scispace - formally typeset
Search or ask a question
Posted ContentDOI

SARS-CoV-2 sensitive to type I interferon pretreatment.

TL;DR: Evaluating type-I Interferon (IFN-I) sensitivity of SARS-CoV-2 relative to the original SARS -CoV indicates the novel CoV is much more sensitive to IFn-I pretreatment, while the absence of open reading frame (ORF) 3b and significant changes to ORF6 suggest the two key IFN antagonists may not maintain equivalent function in SARS.
Abstract: SARS-CoV-2, a novel coronavirus (CoV), has recently emerged causing an ongoing outbreak of viral pneumonia around the world. While genetically distinct from the original SARS-CoV, both group 2B CoVs share similar genome organization and origins to coronaviruses harbored in bats. Importantly, initial guidance has used insights from SARS-CoV infection to inform treatment and public health strategies. In this report, we evaluate type-I Interferon (IFN-I) sensitivity of SARS-CoV-2 relative to the original SARS-CoV. Our results indicate that while SARS-CoV-2 maintains similar viral replication kinetics to SARS-CoV in Vero cell, the novel CoV is much more sensitive to IFN-I pretreatment. Examining transcriptional factor activation and interferon stimulated gene (ISG) induction, SARS-CoV-2 in the context of type I IFN induces phosphorylation of STAT1 and increased ISG proteins. In contrast, the original SARS-CoV has no evidence for STAT1 phosphorylation or ISG protein increases even in the presence of type I IFN pretreatment. Finally, we examined homology between SARS-CoV and SARS-CoV-2 in viral proteins shown to be interferon antagonist. The absence of open reading frame (ORF) 3b and significant changes to ORF6 suggest the two key IFN antagonists may not maintain equivalent function in SARS-CoV-2. Together, the results identify key differences in susceptibility to the IFN-I response between SARS-CoV and SARS-CoV-2. that could help inform disease progression, treatment options, and animal model development. Importance With the ongoing outbreak of COVID-19 disease, differences between the SARS-CoV-2 and the original SARS-CoV could be leveraged to inform disease progression and eventual treatment options. In addition, these findings could have key implications for animal model development as well as further research into how SARS-CoV-2 modulates the type I IFN response early during infection. Article Summary SARS-CoV-2 has similar replication kinetics to SARS-CoV, but demonstrates significant sensitivity to type I interferon treatment.

Summary (1 min read)

Jump to:  and [Summary]

Summary

  • SARS-CoV-2, a novel coronavirus (CoV) that causes COVID-19, has recently emerged causing 2 an ongoing outbreak of viral pneumonia around the world.
  • 64 In this study, the authors further characterize SARS-CoV-2 and compare it to the original SARS-65 CoV.
  • In contrast, the authors find that SARS-CoV-2 is 67 significantly more sensitive to IFN-I pretreatment as compared to SARS-CoV.
  • These results suggest distinct changes between SARS-74 CoV and SARS-CoV-2 in terms of IFN-I antagonism and the authors subsequently examined sequence 75 homology between the SARS-CoV and SARS-CoV-2 viral proteins that may be responsible for 76 these differences.
  • In contrast, SARS-CoV-2 shows a 98 significant reduction in viral replication following IFN-I treatment.
  • The authors still found a reduction in STAT1 156 phosphorylation and ISG induction in IFN-I pretreated SARS-CoV versus SARS-CoV-2 157 infection.

Did you find this useful? Give us your feedback

Figures (4)

Content maybe subject to copyright    Report

Type I interferon susceptibility distinguishes SARS-CoV-2 from SARS-CoV.
Kumari G. Lokugamage
1*
, Adam Hage
1*
, Maren de Vries
3
, Ana M. Valero-Jimenez
3
, Craig
Schindewolf
1
, Meike Dittmann
3
, Ricardo Rajsbaum
1,2
, Vineet D. Menachery
1,2
1
Department of Microbiology and Immunology,
2
Institute for Human Infection and Immunity,
University of Texas Medical Branch, Galveston TX, USA
3
Department of Microbiology, New York University School of Medicine, New York, NY 10016,
USA
*Equal contributions
Corresponding Author: Vineet D. Menachery
Address: University of Texas Medical Branch, 301 University Blvd, Route #0610 Galveston, TX
77555
Email: Vimenach@utmb.edu
Article Summary: SARS-CoV-2 has similar replication kinetics to SARS-CoV, but
demonstrates significant sensitivity to type I interferon treatment.
Running title: SARS-CoV-2 sensitive to type I IFN pretreatment
Keywords: Coronavirus, 2019-nCoV, SARS-CoV-2, COVID-19, SARS-CoV, type I interferon,
IFN
.CC-BY-NC 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted July 13, 2020. ; https://doi.org/10.1101/2020.03.07.982264doi: bioRxiv preprint

Abstract
1
SARS-CoV-2, a novel coronavirus (CoV) that causes COVID-19, has recently emerged causing
2
an ongoing outbreak of viral pneumonia around the world. While distinct from SARS-CoV, both
3
group 2B CoVs share similar genome organization, origins to bat CoVs, and an arsenal of
4
immune antagonists. In this report, we evaluate type-I interferon (IFN-I) sensitivity of SARS-
5
CoV-2 relative to the original SARS-CoV. Our results indicate that while SARS-CoV-2
6
maintains similar viral replication to SARS-CoV, the novel CoV is much more sensitive to IFN-I.
7
In Vero and in Calu3 cells, SARS-CoV-2 is substantially attenuated in the context of IFN-I
8
pretreatment, while SARS-CoV is not. In line with these findings, SARS-CoV-2 fails to
9
counteract phosphorylation of STAT1 and expression of ISG proteins, while SARS-CoV is able
10
to suppress both. Comparing SARS-CoV-2 and influenza A virus in human airway epithelial
11
cultures (HAEC), we observe the absence of IFN-I stimulation by SARS-CoV-2 alone, but detect
12
failure to counteract STAT1 phosphorylation upon IFN-I pretreatment resulting in near ablation
13
of SARS-CoV-2 infection. Next, we evaluated IFN-I treatment post infection and found SARS-
14
CoV-2 was sensitive even after establishing infection. Finally, we examined homology between
15
SARS-CoV and SARS-CoV-2 in viral proteins shown to be interferon antagonists. The absence
16
of an equivalent open reading frame (ORF) 3b and changes to ORF6 suggest the two key IFN-I
17
antagonists may not maintain equivalent function in SARS-CoV-2. Together, the results identify
18
key differences in susceptibility to IFN-I responses between SARS-CoV and SARS-CoV-2 that
19
may help inform disease progression, treatment options, and animal model development.
20
Importance
21
With the ongoing outbreak of COVID-19, differences between SARS-CoV-2 and the original
22
SARS-CoV could be leveraged to inform disease progression and eventual treatment options.
23
In addition, these findings could have key implications for animal model development as well as
24
further research into how SARS-CoV-2 modulates the type I IFN response early during
25
infection.
26
.CC-BY-NC 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted July 13, 2020. ; https://doi.org/10.1101/2020.03.07.982264doi: bioRxiv preprint

Introduction
27
At the end of 2019, a cluster of patients in Hubei Province, China was diagnosed with a
28
viral pneumonia of unknown origins. With community links to the Huanan seafood market in
29
Wuhan, the disease cluster had echoes of the severe acute respiratory syndrome coronavirus
30
(SARS-CoV) outbreak that emerged at the beginning of the century (1). The 2019 etiologic
31
agent was identified as a novel coronavirus, 2019-nCoV, and subsequently renamed SARS-
32
CoV-2 (2). The new virus has nearly 80% nucleotide identity to the original SARS-CoV and the
33
corresponding CoV disease, COVID-19, has many of the hallmarks of SARS-CoV disease
34
including fever, breathing difficulty, bilateral lung infiltration, and death in the most extreme
35
cases (3, 4). In addition, the most severe SARS-CoV-2 disease corresponded to old age (>50
36
years old), health status, and healthcare workers, similar to both SARS- and MERS-CoV (5).
37
Together, the results indicate SARS-CoV-2 infection and disease have strong similarity to the
38
original SARS-CoV epidemic occurring nearly two decades earlier.
39
In the wake of the outbreak, major research efforts have sought to rapidly characterize
40
the novel CoV to aid in treatment and control. Initial modeling studies predicted (6) and
41
subsequent cell culture studies confirmed that spike protein of SARS-CoV-2 utilizes human
42
angiotensin converting enzyme 2 (ACE2) for entry, the same receptor as SARS-CoV (7, 8).
43
Extensive case studies indicated a similar range of disease onset and severe symptoms seen
44
with SARS-CoV (5). Notably, less severe SARS-CoV-2 cases have also been observed and
45
were not captured in the original SARS-CoV outbreak. Importantly, screening and treatment
46
guidance has relied on previous CoV data generated with SARS-CoV and MERS-CoV.
47
Treatments with both protease inhibitors and type-I interferon (IFN-I) have been employed (4);
48
similarly, remdesivir, a drug targeting viral polymerases, has been reported to have efficacy
49
against SARS-CoV-2 similar to findings with both SARS- and MERS-CoV (9-12). Importantly,
50
several vaccine efforts have been initiated with a focus on the SARS-CoV-2 spike protein as the
51
.CC-BY-NC 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted July 13, 2020. ; https://doi.org/10.1101/2020.03.07.982264doi: bioRxiv preprint

major antigenic determinant (13). Together, the similarities with SARS-CoV have been useful in
52
responding to the newest CoV outbreak.
53
The host innate immune response is initiated when viral products are recognized by host
54
cell pattern recognition receptors, including Toll-like receptors (TLRs) and RIG-I-like receptors
55
(RLRs) (14, 15). This response ultimately results in production of IFN-I and other cytokines,
56
which together are essential for an effective antiviral response (16). IFN-I then triggers its own
57
signaling cascade via its receptor, in an autocrine or paracrine manner, which induces
58
phosphorylation of signal transducers and activators of transcription 1 (STAT1) and STAT2.
59
Together, STAT1, STAT2, and a third transcription factor, IRF9, form the Interferon Stimulated
60
Gene Factor 3 (ISGF3) complex, which is essential for induction of many IFN-stimulated genes
61
(ISGs), and ultimately elicit an effective antiviral response (17, 18). To establish productive
62
replication, viruses have developed different mechanisms to escape this antiviral response
63
targeting different parts of the IFN-I response machinery (19).
64
In this study, we further characterize SARS-CoV-2 and compare it to the original SARS-
65
CoV. Using Vero E6 cells, we demonstrate that SARS-CoV-2 maintains similar viral replication
66
kinetics as SARS-CoV following a low dose infection. In contrast, we find that SARS-CoV-2 is
67
significantly more sensitive to IFN-I pretreatment as compared to SARS-CoV. Infection of IFN-I
68
competent Calu3 2B4 cells resulted in reduced SARS-CoV-2 replication compared to SARS-
69
CoV. Similar to Vero cells, Calu3 cells pretreated with IFN-I had a greater reduction of
70
replication of SARS-CoV-2 compared to SARS-CoV. In human airway epithelial cultures, SARS-
71
CoV-2 showed robust replication and an absence of IFN-I stimulation contrasting influenza A
72
virus. However, pretreatment with IFN-I confirmed SARS-CoV-2 sensitivity and inability to
73
control IFN-I responses once initiated. These results suggest distinct changes between SARS-
74
CoV and SARS-CoV-2 in terms of IFN-I antagonism and we subsequently examined sequence
75
homology between the SARS-CoV and SARS-CoV-2 viral proteins that may be responsible for
76
.CC-BY-NC 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted July 13, 2020. ; https://doi.org/10.1101/2020.03.07.982264doi: bioRxiv preprint

these differences. Together, the results suggest SARS-CoV-2 lacks the same capacity to
77
control the IFN-I response as SARS-CoV.
78
Results
79
SARS-CoV-2 is sensitive to IFN-I pre-treatment
80
Our initial studies infected Vero E6 cells using a low multiplicity of infection (MOI) to
81
explore the viral replication kinetics of SARS-CoV-2 relative to SARS-CoV. Following infection,
82
we found that both SARS-CoV and SARS-CoV-2 replicate with similar kinetics, peaking 48
83
hours post infection (Fig. 1A). While SARS-CoV-2 titer was slightly lower than that of SARS-
84
CoV at 24 hours post infection, the results were not statistically different. By 48 hours,
85
replication of both viruses had plateaued and significant cytopathic effect (CPE) was observed
86
for both SARS-CoV and SARS-CoV-2 infections. Together, the results indicated that SARS-CoV
87
and SARS-CoV-2 replicate with similar replication kinetics in Vero E6 cells.
88
We next evaluated the susceptibility of SARS-CoV-2 to IFN-I pretreatment. Treatment
89
with IFN-I (recombinant IFN-
α
) has been attempted as an antiviral approach for a wide variety of
90
pathogens including hepatitis B and C viruses as well as HIV (20). During both the SARS and
91
MERS-CoV outbreaks, IFN-I has been employed with limited effect (21, 22). In this study, we
92
pretreated Vero E6 cells with 1000 U/mL of recombinant IFN-I (IFN-
α
) 18 hours prior to
93
infection. Vero E6 lack the capacity to produce IFN-I, but are able to respond to exogenous
94
treatment (23). Following pretreatment with IFN-I, SARS-CoV infection has a modest reduction
95
in viral titer of 1.5 log
10
plaque forming units (PFU) as compared to untreated control 24 hours
96
post infection (Fig. 1A). However, by 48 hours, SARS-CoV has nearly equivalent viral yields as
97
the untreated conditions (7.2 log
10
PFU versus 7.5 log
10
PFU). In contrast, SARS-CoV-2 shows a
98
significant reduction in viral replication following IFN-I treatment. At both 24 and 48 hours post
99
infection, SARS-CoV-2 had massive 3-log
10
(24 HPI) and 4-log
10
(48 HPI) drops in viral titer as
100
.CC-BY-NC 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted July 13, 2020. ; https://doi.org/10.1101/2020.03.07.982264doi: bioRxiv preprint

Citations
More filters
Journal ArticleDOI
28 May 2020-Cell
TL;DR: It is proposed that reduced innate antiviral defenses coupled with exuberant inflammatory cytokine production are the defining and driving features of COVID-19.

3,286 citations


Cites result from "SARS-CoV-2 sensitive to type I inte..."

  • ...These data demonstrate that addition of IFN-I resulted in a dramatic reduction in virus replication, in agreement with the findings of others (Lokugamage et al., 2020)....

    [...]

Journal ArticleDOI
Nicolas Vabret1, Graham J. Britton1, Conor Gruber1, Samarth Hegde1, Joel Kim1, Maria Kuksin1, Rachel Levantovsky1, Louise Malle1, Alvaro Moreira1, Matthew D. Park1, Luisanna Pia1, Emma Risson1, Miriam Saffern1, Bérengère Salomé1, Myvizhi Esai Selvan1, Matthew P. Spindler1, Jessica Tan1, Verena van der Heide1, Jill Gregory1, Konstantina Alexandropoulos1, Nina Bhardwaj1, Brian D. Brown1, Benjamin Greenbaum1, Zeynep H. Gümüş1, Dirk Homann1, Amir Horowitz1, Alice O. Kamphorst1, Maria A. Curotto de Lafaille1, Saurabh Mehandru1, Miriam Merad1, Robert M. Samstein1, Manasi Agrawal, Mark Aleynick, Meriem Belabed, Matthew Brown1, Maria Casanova-Acebes, Jovani Catalan, Monica Centa, Andrew Charap, Andrew K Chan, Steven T. Chen, Jonathan Chung, Cansu Cimen Bozkus, Evan Cody, Francesca Cossarini, Erica Dalla, Nicolas F. Fernandez, John A. Grout, Dan Fu Ruan, Pauline Hamon, Etienne Humblin, Divya Jha, Julia Kodysh, Andrew Leader, Matthew Lin, Katherine E. Lindblad, Daniel Lozano-Ojalvo, Gabrielle Lubitz, Assaf Magen, Zafar Mahmood2, Gustavo Martinez-Delgado, Jaime Mateus-Tique, Elliot Meritt, Chang Moon1, Justine Noel, Timothy O'Donnell, Miyo Ota, Tamar Plitt, Venu Pothula, Jamie Redes, Ivan Reyes Torres, Mark P. Roberto, Alfonso R. Sanchez-Paulete, Joan Shang, Alessandra Soares Schanoski, Maria Suprun, Michelle Tran, Natalie Vaninov, C. Matthias Wilk, Julio A. Aguirre-Ghiso, Dusan Bogunovic1, Judy H. Cho, Jeremiah J. Faith, Emilie K. Grasset, Peter S. Heeger, Ephraim Kenigsberg, Florian Krammer1, Uri Laserson1 
16 Jun 2020-Immunity
TL;DR: The current state of knowledge of innate and adaptive immune responses elicited by SARS-CoV-2 infection and the immunological pathways that likely contribute to disease severity and death are summarized.

1,350 citations


Cites background or methods from "SARS-CoV-2 sensitive to type I inte..."

  • ...In support of this notion, SARS-CoV-2 infection fails to limit STAT1 phosphorylation, unlike in SARS-CoV-1 infection (Lokugamage et al., 2020)....

    [...]

  • ...Early evidence demonstrated that SARS-CoV-2 is sensitive to IFN-I/III pretreatment in vitro, perhaps to a greater degree than SARS-CoV-1 (Blanco-Melo et al., 2020; Lokugamage et al., 2020; Mantlo et al., 2020; Stanifer et al., 2020)....

    [...]

Journal ArticleDOI
TL;DR: This study shows that Sars-CoV-2 perturbs host innate immune response via both its structural and nonstructural proteins, and thus provides insights into the pathogenesis of SARS-Cov-2.
Abstract: The pandemic of COVID-19 has posed an unprecedented threat to global public health. However, the interplay between the viral pathogen of COVID-19, SARS-CoV-2, and host innate immunity is poorly understood. Here we show that SARS-CoV-2 induces overt but delayed type-I interferon (IFN) responses. By screening 23 viral proteins, we find that SARS-CoV-2 NSP1, NSP3, NSP12, NSP13, NSP14, ORF3, ORF6 and M protein inhibit Sendai virus-induced IFN-β promoter activation, whereas NSP2 and S protein exert opposite effects. Further analyses suggest that ORF6 inhibits both type I IFN production and downstream signaling, and that the C-terminus region of ORF6 is critical for its antagonistic effect. Finally, we find that IFN-β treatment effectively blocks SARS-CoV-2 replication. In summary, our study shows that SARS-CoV-2 perturbs host innate immune response via both its structural and nonstructural proteins, and thus provides insights into the pathogenesis of SARS-CoV-2.

720 citations

Journal ArticleDOI
TL;DR: The recent progress in the understanding of both type I and type III IFN-mediated innate antiviral responses against human coronaviruses is described and the potential use of IFNs as a treatment strategy for COVID-19 is discussed.

662 citations


Cites background or result from "SARS-CoV-2 sensitive to type I inte..."

  • ...Two in vitro studies have already demonstrated that SARS-CoV-2 has greater sensitivity to IFN-I compared with SARS-CoV (Lokugamage et al., 2020; Mantlo et al., 2020)....

    [...]

  • ...Nsp16 of SARS-CoV-2 and of SARS-CoV share 92% amino acid sequence homology, suggesting this evasion strategy is likely retained in the novel virus (Lokugamage et al., 2020)....

    [...]

Journal ArticleDOI
TL;DR: Insight is provided on SARS-CoV-2 evasion of IFN-I response and its potential impact on viral transmission and pathogenesis.

645 citations

References
More filters
Journal ArticleDOI
TL;DR: The epidemiological, clinical, laboratory, and radiological characteristics and treatment and clinical outcomes of patients with laboratory-confirmed 2019-nCoV infection in Wuhan, China, were reported.

36,578 citations


"SARS-CoV-2 sensitive to type I inte..." refers background or methods in this paper

  • ...Treatments with both protease inhibitors and type I interferon (IFN-I) have been employed (4); 48 similarly, remdesivir, a drug targeting viral polymerases, has been reported to have efficacy 49...

    [...]

  • ...corresponding CoV disease, COVID-19, has many of the hallmarks of SARS-CoV disease 34 including fever, breathing difficulty, bilateral lung infiltration, and death in the most extreme 35 cases (3, 4)....

    [...]

Journal ArticleDOI
TL;DR: Human airway epithelial cells were used to isolate a novel coronavirus, named 2019-nCoV, which formed a clade within the subgenus sarbecovirus, Orthocoronavirinae subfamily, which is the seventh member of the family of coronaviruses that infect humans.
Abstract: In December 2019, a cluster of patients with pneumonia of unknown cause was linked to a seafood wholesale market in Wuhan, China. A previously unknown betacoronavirus was discovered through the use of unbiased sequencing in samples from patients with pneumonia. Human airway epithelial cells were used to isolate a novel coronavirus, named 2019-nCoV, which formed a clade within the subgenus sarbecovirus, Orthocoronavirinae subfamily. Different from both MERS-CoV and SARS-CoV, 2019-nCoV is the seventh member of the family of coronaviruses that infect humans. Enhanced surveillance and further investigation are ongoing. (Funded by the National Key Research and Development Program of China and the National Major Project for Control and Prevention of Infectious Disease in China.).

21,455 citations

Journal ArticleDOI
03 Feb 2020-Nature
TL;DR: Identification and characterization of a new coronavirus (2019-nCoV), which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China, and it is shown that this virus belongs to the species of SARSr-CoV, indicates that the virus is related to a bat coronav virus.
Abstract: Since the outbreak of severe acute respiratory syndrome (SARS) 18 years ago, a large number of SARS-related coronaviruses (SARSr-CoVs) have been discovered in their natural reservoir host, bats1–4. Previous studies have shown that some bat SARSr-CoVs have the potential to infect humans5–7. Here we report the identification and characterization of a new coronavirus (2019-nCoV), which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China. The epidemic, which started on 12 December 2019, had caused 2,794 laboratory-confirmed infections including 80 deaths by 26 January 2020. Full-length genome sequences were obtained from five patients at an early stage of the outbreak. The sequences are almost identical and share 79.6% sequence identity to SARS-CoV. Furthermore, we show that 2019-nCoV is 96% identical at the whole-genome level to a bat coronavirus. Pairwise protein sequence analysis of seven conserved non-structural proteins domains show that this virus belongs to the species of SARSr-CoV. In addition, 2019-nCoV virus isolated from the bronchoalveolar lavage fluid of a critically ill patient could be neutralized by sera from several patients. Notably, we confirmed that 2019-nCoV uses the same cell entry receptor—angiotensin converting enzyme II (ACE2)—as SARS-CoV. Characterization of full-length genome sequences from patients infected with a new coronavirus (2019-nCoV) shows that the sequences are nearly identical and indicates that the virus is related to a bat coronavirus.

16,857 citations


"SARS-CoV-2 sensitive to type I inte..." refers background in this paper

  • ...Coupled with 197 a novel furin cleavage site (33), robust upper airway infection (8), and potential transmission 198...

    [...]

  • ...Initial modeling studies predicted (6) and 41 subsequent cell culture studies confirmed that spike protein of SARS-CoV-2 utilizes human 42 angiotensin converting enzyme 2 (ACE2) for entry, the same receptor as SARS-CoV (7, 8)....

    [...]

  • ...While initial work has suggested incompatibility to SARS-CoV-2 infection in 259 mice based on receptor usage (8), the type I IFN response may be a second major barrier that 260 needs to be overcome....

    [...]

Journal ArticleDOI
07 Apr 2020-JAMA
TL;DR: Hospitalised COVID-19 patients are frequently elderly subjects with co-morbidities receiving polypharmacy, all of which are known risk factors for d
Abstract: Background: Hospitalised COVID-19 patients are frequently elderly subjects with co-morbidities receiving polypharmacy, all of which are known risk factors for d

14,343 citations


"SARS-CoV-2 sensitive to type I inte..." refers result in this paper

  • ...Prophylactic and therapeutic remdesivir (GS-5734) treatment in the 355 rhesus macaque model of MERS-CoV infection....

    [...]

  • ...Importantly, screening and treatment 46 guidance has relied on previous CoV data generated with SARS-CoV and MERS-CoV....

    [...]

  • ...However, each of these 256 MERS-CoV mouse models still retained full immune capacity....

    [...]

  • ...In addition, the most severe SARS-CoV-2 disease corresponded to old age (>50 36 years old), health status, and health care workers, similar to both SARS and MERS-CoV (5)....

    [...]

  • ...This level of 206 sensitivity is similar to MERS-CoV and suggests that the novel CoV lacks the same capacity to 207 escape a primed IFN-I response as SARS-CoV (39, 40)....

    [...]

Journal ArticleDOI
TL;DR: This study evaluated the antiviral efficiency of five FAD-approved drugs including ribavirin, penciclovir, nitazoxanide, nafamostat, chloroquine and two well-known broad-spectrum antiviral drugs remdesivir and favipiravir against a clinical isolate of 2019-nCoV in vitro.
Abstract: Dear Editor, In December 2019, a novel pneumonia caused by a previously unknown pathogen emerged in Wuhan, a city of 11 million people in central China. The initial cases were linked to exposures in a seafood market in Wuhan. As of January 27, 2020, the Chinese authorities reported 2835 confirmed cases in mainland China, including 81 deaths. Additionally, 19 confirmed cases were identified in Hong Kong, Macao and Taiwan, and 39 imported cases were identified in Thailand, Japan, South Korea, United States, Vietnam, Singapore, Nepal, France, Australia and Canada. The pathogen was soon identified as a novel coronavirus (2019-nCoV), which is closely related to sever acute respiratory syndrome CoV (SARS-CoV). Currently, there is no specific treatment against the new virus. Therefore, identifying effective antiviral agents to combat the disease is urgently needed. An efficient approach to drug discovery is to test whether the existing antiviral drugs are effective in treating related viral infections. The 2019-nCoV belongs to Betacoronavirus which also contains SARS-CoV and Middle East respiratory syndrome CoV (MERS-CoV). Several drugs, such as ribavirin, interferon, lopinavir-ritonavir, corticosteroids, have been used in patients with SARS or MERS, although the efficacy of some drugs remains controversial. In this study, we evaluated the antiviral efficiency of five FAD-approved drugs including ribavirin, penciclovir, nitazoxanide, nafamostat, chloroquine and two well-known broad-spectrum antiviral drugs remdesivir (GS5734) and favipiravir (T-705) against a clinical isolate of 2019nCoV in vitro. Standard assays were carried out to measure the effects of these compounds on the cytotoxicity, virus yield and infection rates of 2019-nCoVs. Firstly, the cytotoxicity of the candidate compounds in Vero E6 cells (ATCC-1586) was determined by the CCK8 assay. Then, Vero E6 cells were infected with nCoV2019BetaCoV/Wuhan/WIV04/2019 at a multiplicity of infection (MOI) of 0.05 in the presence of varying concentrations of the test drugs. DMSO was used in the controls. Efficacies were evaluated by quantification of viral copy numbers in the cell supernatant via quantitative real-time RT-PCR (qRT-PCR) and confirmed with visualization of virus nucleoprotein (NP) expression through immunofluorescence microscopy at 48 h post infection (p.i.) (cytopathic effect was not obvious at this time point of infection). Among the seven tested drugs, high concentrations of three nucleoside analogs including ribavirin (half-maximal effective concentration (EC50)= 109.50 μM, halfcytotoxic concentration (CC50) > 400 μM, selectivity index (SI) > 3.65), penciclovir (EC50= 95.96 μM, CC50 > 400 μM, SI > 4.17) and favipiravir (EC50= 61.88 μM, CC50 > 400 μM, SI > 6.46) were required to reduce the viral infection (Fig. 1a and Supplementary information, Fig. S1). However, favipiravir has been shown to be 100% effective in protecting mice against Ebola virus challenge, although its EC50 value in Vero E6 cells was as high as 67 μM, suggesting further in vivo studies are recommended to evaluate this antiviral nucleoside. Nafamostat, a potent inhibitor of MERS-CoV, which prevents membrane fusion, was inhibitive against the 2019-nCoV infection (EC50= 22.50 μM, CC50 > 100 μM, SI > 4.44). Nitazoxanide, a commercial antiprotozoal agent with an antiviral potential against a broad range of viruses including human and animal coronaviruses, inhibited the 2019-nCoV at a low-micromolar concentration (EC50= 2.12 μM; CC50 > 35.53 μM; SI > 16.76). Further in vivo evaluation of this drug against 2019-nCoV infection is recommended. Notably, two compounds remdesivir (EC50= 0.77 μM; CC50 > 100 μM; SI > 129.87) and chloroquine (EC50= 1.13 μM; CC50 > 100 μM, SI > 88.50) potently blocked virus infection at low-micromolar concentration and showed high SI (Fig. 1a, b). Remdesivir has been recently recognized as a promising antiviral drug against a wide array of RNA viruses (including SARS/MERS-CoV) infection in cultured cells, mice and nonhuman primate (NHP) models. It is currently under clinical development for the treatment of Ebola virus infection. Remdesivir is an adenosine analogue, which incorporates into nascent viral RNA chains and results in pre-mature termination. Our time-ofaddition assay showed remdesivir functioned at a stage post virus entry (Fig. 1c, d), which is in agreement with its putative antiviral mechanism as a nucleotide analogue. Warren et al. showed that in NHP model, intravenous administration of 10mg/kg dose of remdesivir resulted in concomitant persistent levels of its active form in the blood (10 μM) and conferred 100% protection against Ebola virus infection. Our data showed that EC90 value of remdesivir against 2019-nCoV in Vero E6 cells was 1.76 μM, suggesting its working concentration is likely to be achieved in NHP. Our preliminary data (Supplementary information, Fig. S2) showed that remdesivir also inhibited virus infection efficiently in a human cell line (human liver cancer Huh-7 cells), which is sensitive to 2019-nCoV. Chloroquine, a widely-used anti-malarial and autoimmune disease drug, has recently been reported as a potential broadspectrum antiviral drug. Chloroquine is known to block virus infection by increasing endosomal pH required for virus/ cell fusion, as well as interfering with the glycosylation of cellular receptors of SARS-CoV. Our time-of-addition assay demonstrated that chloroquine functioned at both entry, and at postentry stages of the 2019-nCoV infection in Vero E6 cells (Fig. 1c, d). Besides its antiviral activity, chloroquine has an immune-modulating activity, which may synergistically enhance its antiviral effect in vivo. Chloroquine is widely distributed in the whole body, including lung, after oral administration. The EC90 value of chloroquine against the 2019-nCoV in Vero

5,660 citations

Related Papers (5)
Frequently Asked Questions (1)
Q1. What are the contributions mentioned in the paper "Type i interferon susceptibility distinguishes sars-cov-2 from sars-cov" ?

In this report, the authors evaluate type-I interferon ( IFN-I ) sensitivity of SARS5 CoV-2 relative to the original SARS-CoV. Finally, the authors examined homology between 15 SARS-CoV and SARS-CoV-2 in viral proteins shown to be interferon antagonists. 26. CC-BY-NC 4. 0 International license available under a was not certified by peer review ) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. The absence 16 of an equivalent open reading frame ( ORF ) 3b and changes to ORF6 suggest the two key IFN-I 17 antagonists may not maintain equivalent function in SARS-CoV-2. In addition, these findings could have key implications for animal model development as well as 24 further research into how SARS-CoV-2 modulates the type I IFN response early during 25 infection.