ArticlePDF Available

Advances Toward a Norovirus Antiviral: From Classical Inhibitors to Lethal Mutagenesis

Authors:

Abstract and Figures

Human noroviruses are a leading cause of gastroenteritis worldwide, yet there are no licensed antivirals. There is an urgent need for norovirus therapeutics, particularly for chronic infections in immunocompromised individuals, but also a potential need for prophylactic use in epidemics. Continued research has led to the identification of compounds that inhibit norovirus replication in vitro and, at least in some cases, are also effective in vivo against murine norovirus. Progress has included classical approaches targeting viral proteins and harnessing the antiviral action of interferon, strategies targeting essential host cell factors, and novel strategies exploiting the high mutation rate of noroviruses.
Content may be subject to copyright.
The Journal of Infectious Diseases
SUPPLEMENT ARTICLE
Advances Toward a Norovirus Antiviral: From Classical
Inhibitors to Lethal Mutagenesis
Lucy Thorne,
a
Armando Arias,
a
and Ian Goodfellow
Division of Virology, Department of Pathology, University of Cambridge, Addenbrookes Hospital, United Kingdom
Human noroviruses are a leadi ng cause of gastroenteritis worldwide, yet there are no licensed antivirals. There is an urgent need for
norovirus therapeutics, particularly for chronic infections in immunocompromised individuals, but also a potential need for pro-
phylactic use in epidemics. Continued research has led to the identication of compounds that inhibit norovirus replication in vitro
and, at least in some cases, are also effective in vivo against murine norovirus. Progress has included classical approaches targeting
viral proteins and harnessing the antiviral action of interferon, strategies targeting essential host cell factors, and novel strategies
exploiting the high mutation rate of noroviruses.
Keywords. human norovirus; antivirals; protease; polymerase; interferon λ; favipiravir; lethal mutagenesis.
Human noroviruses (HuNoVs) are a major cause of viral gas-
troenteritis worldwide, and since introduction of the rotavirus
vaccine they have become the leading cause of severe pediatric
gast roenterit is [1]. HuNoVs present a particular problem in
healthcare settings, with close to 4000 hospital outbreaks re-
ported in a 2-year period in the United Kingdom, causing ap-
proximately 9000 days of ward closures, disrupted services, and
a signicant econo mic cost for the UK National Health Service
[2]. Most infections are acute with a low risk of mortality, but
HuNoV is recognized as a signicant risk factor for compli-
cations and increased mortality in immunocompromised indi-
viduals, including transplant recipients and those receiving
immunosuppressive therapies [3]. In man y cases, HuNoV in-
fections in these cohorts can persist for years [4]. Conservative
estimates of mortality rates in develo ping cou ntries are much
higher ; at least 200 000 deat hs per year in children aged <5
years are attributed to HuNoVs [5].
Despite an urgent need, there are currently no licensed anti-
virals or vaccines for HuNoVs. Groups that would most benet
include high-risk populations, such as young and elderly indi-
viduals, chronically infected patients, and medical staff. Health-
care and military settings could also benet from prophylactic
use of antivi rals to potentially contain epidemics. However, the
development of therapeutics has been hindered by the lack of an
efcient cell culture system for HuNoV, which has also slowed
elucidation of the molecular details of noroviru s replication and
viral protein functions, key knowledge for the rational develop-
ment of specic antivirals. A replicon system, in which cells har-
bor self-replicating RNA of the prototype HuNoV, namely
Norwalk virus, has provided an invaluab le tool to evaluate an-
tivirals in vitro [6]. In the past few years, however, there has been
signicant progress in the eld, with the establishment of a plas-
mid-based reverse genetics s y s tem, to allow production of genet-
ically dened HuNoVs, and the rst demonstration of HuNoV
replication in vitro in a cultured B-cell line and in vivo in immu-
nodecient mice, albeit with limited replication in both [79].
These developments may now provide systems to evaluate poten-
tial antivirals, but their usefulness will critically depend on being
robust and reproducible in other laboratories.
Murine norovirus (MNV), which replicates in cultured mac-
rophage and dendritic cells, has been used as a surrogate model
for HuNoV since its discovery in 2003, owing to the availability
of reverse genetics systems and small-animal models. Both
HuNoV and MNV belong to the Caliciviridae family of small,
positive-sense, single-stranded RNA viruses and have similar
genome organization, protein functions, and conserved molec-
ular me chanisms of genome replication and translation. By use
of a combination of MNV and the HuNoV replicon, research
efforts into antivirals have intensied in the last 10 years [6,
10]. This review will focus on the latest developments, which
can be divided into categories of classical antiviral approaches
that target viral proteins or harness the antiviral effects of inter-
feron (IFN), alternative strategies targeting ho st cell processes,
and strategies that exploit the high mutation rate of noroviruses
by lethal mutagenesis.
DEVELOPMENTS IN CLASSICAL ANTIVIRAL
APPROACHES: RENEWE D INTEREST IN IFN AND THE
TARGETING OF VIRAL PROTEINS
Type I and type II IFNs elicit effective antiviral responses
against HuNoV and MNV, emphasizing the critical role of
a
L. T. and A. A. contributed equally to this work.
Correspondence: L. Thorne, Division of Virology, Department of Pathology, University of Cam-
bridge, Addenbrookes Hospital, Hills Rd, Cambridge, CB2 2QQ, UK (lt375@cam.ac.uk).
The Journal of Infectious Diseases
®
2016;213(S1):S2731
© The Author 2016. Published by Oxford University Press for the Infectious Diseases Society of
America. This is an Open Access article distributed under the terms of the Creative Commons
Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted
reuse, distribution, and reproduction in any medium, provided the original work is properly
cited. DOI: 10.1093/infdis/jiv280
Advances Toward Norovirus Antivirals
JID 2016:213 (Suppl 1)
S27
by guest on January 14, 2016http://jid.oxfordjournals.org/Downloaded from
innate immunity in controlling norovirus infections [6, 11]. De-
spite this, clinical use of IFN against HuNoVs has not been de-
scribed. Interest in IFN a s a HuNoV therapeutic has recently
been renewed, owing to a study that found that type III IFN,
IFN-λ, is required to control persistent MNV infecti ons [11].
Treatment with IFN-λ cleared persistent infections in mice
without requiring an adaptive immune response, revealing the
potential of IFN-λ as a treatment for chronic infections in the
immunocompromised.
A classical antiviral strategy is to target e ssential viral pro-
teins, and for HuNoV the viral protease (NS6
pro
) has become
the most widely studied antiviral target [10]. NS6
pro
is a chymo-
trypsin-like cysteine protease responsible for cleavage of the
viral polyprotein to release mature forms of the essential non-
structural replicase proteins, including itself (Figure 1). Resolu-
tion of the NS6
pro
crystal structure in 2006 has since facilitated
structure-guided design of a variety of inhibitors aimed to
mimic natural substrate recognition and react irreversibly
with active site residues. In th e past 2 years, t here has been a
signicant increase in the number of norovirus protease inhib-
itors that exhibit a range of potencies in in vitro enzymatic as-
says and cell-based assays [10]. NS6
pro
shares similarities with
the picornavirus protease (3C
pro
), and some compounds effec-
tive against 3C
pro
exert broad reactivity against NS6
pro
.Most
recently, rupintrivir, originally developed against the rhinovirus
3C
pro
, was found to clear cells of HuNoV replicon RNA and in-
hibit MNV replication in vitro. In enzymatic assays, rupintrivir
inhibited the NS6
pro
of the predominant circulating HuNoV ge-
notype GII.4, suggesting that it may target clinically relevant
strains [12].
Given their essential role in replicating the viral genome, viral
RNAdependent R NA polymerases (RdRps) also present at-
tractive antiviral targets. Polymerase inhibitors have been clin-
ically approved for many RNA viruses, whose RdRps share
conserved structural and functional properties with the norovi-
rus RdRp [13]. RdRp inhibitors can be divided into nucleoside
analogues, which target the active site, and non-nucleoside an-
alogues. The nucleoside analogue 2-C -methylcytidine
(2CMC), originally developed for use against hepatitis C virus
(HCV), inhibits HuNoV replication in vitro, and treatment with
2CMC cleared cells of replicon RNA. 2CMC was also effective
against MNV in vitro and in vivo, in which treatment prevented
diarrhea and mortality in a lethal mouse model of infection [6,
10]. The same authors recently demonstrated that treatme nt
with 2CMC reduced shedding and transmission of MNV and
that prophylactic treat ment completely protected against
MNV infection [14]. While these results are highly promising,
2CMC has not yet been approved for treatment of HCV
Figure 1. Organization of the human norovirus genome and the main antiviral targets. The genome is covalently attached at the 5 end to VPg and is polyadenylated at the 3
end. RNA structures are present at either end of the genome, which interact with host cell factors (shaded shapes) to achieve replication and translation. Essential host cell
factors represent potential antiviral targets for small-molecule inhibitors. The viral genome is divided into 3 open reading frames (ORFs). ORF1 encodes the viral polyprotein,
which is cotranslationally and posttranslationally cleaved by the viral protease, NS6
pro
, to release mature nonstructural proteins, including the viral RNAdependent RNA
polymerase (RdR p). Names given in brackets for the nonstructural proteins represent the alternative nom enclature used for murine norovirus. Both NS6
pro
and the RdRp
are key viral protein targets for a number of inhibitors as shown. ORF2 and ORF3 are translated from the subgenomic RNA and encode the major and minor capsid proteins
respectively. The antiviral effects of type I and II interferons (IFNs) are thought in part to be mediated at the level of translation, although the mechanism of action of IFN-λ has
not yet been determined. Abbreviation: 2CMC, 2-C-methylcytidine.
S28
JID 2016:213 (Suppl 1)
Thorne et al
by guest on January 14, 2016http://jid.oxfordjournals.org/Downloaded from
infection because of concerns regarding its toxicity prole in pa-
tients, although derivatives of 2CMC are currently under devel-
opment and could prove of potential therapeutic use.
Several non-nucleoside inhibitors have been identied with
activi ty against the HuNoV RdRp in enzymatic assays. These
include suramin, NF203, and PPNDS [10]. Crystallographic
studies have revealed the binding site of each compound on
the RdRp, but they have yet to be tested in the cell culture or
animal models of norovirus infections. Recently, a high-
throughput screen against HuNoV GII.4 RdRp activity identi-
ed several compounds that also in hibi ted the Norwalk virus
replicon and MNV replication in vitro, although at much higher
concentrations. These compounds now provide a sc aff old for
rational design and optimization of specic HuNoV RdRp in-
hibitors [15].
TARGETING THE HOST CELL AS AN ANTIVIR AL
APPROACH
Owing to the high error rates of RNA virus replication, norovi-
rus included, the efcacy of antivirals targeting viral proteins is
often limited by the emergence of drug-resistance mutations.
An alternative strategy that circumvents this problem is to target
host cell proteins that are essential for viral replication, provid-
ing a much higher barrier to the generation of resistant mutants.
A number of host cell factors have been identied that interact
with the viral genome and are required for either its replication
or translation; these include La, PTB, DDX3, PCPB2, and
hnRNPs [6]. RNAi-mediated knockdown of these factors re-
duced MNV viral yields in vitro, demonstrating the potential
of targeting host cell proteins to restrict replication. Modulation
of entire cellular processes has also been shown as an antiviral
approach [16]. WP1130, a small-molecule inhibitor of cellular
deubiquitinases, inhibits replication of MNV and several
other RNA viruses, with this activity indirectly mediated by ac-
tivation of the unfolded protein response (UPR). Inhibition of a
specic cellular deubiquitinase (UPS14) resulted in the activa-
tion of inositol-requiring enzyme, a key mediator of the UPR, in
which endoplasmic reticulumassociated protein degradation is
increased and cellular translation decreased. Small-molecule ac-
tivators of the UPR also reduced MNV replication. Derivatives
of WP1130 have recently been identied that have enhanced
broad antiviral activity, without cellular toxicity, although
they have yet to be evaluated in vivo [16].
LETHAL MUTAGENESIS OF NOROVIRUSES
Lethal mutagenesis has recently emerged as a novel alternative
strategy to classical antiviral approaches. Several nucleoside an-
alogues, such as r ibavirin and favipiravir, display antiviral
Figure 2. Lethal mutagenesis as an antiviral strategy to control norovirus. Top, During multiple rounds of virus infection in host cells, diverse virus populations (known as
quasispecies) are formed as a result of low viral RNAdependent RNA polymerase replication fidelity. Genetic diversity is represented as different virus particles containing
different shape and color symbols. Some viruses will contain lethal mutations and are naturally non-viable, represented with a cross. Genetic diversity enables RNA viruses to
have rapid adaptability to the environment and the flexibility to escape from immune responses and antiviral compounds. Bottom, Lethal mutagenesis exploits low replication
fidelity to drive RNA viruses to extinction through an excessive accumulation of mutations. A mutagenic compound (black star) interferes with virus replication, leading to larger
mutation frequencies. As a result of increased error rates during lethal mutagenesis, a larger proportion of viruses will contain lethal mutations. Continuous replication in the
presence of an efficient mutagen can result in complete loss of viral infectivity (virus extinction).
Advances Toward Norovirus Antivirals
JID 2016:213 (Suppl 1)
S29
by guest on January 14, 2016http://jid.oxfordjournals.org/Downloaded from
activities associated with induced mutagenesis in vitro [17, 18].
Excessive accumulation of mutations during replication, beyond
a tolerated value known as the error threshold, can lead to virus
extinction, a proce ss known as lethal m utagenesis (Figure 2).
The in vitro evidence has led to several studies that assessed
the clinical use of lethal mutagenesis. Recent reports on HCV
have suggested that the antiviral activity of ribavirin in vivo
may be a ssociated with increased mutagenesis, although this
is still a controversial topic [18].
With the aim of assessing antiviral therapies based on lethal
mutagenesis, we have recently investigated whether ribavirin
and favipiravir are also mutagenic for noroviruses [ 17]. We
found that treatment of MNV-infected mice with favipiravir re-
sulted in more-rapid clearance rates of infectious virus. Viral
populat ions from treated mice showed highe r mutation fre-
quencies and lower replication tness than viruses isolated
from control mice, suggesting that favipiravir can drive persis-
tent MNV infections to extinction through the accumulation of
debilitating mutations. We also showed that the antiviral activ-
ity of favipiravir is sustained during prolonged treatment peri-
ods (>50 days) without any observed rebound in viral titers. Our
data also sugg ested that favipiravir is more efcient than ribavi-
rin in the control of MNV infections in vivo, possibly because
favipiravir induces more-effective increases in mutat ion fre-
q uency. Ribavirin and favipiravir are purine anal ogues that
can be triphosphorylated within the cell and then incorporated
into nascent viral genomes by the viral RdRp. Their mutagenic
activity resides in their ambiguous pairing behavior, establish-
ing stable base pairing with both uridine and cytidine. This am-
biguity can occur as they are incorporated into the nascent viral
RNA and in the next round of replication acting in the template
strand, resulting altogether in an increase in the number of tran-
sition mutations.
The main advantage of lethal mutagenesis relative to other
antiviral strategies is that it causes a continuous attenuation of
the replicating virus, leading to clearance of infection. In the
absence of the drug, attenuating deleterious mutations may re-
main imprinted in the viral population and in any transmitted
viru s. In addition, it has been demonstrated that signicantly
attenuated and defective variants generated during mutagenesis
can interfere with infection by replication-competent viruses,
thereby facilitating viral extinction [18]. These results raise the
possibility of using favipiravir and lethal mutagenesis as an an-
tiviral strategy to treat chronic HuNoV infections in immuno-
com promised individuals. A recent study has indicated that
ribavirin may be effective in some chronically infected patients,
although whether the antiviral activity observed was due to mu-
tagenesis was not examined [19]. Clearly, further studies in this
area are warranted.
Recent studies encourage the development of new combina-
tional approaches involving a mutagenic compound and a clas-
sical inhibitor, to improve the efcacy of therapies based on
lethal mutagenesis [18]. A possible limitation of such cocktails
may be the greater likelihood of resistance to classical inhibitors
as a consequence of the increased mutation frequencies. Al-
ternative combinati onal approaches may involve sequential
administration of inhibitor and mutagen to reduce the possibil-
ity of resistance emerging, a regimen recently shown to be more
efcient than simultaneous administration [18]. Combinations
of mutag ens and inhi bitors targeting critical host factors may
also present a rational approach, with a lower risk of generating
viral resistance.
SUMMARY
In recent years, there has been signicant progress in the devel-
opment of small-molecule inhibitors for HuNoV (Table 1),
driven by different approaches and accompanied by increased
understanding of the norovirus life cycle and development of
new systems for studying norovirus replication. Importantly,
however, further studies will be required to bring any of these
potential compounds through clinical trials and into the clinic.
To date, the only reported preliminary clinical trial tested nita-
zoxanide, a compound licensed against certain parasites, which
produced a modest decrease in symptom duration in healthy in-
dividuals but, more imp ortantly, resolved symptoms within a
single chronically infected patient [10]. However , no furth er
use has been reported since 2011. Evaluating inhibitors that
are currently in trials or under development for other viruses
with conserved protein functions, including 2CMC derivatives
and favipiravir, may serve to accelerate treatments for HuNoV
through clinical trials.
Su pplementary Data
Supplementary materials are available at http://jid.oxfordjournals.org.
Consisting of data provided by the author to benet the reader, the posted
materials are not copyedited and are the sole responsibility of the author, so
questions or comments should be addressed to the author.
Table 1. Summary of Potential Norovirus Antivirals
Compound Target Mechanism of Action
Classical inhibitor
IFN-λ Host cells Induction of antiviral state, specific
mediators unknown [11]
Rupintrivir Viral protease Irreversible inhibitor of active site [12]
2CMC Viral polymerase Nucleoside analogue [16]
Suramin Viral polymerase Nonnucleoside analogue [10]
NF203 Viral polymerase Nonnucleoside analogue [10]
PPNDS Viral polymerase Nonnucleoside analogue [10]
Host cell inhibitor
WP1130 Cellular
deubiquitinases
Indirect activation of the unfolded
protein response [16]
Chemical mutagen
Favipiravir Viral polymerase Lethal mutagenesis [17]
Ribavirin Viral polymerase Lethal mutagenesis [17]
Abbreviations: 2CMC, 2-C-methylcytidine; IFN-λ, interferon λ.
S30
JID 2016:213 (Suppl 1)
Thorne et al
by guest on January 14, 2016http://jid.oxfordjournals.org/Downloaded from
Notes
Financial support. This work was supported by the Wellcome Trust
(reference WT097997MA; senior fellowship to I. G.).
Potential conicts of interest. All authors: No reported co nicts. All
authors have submitted the ICMJE Form for Disclosure of Potential Con-
icts of Interest. Conicts that the editors consider relevant to the content
of the manuscript have been disclosed.
References
1. Payne DC, Vinje J, Szilagyi PG, et al. Norovirus and medically attended gastroen-
teritis in U.S. children. N Engl J Med 2013; 368:112130.
2. Harris JP, Adams NL, Lopman BA, Allen DJ, Adak GK. The development of web-
based surveillance provides new insights into the burden of norovirus outbreaks in
hospitals in England. Epidemiol Infect 2014; 142:15908.
3. Ronchetti AM, H enry B, Amber t-Balay K, et al. Norovirus-related chronic
diarrhea in a patient treated with alemtuzumab for chronic lymphocytic leukemia.
BMC Infect Dis 2014; 14:239.
4. Woodward JM, Gkrania-Klotsas E, Cordero-Ng AY, et al. The role of ch ronic
norovirus infection in the enteropathy associated with common variable immuno-
deciency. Am J Gastroenterol 2015; 110:3207.
5. Patel MM, Widdowson M-A, Glass RI, Akazawa K, Vinjé J, Parashar UD. System-
atic literature review of role of noroviruses in sporadic gastroenteritis. Emerg Infect
Dis 2008; 14:122431.
6. Arias A, Emmott E, Vashist S, Goodfellow I. Progress towards the prevention and
treatment of norovirus infections. Future Microbiol 2013; 8:1475 87.
7. Jones MK, Watanabe M, Zhu S, et al. Enteric bacteria promote human and mouse
norovirus infection of B cells. Science 2014; 346:7559.
8. Taube S, Kolawole AO, Hohne M, et al. A mouse model for human norovirus.
MBio 2013; 4:00450-13.
9. Katayama K, Murakami K, Sharp TM, et al. Plasmid-based human norovirus
reverse genetics system produces reporter-tagged progeny virus containing infec-
tious genomic RNA. Proc Natl Acad Sci U S A 2014; 111:5.
10. Kaufman SS, Green KY, Korba BE. Treatment of norovir us infections: moving
antivirals from the bench to the bedside. Antiviral Res 2014; 105:8091.
11. Nice TJ, Baldridge MT, McCune BT, et al. Interferon lambda cures persi stent
murine norovirus infection in the absence of adaptive immunity. Science 2014;
27:1258100.
12. Rocha-Pereira J, Nascimento MS, Ma Q, Hilgenfeld R, Neyts J, Jochmans D. The
enterovirus protease inhibitor rupintrivir exerts cross-genotypic anti-norovirus ac-
tivity and clears cells from the norovirus replicon. Antimicrob Agents Chemother
2014; 58:467581.
13. Ferrer-Orta C, Arias A, Escarmís C, Verdaguer N. A comparison of viral RNA-de-
pendent RNA polymerases. Curr Opin Struct Biol 2006; 16:2734.
14. Rocha-Pereira J, Jochmans D, Neyts J. Prophylactic treatment with the nucleoside
analogue 2-C-methylcytidine completely prevents transmission of norovirus.
J Antimicrob Chemother 2015; 70:1907.
15. Eltahla AA, Lim KL, Eden JS, Kelly AG, Mackenzie JM, White PA. Nonnucleoside
inhibitors of norovirus RNA polymerase: scaffolds for rational drug design. Anti-
microb Agents Chemother 2014; 58:311523.
16. Gonzalez-Hernandez MJ, Pal A, Gyan KE, et al. Chemical derivatives of a small
molecule deubiquitinase inhibitor have antiviral activity against several RNA vi-
ruses. PLoS One 2014; 9:e94491.
17. Arias A, Thorne L, Goodfellow I. Favipiravir elicits antiviral mutagenesis during
virus replication in vivo. Elife 2014; 3:e03679.
18. Domingo E, Sheldon J, Perales C. Viral quasispecies evolution. Microbiol Mol Biol
Rev 2012; 76:159216.
19. Woodward JM, Gkrania-Klotsas E, Cordero -Ng AY, et al. The role of chro nic
norovirus infection in the enteropathy associated with common variable immuno-
deciency. Am J Gastroenterol 2015; 110:3207.
Advances Toward Norovirus Antivirals
JID 2016:213 (Suppl 1)
S31
by guest on January 14, 2016http://jid.oxfordjournals.org/Downloaded from
... 80 Murine norovirus in vitro cell cultures have been useful surrogates, and a norovirus replicon system, a cell line containing the human norovirus genome, has been a tool that facilitated evaluation of antiviral targets. 80,81 Current norovirus life cycle targets under investigation are protease inhibitors, polymerase inhibitors, and non-nucleoside inhibitors. 81 Targeting host cell proteins required for viral replication is an attractive option as norovirus, an RNA virus, is prone to replication errors resulting in potential antiviral drug resistance. ...
... 80,81 Current norovirus life cycle targets under investigation are protease inhibitors, polymerase inhibitors, and non-nucleoside inhibitors. 81 Targeting host cell proteins required for viral replication is an attractive option as norovirus, an RNA virus, is prone to replication errors resulting in potential antiviral drug resistance. ...
Article
Full-text available
Norovirus, a leading cause of gastroenteritis outbreaks worldwide, results in substantial direct and indirect healthcare costs. Adults older than 65 years of age bear a significant proportion of the disease burden, and the disease course in this population is often more severe and protracted. In this narrative review, we discuss the epidemiology of norovirus infection, mechanisms of pathogenesis, and transmission pertinent to outbreaks along with infection prevention and control efforts. We also describe the clinical manifestations of norovirus disease with a focus on individuals older than 65 years of age, diagnosis and available treatment options, and the challenges and progress within vaccine development.
... Host factor drugs with the potential to treat norovirus infections include immunomodulators (type I, II, and III interferons) [300][301][302] and small molecule inhibitors that downregulate viral RNA secondary structure-binding host factors [284]. The thiazolide nitazoxanide, tested with varying outcomes in preliminary clinical trials against chronic HuNoV infections [303,304], may activate natural host antiviral defences but may also act through the direct inhibition of virus protein production, maturation, and/or assembly. A comprehensive review of progress made in the development of antivirals (this between 2010 and 2018) was provided in 2018 by Netzler et al. [283]. ...
Article
Full-text available
Human noroviruses are recognised as the major global cause of viral gastroenteritis. Here, we provide an overview of notable advances in norovirus research and provide a short recap of the novel model systems to which much of the recent progress is owed. Significant advances include an updated classification system, the description of alternative virus-like protein morphologies and capsid dynamics, and the further elucidation of the functions and roles of various viral proteins. Important milestones include new insights into cell tropism, host and microbial attachment factors and receptors, interactions with the cellular translational apparatus, and viral egress from cells. No-roviruses have been detected in previously unrecognised hosts and detection itself is facilitated by improved analytical techniques. New potential transmission routes and/or viral reservoirs have been proposed. Recent in vivo and in vitro findings have added to the understanding of host immunity in response to norovirus infection, and vaccine development has progressed to preclinical and even clinical trial testing. Ongoing development of therapeutics includes promising direct-acting small molecules and host-factor drugs.
... Viruses, being obligate parasites, are dependent on the living host cell for their replication (Samji, 2009). The discovery of host factors necessary for virus replication opened broad possibilities for the development of next generation antiviral drugs (Thorne et al., 2016). Instead of targeting viral proteins, this novel pharmacological strategy aims to target essential host proteins and pathways used by the IAVs to fulfill their replicative cycle (Qian et al., 2016). ...
Article
Full-text available
Influenza A virus (IAV) poses a major threat to global public health and is known to employ various strategies to usurp the host machinery for survival. Due to its fast-evolving nature, IAVs tend to escape the effect of available drugs and vaccines thus, prompting the development of novel antiviral strategies. High-throughput mass spectrometric screen of host-IAV interacting partners revealed host Filamin A (FLNA), an actin-binding protein involved in regulating multiple signaling pathways, as an interaction partner of IAV nucleoprotein (NP). In this study, we found that the IAV NP interrupts host FLNA-TRAF2 interaction by interacting with FLNA thus, resulting in increased levels of free, displaced TRAF2 molecules available for TRAF2-ASK1 mediated JNK pathway activation, a pathway critical to maintaining efficient viral replication. In addition, siRNA-mediated FLNA silencing was found to promote IAV replication (87% increase) while FLNA-overexpression impaired IAV replication (65% decrease). IAV NP was observed to be a crucial viral factor required to attain FLNA mRNA and protein attenuation post-IAV infection for efficient viral replication. Our results reveal FLNA to be a host factor with antiviral potential hitherto unknown to be involved in the IAV replication cycle thus, opening new possibilities of FLNA-NP interaction as a candidate anti-influenza drug development target.
... We have previously examined the efficacy of these drugs against USUV during lytic infections, and established an association between the antiviral activities observed and increased virus mutation frequencies [45]. The use of nucleoside drugs eliciting increased virus mutation rates is currently being investigated as an alternative antiviral strategy known as lethal mutagenesis [52,66,67]. An advantage of therapies based on mutagenic nucleosides, such as those used in this study, is that the antiviral effect associated with mutagenesis (i.e., detrimental mutations) is accumulated during viral RNA synthesis, and thus can be propagated to offspring genomes. ...
Article
Full-text available
Chronic viral disease constitutes a major global health problem, with several hundred million people affected and an associated elevated number of deaths. An increasing number of disorders caused by human flaviviruses are related to their capacity to establish a persistent infection. Here we show that Usutu virus (USUV), an emerging zoonotic flavivirus linked to sporadic neurologic disease in humans, can establish a persistent infection in cell culture. Two independent lineages of Vero cells surviving USUV lytic infection were cultured over 82 days (41 cell transfers) without any apparent cytopathology crisis associated. We found elevated titers in the supernatant of these cells, with modest fluctuations during passages but no overall tendency towards increased or decreased infectivity. In addition to full-length genomes, viral RNA isolated from these cells at passage 40 revealed the presence of defective genomes, containing different deletions at the 5’ end. These truncated transcripts were all predicted to encode shorter polyprotein products lacking membrane and envelope structural proteins, and most of non-structural protein 1. Treatment with different broad-range antiviral nucleosides revealed that USUV is sensitive to these compounds in the context of a persistent infection, in agreement with previous observations during lytic infections. The exposure of infected cells to prolonged treatment (10 days) with favipiravir and/or ribavirin resulted in the complete clearance of infectivity in the cellular supernatants (decrease of ~5 log10 in virus titers and RNA levels), although modest changes in intracellular viral RNA levels were recorded (<2 log10 decrease). Drug withdrawal after treatment day 10 resulted in a relapse in virus titers. These results encourage the use of persistently-infected cultures as a surrogate system in the identification of improved antivirals against flaviviral chronic disease.
... The antiviral properties of interferon can be used against norovirus [46]. Novel strategies include targeting essential host cell factors, or exploiting the high mutation rate of noroviruses (ribavirin and favipiravir seem to possess lethal mutagenesis properties) [104][105][106]. The great genetic variability of these viruses, due to the imprecision of the viral polymerase during replication and the potential for strains to recombine, is a barrier to the development of potent drugs against norovirus and sapovirus. ...
... While a number of other norovirus antiviral reports have been published in the last three years, [71][72][73][74][75] there has been significant recent progress in the field, which now warrants an updated review. Herein we include a comprehensive overview of peer-reviewed studies since 2010, including antiviral candidates examined in at least two different systems (e.g. ...
Article
Human noroviruses inflict a significant health burden on society and are responsible for approximately 699 million infections and over 200 000 estimated deaths worldwide each year. Yet despite significant research efforts, approved vaccines or antivirals to combat this pathogen are still lacking. Safe and effective antivirals are not available, particularly for chronically infected immunocompromised individuals, and for prophylactic applications to protect high‐risk and vulnerable populations in outbreak settings. Since the discovery of human norovirus in 1972, the lack of a cell culture system has hindered biological research and antiviral studies for many years. Recent breakthroughs in culturing human norovirus have been encouraging, however, further development and optimization of these novel methodologies are required to facilitate more robust replication levels, that will enable reliable serological and replication studies, as well as advances in antiviral development. In the last few years, considerable progress has been made toward the development of norovirus antivirals, inviting an updated review. This review focuses on potential therapeutics that have been reported since 2010, which were examined across at least two model systems used for studying human norovirus or its enzymes. In addition, we have placed emphasis on antiviral compounds with a defined chemical structure. We include a comprehensive outline of direct‐acting antivirals and offer a discussion of host‐modulating compounds, a rapidly expanding and promising area of antiviral research.
... The development of antivirals for the treatment of norovirus infections consists predominantly of DAAs that target the viral polymerase and the protease (30). In addition, there is an increase in research toward targeting features of the host to limit norovirus replication (31)(32)(33)(34); however, the antiviral effects of TLR7 agonists against norovirus have not been reported. ...
Article
Full-text available
Norovirus infections are a significant health and economic burden globally, accounting for hundreds of millions of cases of acute gastroenteritis every year. In the absence of an approved norovirus vaccine, there is an urgent need to develop antivirals to treat chronic infections, and provide prophylactic therapy to limit viral spread during epidemics and pandemics. Toll-like receptor (TLR) agonists have been explored widely for their antiviral potential and several are progressing through clinical trials for the treatment of human immunodeficiency virus (HIV), hepatitis B virus (HBV) and as adjuvants for norovirus virus-like particle (VLP) vaccines. However, developing norovirus therapies are largely direct-acting antivirals (DAAs) with fewer compounds that target the host. Our aim was to assess the antiviral potential of TLR7 agonist immunomodulators on norovirus infection using the murine norovirus (MNV) and human Norwalk replicon models. TLR7 agonists R-848, Gardiquimod, GS-9620, R-837 and Loxoribine were screened using a plaque reduction assay and each displayed inhibition of MNV replication (EC 50 values: 23.5 nM, 134.4 nM, 0.59 μM, 1.5 μM and 79.4 μM, respectively). RNA-sequencing of TLR7 stimulated cells revealed a predominant upregulation of innate immune response genes and ISGs that are known to drive an antiviral state. Furthermore, the combination of R-848 and the nucleoside analogue (NA) 2’ C-methylcytidine elicited a synergistic antiviral effect against MNV demonstrating that combinational therapy of host-modulators and DAA's could be used to reduce drug cytotoxicity. In summary, we have identified that TLR7 agonists display potent inhibition of norovirus replication and are a therapeutic option to combat norovirus infections.
Article
Human norovirus (HuNoV) causes acute viral gastroenteritis in all age groups, and dehydration and severe diarrhea in the elderly. The World Health Organization reports ∼1.45 million deaths from acute gastroenteritis annually in the world. Rupintrivir, an inhibitory medicine against the human rhinovirus C3 protease, has been reported to inhibit HuNoV 3C protease. However, several HuNoV 3C protease mutations have been revealed to reduce the susceptibility of HuNoV to rupintrivir. The structural details behind rupintrivir-resistance of these single-point mutations (A105V and I109V) are not still clear. Hence, in this study, a combination of computational techniques were used to determine the rupintrivir-resistance mechanism and to propose an inhibitor against wild-type and mutant HuNoV 3C protease through structure-based virtual screening. Dynamic structural results indicated the unstable binding of rupintrivir at the cleft binding site of the wild-type and mutant 3C proteases, leading to its detachment. Our findings presented that the domain II of the HuNoV 3C protease had a critical role in binding of inhibitory molecules. Binding energy computations, steered molecular dynamics and umbrella sampling simulations confirmed that amentoflavone, the novel suggested inhibitor, strongly binds to the cleft site of all protease models and has a good structural stability in the complex system along the molecular dynamic simulations. Our in silico study proposed the selected compound as a potential inhibitor against the HuNoV 3C protease. However, additional experimental and clinical studies are required to corroborate the therapeutic efficacy of the compound.
Chapter
Human norovirus is now considered the main cause of viral gastroenteritis worldwide. Norovirus-related disease is responsible for large numbers of deaths in low-income countries and vast economic losses globally. Frequent outbreaks of norovirus in hospitals (and other different semi-closed settings), constitute a major public health problem as they cause severe disruptions of normal operational service and compromise lives of hospitalized people. In addition to gastroenteritis, human noroviruses are also associated with other serious disorders such as chronic diarrhea, a severe condition which is frequent among different cohorts of immunocompromised individuals and especially prevalent in organ transplant recipients. Despite that there is a critical need for specific drugs to control norovirus disease and its spread in self-contained premises, there are yet no licenced antivirals or vaccines available. In the last few years, there have been isolated a considerable number of molecules with therapeutic potential to the control of norovirus. Several of these molecules exhibit antiviral activities in vivo, in studies carried out during clinical trials or in experiments involving animal models of infection for noroviruses. Here we review some recent advances achieved in the identification and development of antinoroviral drugs, and discuss future perspectives in this field.
Article
Full-text available
Objectives: A severe enteropathy of unknown etiology can be associated with common variable immunodeficiency (CVID). Methods: S tool and archived small intestinal mucosal biopsies from patients with CVID enteropathy were analyzed by PCR for the presence of Norovirus RNA. The PCR products were sequenced to determine the relationship of viral isolates. Stool samples from 10 patients with CVID but no enteropathy served as controls. Results: All eight patients in our CVID cohort with enteropathy showed persistent fecal excretion of Norovirus. Analysis of archived duodenal biopsies revealed a strong association between the presence of Norovirus and villous atrophy over a period of up to 8 years. Analysis of the viral isolates from each patient revealed distinct strains of genogroup II.4. Sequence analysis from consecutive biopsy specimens of one patient demonstrated persistence of the same viral strain over a 6-year period. CVID patients without enteropathy showed no evidence of Norovirus carriage. Viral clearance occurred spontaneously in one patient and followed oral Ribavirin therapy in two further patients, and resulted in complete symptomatic and histological recovery. However, Ribavirin treatment in two further patients was unsuccessful. Conclusions: Norovirus is an important pathogen for patients with CVID and a cause of CVID enteropathy, as viral clearance, symptom resolution, and histological recovery coincide. Ribavirin requires further evaluation as a potential therapy.
Article
Full-text available
Norovirus gastroenteritis is a major public health burden worldwide. Although fecal shedding is important for transmission of enteric viruses, little is known about the immune factors that restrict persistent enteric infection. We report here that while the cytokines interferon α (IFNα) and IFNβ prevented the systemic spread of murine norovirus (MNoV), only IFNλ controlled persistent enteric infection. Infection-dependent induction of IFNλ was governed by the MNoV capsid protein and correlated with diminished enteric persistence. Treatment of established infection with IFNλ cured mice in a manner requiring non-hematopoietic cell expression of the IFNλ receptor, Ifnlr1, and independent of adaptive immunity. These results suggest the therapeutic potential of IFNλ for curing virus infections in the gastrointestinal tract. Copyright © 2014, American Association for the Advancement of Science.
Article
Full-text available
The cell tropism of human noroviruses and the development of an in vitro infection model remain elusive. Although susceptibility to individual human norovirus strains correlates with an individual’s histo-blood group antigen (HBGA) profile, the biological basis of this restriction is unknown. We demonstrate that human and mouse noroviruses infected B cells in vitro and likely in vivo. Human norovirus infection of B cells required the presence of HBGA-expressing enteric bacteria. Furthermore, mouse norovirus replication was reduced in vivo when the intestinal microbiota was depleted by means of oral antibiotic administration. Thus, we have identified B cells as a cellular target of noroviruses and enteric bacteria as a stimulatory factor for norovirus infection, leading to the development of an in vitro infection model for human noroviruses.
Article
Full-text available
Objectives Norovirus outbreaks of acute gastroenteritis are highly prevalent, extensive and can disturb the functioning of health institutions, leading to the closure of hospital wards and causing life-threatening infections in long-term care facilities. There is no vaccine available; hence there is a pressing need for antivirals for the treatment (in immunodeficient patients) and prophylaxis of norovirus infections. We explored in a mouse model whether an inhibitor of norovirus replication can prevent/reduce transmission of the virus.
Article
Full-text available
Significance Human noroviruses are the predominant cause of acute gastroenteritis worldwide, but they remain noncultivatable. A tractable system is needed to understand the host restriction to cultivation. We established a reverse genetics system driven by a mammalian elongation factor-1α promoter without helper virus. This system supports genome replication, particle formation, and particles containing a GFP-marked genomic RNA. RNA from these particles is infectious. The system also produces infectious murine norovirus, confirming its broad applicability to other noroviruses.
Article
Full-text available
Potent and safe inhibitors of norovirus replication are needed for the treatment and prophylaxis of norovirus infections. We here report that the in vitro anti-norovirus activity of the protease inhibitor rupintrivir is extended to murine noroviruses and that rupintrivir clears human cells from their Norwalk replicon after only two passages of antiviral pressure. In addition, we demonstrate that rupintrivir inhibits the human norovirus (genogroup II [GII]) protease and further explain the inhibitory effect of the molecule by means of molecular modeling on the basis of the crystal structure of the Norwalk virus protease. The combination of rupintrivir with the RNA-dependent RNA polymerase inhibitors 2′-C-methylcytidine and favipiravir (T-705) resulted in a merely additive antiviral effect. The fact that rupintrivir is active against noroviruses belonging to genogroup I (Norwalk virus), genogroup V (murine norovirus), and the recombinant 3C-like protease of a GII norovirus suggests that the drug exerts cross-genotypic anti-norovirus activity and will thus most likely be effective against the clinically relevant human norovirus strains. The design of antiviral molecules targeting the norovirus protease could be a valuable approach for the treatment and/or prophylaxis of norovirus infections.
Article
Full-text available
Background Norovirus infection is increasingly recognized as an important cause of persistent gastroenteritis in immunocompromised hosts and can be a potential cause of morbidity in these populations. Case presentation Here, we report a case of norovirus-related chronic diarrhea occurring in a 62-year-old immunocompromised patient treated with alemtuzumab for chronic lymphocytic leukemia. Despite different therapeutic strategies including tapering of immunosuppressive therapy and immunoglobulin administration, diarrhea unfortunately did not resolve and lasted for a total of more than twelve weeks with prolonged norovirus fecal excretion. Conclusions Norovirus infection can occur in the setting of alemtuzumab treatment, even as a single agent, and should be included in the differential diagnoses of acute and chronic diarrhea in these immunocompromised patients. Although the administration of oral immunoglobulin has been described as a promising efficient therapy, this was not the case in our patient. Clinical trials are thus clearly warranted to better define risk factors and efficient therapies for norovirus infection in immunocompromised populations.
Article
Full-text available
Most antiviral treatment options target the invading pathogen and unavoidably encounter loss of efficacy as the pathogen mutates to overcome replication restrictions. A good strategy for circumventing drug resistance, or for pathogens without treatment options, is to target host cell proteins that are utilized by viruses during infection. The small molecule WP1130 is a selective deubiquitinase inhibitor shown previously to successfully reduce replication of noroviruses and some other RNA viruses. In this study, we screened a library of 31 small molecule derivatives of WP1130 to identify compounds that retained the broad-spectrum antiviral activity of the parent compound in vitro but exhibited improved drug-like properties, particularly increased aqueous solubility. Seventeen compounds significantly reduced murine norovirus infection in murine macrophage RAW 264.7 cells, with four causing decreases in viral titers that were similar or slightly better than WP1130 (1.9 to 2.6 log scale). Antiviral activity was observed following pre-treatment and up to 1 hour postinfection in RAW 264.7 cells as well as in primary bone marrow-derived macrophages. Treatment of the human norovirus replicon system cell line with the same four compounds also decreased levels of Norwalk virus RNA. No significant cytotoxicity was observed at the working concentration of 5 µM for all compounds tested. In addition, the WP1130 derivatives maintained their broad-spectrum antiviral activity against other RNA viruses, Sindbis virus, LaCrosse virus, encephalomyocarditis virus, and Tulane virus. Thus, altering structural characteristics of WP1130 can maintain effective broad-spectrum antiviral activity while increasing aqueous solubility.
Article
Full-text available
Norovirus (NoV) is the leading cause of acute gastroenteritis worldwide, causing over 200,000 deaths a year. NoV is nonenveloped, with a single-stranded RNA genome, and is primarily transmitted person to person. The viral RNA-dependent RNA polymerase (RdRp) is critical for the production of genomic and subgenomic RNA and is therefore a prime target for antiviral therapies. Using high-throughput screening, nearly 20,000 “lead-like” compounds were tested for inhibitory activity against the NoV genogroup II, genotype 4 (GII.4) RdRp. The four most potent hits demonstrated half-maximal inhibitory concentrations (IC50s) between 5.0 μM and 9.8 μM against the target RdRp. Compounds NIC02 and NIC04 revealed a mixed mode of inhibition, while NIC10 and NIC12 were uncompetitive RdRp inhibitors. When examined using enzymes from related viruses, NIC02 demonstrated broad inhibitory activity while NIC04 was the most specific GII.4 RdRp inhibitor. The antiviral activity was examined using available NoV cell culture models; the GI.1 replicon and the infectious GV.1 murine norovirus (MNV). NIC02 and NIC04 inhibited the replication of the GI.1 replicon, with 50% effective concentrations (EC50s) of 30.1 μM and 71.1 μM, respectively, while NIC10 and NIC12 had no observable effect on the NoV GI.1 replicon. In the MNV model, NIC02 reduced plaque numbers, size, and viral RNA levels in a dose-dependent manner (EC50s between 2.3 μM and 4.8 μM). The remaining three compounds also reduced MNV replication, although with higher EC50s, ranging from 32 μM to 38 μM. In summary, we have identified novel nonnucleoside inhibitor scaffolds that will provide a starting framework for the development and future optimization of targeted antivirals against NoV.
Article
Noroviruses (NV) are the most common cause of acute gastrointestinal illness in the United States and worldwide. The development of specific antiviral countermeasures has lagged behind that of other viral pathogens, primarily because norovirus disease has been perceived as brief and self-limiting and robust assays suitable for drug discovery have been lacking. The increasing recognition that NV illness can be life-threatening, especially in immunocompromised patients who often require prolonged hospitalization and intensive supportive care, has stimulated new research to develop an effective antiviral therapy. Here, we propose a path forward for evaluating drug therapy in norovirus-infected immunocompromised individuals, a population at high risk for serious and prolonged illness. The clinical and laboratory features of norovirus illness in immunocompromised patients are reviewed, and potential markers of drug efficacy are defined. We discuss the potential design of clinical trials in these patients and how an antiviral therapy that proves effective in immunocompromised patients might also be used in the setting of acute outbreaks, especially in confined settings such as nursing homes, to block the spread of infection and reduce the severity of illness. We conclude by reviewing the current status of approved and experimental compounds that might be evaluated in a hospital setting.