SlideShare a Scribd company logo
IFN-b and multiple sclerosis: From etiology to therapy and back
V. Annibali a,1
, R. Mechelli a,1
, S. Romano a
, M.C. Buscarinu a
, A. Fornasiero a
,
R. Umeton a
, V.A.G. Ricigliano a,b
, F. Orzi c
, E.M. Coccia d
, M. Salvetti a,
*, G. Ristori a
a
Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza
University of Rome, Italy
b
Neuroimmunology Unit, Fondazione Santa Lucia-I.R.C.C.S., Rome, Italy
c
Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy
d
Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanita`, Rome, Italy
1. Introduction
Multiple sclerosis (MS) is an inflammatory demyelinating
disease of the central nervous system (CNS) characterized, in its
most common clinical presentation, by an unpredictable occur-
rence of relapse and remission phases [1–3]. The disease generally
affects young adults [4], with a preference for female gender, as
observed in many other immune-mediated conditions. Being a
multifactorial disorder, its etiology involves both genetic and
environmental risk factors. So far, genome-wide association
studies (GWAS) have shown that genetic predisposition to MS is
determined by more than 100 disease-associated susceptibility
polymorphisms, located in coding and non-coding DNA [5]. Path-
way analyses on MS-related genes demonstrated a relation with
cellular networks specifically involved in immune cell functioning,
antiviral response and interferon (IFN) signaling [6].
Major environmental risk factors for MS include Epstein–Barr
virus (EBV) infection, the reactivation of human endogenous
retroviruses (HERV) in specific conditions, vitamin D deficit and
cigarette smoking, as supported by epidemiological surveys,
serological evidences and other experimental laboratory based
studies [7–13]. Nonetheless, a comprehensive overview of the
events leading to MS development is still lacking.
There is no cure for MS and treatments focus on treating
relapses, slowing the progression of the disease and managing
symptoms. Several immunomodulatory and immunosuppressive
therapeutic agents are currently available for relapsing-remitting
forms (RR) of MS, being interferon beta (IFN-b) the first
therapeutic intervention able to interfere with the course of the
disease and still the most used first-line treatment in RR MS.
Cytokine & Growth Factor Reviews 26 (2015) 221–228
A R T I C L E I N F O
Article history:
Available online 31 October 2014
Keywords:
Multiple sclerosis
Interferon beta
Epstein–Barr virus
Human endogenous retroviruses
Genome-wide Association Studies
A B S T R A C T
Several immunomodulatory treatments are currently available for relapsing-remitting forms of multiple
sclerosis (RRMS). Interferon beta (IFN) was the first therapeutic intervention able to modify the course of
the disease and it is still the most used first-line treatment in RRMS.
Though two decades have passed since IFN-b was introduced in the management of MS, it remains a
valid approach because of its good benefit/risk profile. This is witnessed by new efforts of pharmaceutical
industry to improve this line: a PEGylated form of subcutaneous IFN-b 1a, (Plegridy1
) with a longer half-
life, has been recently approved in RRMS.
This review will survey the various stages of the use of type I IFN in MS, with special attention to the
effect of the treatment on the supposed viral etiologic factors associated to the disease. The antiviral
activities of IFN (that initially prompted its use as immunomodulatory agent in MS), and the mounting
evidences in favor of a viral etiology in MS, allowed us to outline a re-appraisal from etiology to therapy
and back.
ß 2014 Elsevier Ltd. All rights reserved.
Abbreviations: ARR, annualized relapse rate; CIS, clinically isolated syndrome; CNS,
central nervous system; EBNA, Epstein–Barr nuclear antigen; EBV, Epstein–Barr
virus; EDSS, expanded disability status scale; ELISA, enzyme-linked immunosor-
bent assay; GWAS, genome-wide association studies; HERV, human endogenous
retroviruses; HIV, human immunodeficiency virus; IFN, Interferon; IFNAR, IFN–a
receptor; IPA, ingenuity pathway analysis; IRF, interferon regulatory factor; ISGF,
IFN-stimulated gene factor; JAK, Janus-family tyrosine kinases; LMP1, latent
membrane protein-1; MRI, magnetic resonance imaging; MS, multiple sclerosis;
MSRV, MS-associated retroviruses; MxA, myxovirus-induced protein A; NAbs,
neutralizing antibodies; 20
,50
-OAS, 20
,50
-oligoadenylate synthetase; PEG, polyeth-
ylene glycol; RAL, raltegravir; RR, relapsing–remitting forms; STAT, signal
transducers and activators of transcription; SNPs, single nucleotide polymor-
phisms; TLR, toll-like receptors; TYK, tyrosine kinase.
* Corresponding author at: Neurologia, Ospedale S. Andrea, Via di Grottarossa
1035, 00189 Rome, Italy. Tel.: +39 06 33775994; fax: +39 06 33775900.
E-mail address: marco.salvetti@uniroma1.it (M. Salvetti).
1
These authors contributed equally to this work.
Contents lists available at ScienceDirect
Cytokine & Growth Factor Reviews
journal homepage: www.elsevier.com/locate/cytogfr
http://dx.doi.org/10.1016/j.cytogfr.2014.10.010
1359-6101/ß 2014 Elsevier Ltd. All rights reserved.
2. From etiology. . .
The hypothesis of a viral etiology in MS led to several
investigations on a large number of microbes, that, after an initial
enthusiastic attention, failed to be demonstrably associated to MS.
At the moment two agents seem consistently linked to diseases
development: EBV and HERV.
Converging epidemiological, clinical and laboratory studies
support an etiologic role for EBV in MS [14]. EBV is a g-herpesvirus
that infect quite all the adult population and that persists in
infected B cells in a latent or lytic phase [15]. Humans are the
exclusive natural host for EBV which may explain why MS is
unique to humans [16]. Prospective studies have shown that
elevation in serum antibody titers to EBV precedes the occurrence
of MS [17,18]. Epstein–Barr viral load in the peripheral blood of
healthy adults may predict the risk of MS [19], while in children
who develop MS immunoreactivity to EBV, but not to other viruses,
is higher than in controls [20,21]. A history of late EBV infection
and of infectious mononucleosis (IM is often the clinical
manifestation of a late primary EBV infection) is strongly
associated to MS [22]. EBV may be a target of oligoclonal
cerebrospinal fluid IgG [23], CD8+ T cells [24–26] and CD4
response [27], and a vast literature on the cross-reactivity between
EBV and myelin epitopes was produced over the past two decades
[28–30].
Recent works provided further evidences aimed at clarifying
how EBV contributes to disease development. An inadequate
control of EBV at primary infection or at a later stage can lead to
low grade, persistently active EBV infection in CNS infiltrating B
cells [26,31–33]. A ‘‘candidate-interactome’’ aggregate analysis of
genome-wide association data in multiple sclerosis demonstrated
a significant enrichment of potential interactions between the
virus and MS-related genomic regions [34]. The EBV infection of
the MS brain as cause of CNS damage that remains controversial
[35–37], is supported by several recent studies demonstrating a
selective enrichment of EBV-specific CD8+ T cells in the
cerebrospinal fluid of MS patients [38,39] and the presence of
EBV DNA in brain (Mechelli et al., manuscript submitted).
Besides to role of herpesviruses many studies support a
potential contribution for HERV to MS development. Retroviruses
are RNA viruses that may cause a spectrum of diseases of the
nervous system. Their genome contains three genetic domains: env
is responsible for the surface glycoproteins and trans-membrane;
gag encodes the proteins necessary for viral assembly, including
matrix proteins and core shell; pol encodes the enzymes needed for
viral replication, such as reverse transcriptase, protease, ribonu-
clease and integrase [40].
Specific sequences within retroviral genes can lead to the
development of neurovirulence, in particular, the proteins env-
associated, which mediates the binding of the virus to the cell
membrane surface. Neurovirulent retroviruses are able to activate
the host immune response that, through pro-inflammatory
molecules and neurotoxic molecules, ultimately leads to neuronal
death [40].
In 1997, Perron described the isolation and identification of new
retrovirus particles from cell cultures of leptomeninges, choroid
plexus and peripheral B lymphocytes in MS patients. This study
provided molecular evidence that the production of extracellular
virions containing MS-associated retroviruses (MSRV) pol se-
quence was associated with MS. This virus, previously called LM7,
was a new retrovirus which was present in the cerebrospinal fluid
of patients with MS [41]. The same group showed the production of
a specific envelope protein with gliotoxic and pro-inflammatory
actions that may be crucial in MS pathogenesis [42]. Further
studies have tried to explain and confirm the association between
MS and the expression of MSRV envelope (Env), providing
evidences that the retrovirus appears to be related to MS clinical
progression [43,44]. Recently, env antigen was detected in the
serum of 73% of MS patients with similar prevalence in all clinical
forms, and not in subjects affected by other inflammatory diseases.
The different forms of the disease (primary-progressive, RR and
clinically isolated syndrome-CIS) show different ELISA (enzyme-
linked immunosorbent assay) and/or PCR profiles indicative of an
increase with the evolution of the disease [45].
3. To therapy. . .
Interferons were discovered by Isaacs and Lindenmann [46];
they use this term to describe a soluble substance with biological
activity able to interfere with viral replication. Due to their
antiviral activities and considering the plausible viral etiology of
MS, IFNs, regardless of their type, were proposed as immunomod-
ulatory therapeutic agents in MS patients.
The first trials using IFN-g (a type II IFN) were conducted in the
late 1980s and were interrupted because the treated patients
showed an increase of severity and frequency of relapses
[47,48]. These negative results led to study another type of IFNs
(type I), IFN-a and IFN-b, that may act as inhibitors of IFN-g.
Different preparations of IFN-a resulted in reduction of clinical
relapses and activity at magnetic resonance imaging (MRI) in MS
patients, but unacceptable side effects precluded its use in clinical
practice [49–53]. IFN-b was similarly effective in decreasing
disease activity and showed an acceptable risk profile, thus
becoming the first disease-modifying therapy for MS (Table 1).
Subcutaneous IFN-b 1b (Betaseron1
) was the first immuno-
modulatory therapy to receive approval for the treatment of RRMS
in 1993. It is produced by recombinant DNA technology in the
bacterial cell (Escherichia coli) and currently is the only IFN-b
licensed for RR and secondary progressive (SP) MS [54]. In the
registration trial 372 patients were randomized to receive placebo
or IFN-b 1b (50 or 250 mcg subcutaneously every other day) for
2 years. The annualized relapse rate (ARR) was significantly lower
in the IFN-b1b treated groups compared to the placebo group with
a dosage effect. Moreover a significative reduction of activity at
MRI activity was showed. No difference in disease progression
between treatment and placebo groups was demonstrated [55,56].
IFN-b1a IM (Avonex1
) is produced in Chinese hamster ovary
cells and was approved for treatment in RRMS in 1996. In the
Table 1
INF-b formulations approved for RRMS. For detailed description, see text.
Rebif 22/44 Avonex Betaferon/Betaseron/Extavia
IFN subtype Beta 1a Beta 1a Beta 1b
Production CHO CHO E. coli
Aminoacid 166 aa 166 aa 165 aa
Glycosylation 1 N-linked complex 1 N-linked complex None
Administration SC, 3 times/week IM, 1 time/week SC, every other day
Weekly dose 66/132 mcg 30 mcg 875 mcg
CHO = Chinese hamster ovary cells; SC = subcutaneous; IM = intramuscular.
V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228222
pivotal study 301 patients with expanded disability status scale
(EDSS) score of 1.0–3.5 and at least two relapses in the preceding
3 years were randomized to receive placebo or IFN-b 1a (30 mg
intramuscularly once weekly) for 2 years [57,58]. The IFN-b 1a
group showed a significant decrease in the disease activity
compared with placebo.
IFN-b 1a SC (Rebif1
) is also produced in mammalian Chinese
hamster ovary cells using DNA technology. It was approved for
treatment of RRMS in 1998 in Europe and Canada and in 2002 in
the USA. In the PRISM study [59] 560 patients with an EDSS score
between 1.0 and 5.0 and at least two relapses in the preceding
2 years were randomized to receive placebo or IFN-b 1a (22 or
44 mg subcutaneously three times weekly). After 2 years of
treatment, IFN-b 1a showed significant results compared with
placebo in relapse rate and MRI activity with a statistically
significant dose-effect.
While the pivotal clinical trials consistently demonstrated that
both forms of IFN-b reduce ARR by about 1/3 and new brain MRI
lesions over periods of 1–3 years in RRMS, four randomized,
placebo-controlled trials demonstrated poor or no effects on
established progressive MS [54,60–63]. IFN-b has a significant
effect in the earlier inflammatory stages of the disease: three large
clinical trials (CHAMPS, ETOMS and BENEFIT) in CIS patients,
demonstrated an effect on clinical and MRI measures of disease
activity delaying the development to clinically definite (CD) MS
[64–66]. Although these studies had limited comparability because
of different patient populations recruited (BENEFIT and ETOMS
enrolled patients with multifocal manifestations at onset, while
CHAMPS enrolled patients with monofocal forms), the risk of
progression to CDMS was comparably reduced by 40–50%.
Considering the different formulations of IFN-b, head-to-head
trials were conducted to compare the different licensed IFNs
(EVIDENCE and INCOMIN). These studies demonstrated that
increasing the dose of IFN-b (more frequent dosing schedule or
higher dose) gave greater benefit than lower doses, supporting a
dose–response relationship [67,68].
The IFNs-b are generally well tolerated, being the most
frequent side effects injection-site reaction, and a flu-like
syndrome that tends to wanes over time in most patients.
Lymphopenia, hepatic failure, hepatitis, and elevated liver
enzymes have also been reported especially during IFN-b-1b
treatment [69]. Though IFN-b therapy represents a significant
advance in the management of MS, the treatment response is not
uniform and clinical experience shows that about 40% of the MS
patients do not or only poorly respond to IFN-b treatment (‘‘non-
responders’’) [70]. So far there are no established biological
markers able to predict the response. The development of
neutralizing antibodies (NAbs), which at high titers may block
the biological response of the drug with a reduced efficacy, can
contribute to treatment failure [71,72]. Persistent high-titers of
NAbs depend on the formulation and dosing regimen, and occur
more commonly with subcutaneous preparations [73].
IFN was tried in MS as a ‘general’ antiviral approach and gave
positive results as disease modifying therapy. This was not the case
with more ‘specific’ antiviral treatment such as those active on
herpes viruses. Over the last twenty years several clinical trials
have been carried out especially with acyclovir and valacyclovir
[74–76]. Overall, these studies did not obtain significant results in
favor of the drug compared to placebo, though positive trends were
noted. An analysis that also took into account the pharmacokinetic
of acyclovir and valacyclovir suggested an inhibitory effect on
some viruses (Herpes viruses 1, 2, 6 and varicella zoster virus) but
not on others that seem to have a greater correlation with MS (EBV,
Herpes virus 6, and MSRV), thus explaining, at least in part, the
failure of this approach [77]. Further studies with new antiviral
drugs having improved pharmacological characteristics and
antiretroviral activity may result in better outcome and are
currently actively investigated (see Raltegravir in next section).
4. And back. . .
4.1. MS genome-wide association studies and IFN-b pathway
The exact mode of action of IFN-b in MS is likely to be complex
and is not yet fully understood. This topic will be not treated in the
present review, being object of other contributions in this issue. At
the molecular level IFN-b is recognized by the IFN-a receptor
(IFNAR), which is found on many different cell types. The receptor
is a heterodimer formed by IFNAR1 and IFNAR2, which assemble
into a functional receptor complex and initiates the signal
transduction pathway that involves the phosphorylation of several
intracellular mediators. Upon assembly of the IFN receptor
complex, the intracellular domains of IFNAR1 and IFNAR2 active
Janus kinases 1 (JAK1) and tyrosine kinase 2 (TYK2). The JAK1/TYK2
along with IFNAR, phosphorylate signal transducers and activators
of transcription (STAT) 1, and 2, which dimerize and form a
complex with interferon regulatory factor 9 (IRF9). The STA-
T1:2:IRF9 complex is a transcription factor (IFN-stimulated gene
factor, ISGF3), which translocates to the nucleus and binds to the
IFN-stimulated response element (ISRE) of multiple genes
[78]. Different kinds of genes are targeted by ISGF3 complex,
including early genes such as IFN regulatory factor-1 (IRF-1), the
primary positive regulator of IFN production, and IRF-2, an
inhibitor of IFN production. The later genes include the IFN-b
itself and antiviral proteins such as 20
,50
-oligoadenylate synthetase
(20
,50
-OAS) and myxovirus-induced protein A (MxA), which are
specifically induced by type I IFNs. MxA is the established marker
of IFN-b biological activity in IFN-b-treated MS patients [79].
Some genes related to the IFN-b signaling pathway showed
single nucleotide polymorphisms (SNPs) that resulted to be MS-
associated in GWAS [5]. To review this, we highlighted the network
that leads to the connectivity between the IFN-b signaling
pathway and genes exceeding the genome-wide significance
threshold in GWAS published from 2007 in MS (http://www.ge-
nome.gov/gwastudies/). To perform this analysis it was used
Quigen Ingenuity Pathway Analysis (IPA), which was set to run a
‘‘core analysis’’ correlating all MS-associated genes (retrieved on 8/
26/2014) with the known interactors of IFN-b (314 molecules
present in IPA version 21249400); the analysis included only
experimental evidence observed in human samples. Among all
known interactors of IFN-b, 18 genes were also MS-associated
(Table 2 and Fig. 1, that shows most of these connections and how
they relate to IFN-b). The list includes several genes that control
the immune responses (including major histocompatibility
complex alleles, cytokines and co-stimulator molecules), as well
as direct interactors with IFN signaling such as IRF8, NFKB1 and
TYK2 [80,81]. This IPA analysis showed a significant (p-
value < 1.18 Â 10À7
) relationship between the MS-associated
genes and the IFN-b signaling and confirmed previous results
obtained by our group with another approach [6]. Overall, these
data suggest that single unfavorable SNPs (or a combination of
them) affecting components of IFN-b signaling may determine
some deregulation in MS. Further investigation are needed to
clarify the role of these components in MS pathogenesis and
possible corrective effects of exogenous IFN-b on deregulated
pathways (see next section).
4.2. EBV and IFN-b
EBV is a kind of ‘‘one man band’’ in its ability to control the
antiviral immune response of infected cells both in lytic and latent
phase. IFN pathway is no exception, being sabotaged by multiple
V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228 223
mechanisms of immune evasion. In an in vitro setting was
demonstrated that the expression of BRLF1 and BZLF1 (two
immediate-early transcription factors that controls the initiation
of viral lytic gene expression and lytic reactivation from latency)
reduce the IFN-b production down modulating the expression of
IRF3 and IRF7 [82,83]. During the latent phase the up-regulation of
latent membrane protein-1 (LMP1), possibly due to a toll-like
receptors 7 (TLR7) aberrant activation, may blocks TYK2 and the
consequent STATs phosphorylation, inhibiting the expression of
IFN-b stimulated genes [84,85]. Moreover EBV infection of primary
B cells may reduce the cellular antiviral activity inhibiting the TLR9
activation through the expression of LMP1 [86], that in turn may be
Table 2
The MS-associated genes of the IFN-b signaling pathway. For detailed description, see text.
Gene symbol Description Location Type
CD40 CD40 molecule, TNF receptor super family member 5 Plasma membrane Transmembranereceptor
CD86 CD86 molecule Plasma membrane Transmembranereceptor
HLA-B Major histocompatibility complex, class I, B Plasma membrane Transmembranereceptor
HLA-DQA1 Major histocompatibility complex, class II, DQ alpha 1 Plasma membrane Transmembranereceptor
HLA-DQB1 Major histocompatibility complex, class II, DQ beta 1 Plasma membrane Other
HLA-DRA Major histocompatibility complex, class II, DR alpha Plasma membrane Transmembranereceptor
HLA-DRB1 Major histocompatibility complex, class II, DR beta 1 Plasma membrane Transmembranereceptor
IL12A Interleukin 12A Extracellular space Cytokine
IL12B Interleukin 12B Extracellular space Cytokine
IRF8 Interferon regulatory factor 8 Nucleus Transmembranereceptor
MAPK1 Mitogen-activated protein kinase 1 Cytoplasm Kinase
MERTK MER proto-oncogene, tyrosine kinase Plasma membrane Kinase
MMP10 Matrix metallopeptidase 10 (stromelysin 2) Extracellular space Peptidase
MYC V-myc avian myelocytomatosis viral oncogene homolog Nucleus Transmembranereceptor
NFKB1 Nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 Nucleus Transmembranereceptor
PRKRA Protein kinase, interferon-inducible double stranded RNA dependent activator Cytoplasm Other
STAT3 Signal transducer and activator of transcription 3 Nucleus Transmembranereceptor
TYK2 Tyrosinekinase 2 Plasma membrane Kinase
Fig. 1. Representation of the interactions between proteins coded by MS-associated and IFN beta-related genes (see Table 2 for the list of the interaction points strictly shared
by the MS-associated gene list and the IFN interactors).
V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228224
up-regulated by Epstein–Barr nuclear antigen 2 (EBNA2). This
protein is able to control gene expression of viral and cellular
genes, mainly during the first phase of the infection [87]. Some
evidences suggest its potential implication in MS pathology:
EBNA2 expressing cells have been observed in affected brains [31]
and specific EBNA2 genotypes associate with disease status [88].
It seems plausible that IFN-b therapy may compensate for
some of the EBV-induced dysfunctions in the antiviral immune
responses: CD8+ T cells specific for lytic-phase antigens are
detected with high frequency in the peripheral blood of patients
with active disease and are reduced by IFN-b treatment [26], as
well as CD4+ T cell response to EBNA1 peptides pool [89]; in
dendritic cells obtained from MS patients under IFN-b treatment a
reduced TLR9 activation (that promote pro-inflammatory
responses) was observed [90]; a recent work demonstrated an
impaired activation of TLR7 in MS subjects, that decrease the
ability of B cells to mature in plasma cells and that is restored by
IFN-b treatment [91].
4.3. MSRV and IFN-b
IFN-b appears to be capable of interfering with MSRV biology.
An in vitro study showed that IFN-b inhibits the release of MSRV
from peripheral blood mononuclear cells derived from MS patients
[92]. These data were confirmed through a longitudinal evaluation
of patients with MS, during one year of therapy with IFN-b: the
MSRV load in the blood was directly related to the duration of MS
and underwent a considerable reduction to below the limits of
detection within 3 months of IFN therapy; this work suggested to
consider the evaluation of MSRV in plasma as a prognostic marker
to monitor the progression of the disease and the outcome of
therapy [43].
At variance with several trials conducted with anti-herpes
drugs, and notwithstanding evidences of retroviral contribution to
disease pathogenesis, no major attempt has been performed with
anti-retroviral therapy in MS, except for a humanized monoclonal
antibody against the envelope of MSRV, that was tried in a phase I
study [93]. A pilot study, that is ongoing, may herald such an
approach, investigating raltegravir (RAL) in relapsing remitting MS
(ClinicalTrials.gov Identifier: NCT01767701).
RAL is an inhibitor of human immunodeficiency virus (HIV)
integrase, approved in 2007 for clinical use as antiretroviral agent
in HIV infected adults. Clinical studies and subsequent clinical
experience have shown durable virologic suppression, low rates of
adverse effects and long-term safety. Not interacting with the
cytochrome P450 system, RAL may be a good option for
polytherapy. As an inhibitor of retroviral integrase, RAL can be
active against the MSRV that is transactivated by several viruses,
EBV being one of these. RAL is also able to inhibit recombinase and
terminase, two key proteins for EBV [94]. Altogether, RAL seems to
be a good candidate to tackle plausible etiologic agents for MS and
might also add to the effects of IFN.
5. Conclusions
Though two decades have passed since IFN-b was introduced in
the management of MS, it remains a valid approach because of its
good benefit/risk profile. The persisting interest is witnessed by
new efforts that pharmaceutical industry has produced to improve
this line.
Recently, a PEGylated form of subcutaneous IFN-b 1a (Ple-
gridy1
) with a longer half-life (injection frequency every 2 weeks)
has been approved in RRMS. Conjugation of IFN-b 1a with a
molecule of polyethylene glycol (PEG; PEGylation) increases the
size of the product resulting in more solubility, half-life and
efficacy the drug. Compared to placebo, PEG INF has reduced ARR
by about one-third (0.397 in the placebo group versus 0.256 in the
every 2 weeks group). A slight reduction in sustained disability
progression and in several MRI activity measures has also been
demonstrated. The drug was generally well tolerated: the most
common adverse events were influenza-like illness, injection-site
reactions and headache [95]. Due to its frequency of administra-
tion (every 2 weeks), PEG IFN-b1a may have a better safety profile
than other IFNs-b formulations. Moreover, results from the
extension of the phase III study showed that the therapeutic
effects of PLEGRIDY may reach a size that was not attained by
increasing doses of non-pegylated IFN-b and may become even
more relevant over time, suggesting that prolonged treatment with
PLEGRIDY may induce therapeutic effects that go beyond the
immunomodulatory action of IFN-b.
Given the potent antiviral effects of type-1 IFN, the added value
of PLEGRIDY treatment might be related to its ability to target more
efficiently the non-heritable (i.e. viral) cause(s) of MS. This hints at
future therapeutic approaches based on type 1 interferon alone or
in association with specific antiviral drugs that might act as an
etiologic treatment for MS.
The main fields of investigation regard:
(a) etiopathogenesis of multiple sclerosis;
(b) the identification of the world’s largest twin registry in Italian
population; the registry is currently exploited for concordance
studies and for laboratory investigations (studies on twin pairs
discordant for disease); and
(c) clinical trials (especially phase II independent studies) in
patients with multiple sclerosis, Huntington disease and
cerebellar ataxia.
Conflict of interest
MS receives research support and has received fees as speaker
from Sanofi-Aventis, Biogen, Bayer Schering, and Merck Serono.
Acknowledgements
MS is supported from: Italian Multiple Sclerosis Foundation
(Fondazione Italiana Sclerosi Multipla grant number: 2011/R/31)
and Italian Ministry of Health (Ministero della Salute, grant
number: RF-2010-2321254).
References
[1] Amato MP, Ponziani G, Bartolozzi ML, Siracusa G. A prospective study on the
natural history of multiple sclerosis: clues to the conduct and interpretation of
clinical trials. J Neurol Sci 1999;168:96–106.
[2] Scalfari A, Neuhaus A, Degenhardt A, Rice GP, Muraro PA, Daumer M, et al. The
natural history of multiple sclerosis, a geographically based study: relapses
and long-term disability. Brain 2010;133:1914–29.
[3] Bordi I, Umeton R, Ricigliano VA, Annibali V, Mechelli R, Ristori G, et al. A
mechanistic, stochastic model helps understand multiple sclerosis course and
pathogenesis. Int J Genomics 2013;2013:910321.
[4] Koch-Henriksen N. The Danish multiple sclerosis registry: a 50-year follow-up.
Mult Scler 1999;5:293–6.
[5] International Multiple Sclerosis Genetics Consortium (IMSGC), Beecham AH,
Patsopoulos NA, Xifara DK, Davis MF, Kemppinen A, et al. Analysis of immune-
related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat
Genet 2013;45:1353–60.
[6] Ricigliano VA, Umeton R, Germinario L, Alma E, Briani M, Di Segni N, et al.
Contribution of genome-wide association studies to scientific research: a
pragmatic approach to evaluate their impact. PLOS ONE 2013;8:e71198.
[7] Kakalacheva K, Lu¨nemann JD. Environmental triggers of multiple sclerosis.
FEBS Lett 2011;585:3724–9.
[8] Pender MP. The essential role of Epstein–Barr virus in the pathogenesis of
multiple sclerosis. Neuroscientist 2011;17:351–67.
[9] Almohmeed YH, Avenell A, Aucott L, Vickers MA. Systematic review and meta-
analysis of the sero-epidemiological association between Epstein Barr virus
and multiple sclerosis. PLOS ONE 2013;8:e61110.
[10] Simon KC, Munger KL, Ascherio A. Vitamin D and multiple sclerosis: epidemi-
ology, immunology, and genetics. Curr Opin Neurol 2012;25:246–51.
V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228 225
[11] Handel AE, Williamson AJ, Disanto G, Dobson R, Giovannoni G, Ramagopalan
SV. Smoking and multiple sclerosis: an updated meta-analysis. PLoS ONE
2011;6:e16149.
[12] Sutkowski N, Conrad B, Thorley-Lawson DA, Huber BT. Epstein–Barr virus
transactivates the human endogenous retrovirus HERV-K18 that encodes a
superantigen. Immunity 2001;15:579–89.
[13] Antony JM, Deslauriers AM, Bhat RK, Ellestad KK, Power C. Human endogenous
retroviruses and multiple sclerosis: innocent bystanders or disease determi-
nants. Biochim Biophys Acta 2011;1812:162–76.
[14] Lu¨ nemann JD. Epstein–Barr virus in multiple sclerosis: a continuing conun-
drum. Neurology 2012;78:11–2.
[15] Cohen JI. Epstein–Barr virus infection. N Engl J Med 2000;343:481–92.
[16] Haahr S, Ho¨llsberg P. Multiple sclerosis is linked to Epstein–Barr virus infec-
tion. Rev Med Virol 2006;16:297–310.
[17] Levin LI, Munger KL, Rubertone MV, Peck CA, Lennette ET, Spiegelman D, et al.
Temporal relationship between elevation of Epstein–Barr virus antibody titers
and initial onset of neurological symptoms in multiple sclerosis. JAMA
2005;293:2496–500.
[18] DeLorenze GN, Munger KL, Lennette ET, Orentreich N, Vogelman JH, Ascherio
A. Epstein–Barr virus and multiple sclerosis: evidence of association from a
prospective study with long-term follow-up. Arch Neurol 2006;63:839–44.
[19] Wagner HJ, Munger KL, Ascherio A. Plasma viral load of Epstein–Barr virus and
risk of multiple. Eur J Neurol 2004;11:833–4.
[20] Alotaibi S, Kennedy J, Tellier R, Stephens D, Banwell B. Epstein–Barr virus in
pediatric multiple sclerosis. JAMA 2004;291:1875–9.
[21] Banwell B, Krupp L, Kennedy J, Tellier R, Tenembaum S, Ness J, et al. Clinical
features and viral serologies in children with multiple sclerosis: a multina-
tional observational study. Lancet Neurol 2007;6:773–81.
[22] Handel AE, Williamson AJ, Disanto G, Handunnetthi L, Giovannoni G, Rama-
gopalan SV. An updated meta-analysis of risk of multiple sclerosis following
infectious mononucleosis. PLoS ONE 2010;5(9).
[23] Cepok S, Zhou D, Srivastava R, Nessler S, Stei S, Bu¨ ssow K, et al. Identification of
Epstein–Barr virus proteins as putative targets of the immune response in
multiple sclerosis. J Clin Invest 2005;115:1352–60.
[24] Jilek S, Schluep M, Meylan P, Vingerhoets F, Guignard L, Monney A, et al. Strong
EBV-specific CD8+ T-cell response in patients with early multiple sclerosis.
Brain 2008;131:1712–21.
[25] Jilek S, Schluep M, Harari A, Canales M, Lysandropoulos A, Zekeridou A, et al.
HLAB7-restricted EBV specific CD8+ T cells are dysregulated in multiple
sclerosis. J Immunol 2012;188:4671–80.
[26] Angelini DF, Serafini B, Piras E, Severa M, Coccia EM, Rosicarelli B, et al.
Increased CD8+ T cell response to Epstein–Barr virus lytic antigens in the
active phase of multiple sclerosis. PLOS Pathog 2013;9:e1003220.
[27] Lu¨ nemann JD, Edwards N, Muraro PA, Hayashi S, Cohen JI, Mu¨nz C, et al.
Increased frequency and broadened specificity of latent EBV nuclear antigen-
1-specific T cells in multiple sclerosis. Brain 2006;129:1493–506.
[28] Ufret-Vincenty RL, Quigley L, Tresser N, Pak SH, Gado A, Hausmann S, et al. In
vivo survival of viral antigen-specific T cells that induce experimental auto-
immune encephalomyelitis. J Exp Med 1998;188:1725–38.
[29] Lu¨ nemann JD, Jelcic´ I, Roberts S, Lutterotti A, Tackenberg B, Martin R, et al.
EBNA1-specific T cells from patients with multiple sclerosis cross react with
myelin antigens and co-produce IFN-gamma and IL-2. J Exp Med
2008;205:1763–73.
[30] Lang HL, Jacobsen H, Ikemizu S, Andersson C, Harlos K, Madsen L, et al. A
functional and structural basis for TCR cross-reactivity in multiple sclerosis.
Nat Immunol 2002;3:940–3.
[31] Serafini B, Rosicarelli B, Franciotta D, Magliozzi R, Reynolds R, Cinque P, et al.
Dysregulated Epstein–Barr virus infection in the multiple sclerosis brain. J Exp
Med 2007;204:2899–912.
[32] Serafini B, Severa M, Columba-Cabezas S, Rosicarelli B, Veroni C, Chiappetta G,
et al. Epstein–Barr virus latent infection and BAFF expression in B cells in the
multiple sclerosis brain: implications for viral persistence and intrathecal B-
cell activation. J Neuropathol Exp Neurol 2010;69:677–93.
[33] Serafini B, Muzio L, Rosicarelli B, Aloisi F. Radioactive in situ hybridization for
Epstein–Barr virus-encoded small RNA supports presence of Epstein–Barr
virus in the multiple sclerosis brain. Brain 2013;136:e233.
[34] Mechelli R, Umeton R, Policano C, Annibali V, Coarelli G, Ricigliano VA, et al. A
candidate-interactome aggregate analysis of genome-wide association data in
multiple sclerosis. PLOS ONE 2013;8:e63300.
[35] Aloisi F, Serafini B, Magliozzi R, Howell OW, Reynolds R. Detection of Epstein–
Barr virus and B-cell follicles in the multiple sclerosis brain: what you find
depends on how and where you look. Brain 2010;133:e157.
[36] Lassmann H, Niedobitek G, Aloisi F, Middeldorp JM, NeuroproMiSe EBV Working
Group. Epstein–Barr virus in the multiple sclerosis brain: a controversial issue—
report on a focused workshop held in the Centre for Brain Research of the
Medical University of Vienna, Austria. Brain 2011;134:2772–86.
[37] Tzartos JS, Khan G, Vossenkamper A, Cruz-Sadaba M, Lonardi S, Sefia E, et al.
Association of innate immune activation with latent Epstein–Barr virus in
active MS lesions. Neurology 2012;78:15–23.
[38] Jaquie´ry E, Jilek S, Schluep M, Meylan P, Lysandropoulos A, Pantaleo G, et al.
Intrathecal immune responses to EBV in early MS. Eur J Immunol
2010;40:878–87 [Erratum in: Eur J Immunol 2011; 41:1501].
[39] Lossius A, Johansen JN, Vartdal F, Robins H, Ju¯ rate˙ Sˇaltyte˙ B, Holmøy T, et al.
High-throughput sequencing of TCR repertoires in multiple sclerosis reveals
intrathecal enrichment of EBV-reactive CD8(+) T cells. Eur J Immunol 2014,
August [Epub ahead of print].
[40] Power C. Retroviral diseases of the nervous system: pathogenic host response
or viral gene-mediated neurovirulence? Trends Neurosci 2001;24:162–9.
[41] Perron H, Garson JA, Bedin F, Bese`me FG, Paranhos-Baccala G, Komurian-
Pradel F, et al. Molecular identification of a novel retrovirus repeatedly isolated
from patients with multiple sclerosis. Proc Natl Acad Sci U S A 1997;94:
7583–8.
[42] Rieger F, Amouri R, Benjelloun N, Cifuentes-Diaz C, Dobransky T, Lyon-Caen O,
et al. Gliotoxic factor and multiple sclerosis. C R Acad Sci 1996;319:343–50.
[43] Mameli G, Serra C, Astone V, Castellazzi M, Poddighe L, Fainardi E, et al.
Inhibition of multiple-sclerosis-associated retrovirus as biomarker of inter-
feron therapy. J Neurovirol 2008;14:73–7.
[44] Antony JM, van Marle G, Opii W, Butterfield DA, Mallet F, Yong VW, et al.
Human endogenous retrovirus glycoprotein-mediated induction of redox
reactants causes oligodendrocyte death and demyelination. Nat Neurosci
2004;7:1088–95.
[45] Perron H, Germi R, Bernard C, Garcia-Montojo M, Deluen C, Farinelli L, et al.
Human endogenous retrovirus type W envelope expression in blood and brain
cells provides new insights into multiple sclerosis disease. Mult Scler 2012;18:
1721–36.
[46] Isaacs A, Lindenmann J. Virus interference: I. The interferon. Proc R Soc Lond B:
Biol Sci 1957;147:258–67.
[47] Pantich HS, Hirsch RL, Schindler J, Johnson KP. Treatment of multiple sclerosis
with gamma-interferons: exacerbations associated with activation on im-
mune system. Neurology 1987;37:1097–102.
[48] Pantich HS, Hirsch RL, Haley AS, Johnson KP. Exacerbations of multiple
sclerosis in patients treated with gamma-interferon. Lancet 1987;1:893–5.
[49] Knobler RL, Panitch HS, Braheny SL, Sipe JC, Rice GP, Huddlestone JR, et al.
Systemic alpha-interferontherapy of multiple sclerosis. Neurology 1984;34:
1273–9.
[50] Panitch HS. Systemic alpha-interferon in multiple sclerosis. Long-term patient
follow-up. Arch Neurol 1987;44:61–3.
[51] Camenga DL, Johnson KP, Alter M, Engelhardt CD, Fishman PS, Greenstein JI,
et al. Systemic recombinant alpha-2 interferon therapy in relapsing multiple
sclerosis. Arch Neurol 1986;43:1239–46.
[52] Durelli L, Bongioanni MR, Cavallo R, Ferrero B, Ferri R, Ferrio MF, et al. Chronic
systemic high-dose recombinant interferon alfa-2a reduces exacerbation rate.
MRI signs of disease activity, and lymphocyte interferon gamma production in
relapsing–remitting multiple sclerosis. Neurology 1994;44(3 Pt 1):406–13.
[53] Durelli L, Bongioanni MR, Ferrero B, Ferri R, Imperiale D, Bradac GB, et al.
Interferon alpha-2a treatment of relapsing–remitting multiple sclerosis: dis-
ease activity resumes after stopping treatment. Neurology 1996;47:123–9.
[54] European Study Group on Interferon Beta-1b in Secondary Progressive MS.
Placebo-controlled multicentre randomised trial of interferon beta-1b in
treatment of secondary progressive MS. Lancet 1998;352:1491–7.
[55] The IFNB Multiple Sclerosis Study Group. Interferon b-1b is effective in relaps-
ing–remitting multiple sclerosis: I. Clinical results of a multicenter, randomized,
double-blind, placebo-controlled trial. Neurology 1993;43:655–61.
[56] Paty DW, Li DK. Interferon beta lb is effective in relapsing–remitting multiple
sclerosis: II. MRI analysis results of a multicenter, randomized, double-blind,
placebo-controlled trial. Neurology 1993;43:662–7.
[57] Rudick RA, Goodkin DE, Jacobs LD, Cookfair DL, Herndon RM, Richert JR, et al.
Impact of interferon beta-1a on neurologic disability in relapsing multiple
sclerosis. The Multiple Sclerosis Collaborative Research Group (MSCRG). Neu-
rology 1997;49:358–63.
[58] Simon JH, Jacobs LD, Campion M, Wende K, Simonian N, Cookfair DL, et al.
Magnetic resonance studies of intramuscular interferon beta-1a for relapsing
multiple sclerosis. The Multiple Sclerosis Collaborative Research Group. Ann
Neurol 1998;43:79–87.
[59] PRISMS (Prevention of Relapses Disability by Interferon beta-1a Subcutane-
ously in Multiple Sclerosis) Study Group. Randomised double-blind placebo-
controlled study of interferon beta-1a in relapsing/remitting multiple sclero-
sis. Lancet 1998;352:1498–504.
[60] Panitch H, Miller A, Paty D, Weinshenker B, North American Study Group on
Interferon beta-1b in Secondary Progressive MS. Interferon beta-1b in sec-
ondary progressive MS: results from a 3-year controlled study. Neurology
2004;63:1788–95.
[61] Secondary Progressive Efficacy Clinical Trial of Recombinant Interferon-beta-1a
in MS (SPECTRIMS) Study Group. Randomized controlled trial of interferon-beta-
1a in secondary progressive MS: clinical results. Neurology 2001;56:1496–504.
[62] Li DK, Zhao GJ, Paty DW, University of British Columbia MS/MRI Analysis
Research Group The SPECTRIMS Study Group. Randomized controlled trial of
interferon-beta-1a in secondary progressive MS: MRI results. Neurology
2001;56:1505–13.
[63] Cohen JA, Cutter GR, Fischer JS, Goodman AD, Heidenreich FR, Kooijmans MF,
et al. Benefit of interferon beta-1a on MSFC progression in secondary progres-
sive MS. Neurology 2002;59:679–87.
[64] Jacobs LD, Beck RW, Simon JH, Kinkel RP, Brownscheidle CM, Murray TJ, et al.
Intramuscular interferon-beta-1a therapy initiated during a first demyelinat-
ing event in multiple sclerosis. N Engl J Med 2000;343:898–904.
[65] Comi G, Filippi M, Barkhof F, Durelli L, Edan G, Fernandez O, et al. Effect of early
interferon treatment on conversion to definite multiple sclerosis: a random-
ized study. Lancet 2001;357:1576–82.
[66] Kappos L, Polman CH, Freedman MS, Edan G, Hartung HP, Miller DH, et al.
Treatment with interferon beta-1b delays conversion to clinically definite and
McDonald MS in patients with clinically isolated syndromes. Neurology
2006;67:1242–9.
V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228226
[67] Panitch H, Goodin DS, Francis G, Chang P, Coyle PK, O’Connor P, et al.
Randomized, comparative study of interferon b-1a treatment regimens in
MS. The EVIDENCE Trial. Neurology 2002;59:1496–506.
[68] Durelli L, Verdun E, Barbero P, Bergui M, Versino E, Ghezzi A, et al. Every-other-
day interferon beta-1b versus once-weekly interferon beta-1a for multiple
sclerosis: results of a 2-year prospective randomized multicentre study
(INCOMIN). Lancet 2002;359:1453–60.
[69] McGraw CA, Lublin FD. Interferon beta and glatiramer acetate therapy. Neu-
rotherapeutics 2013;10:2–18.
[70] Rudick RA, Lee JC, Simon J, Ransohoff RM, Fisher E. Defining interferon
beta response status in multiple sclerosis patients. Ann Neurol 2004;56:
548–55.
[71] Ross C, Clemmesen K, Svenson M, Sørensen PS, Koch-Henriksen N, Skovgaard
GL, et al. Immunogenicity of interferon-beta in multiple sclerosis patients:
influence of preparation, dosage, dose frequency, and route of administration.
Ann Neurol 2000;48:706–12.
[72] Calabresi PA, Kieseier BC, Arnold DL,BalcerLJ, Boyko A, Pelletier J, et al. Pegylated
interferon b-1a for relapsing–remitting multiple sclerosis (ADVANCE): a ran-
domised, phase 3, double-blind study. Lancet Neurol 2014;13:657–65.
[73] Kappos L, Clanet M, Sandberg-Wollheim M, Radue EW, Hartung HP, Hohlfeld R,
et al. Neutralizing antibodies and efficacy of interferon beta-1a: a 4-year
controlled study. Neurology 2005;65:40–7.
[74] Lycke J, Svennerholm B, Hjelmquisit E, Frise´n L, Badr G, Andersson M, et al.
Acyclovir treatment of relapsing–remitting multiple sclerosis a randomized,
placebo-controlled, double-blind study. J Neurol 1996;243:214–24.
[75] Bech E, Lycke J, Gadeberg P, Hansen HJ, Malmestro¨m C, Andersen O, et al. A
randomized, double-blind, placebo-controlled MRI study of anti-herpes virus
therapy in MS. Neurology 2002;58:31–6.
[76] Friedman JE, Zabriskie JB, Plank C, Ablashi D, Whitman J, Shahan B, et al. A
randomized clinical trial of valacyclovir in multiple sclerosis. Mult Scler
2005;11:286–95.
[77] Lycke J, Andersen O, Svennerholm B, Appelgren L, Dahllof C. Acyclovir con-
centrations in serum and cerebrospinal fluid at steady state. J Antimicrob
Chemother 1989;24:947–54.
[78] Ivashkiv LB, Donlin LT. Regulation of type I interferon responses. Nat Rev
Immunol 2014;14:36–49.
[79] Killestein J, Polman CH. Determinants of interferon b efficacy in patients with
multiple sclerosis. Nat Rev Neurol 2011;7(4):221–8.
[80] Taniguchi T, Takaoka A. A weak signal for strong responses: interferon-alpha/
beta revisited. Nat Rev Mol Cell Biol 2001;2:378–86.
[81] Li P, Wong JJ, Sum C, Sin WX, Ng KQ, Koh MB, et al. IRF8 and IRF3 cooperatively
regulate rapid interferon-b induction in human blood monocytes. Blood
2011;117:2847–54.
[82] Bentz GL, Liu R, Hahn AM, Shackelford J, Pagano JS. Epstein–Barr virus BRLF1
inhibits transcription of IRF3 and IRF7 and suppresses induction of interferon-
beta. Virology 2010;402:121–8.
[83] Hahn AM, Huye LE, Ning S, Webster-Cyriaque J, Pagano JS. Interferon regula-
tory factor 7 is negatively regulated by the Epstein–Barr virus immediate-early
gene, BZLF-1. J Virol 2005;79:10040–52.
[84] Valente RM, Ehlers E, Xu D, Ahmad H, Steadman A, Blasnitz L, et al. Toll-like
receptor 7 stimulates the expression of Epstein–Barr virus latent membrane
protein 1. PLoS ONE 2012;7:e43317.
[85] Ning S, Hahn AM, Huye LE, Pagano JS. Interferon regulatory factor 7 regulates
expression of Epstein–Barr virus latent membrane protein 1: a regulatory
circuit. J Virol 2003;77:9359–68.
[86] Fathallah I, Parroche P, Gruffat H, Zannetti C, Johansson H, Yue J, et al. EBV
latent membrane protein 1 is a negative regulator of TLR9. J Immunol
2010;185:6439–47.
[87] Zhao B, Zou J, Wang H, Johannsen E, Peng CW, Quackenbush J, et al. Epstein–
Barr virus exploits intrinsic B-lymphocyte transcription programs to achieve
immortal cell growth. Proc Natl Acad Sci U S A 2011;108:14902–07.
[88] Mechelli R, Manzari C, Policano C, Annese A, Picardi E, Umeton R, et al. Epstein-
Barr virus genetic variants associated with multiple sclerosis. Neurology 2014.
Manuscript submitted.
[89] Comabella M, Kakalacheva K, Rı´o J, Mu¨ nz C, Montalban X, Lu¨nemann JD. EBV-
specific immune responses in patients with multiple sclerosis responding to
IFNb therapy. Mult Scler 2012;18:605–9.
[90] Balashov KE, Aung LL, Vaknin-Dembinsky A, Dhib-Jalbut S, Weiner HL. Inter-
feron-b inhibits toll-like receptor 9 processing in multiple sclerosis. Ann
Neurol 2010;68:899–906.
[91] Giacomini E, Severa M, Rizzo F, Mechelli R, Annibali V, Ristori G, et al. IFN-b
therapy modulates B-cell and monocyte crosstalk via TLR7 in multiple sclero-
sis patients. Eur J Immunol 2013;43:1963–72.
[92] Serra C, Mameli G, Arru G, Sotgiu S, Rosati G, Dolei A. In vitro modulation of the
multiple sclerosis (MS)-associated retrovirus by cytokines: implications for
MS pathogenesis. J Neurovirol 2003;9:637–43.
[93] Curtin F, Lang AB, Perron H, Laumonier M, Vidal V, Porchet HC, et al. GNbAC1, a
humanized monoclonal antibody against the envelope protein of multiple
sclerosis-associated endogenous retrovirus: a first-in-humans randomized
clinical study. Clin Ther 2012;34:2268–78.
[94] Dreyfus DH. Autoimmune disease: a role for new anti-viral therapies. Auto-
immun Rev 2011;11:88–97.
[95] Sorensen PS, Ross C, Clemmesen KM, Bendtzen K, Frederiksen JL, et al. Clinical
importance of neutralising antibodies against interferon beta in patients with
relapsing–remitting multiple sclerosis. Lancet 2003;362:1184–91.
Viviana Annibali graduated in biological sciences from
Roma Tre University of Rome (Italy) in 2001. In 2006 she
obtained her PhD in cell sciences and technologies and
after a postgraduate master degree in methodologies for
research and development of new therapies from
Sapienza University of Rome, where she recently earned
a clinical pathology specialty. From 2002 she is a re-
search scientist at the CENTERS, S. Andrea Hospital,
Department of Neuroscience, Mental Health and Senso-
ry Organs (NESMOS), Sapienza University of Rome. Dur-
ing her research activities on neurodegenerative
diseases she gained experience in gene expression anal-
ysis at the RNA and protein level in peripheral T cell
subsets. Dr. Annibali has also gained relevant experience in studying molecular
pathways and gene function via genetic and pharmacological approaches. Most
recently, she is investigating the involvement of B cells transcriptome dysfunctions
in multiple sclerosis disease, with particular attention to the role of the cellular and
extracellular microRNAs. She is a member of Italian Association of Neuroimmunol-
ogy (AINI).
Rosella Mechelli received her master degree in Biologi-
cal Sciences in 2000, PhD in genetic and molecular
biology in 2003 from Sapienza University of Rome
where she also earned a postgraduate master degree
in methodologies for the research and development of
new therapies in 2007. During her PhD training she
studied the structure of telomeric chromatin and its
epigenetic modifications. From 2004 she is a research
scientist at the CENTERS, S. Andrea Hospital, Depart-
ment of Neuroscience, Mental Health and Sensory
Organs (NESMOS), Sapienza University of Rome. Her
research interests lie in etiopathogenesis of multiple
sclerosis in monozygotic twins discordant for the dis-
ease. Currently most of her studies are focused on the interaction between heritable
and environmental risk factors, in particular on Epstein–Barr virus genetic variants
and virus–host interactions. She is a member of Italian Association of Neuroim-
munology (AINI).
Silvia Romano obtained a doctor of medicine degree
from Sapienza University of Rome in 2000 and a spe-
cialization degree in Neurology at Sapienza University
of Rome in 2006. She was also a Research Fellow at
Clinical and Behavioral Neurology Laboratory, S. Lucia
Foundation, Rome (2008–2009, project on neurocogni-
tive pattern of multiple sclerosis patients). She obtained
a PhD in Experimental Neurological Sciences in
2010 and then undertook post-doctoral work at the
Center of Experimental Neurological Therapies (CEN-
TERS), a Department Unit of S. Andrea Hospital,
Sapienza University of Rome (2012–2013) working on
demyelinating and hereditary neurodegenerative dis-
ease. Her current position is Researcher of Neurology at Department of Neuros-
ciences, Mental Health and Sensory Organs (NESMOS), at S. Andrea Hospital,
Sapienza University of Rome. Her research activity is focused on (1) etiopathogen-
esis, cognitive impairment and treatment of multiple sclerosis; (2) clinical features
and treatment of patients with hereditary cerebellar ataxias and Huntington
diseases.
Maria Chiara Buscarinu graduated in medicine and
surgery in 2006 and specialized in neurology in
2012 at the University of Sassari. From 2005 to
2011 she worked at the Neurological Clinic of Sassari.
She moved to Rome and she began her PhD in experi-
mental neurology at Sapienza University of Rome. She
carries out ambulatory activity and research at the S.
Andrea Hospital in Rome, with increased interest in
multiple sclerosis and etiopathogenetic factors related
to the disease. She is a member of the Italian Society of
Neuroimmunology (AINI) and the Italian Society of
Neurology (SIN).
Arianna Fornasiero obtained a doctor of medicine de-
gree from Sapienza University of Rome in 2004 and a
Specialization degree in Neurology at Sapienza Univer-
sity of Rome in 2009. From 2010 to 2014 she was
research fellow in experimental neurology at Sapienza
University of Rome. From 2009 to today she work at the
Center of Experimental Neurological Therapies (CEN-
TERS) a Department Unit of S. Andrea Hospital, Sapienza
University of Rome working on demyelinating disease.
Her research activity is focused on etiopathogenesis and
treatment of multiple sclerosis.
V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228 227
Renato Umeton obtained his bachelor’s and master’s
degrees in computer science from University of Calabria.
He earned his PhD in mathematics and informatics
focusing his research on optimization and ontology
studies that found their application in solving problems
in the area of medicine and biology. His experience
included working at Microsoft and at Massachusetts
Institute of Technology, as well as collaborating with
other major institutions such as the Harvard Medical
School and the University of Cambridge in the UK. He
carried out his most recent Postdoc at Sapienza Univer-
sity of Rome – S. Andrea Teaching Hospital, where he
applied his informatics and bioinformatics skills to un-
ravel the genetic and environmental components of the
pathogenesis of multiple sclerosis and other neurology-
related diseases.
Vito AG Ricigliano obtained his medical degree at
‘‘Sapienza’’ University of Rome, Italy, in 2013. He is
currently a MD at Center for Experimental Neurological
Therapies (CENTERS), S. Andrea hospital, Rome, and in
the present year he has been Academic visitor at the
University of Oxford, UK, Nuffield Department of Clini-
cal Neurosciences (NDCN). His research investigates the
role of gene–environment interactions in the etiology of
multiple sclerosis, especially focusing on the character-
ization of the interplay between EBV and the host net-
works, functional interpretation of GWAS data and use
of next-generation techniques (e.g. RNA-sequencing,
exome sequencing).
Francesco Orzi has spent several years dedicated to lab
research, in animal models of neurological diseases, and
in exploiting methods for assessment of brain functional
parameters. Following a 3 years stage (1979–1982) in
the Lab of Dr. L. Sokoloff at NIH, in Bethesda, he became
specifically interested in mapping local cerebral func-
tional changes in animal models. A few studies have
contributed to define the functional circuitry of the
Basal Ganglia in relation to their role in movement
disorders and in motivated behavior. Other studies,
since the early experiences in the laboratory of Dr Klatzo
at NIH, have been carried out in the field of the brain
damage maturation following temporary brain ische-
mia, and in the field of neuroprotection in animal models of brain ischemia. In the
last 15 years he has been fully involved in clinical neurology. Fields of interest are
cerebrovascular diseases and dementias. The focus is on mechanisms that underlie
neuronal degeneration associated with energy defects, dysfunction of the neuro-
muscular unit, and implications for neuroprotection.
Eliana Marina Coccia is head of the Anti-Infectious
Immunity Unit at the Department of Infectious, Parasitic
and Immunomediated Diseases, Istituto Superiore di
Sanita`, Rome-Italy. She received her Ph.D. in biological
sciences in 1984 from University of Rome, working on the
effect of type I IFN on the growth and differentiation of
Friend erythroleukemia cells. From 1984 to 1985, she was
a post-doctoral fellow at the Weizmann Institute of Sci-
ence (Rehovot, Isreal) in Dr. Michel Revel’s group where
she cloned the mouse 2-5A synthetase. In 1991–1992 she
moved at the Pasteur Institute (Paris, France) in the
laboratory of Dr. Ara Hovanessian to investigate the role
of type I IFN on HIV replication. The group lead by E.
Coccia is interested in understanding how type I IFN contribute to the induction of the
immune response against several pathogens, such as HIV, Mycobacterium tuberculosis,
Aspergillus fumigatus and Epstein–Barr virus. In particular the long-term objectives of
her projects is to investigate IFN-driven immune-regulation and aberrant activation
of IFN pathways in microbial infection and autoimmunity, with specific regard to B
lymphocytes and primary dendritic cells.
Marco Salvetti obtained his primary medical qualifica-
tion in 1986 from the Sapienza University of Rome
where he also trained as a clinical neurologist. He
was a postdoctoral fellow at the Max Planck Society
for Multiple Sclerosis in Prof. Hartmut Wekerle’s lab.
Following studies on the fine specificity of the T cell
response to putative autoantigens in multiple sclerosis,
he instituted the world largest twin registry in
1997. From then on, epidemiological, gene expression
and virological studies in twins with multiple sclerosis
begun. At present much of the studies are focused on the
interaction between heritable and environmental fac-
tors in the etiology of multiple sclerosis. This informa-
tion, combined with data from the in vitro screening of off-label activities of
registered drugs (on oligodendrocyte precursors), is exploited for the design of
exploratory clinical trials. These studies are carried out in the context of the clinical
research activity of CENTERS, an institution devoted to nonprofit, phase II trials in
multiple sclerosis and orphan neurological diseases.
Giovanni Ristori obtained in 1985 the degree in medi-
cine, at ‘Universita` Cattolica del Sacro Cuore’, Rome,
Italy and in 1989 the specialization in neurology at
the same University. In 1995 he obtained a PhD in
Neuroscience at Sapienza University of Rome, Italy.
The present position is at Neuroimmunology laboratory
and Neurogenetic Unit, Faculty of Medicine and Psy-
chology, Sapienza University of Rome, Italy.
V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228228

More Related Content

What's hot

EBV and kidney transplant
EBV and kidney transplantEBV and kidney transplant
EBV and kidney transplant
Pediatric Nephrology
 
Infections in immunocompromised patients
Infections in immunocompromised patientsInfections in immunocompromised patients
Infections in immunocompromised patients
dr.Ihsan alsaimary
 
10.1164@rccm.201706 1248 le
10.1164@rccm.201706 1248 le10.1164@rccm.201706 1248 le
10.1164@rccm.201706 1248 le
Julio A. Diaz M.
 
1-s2.0-S1472979215302419-main
1-s2.0-S1472979215302419-main1-s2.0-S1472979215302419-main
1-s2.0-S1472979215302419-main
Karina Chavez
 
Transplantation Immunity. Antitumor Immunity. Autoimmune Diseases
Transplantation Immunity. Antitumor Immunity. Autoimmune DiseasesTransplantation Immunity. Antitumor Immunity. Autoimmune Diseases
Transplantation Immunity. Antitumor Immunity. Autoimmune Diseases
Eneutron
 
Chikv zikv autoimmunity_f
Chikv zikv autoimmunity_fChikv zikv autoimmunity_f
Chikv zikv autoimmunity_f
anayajm
 
West Nile Virus and Other Domestic Nationally Notifiable Arboviral Diseases —...
West Nile Virus and Other Domestic Nationally Notifiable Arboviral Diseases —...West Nile Virus and Other Domestic Nationally Notifiable Arboviral Diseases —...
West Nile Virus and Other Domestic Nationally Notifiable Arboviral Diseases —...
IPN
 
International Journal of Virology & Infectious Diseases
International Journal of Virology & Infectious DiseasesInternational Journal of Virology & Infectious Diseases
International Journal of Virology & Infectious Diseases
SciRes Literature LLC. | Open Access Journals
 
Vaccination in ckd patients
Vaccination in ckd patientsVaccination in ckd patients
Vaccination in ckd patients
Apollo Hospitals
 
Confirmation of Safety of COVID 19 mRNA Vaccination for Cancer Patients
Confirmation of Safety of COVID 19 mRNA Vaccination for Cancer PatientsConfirmation of Safety of COVID 19 mRNA Vaccination for Cancer Patients
Confirmation of Safety of COVID 19 mRNA Vaccination for Cancer Patients
ijtsrd
 
Personalised treatment for asthma
Personalised treatment for asthmaPersonalised treatment for asthma
Personalised treatment for asthma
Nahid Sherbini
 
Medicine of the Future—The Transformation from Reactive to Proactive (P4) Med...
Medicine of the Future—The Transformation from Reactive to Proactive (P4) Med...Medicine of the Future—The Transformation from Reactive to Proactive (P4) Med...
Medicine of the Future—The Transformation from Reactive to Proactive (P4) Med...
Ryan Squire
 
Hypersensitivity pneumonitis
Hypersensitivity pneumonitisHypersensitivity pneumonitis
10.1164@rccm.201701 0053 ed
10.1164@rccm.201701 0053 ed10.1164@rccm.201701 0053 ed
10.1164@rccm.201701 0053 ed
Julio A. Diaz M.
 
Vih y paciente septico
Vih y paciente septicoVih y paciente septico
Vih y paciente septico
Aivan Lima
 
Abordaje de inmunodeficiencias primarias en pacientes con infección por micob...
Abordaje de inmunodeficiencias primarias en pacientes con infección por micob...Abordaje de inmunodeficiencias primarias en pacientes con infección por micob...
Abordaje de inmunodeficiencias primarias en pacientes con infección por micob...
Sociedad Latinoamericana de Alergia, Asma e Inmunología
 
Sesión Académica del CRAIC "Hipersensibilidad a quimioterapéuticos y anticuer...
Sesión Académica del CRAIC "Hipersensibilidad a quimioterapéuticos y anticuer...Sesión Académica del CRAIC "Hipersensibilidad a quimioterapéuticos y anticuer...
Sesión Académica del CRAIC "Hipersensibilidad a quimioterapéuticos y anticuer...
Sociedad Latinoamericana de Alergia, Asma e Inmunología
 
Primary immunodeficiency diseases by dr.gobinda
Primary immunodeficiency diseases by dr.gobindaPrimary immunodeficiency diseases by dr.gobinda
Primary immunodeficiency diseases by dr.gobinda
GOBINDA PRASAD PRADHAN
 
John Boikov H5N1 Essay, Mark - 95 out of 100
John Boikov H5N1 Essay, Mark - 95 out of 100John Boikov H5N1 Essay, Mark - 95 out of 100
John Boikov H5N1 Essay, Mark - 95 out of 100
John Boikov
 
Specific antibody deficiency
Specific antibody deficiencySpecific antibody deficiency

What's hot (20)

EBV and kidney transplant
EBV and kidney transplantEBV and kidney transplant
EBV and kidney transplant
 
Infections in immunocompromised patients
Infections in immunocompromised patientsInfections in immunocompromised patients
Infections in immunocompromised patients
 
10.1164@rccm.201706 1248 le
10.1164@rccm.201706 1248 le10.1164@rccm.201706 1248 le
10.1164@rccm.201706 1248 le
 
1-s2.0-S1472979215302419-main
1-s2.0-S1472979215302419-main1-s2.0-S1472979215302419-main
1-s2.0-S1472979215302419-main
 
Transplantation Immunity. Antitumor Immunity. Autoimmune Diseases
Transplantation Immunity. Antitumor Immunity. Autoimmune DiseasesTransplantation Immunity. Antitumor Immunity. Autoimmune Diseases
Transplantation Immunity. Antitumor Immunity. Autoimmune Diseases
 
Chikv zikv autoimmunity_f
Chikv zikv autoimmunity_fChikv zikv autoimmunity_f
Chikv zikv autoimmunity_f
 
West Nile Virus and Other Domestic Nationally Notifiable Arboviral Diseases —...
West Nile Virus and Other Domestic Nationally Notifiable Arboviral Diseases —...West Nile Virus and Other Domestic Nationally Notifiable Arboviral Diseases —...
West Nile Virus and Other Domestic Nationally Notifiable Arboviral Diseases —...
 
International Journal of Virology & Infectious Diseases
International Journal of Virology & Infectious DiseasesInternational Journal of Virology & Infectious Diseases
International Journal of Virology & Infectious Diseases
 
Vaccination in ckd patients
Vaccination in ckd patientsVaccination in ckd patients
Vaccination in ckd patients
 
Confirmation of Safety of COVID 19 mRNA Vaccination for Cancer Patients
Confirmation of Safety of COVID 19 mRNA Vaccination for Cancer PatientsConfirmation of Safety of COVID 19 mRNA Vaccination for Cancer Patients
Confirmation of Safety of COVID 19 mRNA Vaccination for Cancer Patients
 
Personalised treatment for asthma
Personalised treatment for asthmaPersonalised treatment for asthma
Personalised treatment for asthma
 
Medicine of the Future—The Transformation from Reactive to Proactive (P4) Med...
Medicine of the Future—The Transformation from Reactive to Proactive (P4) Med...Medicine of the Future—The Transformation from Reactive to Proactive (P4) Med...
Medicine of the Future—The Transformation from Reactive to Proactive (P4) Med...
 
Hypersensitivity pneumonitis
Hypersensitivity pneumonitisHypersensitivity pneumonitis
Hypersensitivity pneumonitis
 
10.1164@rccm.201701 0053 ed
10.1164@rccm.201701 0053 ed10.1164@rccm.201701 0053 ed
10.1164@rccm.201701 0053 ed
 
Vih y paciente septico
Vih y paciente septicoVih y paciente septico
Vih y paciente septico
 
Abordaje de inmunodeficiencias primarias en pacientes con infección por micob...
Abordaje de inmunodeficiencias primarias en pacientes con infección por micob...Abordaje de inmunodeficiencias primarias en pacientes con infección por micob...
Abordaje de inmunodeficiencias primarias en pacientes con infección por micob...
 
Sesión Académica del CRAIC "Hipersensibilidad a quimioterapéuticos y anticuer...
Sesión Académica del CRAIC "Hipersensibilidad a quimioterapéuticos y anticuer...Sesión Académica del CRAIC "Hipersensibilidad a quimioterapéuticos y anticuer...
Sesión Académica del CRAIC "Hipersensibilidad a quimioterapéuticos y anticuer...
 
Primary immunodeficiency diseases by dr.gobinda
Primary immunodeficiency diseases by dr.gobindaPrimary immunodeficiency diseases by dr.gobinda
Primary immunodeficiency diseases by dr.gobinda
 
John Boikov H5N1 Essay, Mark - 95 out of 100
John Boikov H5N1 Essay, Mark - 95 out of 100John Boikov H5N1 Essay, Mark - 95 out of 100
John Boikov H5N1 Essay, Mark - 95 out of 100
 
Specific antibody deficiency
Specific antibody deficiencySpecific antibody deficiency
Specific antibody deficiency
 

Similar to IFN- b and multiple sclerosis: From etiology to therapy and back

Infectious Myelopathies P.pptx
Infectious Myelopathies P.pptxInfectious Myelopathies P.pptx
Infectious Myelopathies P.pptx
Mohamed AbdElhady
 
Human Endogenous Retrovirus (paper)
Human Endogenous Retrovirus (paper)Human Endogenous Retrovirus (paper)
Human Endogenous Retrovirus (paper)
Kevin B Hugins
 
Mathematical modeling
Mathematical modelingMathematical modeling
Mathematical modeling
miguelhowella
 
Lombardi et al: XMRV/CFS Inflammatory Signature
Lombardi et al: XMRV/CFS Inflammatory SignatureLombardi et al: XMRV/CFS Inflammatory Signature
Lombardi et al: XMRV/CFS Inflammatory Signature
degarden
 
Human Endogenous Retrovirus (power point)
Human Endogenous Retrovirus (power point)Human Endogenous Retrovirus (power point)
Human Endogenous Retrovirus (power point)
Kevin B Hugins
 
Challenges to Cure: Transmission, Virulence and Pathogenesis of HIV Infection
Challenges to Cure: Transmission, Virulence and Pathogenesis of HIV InfectionChallenges to Cure: Transmission, Virulence and Pathogenesis of HIV Infection
Challenges to Cure: Transmission, Virulence and Pathogenesis of HIV Infection
SSR Institute of International Journal of Life Sciences
 
Walter Reed paper
Walter Reed paperWalter Reed paper
Walter Reed paper
PreveenRamamoorthy
 
Advances in Anti-Dengue Virus Research
Advances in Anti-Dengue Virus ResearchAdvances in Anti-Dengue Virus Research
Advances in Anti-Dengue Virus Research
Alaa Ahmed, M.Sc., Ph.D
 
Pathophysiology- Global Overview of Select Infectious Diseases
Pathophysiology- Global Overview  of Select Infectious DiseasesPathophysiology- Global Overview  of Select Infectious Diseases
Pathophysiology- Global Overview of Select Infectious Diseases
Imhotep Virtual Medical School
 
Epstein barr virus [autosaved]
Epstein barr virus [autosaved]Epstein barr virus [autosaved]
Epstein barr virus [autosaved]
Karthick Kumar
 
Rheumatic feve 2018
Rheumatic feve  2018Rheumatic feve  2018
Rheumatic feve 2018
rajasthan govt
 
Mecanismo les
Mecanismo lesMecanismo les
Mecanismo les
Joel Choca
 
EuroBioForum 2013 - Day 1 | Sergey Suchkov
EuroBioForum 2013 - Day 1 | Sergey SuchkovEuroBioForum 2013 - Day 1 | Sergey Suchkov
EuroBioForum 2013 - Day 1 | Sergey Suchkov
EuroBioForum
 
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
semualkaira
 
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
semualkaira
 
Great information Immune system
Great information Immune systemGreat information Immune system
Great information Immune system
affroz
 
Carvalho 2008
Carvalho 2008Carvalho 2008
Carvalho 2008
Karina Carvalho
 
Epstein-Barr virus genetic variants are associated with multiple sclerosis.
Epstein-Barr virus genetic variants are associated with multiple sclerosis.Epstein-Barr virus genetic variants are associated with multiple sclerosis.
Epstein-Barr virus genetic variants are associated with multiple sclerosis.
Mutiple Sclerosis
 
Analysis in to the Epidemiology and Pathophysiology of Respiratory Syncytial ...
Analysis in to the Epidemiology and Pathophysiology of Respiratory Syncytial ...Analysis in to the Epidemiology and Pathophysiology of Respiratory Syncytial ...
Analysis in to the Epidemiology and Pathophysiology of Respiratory Syncytial ...
Pırıl Erel
 
AIDS PRESANTATION.pptx
AIDS PRESANTATION.pptxAIDS PRESANTATION.pptx
AIDS PRESANTATION.pptx
Mkindi Mkindi
 

Similar to IFN- b and multiple sclerosis: From etiology to therapy and back (20)

Infectious Myelopathies P.pptx
Infectious Myelopathies P.pptxInfectious Myelopathies P.pptx
Infectious Myelopathies P.pptx
 
Human Endogenous Retrovirus (paper)
Human Endogenous Retrovirus (paper)Human Endogenous Retrovirus (paper)
Human Endogenous Retrovirus (paper)
 
Mathematical modeling
Mathematical modelingMathematical modeling
Mathematical modeling
 
Lombardi et al: XMRV/CFS Inflammatory Signature
Lombardi et al: XMRV/CFS Inflammatory SignatureLombardi et al: XMRV/CFS Inflammatory Signature
Lombardi et al: XMRV/CFS Inflammatory Signature
 
Human Endogenous Retrovirus (power point)
Human Endogenous Retrovirus (power point)Human Endogenous Retrovirus (power point)
Human Endogenous Retrovirus (power point)
 
Challenges to Cure: Transmission, Virulence and Pathogenesis of HIV Infection
Challenges to Cure: Transmission, Virulence and Pathogenesis of HIV InfectionChallenges to Cure: Transmission, Virulence and Pathogenesis of HIV Infection
Challenges to Cure: Transmission, Virulence and Pathogenesis of HIV Infection
 
Walter Reed paper
Walter Reed paperWalter Reed paper
Walter Reed paper
 
Advances in Anti-Dengue Virus Research
Advances in Anti-Dengue Virus ResearchAdvances in Anti-Dengue Virus Research
Advances in Anti-Dengue Virus Research
 
Pathophysiology- Global Overview of Select Infectious Diseases
Pathophysiology- Global Overview  of Select Infectious DiseasesPathophysiology- Global Overview  of Select Infectious Diseases
Pathophysiology- Global Overview of Select Infectious Diseases
 
Epstein barr virus [autosaved]
Epstein barr virus [autosaved]Epstein barr virus [autosaved]
Epstein barr virus [autosaved]
 
Rheumatic feve 2018
Rheumatic feve  2018Rheumatic feve  2018
Rheumatic feve 2018
 
Mecanismo les
Mecanismo lesMecanismo les
Mecanismo les
 
EuroBioForum 2013 - Day 1 | Sergey Suchkov
EuroBioForum 2013 - Day 1 | Sergey SuchkovEuroBioForum 2013 - Day 1 | Sergey Suchkov
EuroBioForum 2013 - Day 1 | Sergey Suchkov
 
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
 
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
High-Throughput Sequencing Reveals Micrornas from Plasma Exosomes as Potentia...
 
Great information Immune system
Great information Immune systemGreat information Immune system
Great information Immune system
 
Carvalho 2008
Carvalho 2008Carvalho 2008
Carvalho 2008
 
Epstein-Barr virus genetic variants are associated with multiple sclerosis.
Epstein-Barr virus genetic variants are associated with multiple sclerosis.Epstein-Barr virus genetic variants are associated with multiple sclerosis.
Epstein-Barr virus genetic variants are associated with multiple sclerosis.
 
Analysis in to the Epidemiology and Pathophysiology of Respiratory Syncytial ...
Analysis in to the Epidemiology and Pathophysiology of Respiratory Syncytial ...Analysis in to the Epidemiology and Pathophysiology of Respiratory Syncytial ...
Analysis in to the Epidemiology and Pathophysiology of Respiratory Syncytial ...
 
AIDS PRESANTATION.pptx
AIDS PRESANTATION.pptxAIDS PRESANTATION.pptx
AIDS PRESANTATION.pptx
 

Recently uploaded

Does Over-Masturbation Contribute to Chronic Prostatitis.pptx
Does Over-Masturbation Contribute to Chronic Prostatitis.pptxDoes Over-Masturbation Contribute to Chronic Prostatitis.pptx
Does Over-Masturbation Contribute to Chronic Prostatitis.pptx
walterHu5
 
OCT Training Course for clinical practice Part 1
OCT Training Course for clinical practice Part 1OCT Training Course for clinical practice Part 1
OCT Training Course for clinical practice Part 1
KafrELShiekh University
 
Top 10 Best Ayurvedic Kidney Stone Syrups in India
Top 10 Best Ayurvedic Kidney Stone Syrups in IndiaTop 10 Best Ayurvedic Kidney Stone Syrups in India
Top 10 Best Ayurvedic Kidney Stone Syrups in India
Swastik Ayurveda
 
Best Ayurvedic medicine for Gas and Indigestion
Best Ayurvedic medicine for Gas and IndigestionBest Ayurvedic medicine for Gas and Indigestion
Best Ayurvedic medicine for Gas and Indigestion
Swastik Ayurveda
 
Top-Vitamin-Supplement-Brands-in-India List
Top-Vitamin-Supplement-Brands-in-India ListTop-Vitamin-Supplement-Brands-in-India List
Top-Vitamin-Supplement-Brands-in-India List
SwisschemDerma
 
Cardiac Assessment for B.sc Nursing Student.pdf
Cardiac Assessment for B.sc Nursing Student.pdfCardiac Assessment for B.sc Nursing Student.pdf
Cardiac Assessment for B.sc Nursing Student.pdf
shivalingatalekar1
 
Abortion PG Seminar Power point presentation
Abortion PG Seminar Power point presentationAbortion PG Seminar Power point presentation
Abortion PG Seminar Power point presentation
AksshayaRajanbabu
 
Integrating Ayurveda into Parkinson’s Management: A Holistic Approach
Integrating Ayurveda into Parkinson’s Management: A Holistic ApproachIntegrating Ayurveda into Parkinson’s Management: A Holistic Approach
Integrating Ayurveda into Parkinson’s Management: A Holistic Approach
Ayurveda ForAll
 
Vestibulocochlear Nerve by Dr. Rabia Inam Gandapore.pptx
Vestibulocochlear Nerve by Dr. Rabia Inam Gandapore.pptxVestibulocochlear Nerve by Dr. Rabia Inam Gandapore.pptx
Vestibulocochlear Nerve by Dr. Rabia Inam Gandapore.pptx
Dr. Rabia Inam Gandapore
 
A Classical Text Review on Basavarajeeyam
A Classical Text Review on BasavarajeeyamA Classical Text Review on Basavarajeeyam
A Classical Text Review on Basavarajeeyam
Dr. Jyothirmai Paindla
 
CHEMOTHERAPY_RDP_CHAPTER 1_ANTI TB DRUGS.pdf
CHEMOTHERAPY_RDP_CHAPTER 1_ANTI TB DRUGS.pdfCHEMOTHERAPY_RDP_CHAPTER 1_ANTI TB DRUGS.pdf
CHEMOTHERAPY_RDP_CHAPTER 1_ANTI TB DRUGS.pdf
rishi2789
 
CBL Seminar 2024_Preliminary Program.pdf
CBL Seminar 2024_Preliminary Program.pdfCBL Seminar 2024_Preliminary Program.pdf
CBL Seminar 2024_Preliminary Program.pdf
suvadeepdas911
 
The Electrocardiogram - Physiologic Principles
The Electrocardiogram - Physiologic PrinciplesThe Electrocardiogram - Physiologic Principles
The Electrocardiogram - Physiologic Principles
MedicoseAcademics
 
K CỔ TỬ CUNG.pdf tự ghi chép, chữ hơi xấu
K CỔ TỬ CUNG.pdf tự ghi chép, chữ hơi xấuK CỔ TỬ CUNG.pdf tự ghi chép, chữ hơi xấu
K CỔ TỬ CUNG.pdf tự ghi chép, chữ hơi xấu
HongBiThi1
 
Clinic ^%[+27633867063*Abortion Pills For Sale In Tembisa Central
Clinic ^%[+27633867063*Abortion Pills For Sale In Tembisa CentralClinic ^%[+27633867063*Abortion Pills For Sale In Tembisa Central
Clinic ^%[+27633867063*Abortion Pills For Sale In Tembisa Central
19various
 
share - Lions, tigers, AI and health misinformation, oh my!.pptx
share - Lions, tigers, AI and health misinformation, oh my!.pptxshare - Lions, tigers, AI and health misinformation, oh my!.pptx
share - Lions, tigers, AI and health misinformation, oh my!.pptx
Tina Purnat
 
Netter's Atlas of Human Anatomy 7.ed.pdf
Netter's Atlas of Human Anatomy 7.ed.pdfNetter's Atlas of Human Anatomy 7.ed.pdf
Netter's Atlas of Human Anatomy 7.ed.pdf
BrissaOrtiz3
 
Ketone bodies and metabolism-biochemistry
Ketone bodies and metabolism-biochemistryKetone bodies and metabolism-biochemistry
Ketone bodies and metabolism-biochemistry
Dhayanithi C
 
Diabetic nephropathy diagnosis treatment
Diabetic nephropathy diagnosis treatmentDiabetic nephropathy diagnosis treatment
Diabetic nephropathy diagnosis treatment
arahmanzai5
 
TEST BANK For Basic and Clinical Pharmacology, 14th Edition by Bertram G. Kat...
TEST BANK For Basic and Clinical Pharmacology, 14th Edition by Bertram G. Kat...TEST BANK For Basic and Clinical Pharmacology, 14th Edition by Bertram G. Kat...
TEST BANK For Basic and Clinical Pharmacology, 14th Edition by Bertram G. Kat...
rightmanforbloodline
 

Recently uploaded (20)

Does Over-Masturbation Contribute to Chronic Prostatitis.pptx
Does Over-Masturbation Contribute to Chronic Prostatitis.pptxDoes Over-Masturbation Contribute to Chronic Prostatitis.pptx
Does Over-Masturbation Contribute to Chronic Prostatitis.pptx
 
OCT Training Course for clinical practice Part 1
OCT Training Course for clinical practice Part 1OCT Training Course for clinical practice Part 1
OCT Training Course for clinical practice Part 1
 
Top 10 Best Ayurvedic Kidney Stone Syrups in India
Top 10 Best Ayurvedic Kidney Stone Syrups in IndiaTop 10 Best Ayurvedic Kidney Stone Syrups in India
Top 10 Best Ayurvedic Kidney Stone Syrups in India
 
Best Ayurvedic medicine for Gas and Indigestion
Best Ayurvedic medicine for Gas and IndigestionBest Ayurvedic medicine for Gas and Indigestion
Best Ayurvedic medicine for Gas and Indigestion
 
Top-Vitamin-Supplement-Brands-in-India List
Top-Vitamin-Supplement-Brands-in-India ListTop-Vitamin-Supplement-Brands-in-India List
Top-Vitamin-Supplement-Brands-in-India List
 
Cardiac Assessment for B.sc Nursing Student.pdf
Cardiac Assessment for B.sc Nursing Student.pdfCardiac Assessment for B.sc Nursing Student.pdf
Cardiac Assessment for B.sc Nursing Student.pdf
 
Abortion PG Seminar Power point presentation
Abortion PG Seminar Power point presentationAbortion PG Seminar Power point presentation
Abortion PG Seminar Power point presentation
 
Integrating Ayurveda into Parkinson’s Management: A Holistic Approach
Integrating Ayurveda into Parkinson’s Management: A Holistic ApproachIntegrating Ayurveda into Parkinson’s Management: A Holistic Approach
Integrating Ayurveda into Parkinson’s Management: A Holistic Approach
 
Vestibulocochlear Nerve by Dr. Rabia Inam Gandapore.pptx
Vestibulocochlear Nerve by Dr. Rabia Inam Gandapore.pptxVestibulocochlear Nerve by Dr. Rabia Inam Gandapore.pptx
Vestibulocochlear Nerve by Dr. Rabia Inam Gandapore.pptx
 
A Classical Text Review on Basavarajeeyam
A Classical Text Review on BasavarajeeyamA Classical Text Review on Basavarajeeyam
A Classical Text Review on Basavarajeeyam
 
CHEMOTHERAPY_RDP_CHAPTER 1_ANTI TB DRUGS.pdf
CHEMOTHERAPY_RDP_CHAPTER 1_ANTI TB DRUGS.pdfCHEMOTHERAPY_RDP_CHAPTER 1_ANTI TB DRUGS.pdf
CHEMOTHERAPY_RDP_CHAPTER 1_ANTI TB DRUGS.pdf
 
CBL Seminar 2024_Preliminary Program.pdf
CBL Seminar 2024_Preliminary Program.pdfCBL Seminar 2024_Preliminary Program.pdf
CBL Seminar 2024_Preliminary Program.pdf
 
The Electrocardiogram - Physiologic Principles
The Electrocardiogram - Physiologic PrinciplesThe Electrocardiogram - Physiologic Principles
The Electrocardiogram - Physiologic Principles
 
K CỔ TỬ CUNG.pdf tự ghi chép, chữ hơi xấu
K CỔ TỬ CUNG.pdf tự ghi chép, chữ hơi xấuK CỔ TỬ CUNG.pdf tự ghi chép, chữ hơi xấu
K CỔ TỬ CUNG.pdf tự ghi chép, chữ hơi xấu
 
Clinic ^%[+27633867063*Abortion Pills For Sale In Tembisa Central
Clinic ^%[+27633867063*Abortion Pills For Sale In Tembisa CentralClinic ^%[+27633867063*Abortion Pills For Sale In Tembisa Central
Clinic ^%[+27633867063*Abortion Pills For Sale In Tembisa Central
 
share - Lions, tigers, AI and health misinformation, oh my!.pptx
share - Lions, tigers, AI and health misinformation, oh my!.pptxshare - Lions, tigers, AI and health misinformation, oh my!.pptx
share - Lions, tigers, AI and health misinformation, oh my!.pptx
 
Netter's Atlas of Human Anatomy 7.ed.pdf
Netter's Atlas of Human Anatomy 7.ed.pdfNetter's Atlas of Human Anatomy 7.ed.pdf
Netter's Atlas of Human Anatomy 7.ed.pdf
 
Ketone bodies and metabolism-biochemistry
Ketone bodies and metabolism-biochemistryKetone bodies and metabolism-biochemistry
Ketone bodies and metabolism-biochemistry
 
Diabetic nephropathy diagnosis treatment
Diabetic nephropathy diagnosis treatmentDiabetic nephropathy diagnosis treatment
Diabetic nephropathy diagnosis treatment
 
TEST BANK For Basic and Clinical Pharmacology, 14th Edition by Bertram G. Kat...
TEST BANK For Basic and Clinical Pharmacology, 14th Edition by Bertram G. Kat...TEST BANK For Basic and Clinical Pharmacology, 14th Edition by Bertram G. Kat...
TEST BANK For Basic and Clinical Pharmacology, 14th Edition by Bertram G. Kat...
 

IFN- b and multiple sclerosis: From etiology to therapy and back

  • 1. IFN-b and multiple sclerosis: From etiology to therapy and back V. Annibali a,1 , R. Mechelli a,1 , S. Romano a , M.C. Buscarinu a , A. Fornasiero a , R. Umeton a , V.A.G. Ricigliano a,b , F. Orzi c , E.M. Coccia d , M. Salvetti a, *, G. Ristori a a Centre for Experimental Neurological Therapies (CENTERS), Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy b Neuroimmunology Unit, Fondazione Santa Lucia-I.R.C.C.S., Rome, Italy c Neurology and Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, Italy d Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanita`, Rome, Italy 1. Introduction Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) characterized, in its most common clinical presentation, by an unpredictable occur- rence of relapse and remission phases [1–3]. The disease generally affects young adults [4], with a preference for female gender, as observed in many other immune-mediated conditions. Being a multifactorial disorder, its etiology involves both genetic and environmental risk factors. So far, genome-wide association studies (GWAS) have shown that genetic predisposition to MS is determined by more than 100 disease-associated susceptibility polymorphisms, located in coding and non-coding DNA [5]. Path- way analyses on MS-related genes demonstrated a relation with cellular networks specifically involved in immune cell functioning, antiviral response and interferon (IFN) signaling [6]. Major environmental risk factors for MS include Epstein–Barr virus (EBV) infection, the reactivation of human endogenous retroviruses (HERV) in specific conditions, vitamin D deficit and cigarette smoking, as supported by epidemiological surveys, serological evidences and other experimental laboratory based studies [7–13]. Nonetheless, a comprehensive overview of the events leading to MS development is still lacking. There is no cure for MS and treatments focus on treating relapses, slowing the progression of the disease and managing symptoms. Several immunomodulatory and immunosuppressive therapeutic agents are currently available for relapsing-remitting forms (RR) of MS, being interferon beta (IFN-b) the first therapeutic intervention able to interfere with the course of the disease and still the most used first-line treatment in RR MS. Cytokine & Growth Factor Reviews 26 (2015) 221–228 A R T I C L E I N F O Article history: Available online 31 October 2014 Keywords: Multiple sclerosis Interferon beta Epstein–Barr virus Human endogenous retroviruses Genome-wide Association Studies A B S T R A C T Several immunomodulatory treatments are currently available for relapsing-remitting forms of multiple sclerosis (RRMS). Interferon beta (IFN) was the first therapeutic intervention able to modify the course of the disease and it is still the most used first-line treatment in RRMS. Though two decades have passed since IFN-b was introduced in the management of MS, it remains a valid approach because of its good benefit/risk profile. This is witnessed by new efforts of pharmaceutical industry to improve this line: a PEGylated form of subcutaneous IFN-b 1a, (Plegridy1 ) with a longer half- life, has been recently approved in RRMS. This review will survey the various stages of the use of type I IFN in MS, with special attention to the effect of the treatment on the supposed viral etiologic factors associated to the disease. The antiviral activities of IFN (that initially prompted its use as immunomodulatory agent in MS), and the mounting evidences in favor of a viral etiology in MS, allowed us to outline a re-appraisal from etiology to therapy and back. ß 2014 Elsevier Ltd. All rights reserved. Abbreviations: ARR, annualized relapse rate; CIS, clinically isolated syndrome; CNS, central nervous system; EBNA, Epstein–Barr nuclear antigen; EBV, Epstein–Barr virus; EDSS, expanded disability status scale; ELISA, enzyme-linked immunosor- bent assay; GWAS, genome-wide association studies; HERV, human endogenous retroviruses; HIV, human immunodeficiency virus; IFN, Interferon; IFNAR, IFN–a receptor; IPA, ingenuity pathway analysis; IRF, interferon regulatory factor; ISGF, IFN-stimulated gene factor; JAK, Janus-family tyrosine kinases; LMP1, latent membrane protein-1; MRI, magnetic resonance imaging; MS, multiple sclerosis; MSRV, MS-associated retroviruses; MxA, myxovirus-induced protein A; NAbs, neutralizing antibodies; 20 ,50 -OAS, 20 ,50 -oligoadenylate synthetase; PEG, polyeth- ylene glycol; RAL, raltegravir; RR, relapsing–remitting forms; STAT, signal transducers and activators of transcription; SNPs, single nucleotide polymor- phisms; TLR, toll-like receptors; TYK, tyrosine kinase. * Corresponding author at: Neurologia, Ospedale S. Andrea, Via di Grottarossa 1035, 00189 Rome, Italy. Tel.: +39 06 33775994; fax: +39 06 33775900. E-mail address: marco.salvetti@uniroma1.it (M. Salvetti). 1 These authors contributed equally to this work. Contents lists available at ScienceDirect Cytokine & Growth Factor Reviews journal homepage: www.elsevier.com/locate/cytogfr http://dx.doi.org/10.1016/j.cytogfr.2014.10.010 1359-6101/ß 2014 Elsevier Ltd. All rights reserved.
  • 2. 2. From etiology. . . The hypothesis of a viral etiology in MS led to several investigations on a large number of microbes, that, after an initial enthusiastic attention, failed to be demonstrably associated to MS. At the moment two agents seem consistently linked to diseases development: EBV and HERV. Converging epidemiological, clinical and laboratory studies support an etiologic role for EBV in MS [14]. EBV is a g-herpesvirus that infect quite all the adult population and that persists in infected B cells in a latent or lytic phase [15]. Humans are the exclusive natural host for EBV which may explain why MS is unique to humans [16]. Prospective studies have shown that elevation in serum antibody titers to EBV precedes the occurrence of MS [17,18]. Epstein–Barr viral load in the peripheral blood of healthy adults may predict the risk of MS [19], while in children who develop MS immunoreactivity to EBV, but not to other viruses, is higher than in controls [20,21]. A history of late EBV infection and of infectious mononucleosis (IM is often the clinical manifestation of a late primary EBV infection) is strongly associated to MS [22]. EBV may be a target of oligoclonal cerebrospinal fluid IgG [23], CD8+ T cells [24–26] and CD4 response [27], and a vast literature on the cross-reactivity between EBV and myelin epitopes was produced over the past two decades [28–30]. Recent works provided further evidences aimed at clarifying how EBV contributes to disease development. An inadequate control of EBV at primary infection or at a later stage can lead to low grade, persistently active EBV infection in CNS infiltrating B cells [26,31–33]. A ‘‘candidate-interactome’’ aggregate analysis of genome-wide association data in multiple sclerosis demonstrated a significant enrichment of potential interactions between the virus and MS-related genomic regions [34]. The EBV infection of the MS brain as cause of CNS damage that remains controversial [35–37], is supported by several recent studies demonstrating a selective enrichment of EBV-specific CD8+ T cells in the cerebrospinal fluid of MS patients [38,39] and the presence of EBV DNA in brain (Mechelli et al., manuscript submitted). Besides to role of herpesviruses many studies support a potential contribution for HERV to MS development. Retroviruses are RNA viruses that may cause a spectrum of diseases of the nervous system. Their genome contains three genetic domains: env is responsible for the surface glycoproteins and trans-membrane; gag encodes the proteins necessary for viral assembly, including matrix proteins and core shell; pol encodes the enzymes needed for viral replication, such as reverse transcriptase, protease, ribonu- clease and integrase [40]. Specific sequences within retroviral genes can lead to the development of neurovirulence, in particular, the proteins env- associated, which mediates the binding of the virus to the cell membrane surface. Neurovirulent retroviruses are able to activate the host immune response that, through pro-inflammatory molecules and neurotoxic molecules, ultimately leads to neuronal death [40]. In 1997, Perron described the isolation and identification of new retrovirus particles from cell cultures of leptomeninges, choroid plexus and peripheral B lymphocytes in MS patients. This study provided molecular evidence that the production of extracellular virions containing MS-associated retroviruses (MSRV) pol se- quence was associated with MS. This virus, previously called LM7, was a new retrovirus which was present in the cerebrospinal fluid of patients with MS [41]. The same group showed the production of a specific envelope protein with gliotoxic and pro-inflammatory actions that may be crucial in MS pathogenesis [42]. Further studies have tried to explain and confirm the association between MS and the expression of MSRV envelope (Env), providing evidences that the retrovirus appears to be related to MS clinical progression [43,44]. Recently, env antigen was detected in the serum of 73% of MS patients with similar prevalence in all clinical forms, and not in subjects affected by other inflammatory diseases. The different forms of the disease (primary-progressive, RR and clinically isolated syndrome-CIS) show different ELISA (enzyme- linked immunosorbent assay) and/or PCR profiles indicative of an increase with the evolution of the disease [45]. 3. To therapy. . . Interferons were discovered by Isaacs and Lindenmann [46]; they use this term to describe a soluble substance with biological activity able to interfere with viral replication. Due to their antiviral activities and considering the plausible viral etiology of MS, IFNs, regardless of their type, were proposed as immunomod- ulatory therapeutic agents in MS patients. The first trials using IFN-g (a type II IFN) were conducted in the late 1980s and were interrupted because the treated patients showed an increase of severity and frequency of relapses [47,48]. These negative results led to study another type of IFNs (type I), IFN-a and IFN-b, that may act as inhibitors of IFN-g. Different preparations of IFN-a resulted in reduction of clinical relapses and activity at magnetic resonance imaging (MRI) in MS patients, but unacceptable side effects precluded its use in clinical practice [49–53]. IFN-b was similarly effective in decreasing disease activity and showed an acceptable risk profile, thus becoming the first disease-modifying therapy for MS (Table 1). Subcutaneous IFN-b 1b (Betaseron1 ) was the first immuno- modulatory therapy to receive approval for the treatment of RRMS in 1993. It is produced by recombinant DNA technology in the bacterial cell (Escherichia coli) and currently is the only IFN-b licensed for RR and secondary progressive (SP) MS [54]. In the registration trial 372 patients were randomized to receive placebo or IFN-b 1b (50 or 250 mcg subcutaneously every other day) for 2 years. The annualized relapse rate (ARR) was significantly lower in the IFN-b1b treated groups compared to the placebo group with a dosage effect. Moreover a significative reduction of activity at MRI activity was showed. No difference in disease progression between treatment and placebo groups was demonstrated [55,56]. IFN-b1a IM (Avonex1 ) is produced in Chinese hamster ovary cells and was approved for treatment in RRMS in 1996. In the Table 1 INF-b formulations approved for RRMS. For detailed description, see text. Rebif 22/44 Avonex Betaferon/Betaseron/Extavia IFN subtype Beta 1a Beta 1a Beta 1b Production CHO CHO E. coli Aminoacid 166 aa 166 aa 165 aa Glycosylation 1 N-linked complex 1 N-linked complex None Administration SC, 3 times/week IM, 1 time/week SC, every other day Weekly dose 66/132 mcg 30 mcg 875 mcg CHO = Chinese hamster ovary cells; SC = subcutaneous; IM = intramuscular. V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228222
  • 3. pivotal study 301 patients with expanded disability status scale (EDSS) score of 1.0–3.5 and at least two relapses in the preceding 3 years were randomized to receive placebo or IFN-b 1a (30 mg intramuscularly once weekly) for 2 years [57,58]. The IFN-b 1a group showed a significant decrease in the disease activity compared with placebo. IFN-b 1a SC (Rebif1 ) is also produced in mammalian Chinese hamster ovary cells using DNA technology. It was approved for treatment of RRMS in 1998 in Europe and Canada and in 2002 in the USA. In the PRISM study [59] 560 patients with an EDSS score between 1.0 and 5.0 and at least two relapses in the preceding 2 years were randomized to receive placebo or IFN-b 1a (22 or 44 mg subcutaneously three times weekly). After 2 years of treatment, IFN-b 1a showed significant results compared with placebo in relapse rate and MRI activity with a statistically significant dose-effect. While the pivotal clinical trials consistently demonstrated that both forms of IFN-b reduce ARR by about 1/3 and new brain MRI lesions over periods of 1–3 years in RRMS, four randomized, placebo-controlled trials demonstrated poor or no effects on established progressive MS [54,60–63]. IFN-b has a significant effect in the earlier inflammatory stages of the disease: three large clinical trials (CHAMPS, ETOMS and BENEFIT) in CIS patients, demonstrated an effect on clinical and MRI measures of disease activity delaying the development to clinically definite (CD) MS [64–66]. Although these studies had limited comparability because of different patient populations recruited (BENEFIT and ETOMS enrolled patients with multifocal manifestations at onset, while CHAMPS enrolled patients with monofocal forms), the risk of progression to CDMS was comparably reduced by 40–50%. Considering the different formulations of IFN-b, head-to-head trials were conducted to compare the different licensed IFNs (EVIDENCE and INCOMIN). These studies demonstrated that increasing the dose of IFN-b (more frequent dosing schedule or higher dose) gave greater benefit than lower doses, supporting a dose–response relationship [67,68]. The IFNs-b are generally well tolerated, being the most frequent side effects injection-site reaction, and a flu-like syndrome that tends to wanes over time in most patients. Lymphopenia, hepatic failure, hepatitis, and elevated liver enzymes have also been reported especially during IFN-b-1b treatment [69]. Though IFN-b therapy represents a significant advance in the management of MS, the treatment response is not uniform and clinical experience shows that about 40% of the MS patients do not or only poorly respond to IFN-b treatment (‘‘non- responders’’) [70]. So far there are no established biological markers able to predict the response. The development of neutralizing antibodies (NAbs), which at high titers may block the biological response of the drug with a reduced efficacy, can contribute to treatment failure [71,72]. Persistent high-titers of NAbs depend on the formulation and dosing regimen, and occur more commonly with subcutaneous preparations [73]. IFN was tried in MS as a ‘general’ antiviral approach and gave positive results as disease modifying therapy. This was not the case with more ‘specific’ antiviral treatment such as those active on herpes viruses. Over the last twenty years several clinical trials have been carried out especially with acyclovir and valacyclovir [74–76]. Overall, these studies did not obtain significant results in favor of the drug compared to placebo, though positive trends were noted. An analysis that also took into account the pharmacokinetic of acyclovir and valacyclovir suggested an inhibitory effect on some viruses (Herpes viruses 1, 2, 6 and varicella zoster virus) but not on others that seem to have a greater correlation with MS (EBV, Herpes virus 6, and MSRV), thus explaining, at least in part, the failure of this approach [77]. Further studies with new antiviral drugs having improved pharmacological characteristics and antiretroviral activity may result in better outcome and are currently actively investigated (see Raltegravir in next section). 4. And back. . . 4.1. MS genome-wide association studies and IFN-b pathway The exact mode of action of IFN-b in MS is likely to be complex and is not yet fully understood. This topic will be not treated in the present review, being object of other contributions in this issue. At the molecular level IFN-b is recognized by the IFN-a receptor (IFNAR), which is found on many different cell types. The receptor is a heterodimer formed by IFNAR1 and IFNAR2, which assemble into a functional receptor complex and initiates the signal transduction pathway that involves the phosphorylation of several intracellular mediators. Upon assembly of the IFN receptor complex, the intracellular domains of IFNAR1 and IFNAR2 active Janus kinases 1 (JAK1) and tyrosine kinase 2 (TYK2). The JAK1/TYK2 along with IFNAR, phosphorylate signal transducers and activators of transcription (STAT) 1, and 2, which dimerize and form a complex with interferon regulatory factor 9 (IRF9). The STA- T1:2:IRF9 complex is a transcription factor (IFN-stimulated gene factor, ISGF3), which translocates to the nucleus and binds to the IFN-stimulated response element (ISRE) of multiple genes [78]. Different kinds of genes are targeted by ISGF3 complex, including early genes such as IFN regulatory factor-1 (IRF-1), the primary positive regulator of IFN production, and IRF-2, an inhibitor of IFN production. The later genes include the IFN-b itself and antiviral proteins such as 20 ,50 -oligoadenylate synthetase (20 ,50 -OAS) and myxovirus-induced protein A (MxA), which are specifically induced by type I IFNs. MxA is the established marker of IFN-b biological activity in IFN-b-treated MS patients [79]. Some genes related to the IFN-b signaling pathway showed single nucleotide polymorphisms (SNPs) that resulted to be MS- associated in GWAS [5]. To review this, we highlighted the network that leads to the connectivity between the IFN-b signaling pathway and genes exceeding the genome-wide significance threshold in GWAS published from 2007 in MS (http://www.ge- nome.gov/gwastudies/). To perform this analysis it was used Quigen Ingenuity Pathway Analysis (IPA), which was set to run a ‘‘core analysis’’ correlating all MS-associated genes (retrieved on 8/ 26/2014) with the known interactors of IFN-b (314 molecules present in IPA version 21249400); the analysis included only experimental evidence observed in human samples. Among all known interactors of IFN-b, 18 genes were also MS-associated (Table 2 and Fig. 1, that shows most of these connections and how they relate to IFN-b). The list includes several genes that control the immune responses (including major histocompatibility complex alleles, cytokines and co-stimulator molecules), as well as direct interactors with IFN signaling such as IRF8, NFKB1 and TYK2 [80,81]. This IPA analysis showed a significant (p- value < 1.18 Â 10À7 ) relationship between the MS-associated genes and the IFN-b signaling and confirmed previous results obtained by our group with another approach [6]. Overall, these data suggest that single unfavorable SNPs (or a combination of them) affecting components of IFN-b signaling may determine some deregulation in MS. Further investigation are needed to clarify the role of these components in MS pathogenesis and possible corrective effects of exogenous IFN-b on deregulated pathways (see next section). 4.2. EBV and IFN-b EBV is a kind of ‘‘one man band’’ in its ability to control the antiviral immune response of infected cells both in lytic and latent phase. IFN pathway is no exception, being sabotaged by multiple V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228 223
  • 4. mechanisms of immune evasion. In an in vitro setting was demonstrated that the expression of BRLF1 and BZLF1 (two immediate-early transcription factors that controls the initiation of viral lytic gene expression and lytic reactivation from latency) reduce the IFN-b production down modulating the expression of IRF3 and IRF7 [82,83]. During the latent phase the up-regulation of latent membrane protein-1 (LMP1), possibly due to a toll-like receptors 7 (TLR7) aberrant activation, may blocks TYK2 and the consequent STATs phosphorylation, inhibiting the expression of IFN-b stimulated genes [84,85]. Moreover EBV infection of primary B cells may reduce the cellular antiviral activity inhibiting the TLR9 activation through the expression of LMP1 [86], that in turn may be Table 2 The MS-associated genes of the IFN-b signaling pathway. For detailed description, see text. Gene symbol Description Location Type CD40 CD40 molecule, TNF receptor super family member 5 Plasma membrane Transmembranereceptor CD86 CD86 molecule Plasma membrane Transmembranereceptor HLA-B Major histocompatibility complex, class I, B Plasma membrane Transmembranereceptor HLA-DQA1 Major histocompatibility complex, class II, DQ alpha 1 Plasma membrane Transmembranereceptor HLA-DQB1 Major histocompatibility complex, class II, DQ beta 1 Plasma membrane Other HLA-DRA Major histocompatibility complex, class II, DR alpha Plasma membrane Transmembranereceptor HLA-DRB1 Major histocompatibility complex, class II, DR beta 1 Plasma membrane Transmembranereceptor IL12A Interleukin 12A Extracellular space Cytokine IL12B Interleukin 12B Extracellular space Cytokine IRF8 Interferon regulatory factor 8 Nucleus Transmembranereceptor MAPK1 Mitogen-activated protein kinase 1 Cytoplasm Kinase MERTK MER proto-oncogene, tyrosine kinase Plasma membrane Kinase MMP10 Matrix metallopeptidase 10 (stromelysin 2) Extracellular space Peptidase MYC V-myc avian myelocytomatosis viral oncogene homolog Nucleus Transmembranereceptor NFKB1 Nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 Nucleus Transmembranereceptor PRKRA Protein kinase, interferon-inducible double stranded RNA dependent activator Cytoplasm Other STAT3 Signal transducer and activator of transcription 3 Nucleus Transmembranereceptor TYK2 Tyrosinekinase 2 Plasma membrane Kinase Fig. 1. Representation of the interactions between proteins coded by MS-associated and IFN beta-related genes (see Table 2 for the list of the interaction points strictly shared by the MS-associated gene list and the IFN interactors). V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228224
  • 5. up-regulated by Epstein–Barr nuclear antigen 2 (EBNA2). This protein is able to control gene expression of viral and cellular genes, mainly during the first phase of the infection [87]. Some evidences suggest its potential implication in MS pathology: EBNA2 expressing cells have been observed in affected brains [31] and specific EBNA2 genotypes associate with disease status [88]. It seems plausible that IFN-b therapy may compensate for some of the EBV-induced dysfunctions in the antiviral immune responses: CD8+ T cells specific for lytic-phase antigens are detected with high frequency in the peripheral blood of patients with active disease and are reduced by IFN-b treatment [26], as well as CD4+ T cell response to EBNA1 peptides pool [89]; in dendritic cells obtained from MS patients under IFN-b treatment a reduced TLR9 activation (that promote pro-inflammatory responses) was observed [90]; a recent work demonstrated an impaired activation of TLR7 in MS subjects, that decrease the ability of B cells to mature in plasma cells and that is restored by IFN-b treatment [91]. 4.3. MSRV and IFN-b IFN-b appears to be capable of interfering with MSRV biology. An in vitro study showed that IFN-b inhibits the release of MSRV from peripheral blood mononuclear cells derived from MS patients [92]. These data were confirmed through a longitudinal evaluation of patients with MS, during one year of therapy with IFN-b: the MSRV load in the blood was directly related to the duration of MS and underwent a considerable reduction to below the limits of detection within 3 months of IFN therapy; this work suggested to consider the evaluation of MSRV in plasma as a prognostic marker to monitor the progression of the disease and the outcome of therapy [43]. At variance with several trials conducted with anti-herpes drugs, and notwithstanding evidences of retroviral contribution to disease pathogenesis, no major attempt has been performed with anti-retroviral therapy in MS, except for a humanized monoclonal antibody against the envelope of MSRV, that was tried in a phase I study [93]. A pilot study, that is ongoing, may herald such an approach, investigating raltegravir (RAL) in relapsing remitting MS (ClinicalTrials.gov Identifier: NCT01767701). RAL is an inhibitor of human immunodeficiency virus (HIV) integrase, approved in 2007 for clinical use as antiretroviral agent in HIV infected adults. Clinical studies and subsequent clinical experience have shown durable virologic suppression, low rates of adverse effects and long-term safety. Not interacting with the cytochrome P450 system, RAL may be a good option for polytherapy. As an inhibitor of retroviral integrase, RAL can be active against the MSRV that is transactivated by several viruses, EBV being one of these. RAL is also able to inhibit recombinase and terminase, two key proteins for EBV [94]. Altogether, RAL seems to be a good candidate to tackle plausible etiologic agents for MS and might also add to the effects of IFN. 5. Conclusions Though two decades have passed since IFN-b was introduced in the management of MS, it remains a valid approach because of its good benefit/risk profile. The persisting interest is witnessed by new efforts that pharmaceutical industry has produced to improve this line. Recently, a PEGylated form of subcutaneous IFN-b 1a (Ple- gridy1 ) with a longer half-life (injection frequency every 2 weeks) has been approved in RRMS. Conjugation of IFN-b 1a with a molecule of polyethylene glycol (PEG; PEGylation) increases the size of the product resulting in more solubility, half-life and efficacy the drug. Compared to placebo, PEG INF has reduced ARR by about one-third (0.397 in the placebo group versus 0.256 in the every 2 weeks group). A slight reduction in sustained disability progression and in several MRI activity measures has also been demonstrated. The drug was generally well tolerated: the most common adverse events were influenza-like illness, injection-site reactions and headache [95]. Due to its frequency of administra- tion (every 2 weeks), PEG IFN-b1a may have a better safety profile than other IFNs-b formulations. Moreover, results from the extension of the phase III study showed that the therapeutic effects of PLEGRIDY may reach a size that was not attained by increasing doses of non-pegylated IFN-b and may become even more relevant over time, suggesting that prolonged treatment with PLEGRIDY may induce therapeutic effects that go beyond the immunomodulatory action of IFN-b. Given the potent antiviral effects of type-1 IFN, the added value of PLEGRIDY treatment might be related to its ability to target more efficiently the non-heritable (i.e. viral) cause(s) of MS. This hints at future therapeutic approaches based on type 1 interferon alone or in association with specific antiviral drugs that might act as an etiologic treatment for MS. The main fields of investigation regard: (a) etiopathogenesis of multiple sclerosis; (b) the identification of the world’s largest twin registry in Italian population; the registry is currently exploited for concordance studies and for laboratory investigations (studies on twin pairs discordant for disease); and (c) clinical trials (especially phase II independent studies) in patients with multiple sclerosis, Huntington disease and cerebellar ataxia. Conflict of interest MS receives research support and has received fees as speaker from Sanofi-Aventis, Biogen, Bayer Schering, and Merck Serono. Acknowledgements MS is supported from: Italian Multiple Sclerosis Foundation (Fondazione Italiana Sclerosi Multipla grant number: 2011/R/31) and Italian Ministry of Health (Ministero della Salute, grant number: RF-2010-2321254). References [1] Amato MP, Ponziani G, Bartolozzi ML, Siracusa G. A prospective study on the natural history of multiple sclerosis: clues to the conduct and interpretation of clinical trials. J Neurol Sci 1999;168:96–106. [2] Scalfari A, Neuhaus A, Degenhardt A, Rice GP, Muraro PA, Daumer M, et al. The natural history of multiple sclerosis, a geographically based study: relapses and long-term disability. Brain 2010;133:1914–29. [3] Bordi I, Umeton R, Ricigliano VA, Annibali V, Mechelli R, Ristori G, et al. A mechanistic, stochastic model helps understand multiple sclerosis course and pathogenesis. Int J Genomics 2013;2013:910321. [4] Koch-Henriksen N. The Danish multiple sclerosis registry: a 50-year follow-up. Mult Scler 1999;5:293–6. [5] International Multiple Sclerosis Genetics Consortium (IMSGC), Beecham AH, Patsopoulos NA, Xifara DK, Davis MF, Kemppinen A, et al. Analysis of immune- related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat Genet 2013;45:1353–60. [6] Ricigliano VA, Umeton R, Germinario L, Alma E, Briani M, Di Segni N, et al. Contribution of genome-wide association studies to scientific research: a pragmatic approach to evaluate their impact. PLOS ONE 2013;8:e71198. [7] Kakalacheva K, Lu¨nemann JD. Environmental triggers of multiple sclerosis. FEBS Lett 2011;585:3724–9. [8] Pender MP. The essential role of Epstein–Barr virus in the pathogenesis of multiple sclerosis. Neuroscientist 2011;17:351–67. [9] Almohmeed YH, Avenell A, Aucott L, Vickers MA. Systematic review and meta- analysis of the sero-epidemiological association between Epstein Barr virus and multiple sclerosis. PLOS ONE 2013;8:e61110. [10] Simon KC, Munger KL, Ascherio A. Vitamin D and multiple sclerosis: epidemi- ology, immunology, and genetics. Curr Opin Neurol 2012;25:246–51. V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228 225
  • 6. [11] Handel AE, Williamson AJ, Disanto G, Dobson R, Giovannoni G, Ramagopalan SV. Smoking and multiple sclerosis: an updated meta-analysis. PLoS ONE 2011;6:e16149. [12] Sutkowski N, Conrad B, Thorley-Lawson DA, Huber BT. Epstein–Barr virus transactivates the human endogenous retrovirus HERV-K18 that encodes a superantigen. Immunity 2001;15:579–89. [13] Antony JM, Deslauriers AM, Bhat RK, Ellestad KK, Power C. Human endogenous retroviruses and multiple sclerosis: innocent bystanders or disease determi- nants. Biochim Biophys Acta 2011;1812:162–76. [14] Lu¨ nemann JD. Epstein–Barr virus in multiple sclerosis: a continuing conun- drum. Neurology 2012;78:11–2. [15] Cohen JI. Epstein–Barr virus infection. N Engl J Med 2000;343:481–92. [16] Haahr S, Ho¨llsberg P. Multiple sclerosis is linked to Epstein–Barr virus infec- tion. Rev Med Virol 2006;16:297–310. [17] Levin LI, Munger KL, Rubertone MV, Peck CA, Lennette ET, Spiegelman D, et al. Temporal relationship between elevation of Epstein–Barr virus antibody titers and initial onset of neurological symptoms in multiple sclerosis. JAMA 2005;293:2496–500. [18] DeLorenze GN, Munger KL, Lennette ET, Orentreich N, Vogelman JH, Ascherio A. Epstein–Barr virus and multiple sclerosis: evidence of association from a prospective study with long-term follow-up. Arch Neurol 2006;63:839–44. [19] Wagner HJ, Munger KL, Ascherio A. Plasma viral load of Epstein–Barr virus and risk of multiple. Eur J Neurol 2004;11:833–4. [20] Alotaibi S, Kennedy J, Tellier R, Stephens D, Banwell B. Epstein–Barr virus in pediatric multiple sclerosis. JAMA 2004;291:1875–9. [21] Banwell B, Krupp L, Kennedy J, Tellier R, Tenembaum S, Ness J, et al. Clinical features and viral serologies in children with multiple sclerosis: a multina- tional observational study. Lancet Neurol 2007;6:773–81. [22] Handel AE, Williamson AJ, Disanto G, Handunnetthi L, Giovannoni G, Rama- gopalan SV. An updated meta-analysis of risk of multiple sclerosis following infectious mononucleosis. PLoS ONE 2010;5(9). [23] Cepok S, Zhou D, Srivastava R, Nessler S, Stei S, Bu¨ ssow K, et al. Identification of Epstein–Barr virus proteins as putative targets of the immune response in multiple sclerosis. J Clin Invest 2005;115:1352–60. [24] Jilek S, Schluep M, Meylan P, Vingerhoets F, Guignard L, Monney A, et al. Strong EBV-specific CD8+ T-cell response in patients with early multiple sclerosis. Brain 2008;131:1712–21. [25] Jilek S, Schluep M, Harari A, Canales M, Lysandropoulos A, Zekeridou A, et al. HLAB7-restricted EBV specific CD8+ T cells are dysregulated in multiple sclerosis. J Immunol 2012;188:4671–80. [26] Angelini DF, Serafini B, Piras E, Severa M, Coccia EM, Rosicarelli B, et al. Increased CD8+ T cell response to Epstein–Barr virus lytic antigens in the active phase of multiple sclerosis. PLOS Pathog 2013;9:e1003220. [27] Lu¨ nemann JD, Edwards N, Muraro PA, Hayashi S, Cohen JI, Mu¨nz C, et al. Increased frequency and broadened specificity of latent EBV nuclear antigen- 1-specific T cells in multiple sclerosis. Brain 2006;129:1493–506. [28] Ufret-Vincenty RL, Quigley L, Tresser N, Pak SH, Gado A, Hausmann S, et al. In vivo survival of viral antigen-specific T cells that induce experimental auto- immune encephalomyelitis. J Exp Med 1998;188:1725–38. [29] Lu¨ nemann JD, Jelcic´ I, Roberts S, Lutterotti A, Tackenberg B, Martin R, et al. EBNA1-specific T cells from patients with multiple sclerosis cross react with myelin antigens and co-produce IFN-gamma and IL-2. J Exp Med 2008;205:1763–73. [30] Lang HL, Jacobsen H, Ikemizu S, Andersson C, Harlos K, Madsen L, et al. A functional and structural basis for TCR cross-reactivity in multiple sclerosis. Nat Immunol 2002;3:940–3. [31] Serafini B, Rosicarelli B, Franciotta D, Magliozzi R, Reynolds R, Cinque P, et al. Dysregulated Epstein–Barr virus infection in the multiple sclerosis brain. J Exp Med 2007;204:2899–912. [32] Serafini B, Severa M, Columba-Cabezas S, Rosicarelli B, Veroni C, Chiappetta G, et al. Epstein–Barr virus latent infection and BAFF expression in B cells in the multiple sclerosis brain: implications for viral persistence and intrathecal B- cell activation. J Neuropathol Exp Neurol 2010;69:677–93. [33] Serafini B, Muzio L, Rosicarelli B, Aloisi F. Radioactive in situ hybridization for Epstein–Barr virus-encoded small RNA supports presence of Epstein–Barr virus in the multiple sclerosis brain. Brain 2013;136:e233. [34] Mechelli R, Umeton R, Policano C, Annibali V, Coarelli G, Ricigliano VA, et al. A candidate-interactome aggregate analysis of genome-wide association data in multiple sclerosis. PLOS ONE 2013;8:e63300. [35] Aloisi F, Serafini B, Magliozzi R, Howell OW, Reynolds R. Detection of Epstein– Barr virus and B-cell follicles in the multiple sclerosis brain: what you find depends on how and where you look. Brain 2010;133:e157. [36] Lassmann H, Niedobitek G, Aloisi F, Middeldorp JM, NeuroproMiSe EBV Working Group. Epstein–Barr virus in the multiple sclerosis brain: a controversial issue— report on a focused workshop held in the Centre for Brain Research of the Medical University of Vienna, Austria. Brain 2011;134:2772–86. [37] Tzartos JS, Khan G, Vossenkamper A, Cruz-Sadaba M, Lonardi S, Sefia E, et al. Association of innate immune activation with latent Epstein–Barr virus in active MS lesions. Neurology 2012;78:15–23. [38] Jaquie´ry E, Jilek S, Schluep M, Meylan P, Lysandropoulos A, Pantaleo G, et al. Intrathecal immune responses to EBV in early MS. Eur J Immunol 2010;40:878–87 [Erratum in: Eur J Immunol 2011; 41:1501]. [39] Lossius A, Johansen JN, Vartdal F, Robins H, Ju¯ rate˙ Sˇaltyte˙ B, Holmøy T, et al. High-throughput sequencing of TCR repertoires in multiple sclerosis reveals intrathecal enrichment of EBV-reactive CD8(+) T cells. Eur J Immunol 2014, August [Epub ahead of print]. [40] Power C. Retroviral diseases of the nervous system: pathogenic host response or viral gene-mediated neurovirulence? Trends Neurosci 2001;24:162–9. [41] Perron H, Garson JA, Bedin F, Bese`me FG, Paranhos-Baccala G, Komurian- Pradel F, et al. Molecular identification of a novel retrovirus repeatedly isolated from patients with multiple sclerosis. Proc Natl Acad Sci U S A 1997;94: 7583–8. [42] Rieger F, Amouri R, Benjelloun N, Cifuentes-Diaz C, Dobransky T, Lyon-Caen O, et al. Gliotoxic factor and multiple sclerosis. C R Acad Sci 1996;319:343–50. [43] Mameli G, Serra C, Astone V, Castellazzi M, Poddighe L, Fainardi E, et al. Inhibition of multiple-sclerosis-associated retrovirus as biomarker of inter- feron therapy. J Neurovirol 2008;14:73–7. [44] Antony JM, van Marle G, Opii W, Butterfield DA, Mallet F, Yong VW, et al. Human endogenous retrovirus glycoprotein-mediated induction of redox reactants causes oligodendrocyte death and demyelination. Nat Neurosci 2004;7:1088–95. [45] Perron H, Germi R, Bernard C, Garcia-Montojo M, Deluen C, Farinelli L, et al. Human endogenous retrovirus type W envelope expression in blood and brain cells provides new insights into multiple sclerosis disease. Mult Scler 2012;18: 1721–36. [46] Isaacs A, Lindenmann J. Virus interference: I. The interferon. Proc R Soc Lond B: Biol Sci 1957;147:258–67. [47] Pantich HS, Hirsch RL, Schindler J, Johnson KP. Treatment of multiple sclerosis with gamma-interferons: exacerbations associated with activation on im- mune system. Neurology 1987;37:1097–102. [48] Pantich HS, Hirsch RL, Haley AS, Johnson KP. Exacerbations of multiple sclerosis in patients treated with gamma-interferon. Lancet 1987;1:893–5. [49] Knobler RL, Panitch HS, Braheny SL, Sipe JC, Rice GP, Huddlestone JR, et al. Systemic alpha-interferontherapy of multiple sclerosis. Neurology 1984;34: 1273–9. [50] Panitch HS. Systemic alpha-interferon in multiple sclerosis. Long-term patient follow-up. Arch Neurol 1987;44:61–3. [51] Camenga DL, Johnson KP, Alter M, Engelhardt CD, Fishman PS, Greenstein JI, et al. Systemic recombinant alpha-2 interferon therapy in relapsing multiple sclerosis. Arch Neurol 1986;43:1239–46. [52] Durelli L, Bongioanni MR, Cavallo R, Ferrero B, Ferri R, Ferrio MF, et al. Chronic systemic high-dose recombinant interferon alfa-2a reduces exacerbation rate. MRI signs of disease activity, and lymphocyte interferon gamma production in relapsing–remitting multiple sclerosis. Neurology 1994;44(3 Pt 1):406–13. [53] Durelli L, Bongioanni MR, Ferrero B, Ferri R, Imperiale D, Bradac GB, et al. Interferon alpha-2a treatment of relapsing–remitting multiple sclerosis: dis- ease activity resumes after stopping treatment. Neurology 1996;47:123–9. [54] European Study Group on Interferon Beta-1b in Secondary Progressive MS. Placebo-controlled multicentre randomised trial of interferon beta-1b in treatment of secondary progressive MS. Lancet 1998;352:1491–7. [55] The IFNB Multiple Sclerosis Study Group. Interferon b-1b is effective in relaps- ing–remitting multiple sclerosis: I. Clinical results of a multicenter, randomized, double-blind, placebo-controlled trial. Neurology 1993;43:655–61. [56] Paty DW, Li DK. Interferon beta lb is effective in relapsing–remitting multiple sclerosis: II. MRI analysis results of a multicenter, randomized, double-blind, placebo-controlled trial. Neurology 1993;43:662–7. [57] Rudick RA, Goodkin DE, Jacobs LD, Cookfair DL, Herndon RM, Richert JR, et al. Impact of interferon beta-1a on neurologic disability in relapsing multiple sclerosis. The Multiple Sclerosis Collaborative Research Group (MSCRG). Neu- rology 1997;49:358–63. [58] Simon JH, Jacobs LD, Campion M, Wende K, Simonian N, Cookfair DL, et al. Magnetic resonance studies of intramuscular interferon beta-1a for relapsing multiple sclerosis. The Multiple Sclerosis Collaborative Research Group. Ann Neurol 1998;43:79–87. [59] PRISMS (Prevention of Relapses Disability by Interferon beta-1a Subcutane- ously in Multiple Sclerosis) Study Group. Randomised double-blind placebo- controlled study of interferon beta-1a in relapsing/remitting multiple sclero- sis. Lancet 1998;352:1498–504. [60] Panitch H, Miller A, Paty D, Weinshenker B, North American Study Group on Interferon beta-1b in Secondary Progressive MS. Interferon beta-1b in sec- ondary progressive MS: results from a 3-year controlled study. Neurology 2004;63:1788–95. [61] Secondary Progressive Efficacy Clinical Trial of Recombinant Interferon-beta-1a in MS (SPECTRIMS) Study Group. Randomized controlled trial of interferon-beta- 1a in secondary progressive MS: clinical results. Neurology 2001;56:1496–504. [62] Li DK, Zhao GJ, Paty DW, University of British Columbia MS/MRI Analysis Research Group The SPECTRIMS Study Group. Randomized controlled trial of interferon-beta-1a in secondary progressive MS: MRI results. Neurology 2001;56:1505–13. [63] Cohen JA, Cutter GR, Fischer JS, Goodman AD, Heidenreich FR, Kooijmans MF, et al. Benefit of interferon beta-1a on MSFC progression in secondary progres- sive MS. Neurology 2002;59:679–87. [64] Jacobs LD, Beck RW, Simon JH, Kinkel RP, Brownscheidle CM, Murray TJ, et al. Intramuscular interferon-beta-1a therapy initiated during a first demyelinat- ing event in multiple sclerosis. N Engl J Med 2000;343:898–904. [65] Comi G, Filippi M, Barkhof F, Durelli L, Edan G, Fernandez O, et al. Effect of early interferon treatment on conversion to definite multiple sclerosis: a random- ized study. Lancet 2001;357:1576–82. [66] Kappos L, Polman CH, Freedman MS, Edan G, Hartung HP, Miller DH, et al. Treatment with interferon beta-1b delays conversion to clinically definite and McDonald MS in patients with clinically isolated syndromes. Neurology 2006;67:1242–9. V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228226
  • 7. [67] Panitch H, Goodin DS, Francis G, Chang P, Coyle PK, O’Connor P, et al. Randomized, comparative study of interferon b-1a treatment regimens in MS. The EVIDENCE Trial. Neurology 2002;59:1496–506. [68] Durelli L, Verdun E, Barbero P, Bergui M, Versino E, Ghezzi A, et al. Every-other- day interferon beta-1b versus once-weekly interferon beta-1a for multiple sclerosis: results of a 2-year prospective randomized multicentre study (INCOMIN). Lancet 2002;359:1453–60. [69] McGraw CA, Lublin FD. Interferon beta and glatiramer acetate therapy. Neu- rotherapeutics 2013;10:2–18. [70] Rudick RA, Lee JC, Simon J, Ransohoff RM, Fisher E. Defining interferon beta response status in multiple sclerosis patients. Ann Neurol 2004;56: 548–55. [71] Ross C, Clemmesen K, Svenson M, Sørensen PS, Koch-Henriksen N, Skovgaard GL, et al. Immunogenicity of interferon-beta in multiple sclerosis patients: influence of preparation, dosage, dose frequency, and route of administration. Ann Neurol 2000;48:706–12. [72] Calabresi PA, Kieseier BC, Arnold DL,BalcerLJ, Boyko A, Pelletier J, et al. Pegylated interferon b-1a for relapsing–remitting multiple sclerosis (ADVANCE): a ran- domised, phase 3, double-blind study. Lancet Neurol 2014;13:657–65. [73] Kappos L, Clanet M, Sandberg-Wollheim M, Radue EW, Hartung HP, Hohlfeld R, et al. Neutralizing antibodies and efficacy of interferon beta-1a: a 4-year controlled study. Neurology 2005;65:40–7. [74] Lycke J, Svennerholm B, Hjelmquisit E, Frise´n L, Badr G, Andersson M, et al. Acyclovir treatment of relapsing–remitting multiple sclerosis a randomized, placebo-controlled, double-blind study. J Neurol 1996;243:214–24. [75] Bech E, Lycke J, Gadeberg P, Hansen HJ, Malmestro¨m C, Andersen O, et al. A randomized, double-blind, placebo-controlled MRI study of anti-herpes virus therapy in MS. Neurology 2002;58:31–6. [76] Friedman JE, Zabriskie JB, Plank C, Ablashi D, Whitman J, Shahan B, et al. A randomized clinical trial of valacyclovir in multiple sclerosis. Mult Scler 2005;11:286–95. [77] Lycke J, Andersen O, Svennerholm B, Appelgren L, Dahllof C. Acyclovir con- centrations in serum and cerebrospinal fluid at steady state. J Antimicrob Chemother 1989;24:947–54. [78] Ivashkiv LB, Donlin LT. Regulation of type I interferon responses. Nat Rev Immunol 2014;14:36–49. [79] Killestein J, Polman CH. Determinants of interferon b efficacy in patients with multiple sclerosis. Nat Rev Neurol 2011;7(4):221–8. [80] Taniguchi T, Takaoka A. A weak signal for strong responses: interferon-alpha/ beta revisited. Nat Rev Mol Cell Biol 2001;2:378–86. [81] Li P, Wong JJ, Sum C, Sin WX, Ng KQ, Koh MB, et al. IRF8 and IRF3 cooperatively regulate rapid interferon-b induction in human blood monocytes. Blood 2011;117:2847–54. [82] Bentz GL, Liu R, Hahn AM, Shackelford J, Pagano JS. Epstein–Barr virus BRLF1 inhibits transcription of IRF3 and IRF7 and suppresses induction of interferon- beta. Virology 2010;402:121–8. [83] Hahn AM, Huye LE, Ning S, Webster-Cyriaque J, Pagano JS. Interferon regula- tory factor 7 is negatively regulated by the Epstein–Barr virus immediate-early gene, BZLF-1. J Virol 2005;79:10040–52. [84] Valente RM, Ehlers E, Xu D, Ahmad H, Steadman A, Blasnitz L, et al. Toll-like receptor 7 stimulates the expression of Epstein–Barr virus latent membrane protein 1. PLoS ONE 2012;7:e43317. [85] Ning S, Hahn AM, Huye LE, Pagano JS. Interferon regulatory factor 7 regulates expression of Epstein–Barr virus latent membrane protein 1: a regulatory circuit. J Virol 2003;77:9359–68. [86] Fathallah I, Parroche P, Gruffat H, Zannetti C, Johansson H, Yue J, et al. EBV latent membrane protein 1 is a negative regulator of TLR9. J Immunol 2010;185:6439–47. [87] Zhao B, Zou J, Wang H, Johannsen E, Peng CW, Quackenbush J, et al. Epstein– Barr virus exploits intrinsic B-lymphocyte transcription programs to achieve immortal cell growth. Proc Natl Acad Sci U S A 2011;108:14902–07. [88] Mechelli R, Manzari C, Policano C, Annese A, Picardi E, Umeton R, et al. Epstein- Barr virus genetic variants associated with multiple sclerosis. Neurology 2014. Manuscript submitted. [89] Comabella M, Kakalacheva K, Rı´o J, Mu¨ nz C, Montalban X, Lu¨nemann JD. EBV- specific immune responses in patients with multiple sclerosis responding to IFNb therapy. Mult Scler 2012;18:605–9. [90] Balashov KE, Aung LL, Vaknin-Dembinsky A, Dhib-Jalbut S, Weiner HL. Inter- feron-b inhibits toll-like receptor 9 processing in multiple sclerosis. Ann Neurol 2010;68:899–906. [91] Giacomini E, Severa M, Rizzo F, Mechelli R, Annibali V, Ristori G, et al. IFN-b therapy modulates B-cell and monocyte crosstalk via TLR7 in multiple sclero- sis patients. Eur J Immunol 2013;43:1963–72. [92] Serra C, Mameli G, Arru G, Sotgiu S, Rosati G, Dolei A. In vitro modulation of the multiple sclerosis (MS)-associated retrovirus by cytokines: implications for MS pathogenesis. J Neurovirol 2003;9:637–43. [93] Curtin F, Lang AB, Perron H, Laumonier M, Vidal V, Porchet HC, et al. GNbAC1, a humanized monoclonal antibody against the envelope protein of multiple sclerosis-associated endogenous retrovirus: a first-in-humans randomized clinical study. Clin Ther 2012;34:2268–78. [94] Dreyfus DH. Autoimmune disease: a role for new anti-viral therapies. Auto- immun Rev 2011;11:88–97. [95] Sorensen PS, Ross C, Clemmesen KM, Bendtzen K, Frederiksen JL, et al. Clinical importance of neutralising antibodies against interferon beta in patients with relapsing–remitting multiple sclerosis. Lancet 2003;362:1184–91. Viviana Annibali graduated in biological sciences from Roma Tre University of Rome (Italy) in 2001. In 2006 she obtained her PhD in cell sciences and technologies and after a postgraduate master degree in methodologies for research and development of new therapies from Sapienza University of Rome, where she recently earned a clinical pathology specialty. From 2002 she is a re- search scientist at the CENTERS, S. Andrea Hospital, Department of Neuroscience, Mental Health and Senso- ry Organs (NESMOS), Sapienza University of Rome. Dur- ing her research activities on neurodegenerative diseases she gained experience in gene expression anal- ysis at the RNA and protein level in peripheral T cell subsets. Dr. Annibali has also gained relevant experience in studying molecular pathways and gene function via genetic and pharmacological approaches. Most recently, she is investigating the involvement of B cells transcriptome dysfunctions in multiple sclerosis disease, with particular attention to the role of the cellular and extracellular microRNAs. She is a member of Italian Association of Neuroimmunol- ogy (AINI). Rosella Mechelli received her master degree in Biologi- cal Sciences in 2000, PhD in genetic and molecular biology in 2003 from Sapienza University of Rome where she also earned a postgraduate master degree in methodologies for the research and development of new therapies in 2007. During her PhD training she studied the structure of telomeric chromatin and its epigenetic modifications. From 2004 she is a research scientist at the CENTERS, S. Andrea Hospital, Depart- ment of Neuroscience, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome. Her research interests lie in etiopathogenesis of multiple sclerosis in monozygotic twins discordant for the dis- ease. Currently most of her studies are focused on the interaction between heritable and environmental risk factors, in particular on Epstein–Barr virus genetic variants and virus–host interactions. She is a member of Italian Association of Neuroim- munology (AINI). Silvia Romano obtained a doctor of medicine degree from Sapienza University of Rome in 2000 and a spe- cialization degree in Neurology at Sapienza University of Rome in 2006. She was also a Research Fellow at Clinical and Behavioral Neurology Laboratory, S. Lucia Foundation, Rome (2008–2009, project on neurocogni- tive pattern of multiple sclerosis patients). She obtained a PhD in Experimental Neurological Sciences in 2010 and then undertook post-doctoral work at the Center of Experimental Neurological Therapies (CEN- TERS), a Department Unit of S. Andrea Hospital, Sapienza University of Rome (2012–2013) working on demyelinating and hereditary neurodegenerative dis- ease. Her current position is Researcher of Neurology at Department of Neuros- ciences, Mental Health and Sensory Organs (NESMOS), at S. Andrea Hospital, Sapienza University of Rome. Her research activity is focused on (1) etiopathogen- esis, cognitive impairment and treatment of multiple sclerosis; (2) clinical features and treatment of patients with hereditary cerebellar ataxias and Huntington diseases. Maria Chiara Buscarinu graduated in medicine and surgery in 2006 and specialized in neurology in 2012 at the University of Sassari. From 2005 to 2011 she worked at the Neurological Clinic of Sassari. She moved to Rome and she began her PhD in experi- mental neurology at Sapienza University of Rome. She carries out ambulatory activity and research at the S. Andrea Hospital in Rome, with increased interest in multiple sclerosis and etiopathogenetic factors related to the disease. She is a member of the Italian Society of Neuroimmunology (AINI) and the Italian Society of Neurology (SIN). Arianna Fornasiero obtained a doctor of medicine de- gree from Sapienza University of Rome in 2004 and a Specialization degree in Neurology at Sapienza Univer- sity of Rome in 2009. From 2010 to 2014 she was research fellow in experimental neurology at Sapienza University of Rome. From 2009 to today she work at the Center of Experimental Neurological Therapies (CEN- TERS) a Department Unit of S. Andrea Hospital, Sapienza University of Rome working on demyelinating disease. Her research activity is focused on etiopathogenesis and treatment of multiple sclerosis. V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228 227
  • 8. Renato Umeton obtained his bachelor’s and master’s degrees in computer science from University of Calabria. He earned his PhD in mathematics and informatics focusing his research on optimization and ontology studies that found their application in solving problems in the area of medicine and biology. His experience included working at Microsoft and at Massachusetts Institute of Technology, as well as collaborating with other major institutions such as the Harvard Medical School and the University of Cambridge in the UK. He carried out his most recent Postdoc at Sapienza Univer- sity of Rome – S. Andrea Teaching Hospital, where he applied his informatics and bioinformatics skills to un- ravel the genetic and environmental components of the pathogenesis of multiple sclerosis and other neurology- related diseases. Vito AG Ricigliano obtained his medical degree at ‘‘Sapienza’’ University of Rome, Italy, in 2013. He is currently a MD at Center for Experimental Neurological Therapies (CENTERS), S. Andrea hospital, Rome, and in the present year he has been Academic visitor at the University of Oxford, UK, Nuffield Department of Clini- cal Neurosciences (NDCN). His research investigates the role of gene–environment interactions in the etiology of multiple sclerosis, especially focusing on the character- ization of the interplay between EBV and the host net- works, functional interpretation of GWAS data and use of next-generation techniques (e.g. RNA-sequencing, exome sequencing). Francesco Orzi has spent several years dedicated to lab research, in animal models of neurological diseases, and in exploiting methods for assessment of brain functional parameters. Following a 3 years stage (1979–1982) in the Lab of Dr. L. Sokoloff at NIH, in Bethesda, he became specifically interested in mapping local cerebral func- tional changes in animal models. A few studies have contributed to define the functional circuitry of the Basal Ganglia in relation to their role in movement disorders and in motivated behavior. Other studies, since the early experiences in the laboratory of Dr Klatzo at NIH, have been carried out in the field of the brain damage maturation following temporary brain ische- mia, and in the field of neuroprotection in animal models of brain ischemia. In the last 15 years he has been fully involved in clinical neurology. Fields of interest are cerebrovascular diseases and dementias. The focus is on mechanisms that underlie neuronal degeneration associated with energy defects, dysfunction of the neuro- muscular unit, and implications for neuroprotection. Eliana Marina Coccia is head of the Anti-Infectious Immunity Unit at the Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanita`, Rome-Italy. She received her Ph.D. in biological sciences in 1984 from University of Rome, working on the effect of type I IFN on the growth and differentiation of Friend erythroleukemia cells. From 1984 to 1985, she was a post-doctoral fellow at the Weizmann Institute of Sci- ence (Rehovot, Isreal) in Dr. Michel Revel’s group where she cloned the mouse 2-5A synthetase. In 1991–1992 she moved at the Pasteur Institute (Paris, France) in the laboratory of Dr. Ara Hovanessian to investigate the role of type I IFN on HIV replication. The group lead by E. Coccia is interested in understanding how type I IFN contribute to the induction of the immune response against several pathogens, such as HIV, Mycobacterium tuberculosis, Aspergillus fumigatus and Epstein–Barr virus. In particular the long-term objectives of her projects is to investigate IFN-driven immune-regulation and aberrant activation of IFN pathways in microbial infection and autoimmunity, with specific regard to B lymphocytes and primary dendritic cells. Marco Salvetti obtained his primary medical qualifica- tion in 1986 from the Sapienza University of Rome where he also trained as a clinical neurologist. He was a postdoctoral fellow at the Max Planck Society for Multiple Sclerosis in Prof. Hartmut Wekerle’s lab. Following studies on the fine specificity of the T cell response to putative autoantigens in multiple sclerosis, he instituted the world largest twin registry in 1997. From then on, epidemiological, gene expression and virological studies in twins with multiple sclerosis begun. At present much of the studies are focused on the interaction between heritable and environmental fac- tors in the etiology of multiple sclerosis. This informa- tion, combined with data from the in vitro screening of off-label activities of registered drugs (on oligodendrocyte precursors), is exploited for the design of exploratory clinical trials. These studies are carried out in the context of the clinical research activity of CENTERS, an institution devoted to nonprofit, phase II trials in multiple sclerosis and orphan neurological diseases. Giovanni Ristori obtained in 1985 the degree in medi- cine, at ‘Universita` Cattolica del Sacro Cuore’, Rome, Italy and in 1989 the specialization in neurology at the same University. In 1995 he obtained a PhD in Neuroscience at Sapienza University of Rome, Italy. The present position is at Neuroimmunology laboratory and Neurogenetic Unit, Faculty of Medicine and Psy- chology, Sapienza University of Rome, Italy. V. Annibali et al. / Cytokine & Growth Factor Reviews 26 (2015) 221–228228