ArticlePDF Available

SARS-CoV-2 infection protects against rechallenge in rhesus macaques

Authors:

Abstract and Figures

Immunity from reinfection One of the many open questions about severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is whether an individual who has cleared the virus can be infected a second time and get sick. Chandrashekar et al. and Deng et al. generated rhesus macaque models of SARS-CoV-2 infection and tested whether natural SARS-CoV-2 infection could result in immunity to viral rechallenge. They found that animals indeed developed immune responses that protected against a second infection. Although there are differences between SARS-CoV-2 infection in macaques and in humans, these findings have key implications for public health and economic initiatives if validated in human studies. Science , this issue p. 812 , p. 818
Content may be subject to copyright.
Cite as: A. Chandrashekar et al., Science
10.1126/science.abc4776 (2020).
RESEARCH ARTICLES
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 1
The explosive spread of the COVID-19 pandemic has made
the development of countermeasures an urgent global prior-
ity (18). However, our understanding of the immunopatho-
genesis of SARS-CoV-2 is currently very limited. In
particular, it is currently not known whether SARS-CoV-2
infection induces natural immunity that protects against re-
exposure in humans. Such information is critical for vaccine
strategies, epidemiologic modeling, and public health ap-
proaches. To explore this question, we developed a rhesus
macaque model of SARS-CoV-2 infection, and we assessed
virologic, immunologic, and pathologic features of infection
as well as protective immunity against rechallenge.
Virology and immunology of SARS-CoV-2 infection in
rhesus macaques
We inoculated 9 adult rhesus macaques (6-12 years old) with
a total of 1.1 × 106 PFU (Group 1; N = 3), 1.1 × 105 PFU
(Group 2; N = 3), or 1.1 × 104 PFU (Group 3; N = 3) SARS-
CoV-2, administered as 1 ml by the intranasal (IN) route and
1 ml by the intratracheal (IT) route. Following viral chal-
lenge, we assessed viral RNA levels by RT-PCR in multiple
anatomic compartments. We observed high levels of viral
RNA in bronchoalveolar lavage (BAL) (Fig. 1A) and nasal
swabs (NS) (Fig. 1B), with a median peak of 6.56 (range 5.32-
8.97) log10 RNA copies/ml in BAL and a median peak of 7.00
SARS-CoV-2 infection protects against rechallenge in
rhesus macaques
Abishek Chandrashekar1*, Jinyan Liu1*, Amanda J. Martinot1,2*, Katherine McMahan1*,
Noe B. Mercado1*, Lauren Peter1*, Lisa H. Tostanoski1*, Jingyou Yu1*, Zoltan Maliga3,
Michael Nekorchuk4, Kathleen Busman-Sahay4, Margaret Terry4, Linda M. Wrijil2, Sarah Ducat2,
David R. Martinez5, Caroline Atyeo3,6, Stephanie Fischinger6, John S. Burke6, Matthew D. Slein6,
Laurent Pessaint7, Alex Van Ry7, Jack Greenhouse7, Tammy Taylor7, Kelvin Blade7, Anthony Cook7,
Brad Finneyfrock7, Renita Brown7, Elyse Teow7, Jason Velasco7, Roland Zahn8, Frank Wegmann8,
Peter Abbink1, Esther A. Bondzie1, Gabriel Dagotto1,3, Makda S. Gebre1,3, Xuan He1,
Catherine Jacob-Dolan1,3, Nicole Kordana1, Zhenfeng Li1, Michelle A. Lifton1, Shant H. Mahrokhian1,
Lori F. Maxfield1, Ramya Nityanandam1, Joseph P. Nkolola1, Aaron G. Schmidt6,9, Andrew D. Miller10,
Ralph S. Baric5, Galit Alter6,9, Peter K. Sorger3, Jacob D. Estes4, Hanne Andersen7, Mark G. Lewis7,
Dan H. Barouch1,6,9
1Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. 2Tufts University Cummings
School of Veterinary Medicine, North Grafton, MA 01536, USA. 3Harvard Medical School, Boston, MA 02115, USA. 4Oregon Health & Sciences University, Beaverton,
OR 97006, USA. 5University of North Carolina, Chapel Hill, NC 27599, USA. 6Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA. 7Bioqual,
Rockville, MD 20852, USA. 8Janssen Vaccines & Prevention BV, Leiden, Netherlands. 9Massachusetts Consortium on Pathogen Readiness, Boston, MA 02215, USA.
10Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA.
*These authors contributed equally to this work.
†Corresponding author. Email: dbarouch@bidmc.harvard.edu
An understanding of protective immunity to SARS-CoV-2 is critical for vaccine and public health strategies
aimed at ending the global COVID-19 pandemic. A key unanswered question is whether infection with
SARS-CoV-2 results in protective immunity against re-exposure. We developed a rhesus macaque model of
SARS-CoV-2 infection and observed that macaques had high viral loads in the upper and lower respiratory
tract, humoral and cellular immune responses, and pathologic evidence of viral pneumonia. Following
initial viral clearance, animals were rechallenged with SARS-CoV-2 and showed 5 log10 reductions in
median viral loads in bronchoalveolar lavage and nasal mucosa compared with primary infection.
Anamnestic immune responses following rechallenge suggested that protection was mediated by
immunologic control. These data show that SARS-CoV-2 infection induced protective immunity against re-
exposure in nonhuman primates.
on May 20, 2020 http://science.sciencemag.org/Downloaded from
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 2
(range 5.06-8.55) log10 RNA copies/swab in NS. Viral RNA in
NS increased in all animals from day 1 to day 2, suggesting
viral replication. Viral RNA peaked on day 2 and typically
resolved by day 10-14 in BAL and by day 21-28 in NS. Fol-
lowing day 2, viral loads in BAL and NS appeared compara-
ble in all groups regardless of dose. Viral RNA was
undetectable in plasma (fig. S1). Animals exhibited modestly
decreased appetite and responsiveness suggestive of mild
clinical disease (fig. S2) as well as mild transient neutro-
penia and lymphopenia in the high dose group (fig. S3), but
fever, weight loss, respiratory distress, and mortality were
not observed.
To help differentiate input challenge virus from newly
replicating virus, we developed an RT-PCR assay to assess E
gene subgenomic mRNA (sgmRNA). E gene sgmRNA re-
flects viral replication cellular intermediates that are not
packaged into virions and thus represent putative replicat-
ing virus in cells (9). Compared with total viral RNA (Fig.
1B), sgmRNA levels were lower in NS on day 1 with a medi-
an of 5.11 (range <1.70-5.94) log10 sgmRNA copies/swab, but
then increased by day 2 to a median of 6.50 (range 4.16-7.81)
log10 sgmRNA copies/swab (Fig. 1C).
We next evaluated SARS-CoV-2-specific humoral and
cellular immune responses in these animals. All 9 macaques
developed binding antibody responses to the SARS-CoV-2
Spike (S) protein by ELISA (Fig. 2A) and neutralizing anti-
body (NAb) responses using both a pseudovirus neutraliza-
tion assay (10) (Fig. 2B) and a live virus neutralization assay
(11, 12) (Fig. 2C). NAb titers of approximately 100 were ob-
served in all animals on day 35 regardless of dose group
(range 83-197 by the pseudovirus neutralization assay and
35-326 by the live virus neutralization assay). Antibody re-
sponses of multiple subclasses were observed against the
receptor binding domain (RBD), the prefusion S ectodomain
(S), and the nucleocapsid (N), and antibodies exhibited di-
verse effector functions, including antibody-dependent
complement deposition (ADCD), antibody-dependent cellu-
lar phagocytosis (ADCP), antibody-dependent neutrophil
phagocytosis (ADNP), and antibody-dependent NK cell
degranulation (NK CD107a) and cytokine secretion (NK
MIP1β, NK IFNγ) (13) (Fig. 2D). Cellular immune responses
to pooled S peptides were observed in the majority of ani-
mals by IFN-γ ELISPOT assays on day 35, with a trend to-
ward lower responses in the lower dose groups (Fig. 2E).
Intracellular cytokine staining assays demonstrated induc-
tion of both S-specific CD8+ and CD4+ T cell responses (Fig.
2F).
SARS CoV-2 infection induces acute viral interstitial
pneumonia in rhesus macaques
Only limited pathology data from SARS-CoV-2 infected hu-
mans are currently available. To assess the pathologic char-
acteristics of SARS-CoV-2 infection in rhesus macaques, we
inoculated 4 animals with 1.1 × 105 PFU virus by the IN and
IT routes as above and necropsied them on day 2 (N = 2)
and day 4 (N = 2) following challenge. Multiple regions of
the upper respiratory tract, lower respiratory tract, gastroin-
testinal tract, lymph nodes, and other organs were harvest-
ed for virologic and pathologic analyses. High levels of viral
RNA were observed in all nasal mucosa, pharynx, trachea,
and lung tissues, and lower levels of virus were found in the
gastrointestinal tract, liver, and kidney (fig. S4). Viral RNA
was readily detected in paratracheal lymph nodes but was
only sporadically found in distal lymph nodes and spleen
(fig. S4).
Upper airway mucosae, trachea, and lungs were para-
formaldehyde fixed, paraffin embeded, and evaluated by
histopathology. On day 2 following challenge, both necrop-
sied animals demonstrated multifocal regions of inflamma-
tion and evidence of viral pneumonia, including expansion
of alveolar septae with mononuclear cell infiltrates, consoli-
dation, and edema (Fig. 3, A and B). Regions with edema
also contained numerous polymorphonuclear cells, predom-
inantly neutrophils. Terminal bronchiolar epithelium was
necrotic and sloughed with clumps of epithelial cells detect-
ed within airways and distally within alveolar spaces (Fig. 3,
C and D) with formation of occasional bronchiolar epithelial
syncytial cells (Fig. 3E). Hyaline membranes were occasion-
ally observed within alveolar septa, consistent with damage
to type I and type II pneumocytes (Fig. 3F). Diffusely reac-
tive alveolar macrophages filled alveoli, and some were mul-
tinucleated and labeled positive for nucleocapsid by
immunohistochemistry (Fig. 3G). Alveolar lining cells
(pneumocytes) also prominently labeled positive for nucle-
ocapsid (Fig. 3H).
Multifocal clusters of virus infected cells were present
throughout the lung parenchyma, as detected by immuno-
histochemistry and in situ RNA hybridization (RNAscope)
(14, 15) (Fig. 3I). Both positive-sense and negative-sense viral
RNA was observed by RNAscope (fig. S5), suggesting viral
replication in lung tissue. The dense inflammatory infil-
trates included polymorphonuclear cells detected by endog-
enous myeloperoxidase staining (MPO), CD68 and CD163
positive macrophages, CD4+ and CD8+ T lymphocytes, and
diffuse up-regulation of the type 1 interferon gene MX1 (fig.
S6). SARS-CoV-2 infection led to a significant increase in
polymorphonuclear cell infiltration of lung alveoli compared
with uninfected animals (P = 0.0286) as well as extensive
MX1 staining in approximately 30% of total lung tissue (P =
0.0286) (fig. S7). Inflammatory infiltrates were also detected
in the respiratory epithelial submucosa of larger airways
with transmigration of inflammatory cells into bronchiole
lumen (Fig. 3J). Ciliated epithelial cells also stained positive
for both SARS CoV-2 RNA (Fig. 3K) and SARS nucleocapsid
on May 20, 2020 http://science.sciencemag.org/Downloaded from
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 3
(Fig. 3L). By day 4 following infection, the extent of inflam-
mation and viral pneumonia had diminished, but virus was
still detected in lung parenchyma and neutrophil infiltra-
tion and type 1 interferon responses persisted (fig. S7).
To further characterize infected tissues, we performed
cyclic immunofluorescence (CyCIF) imaging, a method for
multiplex immunophenotyping of paraformaldehyde fixed
tissue specimens (16). Tissues were stained for nucleocapsid
(SARS-N), pan-cytokeratin (to identify epithelial cells), Iba-1
(ionized calcium binding adaptor as a pan-macrophage
marker), CD68 (monocyte/macrophage marker), and CD206
(macrophage marker), in addition to a panel of markers to
identify other immune cells and anatomical structures (ta-
ble S1), and counterstaining for DNA to label all nuclei. Foci
of virus infected cells were randomly dispersed throughout
the lung and were variably associated with inflammatory
infiltrates (Fig. 4, A to D). Some areas of parenchymal con-
solidation and inflammation contained little to no virus
(Fig. 4A, arrows, and fig. S8). Virus infected cells frequently
co-stained with pan-cytokeratin (Fig. 4, E to H), suggesting
that they were alveolar epithelial cells (pneumocytes). Unin-
fected Iba-1+ CD68+ CD206+ activated macrophages were
also frequently detected adjacent to virally infected epitheli-
al cells (Fig. 4, E and I to K). These data demonstrate that
SARS-CoV-2 induced multifocal areas of acute inflammation
and viral pneumonia involving infected pneumocytes, ciliat-
ed bronchial epithelial cells, and likely other cell types.
Protective efficacy against rechallenge with SARS-CoV-
2 in rhesus macaques
On day 35 following initial viral infection (Figs. 1 and 2), we
rechallenged all 9 rhesus macaques with the same doses of
SARS-CoV-2 that were utilized for the primary infection,
namely 1.1 × 106 PFU (Group 1; N = 3), 1.1 × 105 PFU (Group
2; N = 3), or 1.1 × 104 PFU (Group 3; N = 3). We included 3
naïve animals as positive controls in the rechallenge exper-
iment. Very limited viral RNA was observed in BAL on day 1
following rechallenge in two Group 1 animals and in one
Group 2 animal, with no viral RNA detected at subsequent
timepoints (Fig. 5A). In contrast, high levels of viral RNA
were observed in the concurrently challenged naïve animals
(Fig. 5A), as expected. Median peak viral loads in BAL were
>5.1 log10 lower following rechallenge as compared with the
primary challenge (P < 0.0001, two-sided Mann-Whitney
test; Fig. 5B). Viral RNA following rechallenge was higher in
NS compared with BAL, but exhibited dose dependence and
rapid decline (Fig. 5C), and median peak viral loads in NS
were still >1.7 log10 lower following rechallenge as compared
with the primary challenge (P = 0.0011, two-sided Mann-
Whitney test; Fig. 5D).
We speculated that the majority of virus detected in NS
following rechallenge was input challenge virus, and we
therefore assessed sgmRNA levels in NS following rechal-
lenge. Low but detectable levels of sgmRNA were still ob-
served in 4 of 9 animals in NS on day 1 following
rechallenge, but sgmRNA levels declined quickly (Fig. 5E),
and median peak sgmRNA levels in NS were >4.8 log10 lower
following rechallenge as compared with the primary chal-
lenge (P = 0.0003, two-sided Mann-Whitney test; Fig. 5F).
Consistent with these data, plaque assays in BAL and NS
samples following rechallenge showed no recoverable virus
and were lower than following primary infection (P = 0.009
and 0.002, respectively, two-sided Mann-Whitney tests; fig.
S9). Moreover, little or no clinical disease was observed in
the animals following rechallenge (fig. S10).
Following SARS-CoV-2 rechallenge, animals exhibited
rapid anamnestic immune responses, including increased
virus-specific ELISA titers (P = 0.0034, two-sided Mann-
Whitney test), pseudovirus NAb titers (P = 0.0003), and live
virus NAb titers (P = 0.0003) as well as a trend toward in-
creased IFN-γ ELISPOT responses (P = 0.1837) by day 7 after
rechallenge (Fig. 6). In particular, NAb titers were markedly
higher on day 14 following rechallenge compared with day
14 following primary challenge (P < 0.0001, two-sided
Mann-Whitney test) (fig. S11). All animals developed anam-
nestic antibody responses following rechallenge, regardless
of the presence or absence of residual viral RNA or sgmRNA
in BAL or NS, and thus we speculate that the protective effi-
cacy against rechallenge was mediated by rapid immunolog-
ic control.
Discussion
Individuals who recover from certain viral infections typi-
cally develop virus-specific antibody responses that provide
robust protective immunity against re-exposure, but some
viruses do not generate protective natural immunity, such
as HIV-1 (17). Human challenge studies for the common cold
coronavirus 229E have suggested that there may be partial
natural immunity (18). However, there is currently no data
whether humans who have recovered from SARS-CoV-2 in-
fection are protected from re-exposure (World Health Or-
ganization, Scientific Brief, April 24, 2020; https://
www.who.int/news-room/commentaries/detail/immunity-
passports-in-the-context-of-covid-19). This is a critical issue
with profound implications for vaccine development, public
health strategies, antibody-based therapeutics, and epide-
miologic modeling of herd immunity. In this study, we
demonstrate that SARS-CoV-2 infection in rhesus macaques
provided protective efficacy against SARS-CoV-2 rechal-
lenge.
We developed a rhesus macaque model of SARS-CoV-2
infection that recapitulates many aspects of human SARS-
CoV-2 infection, including high levels of viral replication in
the upper and lower respiratory tract (Fig. 1) and clear
on May 20, 2020 http://science.sciencemag.org/Downloaded from
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 4
pathologic evidence of viral pneumonia (Figs. 3 and 4). His-
topathology, immunohistochemistry, RNAscope, and CyCIF
imaging demonstrated multifocal clusters of virus infected
cells in areas of acute inflammation, with evidence for virus
infection of alveolar pneumocytes and ciliated bronchial
epithelial cells. These data suggest the utility of rhesus ma-
caques as a model for SARS-CoV-2 infection for testing vac-
cines and therapeutics and for studying
immunopathogenesis, and our findings complement and
extend recently published data in cynomolgus macaques
(19). However, neither nonhuman primate model led to res-
piratory failure or mortality, and thus further research will
be required to develop a nonhuman primate model of severe
COVID-19 disease.
SARS-CoV-2 infection in rhesus macaques led to hu-
moral and cellular immune responses (Fig. 2) and provided
protection against rechallenge (Fig. 5). Residual low levels of
subgenomic mRNA in nasal swabs in a subset of animals
(Fig. 5) and anamnestic immune responses in all animals
(Fig. 6) following SARS-CoV-2 rechallenge suggest that pro-
tection was mediated by immunologic control and likely
was not sterilizing.
Given the near-complete protection in all animals fol-
lowing SARS-CoV-2 rechallenge, we were unable to deter-
mine immune correlates of protection in this study. SARS-
CoV-2 infection in rhesus monkeys resulted in the induction
of neutralizing antibody titers of approximately 100 by both
a pseudovirus neutralization assay and a live virus neutrali-
zation assay, but the relative importance of neutralizing an-
tibodies, other functional antibodies, cellular immunity, and
innate immunity to protective efficacy against SARS-CoV-2
remains to be determined. Moreover, additional research
will be required to define the durability of natural immuni-
ty.
In summary, SARS-CoV-2 infection in rhesus macaques
induced humoral and cellular immune responses and pro-
vided protective efficacy against SARS-CoV-2 rechallenge.
These data raise the possibility that immunologic approach-
es to the prevention and treatment of SARS-CoV-2 infection
may in fact be possible. However, it is critical to emphasize
that there are important differences between SARS-CoV-2
infection in macaques and humans, with many parameters
still yet to be defined in both species, and thus our data
should be interpreted cautiously. Rigorous clinical studies
will be required to determine whether SARS-CoV-2 infection
effectively protects against SARS-CoV-2 re-exposure in hu-
mans.
REFERENCES AND NOTES
1. F. Wu, S. Zhao, B. Yu, Y.-M. Chen, W. Wang, Z.-G. Song, Y. Hu, Z.-W. Tao, J.-H. Tian,
Y.-Y. Pei, M.-L. Yuan, Y.-L. Zhang, F.-H. Dai, Y. Liu, Q.-M. Wang, J.-J. Zheng, L. Xu,
E. C. Holmes, Y.-Z. Zhang, A new coronavirus associated with human respiratory
disease in China. Nature 579, 265269 (2020). doi:10.1038/s41586-020-2008-
3 Medline
2. P. Zhou, X.-L. Yang, X.-G. Wang, B. Hu, L. Zhang, W. Zhang, H.-R. Si, Y. Zhu, B. Li,
C.-L. Huang, H.-D. Chen, J. Chen, Y. Luo, H. Guo, R.-D. Jiang, M.-Q. Liu, Y. Chen,
X.-R. Shen, X. Wang, X.-S. Zheng, K. Zhao, Q.-J. Chen, F. Deng, L.-L. Liu, B. Yan,
F.-X. Zhan, Y.-Y. Wang, G.-F. Xiao, Z.-L. Shi, A pneumonia outbreak associated
with a new coronavirus of probable bat origin. Nature 579, 270273 (2020).
doi:10.1038/s41586-020-2012-7 Medline
3. M. L. Holshue, C. DeBolt, S. Lindquist, K. H. Lofy, J. Wiesman, H. Bruce, C. Spitters,
K. Ericson, S. Wilkerson, A. Tural, G. Diaz, A. Cohn, L. Fox, A. Patel, S. I. Gerber, L.
Kim, S. Tong, X. Lu, S. Lindstrom, M. A. Pallansch, W. C. Weldon, H. M. Biggs, T.
M. Uyeki, S. K. Pillai, Washington State 2019-nCoV Case Investigation Team,
First Case of 2019 Novel Coronavirus in the United States. N. Engl. J. Med. 382,
929936 (2020). doi:10.1056/NEJMoa2001191 Medline
4. Q. Li, X. Guan, P. Wu, X. Wang, L. Zhou, Y. Tong, R. Ren, K. S. M. Leung, E. H. Y.
Lau, J. Y. Wong, X. Xing, N. Xiang, Y. Wu, C. Li, Q. Chen, D. Li, T. Liu, J. Zhao, M.
Liu, W. Tu, C. Chen, L. Jin, R. Yang, Q. Wang, S. Zhou, R. Wang, H. Liu, Y. Luo, Y.
Liu, G. Shao, H. Li, Z. Tao, Y. Yang, Z. Deng, B. Liu, Z. Ma, Y. Zhang, G. Shi, T. T. Y.
Lam, J. T. Wu, G. F. Gao, B. J. Cowling, B. Yang, G. M. Leung, Z. Feng, Early
Transmission Dynamics in Wuhan, China, of Novel Coronavirus-Infected
Pneumonia. N. Engl. J. Med. 382, 11991207 (2020).
doi:10.1056/NEJMoa2001316 Medline
5. N. Zhu, D. Zhang, W. Wang, X. Li, B. Yang, J. Song, X. Zhao, B. Huang, W. Shi, R. Lu,
P. Niu, F. Zhan, X. Ma, D. Wang, W. Xu, G. Wu, G. F. Gao, W. Tan, China Novel
Coronavirus Investigating and Research Team, A Novel Coronavirus from
Patients with Pneumonia in China, 2019. N. Engl. J. Med. 382, 727733 (2020).
doi:10.1056/NEJMoa2001017 Medline
6. N. Chen, M. Zhou, X. Dong, J. Qu, F. Gong, Y. Han, Y. Qiu, J. Wang, Y. Liu, Y. Wei, J.
Xia, T. Yu, X. Zhang, L. Zhang, Epidemiological and clinical characteristics of 99
cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive
study. Lancet 395, 507513 (2020). doi:10.1016/S0140-6736(20)30211-7
Medline
7. C. Huang, Y. Wang, X. Li, L. Ren, J. Zhao, Y. Hu, L. Zhang, G. Fan, J. Xu, X. Gu, Z.
Cheng, T. Yu, J. Xia, Y. Wei, W. Wu, X. Xie, W. Yin, H. Li, M. Liu, Y. Xiao, H. Gao, L.
Guo, J. Xie, G. Wang, R. Jiang, Z. Gao, Q. Jin, J. Wang, B. Cao, Clinical features of
patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395,
497–506 (2020). doi:10.1016/S0140-6736(20)30183-5 Medline
8. J. F. Chan, S. Yuan, K.-H. Kok, K. K.-W. To, H. Chu, J. Yang, F. Xing, J. Liu, C. C.-Y.
Yip, R. W.-S. Poon, H.-W. Tsoi, S. K.-F. Lo, K.-H. Chan, V. K.-M. Poon, W.-M. Chan,
J. D. Ip, J.-P. Cai, V. C.-C. Cheng, H. Chen, C. K.-M. Hui, K.-Y. Yuen, A familial
cluster of pneumonia associated with the 2019 novel coronavirus indicating
person-to-person transmission: A study of a family cluster. Lancet 395, 514523
(2020). doi:10.1016/S0140-6736(20)30154-9 Medline
9. R. Wölfel, V. M. Corman, W. Guggemos, M. Seilmaier, S. Zange, M. A. Müller, D.
Niemeyer, T. C. Jones, P. Vollmar, C. Rothe, M. Hoelscher, T. Bleicker, S. Brünink,
J. Schneider, R. Ehmann, K. Zwirglmaier, C. Drosten, C. Wendtner, Virological
assessment of hospitalized patients with COVID-2019. Nature (2020).
doi:10.1038/s41586-020-2196-x Medline
10. Z. Y. Yang, W. P. Kong, Y. Huang, A. Roberts, B. R. Murphy, K. Subbarao, G. J.
Nabel, A DNA vaccine induces SARS coronavirus neutralization and protective
immunity in mice. Nature 428, 561564 (2004). doi:10.1038/nature02463
Medline
11. T. Scobey, B. L. Yount, A. C. Sims, E. F. Donaldson, S. S. Agnihothram, V. D.
Menachery, R. L. Graham, J. Swanstrom, P. F. Bove, J. D. Kim, S. Grego, S. H.
Randell, R. S. Baric, Reverse genetics with a full-length infectious cDNA of the
Middle East respiratory syndrome coronavirus. Proc. Natl. Acad. Sci. U.S.A. 110,
1615716162 (2013). doi:10.1073/pnas.1311542110 Medline
12. B. Yount, K. M. Curtis, E. A. Fritz, L. E. Hensley, P. B. Jahrling, E. Prentice, M. R.
Denison, T. W. Geisbert, R. S. Baric, Reverse genetics with a full-length infectious
cDNA of severe acute respiratory syndrome coronavirus. Proc. Natl. Acad. Sci.
U.S.A. 100, 1299513000 (2003). doi:10.1073/pnas.1735582100 Medline
on May 20, 2020 http://science.sciencemag.org/Downloaded from
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 5
13. A. W. Chung, M. P. Kumar, K. B. Arnold, W. H. Yu, M. K. Schoen, L. J. Dunphy, T. J.
Suscovich, N. Frahm, C. Linde, A. E. Mahan, M. Hoffner, H. Streeck, M. E.
Ackerman, M. J. McElrath, H. Schuitemaker, M. G. Pau, L. R. Baden, J. H. Kim, N.
L. Michael, D. H. Barouch, D. A. Lauffenburger, G. Alter, Dissecting Polyclonal
Vaccine-Induced Humoral Immunity against HIV Using Systems Serology. Cell
163, 988998 (2015). doi:10.1016/j.cell.2015.10.027 Medline
14. C. Deleage, A. Schuetz, W. G. Alvord, L. Johnston, X.-P. Hao, D. R. Morcock, R.
Rerknimitr, J. L. K. Fletcher, S. Puttamaswin, N. Phanuphak, R. Dewar, J. M.
McCune, I. Sereti, M. Robb, J. H. Kim, T. W. Schacker, P. Hunt, J. D. Lifson, J.
Ananworanich, J. D. Estes, Impact of early cART in the gut during acute HIV
infection. JCI Insight 1, e87065 (2016). doi:10.1172/jci.insight.87065 Medline
15. C. Deleage, S. W. Wietgrefe, G. Del Prete, D. R. Morcock, X.-P. Hao, M. Piatak Jr.,
J. Bess, J. L. Anderson, K. E. Perkey, C. Reilly, J. M. McCune, A. T. Haase, J. D.
Lifson, T. W. Schacker, J. D. Estes, Defining HIV and SIV Reservoirs in Lymphoid
Tissues. Pathog. Immun. 1, 68106 (2016). doi:10.20411/pai.v1i1.100 Medline
16. J. R. Lin, B. Izar, S. Wang, C. Yapp, S. Mei, P. M. Shah, S. Santagata, P. K. Sorger,
Highly multiplexed immunofluorescence imaging of human tissues and tumors
using t-CyCIF and conventional optical microscopes. eLife 7, e31657 (2018).
doi:10.7554/eLife.31657 Medline
17. M. Altfeld, T. M. Allen, X. G. Yu, M. N. Johnston, D. Agrawal, B. T. Korber, D. C.
Montefiori, D. H. O’Connor, B. T. Davis, P. K. Lee, E. L. Maier, J. Harlow, P. J. R.
Goulder, C. Brander, E. S. Rosenberg, B. D. Walker, HIV-1 superinfection despite
broad CD8+ T-cell responses containing replication of the primary virus. Nature
420, 434439 (2002). doi:10.1038/nature01200 Medline
18. K. A. Callow, H. F. Parry, M. Sergeant, D. A. Tyrrell, The time course of the
immune response to experimental coronavirus infection of man. Epidemiol.
Infect. 105, 435446 (1990). doi:10.1017/S0950268800048019 Medline
19. B. Rockx, T. Kuiken, S. Herfst, T. Bestebroer, M. M. Lamers, B. B. Oude Munnink,
D. de Meulder, G. van Amerongen, J. van den Brand, N. M. A. Okba, D. Schipper,
P. van Run, L. Leijten, R. Sikkema, E. Verschoor, B. Verstrepen, W. Bogers, J.
Langermans, C. Drosten, M. Fentener van Vlissingen, R. Fouchier, R. de Swart, M.
Koopmans, B. L. Haagmans, Comparative pathogenesis of COVID-19, MERS, and
SARS in a nonhuman primate model. Science eabb7314 (2020).
doi:10.1126/science.abb7314 Medline
ACKNOWLEDGMENTS
We thank B. Walker, A. Chakraborty, K. Reeves, B. Chen, J. Feldman, B. Hauser, T.
Caradonna, S. Bondoc, C. Starke, C. Jacobson, D. O’Connor, S. O’Connor, N. Thorn-
burg, E. Borducchi, M. Silva, A. Richardson, C. Caron, and J. Cwiak for generous
advice, assistance, and reagents. Funding: We acknowledge support from the Ragon
Institute of MGH, MIT, and Harvard, Mark and Lisa Schwartz Foundation, Beth Israel
Deaconess Medical Center, Massachusetts Consortium on Pathogen Readiness
(MassCPR), Bill & Melinda Gates Foundation (INV-006131), Janssen Vaccines &
Prevention BV, and the National Institutes of Health (OD024917, AI129797,
AI124377, AI128751, AI126603 to D.H.B.; AI135098 to A.J.M.; AI007387 to L.H.T.;
AI007151 to D.R.M.; AI146779 to A.G.S.; 272201700036I-0-759301900131-1,
AI100625, AI110700, AI132178, AI149644, AI108197 to R.S.B.; CA225088 to P.K.S.;
and OD011092, OD025002 to J.D.E.). We also acknowledge a Fast Grant, Emergent
Ventures, Mercatus Center at George Mason University to A.J.M. and a Burroughs
Wellcome Fund Postdoctoral Enrichment Program Award to D.R.M. Author contri-
butions: D.H.B., H.A., and M.G.L. designed the study. A.C., J.L., K.M., N.B.M., L.P.,
L.H.T., J.Y., P.A., E.A.B., G.D., M.S.G., X.H., C.J.D., N.K., Z.L., M.A.L., L.F.M., and J.P.N.
performed the immunologic and virologic assays. A.J.M., Z.M., M.N., K.B.S., M.T.
L.M.W., S.D., A.D.M., P.K.S., and J.D.E. performed the pathology studies. D.R.M. and
R.S.B. performed the live virus neutralization assays. C.A., S.F., J.S.B., M.D.S., and
G.A. performed the antibody phenotyping. L.P., A.V.R., J.G., T.T., K.B., A.C., B.F.,
R.B., E.T., J.V., H.A., and M.G.L. led the clinical care of the animals and performed the
virologic assays. R.Z. and F.W. participated in study design and interpretation of
data. A.G.S. and B.C. provided purified proteins. D.H.B. wrote the paper with all co-
authors. Competing interests: The authors declare no competing financial interests.
G.A. is an inventor on patent application WO 2017/184733 A1 submitted by Massa-
chusetts General Hospital that covers systems serology. R.Z. and F.W. are employ-
ees of Janssen Vaccines & Prevention BV. Data and materials availability: All data
are available in the manuscript or the supplementary material. Virus stocks are
available from D.H.B. under a material transfer agreement with Beth Israel Deacon-
ess Medical Center. This work is licensed under a Creative Commons Attribution 4.0
International (CC BY 4.0) license, which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly cited. To view a
copy of this license, visit https://creativecommons.org/licenses/by/4.0/. This
license does not apply to figures/photos/artwork or other content included in the
article that is credited to a third party; obtain authorization from the rights holder
before using such material.
SUPPLEMENTARY MATERIALS
science.sciencemag.org/cgi/content/full/science.abc4776/DC1
Materials and Methods
Table S1
Figs. S1 to S11
26 April 2020; accepted 16 May 2020
Published online 20 May 2020
10.1126/science.abc4776
on May 20, 2020 http://science.sciencemag.org/Downloaded from
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 6
Fig. 1. Viral loads in SARS-CoV-2 challenged rhesus macaques.
Rhesus macaques were inoculated by the
intranasal and intratracheal route with 1.1 × 106 PFU (Group 1; N = 3), 1.1 × 105 PFU (Group 2; N = 3), or 1.1 × 104
PFU (Group 3; N = 3) SARS-CoV-2. (
A
) Log10 viral RNA copies/ml (limit 50 copies/ml) were assessed in
bronchoalveolar lavage (BAL) at multiple timepoints following challenge. (
B
and
C
) Log10 viral RNA copies/swab
(B) and log10 sgmRNA copies/swab (limit 50 copies/swab) (C) were assessed in nasal swabs (NS) at multiple
timepoints following challenge. Red horizontal bars reflect median viral loads.
on May 20, 2020 http://science.sciencemag.org/Downloaded from
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 7
(
to
)
Humoral immune responses were assessed
ELISA (A), pseudovirus neutralization assays
(B), live virus neutralization assays (C), and
antibody subclasses and effector functions
to receptor binding domain (RBD), soluble
spike (S) ectodomain, and nucleocapsid (N)
proteins on day 35. Antibody-dependent
complement deposition (ADCD), antibody-
antibody-dependent neutrophil phagocytosis
(ADNP), and antibody-
secretion (NK MIP1β, NK IFNγ) are shown. (
and
) Cellular immune responses were also
assessed following challenge by IFN-γ
intracellular cytokine staining assays (F) in
horizontal bars reflect mean responses.
on May 20, 2020 http://science.sciencemag.org/Downloaded from
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 8
Fig. 3. SARS-CoV-2 induces acute viral interstitial pneumonia.
(
A
to
F
) H&E sections of fixed lung tissue from
SARS-CoV-2 infected rhesus macaques 2 days following challenge showing interstitial edema and regional lung
consolidation (A), intra-alveolar edema and infiltrates of neutrophils (B), bronchiolar epithelial sloughing and
necrosis [(C) and (D)], bronchiolar epithelial syncytial cell formation (E), and hyaline membranes within alveolar
septa (F). (
G
and
H
) IHC for SARS nucleocapsid showing virus infected cells within interstitial spaces including a
viral syncytial cell within the lumen (G) and virus infected alveolar lining cells (H). (
I
) Inflammatory infiltrate
showing multiple cells containing SARS-CoV-2 RNA by RNAscope in situ hybridization. (
J
to
L
) bronchial
respiratory epithelium showing inflammation within the submucosa and transmigration of inflammatory cells into
the ciliated columnar respiratory epithelium of a bronchus (J), SARS-CoV-2 RNA (K), and SARS nucleocapsid (L).
Scale bars (A) = 200 microns; (C, I, K-L) = 100 micron; (G) = 50 micron; (B, D-F, J) = 20 microns, and (H) = 10
microns. H&E = hematoxylin and eosin; IHC = immunohistochemistry; RNAscope = SARS-CoV-2 RNA staining.
on May 20, 2020 http://science.sciencemag.org/Downloaded from
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 9
Fig. 4. SARS-CoV-2 infects alveolar epithelial cells in rhesus macaques.
Cyclic immunofluorescence (CyCIF)
staining of fixed lung tissue from SARS-CoV-2 infected rhesus macaques 2 days following challenge. (
A
) Whole
slide image of a lung stained with Hoechst 33342 to visualize cell nuclei (greyscale); regions of nuclear
consolidation (arrows), and foci of viral replication (box) are highlighted. (
B
) Higher magnification image of inset
box in (A) showing staining for SARS nucleocapsid protein (SARS-N; green) and cell nuclei (grey scale). (
C
) Higher
magnification image of inset box in (B) showing SARS-N (green) and cell nuclei (blue). (
D
) Bright-field IHC for
SARS-N from corresponding lung region depicted in (C). (
E
to
K
) CyCIF staining for DNA (blue; all panels) and
SARS-N [(E), (F), and (H-K); green], CD206 [(E) and (K); magenta], pan-cytokeratin (pan-CK) [(G) and (H); red],
CD68 (I; yellow), or Iba-1 (J; greyscale) showing virus infected epithelial cells and macrophages near an infected
epithelial cell. Scale bars (F-K) = 50 microns.
on May 20, 2020 http://science.sciencemag.org/Downloaded from
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 10
Fig. 5.
Viral loads following SARS-
CoV-
2 rechallenge in rhesus
macaques. On day 35 following
initial infection (Fig. 1), rhesus
macaques were rechallenged with
SARS-CoV-2 by the intranasal and
intratracheal route with 1.1 × 106
PFU (Group 1; N = 3), 1.1 × 105 PFU
(Group 2; N = 3), or 1.1 × 104 PFU
(Group 3; N = 3). Three naïve
animals were included as a positive
control in the rechallenge experi-
ment. (
A
) Log10 viral RNA copies/
ml (limit 50 copies/ml) were
assessed in bronchoalveolar lavage
(BAL) at multiple timepoints
following rechallenge. One of the
naïve animals could not be lavaged.
(
B
) Comparison of viral RNA in BAL
following primary challenge and
rechallenge. (
C
and
E
) Log10 viral
RNA copies/ml (C) and log10
sgmRNA copies/swab (limit 50
copies/ml) (E) were assessed in
nasal swabs (NS) at multiple
timepoints following rechallenge.
(
D
and
F
) Comparison of viral RNA
(D) and sgmRNA (F) in NS following
primary challenge and rechallenge.
Red horizontal bars reflect median
viral loads. P-values reflect two-
sided Mann-Whitney tests.
on May 20, 2020 http://science.sciencemag.org/Downloaded from
First release: 20 May 2020 www.sciencemag.org (Page numbers not final at time of first release) 11
Fig. 6. Anamnestic immune responses following SARS-CoV-2 rechallenge in rhesus
macaques.
Binding antibody ELISAs, pseudovirus neutralization assays, live virus neutralization
assays, and IFN-γ ELISPOT assays are depicted prior to and 7 days following SARS-CoV-2
rechallenge. Red lines reflect mean responses. P-values reflect two-sided Mann-Whitney tests.
on May 20, 2020 http://science.sciencemag.org/Downloaded from
SARS-CoV-2 infection protects against rechallenge in rhesus macaques
and Dan H. Barouch
Aaron G. Schmidt, Andrew D. Miller, Ralph S. Baric, Galit Alter, Peter K. Sorger, Jacob D. Estes, Hanne Andersen, Mark G. Lewis
Nicole Kordana, Zhenfeng Li, Michelle A. Lifton, Shant H. Mahrokhian, Lori F. Maxfield, Ramya Nityanandam, Joseph P. Nkolola,
Zahn, Frank Wegmann, Peter Abbink, Esther A. Bondzie, Gabriel Dagotto, Makda S. Gebre, Xuan He, Catherine Jacob-Dolan,
Greenhouse, Tammy Taylor, Kelvin Blade, Anthony Cook, Brad Finneyfrock, Renita Brown, Elyse Teow, Jason Velasco, Roland
Martinez, Caroline Atyeo, Stephanie Fischinger, John S. Burke, Matthew D. Slein, Laurent Pessaint, Alex Van Ry, Jack
Jingyou Yu, Zoltan Maliga, Michael Nekorchuk, Kathleen Busman-Sahay, Margaret Terry, Linda M. Wrijil, Sarah Ducat, David R.
Abishek Chandrashekar, Jinyan Liu, Amanda J. Martinot, Katherine McMahan, Noe B. Mercado, Lauren Peter, Lisa H. Tostanoski,
published online May 20, 2020
ARTICLE TOOLS http://science.sciencemag.org/content/early/2020/05/19/science.abc4776
MATERIALS
SUPPLEMENTARY http://science.sciencemag.org/content/suppl/2020/05/19/science.abc4776.DC1
CONTENT
RELATED
http://stm.sciencemag.org/content/scitransmed/9/396/eaal3653.full
http://stm.sciencemag.org/content/scitransmed/11/499/eaat0360.full
http://stm.sciencemag.org/content/scitransmed/12/534/eabb1469.full
http://stm.sciencemag.org/content/scitransmed/12/541/eabb5883.full
REFERENCES http://science.sciencemag.org/content/early/2020/05/19/science.abc4776#BIBL
This article cites 19 articles, 3 of which you can access for free
PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions
Terms of ServiceUse of this article is subject to the
is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title
(print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience
No claim to original U.S. Government Works
Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science.
on May 20, 2020 http://science.sciencemag.org/Downloaded from
... The systemic-and tissue-level NK cell depletion achieved by the anti-IL-15 mAb administration was highly successful (Fig 1B) with little to no adverse or off-target impacts, similar to previous reports by our group and others [23,48] and providing a clean model to evaluate the impacts on virology and pathology. The VL in our study in the pharynx peaked at 3DPI (Fig 2), consistent with reports by other groups using RM SARS-CoV-2 models that peak viral load occurs between 1-3DPI [76,77]. Although peak VL were only marginally higher, at all subsequent timepoints post infection, the NK celldepleted group had higher VL compared to the control group, reaching significance at 10DPI. ...
... Another phenotype of SARS-CoV-2-infected animals lacking NK cells was an overt increase in lung pathology and disease (Fig 5). The increased pathology was marked by focal fibrosis, syncytia, endothelialitis, and pneumocyte hyperplasia, all of which are reported by other groups in rodent and NHP models as well as fatal COVID-19 autopsy samples from humans [66,76,78,79]. Metadata analysis has indicated that post-COVID-19 pulmonary fibrosis manifested by the architectural distortion of the lung parenchyma affects 45% of recoverees [80]. ...
... This phenomenon was observed in both groups in our study. Pulmonary edema and the formation of bronchiolar epithelial syncytial cells were also reported in a RM infection (SARS-CoV-2 USA-WA1/2020 strain) model but were far less severe than seen in our NK celldepleted animals [76]. Remarkably the presence of endothelialitis, a characteristic feature of acute SARS-CoV-2 infection (observed 2-4 days post-infection in RMs) was observed in two out of the six CM in the NK cell-depleted group at necropsy and completely absent in the PLOS PATHOGENS Natural killer cells regulate SARS-CoV-2 infection control group [76]. ...
Article
Full-text available
Author summary Natural killer (NK) cells play a critically understudied role in controlling SARS-CoV-2 viral replication, clearance, and disease sequelae. In this manuscript, we investigated the protective role of NK cells in acute infection using a well-established NK cell depletion strategy in cynomolgus macaques (CM) and a SARS-CoV-2 Delta variant infection model. Circulating NK cells exhibited an increased proliferative and activated phenotype following infection, concomitant with peak NK cell expansion at 10 days post-infection (DPI). Importantly, following experimental NK cell depletion, CM exhibited increased viral shedding and delayed viral clearance compared to controls. NK cell-depleted animals also exhibited significantly increased lung pathology and Luminex cytokine analyses of broncho-alveolar lavage (BAL) fluid showed a 5-fold increase in interferon-alpha (IFNα) compared to controls during peak infection. Collectively, our findings suggest that NK cells play a crucial role in controlling SARS-CoV-2 replication and reducing lung damage. These results underscore the potential of NK cell-based vaccines and therapies for COVID-19 and other infectious diseases, warranting further investigation in this area.
... Non-replicating, lentiviral-based pseudoviruses expressing SARS-CoV-2 trimeric spike proteins are employed widely to assess SARS-CoV-2 neutralizing antibody titers in plasma and serum (24)(25)(26)(27). However, it is unclear whether these same assays are compatible with DBS eluates in terms of yielding SARS-CoV-2 neutralizing antibody titers in line with PRNT. ...
Article
Full-text available
In the earliest days of the COVID-19 pandemic, the collection of dried blood spots (DBS) enabled public health laboratories to undertake population-scale seroprevalence studies used to estimate rates of SARS-CoV-2 exposure. With SARS-CoV-2 seropositivity levels now estimated to exceed 94% in the United States, attention has turned to using DBS to assess neutralizing antibodies within cohorts of interest. With this goal in mind, we generated contrived DBS (cDBS) and whole blood-derived DBS from convalescent and vaccinated individuals and subjected DBS eluates to a battery of assays, including a SARS-CoV-2 multiplexed microsphere immunoassay (MIA), a receptor binding domain (RBD)-human ACE2 inhibition assay (iACE2), a cell-based pseudovirus neutralization assay, and real-time PCR-based surrogate neutralization assay (NAB-Sure). The DBS results were benchmarked against paired serum samples tested in a clinically validated SARS-CoV-2 plaque reduction neutralization titer (PRNT) assay. The results of an 8-plex MIA and NAB-Sure assays demonstrated highly significant correlations with PRNT values when evaluated with a panel of 86 paired serum–DBS samples. Both the MIA and NAB-Sure are adaptable to automated liquid handlers for high-throughput capacity. While neutralizing assays were limited to the ancestral SARS-CoV-2 WA1, this study nonetheless represents an important proof of concept demonstrating the potential utility of DBS as a biospecimen type for use in assessing immunity to SARS-CoV-2 at the community and population levels. IMPORTANCE SARS-CoV-2 variants of concern continue to circulate globally and remain a serious health threat to large segments of the population. From a public health standpoint, identifying vulnerable communities based on immune status is critical in terms of vaccine booster recommendations. In this report, we investigated the utility of dried blood spots (DBS) as a biospecimen type from which to estimate SARS-CoV-2 neutralizing antibody titers. Using contrived and whole blood-derived DBS, we demonstrate that SARS-CoV-2 neutralizing antibodies are readily measurable in DBS eluates and correlate with plaque reduction neutralization titer (PRNT) values from paired serum samples. Moreover, several of the methods used to estimate SARS-CoV-2 neutralizing antibodies in DBS eluates are adaptable to high-throughput platforms.
... Serum antibody levels are known to decline over time [85], as do the number of circulating memory cells [86,87]; at the same time, the level of local immune response in the respiratory tract can remain intense. It is known that infection with SARS-CoV-2 protects rhesus macaques from re-infection [88,89]. In this regard, it is particularly important to note that immunization did not induce VAERD, as this is a baseline indicator of vaccine safety and allows further studies to be conducted. ...
Article
Full-text available
Background: Influenza and SARS-CoV-2 viruses are two highly variable pathogens. We have developed a candidate bivalent live vaccine based on the strain of licensed A/Leningrad/17-based cold-adapted live attenuated influenza vaccine (LAIV) of H3N2 subtype, which expressed SARS-CoV-2 immunogenic T-cell epitopes. A cassette encoding fragments of S and N proteins of SARS-CoV-2 was inserted into the influenza NA gene using the P2A autocleavage site. In this study, we present the results of preclinical evaluation of the developed bivalent vaccine in a non-human primate model. Methods: Rhesus macaques (Macaca mulatta) (n = 3 per group) were immunized intranasally with 7.5 lg EID50 of the LAIV/CoV-2 bivalent vaccine, a control non-modified H3N2 LAIV or a placebo (chorioallantoic fluid) using a sprayer device, twice, with a 28-day interval. The blood samples were collected at days 0, 3, 28 and 35 for hematological and biochemical assessment. Safety was also assessed by monitoring body weight, body temperature and clinical signs of the disease. Immune responses to influenza virus were assessed both by determining serum antibody titers in hemagglutination inhibition assay, microneutralization assay and IgG ELISA. T-cell responses were measured both to influenza and SARS-CoV-2 antigens using ELISPOT and flow cytometry. Three weeks after the second immunization, animals were challenged with 105 PFU of Delta SARS-CoV-2. The body temperature, weight and challenge virus shedding were monitored for 5 days post-challenge. In addition, virus titers in various organs and histopathology were evaluated on day 6 after SARS-CoV-2 infection. Results: There was no toxic effect of the immunizations on the hematological and coagulation hemostasis of animals. No difference in the dynamics of the average weight and thermometry results were found between the groups of animals. Both LAIV and LAIV/CoV-2 variants poorly replicated in the upper respiratory tract of rhesus macaques. Nevertheless, despite this low level of virus shedding, influenza-specific serum IgG responses were detected in the group of monkeys immunized with the LAIV/CoV-2 bivalent but not in the LAIV group. Furthermore, T-cell responses to both influenza and SARS-CoV-2 viruses were detected in the LAIV/CoV-2 vaccine group only. The animals were generally resistant to SARS-CoV-2 challenge, with minimal virus shedding in the placebo and LAIV groups. Histopathological changes in vaccinated animals were decreased compared to the PBS group, suggesting a protective effect of the chimeric vaccine candidate. Conclusions: The candidate bivalent vaccine was safe and immunogenic for non-human primates and warrants its further evaluation in clinical trials.
... Broad protective influenza-COVID-19 chimeric mRNA vaccine probes were used to detect a region of the N gene of viral genome (gRNA) [61] and a region of E gene of subgenomic RNA (sgRNA) of SARS-CoV-2 [62], respectively, with sequences as follows: gRNA-F, GACCCCAAAATCAGCGAAAT; gRNA-R, TCTGGTTACTGCCAGTTGAATCTG; gRNA-probe, ACCCCGCATTACGTTTGGTGGACC (Omicron variant gRNA-probe: ACTCCGCATTACGTTTGGTGGACC); sgRNA-F, CGATCTCTTGTAGATCTGT TCTC; sgRNA-R, ATATTGCAGCAGTACGCACACA; sgRNA-probe, ACACTAGCCATCCT-TACTGCGCTTCG. Mice lung tissues were stained with H&E for pathological examination. ...
Article
Full-text available
Influenza and coronavirus disease 2019 (COVID-19) represent two respiratory diseases that have significantly impacted global health, resulting in substantial disease burden and mortality. An optimal solution would be a combined vaccine capable of addressing both diseases, thereby obviating the need for multiple vaccinations. Previously, we conceived a chimeric protein subunit vaccine targeting both influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), utilizing the receptor binding domain of spike protein (S-RBD) and the stalk region of hemagglutinin protein (HA-stalk) components. By integrating the S-RBD from the SARS-CoV-2 Delta variant with the headless hemagglutinin (HA) from H1N1 influenza virus, we constructed stable trimeric structures that remain accessible to neutralizing antibodies. This vaccine has demonstrated its potential by conferring protection against a spectrum of strains in mouse models. In this study, we designed an mRNA vaccine candidate encoding the chimeric antigen. The resultant humoral and cellular immune responses were meticulously evaluated in mouse models. Furthermore, the protective efficacy of the vaccine was rigorously examined through challenges with either homologous or heterologous influenza viruses or SARS-CoV-2 strains. Our findings reveal that the mRNA vaccine exhibited robust immunogenicity, engendering high and sustained levels of neutralizing antibodies accompanied by robust and persistent cellular immunity. Notably, this vaccine effectively afforded complete protection to mice against H1N1 or heterosubtypic H5N8 subtypes, as well as the SARS-CoV-2 Delta and Omicron BA.2 variants. Additionally, our mRNA vaccine design can be easily adapted from Delta RBD to Omicron RBD antigens, providing protection against emerging variants. The development of two-in-one vaccine targeting both influenza and COVID-19, incorporating the mRNA platform, may provide a versatile approach to combating future pandemics.
... The levels of genomic RNA (gRNA) and subgenomic RNA (sgRNA) were analyzed by quantitative reverse transcription-PCR (qRT-PCR) assays, which were performed using a FastKing One Step Probe RT-qPCR kit (Tiangen Biotech, China) on a CFX96 Touch real-time PCR detection system (Bio-Rad, USA) according to the manufacturer's protocol. The primers and probes were designed to target the N gene of the gRNA [63] and the E gene of sgRNA [64], with sequences as follows: gRNA-F, GACCCCAAAACCAGCGAAAT; gRNA-probe, ACTCCGCATTACG TTTGGTGGACC; gRNA-R, TCTGGTTACTGCCAGTTGAATCTG; sgRNA-F, TGATCTCT TGTAGATCTGTTCTC; sgRNA-probe, ACACTAGCCATCCTTACTGCGCTTCG; sgRN A-R, ATATTGCAGCAGTACGCACACA. ...
Article
Full-text available
Protective vaccines are crucial for preventing and controlling coronavirus disease 2019 (COVID-19). Updated vaccines are needed to confront the continuously evolving and circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. These vaccines should be safe, effective, amenable to easily scalable production, and affordable. Previously, we developed receptor binding domain (RBD) dimer-based protein subunit vaccines (ZF2001 and updated vaccines) in mammalian cells. In this study, we explored a strategy for producing RBD-dimer immunogens in Pichia pastoris. We found that wild-type P. pastoris produced hyperglycosylated RBD-dimer protein containing four N-glycosylation sites in P. pastoris. Therefore, we engineered the wild type P. pastoris (GS strain) into GSΔOCH1pAO by deleting the OCH1 gene (encoding α-1,6-mannosyltransferase enzyme) to decrease glycosylation, as well as by overexpressing the HIS4 gene (encoding histidine dehydrogenase) to increase histidine synthesis for better growth. In addition, RBD-dimer protein was truncated to remove the R328/F329 cleavage sites in P. pastoris. Several homogeneous RBD-dimer proteins were produced in the GSΔOCH1pAO strain, demonstrating the feasibility of using the P. pastoris expression system. We further resolved the cryo-EM structure of prototype-Beta RBD-dimer complexed with the neutralizing antibody CB6 to reveal the completely exposed immune epitopes of the RBDs. In a murine model, we demonstrated that the yeast-produced RBD-dimer induces robust and protective antibody responses, which is suitable for boosting immunization. This study developed the yeast system for producing SARS-CoV-2 RBD-dimer immunogens, providing a promising platform and pipeline for the future continuous updating and production of SARS-CoV-2 vaccines.
Article
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV 2) pandemic has caused more than 7 million deaths globally. Despite the presence of infection- and vaccine-induced immunity, SARS-CoV-2 infections remain a major global health concern because of the emergence of SARS-CoV-2 variants that can cause severe acute coronavirus disease 2019 (COVID-19) or enhance Long Covid disease phenotypes. About 5 to 10% of SARS-CoV-2–infected individuals develop Long Covid, which, similar to acute COVID 19, often affects the lung. However, Long Covid can also affect other peripheral organs, especially the brain. The causal relationships between acute disease phenotypes, long-term symptoms, and involvement of multiple organ systems remain elusive, and animal model systems mimicking both acute and post-acute phases are imperative. Here, we review the current state of Long Covid animal models, including current and possible future applications.
Article
Background Patients receiving hemodialysis (HD) and kidney transplant recipients are immunocompromised populations prioritized for coronavirus disease 2019 (COVID-19) vaccination, however, there were few clinical trials with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccine responses. Compared with controls, individuals with chronic kidney disease and those on immunosuppressants have lower antibody titers and rates of seroconversion after vaccination. There is a lack of data on their humoral response to COVID-19 immunization. To study the effect of different types of available COVID-19 vaccines in Egypt (AstraZeneca, Sinopharm, Pfizer/BioNTech, and Sputnik) on neutralizing antibodies against COVID-19 in HD and kidney transplantation patients compared with the healthy population. Patients and methods A total of 84 participants; 28 HD patients, 28 kidney transplant recipients, and 28 healthy medical staff members were recruited to test the serological reaction. Six months following the second dose of the COVID-19 vaccine, we evaluated antibody titers against SARS-CoV-2 by Elecsys Anti-SARS-CoV-2 S (Roch) and collected data from the patients, including their comorbidities and the length of time since their kidney transplant. Results All the study groups were comparable as regards age, sex, and BMI, however, hemoglobin was significantly higher in the control group. Antibody response to vaccination was strongest in the control group (100%), followed by HD patients (85%), with transplant recipients showing a significantly weaker response (60%). The Pfizer vaccine generated higher neutralizing antibody levels compared with other vaccines in this study. Yet, the difference was not statistically significant. Additionally, no significant difference in response between the different vaccine types. the transplant group displayed significantly lower levels compared with the control group ( P <0.001) and a trend towards lower levels compared with the dialysis group ( P =0.06). Conclusion Our study found that all tested vaccines generated comparable levels of neutralizing antibodies in healthy individuals and those with chronic kidney disease (dialysis patients). While 85% of dialysis patients achieved seroconversion (positive antibody test) similar to the healthy control group, only 60% of kidney transplant recipients did. the duration post-transplant may be associated with higher rate of seroconversion. the transplant group displayed lower levels of antibodies compared with the control and the dialysis group which suggests a weaker immune response in transplant patients.
Article
Full-text available
The continuous emergence of new SARS-CoV-2 variants requires that COVID vaccines be updated to match circulating strains. We generated B/HPIV3-vectored vaccines expressing 6P-stabilized S protein of the ancestral, B.1.617.2/Delta, or B.1.1.529/Omicron variants as pediatric vaccines for intranasal immunization against HPIV3 and SARS-CoV-2 and characterized these in hamsters. Following intranasal immunization, these B/HPIV3 vectors replicated in the upper and lower respiratory tract and induced mucosal and serum anti-S IgA and IgG. B/HPIV3 expressing ancestral or B.1.617.2/Delta-derived S-6P induced serum antibodies that effectively neutralized SARS-CoV-2 of the ancestral and B.1.617.2/Delta lineages, while the cross-neutralizing potency of B.1.1.529/Omicron S-induced antibodies was lower. Despite the lower cross-neutralizing titers induced by B/HPIV3 expressing S-6P from B.1.1.529/Omicron, a single intranasal dose of all three versions of B/HPIV3 vectors was protective against matched or heterologous WA1/2020, B.1.617.2/Delta or BA.1 (B.1.1.529.1)/Omicron challenge; hamsters were protected from challenge virus replication in the lungs, while low levels of challenge virus were detectable in the upper respiratory tract of a small number of animals. Immunization also protected against lung inflammatory response after challenge, with mild inflammatory cytokine induction associated with the slightly lower level of cross-protection of WA1/2020 and B.1.617.2/Delta variants against the BA.1/Omicron variant. Serum antibodies elicited by all vaccine candidates were broadly reactive against 20 antigenic variants, but the antigenic breadth of antibodies elicited by B/HPIV3-expressed S-6P from the ancestral or B.1.617.2/Delta variant exceeded that of the S-6P B.1.1.529/Omicron expressing vector. These results will guide development of intranasal B/HPIV3 vectors with S antigens matching circulating SARS-CoV-2 variants.
Article
Full-text available
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Article
Full-text available
Coronavirus in nonhuman primates We urgently need vaccines and drug treatments for coronavirus disease 2019 (COVID-19). Even under these extreme circumstances, we must have animal models for rigorous testing of new strategies. Rockx et al. have undertaken a comparative study of three human coronaviruses in cynomolgus macaques: severe acute respiratory syndrome–coronavirus (SARS-CoV) (2002), Middle East respiratory syndrome (MERS)–CoV (2012), and SARS-CoV-2 (2019), which causes COVID-19 (see the Perspective by Lakdawala and Menachery). The most recent coronavirus has a distinct tropism for the nasal mucosa but is also found in the intestinal tract. Although none of the older macaques showed the severe symptoms that humans do, the lung pathology observed was similar. Like humans, the animals shed virus for prolonged periods from their upper respiratory tracts, and like influenza but unlike the 2002 SARS-CoV, this shedding peaked early in infection. It is this cryptic virus shedding that makes case detection difficult and can jeopardize the effectiveness of isolation. Science , this issue p. 1012 ; see also p. 942
Article
Full-text available
Coronavirus disease 2019 (COVID-19) is an acute respiratory tract infection that emerged in late 20191,2. Initial outbreaks in China involved 13.8% cases with severe, and 6.1% with critical courses³. This severe presentation corresponds to the usage of a virus receptor that is expressed predominantly in the lung2,4. By causing an early onset of severe symptoms, this same receptor tropism is thought to have determined pathogenicity, but also aided the control, of severe acute respiratory syndrome (SARS) in 2003⁵. However, there are reports of COVID-19 cases with mild upper respiratory tract symptoms, suggesting the potential for pre- or oligosymptomatic transmission6–8. There is an urgent need for information on body site-specific virus replication, immunity, and infectivity. Here we provide a detailed virological analysis of nine cases, providing proof of active virus replication in upper respiratory tract tissues. Pharyngeal virus shedding was very high during the first week of symptoms (peak at 7.11 × 10⁸ RNA copies per throat swab, day 4). Infectious virus was readily isolated from throat- and lung-derived samples, but not from stool samples, in spite of high virus RNA concentration. Blood and urine never yielded virus. Active replication in the throat was confirmed by viral replicative RNA intermediates in throat samples. Sequence-distinct virus populations were consistently detected in throat and lung samples from the same patient, proving independent replication. Shedding of viral RNA from sputum outlasted the end of symptoms. Seroconversion occurred after 7 days in 50% of patients (14 days in all), but was not followed by a rapid decline in viral load. COVID-19 can present as a mild upper respiratory tract illness. Active virus replication in the upper respiratory tract puts the prospects of COVID-19 containment in perspective.
Article
Full-text available
Since the SARS outbreak 18 years ago, a large number of severe acute respiratory syndrome-related coronaviruses (SARSr-CoV) have been discovered in their natural reservoir host, bats1–4. Previous studies indicated that some of those bat SARSr-CoVs have the potential to infect humans5–7. Here we report the identification and characterization of a novel coronavirus (2019-nCoV) which caused an epidemic of acute respiratory syndrome in humans in Wuhan, China. The epidemic, which started from 12 December 2019, has caused 2,050 laboratory-confirmed infections with 56 fatal cases by 26 January 2020. Full-length genome sequences were obtained from five patients at the early stage of the outbreak. They are almost identical to each other and share 79.5% sequence identify to SARS-CoV. Furthermore, it was found that 2019-nCoV is 96% identical at the whole-genome level to a bat coronavirus. The pairwise protein sequence analysis of seven conserved non-structural proteins show that this virus belongs to the species of SARSr-CoV. The 2019-nCoV virus was then isolated from the bronchoalveolar lavage fluid of a critically ill patient, which can be neutralized by sera from several patients. Importantly, we have confirmed that this novel CoV uses the same cell entry receptor, ACE2, as SARS-CoV.
Article
Full-text available
Emerging infectious diseases, such as SARS and Zika, present a major threat to public health1–3. Despite intense research efforts, how, when and where new diseases appear are still the source of considerable uncertainly. A severe respiratory disease was recently reported in the city Wuhan, Hubei province, China. Up to 25th of January 2020, at least 1,975 cases have been reported since the first patient was hospitalized on the 12th of December 2019. Epidemiological investigation suggested that the outbreak was associated with a seafood market in Wuhan. We studied one patient who was a worker at the market, and who was admitted to Wuhan Central Hospital on 26th of December 2019 experiencing a severe respiratory syndrome including fever, dizziness and cough. Metagenomic RNA sequencing4 of a bronchoalveolar lavage fluid sample identified a novel RNA virus from the family Coronaviridae, designed here as WH-Human-1 coronavirus. Phylogenetic analysis of the complete viral genome (29,903 nucleotides) revealed that the virus was most closely related (89.1% nucleotide similarity) to a group of SARS-like coronaviruses (genus Betacoronavirus, subgenus Sarbecovirus) previously sampled from bats in China. This outbreak highlights the ongoing capacity of viral spill-over from animals to cause severe disease in humans.
Article
Full-text available
Background: The initial cases of novel coronavirus (2019-nCoV)-infected pneumonia (NCIP) occurred in Wuhan, Hubei Province, China, in December 2019 and January 2020. We analyzed data on the first 425 confirmed cases in Wuhan to determine the epidemiologic characteristics of NCIP. Methods: We collected information on demographic characteristics, exposure history, and illness timelines of laboratory-confirmed cases of NCIP that had been reported by January 22, 2020. We described characteristics of the cases and estimated the key epidemiologic time-delay distributions. In the early period of exponential growth, we estimated the epidemic doubling time and the basic reproductive number. Results: Among the first 425 patients with confirmed NCIP, the median age was 59 years and 56% were male. The majority of cases (55%) with onset before January 1, 2020, were linked to the Huanan Seafood Wholesale Market, as compared with 8.6% of the subsequent cases. The mean incubation period was 5.2 days (95% confidence interval [CI], 4.1 to 7.0), with the 95th percentile of the distribution at 12.5 days. In its early stages, the epidemic doubled in size every 7.4 days. With a mean serial interval of 7.5 days (95% CI, 5.3 to 19), the basic reproductive number was estimated to be 2.2 (95% CI, 1.4 to 3.9). Conclusions: On the basis of this information, there is evidence that human-to-human transmission has occurred among close contacts since the middle of December 2019. Considerable efforts to reduce transmission will be required to control outbreaks if similar dynamics apply elsewhere. Measures to prevent or reduce transmission should be implemented in populations at risk. (Funded by the Ministry of Science and Technology of China and others.).
Article
Full-text available
In December 2019, a cluster of patients with pneumonia of unknown cause was linked to a seafood wholesale market in Wuhan, China. A previously unknown betacoronavirus was discovered through the use of unbiased sequencing in samples from patients with pneumonia. Human airway epithelial cells were used to isolate a novel coronavirus, named 2019-nCoV, which formed another clade within the subgenus sarbecovirus, Orthocoronavirinae subfamily. Different from both MERS-CoV and SARS-CoV, 2019-nCoV is the seventh member of the family of coronaviruses that infect humans. Enhanced surveillance and further investigation are ongoing. (Funded by the National Key Research and Development Program of China and the National Major Project for Control and Prevention of Infectious Disease in China.).
Article
Full-text available
Background: An ongoing outbreak of pneumonia associated with a novel coronavirus was reported in Wuhan city, Hubei province, China. Affected patients were geographically linked with a local wet market as a potential source. No data on person-to-person or nosocomial transmission have been published to date. Methods: In this study, we report the epidemiological, clinical, laboratory, radiological, and microbiological findings of five patients in a family cluster who presented with unexplained pneumonia after returning to Shenzhen, Guangdong province, China, after a visit to Wuhan, and an additional family member who did not travel to Wuhan. Phylogenetic analysis of genetic sequences from these patients were done. Findings: From Jan 10, 2020, we enrolled a family of six patients who travelled to Wuhan from Shenzhen between Dec 29, 2019 and Jan 4, 2020. Of six family members who travelled to Wuhan, five were identified as infected with the novel coronavirus. Additionally, one family member, who did not travel to Wuhan, became infected with the virus after several days of contact with four of the family members. None of the family members had contacts with Wuhan markets or animals, although two had visited a Wuhan hospital. Five family members (aged 36-66 years) presented with fever, upper or lower respiratory tract symptoms, or diarrhoea, or a combination of these 3-6 days after exposure. They presented to our hospital (The University of Hong Kong-Shenzhen Hospital, Shenzhen) 6-10 days after symptom onset. They and one asymptomatic child (aged 10 years) had radiological ground-glass lung opacities. Older patients (aged >60 years) had more systemic symptoms, extensive radiological ground-glass lung changes, lymphopenia, thrombocytopenia, and increased C-reactive protein and lactate dehydrogenase levels. The nasopharyngeal or throat swabs of these six patients were negative for known respiratory microbes by point-of-care multiplex RT-PCR, but five patients (four adults and the child) were RT-PCR positive for genes encoding the internal RNA-dependent RNA polymerase and surface Spike protein of this novel coronavirus, which were confirmed by Sanger sequencing. Phylogenetic analysis of these five patients' RT-PCR amplicons and two full genomes by next-generation sequencing showed that this is a novel coronavirus, which is closest to the bat severe acute respiatory syndrome (SARS)-related coronaviruses found in Chinese horseshoe bats. Interpretation: Our findings are consistent with person-to-person transmission of this novel coronavirus in hospital and family settings, and the reports of infected travellers in other geographical regions. Funding: The Shaw Foundation Hong Kong, Michael Seak-Kan Tong, Respiratory Viral Research Foundation Limited, Hui Ming, Hui Hoy and Chow Sin Lan Charity Fund Limited, Marina Man-Wai Lee, the Hong Kong Hainan Commercial Association South China Microbiology Research Fund, Sanming Project of Medicine (Shenzhen), and High Level-Hospital Program (Guangdong Health Commission).
Article
An outbreak of novel coronavirus (2019-nCoV) that began in Wuhan, China, has spread rapidly, with cases now confirmed in multiple countries. We report the first case of 2019-nCoV infection confirmed in the United States and describe the identification, diagnosis, clinical course, and management of the case, including the patient's initial mild symptoms at presentation with progression to pneumonia on day 9 of illness. This case highlights the importance of close coordination between clinicians and public health authorities at the local, state, and federal levels, as well as the need for rapid dissemination of clinical information related to the care of patients with this emerging infection.
Article
Background: In December, 2019, a pneumonia associated with the 2019 novel coronavirus (2019-nCoV) emerged in Wuhan, China. We aimed to further clarify the epidemiological and clinical characteristics of 2019-nCoV pneumonia. Methods: In this retrospective, single-centre study, we included all confirmed cases of 2019-nCoV in Wuhan Jinyintan Hospital from Jan 1 to Jan 20, 2020. Cases were confirmed by real-time RT-PCR and were analysed for epidemiological, demographic, clinical, and radiological features and laboratory data. Outcomes were followed up until Jan 25, 2020. Findings: Of the 99 patients with 2019-nCoV pneumonia, 49 (49%) had a history of exposure to the Huanan seafood market. The average age of the patients was 55·5 years (SD 13·1), including 67 men and 32 women. 2019-nCoV was detected in all patients by real-time RT-PCR. 50 (51%) patients had chronic diseases. Patients had clinical manifestations of fever (82 [83%] patients), cough (81 [82%] patients), shortness of breath (31 [31%] patients), muscle ache (11 [11%] patients), confusion (nine [9%] patients), headache (eight [8%] patients), sore throat (five [5%] patients), rhinorrhoea (four [4%] patients), chest pain (two [2%] patients), diarrhoea (two [2%] patients), and nausea and vomiting (one [1%] patient). According to imaging examination, 74 (75%) patients showed bilateral pneumonia, 14 (14%) patients showed multiple mottling and ground-glass opacity, and one (1%) patient had pneumothorax. 17 (17%) patients developed acute respiratory distress syndrome and, among them, 11 (11%) patients worsened in a short period of time and died of multiple organ failure. Interpretation: The 2019-nCoV infection was of clustering onset, is more likely to affect older males with comorbidities, and can result in severe and even fatal respiratory diseases such as acute respiratory distress syndrome. In general, characteristics of patients who died were in line with the MuLBSTA score, an early warning model for predicting mortality in viral pneumonia. Further investigation is needed to explore the applicability of the MuLBSTA score in predicting the risk of mortality in 2019-nCoV infection. Funding: National Key R&D Program of China.