ArticlePDF Available

Inhibitors of cellular kinases with broad-spectrum antiviral activity for hemorrhagic fever viruses

Authors:

Abstract and Figures

Host cell kinases are important for the replication of a number of hemorrhagic fever viruses. We tested a panel of kinase inhibitors for their ability to block the replication of multiple hemorrhagic fever viruses. OSU-03012 inhibited the replication of Lassa, Ebola, Marburg and Nipah viruses, whereas BIBX 1382 dihydrochloride inhibited Lassa, Ebola and Marburg viruses. BIBX 1382 blocked both Lassa and Ebola virus glycoprotein-dependent cell entry. These compounds may be used as tools to understand conserved virus-host interactions, and implicate host cell kinases that may be targets for broad spectrum therapeutic intervention. Copyright © 2015. Published by Elsevier B.V.
Content may be subject to copyright.
Inhibitors of cellular kinases with broad-spectrum antiviral activity for
hemorrhagic fever viruses
q
Emma L. Mohr
a,b
, Laura K. McMullan
a
, Michael K. Lo
a
, Jessica R. Spengler
a
, Éric Bergeron
a
,
César G. Albariño
a
, Punya Shrivastava-Ranjan
a
, Cheng-Feng Chiang
a
, Stuart T. Nichol
a
,
Christina F. Spiropoulou
a,
, Mike Flint
a
a
Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease
Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA 30333, USA
b
Emory University Department of Pediatrics, Emory-Children’s Center, 2015 Uppergate Drive, Atlanta, GA 30322, USA
article info
Article history:
Received 6 April 2015
Revised 7 May 2015
Accepted 11 May 2015
Available online 16 May 2015
Keywords:
Ebola
Lassa
Antiviral
AR-12
BIBX
abstract
Host cell kinases are important for the replication of a number of hemorrhagic fever viruses. We tested a
panel of kinase inhibitors for their ability to block the replication of multiple hemorrhagic fever viruses.
OSU-03012 inhibited the replication of Lassa, Ebola, Marburg and Nipah viruses, whereas BIBX 1382
dihydrochloride inhibited Lassa, Ebola and Marburg viruses. BIBX 1382 blocked both Lassa and Ebola
virus glycoprotein-dependent cell entry. These compounds may be used as tools to understand conserved
virus–host interactions, and implicate host cell kinases that may be targets for broad spectrum therapeu-
tic intervention.
Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creative-
commons.org/licenses/by-nc-nd/4.0/).
1. Introduction
Viral hemorrhagic fevers (VHFs) cause significant morbidity and
mortality globally. The infectious etiologies of VHFs include mem-
bers of several RNA virus families, including the Arenaviridae,
Bunyaviridae,Filoviridae, and Flaviviridae. The leading cause of
VHF worldwide is Lassa virus (LASV), an arenavirus endemic to
West Africa that causes 300,000–500,000 infections annually
(Ogbu et al., 2007). Ebola virus (EBOV), Marburg virus (MARV),
Junin virus (JUNV), Alkhurma hemorrhagic fever virus (AHFV,
called Alkhumra virus in some reports), and Crimean Congo hem-
orrhagic fever virus (CCHFV), as well as the encephalitic Nipah
virus (NiV), cause sporadic outbreaks, often with high
case-fatality rates (Aljofan, 2013; Kortekaas et al., 2010; MacNeil
and Rollin, 2012; Madani, 2005). These highly pathogenic agents
are all classified as biosafety level 4 (BSL-4) pathogens; there are
no approved therapeutics or vaccines, and medical care for patients
is generally only supportive. Despite the challenges inherent in
studying BSL-4 agents, research into therapies for these viruses is
critical because of the potential for large outbreaks with high
case-fatality rates, as demonstrated by the 2013–2015 EBOV out-
break in West Africa.
Host cell kinases have been implicated in the replication of sev-
eral BSL-4 viruses. One signaling pathway, the phosphatidylinositol
3-kinase (PI3K)/Akt pathway, was reported to be essential for the
propagation of LASV and EBOV in cell culture. Inhibition of the
PI3K/Akt pathway by the small molecule BEZ-235 impeded the
budding of LASV virus-like particles (VLPs) (Urata et al., 2012).
Another inhibitor, LY294002, blocked EBOV entry (Saeed et al.,
2008) and an early event in JUNV infection (Linero and Scolaro,
2009). The replication of Andes virus, a bunyavirus, was blocked
by temsirolimus, an inhibitor of mTOR, another kinase in the
PI3K/Akt pathway (McNulty et al., 2013). We therefore hypothe-
sized that a cellular kinase could be essential for the replication
of multiple highly pathogenic viruses. Identifying such a kinase
http://dx.doi.org/10.1016/j.antiviral.2015.05.003
0166-3542/Published by Elsevier B.V.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Abbreviations: AHFV, Alkhurma hemorrhagic fever virus; BSL-4, biosafety level
4; CC
50
, 50% cytotoxic concentration; CCHFV, Crimean Congo hemorrhagic fever
virus; EBOV, Ebola virus; EC
50
, 50% effective concentration; GFP, green fluorescent
protein; LASV, Lassa virus; MARV, Marburg virus; NiV, Nipah virus; PBS, phosphate
buffered saline; SI, selectivity index; VHF, viral hemorrhagic fever; VLP, virus-like
particle; VSV, vesicular stomatitis virus.
q
The findings and conclusions in this report are those of the authors and do not
necessarily represent the official position of the Centers for Disease Control and
Prevention.
Corresponding author. Tel.: +1 404 639 1294.
E-mail address: ccs8@cdc.gov (C.F. Spiropoulou).
Antiviral Research 120 (2015) 40–47
Contents lists available at ScienceDirect
Antiviral Research
journal homepage: www.elsevier.com/locate/antiviral
might shed light on conserved virus–host interactions. In addition,
therapies targeting such kinases could have broad-spectrum
antiviral activity, a desirable property given the difficulty of devel-
oping therapies for individual hemorrhagic fever viruses. Here, we
report the identification of 2 inhibitors of cellular kinases which
impeded the replication of multiple highly pathogenic viruses.
2. Materials and methods
2.1. Biosafety
All work with infectious virus was conducted in a BSL-4 labora-
tory at the Centers for Disease Control and Prevention (CDC,
Atlanta, GA). All laboratorians adhered to international practices
appropriate for this biosafety level. Experiments involving cDNA
encoding viral sequences were approved by the CDC Institutional
Biosafety Committee.
2.2. Cell lines, viruses, and compounds
A549, Vero-E6, HeLa, and HT-1080 cells were from the CDC
Biologics Branch and HEK-293 cells were from ATCC. These cell lines
were maintained in Dulbecco’s modified Eagle’s medium (DMEM;
Life Technologies, Grand Island, NY, USA) supplemented with 10%
(v/v) fetal calf serum (FCS; Hyclone, Thermo Scientific, Waltham,
MA, USA) and penicillin–streptomycin (Life Technologies). Huh7
cells were from Apath, LLC (Brooklyn, NY, USA) and were propa-
gated in DMEM, 10% (v/v) FCS, and 1 non-essential amino acids
(Life Technologies). Viruses were from the CDC Viral Special
Pathogens Branch reference collection: LASV (strain Josiah); EBOV
(strain Mayinga); AHFV (strain 200300001); CCHFV (strain
IbAr10200). The Kinase Inhibitor Toolbox library and BIBX 1382
dihydrochloride were from Tocris Bioscience (Bristol, UK). The
PI3K Signaling Inhibitor Library and OSU-03012 were from Selleck
Chemicals (Houston, TX, USA). Compounds were diluted in
dimethylsulfoxide (DMSO; Sigma–Aldrich, St. Louis, MO, USA) as
indicated.
2.3. Assays for antiviral activity and cell viability
To test for the inhibition of LASV replication, A549 cells were
seeded at a density of 1 10
4
cells/well of a 96-well plate the
day prior to infection. Compounds were added to the cells, and
1 h later, the cells were infected with LASV at an MOI of 0.2.
After 48 h, the monolayers were fixed with 10% (v/v) formalin
(Sigma–Aldrich) and
c
-irradiated with 2 10
6
rads. The cells were
permeabilized with 0.1% (v/v) Triton X-100 in phosphate buffered
saline (PBS) for 10 min at room temperature, and LASV proteins
were detected with monoclonal antibodies directed against the
LASV glycoprotein and nucleoprotein (1:10,000 dilution in PBS
supplemented with 2% w/v bovine serum albumin) and goat
anti-mouse Alexa 488 (1:1000; Life Technologies). Cells were
stained with CellMask Red and NucBlue (Life Technologies) and
immunofluorescence microscopy was performed using the
Operetta Imaging System (PerkinElmer, Waltham, MA).
The assay for the inhibition of AHFV-induced cytopathic effect
in A549 cells was as described previously (Flint et al., 2014). The
recombinant reporter viruses NiV-luc and EBOV and MARV
expressing green fluorescent protein (GFP) reporter (EBOV-GFP
and MARV-GFP) have also been described (Albariño et al., 2013;
Lo et al., 2014; Towner et al., 2005). The assay for the inhibition
of CCHFV-induced cytopathic effects was based on one described
previously (Paragas et al., 2004). Briefly, compounds were added
to SW13 cells in an 80% confluent monolayer in a 96-well plate, fol-
lowed by infection with CCHFV at an MOI of 0.1, 1 h later. Cell
viability, as measured by CellTiter-Glo (Promega, Madison, WI,
USA), was measured 72 h post infection.
Cell viability was determined concurrently with the virus inhi-
bition assays, but on compound-treated and mock-infected cells,
using CellTiter-Glo (Promega) or PrestoBlue (Life Technologies)
according to the manufacturer’s instructions. Viability was also
assessed by nuclei number, as determined by counting the
NucBlue-stained organelles with Harmony image analysis software
(PerkinElmer). For each assay, values were normalized to
vehicle-only DMSO controls.
For compound titrations, GraphPad Prism (GraphPad Software,
La Jolla, CA, USA) was used to fit a 4-parameter equation to semilog
plots of the concentration–response data and to interpolate the
concentration of compound that inhibited 50% of the virus replica-
tion (EC
50
). The 50% cytotoxic concentration (CC
50
) was similarly
derived using viability data from mock-infected cells. The selectiv-
ity index (SI) was calculated by dividing the CC
50
by the EC
50
.
2.4. Viral titer reduction assay
Titer reduction assays for LASV and EBOV were performed in
A549 and Huh7 cells, respectively. Cells were treated with com-
pounds for 1 h prior to infection. Two days later, culture super-
natants were harvested and virus titrations were performed in
Vero-E6 cells. Three days post infection, the cells were fixed, per-
meabilized, and stained to visualize viral proteins. End-point viral
titers were determined, and the 50% tissue culture infectious dose
(TCID
50
) was calculated using the method of Reed and Muench
(Reed and Muench, 1938).
2.5. Quantitative reverse transcription polymerase chain reaction
assay
Cells were seeded and treated with compounds for 1 h before
infection with LASV at an MOI of 0.1. The medium was removed
24 h post-infection, lysis buffer (MagMax Total RNA isolation kit;
Life Technologies) was added, and RNA was extracted using a
MagMax-96 deep-well magnetic particle processor (Life
Technologies). Quantitative reverse transcription polymerase chain
reaction (qRT-PCR) was performed with the Express One-Step
Superscript qRT-PCR kit (Life Technologies) and analyzed on an
Applied Biosciences 7500 real-time PCR machine (Life
Technologies). LASV nucleoprotein RNA was quantitated using for-
ward (5
0
-AATCAGTTCGGGACCATGC-3
0
) and reverse (5
0
-GTGTTGG
GATACTTTGCTGTG-3
0
) primers and a probe oligonucleotide (5
0
-/5
6-FAM/AGTCAACCT/ZEN/GCCCCTGTTTTGTCA/Iowa Black FQ/-3
0
)
from Integrated DNA Technologies (Coralville, IA). Levels of glycer-
aldehyde 3-phosphate dehydrogenase (GAPDH) RNA, or 18S ribo-
somal RNA in Vero-E6 cells, were determined using control
primer-probe sets (Life Technologies). Viral RNA levels were nor-
malized to GAPDH or 18S RNA and expressed relative to infected,
vehicle-treated controls.
2.6. VLP assembly assay
HEK-293 cells were transfected with plasmids encoding
FLAG-tagged LASV Z protein, or the non-budding G2A mutant
(Perez et al., 2004), using Lipofectamine 2000 (Life Technologies).
Following overnight incubation, the transfection media was
removed and compounds were added to a final DMSO concentra-
tion of 0.5%. After 48 h, cell lysates were prepared with radioim-
munoprecipitation assay (RIPA) buffer supplemented with
protease inhibitors (Complete Protease Inhibitor, Roche,
Indianapolis, IN, USA). The culture medium was clarified by cen-
trifugation at 4000gfor 20 min, and VLPs were concentrated from
the supernatants by centrifugation at 300,000gthrough a 20%
E.L. Mohr et al. / Antiviral Research 120 (2015) 40–47 41
sucrose cushion. Cell lysates and pelleted VLPs were analyzed by
Western blotting with anti-FLAG M2-peroxidase antibody
(Sigma–Aldrich) or anti-bactin antibody (Genscript, Piscataway,
NJ, USA).
2.7. Assay for LASV and EBOV glycoprotein-dependent entry
HIV pseudotyped particles bearing the glycoproteins of LASV,
EBOV from the 1976 Zaire (Genbank accession No. U23187.1)
and 2014 West African (KP178538.1) outbreaks, vesicular stomati-
tis virus (VSV) G protein, or no glycoprotein were prepared and
used as previously (Flint et al., 2004). Briefly, LentiX-293T cells
(Clontech, Mountain View, CA, USA) were co-transfected with plas-
mid DNA encoding the HIV genome containing the firefly luciferase
gene (pNL4–3.Luc.R
-
.E
-
) and expression vectors encoding the viral
glycoprotein or empty vector in a 32:1 ratio. Pseudotyped viruses
were quantitated by determining HIV matrix protein (p24) con-
tent. Viral glycoprotein-dependent entry assays were performed
using HT-1080 cells and 6 ng of p24 pseudotyped particles, with
firefly luciferase activity determined using BrightGlo (Promega)
72 h post-transduction. Cell viability was concurrently determined
by CellTiter-Glo (Promega) in compound-treated and
mock-transduced cells.
3. Results
3.1. Identification of kinase inhibitors with activity against
hemorrhagic fever viruses
We hypothesized that a cellular kinase would be essential for
the replication of multiple BSL-4 viruses. Therefore, we initially
tested a total of 163 kinase inhibitors, focusing on the PI3K/Akt
Table 1
Compounds inhibiting LASV replication in A549 cells.
Compound % infected cells
a
% cell viability
a
Reported targets
b
5
l
M 500 nM 50 nM 5
l
M 500 nM 50 nM
BEZ-235 14 ± 3 19 ± 5 43 ± 7 31 ± 4 32 ± 1 48 ± 3 PI3K, mTOR
OSU-03012 0 ± 0 32 ± 9 94 ± 18 60 ± 4 82 ± 1 94 ± 4 PDK-1, PAK
AZD8055 4 ± 3 17 ± 15 18 ± 4 34 ± 4 36 ± 1 55 ± 2 mTOR
TWS119 24 ± 9 41 ± 16 41 ± 3 71 ± 7 91 ± 2 93 ± 1 GSK-3
SU 4312 32 ± 6 80 ± 19 83 ± 29 96 ± 3 100 ± 2 98 ± 3 VEGFR
BIBX 1382 dihydrochloride 8 ± 1 69 ± 5 75 ± 7 70 ± 4 85 ± 1 94 ± 3 ErbB1, ErbB2, ErbB4
Ki 8751 23 ± 1 70 ± 14 90 ± 18 57 ± 5 99 ± 2 100 ± 4 VEGFR
10-DEBC hydrochloride 11 ± 5 85 ± 25 81 ± 7 69 ± 3 97 ± 2 95 ± 1 Akt, PKB
a
Mean % infected cells or % viability ± standard deviation from 3 replicate wells, normalized to the vehicle-only control.
b
PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; PAK, p21-activated kinase; mTOR, mammalian target of rapamycin; PDK-1, 3-phosphoinositide-dependent kinase
1; GSK-3, glycogen synthase kinase 3; VEGFR, vascular endothelial growth factor receptor; EGFR, epidermal growth factor receptor; PKB, protein kinase B; n/a, not applicable.
A
CB
1852 617 206 69 23 8
Mock-infected DMSO
OSU-03012 concentr on (nM)
% infected cells
% viability
Fig. 1. OSU-03012 inhibits LASV and EBOV in cell culture. (A) OSU-03012 induced a concentration-dependent reduction in LASV replication. A549 cells were treated for 1 h
with varying concentrations of OSU-03012 before infection with LASV at an MOI of 0.2. Two days post infection, the cells were fixed, permeabilized, and stained. Green, LASV
proteins; blue, cell nuclei; red, cell cytoplasm. (B) A representative concentration–response curve showing the quantitation of LASV-infected cells (%) normalized to the
vehicle-only control; each point is the mean of 9 fields from each of quadruplicate wells, with error bars indicating the standard deviation. Cell viability (%) of compound-
treated, mock-infected cells is also shown; each point is the mean of quadruplicate wells, with error bars indicating standard deviation. (C) A representative concentration–
response curve showing viability of mock-infected cells, or inhibition of GFP expression in Huh 7 cells infected with the EBOV-GFP reporter virus. Points represent mean
values, and error bars indicate standard deviations calculated from 4 replicate wells.
42 E.L. Mohr et al. / Antiviral Research 120 (2015) 40–47
pathway, for their ability to inhibit the replication of LASV. A549
cells were treated with compounds 1 h prior to mock-infection or
infection with LASV at an MOI of 0.2. Two days post infection,
the cells were fixed, permeabilized, and stained to visualize LASV
proteins and the mock-infected cells were tested for viability
(Supplemental Table 1). BEZ-235, a known inhibitor of LASV VLP
formation (Urata et al., 2012), was used as a positive control. In
addition to BEZ-235, 7 compounds had anti-LASV activity with
minimal cytotoxicity (Table 1). These compounds were tested for
their ability to inhibit the replication of EBOV-GFP, MARV-GFP,
and NiV-luc recombinant reporter viruses, as well as AHFV and
CCHFV. Two compounds, OSU-03012 (also known as AR-12) and
BIBX 1382 dihydrochloride, demonstrated inhibition of both
LASV and other viruses and were further characterized.
Table 2
OSU-03012 activity against selected viruses.
Assay type
a
Cell line Multiplicity of infection Duration (h) EC
50b
(
l
M) CC
50
CellTiter-Glo (
l
M) SI CellTiter-Glo
LASV Immunofluorescence A549 0.2 48 0.5 ± 0.1 5.7 ± 2.5 11
AHFV CPE inhibition A549 0.1 72 2.9 ± 0.8 6.5 ± 1.1 2
NiV-Luc Luc reporter 293T 0.2 48 0.4 ± 0.2 8.2 ± 0.9 21
EBOV-GFP GFP reporter Huh7 0.2 48 0.3 ± 0.07 6.4 ± 1.1 23
MARV-GFP GFP reporter Huh7 0.2 48 0.3 ± 0.1 7.1 ± 0.6 20
CCHFV CPE inhibition SW13 0.1 72 No inhibition 6.7 ± 0.8 n/a
a
CPE, cytopathic effect; Luc, luciferase; GFP, green fluorescent protein.
b
Mean EC
50
and CC
50
values ± the standard deviation from at least 3 independent determinations are shown.
Table 3
Activity of BIBX 1382 dihydrochloride against selected viruses.
EC
50a
(
l
M) CC
50
CellTiter-Glo (
l
M) SI CellTiter-Glo CC
50
PrestoBlue (
l
M) SI PrestoBlue CC
50
nuclei number (
l
M) SI nuclei number
LASV 3.2 ± 2.4 15.3 ± 6.7 6 21.0 ± 1.4 7 63.4 ± 16.7 24
EBOV-GFP 1.1 ± 0.6 19.8 ± 0.3 18 ND ND ND ND
MARV-GFP 1.8 ± 0.2 29.1 ± 16.4 19 ND ND ND ND
ND: not done.
a
Mean EC
50
and CC
50
values ± the standard deviation from at least 3 independent determinations are shown.
BA
DC
2831XBIB21030-USO
LASVEBOV
EC₅₀ = 257 nM
CC₅₀ = 5700 nM
SI = 22
EC₅₀ = 3250 nM
CC₅₀ = 15300 nM
SI = 5
EC₅₀ = 172 nM
CC₅₀ = 6400 nM
SI = 37
EC₅₀ = 2012 nM
CC₅₀ = 19800 nM
SI = 10
Fig. 2. OSU-03012 (A and C) and BIBX 1382 (B and D) inhibit LASV and EBOV titers in a concentration-dependent manner. Cells were in treated with varying concentrations of
compound for 1 h. A549 cells were then infected with LASV at an MOI of 0.2 (A and B), and Huh7 cells were infected with EBOV at an MOI of 0.02 (C and D). Two days post
infection, culture supernatants were harvested and viral titers determined in Vero-E6 cells. Mean titers are shown with error bars indicating standard errors calculated from 3
replicate wells.
E.L. Mohr et al. / Antiviral Research 120 (2015) 40–47 43
Fig. 3. OSU-03012 and BIBX 1382 induce a concentration-dependent decrease in LASV RNA in multiple cell lines. (A) A549, (B) Huh7, (C) Vero-E6, and (D) HT-1080 cells were
treated with the indicated compound concentrations for 1 h, and then infected with LASV at an MOI of 0.1. RNA was extracted 24 h post infection, and qRT-PCR for LASV RNA
or endogenous cell RNA was performed (white bars). Mock-infected cell monolayers were processed for viability assays using CellTiter-Glo (black bars). Columns represent
mean values, and error bars indicate standard deviations calculated from 8 replicate wells.
44 E.L. Mohr et al. / Antiviral Research 120 (2015) 40–47
3.2. OSU-03012 and BIBX 1382 inhibit multiple highly pathogenic
viruses in cell culture
OSU-03012 was reported to be an inhibitor of
3-phosphinositide-dependent protein kinase 1 (PDK-1) and an
inducer of apoptosis (Zhu et al., 2004). It reduced LASV replication
in A549 cells in a concentration-dependent manner (Fig. 1A and B),
with an EC
50
of 0.5
l
M(Table 2). We used 3 different methods to
measure cell viability to ensure the CC
50
value was accurately
determined: by determining cellular ATP (CellTiter-Glo) and reduc-
ing activity (PrestoBlue), and by counting nuclei by high content
image analysis. The CC
50
values were comparable for each assay
(5.7 ± 2.5
l
M for CellTiter-Glo, 5.4 ± 1.7
l
M for PrestoBlue,
3.1 ± 0.8
l
M for nuclei number; Table 2). The SI for OSU-03012
inhibition of LASV in A549 cells was 6–11, indicating an antiviral
effect rather than reduced virus growth due to cell death.
OSU-03012 also inhibited NiV-Luc, EBOV-GFP, and MARV-GFP with
EC
50
values around 0.3
l
M(Fig. 1C and Table 2) and SI values of
approximately 20. OSU-03012 had no detectable effect on CCHFV
replication, and any inhibition of AHFV was difficult to distinguish
from cytotoxic effects (SI = 2–3; Table 2).
BIBX 1382 was reported to be an inhibitor of the ErbB kinases,
including ErbB1, the epidermal growth factor receptor (EGFR)
kinase (Egeblad et al., 2001). In our study, it inhibited LASV,
EBOV-GFP, and MARV-GFP, with EC
50
values ranging 1.1–3.2
l
M
and SI values 6–24 (Table 3). BIBX 1382 had no specific antiviral
effects against NiV-Luc, CCHFV, or AHFV (data not shown).
Next, we tested the effect of OSU-03012 and BIBX 1382 on viral
titers. A549 or Huh7 cells were treated with the compounds for 1 h
prior to infection with LASV or wild-type EBOV, respectively. Two
days post infection, the cell supernatants were harvested and viral
titers were determined. Both OSU-03012 and BIBX 1382 reduced
LASV and EBOV titers by 2–3 logs in a concentration-dependent
manner (Fig. 2A–D) at concentrations below their CC
50
values
(compare concentrations used to CC
50
values in Tables 2 and 3).
To further confirm the antiviral effects of OSU-03012 and BIBX
1382, we measured LASV RNA in various infected cell lines. A549,
Vero-E6, HT-1080, and Huh7 cell lines were treated with the com-
pounds for 1 h, and then infected with LASV at an MOI of 0.1. RNA
was extracted from the infected cells 24 h post infection, and LASV
RNA was quantified by qRT-PCR. The viability of compound-treated
and mock-infected cells was determined simultaneously. Both
compounds induced a concentration-dependent reduction in
LASV RNA in each of the cell lines, with minimal effects on cell via-
bility (Fig. 3).
3.3. Mechanism of action studies
To understand the antiviral mechanisms of OSU-03012 and
BIBX 1382, we used assays that recapitulate individual steps of
the viral replication cycle. First, we tested the ability of these com-
pounds to inhibit LASV and EBOV glycoprotein-dependent entry by
using HIV particles pseudotyped with the viral glycoproteins.
Amiodarone, a recently reported inhibitor of EBOV-glycoprotein
dependent entry, was used as a control (Gehring et al., 2014).
OSU-03012 did not affect entry mediated by any of the glycopro-
teins tested (data not shown). In contrast, BIBX 1382 inhibited
EBOV-glycoprotein dependent entry, with similar potency against
the 1976 and 2014 EBOV sequences (Fig. 4 and Table 4). LASV
glycoprotein-dependent entry was also affected (Fig. 4 and
Table 4). OSU-03012, BIBX 1382, and amiodarone did not inhibit
VSV-G-dependent entry (Fig. 4).
When expressed in cell culture, the LASV Z protein assembles
into VLPs that are released into the culture medium (Perez et al.,
2003; Strecker et al., 2003). To determine if OSU-03012 and BIBX
1382 affected VLP formation, HEK-293 cells were transfected with
plasmids encoding LASV Z, then treated with each compound. VLPs
were harvested from the culture medium, centrifuged through
sucrose cushions, and detected by Western blotting. A mutant Z
protein (G2A) lacking the myristoylation site required for VLP
Fig. 4. Effect of (A) BIBX 1382 and (B) amiodarone on LASV and EBOV glycoprotein-
dependent entry. HT-1080 cells were transduced with HIV particles pseudotyped
with EBOV, LASV, or vesicular stomatitis virus (VSV) glycoproteins in cell culture
medium containing inhibitor compounds. Cell culture media containing the
pseudotype particles and compounds were removed 6 h post transduction and
were replaced with cell culture media without compound. Transduction was
measured by firefly luciferase activity 3 days post transduction. Mock-transduced,
compound-treated cell monolayers were processed for viability assays using
CellTiter-Glo. A representative of 3 independent experiments is shown. Points
represent mean values, and error bars indicate standard deviations calculated from
4 replicate wells.
Table 4
Effects of BIBX 1382 dihydrochloride and amiodarone on EBOV, LASV, and VSV
glycoprotein-dependent entry, and on cell viability.
Virus
glycoprotein
BIBX 1382 Amiodarone HCl
EC
50
(
l
M)
a
CC
50
(
l
M)
a
SI EC
50
(
l
M)
a
CC
50
(
l
M)
a
SI
EBOV (1976)
b
1.2 ± 0.2 24.3 ± 3.9 20 1.4 ± 0.4 47.1 ± 18 34
EBOV (2014) 1.6 ± 0.3 15 1.6 ± 0.4 29
LASV 7.5 ± 3.0 3 10.7 ± 1.3 4
VSV 30.9 ± 8.5 <1 35.4 ± 13.7 1
a
Mean EC
50
and CC
50
values ± the standard deviation from at least 3 independent
determinations are shown.
b
EBOV (1976): EBOV strain isolated during the 1976 Zaire outbreak. EBOV
(2014): EBOV strain isolated during the 2014–2015 outbreak in West Africa.
E.L. Mohr et al. / Antiviral Research 120 (2015) 40–47 45
formation (Perez et al., 2004) served as a negative control. Each
transfected cell monolayer expressed similar amounts of
wild-type Z or G2A mutant regardless of treatment (Fig. 5). As
expected, the G2A mutant was not secreted, while the wild-type
Z protein assembled into VLPs. Neither OSU-03012 nor BIBX
1382 inhibited the release of LASV Z VLPs (Fig. 5).
4. Discussion
Host cell kinases have been implicated in the replication of sev-
eral BSL-4 viruses (Linero and Scolaro, 2009; Saeed et al., 2008;
Urata et al., 2012). Here, we identified 2 inhibitors of cellular
kinases that inhibit multiple such viruses in cell culture.
OSU-03012 (AR-12) is a celecoxib derivative that does not inhi-
bit cyclooxygenase-2, but has been reported to inhibit PDK-1 and
p21-activated kinase (Porchia et al., 2007; Zhu et al., 2004). We
tested several other reported inhibitors of PDK-1 for their ability
to inhibit LASV replication, including PHT-427, BX-912, BX-795,
PS 48, and GSK 2334470. None of these had a specific anti-LASV
activity (data not shown), suggesting that the antiviral effect of
OSU-03012 is probably not mediated through PDK-1 inhibition. It
is important to note that kinase inhibitors, especially those that
bind within the highly-conserved ATP binding site, frequently inhi-
bit multiple cellular kinases. OSU-03012 was reported to induce
apoptosis or autophagy (Gao et al., 2008; Johnson et al., 2005;
Lee et al., 2009; Liu et al., 2013; Zhu et al., 2004) and was the sub-
ject of a phase I clinical trial in patients with advanced or recurrent
solid tumors or lymphoma. Recently, OSU-03012 was reported to
reduce expression of the endoplasmic reticulum chaperone
HSPA5 (also called GRP78 or BiP) (Booth et al., 2015). Such a reduc-
tion could inhibit the assembly of enveloped viruses that rely on
HSPA5 activity for glycoprotein folding. HSPA5 is incorporated into
the EBOV virion (Spurgers et al., 2010), and knocking down HSPA5
in mice conferred protection from lethal challenge with EBOV (Reid
et al., 2014). Although OSU-03012 was reported to decrease the
expression of NPC1 and LAMP1 (Booth et al., 2015), molecules
involved in the entry of EBOV and LASV (Carette et al., 2011;
Cote et al., 2011; Jae et al., 2014), it did not inhibit EBOV or LASV
glycoprotein-dependent entry in our HIV pseudotype system
(Fig. 4). We are currently investigating the effect of OSU-03012
on the maturation of the EBOV and LASV glycoproteins.
BIBX 1382 was reported to be an inhibitor of the ErbB kinases,
including EGFR (Egeblad et al., 2001). Further work is required to
understand the mechanism by which BIBX 1382 inhibits LASV
and EBOV glycoprotein-dependent entry. These two viruses appear
to use different routes of entry, with LASV apparently using
clathrin-mediated endocytosis and EBOV using macropinocytosis
(Nanbo et al., 2010; Saeed et al., 2010; Vela et al., 2007). Since
BIBX 1382 appears to inhibit the entry of both viruses, this sug-
gests that some aspect of the entry mechanism is shared between
the two. The development of BIBX 1382 was halted after a phase I
study revealed low bioavailability and a dose-limiting increase of
liver enzymes (Dittrich et al., 2002). It may therefore be difficult
to justify repurposing this compound as a candidate VHF
therapeutic.
In conclusion, we have identified 2 compounds with antiviral
activity against LASV, EBOV and MARV in cell culture. Our data
suggest that BIBX 1382 is an inhibitor of LASV and EBOV entry.
OSU-03012 reduces LASV and EBOV virus titers and RNA levels,
but does not inhibit entry or LASV Z-protein mediated assembly.
We believe that testing OSU-03012 for antiviral efficacy in appro-
priate animal models is warranted. In addition, OSU-03012 and
BIBX 1382 may be useful tools for understanding interactions
between these viruses and their host cells.
Acknowledgements
We thank Tanya Klimova for assistance with editing this manu-
script. The findings and conclusions in this report are those of the
authors and do not necessarily represent the official position of the
Centers for Disease Control and Prevention.
Appendix A. Supplementary data
Supplementary data associated with this article can be found, in
the online version, at http://dx.doi.org/10.1016/j.antiviral.2015.05.
003.
References
Albariño, C.G., Uebelhoer, L.S., Vincent, J.P., Khristova, M.L., Chakrabarti, A.K.,
McElroy, A., Nichol, S.T., Towner, J.S., 2013. Development of a reverse genetics
system to generate recombinant Marburg virus derived from a bat isolate.
Virology 446, 230–237.
Aljofan, M., 2013. Hendra and Nipah infection: emerging paramyxoviruses. Virus
Res. 177, 119–126.
Booth, L., Roberts, J.L., Cash, D.R., Tavallai, S., Jean, S., Fidanza, A., Cruz-Luna, T.,
Siembiba, P., Cycon, K.A., Cornelissen, C.N., Dent, P., 2015. GRP78/BiP/HSPA5/
Dna K is a universal therapeutic target for human disease. J. Cell. Physiol. 230,
1661–1676.
Carette, J.E., Raaben, M., Wong, A.C., Herbert, A.S., Obernosterer, G., Mulherkar, N.,
Kuehne, A.I., Kranzusch, P.J., Griffin, A.M., Ruthel, G., Dal Cin, P., Dye, J.M.,
Whelan, S.P., Chandran, K., Brummelkamp, T.R., 2011. Ebola virus entry requires
the cholesterol transporter Niemann-Pick C1. Nature 477, 340–343.
Cote, M., Misasi, J., Ren, T., Bruchez, A., Lee, K., Filone, C.M., Hensley, L., Li, Q., Ory, D.,
Chandran, K., Cunningham, J., 2011. Small molecule inhibitors reveal Niemann-
Pick C1 is essential for Ebola virus infection. Nature 477, 344–348.
Dittrich, C., Greim, G., Borner, M., Weigang-Kohler, K., Huisman, H., Amelsberg, A.,
Ehret, A., Wanders, J., Hanauske, A., Fumoleau, P., 2002. Phase I and
pharmacokinetic study of BIBX 1382 BS, an epidermal growth factor receptor
(EGFR) inhibitor, given in a continuous daily oral administration. Eur. J. Cancer
38, 1072–1080.
Egeblad, M., Mortensen, O.H., van Kempen, L.C., Jaattela, M., 2001. BIBX1382BS, but
not AG1478 or PD153035, inhibits the ErbB kinases at different concentrations
in intact cells. Biochem. Biophys. Res. Commun. 281, 25–31.
Flint, M., Logvinoff, C., Rice, C.M., McKeating, J.A., 2004. Characterization of
infectious retroviral pseudotype particles bearing hepatitis C virus
glycoproteins. J. Virol. 78, 6875–6882.
Flint, M., McMullan, L.K., Dodd, K.A., Bird, B.H., Khristova, M.L., Nichol, S.T.,
Spiropoulou, C.F., 2014. Inhibitors of the tick-borne, hemorrhagic fever-
associated flaviviruses. Antimicrob. Agents Chemother. 58, 3206–3216.
Gao, M., Yeh, P.Y., Lu, Y.S., Hsu, C.H., Chen, K.F., Lee, W.C., Feng, W.C., Chen, C.S., Kuo,
M.L., Cheng, A.L., 2008. OSU-03012, a novel celecoxib derivative, induces
reactive oxygen species-related autophagy in hepatocellular carcinoma. Cancer
Res. 68, 9348–9357.
Gehring, G., Rohrmann, K., Atenchong, N., Mittler, E., Becker, S., Dahlmann, F.,
Pohlmann, S., Vondran, F.W., David, S., Manns, M.P., Ciesek, S., von Hahn, T.,
2014. The clinically approved drugs amiodarone, dronedarone and verapamil
inhibit filovirus cell entry. J. Antimicrob. Chemother. 69, 2123–2131.
Jae, L.T., Raaben, M., Herbert, A.S., Kuehne, A.I., Wirchnianski, A.S., Soh, T.K., Stubbs,
S.H., Janssen, H., Damme, M., Saftig, P., Whelan, S.P., Dye, J.M., Brummelkamp,
VLPs FLAG
Cells FLAG
OSU-03012 (µ
DMSO
DMSO
6
3
1.5
0.75
BIBX-1382 (µ
DMSO
DMSO
15
7.5
3.75
1.875
Compound:
Z-WT
Z-G2A
Cells
Z-WT
Z-WT
Z-WT
Z-WT
Z-WT
Z-G2A
Z-WT
Z-WT
Z-WT
Z-WT
Fig. 5. Neither OSU-03012 nor BIBX 1382 inhibit the formation of LASV Z protein
virus-like particles (VLPs). HEK-293 cells were transfected with plasmids express-
ing LASV Z protein or a G2A mutant, and treated with varying concentrations of
compound, or with DMSO as a vehicle-only control. After 48 h, the cells were lysed
and analyzed for the expression of Z protein or actin; VLPs in the culture
supernatant were collected by centrifugation through a sucrose cushion before
analysis. A representative of 2 independent experiments is shown.
46 E.L. Mohr et al. / Antiviral Research 120 (2015) 40–47
T.R., 2004. Virus entry. Lassa virus entry requires a trigger-induced receptor
switch. Science 344, 1506–1510.
Johnson, A.J., Smith, L.L., Zhu, J., Heerema, N.A., Jefferson, S., Mone, A., Grever, M.,
Chen, C.S., Byrd, J.C., 2005. A novel celecoxib derivative, OSU03012, induces
cytotoxicity in primary CLL cells and transformed B-cell lymphoma cell line via
a caspase- and Bcl-2-independent mechanism. Blood 105, 2504–2509.
Kortekaas, J., Ergonul, O., Moormann, R.J., 2010. Interventions against West Nile
virus, Rift Valley fever virus, and Crimean-Congo hemorrhagic fever virus:
where are we? Vector Borne Zoonotic Dis. 10, 709–718.
Lee, T.X., Packer, M.D., Huang, J., Akhmametyeva, E.M., Kulp, S.K., Chen, C.S.,
Giovannini, M., Jacob, A., Welling, D.B., Chang, L.S., 2009. Growth inhibitory and
anti-tumour activities of OSU-03012, a novel PDK-1 inhibitor, on vestibular
schwannoma and malignant schwannoma cells. Eur. J. Cancer 45, 1709–1720.
Linero, F.N., Scolaro, L.A., 2009. Participation of the phosphatidylinositol 3-kinase/
Akt pathway in Junin virus replication in vitro. Virus Res. 145, 166–170.
Liu, J., Qin, C.K., Lv, W., Zhao, Q., Qin, C.Y., 2013. OSU-03012, a non-Cox inhibiting
celecoxib derivative, induces apoptosis of human esophageal carcinoma cells
through a p53/Bax/cytochrome c/caspase-9-dependent pathway. Anticancer
Drugs 24, 690–698.
Lo, M.K., Nichol, S.T., Spiropoulou, C.F., 2014. Evaluation of luciferase and GFP-
expressing Nipah viruses for rapid quantitative antiviral screening. Antiviral
Res. 106, 53–60.
MacNeil, A., Rollin, P.E., 2012. Ebola and Marburg hemorrhagic fevers: neglected
tropical diseases? PLoS Negl. Trop. Dis. 6, e1546.
Madani, T.A., 2005. Alkhurma virus infection, a new viral hemorrhagic fever in Saudi
Arabia. J. Infect. 51, 91–97.
McNulty, S., Flint, M., Nichol, S.T., Spiropoulou, C.F., 2013. Host mTORC1 signaling
regulates andes virus replication. J. Virol. 87, 912–922.
Nanbo, A., Imai, M., Watanabe, S., Noda, T., Takahashi, K., Neumann, G., Halfmann, P.,
Kawaoka, Y., 2010. Ebolavirus is internalized into host cells via
macropinocytosis in a viral glycoprotein-dependent manner. PLoS Pathog. 6,
e1001121.
Ogbu, O., Ajuluchukwu, E., Uneke, C.J., 2007. Lassa fever in West African sub-region:
an overview. J. Vector Borne Dis. 44, 1–11.
Paragas, J., Whitehouse, C.A., Endy, T.P., Bray, M., 2004. A simple assay for
determining antiviral activity against Crimean-Congo hemorrhagic fever virus.
Antiviral Res. 62, 21–25.
Perez, M., Craven, R.C., de la Torre, J.C., 2003. The small RING finger protein Z drives
arenavirus budding: implications for antiviral strategies. Proc. Natl. Acad. Sci.
USA 100, 12978–12983.
Perez, M., Greenwald, D.L., de la Torre, J.C., 2004. Myristoylation of the RING finger Z
protein is essential for arenavirus budding. J. Virol. 78, 11443–11448.
Porchia, L.M., Guerra, M., Wang, Y.C., Zhang, Y., Espinosa, A.V., Shinohara, M., Kulp,
S.K., Kirschner, L.S., Saji, M., Chen, C.S., Ringel, M.D., 2007. 2-amino-N-{4-[5-(2-
phenanthrenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]-phenyl} acetamide
(OSU-03012), a celecoxib derivative, directly targets p21-activated kinase.
Mol. Pharmacol. 72, 1124–1131.
Reed, L.J., Muench, H., 1938. A simple method of estimating fifty per cent endpoints.
Am. J. Hyg. 27, 493–497.
Reid, S.P., Shurtleff, A.C., Costantino, J.A., Tritsch, S.R., Retterer, C., Spurgers, K.B.,
Bavari, S., 2014. HSPA5 is an essential host factor for Ebola virus infection.
Antiviral Res. 109, 171–174.
Saeed, M.F., Kolokoltsov, A.A., Albrecht, T., Davey, R.A., 2010. Cellular entry of Ebola
virus involves uptake by a macropinocytosis-like mechanism and subsequent
trafficking through early and late endosomes. PLoS Pathog. 6, e1001110.
Saeed, M.F., Kolokoltsov, A.A., Freiberg, A.N., Holbrook, M.R., Davey, R.A., 2008.
Phosphoinositide-3 kinase-Akt pathway controls cellular entry of Ebola virus.
PLoS Pathog. 4, e1000141.
Spurgers, K.B., Alefantis, T., Peyser, B.D., Ruthel, G.T., Bergeron, A.A., Costantino, J.A.,
Enterlein, S., Kota, K.P., Boltz, R.C., Aman, M.J., Delvecchio, V.G., Bavari, S., 2010.
Identification of essential filovirion-associated host factors by serial proteomic
analysis and RNAi screen. Mol. Cell. Proteomics 9, 2690–2703.
Strecker, T., Eichler, R., Meulen, J., Weissenhorn, W., Dieter Klenk, H., Garten, W.,
Lenz, O., 2003. Lassa virus Z protein is a matrix protein and sufficient for the
release of virus-like particles [corrected]. J. Virol. 77, 10700–10705.
Towner, J.S., Paragas, J., Dover, J.E., Gupta, M., Goldsmith, C.S., Huggins, J.W., Nichol,
S.T., 2005. Generation of eGFP expressing recombinant Zaire Ebolavirus for
analysis of early pathogenesis events and high-throughput antiviral drug
screening. Virology 332, 20–27.
Urata, S., Ngo, N., de la Torre, J.C., 2012. The PI3K/Akt pathway contributes to
arenavirus budding. J. Virol. 86, 4578–4585.
Vela, E.M., Zhang, L., Colpitts, T.M., Davey, R.A., Aronson, J.F., 2007. Arenavirus entry
occurs through a cholesterol-dependent, non-caveolar, clathrin-mediated
endocytic mechanism. Virology 369, 1–11.
Zhu, J., Huang, J.W., Tseng, P.H., Yang, Y.T., Fowble, J., Shiau, C.W., Shaw, Y.J., Kulp,
S.K., Chen, C.S., 2004. From the cyclooxygenase-2 inhibitor celecoxib to a novel
class of 3-phosphoinositide-dependent protein kinase-1 inhibitors. Cancer Res.
64, 4309–4318.
E.L. Mohr et al. / Antiviral Research 120 (2015) 40–47 47
... To determine the suitability of rLUJV/ZsG for in vitro screening assays, potential LUJV inhibitors were screened using rLUJV/ZsG in Huh7 cells, which have been previously used in the discovery and evaluation of antiviral compounds for other VHF pathogens [16,18,19]. The assays were optimized using ribavirin, a well-documented broad-spectrum antiviral. ...
... BX-795 inhibits 3-phosphoinositide-dependent kinase 1, IKK-related kinase, TANK-binding kinase 1, and IKKε; afatanib is a tyrosine kinase inhibitor. AR-12 has previously been shown to inhibit both LASV [18] and JUNV [43], and BX-795 has shown promise as a potent inhibitor of herpes simplex virus 1 (HSV-1) and HSV-2 [44]. Against rLUJV/ZsG, BX-795 (EC 50 0.60 ± 0.27 µM, CC 50 9.91 ± 1.09 µM) performed the best, with an SI value of 16.6. ...
... BX-795 inhibits 3-phosphoinositide-dependent kinase 1, IKK-related kinase, TANK-binding kinase 1, and IKKε; afatanib is a tyrosine kinase inhibitor. AR-12 has previously been shown to inhibit both LASV [18] and JUNV [43], and BX-795 has shown promise as a potent inhibitor of herpes simplex virus 1 (HSV-1) and HSV-2 [44]. Against rLUJV/ZsG, BX-795 (EC50 0.60 ± 0.27 μM, CC50 9.91 ± 1.09 μM) performed the best, with an SI value of 16.6. ...
Article
Full-text available
Lujo virus (LUJV), a highly pathogenic arenavirus, was first identified in 2008 in Zambia. To aid the identification of effective therapeutics for LUJV, we developed a recombinant reporter virus system, confirming reporter LUJV comparability with wild-type virus and its utility in high-throughput antiviral screening assays. Using this system, we evaluated compounds with known and unknown efficacy against related arenaviruses, with the aim of identifying LUJV-specific and potential new pan-arenavirus antivirals. We identified six compounds demonstrating robust anti-LUJV activity, including several compounds with previously reported activity against other arenaviruses. These data provide critical evidence for developing broad-spectrum antivirals against high-consequence arenaviruses.
... NCT00978523). AR-12 In vitro (195,196,201,203) ZIKV, DENV ...
... In vivo ( In vitro (169,172) inhibits replication of several viruses requiring cellular kinases in vitro (Lassa, Ebola, Marburg, and Nipah viruses [196]). It also inhibits replication of DENV and ZIKV, two flaviviruses that are known to require GRP78 and activate the PI3K/AKT pathway (197)(198)(199). ...
Article
Full-text available
Viruses are intracellular parasites that subvert the functions of their host cells to accomplish their infection cycle. The endoplasmic reticulum (ER)-residing chaperone proteins are central for the achievement of different steps of the viral cycle, from entry and replication to assembly and exit. The most abundant ER chaperones are GRP78 (78-kDa glucose-regulated protein), GRP94 (94-kDa glucose-regulated protein), the carbohydrate or lectin-like chaperones calnexin (CNX) and calreticulin (CRT), the protein disulfide isomerases (PDIs), and the DNAJ chaperones. This review will focus on the pleiotropic roles of ER chaperones during viral infection. We will cover their essential role in the folding and quality control of viral proteins, notably viral glycoproteins which play a major role in host cell infection. We will also describe how viruses co-opt ER chaperones at various steps of their infectious cycle but also in order to evade immune responses and avoid apoptosis. Finally, we will discuss the different molecules targeting these chaperones and the perspectives in the development of broad-spectrum antiviral drugs.
... • 16G8 and 17C8 as inhibitors of LASV, JUNV, MACV GTOV replication via entry interference (IC 50 ≈200-350 nM) [180]; • bergamottin and casticin as entry inhibitors of CHAPV, GTOV, JUNV, LASV, LUJV, and MACV transduction in the micromolar range [181]; • BEZ-235, a dual phosphatidylinositol 3-kinase (PI3K)/ mammalian target of rapamycin (Rpm) (mTOR) inhibitor and LY294002, a PI3K inhibitor, as inhibitors of LASV particle production [182]; • BIBX 1382 and OSU-03012, two kinase inhibitors, as inhibitors of LASV replication [183]; • cap-dependent endonuclease inhibitors that inhibit JUNV and LASV replication at micromolar concentrations [184]; • CP100356, a specific P-glycoprotein inhibitor, as an inhibitor of LASV replication (IC 50 = 0.062 μM) and JUNV, LASV, MACV, and SBAV pseudotype transduction [185]; • F1204 and F1781, benzotriazole derivates that target the LASV endonuclease activity in a minigenome system [166]; • F1920 and F1965, which inhibit LASV pseudotype transduction [161]; • PF-429242, an inhibitor of cellular site 1 protease (S1P)mediated processing of LASV GPV, as a disruptor of LASV maturation in the low micromolar range [186]; • quercetin, a flavonol that interrupts the PI3K/Akt pathway, as an inhibitor of JUNV replication via entry interference [187]; • BEZ-235 and LY294002, which together target the PI3K/ Akt pathway inhibit viral budding of LASV and replication [182]; ...
Article
Introduction Mammarenaviruses are negative-sense bisegmented enveloped RNA viruses that are endemic in Africa, the Americas, and Europe. Several are highly virulent, causing acute human diseases associated with high case fatality rates, and are considered to be significant with respect to public health impact or bioterrorism threat. Areas covered This review summarizes the status quo of treatment development, starting with drugs that are in advanced stages of evaluation in early clinical trials, followed by promising candidate medical countermeasures emerging from bench analyses and investigational animal research. Expert opinion Specific therapeutic treatments for diseases caused by mammarenaviruses remain limited to the off-label use of ribavirin and transfusion of convalescent sera. Progress in identifying novel candidate medical countermeasures against mammarenavirus infection has been slow in part because of the biosafety and biosecurity requirements. However, novel methodologies and tools have enabled increasingly efficient high-throughput molecular screens of regulatory-agency-approved small-molecule drugs and led to the identification of several compounds that could be repurposed for the treatment of infection with several mammarenaviruses. Unfortunately, most of them have not yet been evaluated in vivo. The most promising treatment under development is a monoclonal antibody cocktail that is protective against multiple lineages of the Lassa virus in nonhuman primate disease models.
... Secondly, they checked that OSU-03012 and BIBX 1382 affect the formation of virus-like particles (VLP) of LASV Z protein. Both OSU-03012 and BIBX did not inhibit the release of VLP of LASV Z protein [20]. ...
Chapter
Nipah virus (NiV) is a bat-borne, highly pathogenic RNA virus belonging to the family Paramyxoviridae. It is known for causing lethal encephalitis in humans with a high fatality rate. With time, the world has faced numerous outbreaks in various regions such as Malaysia, Bangladesh, Philippines, and India. In this chapter, we have summarized experimentally tested antivirals and computational approaches to predict potential inhibitors against NiV. Various studies have been conducted in vitro and in vivo to find the potent novel molecules or repurposed drugs. This section describes the drug’s primary function in case of repurposing, targets, screening method, inhibition efficiency, etc., from important studies. The computational section describes the approaches to identifying the potent inhibitors against NiV. These approaches include machine learning and QSAR-based prediction, molecular docking, molecular dynamics, integrated structure- and network-based approach, Drug–target–drug network-based approach, etc. In conclusion, this work will be helpful for the researchers in examining antivirals against NiV.KeywordsNipah virusAntiviralsInhibitorsComputationalPredictionRepurposed drugs
... Several selective estrogen receptor modulators (SERMs) were found to inhibit EBOV entry by inducing endolysosomal calcium accumulation [85]. BIBX 1382, previously known as a ErbB kinase inhibitor, was also shown to block EBOV entry [86]. ...
Article
Full-text available
Since the 2014–2016 epidemic, Ebola virus (EBOV) has spread to several countries and has become a major threat to global health. EBOV is a risk group 4 pathogen, which imposes significant obstacles for the development of countermeasures against the virus. Efforts have been made to develop anti-EBOV immunization and therapeutics, with three vaccines and two antibody-based therapeutics approved in recent years. Nonetheless, the high fatality of Ebola virus disease highlights the need to continuously develop antiviral strategies for the future management of EBOV outbreaks in conjunction with vaccination programs. This review aims to highlight potential EBOV therapeutics and their target(s) of inhibition, serving as a summary of the literature to inform readers of the novel candidates available in the continued search for EBOV antivirals.
Chapter
Full-text available
Severe fever with thrombocytopenia syndrome virus (SFTSV) also known as Dabie bandavirus of the family Phenuiviridae is a negative-strand RNA virus and a tick-borne virus. Replication of SFTSV into systemic circulation and occurrence of viremia cause cytokine storm and T-cell overstimulation. The event of viremia-induced thrombocytopenia causes reduced platelet count and splenic macrophages, followed by endothelial damages and compromised immune system that cause multi-organ damages. Limited options for specific anti-SFTSV drugs pose significant challenges associated with clinical management of SFTSV infection. This book chapter chiefly emphasizes upon the genetic diversity, geographical distribution, pathogenesis associated with various clinical aspects like symptoms, diagnosis, and available clinical management options. In addition, current research linked with anti-SFTSV drug development is comprehensively portrayed in this review.KeywordsSevere Fever with Thrombocytopenia Syndrome virus (SFTSV)Dabie bandavirusHuaiyangshan BanyangvirusPhenuiviridaeClinical symptoms of SFTSV infectionClinical diagnosis of SFTSVSFTS L proteinSFTSV drug targetMolecular DockingMolecular Dynamics
Article
Seoul virus (SEOV) is an emerging global health threat that can cause hemorrhagic fever with renal syndrome (HFRS), which results in case fatality rates of ∼2%. There are no approved treatments for SEOV infections. We developed a cell-based assay system to identify potential antiviral compounds for SEOV and generated additional assays to characterize the mode of action of any promising antivirals. To test if candidate antivirals targeted SEOV glycoprotein-mediated entry, we developed a recombinant reporter vesicular stomatitis virus expressing SEOV glycoproteins. To facilitate the identification of candidate antiviral compounds targeting viral transcription/replication, we successfully generated the first reported minigenome system for SEOV. This SEOV minigenome (SEOV-MG) screening assay will also serve as a prototype assay for discovery of small molecules inhibiting replication of other hantaviruses, including Andes and Sin Nombre viruses. Ours is a proof-of-concept study in which we tested several compounds previously reported to have activity against other negative-strand RNA viruses using our newly developed hantavirus antiviral screening systems. These systems can be used under lower biocontainment conditions than those needed for infectious viruses, and identified several compounds with robust anti-SEOV activity. Our findings have important implications for the development of anti-hantavirus therapeutics.
Chapter
New antiviral drugs are urgently needed. Recent outbreaks caused by viruses with great epidemiological impact such as Zika, or extraordinary virulence such as Ebola, Nipah, Lassa, Crimean-Congo haemorrhagic fever highlight the current lack of clinically proven vaccines and treatments for these potentially catastrophic agents. Antiviral Discovery for Highly Pathogenic Emerging Viruses comprehensively outlines the state of the art in antiviral drug discovery including identification of targets, screening strategies and the current pipeline of antiviral candidates including regulatory issues. The book also addresses the challenges faced in proceeding from pre-clinical studies to animal models and clinical trials with these highly pathogenic agents. Ideal for drug discovery scientists and medicinal chemists with an interest in antiviral drug discovery and development, this book provides a complete overview of the latest progress in the field, recent advances and the challenges that remain in developing these highly pathogenic agents. Illustrated throughout with case studies this book is a valuable resource in this complex and multidisciplinary field.
Article
Full-text available
Streptococcus pyogenes (group A Streptococcus (GAS) is an important human pathogen that can cause severe invasive infection, such as necrotizing fasciitis and streptococcal toxic shock syndrome. The mortality rate of streptococcal toxic shock syndrome ranges from 20% to 50% in spite of antibiotics administration. AR-12, a pyrazole derivative, has been reported to inhibit the infection of viruses, intracellular bacteria, and fungi. In this report, we evaluated the bactericidal activities and mechanisms of AR-12 on GAS infection. Our in vitro results showed that AR-12 dose-dependently reduced the GAS growth, and 2.5 μg/mL of AR-12 significantly killed GAS within 2 h. AR-12 caused a remarkable reduction in nucleic acid and protein content of GAS. The expression of heat shock protein DnaK and streptococcal exotoxins was also inhibited by AR-12. Surveys of the GAS architecture by scanning electron microscopy revealed that AR-12-treated GAS displayed incomplete septa and micro-spherical structures protruding out of cell walls. Moreover, the combination of AR-12 and gentamicin had a synergistic antibacterial activity against GAS replication for both in vitro and in vivo infection. Taken together, these novel findings obtained in this study may provide a new therapeutic strategy for invasive GAS infection.
Chapter
Arthropod-borne viruses, also known as arbovirusesArbovirus, are transmitted by bites of infected mosquitoMosquitoes or tick vectorsVectors. In this context, the FlavivirusFlavivirus genus is mainly transmitted by mosquitoes from the Aedes genus, being the Ae. africanus, Ae. aegypti, and Ae. AlbopictusAe. Albopictus species are responsible for transmitting the Zika virus (ZIKV). ItZika virus (ZIKV) is a lipid-enveloped virus constitute of an RNARibonucleic acid (RNA)genomeGenome, which is translated into a polyprotein encoding three structural proteinsStructural proteins {(capsid (C), membrane (M), and envelope (E)} and seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5). Several biological targets have been identified for developing antiviralAntivirals agents against ZIKV, which could prevent virus entry, assembly, or release of new virion particles, by interactions with structural proteinsStructural proteins. Drugs targeting non-structural proteins could inhibit the ZIKV-replication cycle. Nature has provided excellent compounds for designing new potent analogs. The medicinal chemistryMedicinal chemistry emerges as an impressive, rationally design and to develop new antiviralAntivirals agents. Additionally, virtual and high-throughput screenings have contributed greatly to the identification of previously approved-drugs that could be repurposed for ZIKV, as antiviralAntivirals agents. In this chapter, we offer a literature review about the most relevant and recent advances made on the medicinal chemistryMedicinal chemistry of ZIKV research. We focus on natural, nature-basedNature-based, semi-synthetic, and synthetic antiviral compounds, as well as repurposed drugs and other inhibitors targeting ZIKV.
Article
Full-text available
The chaperone GRP78 / Dna K is conserved throughout evolution down to prokaryotes. The GRP78 inhibitor OSU-03012 (AR-12) interacted with sildenafil (Viagra) or tadalafil (Cialis) to rapidly reduce GRP78 levels in eukaryotes and as a single agent reduce Dna K levels in prokaryotes. Similar data with the drug combination were obtained for: HSP70, HSP90, GRP94, GRP58, HSP27, HSP40 and HSP60. OSU-03012/sildenafil treatment killed brain cancer stem cells and decreased the expression of: NPC1 and TIM1; LAMP1; and NTCP1, receptors for Ebola / Marburg / Hepatitis A, Lassa fever, and Hepatitis B viruses, respectively. Pre-treatment with OSU-03012/sildenafil reduced expression of the coxsakie and adenovirus receptor in parallel with it also reducing the ability of a serotype 5 adenovirus or coxsakie virus B4 to infect and to reproduce. Similar data were obtained using Chikungunya, Mumps, Measles, Rubella, RSV, CMV and Influenza viruses. OSU-03012 as a single agent at clinically relevant concentrations killed laboratory generated antibiotic resistant E. coli and clinical isolate multi-drug resistant N. gonorrhoeae and MRSE which was in bacteria associated with reduced Dna K and Rec A expression. The PDE5 inhibitors sildenafil or tadalafil enhanced OSU-03012 killing in N. gonorrhoeae and MRSE and low marginally toxic doses of OSU-03012 could restore bacterial sensitivity in N. gonorrhoeae to multiple antibiotics. Thus Dna K and bacterial phosphodiesterases are novel antibiotic targets, and inhibition of GRP78 is of therapeutic utility for cancer and also for bacterial and viral infections. This article is protected by copyright. All rights reserved
Article
Full-text available
Lassa virus spreads from a rodent to humans and can lead to lethal hemorrhagic fever. Despite its broad tropism, chicken cells were reported 30 years ago to resist infection. We found that Lassa virus readily engaged its cell-surface receptor α-dystroglycan in avian cells, but virus entry in susceptible species involved a pH-dependent switch to an intracellular receptor, the lysosome-resident protein LAMP1. Iterative haploid screens revealed that the sialyltransferase ST3GAL4 was required for the interaction of the virus glycoprotein with LAMP1. A single glycosylated residue in LAMP1, present in susceptible species but absent in birds, was essential for interaction with the Lassa virus envelope protein and subsequent infection. The resistance of Lamp1-deficient mice to Lassa virus highlights the relevance of this receptor switch in vivo.
Article
Full-text available
Filoviruses such as Ebola virus and Marburg virus cause a severe haemorrhagic fever syndrome in humans for which there is no specific treatment. Since filoviruses use a complex route of cell entry that depends on numerous cellular factors, we hypothesized that there may be drugs already approved for human use for other indications that interfere with signal transduction or other cellular processes required for their entry and hence have anti-filoviral properties. We used authentic filoviruses and lentiviral particles pseudotyped with filoviral glycoproteins to identify and characterize such compounds. We discovered that amiodarone, a multi-ion channel inhibitor and adrenoceptor antagonist, is a potent inhibitor of filovirus cell entry at concentrations that are routinely reached in human serum during anti-arrhythmic therapy. A similar effect was observed with the amiodarone-related agent dronedarone and the L-type calcium channel blocker verapamil. Inhibition by amiodarone was concentration dependent and similarly affected pseudoviruses as well as authentic filoviruses. Inhibition of filovirus entry was observed with most but not all cell types tested and was accentuated by the pre-treatment of cells, indicating a host cell-directed mechanism of action. The New World arenavirus Guanarito was also inhibited by amiodarone while the Old World arenavirus Lassa and members of the Rhabdoviridae (vesicular stomatitis virus) and Bunyaviridae (Hantaan) families were largely resistant. The ion channel blockers amiodarone, dronedarone and verapamil inhibit filoviral cell entry.
Article
Full-text available
Nipah virus (NiV) outbreaks have occurred in Malaysia, India, and Bangladesh, and the virus continues to cause annual outbreaks of fatal human encephalitis in Bangladesh due to spillover from its bat host reservoir. Due to its high pathogenicity, its potential use for bio/agro-terrorism, and to the current lack of approved therapeutics, NiV is designated as an overlap select agent requiring biosafety level-4 containment. Although the development of therapeutic monoclonal antibodies and soluble protein subunit vaccines have shown great promise, the paucity of effective antiviral drugs against NiV merits further exploration of compound libraries using rapid quantitative antiviral assays. As a proof-of-concept study, we evaluated the use of fluorescent and luminescent reporter NiVs for antiviral screening. We constructed and rescued NiVs expressing either Renilla luciferase or green fluorescent protein, and characterized their reporter signal kinetics in different cell types as well as in the presence of several inhibitors. The 50 percent effective concentrations (EC50s) derived for inhibitors against both reporter viruses are within range of EC50s derived from virus yield-based dose-response assays against wild-type NiV (within 1 Log10), thus demonstrating that both reporter NiVs can serve as robust antiviral screening tools. Utilizing these live NiV-based reporter assays requires modest instrumentation, and circumvents the time and labor-intensive steps associated with cytopathic effect or viral antigen-based assays. These reporter NiVs will not only facilitate antiviral screening, but also the study of host cell components that influence the virus life cycle.
Article
Full-text available
No antiviral therapies are available for the tick-borne flaviviruses associated with hemorrhagic fevers: Kyasanur Forest disease virus (KFDV), both classical and the Alkhurma hemorrhagic fever virus (AHFV) subtype, and Omsk hemorrhagic fever virus (OHFV). We tested compounds reported to have antiviral activity against members of the Flaviviridae family for their ability to inhibit AHFV replication. 6-azauridine (6-azaU), 2' -C-methylcytidine (2' -CMC), and interferon-α2a (IFNα) inhibited the replication of AHFV and also KFDV, OHFV and Powassan virus. The combination of IFNα and 2' -CMC exerted an additive antiviral effect on AHFV and the combination of IFNα and 6-azaU was moderately synergistic. The combination of 2' -CMC and 6-azaU was complex, being strongly synergistic but with a moderate level of antagonism. The antiviral activity of 6-azaU was reduced by the addition of cytidine, but not guanosine, suggesting that it acted by inhibiting pyrimidine biosynthesis. To investigate the mechanism of action of 2' -CMC, AHFV variants with reduced susceptibility to 2' -CMC were selected. We used a replicon system to assess the substitutions present in the selected AHFV population. A double NS5 mutant S603T/C666S and a triple mutant S603T/C666S/M644V were more resistant to 2' -CMC than the wild-type replicon. The S603T/C666S mutant had a reduced level of replication which was increased when M644V was also present, though the replication of this triple mutant was still below that of wild-type. The S603 and C666 residues were predicted to lie in the active site of the AHFV NS5 polymerase, implicating the catalytic center of the enzyme as the binding site for 2' -CMC.
Article
Development of novel strategies targeting the highly virulent ebolaviruses is urgently required. A proteomic study identified the ER chaperone HSPA5 as an ebolavirus-associated host protein. Here, we show using the HSPA5 inhibitor (-)- epigallocatechin gallate (EGCG) that the chaperone is essential for virus infection, thereby demonstrating a functional significance for the association. Furthermore, in vitro and in vivo gene targeting impaired viral replication and protected animals in a lethal infection model. These findings demonstrate that HSPA5 is vital for replication and can serve as a viable target for the design of host-based countermeasures.
Article
Recent investigations have shown the Egyptian fruit bat (Rousettus aegyptiacus) to be a natural reservoir for marburgviruses. To better understand the life cycle of these viruses in the natural host, a new reverse genetics system was developed for the reliable rescue of a Marburg virus (MARV) originally isolated directly from a R. aegyptiacus bat (371Bat). To develop this system, the exact terminal sequences were first determined by 5' and 3' RACE, followed by the cloning of viral proteins NP, VP35, VP30 and L into expression plasmids. Novel conditions were then developed to efficiently replicate virus mini-genomes followed by the construction of full-length genomic clones from which recombinant wild type and GFP-containing MARVs were rescued. Surprisingly, when these recombinant MARVs were propagated in primary human macrophages, a dramatic difference was found in their ability to grow and to elicit anti-viral cytokine responses.
Article
Since their first emergence in mid 1990s henipaviruses continued to re emerge in Australia and South East Asia almost every year. In total there has been more than 8 Nipah and 18 Hendra virus outbreaks reported in South East Asia and Australia, respectively. These outbreaks are associated with significant economic and health damages that most high risks countries (particularly in South East Asia) cannot bear the burden of such economical threats. Up until recently, there were no actual therapeutics available to treat or prevent these lethal infections. However, an international collaborative research has resulted in the identification of a potential equine Hendra vaccine capable of providing antibody protection against Hendra virus infections. Consequently, with the current findings and after nearly 2 decades since their first detection, are we there yet? This review recaps the chronicle of the henipavirus emergence and briefly evaluates potential anti-henipavirus vaccines and antivirals.
Article
OSU-03012 is a celecoxib derivative devoid of cyclooxygenase-2 inhibitory activity. It was previously reported to inhibit the growth of some tumor cells through the AKT-signaling pathway. In the current study, we assessed the ability of OSU-03012 to induce apoptosis in human esophageal carcinoma cells and the mechanism by which this occurs. A cell proliferation assay indicated that OSU-03012 inhibited the growth of human esophageal carcinoma cell lines with an IC50 below 2 μmol/l and had the most effective cytotoxicity against Eca-109 cells. Terminal deoxynucleotidyl transferase-mediated nick-end labeling assay and flow cytometry analysis showed that OSU-03012 could induce the apoptosis in Eca-109 cells. After treatment of Eca-109 cells with 2 μmol/l OSU-03012 for 24 h, the apoptosis index increased from 14.07 to 53.72%. OSU-03012 treatment resulted in a 30-40% decrease in the mitochondrial membrane potential and caused cytochrome c release into the cytosol. Further studies with caspase-9-specific and caspase-8-specific inhibitors (z-LEHDfmk and z-IETDfmk, respectively) pointed toward the involvement of the caspase-9 pathway, but not the caspase-8 pathway, in the execution of OSU-03012-induced apoptosis. Immunoblot analysis demonstrated that OSU-03012-induced cellular apoptosis was associated with upregulation of Bax, cleaved caspase-3, and cleaved caspase-9. Ser-15 of p53 was phosphorylated after 24 h of treatment of the cancer cells with OSU-03012. This increase in p53 was associated with the decrease in Bcl-2 and increase in Bax. An inhibitor of p53, pifithrin-α, attenuated the anticancer effects of OSU-03012 and downregulated the expression of Bax and cleaved caspase-9. Altogether, our results show that OSU-03012 could induce apoptosis in human esophageal carcinoma cells through a p53/Bax/cytochrome c/caspase-9-dependent pathway.