Critical analysis (5 pages)

Neutralizing antibody vaccine for pandemic
and pre-emergent coronaviruses

Kevin O. Saunders, Esther Lee, Robert Parks, David R. Martinez, Dapeng Li, Haiyan Chen,
Robert J. Edwards, Sophie Gobeil, Maggie Barr, Katayoun Mansouri, S. Munir Alam,
Laura L. Sutherland, Fangping Cai, Aja M. Sanzone, Madison Berry, Kartik Manne,
Kevin W. Bock, Mahnaz Minai, Bianca M. N ag at a , A ny wa y B. Kapingidza, Mihai Azoitei,
Longping V. Tse, Trevor D. Scobey, Rachel L. Spreng, R. Wes Rountree, C. Todd DeMarco,
Thomas N. D en ny , C hr is to ph er W. Woods, Elizabeth W. Petzold, Juanjie Tang,
Thomas H. Oguin I II , G re go ry D. Sempowski, Matthew Gagne, Daniel C. Douek,
Mark A. Tomai, Christopher B. Fox, Robert Seder, Kevin Wiehe, Drew Weissman,
Norbert Pardi, Hana Golding, Surender K hu ra na , P ri yamvada Acharya, Hanne Andersen,
Mark G. Lewis, Ian N. Moore, David C. Montefiori, Ralph S. Baric & Barton F. Haynes

This is a PDF file of a peer-reviewed paper that has been accepted for publication.
Although unedited, the content has been subjected to preliminary formatting.
Nature is providing this early version of the typeset paper as a service to our authors
and readers. The text and figures will undergo copyediting and a proof review before
the paper is published in its final form. Please note that during the production process
errors may be discovered which could affect the content, and all legal disclaimers
apply.

Received: 7 February 2021

Accepted: 29 April 2021

Accelerated Article Preview Published
online 10 May 2021

Cite this article as: Saunders, K. O. et al.
Neutralizing antibody vaccine for pandemic
and pre-emergent coronaviruses. Nature
https://doi.org/10.1038/s41586-021-
03594-0 (2021).

https://doi.org/10.1038/s41586-021-03594-0

Nature | www.nature.com

Accelerated Article Preview

AC
CE

LE
RA

TE
D

AR
TI

CL
E

PR
EV

IE
W

https://doi.org/10.1038/s41586-021-03594-0

Nature | www.nature.com | 1

Article

Neutralizing antibody vaccine for pandemic
and pre-emergent coronaviruses

Kevin O. Saunders1,2,3,4 ✉, Esther Lee1,5, Robert Parks1,5, David R. Martinez6, Dapeng Li1,5,
Haiyan Chen1,5, Robert J. Edwards1,5, Sophie Gobeil1,5, Maggie Barr1,5, Katayoun Mansouri1,5,
S. Munir Alam1,5, Laura L. Sutherland1,5, Fangping Cai1,5, Aja M. Sanzone1,5, Madison Berry1,5,
Kartik Manne1,5, Kevin W. Bock7, Mahnaz Minai7, Bianca M. Nagata7, Anyway B. Kapingidza1,5,
Mihai Azoitei1,5, Longping V. Tse6, Trevor D. Scobey6, Rachel L. Spreng1,5, R. Wes Rountree1,5,
C. Todd DeMarco1,5, Thomas N. Denny1,5, Christopher W. Woods1,5,8, Elizabeth W. Petzold8,
Juanjie Tang9, Thomas H. Oguin III1,5, Gregory D. Sempowski1,5, Matthew Gagne10,
Daniel C. Douek10, Mark A. Tomai11, Christopher B. Fox12, Robert Seder10, Kevin Wiehe1,5,
Drew Weissman13, Norbert Pardi13, Hana Golding9, Surender Khurana9,
Priyamvada Acharya1,2, Hanne Andersen14, Mark G. Lewis14, Ian N. Moore7,
David C. Montefiori1,2, Ralph S. Baric6 & Barton F. Haynes1,3,5 ✉

Betacoronaviruses (betaCoVs) caused the severe acute respiratory syndrome (SARS)
and Middle East Respiratory Syndrome (MERS) outbreaks, and the SARS-CoV-2
pandemic1–4. Vaccines that elicit protective immunity against SARS-CoV-2 and
betaCoVs circulating in animals have the potential to prevent future betaCoV
pandemics. Here, we show that macaque immunization with a multimeric SARS-CoV-2
receptor binding domain (RBD) nanoparticle adjuvanted with 3M-052/Alum elicited
cross-neutralizing antibody (cross-nAb) responses against batCoVs, SARS-CoV-1,
SARS-CoV-2, and SARS-CoV-2 variants B.1.1.7, P.1, and B.1.351. Nanoparticle
vaccination resulted in a SARS-CoV-2 reciprocal geometric mean neutralization ID50
titer of 47,216, and protection against SARS-CoV-2 in macaque upper and lower
respiratory tracts. Importantly, nucleoside-modified mRNA encoding a stabilized
transmembrane spike or monomeric RBD also induced SARS-CoV-1 and batCoV
cross-nAbs, albeit at lower titers. These results demonstrate current mRNA vaccines
may provide some protection from future zoonotic betaCoV outbreaks, and provide a
platform for further development of pan-betaCoV vaccines.

SARS coronavirus 1 (SARS-CoV-1), SARS coronavirus 2 (SARS-CoV-2),
and MERS coronavirus (MERS-CoV) emerged from transmission events
where humans were infected with bat or camel CoVs5–8. BetaCoVs that
circulate in civets, bats, and Malayan pangolins are genetically similar
to SARS-CoV-1 and SARS-CoV-2, and use human ACE2 as a receptor5,6,9–11.
These SARS-related animal coronaviruses have the potential to be trans-
mitted to humans12. Cross-nAbs capable of neutralizing multiple beta-
CoVs and preventing or treating betaCoV infection have been isolated
from SARS-CoV-1 infected humans13–24, providing proof-of-concept for
development of betaCoV vaccines against Sarbecoviruses25.

In mice, vaccine induction of cross-nAbs has been reported for
CoV pseudoviruses26,27. However, it is unknown whether spike vac-
cination of primates can elicit cross-nAbs against SARS-CoV-1, bat
betaCoVs, or SARS-CoV-2 escape viruses. A target of cross-nAbs is
the RBD of spike14,24,25. One such RBD cross-nAb is antibody DH1047,

which cross-neutralizes SARS-CoV-1, SARS-CoV-2 and bat CoVs15. RBD
immunogenicity can be augmented by arraying multiple copies on
nanoparticles, mimicking virus-like particles26–29. Thus, we designed
a 24-mer SARS-CoV-2 RBD-ferritin nanoparticle vaccine. The RBD nan-
oparticle was constructed by expressing recombinant SARS-CoV-2
RBD with a C-terminal sortase A donor sequence, and by expressing a
24-subunit, self-assembling protein nanoparticle Helicobacter pylori
ferritin with an N-terminal sortase A acceptor sequence30. The RBD
and ferritin nanoparticle were conjugated together by a sortase A reac-
tion (Fig. 1a, Extended Data Fig. 1)30. Analytical size exclusion chro-
matography and negative stain electron microscopy confirmed that
RBD was conjugated to the surface of the ferritin nanoparticle (Fig. 1a,
Extended Data Fig. 1b,c). The RBD sortase A conjugated nanoparti-
cle (RBD-scNP) bound to human ACE2, the receptor for SARS-CoV-2,
and to potently neutralizing SARS-CoV-2-specific RBD antibodies

https://doi.org/10.1038/s41586-021-03594-0

Received: 7 February 2021

Accepted: 29 April 2021

Published online: 10 May 2021

1Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA. 2Department of Surgery, Duke University, Durham, NC, USA. 3Department of Immunology, Duke
University School of Medicine, Durham, NC, USA. 4Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA. 5Department of Medicine, Duke
University School of Medicine, Durham, NC, USA. 6Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. 7Infectious Disease Pathogenesis Section,
Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA. 8Center for Applied Genomics and Precision Medicine,
Duke University Medical Center, Durham, NC, USA. 9Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD, USA.
10Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, USA. 11Corporate Research Materials Lab, 3M Company, St Paul, MN, USA.
12Infectious Disease Research Institute, Seattle, WA, USA. 13Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA. 14BIOQUAL, Rockville, MD, USA.
✉e-mail: [email protected]; [email protected]

AC
CE

LE
RA

TE
D

AR
TI

CL
E

PR
EV

IE
W

https://doi.org/10.1038/s41586-021-03594-0

mailto:[email protected]

mailto:[email protected]

2 | Nature | www.nature.com

Article
(Abs) DH1041, DH1042, DH1043, DH1044, and DH104515 (Fig. 1b). The
cross-nAb DH1047 also bound to the RBD-scNP (Fig. 1b). The RBD-scNP
lacked binding to SARS-CoV-2 spike Abs that bound outside of the RBD
(Fig. 1b).

Five cynomolgus macaques were immunized three times intramus-
cularly four weeks apart with 100 μg of RBD-scNP adjuvanted with
5 μg of the TLR7/8 agonist 3M-052 absorbed to 500 μg of alum (Fig. 1c
and Extended Data Fig. 1d,e)31. Immunizations were well-tolerated
in macaques (Extended Data Fig. 2). Immunization with RBD-scNP
adjuvanted with 3M-052/Alum elicited binding IgG against SARS-CoV-2
RBD and stabilized Spike ectodomain (S-2P) (Fig. 1d), but immuni-
zation with 3M-052/Alum alone did not (Extended Data Fig. 3a,b).
Boosting once maximally increased SARS-CoV-2 binding IgG titers
(Fig. 1d). ACE2 competitive binding assays demonstrated the pres-
ence of ACE2-binding site Abs in the serum of vaccinated macaques
(Fig. 1e). Similarly, plasma Abs blocked the binding of ACE2-binding
site-focused, RBD neutralizing antibody DH1041 (Fig. 1e). Vaccine
induction of nAbs was assessed against a SARS-CoV-2 pseudovirus
with an aspartic acid to glycine substitution at position 614 (D614G)32.
Two RBD-scNP immunizations induced potent serum nAbs with fifty
percent inhibitory reciprocal serum dilution (ID50) neutralization
titers ranging from 21,292 to 162,603 (Fig. 1f,g). We compared these nAb
titers to those elicited by cynomolgus macaques immunized twice with
50 μg of lipid-encapsulated nucleoside-modified mRNA (mRNA-LNP)
encoding stabilized transmembrane (TM) Spike (S-2P) that is analogous
to COVID-19 vaccines authorized for emergency use (Extended Data
Fig. 1f ). Serum neutralization titers against SARS-CoV-2 D614G pseu-
dovirus elicited by RBD-scNP immunization were significantly higher
than titers elicited by two S-2P mRNA-LNP immunizations (Fig. 1i, group
geometric mean ID50 47,216 versus 6,469, P = 0.0079 Exact Wilcoxon
test, n=5)33,34. When compared to natural human infection, RBD-scNP
vaccination elicited higher ID50 neutralization titers (Fig. 1j). Thus,
RBD-scNP adjuvanted with 3M-052/Alum elicits significantly higher
neutralizing titers in macaques compared to current vaccine platforms
or natural human infection.

The SARS-CoV-2 variant B.1.1.7 or United Kingdom variant is spread-
ing globally, and has been suggested to have higher infectivity than
the Wuhan-1 strain35,36. B.1.351 lineage viruses are widespread in the
Republic of South Africa36–38, and along with Brazilian variant P.1 are
of concern due to their neutralization-resistant phenotype mediated
by mutations in the RBD at K417N, E484K, and N501Y39. Each of these
mutations were distal to the cross-nAb DH1047 binding site owing to
its long HCDR3 used to contact RBD; however, the E484K mutation was
within the binding site of RBD nAb DH1041 (Fig. 2a,b)15. Thus, DH1041
binding to SARS-CoV-2 RBD was knocked out by the E484K mutation,
but DH1047 binding to RBD was unaffected by K417N, E484K, or N501Y
(Fig. 2c,d and Extended Data Fig. 3).

We determined whether RBD-scNP or mRNA-LNP immunization
elicited nAbs against these particular SARS-CoV-2 variants. RBD-scNP
macaque serum potently neutralized a pseudovirus bearing the
D614G spike and the B.1.1.7 spike (Fig. 2d,e). Similarly, S-2P mRNA-LNP
immunization elicited equivalent titers of nAbs against the B.1.1.7
and D614G variants of SARS-CoV-2, although titers were lower than
RBD-scNP immunization (Fig. 2d,e). Macaque serum from RBD-scNP
or mRNA-LNP immunization neutralized SARS-CoV-2 WA-1, B.1.351, and
P.1 pseudoviruses, with ID80 titers being more potent for the RBD-scNP
group (Fig. 2f-i). On average, the RBD-scNP group neutralization titers
decreased by 3-fold against B.1.351 or P.1, whereas the mRNA-LNP group
decreased by 6-fold for B.1.351 and 10-fold for P.1 based on ID50 titers
(Fig. 2g,i). Additionally, we observed RBD-scNP and S-2P mRNA-LNP
immune plasma IgG binding to SARS-CoV-2 S was unaffected by muta-
tions observed in Danish minks, B.1.351, P.1, and B.1.1.7 SARS-CoV-2
strains36,37,40 (Extended Data Fig. 3). In summary, both vaccines tested
here elicited nAbs that were unaffected by the mutations in the B.1.1.7
strain. However, nAbs elicited by RBD-scNP more potently neutralized

the difficult-to-neutralize B.1.351 and P.1 virus strains than nAbs elicited
with S-2P mRNA-LNP immunization.

SARS-related CoVs that circulate in humans and animals remain a
threat for future outbreaks12,41,42. Therefore, we examined neutrali-
zation of SARS-CoV-1 and SARS-related group 2b batCoV-WIV-1 and
SARS-related batCoV-SHC014 viruses by immune sera from macaques
vaccinated with the RBD-scNP, mRNA-LNP encoding monomeric
RBD or S-2P (Extended Data Fig. 1e-g)6,9,41,42. After two immunizations,
RBD-scNP, S-2P mRNA-LNP, and the RBD monomer mRNA-LNP elic-
ited nAbs against SARS-CoV-1, batCoV-WIV-1, and batCoV-SHC014
(Fig. 3a, Extended Data Fig. 4). Neutralization was more potent for
replication-competent SARS-CoV-2 virus compared to the other
three SARS-related viruses (Fig. 3a, Extended Data Fig. 4), with neu-
tralization titers varying up to 4-fold within the RBD-scNP group
(Extended Data Fig. 4). Overall, RBD-scNP immunization elicited the
highest neutralization titers (Fig. 3a, Extended Data Fig. 4). Small
increases in neutralization potency were gained by boosting a third
time with the RBD-scNP (Fig. 3b). Moreover, RBD-scNP immunization
elicited cross-reactive plasma IgG against SARS-CoV-2, SARS-CoV-1,
batCoV-RaTG13, batCoV-SHC014, pangolin CoV-GXP4L Spike proteins
(Fig. 3c and Extended Data Fig. 5a,c). Binding antibody titers were high
for these spikes, even in instances where neutralization titers were low
suggesting non-nAbs contributed to binding titers. RBD-scNP immune
plasma IgG did not bind spike from the four endemic human CoVs or
MERS-CoV (Extended Data Fig. 5a,c). The lack of binding by plasma IgG
to these latter five S ectodomains was consistent with RBD sequence
divergence among groups 1, 2a, 2b and 2c coronaviruses (Fig. 3f and
Extended Data Fig. 6-7). The SARS-CoV-2 spike induced cross-nAbs
against multiple group 2b SARS-related betaCoVs, with the highest
titers induced by RBD-scNP.

Immune sera from RBD-scNP-immunized macaques exhibited a
similar cross-neutralizing profile as the cross-nAb DH1047. DH1047
bound with <0.02 nM affinity to monomeric SARS-CoV-2 RBD (Extended Data Fig. 5b), and bound the RBD-scNP (Fig. 1b). The cross-reactive DH1047 epitope is adjacent to the N-terminus of the ACE2-binding site, distinguishing it from dominant ACE2 binding site-focused nAbs such as DH1041 (Fig. 3d)15, and it has high group 2b sequence conserva- tion (Fig. 3e). Overall RBD sequences within betaCoV groups are more conserved than sequences from different groups (Fig. 3f and Extended Data Fig. 6-7). The presence of DH1047-like antibodies was determined with DH1047 blocking assays. Plasma from all RBD-scNP-immunized macaques blocked the binding of ACE2 and DH1047 to SARS-CoV-2 S-2P ectodomain (Fig. 3g and Extended Data Fig. 5d). The DH1047-blocking antibodies were cross-reactive as they also potently blocked DH1047 binding to batCoV-SHC014 S-2P (Fig. 3g). RBD-scNP immunization elic- ited higher magnitudes of DH1047 blocking Abs than S-2P mRNA-LNP immunization of macaques, Pfizer/BNT162b2 mRNA-LNP immuniza- tion of humans, or SARS-CoV-2 human infection (Fig. 3h and Extended Data Fig. 5d). ACE2 blocking was high in all groups (Fig. 3h). While 5 of 5 RBD-scNP-vaccinated macaques exhibited potent DH1047 serum blocking activity, 3 of 4 immunized humans and 9 of 22 of COVID-19 convalescent humans had detectable serum DH1047 blocking activity (Fig. 3h). Thus, the DH1047-like antibody response was subdominant in infected or immunized humans and S-2P mRNA-LNP-immunized macaques, but was a dominant antibody response to RBD-scNP vac- cination. To determine vaccine protection against coronavirus infection, we challenged RBD-scNP-vaccinated and S-2P mRNA-LNP primed/ RBD-scNP boosted monkeys with 105 plaque forming units of SARS-CoV-2 virus via intratracheal and intranasal routes after their last boost (Fig. 4a). NAbs were detectable in all macaques 2 weeks after the final immunization (Fig. 3b and Extended Data Fig. 4b,c). Bronchoalveolar lavage (BAL) fluid was collected 2 days post chal- lenge (Fig. 4a). Infectious SARS-CoV-2 was detectable in BAL fluid from 5 of 6 unimmunized macaques, and undetectable in all RBD-scNP and AC CE LE RA TE D AR TI CL E PR EV IE W AC CE LE RA TE D AR TI CL E PR EV IE W Nature | www.nature.com | 3 S-2P mRNA-LNP/RBD-scNP-immunized macaques (Fig. 4b). Copies of Envelope (E) and Nucleocapsid (N) subgenomic RNA in fluid from nasal swabs and bronchoalveolar lavage (BAL) two and four days after challenge was used to quantify SARS-CoV-2 replication (Fig. 4a). On day 2 after challenge, unimmunized macaques had an average of 1.3x105 and 1.2x104 copies/mL of E sgRNA in the nasal swab and BAL fluids, respectively (Fig. 4c,d). In contrast, RBD-scNP-vaccinated monkeys and 4 of 5 S-2P mRNA-LNP monkeys had undetectable levels of E sgRNA in the upper and lower respiratory tract (Fig. 4c,d). We sampled mon- keys again 2 days later, and found no detectable E sgRNA in any vac- cinated monkey BAL or nasal swab samples (Fig. 4b,c). Similarly, all RBD-scNP-vaccinated macaque had undetectable N sgRNA in BAL and the nasal swab fluid, except one macaque that had 234 copies/mL of N sgRNA detected on day 2 in nasal swab fluid (Fig. 4e,f ). Virus replication was undetectable in this macaque by the fourth day after challenge (Fig. 4e). Additionally, all but one mRNA-LNP/RBD-scNP-immunized macaque had undetectable N sgRNA in BAL or nasal swab samples (Fig. 4e,f ). Moreover, SARS-CoV-2 nucleocapsid antigen was undetect- able in the lung tissue of all vaccinated macaques, but was detected in all control macaques (Fig. 4g and Extended Data Fig. 8). Hematoxylin and eosin staining of lung tissue showed a reduction in inflammation in immunized macaques compared to control macaques (Extended Data Fig. 8 and Extended Data Table 1). Finally, mucosal immunity to SARS-CoV-2 were examined when possible both before and after SARS-CoV-2 challenge (Extended Data Fig. 9). IgG from concentrated BAL bound to spike and blocked ACE-2, DH1041, and DH1047 binding to spike (Extended Data Fig. 9b-d). Each response was higher in the BAL from monkeys immunized three times with RBD-scNP compared to monkeys immunized two times with S-2P mRNA-LNP and boosted once with RBD-scNP, although the BAL was collected from each group at different timepoints. Unconcentrated nasal wash samples from monkeys immunized with either RBD-scNPs or S-2P mRNA-LNP prime/RBD-scNP boost showed similar low levels of spike-binding IgG post challenge (Extended Data Fig. 9e). Nonetheless, RBD-scNP-immunization elicited RBD-specific mucosal antibodies. As three coronavirus epidemics have occurred in the past 20 years, there is a need to develop effective pancoronavirus vaccines prior to the next pandemic25. The epitopes of betaCoV cross-nAbs, such as DH1047, provide clear targets for vaccines aiming to protect against multiple CoVs13–15,43. We have shown that immunization with RBD-scNP adju- vanted with a toll-like receptor agonist 3M-052, and spike mRNA-LNP to a lesser extent, induces cross-nAbs against multiple SARS-related human and bat betaCoVs in primates. These results demonstrate that SARS-CoV-2 vaccination with either the RBD-scNP or spike mRNA-LNP vaccines similar to those authorized for use in humans, will likely elicit cross-nAbs with the potential to prevent future group 2b betaCoV spillo- ver from bats to humans12,26. The emergence of SARS-CoV-2 neutralization-resistant and highly infectious variants continues to be a concern for vaccine efficacy. RBD-scNP and SARS-CoV-2 spike mRNA-LNP immunizations elicited SARS-CoV-2 nAbs against SARS-CoV-2 D614G, B.1.1.7, P.1, and B.1.351 strains. The nAbs elicited by RBD-scNP and S-2P mRNA-LNP were of different specificities since RBD-scNP-induced nAbs showed a smaller reduction in neutralization potency across the different variants compared to S-2P mRNA-LNP immune sera. Our results are consistent with the demonstration that current COVID-19 vaccines have reduced efficacy against the B.1.351 SARS-CoV-2 variant44–49. The RBD-scNP vaccine is a promising platform for pancoronavi- rus vaccine development for the following reasons. The RBD-scNP vaccine induced apparent sterilizing immunity in the upper respira- tory tract, which has not been routinely achieved with SARS-CoV-2 vaccination in macaques50,51. Additionally, the extraordinarily high neutralization titers achieved by RBD-scNP vaccination bode well for an extended duration of protection. Despite the induction of high levels of antibody we observed no evidence of increased immunopathology, inflammatory cytokines or virus replication indicative vaccine-elicited antibody-dependent enhancement. Lack of in vivo infection enhance- ment is consistent with studies using SARS-CoV-2 monoclonal Abs15. 3M-052 adsorbed to Alum is in clinical testing (NCT04177355) gen- erating a potential translational pathway for RBD-scNP adjuvanted with 3M-052. The RBD-scNP/3M-052 vaccine represents a platform for producing pancoronavirus vaccines that could prevent, rapidly temper, or extinguish the next spillover of a coronavirus into humans. Online content Any methods, additional references, Nature Research reporting sum- maries, source data, extended data, supplementary information, acknowledgements, peer review information; details of author contri- butions and competing interests; and statements of data and code avail- ability are available at https://doi.org/10.1038/s41586-021-03594-0. 1. Wang, C., Horby, P. W., Hayden, F. G. & Gao, G. F. A novel coronavirus outbreak of global health concern. Lancet 395, 470-473, https://doi.org/10.1016/s0140-6736(20)30185-9 (2020). 2. Zaki, A. M., van Boheemen, S., Bestebroer, T. M., Osterhaus, A. D. & Fouchier, R. A. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med 367, 1814-1820, https://doi.org/10.1056/NEJMoa1211721 (2012). 3. Zhong, N. S. et al. Epidemiology and cause of severe acute respiratory syndrome (SARS) in Guangdong, People’s Republic of China, in February, 2003. Lancet 362, 1353-1358, https://doi.org/10.1016/s0140-6736(03)14630-2 (2003). 4. Zhu, N. et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N Engl J Med 382, 727-733, https://doi.org/10.1056/NEJMoa2001017 (2020). 5. Zhou, P. et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 579, 270-273, https://doi.org/10.1038/s41586-020-2012-7 (2020). 6. Li, W. et al. Bats are natural reservoirs of SARS-like coronaviruses. Science 310, 676-679, https://doi.org/10.1126/science.1118391 (2005). 7. Olival, K. J. et al. Host and viral traits predict zoonotic spillover from mammals. Nature 546, 646-650, https://doi.org/10.1038/nature22975 (2017). 8. Sabir, J. S. et al. Co-circulation of three camel coronavirus species and recombination of MERS-CoVs in Saudi Arabia. Science 351, 81-84, https://doi.org/10.1126/science.aac8608 (2016). 9. Ge, X. Y. et al. Isolation and characterization of a bat SARS-like coronavirus that uses the ACE2 receptor. Nature 503, 535-538, https://doi.org/10.1038/nature12711 (2013). 10. Guan, Y. et al. Isolation and characterization of viruses related to the SARS coronavirus from animals in southern China. Science 302, 276-278, https://doi.org/10.1126/ science.1087139 (2003). 11. Xiao, K. et al. Isolation of SARS-CoV-2-related coronavirus from Malayan pangolins. Nature 583, 286-289, https://doi.org/10.1038/s41586-020-2313-x (2020). 12. Menachery, V. D., Graham, R. L. & Baric, R. S. Jumping species-a mechanism for coronavirus persistence and survival. Curr Opin Virol 23, 1-7, https://doi.org/10.1016/j. coviro.2017.01.002 (2017). 13. Pinto, D. et al. Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Nature 583, 290-295, https://doi.org/10.1038/s41586-020-2349-y (2020). 14. Wec, A. Z. et al. Broad neutralization of SARS-related viruses by human monoclonal antibodies. Science 369, 731-736, https://doi.org/10.1126/science.abc7424 (2020). 15. Li, D. et al. The functions of SARS-CoV-2 neutralizing and infection-enhancing antibodies in vitro and in mice and nonhuman primates. bioRxiv, https://doi.org/10.1101/ 2020.12.31.424729 (2021). 16. Shi, R. et al. A human neutralizing antibody targets the receptor-binding site of SARS-CoV-2. Nature 584, 120-124, https://doi.org/10.1038/s41586-020-2381-y (2020). 17. Ju, B. et al. Human neutralizing antibodies elicited by SARS-CoV-2 infection. Nature 584, 115-119, https://doi.org/10.1038/s41586-020-2380-z (2020). 18. Robbiani, D. F. et al. Convergent antibody responses to SARS-CoV-2 in convalescent individuals. Nature 584, 437-442, https://doi.org/10.1038/s41586-020-2456-9 (2020). 19. Zost, S. J. et al. Potently neutralizing and protective human antibodies against SARS-CoV-2. Nature 584, 443-449, https://doi.org/10.1038/s41586-020-2548-6 (2020). 20. Liu, L. et al. Potent neutralizing antibodies against multiple epitopes on SARS-CoV-2 spike. Nature 584, 450-456, https://doi.org/10.1038/s41586-020-2571-7 (2020). 21. Hansen, J. et al. Studies in humanized mice and convalescent humans yield a SARS-CoV-2 antibody cocktail. Science 369, 1010-1014, https://doi.org/10.1126/science.abd0827 (2020). 22. Brouwer, P. J. M. et al. Potent neutralizing antibodies from COVID-19 patients define multiple targets of vulnerability. Science 369, 643-650, https://doi.org/10.1126/science. abc5902 (2020). 23. Rogers, T. F. et al. Isolation of potent SARS-CoV-2 neutralizing antibodies and protection from disease in a small animal model. Science 369, 956-963, https://doi.org/10.1126/ science.abc7520 (2020). 24. Piccoli, L. et al. Mapping Neutralizing and Immunodominant Sites on the SARS-CoV-2 Spike Receptor-Binding Domain by Structure-Guided High-Resolution Serology. Cell 183, 1024-1042.e1021, https://doi.org/10.1016/j.cell.2020.09.037 (2020). 25. Burton, D. R. & Walker, L. M. Rational Vaccine Design in the Time of COVID-19. Cell Host Microbe 27, 695-698, https://doi.org/10.1016/j.chom.2020.04.022 (2020). 26. Cohen, A. A. et al. Mosaic nanoparticles elicit cross-reactive immune responses to zoonotic coronaviruses in mice. Science, https://doi.org/10.1126/science.abf6840 (2021). AC CE LE RA TE D AR TI CL E PR EV IE W AC CE LE RA TE D AR TI CL E PR EV IE W https://doi.org/10.1038/s41586-021-03594-0 https://doi.org/10.1016/s0140-6736(20)30185-9 https://doi.org/10.1056/NEJMoa1211721 https://doi.org/10.1016/s0140-6736(03)14630-2 https://doi.org/10.1056/NEJMoa2001017 https://doi.org/10.1038/s41586-020-2012-7 https://doi.org/10.1126/science.1118391 https://doi.org/10.1038/nature22975 https://doi.org/10.1126/science.aac8608 https://doi.org/10.1038/nature12711 https://doi.org/10.1126/science.1087139 https://doi.org/10.1126/science.1087139 https://doi.org/10.1038/s41586-020-2313-x https://doi.org/10.1016/j.coviro.2017.01.002 https://doi.org/10.1016/j.coviro.2017.01.002 https://doi.org/10.1038/s41586-020-2349-y https://doi.org/10.1126/science.abc7424 https://doi.org/10.1101/2020.12.31.424729 https://doi.org/10.1101/2020.12.31.424729 https://doi.org/10.1038/s41586-020-2381-y https://doi.org/10.1038/s41586-020-2380-z https://doi.org/10.1038/s41586-020-2456-9 https://doi.org/10.1038/s41586-020-2548-6 https://doi.org/10.1038/s41586-020-2571-7 https://doi.org/10.1126/science.abd0827 https://doi.org/10.1126/science.abc5902 https://doi.org/10.1126/science.abc5902 https://doi.org/10.1126/science.abc7520 https://doi.org/10.1126/science.abc7520 https://doi.org/10.1016/j.cell.2020.09.037 https://doi.org/10.1016/j.chom.2020.04.022 https://doi.org/10.1126/science.abf6840 27. Ma, X. et al. Nanoparticle Vaccines Based on the Receptor Binding Domain (RBD) and Heptad Repeat (HR) of SARS-CoV-2 Elicit Robust Protective Immune Responses. Immunity 53, 1315-1330 e1319, https://doi.org/10.1016/j.immuni.2020.11.015 (2020). 28. Bangaru, S. et al. Structural analysis of full-length SARS-CoV-2 spike protein from an advanced vaccine candidate. Science 370, 1089-1094, https://doi.org/10.1126/science. abe1502 (2020). 29. Walls, A. C. et al. Elicitation of Potent Neutralizing Antibody Responses by Designed Protein Nanoparticle Vaccines for SARS-CoV-2. Cell 183, 1367-1382 e1317, https:// doi.org/10.1016/j.cell.2020.10.043 (2020). 30. Saunders, K. O. et al. Targeted selection of HIV-specific antibody mutations by engineering B cell maturation. Science 366, https://doi.org/10.1126/science.aay7199 (2019). 31. Fox, C. B. et al. Adsorption of a synthetic TLR7/8 ligand to aluminum oxyhydroxide for enhanced vaccine adjuvant activity: A formulation approach. J Control Release 244, 98-107, https://doi.org/10.1016/j.jconrel.2016.11.011 (2016). 32. Korber, B. et al. Tracking Changes in SARS-CoV-2 Spike: Evidence that D614G Increases …

Place your order
(550 words)

Approximate price: $22

Calculate the price of your order

550 words
We'll send you the first draft for approval by September 11, 2018 at 10:52 AM
Total price:
$26
The price is based on these factors:
Academic level
Number of pages
Urgency
Basic features
  • Free title page and bibliography
  • Unlimited revisions
  • Plagiarism-free guarantee
  • Money-back guarantee
  • 24/7 support
On-demand options
  • Writer’s samples
  • Part-by-part delivery
  • Overnight delivery
  • Copies of used sources
  • Expert Proofreading
Paper format
  • 275 words per page
  • 12 pt Arial/Times New Roman
  • Double line spacing
  • Any citation style (APA, MLA, Chicago/Turabian, Harvard)

Our guarantees

Delivering a high-quality product at a reasonable price is not enough anymore.
That’s why we have developed 5 beneficial guarantees that will make your experience with our service enjoyable, easy, and safe.

Money-back guarantee

You have to be 100% sure of the quality of your product to give a money-back guarantee. This describes us perfectly. Make sure that this guarantee is totally transparent.

Read more

Zero-plagiarism guarantee

Each paper is composed from scratch, according to your instructions. It is then checked by our plagiarism-detection software. There is no gap where plagiarism could squeeze in.

Read more

Free-revision policy

Thanks to our free revisions, there is no way for you to be unsatisfied. We will work on your paper until you are completely happy with the result.

Read more

Privacy policy

Your email is safe, as we store it according to international data protection rules. Your bank details are secure, as we use only reliable payment systems.

Read more

Fair-cooperation guarantee

By sending us your money, you buy the service we provide. Check out our terms and conditions if you prefer business talks to be laid out in official language.

Read more
Open chat
1
You can contact our live agent via WhatsApp! Via + 1 929 473-0077

Feel free to ask questions, clarifications, or discounts available when placing an order.

Order your essay today and save 20% with the discount code GURUH