ArticlePDF Available

Medical Countermeasures Analysis of 2019-nCoV and Vaccine Risks for Antibody-Dependent Enhancement (ADE)

Authors:

Figures

Content may be subject to copyright.
Medical Countermeasures Analysis of 2019-nCoV and Vaccine Risks for Antibody-
dependent Enhancement (ADE)
Darrell O. Ricke, PhD1 & Robert W. Malone2, MD
1Biological and Chemical Technologies
Massachusetts Institute of Technology Lincoln Laboratory
Lexington, MA USA
2Chief'Medical'Officer,'Alchem'Laboratories'
13305'Rachel'Boulevard,'Alachua'FL'32615'USA'
rmalone@alchem.com'
ORCID:'0000-0003-0340-7490'
Corresponding author: Darrell O. Ricke, Ph.D.
Email: Darrell.Ricke@ll.mit.edu
ORCID for Darrell Ricke is 0000-0002-2842-2809
Summary
Background In 80% of patients, COVID-19 presents as mild disease1,2. 20% of cases develop
severe (13%) or critical (6%) illness. More severe forms of COVID-19 present as clinical severe
acute respiratory syndrome, but include a T-predominant lymphopenia3, high circulating levels
of proinflammatory cytokines and chemokines, accumulation of neutrophils and macrophages in
lungs, and immune dysregulation including immunosuppression4.
Methods All major SARS-CoV-2 proteins were characterized using an amino acid residue
variation analysis method. Results predict that most SARS-CoV-2 proteins are evolutionary
constrained, with the exception of the spike (S) protein extended outer surface. Results were
interpreted based on known SARS-like coronavirus virology and pathophysiology, with a focus
on medical countermeasure development implications.
Findings Non-neutralizing antibodies to variable S domains may enable an alternative infection
pathway via Fc receptor-mediated uptake. This may be a gating event for the immune response
dysregulation observed in more severe COVID-19 disease. Prior studies involving vaccine
candidates for FCoV5,6 SARS-CoV-17-10 and Middle East Respiratory Syndrome coronavirus
(MERS-CoV) 11 demonstrate vaccination-induced antibody-dependent enhancement of disease
(ADE), including infection of phagocytic antigen presenting cells (APC). T effector cells are
believed to play an important role in controlling coronavirus infection; pan-T depletion is present
in severe COVID-19 disease3 and may be accelerated by APC infection. Sequence and structural
conservation of S motifs suggests that SARS and MERS vaccine ADE risks may foreshadow
SARS-CoV-2 S-based vaccine risks. Autophagy inhibitors may reduce APC infection and T-cell
depletion12 13. Amino acid residue variation analysis identifies multiple constrained domains
suitable as T cell vaccine targets. Evolutionary constraints on proven antiviral drug targets
present in SARS-CoV-1 and SARS-CoV-2 may reduce risk of developing antiviral drug escape
mutants.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
© 2020 by the author(s). Distributed under a Creative Commons CC BY license.
Interpretation Safety testing of COVID-19 S protein-based B cell vaccines in animal models is
strongly encouraged prior to clinical trials to reduce risk of ADE upon virus exposure.
Introduction
COVID-19 is caused by the SARS-CoV-2 (2019-nCoV) betacoronavirus. The SARS-CoV-2 is a
novel betacoronavirus with sequenced genomes ranging from 29.8k to 29.9k RNA bases. The
SARS-CoV-2 genome encodes replicase proteins, structural proteins, and accessory proteins14
(Table 1). The ORF1a and ORF1ab polyproteins are proteolytically cleaved into 16 non-
structural proteins designated nsp1-1614 (Table 1). Like SARS, COVID-19 manifests as a
virulent zoonotic virus-mediated disease in humans with currently 81,191 confirmed cases and
2,768 deaths as of Feb. 26, 202015.
Zoonotic MERS-CoV, SARS-CoV-1, and SARS-CoV-2 are evolutionarily related, and share
many similarities in human disease characteristics and progression. The mild variant first phase
of viral progression generally presents with mild flu-like symptoms. Most patients never
progress beyond this phase, and typically recover quickly and uneventfully. In a mouse animal
model, phagocytic cells contribute to the antibody-mediated elimination of SARS-CoV-116, and
it may be that innate responses are sufficient to suppress MERS-CoV and SARS-CoV-2 in the
majority of patients. For some individuals (18.5%17), infection progresses to a second severe-
critical variant phase. Progression to the second phase often coincides with the typical timing of
onset of adaptive humoral immunity antibody response (approximately 7-14 days post infection).
MERS-CoV can infect monocyte-derived macrophages (MDMs), monocyte-derived dendric
cells (MoDCs), and T-cells18,19, but the infectivity of SARS-CoV-2 in these cell populations
(with or without non-neutralizing antibody) has not been characterized. For patients with severe
and critical symptoms, the pathophysiology is consistent with increased infection of phagocytic
immune cells (immature MDMs and MoDCs); see Figure 1 for a diagram of the postulated
cascade mechanism. Chemokines released from infected cells may attract additional dendritic
cells and immature macrophages that are susceptible to infection, leading to a possible infection
amplifying cascade of immune cell infection and dysregulation. For some patients with severe
symptoms, excessive activation of macrophages may contribute to a chemokine and cytokine
storm20-22. Individuals with SARS have pronounced peripheral T-cell lymphocytopenia with
reduced CD4+ and CD8+ T-cells23,24, just as is observed with COVID-193. MERS-CoV and
SARS-CoV are also associated with T-cell apoptosis25,26. Infection of macrophages and some T-
cells along with viral dysregulation of cellular pathways result in compromised innate and
humoral immunity in patients during this second and more severe phase of infection 27. High
virus titer in blood plus the possibility of infected immune cell migration throughput the body
may account for the additional disease pathophysiologic and clinical observations observed with
these viruses. MHC I and interleukin (IL)-12 receptor B1 (IL-12RB1) genetic differences
associated with disease progression has been characterized for SARS28-30. Patients with low or
deficient serum levels of the innate immune response pattern recognition molecule mannose-
binding lectin (MBL) have increased frequency in SARS patients versus controls31. MHC
downregulation by epigenetic modifications seen with MERS-CoV infections may enhance
avoidance of T-killer cell responses, and direct infection of some T-cells18 may play a role in
increased mortality rate seen for MERS32. Other disease differences may simply be the different
population of cells with target host receptors angiotensin I converting enzyme 2 (ACE2) for
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
SARS-CoV-1 and SARS-CoV-233, and dipeptidyl peptidase IV (DPP4) for MERS-CoV. ACE2
is expressed in high density in lungs34.
Characterizing variability and evolution of viral proteins must inform medical countermeasure
(MCM) design and development strategies for RNA viruses such as SARS-CoV-2. For viral
progeny, deleterious mutations are rapidly selected against35. Neutral mutations36 provide a
framework for antigenic drift to facilitate escape from immune responses; these residues will
continue to mutate over time. The critical-spacer model proposes that proteins have either amino
acid residue side-chains critical for function or have variable side-chains which may function for
positioning/folding of critical residues37. The divergence model of protein evolution proposes
that the number of critical residues for a protein is consistent for evolutionarily closely related
proteins38. Herein, these concepts are applied to SARS-CoV-2 proteins by leveraging closely
related coronavirus protein sequences to provide insights into viral vulnerabilities that can be
exploited when designing MCMs. The majority of the SARS-CoV-2 proteins exhibit very high
proportions of critical residues to total residues (see Table 2 and Figure 2); hence, these viral
enzymes are excellent small molecule targets. Such small molecule drug therapeutics or
prophylactics have good chances of being effective against SARS-CoV, SARS-CoV-2, and
SARS-like CoVs if they target these highly conserved domains. Non-exposed replicase and
accessory proteins have abundant highly conserved long peptide targets for selecting continuous
segments of critical residues for T-cell epitope vaccines39. In contrast, the extracellular domain
of the S protein exhibits exposed surface areas with high amino acid residue variability.
Increased risk for antibody-dependent enhancement (ADE) from vaccines targeting SARS-CoV-
2, SARS-CoV-1, and MERS-CoV exposed residues is indicated by observed ADE in animal
models and the antibody facilitated infection of phagocytic immune cells frequently observed
with coronaviruses16,40. Peptides and antibodies targeting HR2 and cell fusion have been shown
to block SARS-CoV-1 and MERS-CoV infections in cell lines41-47 and animal models48-50.
Based on the conservation of these domains observed with divergence-based modeling, testing of
similar peptides and antibodies to these targets for SARS-CoV-2 may yield new insights and
opportunities for MCM development. Likewise, drugs that target the phagocytic pathway
associated with Fc-receptor mediated endocytosis are promising candidates for blocking the
cascade of immune cell infections that results in immune dysregulation in COVID-19 patients.
Methods
2019-nCoV protein sequences from GenBank entry MN908947.3 were searched against the non-
redundant (nr) and PDB database using the NCBI BLASTP web interface. Hit protein sequences
were downloaded. Protein multiple sequence alignments were created with the Dawn program51.
Additional 2019-nCoV sequences were added to existing alignments with the Jalview program52.
Identified protein structures were downloaded from RCSB PDB database53. Dawn variation
results were visualized with the Jmol program54.
Role of the funding source
The funder of the study had no role in study design, data collection, data analysis, data
interpretation, or writing of the report. The corresponding authors had full access to all the data
in the study and had final responsibility for the decision to submit for publication.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
Results
Dawn variation results for 2019-nCoV amino acid residues were classified into residues with no
observed variability (candidate critical residues; colored dark green in Figure 2) and to residues
with 5 or more amino acid substitutions (candidate spacer residues; colored dark blue in Figure
2). Amino acids residues colored yellow are considered constrained, allowing only a subset of
possible amino acid substitutions. Amino acid residues with conservative substitutions are also
considered critical residues, and are colored light green in Figure 2; positions with > 95%
conservation of a single residue were included in this category to accommodate potential
sequencing errors and possible adaptative mutations. Twelve of the nsp replicase proteins have a
ratio of critical to total residues of 0.9 or higher (Table 1); this is illustrated in Figure 2 for 2019-
nCoV proteins with high proportions of critical residues in Figure 2 (dark green residues). In
sharp contrast, the S protein exhibits regions of extensive variability of exposed surface residues
(Figure 2).
Discussion
Variation Results
The observed amino acid variations in SARS-CoV-2 proteins are consistent with expected
natural variations in the context of random mutations and selection in the context of host immune
responses. For the nonstructural replicase proteins, the majority have fractions of critical
residues above 88% (Table 2). Long continuous stretches of invariant residues are excellent
candidates for T-cell vaccines epitope selection, and also for exploratory anti-viral small
inhibitory RNA (siRNAs)55 development. With a large RNA genome, the virus has evolved over
time by deleting unnecessary spacer residues. The S protein S1 extended domain shows the
highest number of exposed surface highly variable residues, in sharp contrast to the replicase
enzymes (Figure 2). These spacer residues may function as exposed antigens for antibody
responses with the possible adaptive benefit of suppressing immune responses to less
immunogenic surface antigens. Many of these S protein antigens may lead to non-neutralizing
antibodies. Alternately, evolutionary selection for mutations to these residues may facilitate
antigenic drift to escape immune responses. It seems unusual to have the excessive number of
spacer residues on the S1 extended domain, unless it provides 2019-nCoV with an additional
selective advantage associated with non-neutralizing antibodies bound to this domain.
Coronaviruses have Multiple Options for Cell Infection
The 2019-nCoV S protein contains receptor-binding domains (RBD) targeting human
angiotensin I converting enzyme 2 (ACE2)56,57; this is the initial route for infecting host cells.
To take advantage of antibody responses, coronaviruses also leverage antibody Fc uptake to
infect immune cells58. Coronaviruses use the S protein subunit 2 FP, HR1, and HR2 to infect
immune cells upon proteolytic cleavage of S within endosomes. HR1 and HR2 form a canonical
6-helix bundle involved in membrane fusion41. Jaume et al.58 found that antibody-mediated
infection was dependent on Fc receptor II and not the endosomal/lysosomal pathway utilized by
ACE2 targeting. Viral infection of complement receptor (CR) cells is an additional possible
route of infecting cells59. This multi-pronged approach provides coronaviruses like SARS-CoV-
1, MERS-CoV, and SARS-CoV-2 with more than one mechanism for infecting host cells. This
leads to the hypothesis that antibody mediated uptake of virus is the potential mechanism that
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
induces ADE to vaccines and can also be mediated by maternally transferred antibodies
(matAbs)60-63.
Macrophages and Immune Dysregulation
Lymphopenia is a common feature in patients with SARS23,64 or COVID-1965,66. Two receptors
have been identified for SARS-CoV-1 including ACE267 and C-type lectin domain family 4
member M (CLEC4M, CD209L, CD299, DC-SIGN2, DC-SIGNR, HP10347, and L-SIGN)68
with CLEC4M expressed in human lymph nodes69. Individuals homozygous for CLEC4M
tandem repeats are less susceptible to SARS infection70. In a mouse model, depletion of CD4+ T
cells resulted in an enhanced immune-mediated interstitial pneumonitis when challenged with
SARS-CoV-171. In contrast, depletion of CD4+ and CD8+ T cells as well as antibodies enabled
innate defense mechanisms to control the SARS-CoV-1 virus without immune dysregulation71.
Similar results were also observed in mice with SARS-CoV-1 challenge, but treatment with
liposomes containing clodronate, which deplete alveolar macrophages (AM), prevented immune
deficient virus-specific T cell response72. In a macaque model, anti-spike IgG causes acute lung
injury by skewing macrophage response towards proinflammatory monocyte/macrophage
recruitment and accumulation during acute SARS-CoV-1 infection73. These observations are
likely linked by antibody-dependent enhancement of coronavirus infection of macrophages58,74.
In SARS patients, severe SARS was associated with a more robust IgG response75; early
responders (antibody detectable within 2 weeks) had a higher death rate76,77. The
pathophysiology of severe and critical SARS and COVID-19 diseases fits a proposed model of
antibody-dependent infection of macrophages as the key gate step in disease progression from
mild to severe and critical symptoms, and may explain the observed dysregulated immune
responses78 including apoptosis contributing to development of pan-T cell lymphopenia,
proinflammatory cascade with macrophage accumulation, and cytokine and chemokine
accumulations in lungs with a cytokine storm in some patients.
Vaccine Risks for Antibody-dependent Enhancement (ADE)
Many of the viruses associated with ADE have cell membrane fusion mechanisms61. For
influenza A H1N1, vaccine-induced anti-HA2 antibodies promote virus fusion causing vaccine-
associated enhanced respiratory disease (VAERD)79. ADE was observed for the respiratory
syncytial virus (RSV) in the Bonnet monkey model60. Van Erp et al.60 recommends avoidance of
induction of respiratory syncytial virus (RSV) non-neutralizing antibodies or subneutralizing
antibodies to avoid ADE. In a mouse model, attempts to create vaccines for SARS-CoV-1 lead
to pulmonary immunopathology upon challenge with SARS-CoV-19; these vaccines included
inactivated whole viruses, inactivated viruses with adjuvant, and a recombinant DNA spike (S)
protein vaccine in a virus-like particle (VLP)-based vaccine. Enhanced hepatitis was observed in
a ferret model with a vaccine with recombinant modified vaccinia virus Ankara (rMVA)
expressing the SARS-CoV-1 S protein80. Jaume et al.58 point out the potential pitfalls associated
with immunizations against SARS-CoV-1. This leads to the prediction that new attempts to
create either SARS-CoV-1 vaccines81, MERS-CoV vaccines11, or SARS-CoV-2 vaccines have
potentially higher risks for inducing ADE in humans facilitated by antibody infection of
phagocytic immune cells. This potential ADE risk is independent of the vaccine technology82 or
targeting strategy selected due to predicted phagocytic immune cell infections upon antibody
uptake.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
Convalescent plasma therapy has been provided to SARS83 and COVID-1984 patients. Candidate
patients for convalescent plasma therapy are already experiencing advanced clinical disease
symptoms, potentially mitigating ADE risk. For Hong Kong SARS patients, convalescent
plasma therapy had improved outcomes (6.4% mortality rate) when it was provided before day
14 versus after (21.9% mortality rate) compared to the overall SARS-related mortality rate in of
17%. This is also being seen for initial COVID-19 patients treated with convalescent plasma
therapy84.
Antibody Targets
Analyzing the Cryo-EM structures of MERS-CoV and SARS-CoV-1 spike (S) glycoproteins,
Yuan et al. 85 suggest that the fusion peptide (FP) and the heptad repeat 1 region (HR1) are
potential targets for eliciting broadly neutralizing antibodies based on exposure on the surface of
the stem region, lack of N-linked glycosylation sites in this region, and sequence conservation.
Antibodies that interrupt virus-cell fusion will likely block the infection of immune cells using
Fc-mediated uptake of virus58. This has been demonstrated for SARS-CoV-1 for antibodies to
the HR2 region86-88. Likewise, 2019-nCoV antibodies that block cell fusion are predicted to not
share the same ADE risk of other 2019-nCoV antibodies. Antibodies that target the S RBD89
may have an ADE risk unless combined with a second cell fusion blocking antibody.
Targeting Cell Fusion
In addition to antibodies, peptides targeting HR2 have been shown to effectively block infection
in cell and animal models. Multiple peptides based on the heptad repeat regions (HR1 and HR2)
have been shown to suppress SARS-CoV-1 cell entry42-46. Specific combinations of two
peptides show synergistic viral inhibition43. An HR2 peptide was effective in a mouse model
administered intranasally against human coronavirus 229E (HCoV-229E)48. An HR2 peptide
combined with human interferon-a (IFN-a) also have significant synergistic antiviral effect
against feline coronavirus (FCoV)90. Based on anti-HIV-1 peptide, T-2091, Lambert et al.
demonstrate that analogous peptides inhibit respiratory syncytial virus (RSV), human
parainfluenza virus type 3 (HPIV-3), and measles virus (MV)92. An HR2 peptide can effectively
inhibit MERS-CoV replication49. Gao et al.41 identified an HR2 peptide that inhibits MERS-
CoV fusion in their pseudotyped-virus system. MERS-CoV HR1 entry inhibitor peptides have
been modified to form intra-molecular salt-bridges and increase peptide solubility50. The peptide
MERS HP2P-M2 protected C57BL/6 mice and mice deficient for VDJ recombination-activating
protein 1 (RAG1); this protection was enhanced by combing this peptide with interferon-b50.
Similar results are demonstrated for additional mouse models93,94. Lipopeptides have been
design to target cell fusion peptides47. An analogous fusion inhibitor, enfuvirtide (T-20), has
been approved for treatment of HIV-1 infections91. This provides a path forward for peptide-
based MCMs for 2019-nCoV. A set of SARS-CoV-1 inhibitory peptides that could be adapted
or directly tested on SARS-CoV-2 are illustrated in Figure 3. The SARS-CoV-1 HR2 peptides
can be directly tested on 2019-nCoV without modification due to sequence identity in this region
of the S protein.
B cell Vaccine Designs
B cell vaccines that target the S protein cell fusion mechanisms have the highest chance of
raising neutralizing antibodies with minimal or no ADE risk. Antibodies targeting other portions
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
of the S protein or other 2019-nCoV exposed proteins may enable infection of phagocytic
immune cells even if they are neutralizing.
T cell Vaccine Designs
Variation results identified multiple continuous linear segments of critical residues from which T
cell epitopes can be selected in SARS-CoV-2 replicase enzymes and accessory proteins (Figure
2). Antibodies developed against these epitopes are highly unlikely to enable antibody enhanced
infection of phagocytic immune cells because they are not exposed on the surface of 2019-nCoV.
Targeting Autophagy
Coronavirus replication exploits aspects of normal cellular autophagy95. SKP2 attenuates
autophagy through Beclin1-ubiquination; its inhibition by the licensed drug niclosamide, a
treatment for tapeworms, drastically reduced the replications of MERS-CoV in cell culture13.
Compounds that block autophagy are worth investigating as SARS-CoV-2 MCM.
Targeting Viral Enzymes
2019-nCoV enzyme proteins are highly conserved with minimal spacer residues (Table 2 and
Figure 2). The variation results indicate that available SARS-CoV-1 protein structures (Table 2)
can be directly used for in silico docking and high throughput compound screens. SARS-CoV-2
protein structures are becoming rapidly available96 for compound screening approaches. The
high conservation around enzyme pockets holds promise that compound inhibitors against
SARS-CoV-2 will also be effective against SARS-CoV-1 and SARS-like CoV enzymes.
Summary
Given past data on multiple SARS-CoV-1 and MERS-CoV vaccine efforts which have failed due
to ADE in animal models9,11, it is reasonable to hypothesize a similar ADE risk for SARS-CoV-2
vaccine efforts unless they specifically target domains which will block virus-immune cell
fusion. MCMs based on vaccines, antibodies, or peptides that block cell fusion could minimize
predicted ADE risks. Synergy has been observed for combinations of CoV countermeasures
including interferon-a and -b. Small molecules targeting viral enzymes should also be pursued.
Data Availability
Protein multiple sequence alignments and associated variation files are included in Ricke,
Darrell, 2020, "Medical Countermeasures Analysis of 2019-nCoV / SARS-CoV-2 for COVID-
19", https://doi.org/10.7910/DVN/XWVOA8, Harvard Dataverse, V1.
Acknowledgements
The author acknowledges Nora Smith for literature search assistance, Irene Stapleford for
graphic art assistance, and Dr. Casandra Philipson for proof reading feedback.
Conflicts of Interest
Dr. Ricke and Dr. Malone have nothing to disclose.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
DISTRIBUTION STATEMENT A. Approved for public release. Distribution is unlimited.
This material is based upon work supported under U.S. Air Force Contract No. FA8702-15-D-
0001. Any opinions, findings, conclusions or recommendations expressed in this material are
those of the author(s) and do not necessarily reflect the views of the Under Secretary of Defense
for Research and Engineering.
References
1 Huang, C. et al. Clinical features of patients infected with 2019 novel coronavirus in
Wuhan, China. Lancet 395, 497-506, doi:https://doi.org/10.1016/S0140-6736(20)30183-
5 (2020).
2 Wang, D. et al. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel
Coronavirus–Infected Pneumonia in Wuhan, China. JAMA, doi:10.1001/jama.2020.1585
(2020).
3 Nicholls, J. M. et al. Lung pathology of fatal severe acute respiratory syndrome. Lancet
361, 1773-1778, doi:https://doi.org/10.1016/S0140-6736(03)13413-7 (2003).
4 Zhang, Y. The Epidemiological Characteristics of an Outbreak of 2019 Novel Coronavirus
Diseases (COVID-19) China, 2020,
<http://weekly.chinacdc.cn/en/article/id/e53946e2-c6c4-41e9-9a9b-fea8db1a8f51> (
5 Satoh, R. et al. Screening and identification of T helper 1 and linear immunodominant
antibody-binding epitopes in the spike 2 domain and the nucleocapsid protein of feline
infectious peritonitis virus. Vaccine 29, 1791-1800,
doi:https://doi.org/10.1016/j.vaccine.2010.12.106 (2011).
6 Scott, F. W. Immunization against feline coronaviruses. Adv Exp Med Biol 218, 569-576
(1987).
7 Peiris, J. S. M., Guan, Y. & Yuen, K. Y. Severe acute respiratory syndrome. Nat Med 10,
S88-S97, doi:10.1038/nm1143 (2004).
8 Hui, D. S. C. & Zumla, A. Severe Acute Respiratory Syndrome: Historical, Epidemiologic,
and Clinical Features. Infect Dis Clin North Am 33, 869-889,
doi:https://doi.org/10.1016/j.idc.2019.07.001 (2019).
9 Tseng, C.-T. et al. Immunization with SARS coronavirus vaccines leads to pulmonary
immunopathology on challenge with the SARS virus. PLoS One 7, e35421,
doi:10.1371/journal.pone.0035421 (2012).
10 Wang, S.-F. et al. Antibody-dependent SARS coronavirus infection is mediated by
antibodies against spike proteins. Biochem Biophys Res Commun 451, 208-214,
doi:https://doi.org/10.1016/j.bbrc.2014.07.090 (2014).
11 Agrawal, A. S. et al. Immunization with inactivated Middle East Respiratory Syndrome
coronavirus vaccine leads to lung immunopathology on challenge with live virus. Hum
Vaccin Immunother 12, 2351-2356, doi:10.1080/21645515.2016.1177688 (2016).
12 Yang, Y.-p. et al. Application and interpretation of current autophagy inhibitors and
activators. Acta Pharmacol Sin 34, 625-635, doi:10.1038/aps.2013.5 (2013).
13 Gassen, N. C. et al. SKP2 attenuates autophagy through Beclin1-ubiquitination and its
inhibition reduces MERS-Coronavirus infection. Nat Commun 10, 5770,
doi:10.1038/s41467-019-13659-4 (2019).
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
14 Chen, Y., Liu, Q. & Guo, D. Emerging coronaviruses: Genome structure, replication, and
pathogenesis. J Med Virol, doi:10.1002/jmv.25681 (2020).
15 Coronavirus COVID-19 Global Cases by Johns Hopkins CSSE,
<https://gisanddata.maps.arcgis.com/apps/opsdashboard/index.html#/bda7594740fd4
0299423467b48e9ecf6> (
16 Yasui, F. et al. Phagocytic cells contribute to the antibody-mediated elimination of
pulmonary-infected SARS coronavirus. Virology 454-455, 157-168,
doi:https://doi.org/10.1016/j.virol.2014.02.005 (2014).
17 Zhang, Y. Vital Surveillances: The Epidemiological Characteristics of an Outbreak of 2019
Novel Coronavirus Diseases (COVID-19) China, 2020,
<http://weekly.chinacdc.cn/en/article/id/e53946e2-c6c4-41e9-9a9b-fea8db1a8f51> (
18 Chu, H. et al. Middle East Respiratory Syndrome Coronavirus Efficiently Infects Human
Primary T Lymphocytes and Activates the Extrinsic and Intrinsic Apoptosis Pathways. J
Infect Dis 213, 904-914, doi:10.1093/infdis/jiv380 (2015).
19 Zhou, J., Chu, H., Chan, J. F.-W. & Yuen, K.-Y. Middle East respiratory syndrome
coronavirus infection: virus-host cell interactions and implications on pathogenesis. Virol
J 12, 218, doi:10.1186/s12985-015-0446-6 (2015).
20 Huang, K.-J. et al. An interferon-γ-related cytokine storm in SARS patients. J Med Virol
75, 185-194, doi:10.1002/jmv.20255 (2005).
21 Tisoncik, J. R. et al. Into the eye of the cytokine storm. Microbiol Mol Biol Rev 76, 16-32,
doi:10.1128/MMBR.05015-11 (2012).
22 Channappanavar, R. & Perlman, S. Pathogenic human coronavirus infections: causes and
consequences of cytokine storm and immunopathology. Semin Immunopathol 39, 529-
539, doi:10.1007/s00281-017-0629-x (2017).
23 Wong, R. S. M. et al. Haematological manifestations in patients with severe acute
respiratory syndrome: retrospective analysis. BMJ 326, 1358-1362,
doi:10.1136/bmj.326.7403.1358 (2003).
24 Li, T. et al. Significant Changes of Peripheral T Lymphocyte Subsets in Patients with
Severe Acute Respiratory Syndrome. J Infect Dis 189, 648-651, doi:10.1086/381535
(2004).
25 Yang, Y. et al. Bcl-xL inhibits T-cell apoptosis induced by expression of SARS coronavirus
E protein in the absence of growth factors. Biochem J 392, 135-143,
doi:10.1042/BJ20050698 (2005).
26 Li, G. et al. Coronavirus infections and immune responses. J Med Virol,
doi:10.1002/jmv.25685 (2020).
27 Gu, J. & Korteweg, C. Pathology and pathogenesis of severe acute respiratory syndrome.
Am J Pathol 170, 1136-1147, doi:10.2353/ajpath.2007.061088 (2007).
28 Ng, M. H. L. et al. Association of Human-Leukocyte-Antigen Class I (B*0703) and Class II
(DRB1*0301) Genotypes with Susceptibility and Resistance to the Development of
Severe Acute Respiratory Syndrome. J Infect Dis 190, 515-518, doi:10.1086/421523
(2004).
29 Lin, M. et al. Association of HLA class I with severe acute respiratory syndrome
coronavirus infection. BMC Med Genet 4, 9, doi:10.1186/1471-2350-4-9 (2003).
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
30 Tang, F. et al. IL-12 RB1 genetic variants contribute to human susceptibility to severe
acute respiratory syndrome infection among Chinese. PLoS One 3, e2183,
doi:10.1371/journal.pone.0002183 (2008).
31 Ip, W. K. E. et al. Mannose-Binding Lectin in Severe Acute Respiratory Syndrome
Coronavirus Infection. J Infect Dis 191, 1697-1704, doi:10.1086/429631 (2005).
32 Menachery, V. D. et al. MERS-CoV and H5N1 influenza virus antagonize antigen
presentation by altering the epigenetic landscape. Proc Natl Acad Sci U S A 115, E1012-
E1021, doi:10.1073/pnas.1706928115 (2018).
33 Wan, Y., Shang, J., Graham, R., Baric, R. S. & Li, F. Receptor recognition by novel
coronavirus from Wuhan: An analysis based on decade-long structural studies of SARS. J
Virol, JVI.00127-00120, doi:10.1128/JVI.00127-20 (2020).
34 Zhao, Y. et al. Single-cell RNA expression profiling of ACE2, the putative receptor of
Wuhan 2019-nCov. bioRxiv, doi:10.1101/2020.01.26.919985 (2020).
35 Darwin, C. On the Origin of Species. (1859).
36 Kimura, M. Evolutionary Rate at the Molecular Level. Nature 217, 624-626 (1968).
37 Bottema, C. D. K. et al. Missense mutations and evolutionary conservation of amino
acids: evidence that many of the amino acids in factor IX function as "spacer" elements.
Am J Hum Genet 49, 820-838 (1991).
38 Ricke, D. O. Divergence Model of Protein Evolution. bioRxiv,
doi:http://dx.doi.org/10.1101/045930 (2016).
39 Liu, W. J. et al. T-cell immunity of SARS-CoV: Implications for vaccine development
against MERS-CoV. Antiviral Res 137, 82-92,
doi:https://doi.org/10.1016/j.antiviral.2016.11.006 (2017).
40 Maier, H. J. & Britton, P. Involvement of autophagy in coronavirus replication. Viruses 4,
3440-3451, doi:10.3390/v4123440 (2012).
41 Gao, J. et al. Structure of the fusion core and inhibition of fusion by a heptad repeat
peptide derived from the S protein of Middle East respiratory syndrome coronavirus. J
Virol 87, 13134-13140, doi:10.1128/JVI.02433-13 (2013).
42 Yuan, K. et al. Suppression of SARS-CoV entry by peptides corresponding to heptad
regions on spike glycoprotein. Biochem Biophys Res Commun 319, 746-752,
doi:https://doi.org/10.1016/j.bbrc.2004.05.046 (2004).
43 Liu, I. J. et al. Identification of a minimal peptide derived from heptad repeat (HR) 2 of
spike protein of SARS-CoV and combination of HR1-derived peptides as fusion inhibitors.
Antiviral Res 81, 82-87, doi:https://doi.org/10.1016/j.antiviral.2008.10.001 (2009).
44 Lai, S.-C. et al. Characterization of neutralizing monoclonal antibodies recognizing a 15-
residues epitope on the spike protein HR2 region of severe acute respiratory syndrome
coronavirus (SARS-CoV). J Biomed Sci 12, 711-727, doi:10.1007/s11373-005-9004-3
(2005).
45 Bosch, B. J. et al. Severe acute respiratory syndrome coronavirus (SARS-CoV) infection
inhibition using spike protein heptad repeat-derived peptides. Proc Natl Acad Sci U S A
101, 8455-8460, doi:10.1073/pnas.0400576101 (2004).
46 Zhu, J. et al. Following the rule: formation of the 6-helix bundle of the fusion core from
severe acute respiratory syndrome coronavirus spike protein and identification of
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
potent peptide inhibitors. Biochemical and Biophysical Research Communications 319,
283-288, doi:https://doi.org/10.1016/j.bbrc.2004.04.141 (2004).
47 Wang, C. et al. De Novo Design of α-Helical Lipopeptides Targeting Viral Fusion Proteins:
A Promising Strategy for Relatively Broad-Spectrum Antiviral Drug Discovery. J Med
Chem 61, 8734-8745, doi:10.1021/acs.jmedchem.8b00890 (2018).
48 Xia, S. et al. Peptide-Based Membrane Fusion Inhibitors Targeting HCoV-229E Spike
Protein HR1 and HR2 Domains. Int J Mol Sci 19, 487, doi:10.3390/ijms19020487 (2018).
49 Lu, L. et al. Structure-based discovery of Middle East respiratory syndrome coronavirus
fusion inhibitor. Nat Commun 5, 3067, doi:10.1038/ncomms4067 (2014).
50 Xia, S. et al. Middle East respiratory syndrome coronavirus (MERS-CoV) entry inhibitors
targeting spike protein. Virus Res 194, 200-210,
doi:https://doi.org/10.1016/j.virusres.2014.10.007 (2014).
51 Ricke, D. O. & Shcherbina, A. Dawn: Rapid large-scale protein multiple sequence
alignment and conservation analysis. 2015 IEEE High Performance Extreme Computing
Conference (HPEC), doi:10.1109/HPEC.2015.7322463 (2015).
52 Waterhouse, A. M., Procter, J. B., Martin, D. M. A., Clamp, M. & Barton, G. J. Jalview
Version 2—a multiple sequence alignment editor and analysis workbench.
Bioinformatics 25, 1189-1191, doi:10.1093/bioinformatics/btp033 (2009).
53 Berman, H. M. et al. The Protein Data Bank. Nucleic Acids Res 28, 235-242 (2000).
54 Jmol: an open-source Java viewer for chemical structures in 3D, <www.jmol.org> (
55 Pyrc, K., Berkhout, B. & Hoek, L. v. d. Antiviral Strategies Against Human Coronaviruses.
Infect Disorders - Drug Targets 7, 59-66,
doi:http://dx.doi.org/10.2174/187152607780090757 (2007).
56 Xu, X. et al. Evolution of the novel coronavirus from the ongoing Wuhan outbreak and
modeling of its spike protein for risk of human transmission. Sci China Life Sci,
doi:10.1007/s11427-020-1637-5 (2020).
57 Letko, M. & Munster, V. Functional assessment of cell entry and receptor usage for
lineage B β-coronaviruses, including 2019-nCoV. bioRxiv, 2020.2001.2022.915660,
doi:10.1101/2020.01.22.915660 (2020).
58 Jaume, M. et al. Anti-severe acute respiratory syndrome coronavirus spike antibodies
trigger infection of human immune cells via a pH- and cysteine protease-independent
FcγR pathway. J Virol 85, 10582-10597, doi:10.1128/JVI.00671-11 (2011).
59 Wang, F. S. et al. Acquired but reversible loss of erythrocyte complement receptor 1
(CR1, CD35) and its longitudinal alteration in patients with severe acute respiratory
syndrome. Clin Exp Immunol 139, 112-119, doi:10.1111/j.1365-2249.2005.02681.x
(2005).
60 van Erp, E. A. et al. In Vitro Enhancement of Respiratory Syncytial Virus Infection by
Maternal Antibodies Does Not Explain Disease Severity in Infants. J Virol 91, e00851-
00817, doi:10.1128/JVI.00851-17 (2017).
61 Smatti, M. K., Al Thani, A. A. & Yassine, H. M. Viral-Induced Enhanced Disease Illness.
Front Microbiol 9, 2991, doi:10.3389/fmicb.2018.02991 (2018).
62 Jares Baglivo, S. & Polack, F. P. The long road to protect infants against severe RSV lower
respiratory tract illness. F1000Res 8, F1000 Faculty Rev-1610,
doi:10.12688/f1000research.18749.1 (2019).
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
63 Winarski, K. L. et al. Antibody-dependent enhancement of influenza disease promoted
by increase in hemagglutinin stem flexibility and virus fusion kinetics. Proc Natl Acad Sci
116, 15194, doi:10.1073/pnas.1821317116 (2019).
64 Panesar, N. S. Lymphopenia in SARS. Lancet 361, 1985, doi:10.1016/S0140-
6736(03)13557-X (2003).
65 Xu, Z. et al. Pathological findings of COVID-19 associated with acute respiratory distress
syndrome. Lancet Respir Med, doi:10.1016/S2213-2600(20)30076-X.
66 Guan, W.-j. et al. Clinical characteristics of 2019 novel coronavirus infection in China.
medRxiv, 2020.2002.2006.20020974, doi:10.1101/2020.02.06.20020974 (2020).
67 Li, W. et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS
coronavirus. Nature 426, 450-454, doi:10.1038/nature02145 (2003).
68 Jeffers, S. A. et al. CD209L (L-SIGN) is a receptor for severe acute respiratory syndrome
coronavirus. Proc Natl Acad Sci U S A 101, 15748, doi:10.1073/pnas.0403812101 (2004).
69 Liu, H., Yu, W., Liou, L.-Y. & Rice, A. P. Isolation and characterization of the human DC-
SIGN and DC-SIGNR promoters. Gene 313, 149-159, doi:https://doi.org/10.1016/S0378-
1119(03)00674-7 (2003).
70 Chan, V. S. F. et al. Homozygous L-SIGN (CLEC4M) plays a protective role in SARS
coronavirus infection. Nat Genet 38, 38-46, doi:10.1038/ng1698 (2006).
71 Chen, J. et al. Cellular Immune Responses to Severe Acute Respiratory Syndrome
Coronavirus (SARS-CoV) Infection in Senescent BALB/c Mice: CD4+ T Cells Are Important
in Control of SARS-CoV Infection. J Virol 84, 1289, doi:10.1128/JVI.01281-09 (2010).
72 Zhao, J., Zhao, J., Van Rooijen, N. & Perlman, S. Evasion by stealth: inefficient immune
activation underlies poor T cell response and severe disease in SARS-CoV-infected mice.
PLoS Pathog 5, e1000636, doi:10.1371/journal.ppat.1000636 (2009).
73 Liu, L. et al. Anti-spike IgG causes severe acute lung injury by skewing macrophage
responses during acute SARS-CoV infection. JCI Insight 4, e123158,
doi:10.1172/jci.insight.123158 (2019).
74 Dandekar, A. A. & Perlman, S. Immunopathogenesis of coronavirus infections:
implications for SARS. Nat Rev Immunol 5, 917-927, doi:10.1038/nri1732 (2005).
75 Lee, N. et al. Anti-SARS-CoV IgG response in relation to disease severity of severe acute
respiratory syndrome. J Clin Virol 35, 179-184,
doi:https://doi.org/10.1016/j.jcv.2005.07.005 (2006).
76 Ho, M.-S. et al. Neutralizing Antibody Response and SARS Severity. Emerg Infect Dis,
doi:https://dx.doi.org/10.3201/eid1111.040659 (2005).
77 Zhang, L. et al. Antibody responses against SARS coronavirus are correlated with disease
outcome of infected individuals. J Med Virol 78, 1-8, doi:10.1002/jmv.20499 (2006).
78 Channappanavar, R. et al. Dysregulated Type I Interferon and Inflammatory Monocyte-
Macrophage Responses Cause Lethal Pneumonia in SARS-CoV-Infected Mice. Cell Host
Microbe 19, 181-193, doi:10.1016/j.chom.2016.01.007 (2016).
79 Khurana, S. et al. Vaccine-Induced Anti-HA2 Antibodies Promote Virus Fusion and
Enhance Influenza Virus Respiratory Disease. Sci Transl Med 5, 200ra114,
doi:10.1126/scitranslmed.3006366 (2013).
80 Weingartl, H. et al. Immunization with Modified Vaccinia Virus Ankara-Based
Recombinant Vaccine against Severe Acute Respiratory Syndrome Is Associated with
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
Enhanced Hepatitis in Ferrets. J Virol 78, 12672, doi:10.1128/JVI.78.22.12672-
12676.2004 (2004).
81 Severe Acute Respiratory Syndrome (SARS) Vaccine,
<https://www.bcm.edu/departments/pediatrics/sections-divisions-centers/tropical-
medicine/research/vaccine-development/sarsvaccine> (2020).
82 Rauch, S., Jasny, E., Schmidt, K. E. & Petsch, B. New Vaccine Technologies to Combat
Outbreak Situations. Front Immunol 9, 1963, doi:10.3389/fimmu.2018.01963 (2018).
83 Cheng, Y. et al. Use of convalescent plasma therapy in SARS patients in Hong Kong. Eur J
Clin Microbiol Infect Dis 24, 44-46, doi:10.1007/s10096-004-1271-9 (2005).
84 Ravindranath, P. Convalescent plasma therapy tested on critically ill COVID-19 patients,
<https://journosdiary.com/2020/02/15/convalescent-plasma-therapy-covid-19/> (
85 Yuan, Y. et al. Cryo-EM structures of MERS-CoV and SARS-CoV spike glycoproteins reveal
the dynamic receptor binding domains. Nat Commun 8, 15092,
doi:10.1038/ncomms15092 (2017).
86 Lip, K.-M. et al. Monoclonal Antibodies Targeting the HR2 Domain and the Region
Immediately Upstream of the HR2 of the S Protein Neutralize In Vitro Infection of Severe
Acute Respiratory Syndrome Coronavirus. J Virol 80, 941, doi:10.1128/JVI.80.2.941-
950.2006 (2006).
87 Tripet, B., Kao, D. J., Jeffers, S. A., Holmes, K. V. & Hodges, R. S. Template-based coiled-
coil antigens elicit neutralizing antibodies to the SARS-coronavirus. J Struct Biol 155, 176-
194, doi:https://doi.org/10.1016/j.jsb.2006.03.019 (2006).
88 Keng, E. C. T. et al. Amino Acids 1055 to 1192 in the S2 Region of Severe Acute
Respiratory Syndrome Coronavirus S Protein Induce Neutralizing Antibodies:
Implications for the Development of Vaccines and Antiviral Agents. J Virol 79, 3289-
3296, doi:10.1128/JVI.79.6.3289-3296.2005 (2005).
89 Tian, X. et al. Potent binding of 2019 novel coronavirus spike protein by a SARS
coronavirus-specific human monoclonal antibody. Emerg Microbes Infect 9, 382-385,
doi:10.1080/22221751.2020.1729069 (2020).
90 Liu, I. J., Tsai, W.-T., Hsieh, L.-E. & Chueh, L.-L. Peptides corresponding to the predicted
heptad repeat 2 domain of the feline coronavirus spike protein are potent inhibitors of
viral infection. PLoS One 8, e82081, doi:10.1371/journal.pone.0082081 (2013).
91 Kilby, J. M. et al. Potent suppression of HIV-1 replication in humans by T-20, a peptide
inhibitor of gp41-mediated virus entry. Nat Med 4, 1302-1307, doi:10.1038/3293 (1998).
92 Lambert, D. M. et al. Peptides from conserved regions of paramyxovirus fusion (F)
proteins are potent inhibitors of viral fusion. Proc Natl Acad Sci 93, 2186,
doi:10.1073/pnas.93.5.2186 (1996).
93 Channappanavar, R. et al. Protective Effect of Intranasal Regimens Containing Peptidic
Middle East Respiratory Syndrome Coronavirus Fusion Inhibitor Against MERS-CoV
Infection. J Infect Dis 212, 1894-1903, doi:10.1093/infdis/jiv325 (2015).
94 Jiang, S. et al. Intranasally administered peptidic viral fusion inhibitor protected hDPP4
transgenic mice from MERS-CoV infection. Lancet 386, S44, doi:10.1016/S0140-
6736(15)00625-X (2015).
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
95 Prentice, E., Jerome, W. G., Yoshimori, T., Mizushima, N. & Denison, M. R. Coronavirus
Replication Complex Formation Utilizes Components of Cellular Autophagy. J Biol Chem
279, 10136-10141 (2004).
96 Liu, X., Zhang, B., Jin, Z., Yang, H. & Rao, Z. The crystal structure of COVID-19 main
protease in complex with an inhibitor N3, <http://www.rcsb.org/structure/6LU7>
(2020).
97 Narayanan, K. et al. Severe Acute Respiratory Syndrome Coronavirus nsp1 Suppresses
Host Gene Expression, Including That of Type I Interferon, in Infected Cells. J Virol 82,
4471, doi:10.1128/JVI.02472-07 (2008).
98 Narayanan, K. et al. Severe acute respiratory syndrome coronavirus nsp1 suppresses
host gene expression, including that of type I interferon, in infected cells. J Virol 82,
4471-4479, doi:10.1128/JVI.02472-07 (2008).
99 Serrano, P. et al. Nuclear Magnetic Resonance Structure of the N-Terminal Domain of
Nonstructural Protein 3 from the Severe Acute Respiratory Syndrome Coronavirus. J
Virol 81, 12049, doi:10.1128/JVI.00969-07 (2007).
100 Saikatendu, K. S. et al. Structural Basis of Severe Acute Respiratory Syndrome
Coronavirus ADP-Ribose--Phosphate Dephosphorylation by a Conserved Domain of
nsP3. Structure 13, 1665-1675, doi:https://doi.org/10.1016/j.str.2005.07.022 (2005).
101 Ratia, K. et al. Severe acute respiratory syndrome coronavirus papain-like protease:
structure of a viral deubiquitinating enzyme. Proc Natl Acad Sci U S A 103, 5717-5722,
doi:10.1073/pnas.0510851103 (2006).
102 Beachboard, D. C., Anderson-Daniels, J. M. & Denison, M. R. Mutations across murine
hepatitis virus nsp4 alter virus fitness and membrane modifications. J Virol 89, 2080-
2089, doi:10.1128/JVI.02776-14 (2015).
103 Yang, H. et al. The crystal structures of severe acute respiratory syndrome virus main
protease and its complex with an inhibitor. Proc Natl Acad Sci 100, 13190,
doi:10.1073/pnas.1835675100 (2003).
104 Angelini, M. M., Akhlaghpour, M., Neuman, B. W. & Buchmeier, M. J. Severe acute
respiratory syndrome coronavirus nonstructural proteins 3, 4, and 6 induce double-
membrane vesicles. mBio 4, e00524-00513, doi:10.1128/mBio.00524-13 (2013).
105 Cottam, E. M., Whelband, M. C. & Wileman, T. Coronavirus NSP6 restricts
autophagosome expansion. Autophagy 10, 1426-1441, doi:10.4161/auto.29309 (2014).
106 Kirchdoerfer, R. N. & Ward, A. B. Structure of the SARS-CoV nsp12 polymerase bound to
nsp7 and nsp8 co-factors. Nat Commun 10, 2342, doi:10.1038/s41467-019-10280-3
(2019).
107 Zhai, Y. et al. Insights into SARS-CoV transcription and replication from the structure of
the nsp7–nsp8 hexadecamer. Nat Struct Mol Biol 12, 980-986, doi:10.1038/nsmb999
(2005).
108 Sutton, G. et al. The nsp9 Replicase Protein of SARS-Coronavirus, Structure and
Functional Insights. Structure 12, 341-353, doi:https://doi.org/10.1016/j.str.2004.01.016
(2004).
109 Ma, Y. et al. Structural basis and functional analysis of the SARS coronavirus nsp14-
nsp10 complex. Proc Natl Acad Sci U S A 112, 9436-9441, doi:10.1073/pnas.1508686112
(2015).
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
110 Decroly, E. et al. Crystal Structure and Functional Analysis of the SARS-Coronavirus RNA
Cap 2ʹ-O-Methyltransferase nsp10/nsp16 Complex. PLoS Pathog 7, e1002059,
doi:10.1371/journal.ppat.1002059 (2011).
111 Jia, Z. et al. Delicate structural coordination of the Severe Acute Respiratory Syndrome
coronavirus Nsp13 upon ATP hydrolysis. Nucleic Acids Res 47, 6538-6550,
doi:10.1093/nar/gkz409 (2019).
112 Surya, W., Li, Y. & Torres, J. Structural model of the SARS coronavirus E channel in LMPG
micelles. Biochim Biophys Acta Biomembranes 1860, 1309-1317,
doi:https://doi.org/10.1016/j.bbamem.2018.02.017 (2018).
113 Shi, P. et al. PEDV nsp16 negatively regulates innate immunity to promote viral
proliferation. Virus Res 265, 57-66, doi:https://doi.org/10.1016/j.virusres.2019.03.005
(2019).
114 Wilson, L., Gage, P. & Ewart, G. Hexamethylene amiloride blocks E protein ion channels
and inhibits coronavirus replication. Virology 353, 294-306,
doi:https://doi.org/10.1016/j.virol.2006.05.028 (2006).
115 Li, Y., Surya, W., Claudine, S. & Torres, J. Structure of a conserved Golgi complex-
targeting signal in coronavirus envelope proteins. J Biol Chem 289, 12535-12549,
doi:10.1074/jbc.M114.560094 (2014).
116 Schoeman, D. & Fielding, B. C. Coronavirus envelope protein: current knowledge. Virol J
16, 69, doi:10.1186/s12985-019-1182-0 (2019).
117 Nieto-Torres, J. L. et al. Severe acute respiratory syndrome coronavirus envelope
protein ion channel activity promotes virus fitness and pathogenesis. PLoS Pathog 10,
e1004077, doi:10.1371/journal.ppat.1004077 (2014).
118 Nieto-Torres, J. L. et al. Severe acute respiratory syndrome coronavirus E protein
transports calcium ions and activates the NLRP3 inflammasome. Virology 485, 330-339,
doi:10.1016/j.virol.2015.08.010 (2015).
119 Jimenez-Guardeño, J. M. et al. The PDZ-Binding Motif of Severe Acute Respiratory
Syndrome Coronavirus Envelope Protein Is a Determinant of Viral Pathogenesis. PLoS
Pathog 10, e1004320, doi:10.1371/journal.ppat.1004320 (2014).
120 Jimenez-Guardeño, J. M. et al. Identification of the Mechanisms Causing Reversion to
Virulence in an Attenuated SARS-CoV for the Design of a Genetically Stable Vaccine.
PLoS Pathog 11, e1005215, doi:10.1371/journal.ppat.1005215 (2015).
121 Tseng, Y.-T., Chang, C.-H., Wang, S.-M., Huang, K.-J. & Wang, C.-T. Identifying SARS-CoV
Membrane Protein Amino Acid Residues Linked to Virus-Like Particle Assembly. PLoS
One 8, e64013, doi:10.1371/journal.pone.0064013 (2013).
122 Chen, C.-Y. et al. Structure of the SARS Coronavirus Nucleocapsid Protein RNA-binding
Dimerization Domain Suggests a Mechanism for Helical Packaging of Viral RNA. J Mol
Biol 368, 1075-1086, doi:https://doi.org/10.1016/j.jmb.2007.02.069 (2007).
123 Nelson, C. A., Pekosz, A., Lee, C. A., Diamond, M. S. & Fremont, D. H. Structure and
Intracellular Targeting of the SARS-Coronavirus Orf7a Accessory Protein. Structure 13,
75-85, doi:https://doi.org/10.1016/j.str.2004.10.010 (2005).
124 Hänel, K., Stangler, T., Stoldt, M. & Willbold, D. Solution structure of the X4 protein
coded by the SARS related coronavirus reveals an immunoglobulin like fold and suggests
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
a binding activity to integrin I domains. J Biomed Sci 13, 281-293, doi:10.1007/s11373-
005-9043-9 (2006).
125 Fielding, B. C. et al. Characterization of a unique group-specific protein (U122) of the
severe acute respiratory syndrome coronavirus. J Virol 78, 7311-7318,
doi:10.1128/JVI.78.14.7311-7318.2004 (2004).
126 Arnaout, M. A. Leukocyte Adhesion Molecules Deficiency: Its Structural Basis,
Pathophysiology and Implications for Modulating the Inflammatory Response. Immunol
Rev 114, 145-180, doi:10.1111/j.1600-065X.1990.tb00564.x (1990).
127 Teoh, K.-T. et al. The SARS coronavirus E protein interacts with PALS1 and alters tight
junction formation and epithelial morphogenesis. Mol Biol Cell 21, 3838-3852,
doi:10.1091/mbc.E10-04-0338 (2010).
128 Almeida, M. S., Johnson, M. A., Herrmann, T., Geralt, M. & Wüthrich, K. Novel β-Barrel
Fold in the Nuclear Magnetic Resonance Structure of the Replicase Nonstructural
Protein 1 from the Severe Acute Respiratory Syndrome Coronavirus. J Virol 81, 3151,
doi:10.1128/JVI.01939-06 (2007).
129 Lin, M.-H. et al. Disulfiram can inhibit MERS and SARS coronavirus papain-like proteases
via different modes. Antiviral Res 150, 155-163,
doi:https://doi.org/10.1016/j.antiviral.2017.12.015 (2018).
130 Chen, Y. et al. Biochemical and Structural Insights into the Mechanisms of SARS
Coronavirus RNA Ribose 2ʹ-O-Methylation by nsp16/nsp10 Protein Complex. PLoS
Pathog 7, e1002294, doi:10.1371/journal.ppat.1002294 (2011).
131 Ma, Y. et al. Structural basis and functional analysis of the SARS coronavirus nsp14–
nsp10 complex. Proc Nat Acad Sci U S A 112, 9436, doi:10.1073/pnas.1508686112
(2015).
132 Xu, X. et al. New Antiviral Target Revealed by the Hexameric Structure of Mouse
Hepatitis Virus Nonstructural Protein nsp15. J Virol 80, 7909, doi:10.1128/JVI.00525-06
(2006).
133 Saikatendu, K. S. et al. Ribonucleocapsid Formation of Severe Acute Respiratory
Syndrome Coronavirus through Molecular Action of the N-Terminal Domain of N
Protein. J Virol 81, 3913, doi:10.1128/JVI.02236-06 (2007).
134 Xue, X. et al. Regulator-dependent mechanisms of C3b processing by factor I allow
differentiation of immune responses. Nat Struct Mol Biol 24, 643-651,
doi:10.1038/nsmb.3427 (2017).
135 Wrapp, D. et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion
conformation. Science, eabb2507, doi:10.1126/science.abb2507 (2020).
136 Sainz, B., Jr. et al. Inhibition of severe acute respiratory syndrome-associated
coronavirus (SARS-CoV) infectivity by peptides analogous to the viral spike protein. Virus
Res 120, 146-155, doi:10.1016/j.virusres.2006.03.001 (2006).
137 Liu, S. et al. Interaction between heptad repeat 1 and 2 regions in spike protein of SARS-
associated coronavirus: implications for virus fusogenic mechanism and identification of
fusion inhibitors. Lancet 363, 938-947, doi:https://doi.org/10.1016/S0140-
6736(04)15788-7 (2004).
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
138 Ujike, M. et al. Heptad Repeat-Derived Peptides Block Protease-Mediated Direct Entry
from the Cell Surface of Severe Acute Respiratory Syndrome Coronavirus but Not Entry
via the Endosomal Pathway. J Virol 82, 588, doi:10.1128/JVI.01697-07 (2008).
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
Tables
Table 1. 2019-nCoV proteins*.
Protein
Function
Cofactors
References
nsp1
cellular mRNA degradation, inhibiting type I
interferon (IFN) expression
97,98
nsp2
unknown
nsp3
multidomain protein
nsp3a
interacts with single-stranded RNA
99
nsp3b
ADP-ribose 1"-phosphatase
100
nsp3d
papain-like protease (Plpro), deubiquitinating
enzyme (DUB)
101
nsp4
double-membrane vesicles (DMV) formation
102
nsp5
3C-like protease (3CLpro)
103
nsp6
restricting autophagosome expansion, DMV
formation
104,105
nsp7
RNA binding
nsp8:nsp12
106,107
nsp8
RNA binding; primase
nsp7:nsp12, nsp9
106,107
nsp9
RNA binding, dimerization
nsp8
108
nsp10
scaffold cofactor
nsp10, nsp16
109,110
nsp11
unknown
nsp12
RNA-dependent RNA polymerase (RdRp)
nsp7:nsp8, nsp14
106
nsp13
RNA helicase, 5' triphosphatase
111
nsp14
3'-5' exoribonuclease (ExoN), guanine-N7
methyl transferase (N7-Mtase) for mRNA
capping, nsp12:nsp14 RNA synthesis and
proofreading
109
nsp15
endoribonuclease
112
nsp16
nsp16:nsp10 RNA cap 2'-O-methyltransferase,
negatively regulates innate immunity
110,113
E
forms homopentameric ion channels (IC) with
poor ion selectivity, Golgi complex-targeting
signal, PDZ-binding motif (PBM)
114-120
M
membrane protein
121
N
packages viral RNA
122
ORF3a
ORF6
ORF7a
Ig-like domain, ER retention signal
123-126
ORF7b
ORF8
ORF10
unknown
S
receptor binding, cell fusion
85
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
*The E protein IC releases calcium from the endoplasmic reticulum intermediate compartment
(ERGIC), leading to NLRP3 inflammasome activation117,118. The E protein has a PDZ-binding
motif (PBM)116 that interacts with syntenin PDZ motifs to activate p38 mitogen-activated protein
kinase (MAPK) pathway and promotes an acute proinflammatory response119 and a virus PBM
domain is required for virulence120. The E protein PDZ-binding motif binds to PALS1 and alters
tight junction formation and epithelial morphogenesis127. The envelope (E) protein includes two
pathways to promote inflammation; these may contribute to the ADE response. ORF7a protein
has Ig-like domain123. Hänel et al.124 suggest that this ORF7a possess binding activity for aL
integrin I domain of LFA-1 suggesting that this might block newly synthesized LFA-1 molecules
from reaching the cell surface because ORF7a contains an ER retention signal125. Loss of LFA-1
negatively impacts immune responses126. This suggests possible interference of ORF7a with
immune surveillance mechanisms.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
Table 2. 2019-nCoV Variance Analysis
Protein
V1: Critical
V2
V3
V4
V5+: Spacers
Fraction
Structure
nsp1
112
40
19
3
7
0.84
2GDT:A128
nsp2
279
187
101
46
25
0.73
nsp3
996
514
239
115
92
0.77
2GRI:A99
nsp3a
82
35
20
22
12
0.68
2ACF:A100
Plpro
212
68
24
10
5
0.88
5Y3E:A129
nsp4
337
112
34
13
4
0.90
nsp5
254
46
4
2
0
0.98
6LU796
nsp6
209
64
15
2
0
0.94
nsp7
69
13
1
0
0
0.99
2AHM:A10
7
nsp8
170
26
2
0
0
0.99
2AHM:G10
7
nsp9
95
16
2
0
0
0.98
1UW7:A10
8
nsp10
109
27
3
0
0
0.98
3R24:B130
nsp12
5,226
1374
346
105
50
0.93
nsp13
538
61
2
1
0
1.00
6JYT:A111
nsp14
442
78
7
0
0
0.99
5C8T:B131
nsp15
246
76
17
6
1
0.93
2GTH:A132
nsp16
230
55
8
1
2
0.96
3R24:A130
E
24
33
17
5
3
0.70
5X29:A112
M
178
29
11
4
0
0.93
N
294
76
33
15
4
0.88
2OFZ:A133
ORF3a
107
79
54
20
15
0.68
ORF6
17
21
22
3
0
0.60
ORF7a
55
28
30
10
4
0.65
1XAK:A123
ORF7b
5
33
11
4
1
0.70
ORF8
59
39
15
8
0
0.81
5O32:I134
ORF10
38
0
0
0
0
1.00
S
650
263
123
107
152
0.71
6CRZ:A135
Figures
Figure 1. Disease progression model with normal immune responses during the initial mild
symptoms phase (see 1-3). Antigen presenting cells migrate to the lymph nodes to activate T-
cells (2a). The progression gate to severe and critical disease is the infection of phagocytic
immune cells (3a) leading to immune dysregulation (4b). In the lungs, chemokines attract
additional dendritic cells and immature macrophages that are subsequently infected in an positive
feedback-loop infection cascade (4b). Virus and infected phagocytic immune cells disseminate
throughout the body infecting additional organs (5 & 6). Levels of chemokine and cytokines in
the lungs from infected cells can create a cytokine storm (6).
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
Figure 2. 2019-nCoV variation results. Amino acid residue color code: dark green (critical
residues), light green (critical residues with conservative substitutions or variant in less than 10
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
sequences, yellow (3 variants), light blue (4 variants; likely spacer residues), and blue (5+
variants; spacer residues).
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
nsp1
nsp3
nsp3a
PL-pro protease
nsp5-3CL protease
nsp7 & nsp8
nsp9
nsp10
nsp13
nsp14
nsp15
nsp16
S
E
ORF7a
N
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
Figure 3. SARS-CoV-1 Inhibitory Peptides N4643, HR1-142, HR2-1842, WW-III136, WW-IV136, sHR2-245, sHR2-
845, HRC187, HRC287, CP-1137, SR9138, P643, and CB-11944. SARS-CoV-2 residues different from SARS-CoV-
1 are underlined for adapting SARS-CoV-1 inhibitory peptides.
SARS2 907-NGIGVTQNVLYENQKLIANQFNSAIGKIQDSLSSTASALGKLQDVVNQNAQALNTLVKQ-965
SARS1 889-NGIGVTQNVLYENQKQIANQFNKAISQIQESLTTTSTALGKLQDVVNQNAQALNTLVKQ-947
N46 QKQIANQFNKAISQIQESLTTTSTALGKLQDVVNQNAQALNTLVKQ
HR1-1 NGIGVTQNVLYENQKQIANQFNKAISQIQESLTTTSTA
SARS2 1046-GYHLMSFPQSAPHGVVFLHVTY-1067
SARS1 1028-GYHLMSFPQAAPHGVVFLHVTY-1049
WW-III GYHLMSFPQAAPHGVVFLHVTW
SARS2 1093-GVFVSNGTHWFVTQRNFYE-1111
SARS1 1075-GVFVFNGTSWFITQRNFFS-1093
WW-IV GVFVFNGTSWFITQRNFFS
SARS2 1144-ELDSFKEELDKYFKNHTSPDVDLGDISGINASVVNIQKEIDRLNEVAKNLNESLIDLQELGKYEQYIK-1211
SARS1 1126-ELDSFKEELDKYFKNHTSPDVDLGDISGINASVVNIQKEIDRLNEVAKNLNESLIDLQELGKYEQYIK-1193
sHR2-8 ELDSFKEELDKYFKNHTSPDVDLGDISGINASVVNIQKEIDRLNEVAKNLNESLIDLQELGKYEQYIK
sHR2-2 PKEELDKYFKNHTSPDVDLGDISGINASVVNIQKEIDRLNEVAKNLNESLIDLQELGKYE
HR2-18 IQKEIDRLNEVAKNLNESLIDLQELGK
HRC2 QKEIDRLNEVIKNLNESIIDLQEL
HRC1 NASIVNLQKEIDRLNEVIKNLNES
CP-1 GINASVVNIQKEIDRLNEVAKNLNESLIDLQELGKYE
P6 GINASVVNIQKEIDRLNEVAKNL
SR9 ISGINASVVNIQKEIDRLNEVAKNLNESLIDLQEL
CB-119 SPDVDLGDISGINAS
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 8 March 2020 doi:10.20944/preprints202003.0138.v1
... These findings suggested a T cell-mediated cross-reactivity between circulating 'common cold' coronaviruses and SARS-CoV-2. This cross-reactivity could be responsible for either enhanced protection against COVID-19 (13,14) or, in contrast, for antibody-dependent enhancement (ADE) of COVID-19 (15,16). ...
... This cross-reactivity does not necessarily need to mediate an enhanced protection against COVID-19 (30). In contrast, it may promote ADE of the disease (15), which is often associated with the infection of immune cells and leads to immune cell apoptosis (16). Severe forms of COVID-19 are associated with substantially decreased levels of immune cells (31). ...
Article
Full-text available
The adaptive immune response to severe acute respiratory coronavirus 2 (SARS-CoV-2) is important for vaccine development and in the recovery from coronavirus disease 2019 (COVID-19). Men and cancer patients have been reported to be at higher risks of contracting the virus and developing the more severe forms of COVID-19. Prostate cancer (PCa) may be associated with both of these risks. We show that CD4+ T cells of SARS-CoV-2-unexposed patients with hormone-refractory (HR) metastatic PCa had decreased CD4+ T cell immune responses to antigens from SARS-CoV-2 spike glycoprotein but not from the spiked glycoprotein of the ‘common cold’-associated human coronavirus 229E (HCoV-229E) as compared with healthy male volunteers who responded comparably to both HCoV-229E- and SARS-CoV-2-derived antigens. Moreover, the HCoV-229E spike glycoprotein antigen-elicited CD4+ T cell immune responses cross-reacted with the SARS-CoV-2 spiked glycoprotein antigens. PCa patients may have impaired responses to the vaccination, and the cross-reactivity can mediate antibody-dependent enhancement (ADE) of COVID-19. These findings highlight the potential for increased vulnerability of PCa patients to COVID-19.
... This increase allows them to enter cells that express FcγR. Some have suggested that amino acid diversity and antigen drift due to mutations may lead to antibody-dependent progression (Negro 2020;Ricke and Malone 2020). ...
Article
Full-text available
Coronavirus disease 2019 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We have used bioinformatics to investigate seventeen mutations in the spike protein of SARS-CoV-2, as this mediates infection of human cells and is the target of most vaccine strategies and antibody-based therapies. Two mutations, H146Y and S221W, were identified as being most pathogenic. Mutations at positions D614G, A829T, and P1263L might also have deleterious effects on protein function. We hypothesized that candidate small molecules may be repurposed to combat viral infection. We investigated changes in binding energies of the ligands and the mutant proteins by assessing molecular docking. For an understanding of cellular function and organization, protein–protein interactions are also critical. Protein–protein docking for naïve and mutated structures of SARS-CoV-2 S protein was evaluated for their binding energy with the angiotensin-converting enzyme 2 (ACE2). These interactions might limit the binding of the SARS-CoV-2 spike protein to the ACE2 receptor or may have a deleterious effect on protein function that may limit infection. These results may have important implications for the transmission of SARS-CoV-2, its pathogenesis, and the potential for drug repurposing and immune therapies.
... This cross-reactivity does not necessarily need to mediate an enhanced protection against COVID-19 (DiPiazza et al., 2016). In contrast, it may promote ADE of the disease (Hotez et al., 2020;Iwasaki and Yang, 2020;Tetro, 2020), which is often associated with the infection of immune cells and leads to immune cell apoptosis (Ricke, 2020). Severe forms of COVID-19 are associated with substantially decreased levels of immune cells (Cao, 2020;Qin et al., 2020). ...
Preprint
Full-text available
The adaptive immune response to severe acute respiratory coronavirus 2 (SARS-CoV-2) is important for vaccine development and in the recovery from coronavirus disease 2019 (COVID-19). Men and cancer patients have been reported to be at higher risks of contracting the virus and developing the more severe forms of COVID-19. Prostate cancer (PCa) may be associated with both of these risks. We show that CD4 ⁺ T cells of SARS-CoV-2-unexposed patients with hormone-refractory (HR) metastatic PCa had decreased CD4 ⁺ T cell immune responses to antigens from SARS-CoV-2 spike glycoprotein but not from the spiked glycoprotein of the 'common cold'-associated human coronavirus 229E (HCoV-229E) as compared with healthy male volunteers who responded comparably to both HCoV-229E- and SARS-CoV-2-derived antigens. Moreover, the HCoV-229E spike glycoprotein antigen-elicited CD4 ⁺ T cell immune responses cross-reacted with the SARS-CoV-2 spiked glycoprotein antigens. PCa patients may have impaired responses to the vaccination, and the cross-reactivity can mediate antibody-dependent enhancement (ADE) of COVID-19. These findings highlight the potential for increased vulnerability of PCa patients to COVID-19.
... These preparations also did not interfere with the SARS-CoV-2 neutralising capacity of serum IgG. This latter result suggests that IVIG batches manufactured after the COVID-19 outbreak should not exclude donors that have recovered from COVID-19, provided that they do not present potentially deleterious anti-self-reactivity or antibodydependent SARS-CoV-2 enhancement activity (24). It is nevertheless important to consider that IVIG infusions were reported to be ineffective in non-COVID-19-related SARS (25). ...
Article
Full-text available
The capacity of pre-existing immunity to human common coronaviruses (HCoV) to cross-protect against de novo COVID-19is yet unknown. In this work, we studied the sera of 175 COVID-19 patients, 76 healthy donors and 3 intravenous immunoglobulins (IVIG) batches. We found that most COVID-19 patients developed anti-SARS-CoV-2 IgG antibodies before IgM. Moreover, the capacity of their IgGs to react to beta-HCoV, was present in the early sera of most patients before the appearance of anti-SARS-CoV-2 IgG. This implied that a recall-type antibody response was generated. In comparison, the patients that mounted an anti-SARS-COV2 IgM response, prior to IgG responses had lower titres of anti-beta-HCoV IgG antibodies. This indicated that pre-existing immunity to beta-HCoV was conducive to the generation of memory type responses to SARS-COV-2. Finally, we also found that pre-COVID-19-era sera and IVIG cross-reacted with SARS-CoV-2 antigens without neutralising SARS-CoV-2 infectivity in vitro . Put together, these results indicate that whilst pre-existing immunity to HCoV is responsible for recall-type IgG responses to SARS-CoV-2, it does not lead to cross-protection against COVID-19.
... 10 It is likely to occur when antibody levels or specificities do not permit neutralisation. 11 For these reasons, it is important to determine neutralising antibody titres in donated plasma, as well as a practical cut-off titre level, to evaluate not only its safety but also its effectiveness for convalescent plasma transfusion. ...
Article
Full-text available
Introduction The lack of approved specific therapeutic agents to treat coronavirus disease (COVID‐19) associated with severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infection has led to the rapid implementation of convalescent plasma therapy (CPT) trials in many countries, including the United Kingdom. Effective CPT is likely to require high titres of neutralising antibody (nAb) in convalescent donations. Understanding the relationship between functional neutralising antibodies and antibody levels to specific SARS‐CoV‐2 proteins in scalable assays will be crucial for the success of a large‐scale collection. We assessed whether neutralising antibody titres correlated with reactivity in a range of enzyme‐linked immunosorbent assays (ELISA) targeting the spike (S) protein, the main target for human immune response. Methods Blood samples were collected from 52 individuals with a previous laboratory‐confirmed SARS‐CoV‐2 infection. These were assayed for SARS‐CoV‐2 nAbs by microneutralisation and pseudo‐type assays and for antibodies by four different ELISAs. Receiver operating characteristic (ROC) analysis was used to further identify sensitivity and specificity of selected assays to identify samples containing high nAb levels. Results All samples contained SARS‐CoV‐2 antibodies, whereas neutralising antibody titres of greater than 1:20 were detected in 43 samples (83% of those tested) and >1:100 in 22 samples (42%). The best correlations were observed with EUROimmun immunoglobulin G (IgG) reactivity (Spearman Rho correlation coefficient 0.88; p < 0.001). Based on ROC analysis, EUROimmun would detect 60% of samples with titres of >1:100 with 100% specificity using a reactivity index of 9.1 (13/22). Discussion Robust associations between nAb titres and reactivity in several ELISA‐based antibody tests demonstrate their possible utility for scaled‐up production of convalescent plasma containing potentially therapeutic levels of anti‐SARS‐CoV‐2 nAbs.
Article
Background: Convalescent plasma may reduce mortality in patients with viral respiratory diseases, and is being investigated as a potential therapy for coronavirus disease 2019 (COVID-19). A thorough understanding of the current body of evidence regarding benefits and risks of this intervention is required. Objectives: To assess the effectiveness and safety of convalescent plasma transfusion in the treatment of people with COVID-19; and to maintain the currency of the evidence using a living systematic review approach. Search methods: To identify completed and ongoing studies, we searched the World Health Organization (WHO) COVID-19 Global literature on coronavirus disease Research Database, MEDLINE, Embase, Cochrane COVID-19 Study Register, and the Epistemonikos COVID-19 L*OVE Platform. We searched monthly until 03 March 2022. Selection criteria: We included randomised controlled trials (RCTs) evaluating convalescent plasma for COVID-19, irrespective of disease severity, age, gender or ethnicity. We excluded studies that included populations with other coronavirus diseases (severe acute respiratory syndrome (SARS) or Middle East respiratory syndrome (MERS)), as well as studies evaluating standard immunoglobulin. Data collection and analysis: We followed standard Cochrane methodology. To assess bias in included studies we used RoB 2. We used the GRADE approach to rate the certainty of evidence for the following outcomes: all-cause mortality at up to day 28, worsening and improvement of clinical status (for individuals with moderate to severe disease), hospital admission or death, COVID-19 symptoms resolution (for individuals with mild disease), quality of life, grade 3 or 4 adverse events, and serious adverse events. Main results: In this fourth review update version, we included 33 RCTs with 24,861 participants, of whom 11,432 received convalescent plasma. Of these, nine studies are single-centre studies and 24 are multi-centre studies. Fourteen studies took place in America, eight in Europe, three in South-East Asia, two in Africa, two in western Pacific and three in eastern Mediterranean regions and one in multiple regions. We identified a further 49 ongoing studies evaluating convalescent plasma, and 33 studies reporting as being completed. Individuals with a confirmed diagnosis of COVID-19 and moderate to severe disease 29 RCTs investigated the use of convalescent plasma for 22,728 participants with moderate to severe disease. 23 RCTs with 22,020 participants compared convalescent plasma to placebo or standard care alone, five compared to standard plasma and one compared to human immunoglobulin. We evaluate subgroups on detection of antibodies detection, symptom onset, country income groups and several co-morbidities in the full text. Convalescent plasma versus placebo or standard care alone Convalescent plasma does not reduce all-cause mortality at up to day 28 (risk ratio (RR) 0.98, 95% confidence interval (CI) 0.92 to 1.03; 220 per 1000; 21 RCTs, 19,021 participants; high-certainty evidence). It has little to no impact on need for invasive mechanical ventilation, or death (RR 1.03, 95% CI 0.97 to 1.11; 296 per 1000; 6 RCTs, 14,477 participants; high-certainty evidence) and has no impact on whether participants are discharged from hospital (RR 1.00, 95% CI 0.97 to 1.02; 665 per 1000; 6 RCTs, 12,721 participants; high-certainty evidence). Convalescent plasma may have little to no impact on quality of life (MD 1.00, 95% CI -2.14 to 4.14; 1 RCT, 483 participants; low-certainty evidence). Convalescent plasma may have little to no impact on the risk of grades 3 and 4 adverse events (RR 1.17, 95% CI 0.96 to 1.42; 212 per 1000; 6 RCTs, 2392 participants; low-certainty evidence). It has probably little to no effect on the risk of serious adverse events (RR 1.14, 95% CI 0.91 to 1.44; 135 per 1000; 6 RCTs, 3901 participants; moderate-certainty evidence). Convalescent plasma versus standard plasma We are uncertain whether convalescent plasma reduces or increases all-cause mortality at up to day 28 (RR 0.73, 95% CI 0.45 to 1.19; 129 per 1000; 4 RCTs, 484 participants; very low-certainty evidence). We are uncertain whether convalescent plasma reduces or increases the need for invasive mechanical ventilation, or death (RR 5.59, 95% CI 0.29 to 108.38; 311 per 1000; 1 study, 34 participants; very low-certainty evidence) and whether it reduces or increases the risk of serious adverse events (RR 0.80, 95% CI 0.55 to 1.15; 236 per 1000; 3 RCTs, 327 participants; very low-certainty evidence). We did not identify any study reporting other key outcomes. Convalescent plasma versus human immunoglobulin Convalescent plasma may have little to no effect on all-cause mortality at up to day 28 (RR 1.07, 95% CI 0.76 to 1.50; 464 per 1000; 1 study, 190 participants; low-certainty evidence). We did not identify any study reporting other key outcomes. Individuals with a confirmed diagnosis of SARS-CoV-2 infection and mild disease We identified two RCTs reporting on 536 participants, comparing convalescent plasma to placebo or standard care alone, and two RCTs reporting on 1597 participants with mild disease, comparing convalescent plasma to standard plasma. Convalescent plasma versus placebo or standard care alone We are uncertain whether convalescent plasma reduces all-cause mortality at up to day 28 (odds ratio (OR) 0.36, 95% CI 0.09 to 1.46; 8 per 1000; 2 RCTs, 536 participants; very low-certainty evidence). It may have little to no effect on admission to hospital or death within 28 days (RR 1.05, 95% CI 0.60 to 1.84; 117 per 1000; 1 RCT, 376 participants; low-certainty evidence), on time to COVID-19 symptom resolution (hazard ratio (HR) 1.05, 95% CI 0.85 to 1.30; 483 per 1000; 1 RCT, 376 participants; low-certainty evidence), on the risk of grades 3 and 4 adverse events (RR 1.29, 95% CI 0.75 to 2.19; 144 per 1000; 1 RCT, 376 participants; low-certainty evidence) and the risk of serious adverse events (RR 1.14, 95% CI 0.66 to 1.94; 133 per 1000; 1 RCT, 376 participants; low-certainty evidence). We did not identify any study reporting other key outcomes. Convalescent plasma versus standard plasma We are uncertain whether convalescent plasma reduces all-cause mortality at up to day 28 (OR 0.30, 95% CI 0.05 to 1.75; 2 per 1000; 2 RCTs, 1597 participants; very low-certainty evidence). It probably reduces admission to hospital or death within 28 days (RR 0.49, 95% CI 0.31 to 0.75; 36 per 1000; 2 RCTs, 1595 participants; moderate-certainty evidence). Convalescent plasma may have little to no effect on initial symptom resolution at up to day 28 (RR 1.12, 95% CI 0.98 to 1.27; 1 RCT, 416 participants; low-certainty evidence). We did not identify any study reporting other key outcomes. This is a living systematic review. We search monthly for new evidence and update the review when we identify relevant new evidence. Authors' conclusions: For the comparison of convalescent plasma versus placebo or standard care alone, our certainty in the evidence that convalescent plasma for individuals with moderate to severe disease does not reduce mortality and has little to no impact on clinical improvement or worsening is high. It probably has little to no effect on SAEs. For individuals with mild disease, we have low certainty evidence for our primary outcomes. There are 49 ongoing studies, and 33 studies reported as complete in a trials registry. Publication of ongoing studies might resolve some of the uncertainties around convalescent plasma therapy for people with asymptomatic or mild disease.
Preprint
Full-text available
*** THIS PAPER HAS BEEN PLACED HERE FOR PUBLIC PEER-REVIEW *** *** After public peer-review an attempt will be made for journal submission, any suggestions for interested journals are welcome. *** *** All comments, corrections, questions, criticism, or other feedback can be emailed to the author, or placed under Comments below. Thank you all in advance for your help in assessing and improving this study's methods and design.*** Policy makers and mainstream news anchors have promised the public that the COVID-19 vaccine rollout worldwide would reduce symptoms, and thereby cases and deaths associated with COVID-19. While this vaccine rollout is still in progress, there is a large amount of public data available that permits an analysis of the effect of the vaccine rollout on COVID-19 related cases and deaths. Has this public policy treatment produced the desired effect? One manner to respond to this question can begin by implementing a Bayesian causal analysis comparing both pre- and post-treatment periods. This study analyzed publicly available COVID-19 data from OWID utlizing the R package CausalImpact to determine the causal effect of the administration of vaccines on two dependent variables that have been measured cumulatively throughout the pandemic: total deaths per million (y1) and total cases per million (y2). After eliminating all results from countries with p > 0.05, there were 128 countries for y1 and 103 countries for y2 to analyze in this fashion, comprising 145 unique countries in total (avg. p < 0.004). Results indicate that the treatment (vaccine administration) has a strong and statistically significant propensity to causally increase the values in either y1 or y2 over and above what would have been expected with no treatment. y1 showed an increase/decrease ratio of (+115/-13), which means 89.84% of statistically significant countries showed an increase in total deaths per million associated with COVID-19 due directly to the causal impact of treatment initiation. y2 showed an increase/decrease ratio of (+105/-16) which means 86.78% of statistically significant countries showed an increase in total cases per million of COVID-19 due directly to the causal impact of treatment initiation. Causal impacts of the treatment on y1 ranges from -19% to +19015% with an average causal impact of +463.13%. Causal impacts of the treatment on y2 ranges from -46% to +12240% with an average causal impact of +260.88%. Hypothesis 1 Null can be rejected for a large majority of countries. This study subsequently performed correlational analyses on the causal impact results, whose effect variables can be represented as y1.E and y2.E respectively, with the independent numeric variables of: days elapsed since vaccine rollout began (n1), total vaccination doses per hundred (n2), total vaccine brands/types in use (n3) and the independent categorical variables continent (c1), country (c2), vaccine variety (c3). All categorical variables showed statistically significant (avg. p: < 0.001) postive Wilcoxon signed rank values (y1.E V:[c1 3.04; c2: 8.35; c3: 7.22] and y2.E V:[c1 3.04; c2: 8.33; c3: 7.19]). This demonstrates that the distribution of y1.E and y2.E was non-uniform among categories. The Spearman correlation between n2 and y2.E was the only numerical variable that showed statistically significant results (y2.E ~ n2: rho: 0.34 CI95%[0.14, 0.51], p: 4.91e-04). This low positive correlation signifies that countries with higher vaccination rates do not have lower values for y2.E, slightly the opposite in fact. Still, the specifics of the reasons behind these differences between countries, continents, and vaccine types is inconclusive and should be studied further as more data become available. Hypothesis 2 Null can be rejected for c1, c2, c3 and n2 and cannot be rejected for n1, and n3. The statistically significant and overwhelmingly positive causal impact after vaccine deployment on the dependent variables total deaths and total cases per million should be highly worrisome for policy makers. They indicate a marked increase in both COVID-19 related cases and death due directly to a vaccine deployment that was originally sold to the public as the “key to gain back our freedoms.” The effect of vaccines on total cases per million and its low positive association with total vaccinations per hundred signifies a limited impact of vaccines on lowering COVID-19 associated cases. These results should encourage local policy makers to make policy decisions based on data, not narrative, and based on local conditions, not global or national mandates. These results should also encourage policy makers to begin looking for other avenues out of the pandemic aside from mass vaccination campaigns. Some variables that could be included in future analyses might include vaccine lot by country, the degree of prevalence of previous antibodies against SARS-CoV or SARS-CoV-2 in the population before vaccine administration begins, and the Causal Impact of ivermectin on the same variables used in this study.
Article
Full-text available
Coronaviridae is a typical type of viral family comprised of an exceptionally huge RNA genome with a characteristic appearance and a noteworthy propensity to move from creatures to people. Since the start of the 21st century, three exceptionally contagious and pathogenic coronaviruses have traversed the species boundary and caused lethal pneumonia, exacting serious episodes, and causing human wellbeing crises to an unfathomable extent. The COVID-19 pandemic, presumably, is the most wrecking one, over the most recent 100 years after Spanish influenza. To the quick assessment of the various methodologies for a capability to evoke defensive resistance and well-being to shorten undesirable resistant potentiation, which assumes a significant function in this virus's pathogenesis. Considering the forthcoming public health crisis, researchers around the globe are consolidating their logical scientific possessions and offering initial information in an unprecedented way. In this review, we have tried to summarize some of the SAR-nCoV2 treatments that essentially focus on the potential drugs including Lopinavir/Ritonavir, Favipiravir, Remdesivir, Chloroquine phosphate, Hydroxychloroquine, Arbidol, and so on. Other potential vaccine developments with their difficulties associated and their current status are also being addressed.
Article
Background: Convalescent plasma and hyperimmune immunoglobulin may reduce mortality in patients with viral respiratory diseases, and are being investigated as potential therapies for coronavirus disease 2019 (COVID-19). A thorough understanding of the current body of evidence regarding benefits and risks of these interventions is required. OBJECTIVES: Using a living systematic review approach, to assess whether convalescent plasma or hyperimmune immunoglobulin transfusion is effective and safe in the treatment of people with COVID-19; and to maintain the currency of the evidence. Search methods: To identify completed and ongoing studies, we searched the World Health Organization (WHO) COVID-19 Global literature on coronavirus disease Research Database, MEDLINE, Embase, the Cochrane COVID-19 Study Register, the Epistemonikos COVID-19 L*OVE Platform, and trial registries. Searches were done on 17 March 2021. Selection criteria: We included randomised controlled trials (RCTs) evaluating convalescent plasma or hyperimmune immunoglobulin for COVID-19, irrespective of disease severity, age, gender or ethnicity. For safety assessments, we also included non-controlled non-randomised studies of interventions (NRSIs) if 500 or more participants were included. We excluded studies that included populations with other coronavirus diseases (severe acute respiratory syndrome (SARS) or Middle East respiratory syndrome (MERS)), as well as studies evaluating standard immunoglobulin. Data collection and analysis: We followed standard Cochrane methodology. To assess bias in included studies, we used the Cochrane 'Risk of Bias 2' tool for RCTs, and for NRSIs, the assessment criteria for observational studies, provided by Cochrane Childhood Cancer. We rated the certainty of evidence, using the GRADE approach, for the following outcomes: all-cause mortality, improvement and worsening of clinical status (for individuals with moderate to severe disease), development of severe clinical COVID-19 symptoms (for individuals with asymptomatic or mild disease), quality of life (including fatigue and functional independence), grade 3 or 4 adverse events, and serious adverse events. Main results: We included 13 studies (12 RCTs, 1 NRSI) with 48,509 participants, of whom 41,880 received convalescent plasma. We did not identify any completed studies evaluating hyperimmune immunoglobulin. We identified a further 100 ongoing studies evaluating convalescent plasma or hyperimmune immunoglobulin, and 33 studies reporting as being completed or terminated. Individuals with a confirmed diagnosis of COVID-19 and moderate to severe disease Eleven RCTs and one NRSI investigated the use of convalescent plasma for 48,349 participants with moderate to severe disease. Nine RCTs compared convalescent plasma to placebo treatment or standard care alone, and two compared convalescent plasma to standard plasma (results not included in abstract). Effectiveness of convalescent plasma We included data on nine RCTs (12,875 participants) to assess the effectiveness of convalescent plasma compared to placebo or standard care alone. Convalescent plasma does not reduce all-cause mortality at up to day 28 (risk ratio (RR) 0.98, 95% confidence interval (CI) 0.92 to 1.05; 7 RCTs, 12,646 participants; high-certainty evidence). It has little to no impact on clinical improvement for all participants when assessed by liberation from respiratory support (RR not estimable; 8 RCTs, 12,682 participants; high-certainty evidence). It has little to no impact on the chance of being weaned or liberated from invasive mechanical ventilation for the subgroup of participants requiring invasive mechanical ventilation at baseline (RR 1.04, 95% CI 0.57 to 1.93; 2 RCTs, 630 participants; low-certainty evidence). It does not reduce the need for invasive mechanical ventilation (RR 0.98, 95% CI 0.89 to 1.08; 4 RCTs, 11,765 participants; high-certainty evidence). We did not identify any subgroup differences. We did not identify any studies reporting quality of life, and therefore, do not know whether convalescent plasma has any impact on quality of life. One RCT assessed resolution of fatigue on day 7, but we are very uncertain about the effect (RR 1.21, 95% CI 1.02 to 1.42; 309 participants; very low-certainty evidence). Safety of convalescent plasma We included results from eight RCTs, and one NRSI, to assess the safety of convalescent plasma. Some of the RCTs reported on safety data only for the convalescent plasma group. We are uncertain whether convalescent plasma increases or reduces the risk of grade 3 and 4 adverse events (RR 0.90, 95% CI 0.58 to 1.41; 4 RCTs, 905 participants; low-certainty evidence), and serious adverse events (RR 1.24, 95% CI 0.81 to 1.90; 2 RCTs, 414 participants; low-certainty evidence). A summary of reported events of the NRSI (reporting safety data for 20,000 of 35,322 transfused participants), and four RCTs reporting safety data only for transfused participants (6125 participants) are included in the full text. Individuals with a confirmed diagnosis of SARS-CoV-2 infection and asymptomatic or mild disease We identified one RCT reporting on 160 participants, comparing convalescent plasma to placebo treatment (saline). Effectiveness of convalescent plasma We are very uncertain about the effect of convalescent plasma on all-cause mortality (RR 0.50, 95% CI 0.09 to 2.65; very low-certainty evidence). We are uncertain about the effect of convalescent plasma on developing severe clinical COVID-19 symptoms (RR not estimable; low-certainty evidence). We identified no study reporting quality of life. Safety of convalescent plasma We do not know whether convalescent plasma is associated with a higher risk of grade 3 or 4 adverse events (very low-certainty evidence), or serious adverse events (very low-certainty evidence). This is a living systematic review. We search weekly for new evidence and update the review when we identify relevant new evidence. Please refer to the Cochrane Database of Systematic Reviews for the current status of this review. Authors' conclusions: We have high certainty in the evidence that convalescent plasma for the treatment of individuals with moderate to severe disease does not reduce mortality and has little to no impact on measures of clinical improvement. We are uncertain about the adverse effects of convalescent plasma. While major efforts to conduct research on COVID-19 are being made, heterogeneous reporting of outcomes is still problematic. There are 100 ongoing studies and 33 studies reporting in a study registry as being completed or terminated. Publication of ongoing studies might resolve some of the uncertainties around hyperimmune immunoglobulin therapy for people with any disease severity, and convalescent plasma therapy for people with asymptomatic or mild disease.
Article
The COVID-19 pandemic has drawn attention to food insecurity in developed countries. Despite adequate levels of agricultural production, consumers experienced demand-induced scarcity. Understanding the effects on nutrition and the environment is limited, yet critical to informing ecologically embedded mitigation strategies. To identify mitigation strategies, we investigated wheat flour and egg retail shortages in the United Kingdom (UK), focusing on consumer behavior during the COVID-19 lockdown. The 6 Steps for Quality Intervention Development (6SQuID) framework informed the methodology. Mixed qualitative and quantitative methods were used to pinpoint the causes of the shortages, and ecological impacts of consumer behavior were related using survey results (n = 243) and environmental and nutritional databases. This research confirmed consumers’ narrowed consideration set, willingness to pay, and significant reliance on processed foods which indicates agronomic biofortification, breeding strategies, selective imports and improved processed food quality are important mitigation strategies. We identified positive and negative synergies in consumer, producer and retailer behavior and related these to mitigation strategies in support of a circular bio-economy for food production. We found that the substitutes or alternative foods consumed during the COVID-19 lockdown were nutritionally inadequate. We identified the most ecological substitute for wheat flour to be corn flour; and for eggs, yogurt. Our findings also indicate that selenium deficiency is a risk for the UK population, especially to the increasing fifth of the population that is vegetarian. Due to the need to implement short-, medium-, and long-term mitigation strategies, a coordinated effort is required by all stakeholders.
Article
Full-text available
The newly identified 2019 novel coronavirus (2019-nCoV) has caused more than 11,900 laboratory-confirmed human infections, including 259 deaths, posing a serious threat to human health. Currently, however, there is no specific antiviral treatment or vaccine. Considering the relatively high identity of receptor-binding domain (RBD) in 2019-nCoV and SARS-CoV, it is urgent to assess the cross-reactivity of anti-SARS CoV antibodies with 2019-nCoV spike protein, which could have important implications for rapid development of vaccines and therapeutic antibodies against 2019-nCoV. Here, we report for the first time that a SARS-CoV-specific human monoclonal antibody, CR3022, could bind potently with 2019-nCoV RBD (KD of 6.3 nM). The epitope of CR3022 does not overlap with the ACE2 binding site within 2019-nCoV RBD. These results suggest that CR3022 may have the potential to be developed as candidate therapeutics, alone or in combination with other neutralizing antibodies, for the prevention and treatment of 2019-nCoV infections. Interestingly, some of the most potent SARS-CoV-specific neutralizing antibodies (e.g. m396, CR3014) that target the ACE2 binding site of SARS-CoV failed to bind 2019-nCoV spike protein, implying that the difference in the RBD of SARS-CoV and 2019-nCoV has a critical impact for the cross-reactivity of neutralizing antibodies, and that it is still necessary to develop novel monoclonal antibodies that could bind specifically to 2019-nCoV RBD.
Article
Full-text available
Structure of the nCoV trimeric spike The World Health Organization has declared the outbreak of a novel coronavirus (2019-nCoV) to be a public health emergency of international concern. The virus binds to host cells through its trimeric spike glycoprotein, making this protein a key target for potential therapies and diagnostics. Wrapp et al. determined a 3.5-angstrom-resolution structure of the 2019-nCoV trimeric spike protein by cryo–electron microscopy. Using biophysical assays, the authors show that this protein binds at least 10 times more tightly than the corresponding spike protein of severe acute respiratory syndrome (SARS)–CoV to their common host cell receptor. They also tested three antibodies known to bind to the SARS-CoV spike protein but did not detect binding to the 2019-nCoV spike protein. These studies provide valuable information to guide the development of medical counter-measures for 2019-nCoV. Science , this issue p. 1260
Preprint
Full-text available
Background: Since December 2019, acute respiratory disease (ARD) due to 2019 novel coronavirus (2019-nCoV) emerged in Wuhan city and rapidly spread throughout China. We sought to delineate the clinical characteristics of these cases. Methods: We extracted the data on 1,099 patients with laboratory-confirmed 2019-nCoV ARD from 552 hospitals in 31 provinces/provincial municipalities through January 29th, 2020. Results: The median age was 47.0 years, and 41.90% were females. Only 1.18% of patients had a direct contact with wildlife, whereas 31.30% had been to Wuhan and 71.80% had contacted with people from Wuhan. Fever (87.9%) and cough (67.7%) were the most common symptoms. Diarrhea is uncommon. The median incubation period was 3.0 days (range, 0 to 24.0 days). On admission, ground-glass opacity was the typical radiological finding on chest computed tomography (50.00%). Significantly more severe cases were diagnosed by symptoms plus reverse-transcriptase polymerase-chain-reaction without abnormal radiological findings than non-severe cases (23.87% vs. 5.20%, P<0.001). Lymphopenia was observed in 82.1% of patients. 55 patients (5.00%) were admitted to intensive care unit and 15 (1.36%) succumbed. Severe pneumonia was independently associated with either the admission to intensive care unit, mechanical ventilation, or death in multivariate competing-risk model (sub-distribution hazards ratio, 9.80; 95% confidence interval, 4.06 to 23.67). Conclusions: The 2019-nCoV epidemic spreads rapidly by human-to-human transmission. Normal radiologic findings are present among some patients with 2019-nCoV infection. The disease severity (including oxygen saturation, respiratory rate, blood leukocyte/lymphocyte count and chest X-ray/CT manifestations) predict poor clinical outcomes.
Article
Full-text available
Significance Next-generation influenza vaccines and broadly neutralizing antibodies (bNAbs) are in clinical development. Some of these do not block virus–receptor interactions and thus are predicted to provide protection via alternative mechanisms at the postentry stage or use Fc-dependent mechanisms. Nonneutralizing antibodies have the potential to mediate enhancement of respiratory disease (ERD). Our study describes ADE with two different functional MAbs that destabilized HA stem domain, increased influenza virus fusion kinetics, and led to enhanced lung pathology and ERD in a dose-dependent manner in a mice model. This study underlines careful preclinical evaluation of next-generation influenza vaccines or antibody-based therapeutics that do not block influenza virus receptor binding.
Article
Full-text available
To date, an effective therapeutic treatment that confers strong attenuation toward coronaviruses (CoVs) remains elusive. Of all the potential drug targets, the helicase of CoVs is considered to be one of the most important. Here, we first present the structure of the full-length Nsp13 helicase of SARS-CoV (SARS-Nsp13) and investigate the structural coordination of its five domains and how these contribute to its translocation and unwinding activity. A translocation model is proposed for the Upf1-like helicase members according to three different structural conditions in solution characterized through H/D exchange assay, including substrate state (SARS-Nsp13-dsDNA bound with AMPPNP), transition state (bound with ADP-AlF4-) and product state (bound with ADP). We observed that the β19-β20 loop on the 1A domain is involved in unwinding process directly. Furthermore, we have shown that the RNA dependent RNA polymerase (RdRp), SARS-Nsp12, can enhance the helicase activity of SARS-Nsp13 through interacting with it directly. The interacting regions were identified and can be considered common across CoVs, which provides new insights into the Replication and Transcription Complex (RTC) of CoVs.
Article
Full-text available
Recent history is punctuated by the emergence of highly pathogenic coronaviruses such as SARS- and MERS-CoV into human circulation. Upon infecting host cells, coronaviruses assemble a multi-subunit RNA-synthesis complex of viral non-structural proteins (nsp) responsible for the replication and transcription of the viral genome. Here, we present the 3.1 Å resolution structure of the SARS-CoV nsp12 polymerase bound to its essential co-factors, nsp7 and nsp8, using single particle cryo-electron microscopy. nsp12 possesses an architecture common to all viral polymerases as well as a large N-terminal extension containing a kinase-like fold and is bound by two nsp8 co-factors. This structure illuminates the assembly of the coronavirus core RNA-synthesis machinery, provides key insights into nsp12 polymerase catalysis and fidelity and acts as a template for the design of novel antiviral therapeutics.
Article
Full-text available
Background Coronaviruses (CoVs) primarily cause enzootic infections in birds and mammals but, in the last few decades, have shown to be capable of infecting humans as well. The outbreak of severe acute respiratory syndrome (SARS) in 2003 and, more recently, Middle-East respiratory syndrome (MERS) has demonstrated the lethality of CoVs when they cross the species barrier and infect humans. A renewed interest in coronaviral research has led to the discovery of several novel human CoVs and since then much progress has been made in understanding the CoV life cycle. The CoV envelope (E) protein is a small, integral membrane protein involved in several aspects of the virus’ life cycle, such as assembly, budding, envelope formation, and pathogenesis. Recent studies have expanded on its structural motifs and topology, its functions as an ion-channelling viroporin, and its interactions with both other CoV proteins and host cell proteins. Main body This review aims to establish the current knowledge on CoV E by highlighting the recent progress that has been made and comparing it to previous knowledge. It also compares E to other viral proteins of a similar nature to speculate the relevance of these new findings. Good progress has been made but much still remains unknown and this review has identified some gaps in the current knowledge and made suggestions for consideration in future research. Conclusions The most progress has been made on SARS-CoV E, highlighting specific structural requirements for its functions in the CoV life cycle as well as mechanisms behind its pathogenesis. Data shows that E is involved in critical aspects of the viral life cycle and that CoVs lacking E make promising vaccine candidates. The high mortality rate of certain CoVs, along with their ease of transmission, underpins the need for more research into CoV molecular biology which can aid in the production of effective anti-coronaviral agents for both human CoVs and enzootic CoVs.
Preprint
Over the past 20 years, several coronaviruses have crossed the species barrier into humans, causing outbreaks of severe, and often fatal, respiratory illness. Since SARS-CoV was first identified in animal markets, global viromics projects have discovered thousands of coronavirus sequences in diverse animals and geographic regions. Unfortunately, there are few tools available to functionally test these novel viruses for their ability to infect humans, which has severely hampered efforts to predict the next zoonotic viral outbreak. Here we developed an approach to rapidly screen lineage B betacoronaviruses, such as SARS-CoV and the recent 2019-nCoV, for receptor usage and their ability to infect cell types from different species. We show that host protease processing during viral entry is a significant barrier for several lineage B viruses and that bypassing this barrier allows several lineage B viruses to enter human cells through an unknown receptor. We also demonstrate how different lineage B viruses can recombine to gain entry into human cells and confirm that human ACE2 is the receptor for the recently emerging 2019-nCoV.