the understanding of Ampligen® safety profile across diverse animal species and focuses on the unique TLR2 receptor/Ampligen® interaction.
  79. Greene JJ, Ts'o POP, Strayer DR, Carter WA (1984) Therapeutic applications of double-stranded RNAs. Interferons and Their Applications 71: 535-555.
  80. De Clercq E (2006) Interferon and its inducers--a never-ending story: "old" and "new" data in a new perspective. J Infect Dis 194 Suppl 1: S19-26.
  81. Sumita M, Desaulniers JP, Chang YC, Chui HM, Clos L 2nd, et al. (2005) Effects of nucleotide substitution and modification on the stability and structure of helix 69 from 28S rRNA. RNA 11: 1420-1429.
  82. Fukuda K, Straus SE, Hickie I, Sharpe MC, Dobbins JG, et al. (1994) The chronic fatigue syndrome: a comprehensive approach to its definition and study. Intern CFS Study Group. Ann Intern Med 121:953-959.
  83. Holmes GP, Kaplan JE, Gantz NM, Komaroff AL, Schonberger LB, et al. (1988) Chronic fatigue syndrome: a working case definition. Ann Intern Med 108: 387-389.
  84. Komaroff AL, Fagioli LR, Geiger AM, Doolittle TH, Lee J, et al. (1996) An examination of the working case definition of chronic fatigue syndrome. Am J Med 100: 56-64.
  85. Maes M (2013) Inflammatory and oxidative and nitrosative stress cascades as new drug targets in myalgic encephalomyelitis and chronic fatigue syndrome. Mod Trends Pharmacopsychiatri 28: 162-174.
  86. Nijs J, Frémont M (2008) Intracellular immune dysfunction in myalgic encephalomyelitis/chronic fatigue syndrome: state of the art and therapeutic implications. Expert Opin Ther Targets 12: 281-289.
  87. Chakrabarti A, Jha BK, Silverman RH (2011) New insights into the role of RNase L in innate immunity. J Interferon Cytokine Res 31: 49-57.
  88. El Bakkouri K, Englebienne P, De Meirleir K, Herst CVT (2003) Methods of treatment of chronic immune disease. World Patent ?WO03061605, 2003.
  89. Nicolson GL, Haier J (2010) Role of chronic bacterial and viral infections in neurodegenerative, neurobehavioural, psychiatric, autoimmune and fatiguing illnesses: Part 2. BJMP 3: 301.
  90. Liang SL, Quirk D, Zhou A (2006) RNase L: its biological roles and regulation. IUBMB Life 58: 508-514.
  91. Ireland DD, Stohlman SA, Hinton DR, Kapil P, Silverman RH, et al. (2009) RNase L mediated protection from virus induced demyelination. PLoS Pathog 5: e1000602.
  92. Hemispherx Biopharma Ampligen for the treatment of chronic fatigue syndrome. 2012.www.fda.gov/downloads/AdvisoryCommitees/CommitteesMeetingMaterials/Drugs/ArthritisAdvisoryCommitee/UCM334430.pdf
  93. Twisk FN (2014) The status of and future research into Myalgic Encephalomyelitis and Chronic Fatigue Syndrome: the need of accurate diagnosis, objective assessment, and acknowledging biological and clinical subgroups. Front Physiol 5: 109.
  94. Rimes KA, Chalder T (2005) Treatments for chronic fatigue syndrome. Occup Med (Lond) 55: 32-39.
  95. Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, et al. (2004) Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethal encephalitis. Nat Med 10: 1366-1373.
  96. Daffis S, Samuel MA, Suthar MS, Gale M Jr, Diamond MS (2008) Toll-like receptor 3 has a protective role against West Nile virus infection. J Virol 82: 10349-10358.
  97. Nath A, Tyler KL (2013) Novel approaches and challenges to treatment of central nervous system viral infections. Ann Neurol 74: 412-422.
  98. No Author listed (2004) Mismatched double-stranded RNA, polyI:polyC12U. Drugs R.D. 5: 297-304.
  99. Morris G, Maes M (2013) Myalgic encephalomyelitis/chronic fatigue syndrome and encephalomyelitis disseminata/multiple sclerosis show remarkable levels of similarity in phenomenology and neuroimmune characteristics. BMC Med 11:205.
  100. Karussis D (2014) The diagnosis of multiple sclerosis and the various related demyelinating syndromes: a critical review. J Autoimmun 48-49: 134-42.
  101. Wootla B, Eriguchi M, Rodriguez M (2012) Is multiple sclerosis an autoimmune disease? Autoimmune Dis 2012: 969657.
  102. Brenu EW, Tajouri L, Ashton KJ, Staines DR, Marshall-Gradisnik SM (2013) Chronic fatigue syndrome/myalgic encephalomyelitis and parallels with autoimmune disorders.Edited by Spaska Angelova Stanilova 205.
  103. Hernández-Pedro NY, Espinosa-Ramirez G, de la Cruz VP, Pineda B, Sotelo J (2013) Initial immunopathogenesis of multiple sclerosis: innate immune response. Clin Dev Immunol 2013: 413465.
  104. Gooshe M, Abdolghaffari AH, Gambuzza ME, Rezaei N (2014) The role of Toll-like receptors in multiple sclerosis and possible targeting for therapeutic purposes. Rev Neurosci 25: 713-739.
  105. Hansen BS, Hussain RZ, Lovett-Racke AE, Thomas JA, Racke MK (2006) Multiple toll-like receptor agonists act as potent adjuvants in the induction of autoimmunity. J Neuroimmunol 172: 94-103.
  106. Podda G, Nyirenda M, Crooks J, Gran B (2013) Innate immune responses in the CNS: role of toll-like receptors, mechanisms, and therapeutic opportunities in multiple sclerosis. J Neuroimmune Pharmacol 8: 791-806.
  107. Airas L, Niemelä J, Yegutkin G, Jalkanen S (2007) Mechanism of action of IFN-beta in the treatment of multiple sclerosis: a special reference to CD73 and adenosine. Ann N Y Acad Sci 1110: 641-648.
  108. McFarlin DE, Bever CT, Salazar AM, Levy HB (1985) A preliminary trial of poly(I,C)-LC in multiple sclerosis. J Biol Response Mod 4: 544-548.
  109. Bever CT Jr, Salazar AM, Neely E, Ferraraccio BE, Rose JW, et al. (1986) Preliminary trial of poly ICLC in chronic progressive multiple sclerosis. Neurology 36: 494-498.
  110. Tzima S, Victoratos P, Kranidioti K, Alexiou M, Kollias G (2009) Myeloid heme oxygenase-1 regulates innate immunity and autoimmunity by modulating IFN-beta production. J Exp Med 206: 1167-1179.
  111. Minagar A (2013) Current and future therapies for multiple sclerosis. Scientifica (Cairo) 2013: 249101.
  112. Creeke PI, Farrell RA (2013) Clinical testing for neutralizing antibodies to interferon-ß in multiple sclerosis. Ther Adv Neurol Disord 6: 3-17.
  113. Castro-Borrero W, Graves D, Frohman TC, Flores AB, Hardeman P, et al. (2012) Current and emerging therapies in multiple sclerosis: a systematic review. Ther Adv Neurol Disord 5: 205-220.
  114. Mitchell WM, Nicodemus CF, Carter WA, Horvath JC, Strayer DR (2014) Discordant biological and toxicological species responses to TLR3 activation. Am J Pathol 184: 1062-1072.
  115. Lundberg AM, Drexler SK, Monaco C, Williams LM, Sacre SM, et al. (2007) Key differences in TLR3/poly I:C signaling and cytokine induction by human primary cells: a phenomenon absent from murine cell systems. Blood 110: 3245-3252.
Citation: Gambuzza ME, Sofo V, Salmeri FM, Soraci L, Marino S, et al. (2015) A Toll-Like Receptor 3-Agonist as Promising Candidate in Multiple Sclerosis Treatment. J Clin Cell Immunol 6:339.

Copyright: © 2015 Gambuzza ME, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top