scispace - formally typeset
Search or ask a question
Posted ContentDOI

SARS-CoV-2 variant B.1.617 is resistant to Bamlanivimab and evades antibodies induced by infection and vaccination

TL;DR: In this article, the authors analyzed whether B.1.617 is more adept in entering cells and/or evades antibody responses, and revealed that antibody evasion may contribute to the rapid spread of this variant.
Abstract: The emergence of SARS-CoV-2 variants threatens efforts to contain the COVID-19 pandemic. The number of COVID-19 cases and deaths in India has risen steeply in recent weeks and a novel SARS-CoV-2 variant, B.1.617, is believed to be responsible for many of these cases. The spike protein of B.1.617 harbors two mutations in the receptor binding domain, which interacts with the ACE2 receptor and constitutes the main target of neutralizing antibodies. Therefore, we analyzed whether B.1.617 is more adept in entering cells and/or evades antibody responses. B.1.617 entered two out of eight cell lines tested with slightly increased efficiency and was blocked by entry inhibitors. In contrast, B.1.617 was resistant against Bamlanivimab, an antibody used for COVID-19 treatment. Finally, B.1.617 evaded antibodies induced by infection or vaccination, although with moderate efficiency. Collectively, our study reveals that antibody evasion of B.1.617 may contribute to the rapid spread of this variant.

Summary (4 min read)

INTRODUCTION

  • The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the devastating coronavirus disease 2019 (COVID-19) pandemic, which more than one year after its emergence is associated with record numbers in cases and deaths (W.H.O., 2021) .
  • Strategies to fight the COVID-19 pandemic either by vaccines or non-pharmaceutical interventions have been threatened by the emergence of SARS-CoV-2 variants of concern (VOC).
  • These variants harbor mutations that confer increased transmissibility or immune evasion (Plante et al., 2021) .
  • The S protein is incorporated into the viral membrane and facilitates viral entry into target cells.
  • It is believed that many cases are due to infection with a novel variant, B.1.617, that harbors eight mutations in the S protein, including mutations L452R and E484Q within the RBD, which introduce changes at amino acid positions known to modulate antibody-mediated neutralization (Li et al., 2021; Li et al., 2020) .

The S protein of variant B.1.617 mediates increased entry into certain human intestinal and lung cell lines

  • These pseudotyped particles faithfully mimic cell entry of SARS-CoV-2 and have been previously used to identify host factors required for SARS-CoV-2 cell entry and to study neutralization of SARS-CoV-2 by antibodies (Hoffmann et al., 2021a; Hoffmann et al., 2020; Riepler et al., 2020; Schmidt et al., 2020) .
  • The only exceptions were Caco-2 and Calu-3 cells (and 293T cells in case of B.1.351), which are derived from human intestine and lung, respectively, and for which the S protein of B.1.617 (and B.1.351 ) mediated entry with moderately increased efficiency .
  • In addition, the authors investigated S protein-driven cell entry into BHK-21 cells, which were transfected either with empty plasmid or ACE2 expression plasmid.

Soluble ACE2 and Camostat inhibit cell entry driven by the S protein of variant B.1.617

  • The authors next examined whether entry of B.1.617 can be blocked by inhibitors targeting the RBD (soluble ACE2) and proteolytic activation of the S protein.
  • Soluble ACE2 binds to the RBD and blocks subsequent engagement of membrane bound ACE2.
  • The authors analyzed whether these antibodies were able to inhibit host cell entry driven by the S protein of variant B.1.617.
  • These results suggest that Casirivimab and particularly Bamlanivimab monotherapy may not be suitable for treatment of patients infected with variant B1.617.

SARS-CoV-2 infection induces the generation of neutralizing antibodies in most infected patients

  • And it is believed that these antibody responses are important for protection from re-infection (Rodda et al., 2021; Wajnberg et al., 2020) .
  • Therefore, the authors determined whether variant B.1.617 evades inhibition by antibodies, which might contribute to its increasing transmission dynamics.
  • For this, the authors analyzed antibody-mediated neutralization using plasma samples obtained from 15 COVID-19 patients at the intensive care unit of Göttingen University Hospital (Table S1 ).
  • These plasma samples were prescreened for neutralizing activity and tested for their ability to block host cell entry driven by WT S protein and the S protein of variant B.1.617.
  • The S protein of variant B.1.351 served as control since this S protein efficiently evades antibody-mediated neutralization (Hoffmann et al., 2021a) .

Diminished neutralization by plasma from Comirnaty/BNT162b2 vaccinated patients

  • Vaccination with Comirnaty/BNT162b2 has been shown to be safe and to protect against COVID-19 with high efficiency (Polack et al., 2020) .
  • The vaccine induces antibody and T cell responses (Grifoni et al., 2020; Peng et al., 2020) and the neutralizing antibodies triggered by vaccination are believed to be important for vaccine-induced protection against SARS-CoV-2 infection.
  • Therefore, the authors analyzed whether cell entry driven by the S protein of variant B.1.617 can be efficiently inhibited by plasma from Comirnaty/BNT162b2 vaccinated individuals (Table S2 ).
  • To address this question, the authors analyzed neutralization by 15 plasma samples obtained from vaccinees two to three weeks after they had received the second vaccine dose.
  • Thus, variant B.1.617 can partially evade control by antibodies induced by vaccination with Comirnaty/BNT162b2.

DISCUSSION

  • The recent surge in COVID-19 cases and deaths in India is paralleled by the spread of the novel SARS-CoV-2 variant B.1.617.
  • The B.1.617 S protein facilitated moderately increased entry into the human lung and intestinal cell lines Calu-3 and Caco-2, respectively, and this effect was much less pronounced when ACE2 was overexpressed in Calu-3 cells.
  • Another explanation for the increased spread of variant B.1.617 in India might be immune evasion, i.e. the ability to spread in a population in which a substantial portion of individuals has preexisting immune responses against SARS-CoV-2.
  • The RBD of the B.1.617 S protein harbors two mutations associated with (L452R) or suspected (E484Q) of antibody evasion.

Cell

  • Authentication of cell lines was performed by STR-typing, amplification and sequencing of a fragment of the cytochrome c oxidase gene, microscopic examination and/or according to their growth characteristics.
  • In addition, cell lines were routinely tested for contamination by mycoplasma.

Expression plasmids

  • In order to generate the expression vector for the S protein of SARS-CoV-2 variant B.1.617, the required mutations were inserted into the WT SARS-CoV-2 S sequence by overlap extension PCR.
  • The resulting open reading frame was further inserted into the pCG1 plasmid (kindly provided by Roberto Cattaneo, Mayo Clinic College of Medicine, Rochester, MN, USA), making use of the unique BamHI and XbaI restriction sites.
  • Sequence integrity was verified by sequencing using a commercial sequencing service (Microsynth SeqLab).
  • Transfection of 293T cells was carried out by the calcium-phosphate precipitation method, while BHK-21 cells were transfected using Lipofectamine LTX (Thermo Fisher Scientific).

Preparation of vesicular stomatitis virus pseudotypes

  • For this study, the authors employed rhabdoviral pseudotype particles that are based on a replicationdeficient VSV vector that lacks the genetic information for VSV-G and instead codes for two reporter proteins, enhanced green fluorescent protein and firefly luciferase (FLuc), VSV * ΔG-FLuc (kindly provided by Gert Zimmer, Institute of Virology and Immunology, Mittelhäusern, Switzerland) (Berger Rentsch and Zimmer, 2011) .
  • Thereafter, cells received culture medium containing anti-VSV-G antibody (culture supernatant from I1-hybridoma cells; ATCC no.
  • CRL-2700; except for cells expressing VSV-G, which received only medium) and incubated for 16-18 h.
  • Finally, the clarified supernatant was aliquoted and stored at -80 °C.

Production of soluble ACE2

  • For the production of soluble ACE2 fused to the Fc portion of human immunoglobulin G (IgG), sol-ACE2, 293T cells were grown in a T-75 flask and transfected with 20 µg of sol-ACE2 expression plasmid.
  • Then, the culture supernatant was collected and the cells received fresh medium and were further incubated.
  • After an additional 24 h, the culture supernatant was harvested and centrifuged as described before.
  • Next, the clarified supernatants from both harvests were combined, loaded onto Vivaspin protein concentrator columns with a molecular weight cutoff of 30 kDa and centrifuged at 4,000 x g at 4 °C until a concentration factor of 20 was achieved.
  • Finally, the concentrated sol-ACE2 was aliquoted and stored at -80 °C.

Collection of serum and plasma samples

  • All plasma samples were heat-inactivated (56 °C, 30 min) before analysis.
  • Further, all plasma were pre-screened for the presence of neutralizing activity against WT SARS-CoV-2 S using a pseudotype neutralization test.
  • Convalescent plasma samples were collected from COVID-19 patients treated at the intensive care unit of the University Medicine Göttingen under approval given by the ethic committee of the University Medicine Göttingen (SeptImmun Study 25/4/19 Ü).
  • Plasma from individuals vaccinated with BioNTech/Pfizer vaccine BNT162b2/ Comirnaty were obtained 24-31 days after the second dose.
  • The study was approved by the Institutional Review Board of Hannover Medical School (8973_BO_K_2020).

Transduction of target cells

  • All transduction experiments were carried out in 96-well format at a cell confluency of 50-80%.
  • For experiments addressing cell tropism and entry efficiency, Vero, Caco-2, Calu-3, Calu-3 (ACE2), 293T, A549 (ACE2), A549 (ACE2+TMPRSS2) and Huh-7 target cells were inoculated with identical volumes of pseudotype preparations.
  • For this, the culture supernatant was aspirated.

Pseudotype particle neutralization test

  • In all cases, particles incubated only with medium served as control.
  • Transduction efficiency was determined at 16-18 h postinoculation as described above.

Data analysis

  • The results presented in this study represent average (mean) data obtained from three biological Data normalization was performed in the following fashion: (i) For comparison of entry efficiency by the different S proteins, transduction was normalized against WT SARS-CoV-2 S (set as 100%).
  • Alternatively, transduction was normalized against the background signal (luminescence measured for cells inoculated with particles bearing no viral glycoprotein; set as 1).
  • Evidence of escape of SARS-CoV-2 variant B.1.351 from natural and vaccine-induced sera.
  • A pneumonia outbreak associated with a new coronavirus of probable bat origin.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

1
SARS-CoV-2 variant B.1.617 is resistant to Bamlanivimab and evades
1
antibodies induced by infection and vaccination
2
3
Markus Hoffmann,
1,2,6,*
Heike Hofmann-Winkler,
1,6
Nadine Krüger,
1,6
Amy Kempf,
1,2
4
Inga Nehlmeier,
1
Luise Graichen,
1,2
Anzhalika Sidarovich,
1,2
Anna-Sophie Moldenhauer,
1
5
Martin S. Winkler,
3
Sebastian Schulz,
4
Hans-Martin Jäck,
4
Metodi V. Stankov,
5
6
Georg M. N. Behrens,
5
Stefan Pöhlmann
1,2,*
7
8
1
Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077 Göttingen, Germany
9
2
Faculty of Biology and Psychology, Georg-August-University Göttingen, Wilhelmsplatz 1,
10
37073 Göttingen, Germany
11
3
Department of Anaesthesiology, University of Göttingen Medical Center, Göttingen, Georg-
12
August University of Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
13
4
Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander
14
University of Erlangen-Nürnberg, Glückstraße 6, 91054 Erlangen, Germany
15
5
Department for Rheumatology and Immunology, Hannover Medical School, Carl-Neuberg-Str.
16
1, 30625 Hannover, Germany
17
18
6
These authors contributed equally
19
7
Lead contact
20
*Correspondence: mhoffmann@dpz.eu (M.H.), spoehlmann@dpz.eu (S.P.)
21
22
23
24
.CC-BY-NC-ND 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted May 5, 2021. ; https://doi.org/10.1101/2021.05.04.442663doi: bioRxiv preprint

2
SUMMARY
25
The emergence of SARS-CoV-2 variants threatens efforts to contain the COVID-19
26
pandemic. The number of COVID-19 cases and deaths in India has risen steeply in recent
27
weeks and a novel SARS-CoV-2 variant, B.1.617, is believed to be responsible for many of
28
these cases. The spike protein of B.1.617 harbors two mutations in the receptor binding
29
domain, which interacts with the ACE2 receptor and constitutes the main target of
30
neutralizing antibodies. Therefore, we analyzed whether B.1.617 is more adept in entering
31
cells and/or evades antibody responses. B.1.617 entered two out of eight cell lines tested
32
with slightly increased efficiency and was blocked by entry inhibitors. In contrast, B.1.617
33
was resistant against Bamlanivimab, an antibody used for COVID-19 treatment. Finally,
34
B.1.617 evaded antibodies induced by infection or vaccination, although with moderate
35
efficiency. Collectively, our study reveals that antibody evasion of B.1.617 may contribute to
36
the rapid spread of this variant.
37
38
39
40
41
42
43
44
45
46
47
48
.CC-BY-NC-ND 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted May 5, 2021. ; https://doi.org/10.1101/2021.05.04.442663doi: bioRxiv preprint

3
INTRODUCTION
49
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the
50
devastating coronavirus disease 2019 (COVID-19) pandemic, which more than one year after its
51
emergence is associated with record numbers in cases and deaths (W.H.O., 2021). Effective
52
antivirals are largely lacking, although recombinant antibodies targeting the viral spike protein
53
(S) can significantly reduce viral load und have received emergency use authorization (EUA)
54
(Chen et al., 2021a; Gottlieb et al., 2021). Drugs that target the dysregulated cytokine responses
55
characteristic for COVID-19 are available but their clinical benefit is oversee-able (Tomazini et
56
al., 2020; W.H.O. REACT Working Group et al., 2020). While progress in treatment
57
development is moderate, mRNA- and vector-based vaccines are available that provide efficient
58
protection against disease (Baden et al., 2021; Polack et al., 2020). As a consequence, vaccination
59
is viewed as the key instrument in combating and ultimately ending the COVID-19 pandemic.
60
Strategies to fight the COVID-19 pandemic either by vaccines or non-pharmaceutical
61
interventions have been threatened by the emergence of SARS-CoV-2 variants of concern
62
(VOC). These variants harbor mutations that confer increased transmissibility or immune evasion
63
(Plante et al., 2021). Studies of mutations present in VOC have mainly focused on the viral S
64
protein. The S protein is incorporated into the viral membrane and facilitates viral entry into
65
target cells. For this, the surface unit, S1, of the S protein first binds to the cellular receptor ACE2
66
(Hoffmann et al., 2020; Zhou et al., 2020) via its receptor binding domain (RBD). Subsequently,
67
the S protein is activated by TMPRSS2 or related cellular proteases (Hoffmann et al., 2021b;
68
Hoffmann et al., 2020) and the transmembrane unit, S2, of the S protein facilitates fusion of the
69
viral and a cellular membrane, allowing delivery of the viral genome into the host cell. These
70
processes are essential for SARS-CoV-2 infection and are targeted by drugs and neutralizing
71
antibodies.
72
.CC-BY-NC-ND 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted May 5, 2021. ; https://doi.org/10.1101/2021.05.04.442663doi: bioRxiv preprint

4
The prototypic VOC with increased fitness is variant B.1.1.7, which emerged in the
73
United Kingdom and is now spreading in many countries. B.1.1.7 replicates to higher levels in
74
patients and is more efficiently transmitted between humans as compared to the previously
75
circulating viruses (Frampton et al., 2021; Graham et al., 2021; Leung et al., 2021). The increased
76
transmissibility might be linked to mutation N501Y in the RBD that might increase binding to
77
ACE2 (Ali et al., 2021; Luan et al., 2021). However, the exact mechanisms underlying more
78
robust transmission of B.1.1.7 remain to be elucidated. In contrast, differences in antibody-
79
mediated neutralization between previously circulating viruses and variant B.1.1.7 are minor,
80
with B.1.1.7 being slightly less sensitive to neutralization (Chen et al., 2021b; Collier et al., 2021;
81
Hoffmann et al., 2021a; Kuzmina et al., 2021; Muik et al., 2021; Planas et al., 2021; Shen et al.,
82
2021; Supasa et al., 2021; Wang et al., 2021a; Xie et al., 2021). In sum, B.1.1.7 shows increased
83
fitness and will outcompete previously circulating viruses in an immunologically naïve
84
population.
85
In populations with a high percentage of individuals with pre-existing immune responses
86
against SARS-CoV-2, viral variants that can evade immune control have a selective advantage.
87
Variant B.1.351 that became dominant in South Africa (Tegally et al., 2021) and variant P.1 that
88
became dominant in Brazil (Faria et al., 2021) are such variants (Chen et al., 2021b; Dejnirattisai
89
et al., 2021; Edara et al., 2021; Garcia-Beltran et al., 2021; Hoffmann et al., 2021a; Kuzmina et
90
al., 2021; Planas et al., 2021; Wang et al., 2021a; Zhou et al., 2021). These variants harbor
91
mutations in the S protein that reduce neutralization by antibodies, including E484K, which is
92
located in the RBD and is present in both, B.1.351 and P1 (Li et al., 2021; Liu et al., 2021; Wang
93
et al., 2021c). At present, evasion form antibodies is most prominent for variant B.1.351 but it is
94
unclear whether variants can arise that exhibit increased or even complete neutralization
95
resistance.
96
.CC-BY-NC-ND 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted May 5, 2021. ; https://doi.org/10.1101/2021.05.04.442663doi: bioRxiv preprint

5
India has seen a steep increase in COVID-19 cases and deaths in the recent weeks
97
(W.H.O., 2021). It is believed that many cases are due to infection with a novel variant, B.1.617,
98
that harbors eight mutations in the S protein, including mutations L452R and E484Q within the
99
RBD, which introduce changes at amino acid positions known to modulate antibody-mediated
100
neutralization (Li et al., 2021; Li et al., 2020). However, it is at present unknown whether
101
B.1.617 evades antibody-mediated neutralization. Similarly, it is unknown whether the variant
102
exhibits an altered dependence on host cell factors for entry, which may alter cell tropism, entry
103
efficiency and sensitivity to entry inhibitors.
104
Here, we report that the S protein of B.1.617 mediates moderately enhanced entry into the
105
human lung- and intestine-derived cell lines Calu-3 and Caco-2, respectively, and that entry is
106
inhibited by soluble ACE2 and Camostat, the latter of which targets TMPRSS2. In contrast, entry
107
driven by the B.1.617 S protein was fully resistant to neutralization by a monoclonal antibody
108
with EUA for COVID-19 treatment (Bamlanivimab). Finally, B.1.617 S protein-driven entry was
109
partially resistant against neutralization by antibodies elicited upon infection or vaccination with
110
the Comirnaty/BNT162b2 vaccine.
111
112
113
114
115
116
117
118
119
120
.CC-BY-NC-ND 4.0 International licenseavailable under a
was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprint (whichthis version posted May 5, 2021. ; https://doi.org/10.1101/2021.05.04.442663doi: bioRxiv preprint

Citations
More filters
Journal ArticleDOI
TL;DR: In this article, the authors provide an up-to-date comparative analysis of the characteristics, adverse events, efficacy, effectiveness and impact of the variants of concern for nineteen COVID-19 vaccines.

408 citations

Journal ArticleDOI
20 Jul 2021
TL;DR: In this paper, the signature mutations possessed by these strains were L452R, T478K, E484Q, D614G and P681R in the spike protein, including within the receptor-binding domain.
Abstract: As the global severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic expands, genomic epidemiology and whole genome sequencing are being used to investigate its transmission and evolution. Against the backdrop of the global emergence of “variants of concern” (VOCs) during December 2020 and an upsurge in a state in the western part of India since January 2021, whole genome sequencing and analysis of spike protein mutations using sequence and structural approaches were undertaken to identify possible new variants and gauge the fitness of the current circulating strains. Phylogenetic analysis revealed that newly identified lineages B.1.617.1 and B.1.617.2 were predominantly circulating. The signature mutations possessed by these strains were L452R, T478K, E484Q, D614G and P681R in the spike protein, including within the receptor-binding domain (RBD). Of these, the mutations at residue positions 452, 484 and 681 have been reported in other globally circulating lineages. The structural analysis of RBD mutations L452R, T478K and E484Q revealed that these may possibly result in increased ACE2 binding while P681R in the furin cleavage site could increase the rate of S1-S2 cleavage, resulting in better transmissibility. The two RBD mutations, L452R and E484Q, indicated decreased binding to select monoclonal antibodies (mAbs) and may affect their neutralization potential. Further in vitro/in vivo studies would help confirm the phenotypic changes of the mutant strains. Overall, the study revealed that the newly emerged variants were responsible for the second wave of COVID-19 in Maharashtra. Lineage B.1.617.2 has been designated as a VOC delta and B.1.617.1 as a variant of interest kappa, and they are being widely reported in the rest of the country as well as globally. Continuous monitoring of these and emerging variants in India is essential.

401 citations

Journal ArticleDOI
10 Jun 2021-Nature
TL;DR: The authors showed that serum samples taken from twenty human volunteers, two or four weeks after their second dose of the BNT162b2 vaccine, neutralize engineered SARS-CoV-2 with a USA-WA1/2020 genetic background (a virus strain isolated in January 2020) and spike glycoproteins from the recently identified B.1.617.
Abstract: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is continuing to evolve around the world, generating new variants that are of concern on the basis of their potential for altered transmissibility, pathogenicity, and coverage by vaccines and therapeutic agents1–5. Here we show that serum samples taken from twenty human volunteers, two or four weeks after their second dose of the BNT162b2 vaccine, neutralize engineered SARS-CoV-2 with a USA-WA1/2020 genetic background (a virus strain isolated in January 2020) and spike glycoproteins from the recently identified B.1.617.1, B.1.617.2, B.1.618 (all of which were first identified in India) or B.1.525 (first identified in Nigeria) lineages. Geometric mean plaque reduction neutralization titres against the variant viruses—particularly the B.1.617.1 variant—seemed to be lower than the titre against the USA-WA1/2020 virus, but all sera tested neutralized the variant viruses at titres of at least 1:40. The susceptibility of the variant strains to neutralization elicited by the BNT162b2 vaccine supports mass immunization as a central strategy to end the coronavirus disease 2019 (COVID-19) pandemic globally. Samples of serum from individuals immunized with the BNT162b2 vaccine show neutralization activity against engineered SARS-CoV-2s bearing the spike mutations from B.1.617 and other variants.

280 citations

Journal ArticleDOI
10 Jun 2021-Cell
TL;DR: An overview of SARS-CoV-2-neutralizing mAbs, including their origin, specificity, structure, antiviral and immunological mechanisms of action, and resistance to circulating variants, as well as a snapshot of the clinical trials of approved or late-stage mAb therapeutics can be found in this paper.

247 citations

Posted ContentDOI
27 May 2021-bioRxiv
TL;DR: The SARS-CoV-2 B.1.617 lineage emerged in October 2020 in India1-6.7.1 and has since then become dominant in some indian regions and further spread to many countries.
Abstract: The SARS-CoV-2 B.1.617 lineage emerged in October 2020 in India1-6. It has since then become dominant in some indian regions and further spread to many countries. The lineage includes three main subtypes (B1.617.1, B.1617.2 and B.1.617.3), which harbour diverse Spike mutations in the N-terminal domain (NTD) and the receptor binding domain (RBD) which may increase their immune evasion potential. B.1.617.2 is believed to spread faster than the other versions. Here, we isolated infectious B.1.617.2 from a traveller returning from India. We examined its sensitivity to monoclonal antibodies (mAbs) and to antibodies present in sera from COVID-19 convalescent individuals or vaccine recipients, in comparison to other viral lineages. B.1.617.2 was resistant to neutralization by some anti-NTD and anti-RBD mAbs, including Bamlanivimab, which were impaired in binding to the B.1.617.2 Spike. Sera from convalescent patients collected up to 12 months post symptoms and from Pfizer Comirnaty vaccine recipients were 3 to 6 fold less potent against B.1.617.2, relative to B.1.1.7. Sera from individuals having received one dose of AstraZeneca Vaxzevria barely inhibited B.1.617.2. Thus, B.1.617.2 spread is associated with an escape to antibodies targeting non-RBD and RBD Spike epitopes.

200 citations

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

16,857 citations

Journal ArticleDOI
16 Apr 2020-Cell
TL;DR: It is demonstrated that SARS-CoV-2 uses the SARS -CoV receptor ACE2 for entry and the serine protease TMPRSS2 for S protein priming, and it is shown that the sera from convalescent SARS patients cross-neutralized Sars-2-S-driven entry.

15,362 citations

Journal ArticleDOI
TL;DR: A two-dose regimen of BNT162b2 conferred 95% protection against Covid-19 in persons 16 years of age or older and safety over a median of 2 months was similar to that of other viral vaccines.
Abstract: Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the resulting coronavirus disease 2019 (Covid-19) have afflicted tens of millions of people in a world...

10,274 citations

Journal ArticleDOI
25 Jun 2020-Cell
TL;DR: Using HLA class I and II predicted peptide ‘megapools’, circulating SARS-CoV-2−specific CD8+ and CD4+ T cells were identified in ∼70% and 100% of COVID-19 convalescent patients, respectively, suggesting cross-reactive T cell recognition between circulating ‘common cold’ coronaviruses and SARS.

3,043 citations

Journal ArticleDOI
TL;DR: A molecular explanation why SARS-CoV infections cause severe and often lethal lung failure and suggest a rational therapy for SARS and possibly other respiratory disease viruses is provided.
Abstract: During several months of 2003, a newly identified illness termed severe acute respiratory syndrome (SARS) spread rapidly through the world. A new coronavirus (SARS-CoV) was identified as the SARS pathogen, which triggered severe pneumonia and acute, often lethal, lung failure. Moreover, among infected individuals influenza such as the Spanish flu and the emergence of new respiratory disease viruses have caused high lethality resulting from acute lung failure. In cell lines, angiotensin-converting enzyme 2 (ACE2) has been identified as a potential SARS-CoV receptor. The high lethality of SARS-CoV infections, its enormous economic and social impact, fears of renewed outbreaks as well as the potential misuse of such viruses as biologic weapons make it paramount to understand the pathogenesis of SARS-CoV. Here we provide the first genetic proof that ACE2 is a crucial SARS-CoV receptor in vivo. SARS-CoV infections and the Spike protein of the SARS-CoV reduce ACE2 expression. Notably, injection of SARS-CoV Spike into mice worsens acute lung failure in vivo that can be attenuated by blocking the renin-angiotensin pathway. These results provide a molecular explanation why SARS-CoV infections cause severe and often lethal lung failure and suggest a rational therapy for SARS and possibly other respiratory disease viruses.

2,983 citations

Related Papers (5)