scispace - formally typeset
Open AccessPosted ContentDOI

Emerging SARS-CoV-2 variants of concern evade humoral immune responses from infection and vaccination

Reads0
Chats0
TLDR
In this article, the authors assessed the recognition of three SARS-CoV-2 variants of concern (B.1.7, B.1,351 and P.351) in cohorts of COVID-19 patients ranging in disease severity and recipients of the Pfizer/BioNTech vaccine (n = 50).
Abstract
Emerging SARS-CoV-2 variants pose a threat to human immunity induced by natural infection and vaccination. We assessed the recognition of three variants of concern (B.1.1.7, B.1.351 and P.1) in cohorts of COVID-19 patients ranging in disease severity (n = 69) and recipients of the Pfizer/BioNTech vaccine (n = 50). Spike binding and neutralization against all three VOC was substantially reduced in the majority of samples, with the largest 4-7-fold reduction in neutralization being observed against B.1.351. While hospitalized COVID-19 patients and vaccinees maintained sufficient neutralizing titers against all three VOC, 39% of non-hospitalized patients did not neutralize B.1.351. Moreover, monoclonal neutralizing antibodies (NAbs) show sharp reductions in their binding kinetics and neutralizing potential to B.1.351 and P.1, but not to B.1.1.7. These data have implications for the degree to which pre-existing immunity can protect against subsequent infection with VOC and informs policy makers of susceptibility to globally circulating SARS-CoV-2 VOC.

read more

Content maybe subject to copyright    Report

Emerging SARS-CoV-2 variants of concern evade humoral immune responses from infection and
1
vaccination
2
3
4
Tom G. Caniels
1†
, Ilja Bontjer
1†
, Karlijn van der Straten
1†
, Meliawati Poniman
1
, Judith A. Burger
1
, Brent
5
Appelman
2
, Ayesha H.A. Lavell
3
, Melissa Oomen
1
, Gert-Jan Godeke
4
, Coralie Valle
4
, Ramona Mögling
4
,
6
Hugo D.G. van Willigen
1
, Elke Wynberg
5
, Michiel Schinkel
2
, Lonneke A. van Vught
2
, Denise Guerra
1
,
7
Jonne L. Snitselaar
1
, Devidas N. Chaturbhuj
6
, Isabel Cuella Martin
1
, Amsterdam UMC COVID-19 S3/HCW
8
study group, John P. Moore
6
, Menno D. de Jong
1
, Chantal Reusken
4
, Jonne J. Sikkens
3
, Marije K.
9
Bomers
3
, Godelieve J. de Bree
7
, Marit J. van Gils
1
*, Dirk Eggink
1,4
*, Rogier W. Sanders
1,6
*
10
11
1
Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for
12
Infection and Immunity, Amsterdam, the Netherlands.
13
2
Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam,
14
Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands.
15
3
Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Institute
16
for Infection and Immunity, Amsterdam, the Netherlands.
17
4
Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven,
18
the Netherlands.
19
5
Department of Infectious Diseases, Public Health Service of Amsterdam, GGD, Amsterdam, the
20
Netherlands.
21
6
Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York,
22
USA.
23
7
Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for
24
Infection and Immunity, Amsterdam, the Netherlands.
25
26
† These authors contributed equally to this work.
27
28
* Correspondence to: r.w.sanders@amsterdamumc.nl, dirk.eggink@rivm.nl,
29
m.j.vangils@amsterdamumc.nl
30
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted June 1, 2021. ; https://doi.org/10.1101/2021.05.26.21257441doi: medRxiv preprint
NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice.

Abstract
31
Emerging SARS-CoV-2 variants pose a threat to human immunity induced by natural infection and
32
vaccination. We assessed the recognition of three variants of concern (B.1.1.7, B.1.351 and P.1) in
33
cohorts of COVID-19 patients ranging in disease severity (n = 69) and recipients of the Pfizer/BioNTech
34
vaccine (n = 50). Spike binding and neutralization against all three VOC was substantially reduced in the
35
majority of samples, with the largest 4-7-fold reduction in neutralization being observed against B.1.351.
36
While hospitalized COVID-19 patients and vaccinees maintained sufficient neutralizing titers against all
37
three VOC, 39% of non-hospitalized patients did not neutralize B.1.351. Moreover, monoclonal neutralizing
38
antibodies (NAbs) show sharp reductions in their binding kinetics and neutralizing potential to B.1.351 and
39
P.1, but not to B.1.1.7. These data have implications for the degree to which pre-existing immunity can
40
protect against subsequent infection with VOC and informs policy makers of susceptibility to globally
41
circulating SARS-CoV-2 VOC.
42
43
Introduction
44
With over 140 million confirmed infections and over three million deaths as of April 2021, the coronavirus
45
disease 2019 (COVID-19) pandemic shows few signs of abating (1). While severe acute respiratory
46
syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, remained relatively stable
47
genetically and antigenically during the first stage of the pandemic, higher levels of genetic variation have
48
been observed during the second wave of the pandemic with considerable genetic changes compared to
49
the original Wuhan Hu-1 strain of SARS-CoV-2. These genetic changes include substitutions within the
50
functional domains of the SARS-CoV-2 spike (S) protein resulting in altered phenotypes of the virus. The
51
WHO has currently defined three VOC based on possible increase in transmissibility or change in COVID-
52
19 epidemiology, increase in virulence or change in clinical disease presentation, or decrease in
53
effectiveness of available diagnostics, vaccines and therapeutics. These VOC include B.1.1.7
54
(20I/N501Y.V1, first detected in the United Kingdom), B.1.351 (20H/N501Y.V2, first detected in South
55
Africa) and B.1.1.28.P1 (P.1, 20J/N501Y.V3, first detected in Brazil), which have since spread globally and
56
as of April 2021, cases with these variants have been reported in 132, 82 and 52 countries, respectively
57
(25). The emergence of these variants has raised concerns as to whether immunity, be it natural
58
immunity from prior infection or immunity from vaccination, can protect against these different variants.
59
Moreover, it is unclear whether therapeutic monoclonal NAbs isolated from convalescent donors retain
60
therapeutic efficacy against these new variants.
61
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted June 1, 2021. ; https://doi.org/10.1101/2021.05.26.21257441doi: medRxiv preprint

The S protein of coronaviruses is the main target of NAbs and consists of a membrane-proximal
62
S2 domain containing the fusion peptide and a membrane-distal S1 domain containing the receptor
63
binding domain (RBD) and the N-terminal domain (NTD) (Fig. 1A). The S protein mediates viral entry
64
through interaction with the angiotensin-converting enzyme 2 (ACE2) receptor on host cells (6, 7).
65
Therefore, substitutions present in the RBD of these emerging VOC are particularly worrisome as they
66
might improve binding to the human receptor and thus increase viral fitness and transmissibility. Indeed,
67
B.1.1.7, B.1.351 and P.1 all share the N501Y RBD mutation, which contributes to an enhanced interaction
68
with the human ACE2 receptor resulting in increased infectivity and transmissibility (Fig. 1A) (8, 9). In
69
addition, both B.1.351 and P.1 contain the E484K substitution and a substitution at position 417 (K417N in
70
B.1.351 and K417T in P.1) both of which have been implicated in escape from NAbs by several studies
71
(Fig. 1A) (10, 11). More recently, E484K has also been observed in B.1.1.7 variants that have adopted this
72
mutation independently, leading to a more substantial loss of neutralizing titers in vaccinated individuals
73
than for B.1.1.7 alone (12). Additional substitutions mostly arise in the NTD region, located apically on the
74
S1 domain. Although the exact functional implication of substitutions within the NTD is not clear, it has
75
been shown that this domain is an important target for NAbs (13, 14). VOC B.1.351 and P.1 both carry five
76
mutations in this region of which they share one (L18F). Early reports on the B.1.351 lineage reported
77
variants with a substitution at position 242 and 246 (L242H and R246I), while other sublineages had a
78
three amino acid deletion (Δ242-244) (4). B.1.1.7 does not have substitutions in its NTD but has deletions
79
in this region (Δ69-70, Δ144) (Fig. 1A). Other mutations are identified in the S2 domain of the S protein
80
and their impact on antibody recognition and infectivity is not yet well understood, partly because the vast
81
majority of NAbs target the RBD or NTD. Finally, all these variants have the D614G mutation that defines
82
the B.1 lineage and became dominant throughout 2020, now being present in the large majority of
83
sequenced SARS-CoV-2 variants (> 99%) (Fig. 1A) (15).
84
All three VOC have been linked to increased infectivity and transmissibility, and the first reports
85
about specific substitutions in B.1.351 and P.1 associated with escape from immunity by infection or
86
mRNA vaccines have become available (10, 1618). These first reports suggest that pre-existing immunity
87
was generally sufficient to neutralize B.1.1.7 to similar levels as WT in mRNA vaccine recipients and in
88
convalescent individuals up to nine months after infection (17, 19). In case of B.1.351, the impact is more
89
substantial with neutralizing titers from convalescent patients being reduced by ~6 to 13-fold while vaccine
90
recipients are reported to have a ~10 to 14-fold reduction (10, 17, 18). Moreover, it is reported that ~40%
91
of convalescent patients do not have any neutralizing activity against B.1.351 nine months after primary
92
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted June 1, 2021. ; https://doi.org/10.1101/2021.05.26.21257441doi: medRxiv preprint

infection (17). A recent study showed that although B.1.351 and P.1 have similar mutations in their RBD,
93
sera from convalescent patients as well as mRNA vaccine recipients showed a neutralization reduction of
94
~3-fold against P.1 while this was 7-13-fold for B.1.351 (20, 21).
95
Although these studies have provided valuable initial insights in altered antigenic properties of
96
these VOC, few studies have compared all three VOC side-by-side. Furthermore, comparing immune
97
responses from vaccine recipients and individuals who have suffered from mild or severe COVID-19 as
98
well as reactivity of NAbs to all three VOC within the same study will provide useful insights that can be
99
used for additional surveillance of SARS-CoV-2 variants, the use of monoclonal antibodies for treatment
100
and modifications of vaccines in order to increase immune coverage of immunity to include yet unknown
101
emerging SARS-CoV-2 variants.
102
103
Results
104
Recognition of SARS-CoV-2 VOC by convalescent and vaccine sera is reduced
105
Here, we assessed the impact of VOC B.1.1.7, B.1.351 and P.1 on humoral immunity elicited either by
106
mRNA vaccination or by natural infection with SARS-CoV-2. To this end, we studied two cohorts of
107
individuals: the COSCA cohort that included convalescent COVID-19 patients (n = 69) and the S3 cohort
108
that included health care workers (HCW) who were vaccinated twice with the Pfizer/BioNTech COVID-19
109
vaccine (n = 50) (Table 1). The COSCA cohort included hospitalized and non-hospitalized patients in the
110
Netherlands who were enrolled and sampled four to six weeks after symptom onset (i.e. close to the
111
expected peak of humoral immunity) between March 2020 and January 2021 (Table 1). Based on the
112
rarity of B.1.351 and P.1 in the Netherlands, no individuals are expected to have been infected with either
113
VOC. However, infections with B.1.1.7 became more prevalent towards the end of 2020, with the
114
estimated prevalence of B.1.1.7 reaching 24% in the Netherlands in the week of the last inclusion
115
(January 2021) (22). As genomic data was not collected as part of this study, we cannot exclude that a
116
few patients were infected with B.1.1.7. The S3 cohort consists of health care workers without prior SARS-
117
CoV-2 infection who received the approved Pfizer/BioNTech COVID-19 mRNA vaccine twice with a three
118
week interval and sampled four weeks after the second vaccination (Table 1).
119
We first assessed S protein binding titers of convalescent and vaccinee sera in a custom multiplex
120
protein microarray that has been extensively validated for clinical use (table S1) (23). We generated S
121
proteins, using previously described stabilization approaches, from all three VOC that had been identified
122
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted June 1, 2021. ; https://doi.org/10.1101/2021.05.26.21257441doi: medRxiv preprint

at the time of assessment as well as a control wild-type (WT) S protein from the Wuhan Hu-1 virus
123
(GenBank: MN908947.3) isolated in December 2019 (6, 24, 25). Overall, the antibody responses against
124
each S protein were heterogeneous and differed up to ~1200-fold between the strongest and weakest
125
responders. The recognition of the three VOC by convalescent patients was significantly reduced
126
compared to WT by an average of 2.4-fold, 3-fold and 4-fold for B.1.1.7, B.1.351 and P.1, respectively (p <
127
0.0001 for all, Fig. 1, B and C). Binding titers elicited by the mRNA vaccine were more homogeneous than
128
those elicited by natural infection and differed ~10-fold between responders, with all participants having
129
half-maximal binding titers (ED
50
s) exceeding 10
3
, except for one poor responder (Fig. 1B). Since the
130
variability of binding titers in convalescent sera is considerable, we examined whether this variability was
131
related to severity of disease (i.e. hospital admission, Fig. 1D). We observed a highly significant ~8-fold
132
difference in ED
50
s between non-hospitalized patients and hospitalized patients, which is in line with
133
previous reports of WT SARS-CoV-2 binding titers correlating with severity of disease (p < 0.0001) (26,
134
27). This difference in binding titers between non-hospitalized patients and hospitalized patients was
135
consistent for all three SARS-CoV-2 VOC studied (Fig. 1D). When comparing immune responses of
136
vaccine recipients with convalescent sera, we observed similar S protein binding titers between vaccinee
137
and hospitalized patients, which are an average ~4 to 11-fold higher compared to non-hospitalized
138
patients for all VOC (Fig. 1D). Taken together, these data indicate that vaccine recipients as well as
139
COVID-19 patients exhibit reduced binding to S proteins of the currently circulating VOC, with hospitalized
140
patients and vaccine recipients exhibiting higher binding titers overall compared to non-hospitalized
141
patients.
142
143
SARS-CoV-2 VOC are substantially less sensitive to serum NAbs
144
We next tested the neutralizing activity of convalescent and vaccinee sera (Fig. 2, table S1). To this end,
145
we generated lentiviral-based pseudoviruses of the currently widespread SARS-CoV-2 D614G (WT)
146
variant as well as B.1.1.7, B.1.351 and P.1. We detected substantial neutralizing activity against the WT
147
virus (half maximal neutralization titer, ID
50
> 100) in 96% of convalescent patients irrespective of
148
hospitalization, and in all vaccine recipients, except the one poor responder (Fig. 2A). Indeed, the overall
149
binding titers correlated well with the neutralization titers for all VOC (fig. S1C). Non-hospitalized patients
150
had the most heterogeneous responses, with WT neutralizing titers differing by up to ~150-fold, while titers
151
against WT were much more homogeneous in hospitalized patients (up to ~20-fold difference) and
152
vaccine recipients (up to ~12-fold difference) (Fig. 2A). Consistent with the binding results, we observed a
153
All rights reserved. No reuse allowed without permission.
(which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
The copyright holder for this preprintthis version posted June 1, 2021. ; https://doi.org/10.1101/2021.05.26.21257441doi: medRxiv preprint

Citations
More filters
Posted ContentDOI

Mapping the antigenic diversification of SARS-CoV-2

TL;DR: SARS-CoV-2 antigenic drift is studied by assessing neutralizing activity against variants-of-concern (VOCs) of a unique set of sera from patients infected with a range of VOCs to reveal Omicron forms a new antigenic cluster associated with immune escape and likely requiring vaccine updates to ensure vaccine effectiveness.
Journal ArticleDOI

Mutations of SARS-CoV-2 spike protein: Implications on immune evasion and vaccine-induced immunity.

TL;DR: In this article, the authors discuss the implications of SARS-CoV-2 spike protein mutations on immune evasion and vaccine-induced immunity and forward directions which could contribute to future studies focusing on designing effective vaccines and/or immunotherapies to consider viral evolution.
References
More filters
Journal ArticleDOI

An interactive web-based dashboard to track COVID-19 in real time.

TL;DR: The outbreak of the 2019 novel coronavirus disease (COVID-19) has induced a considerable degree of fear, emotional stress and anxiety among individuals around the world.
Journal ArticleDOI

Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation.

TL;DR: The authors show that this protein binds at least 10 times more tightly than the corresponding spike protein of severe acute respiratory syndrome (SARS)–CoV to their common host cell receptor, and test several published SARS-CoV RBD-specific monoclonal antibodies found that they do not have appreciable binding to 2019-nCoV S, suggesting that antibody cross-reactivity may be limited between the two RBDs.
Journal ArticleDOI

Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein.

TL;DR: It is demonstrating that cross-neutralizing antibodies targeting conserved S epitopes can be elicited upon vaccination, and it is shown that SARS-CoV-2 S uses ACE2 to enter cells and that the receptor-binding domains of Sars- coV- 2 S and SARS S bind with similar affinities to human ACE2, correlating with the efficient spread of SATS among humans.
Journal ArticleDOI

Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England.

TL;DR: Using a variety of statistical and dynamic modeling approaches, the authors estimate that this variant has a 43 to 90% (range of 95% credible intervals, 38 to 130%) higher reproduction number than preexisting variants, and a fitted two-strain dynamic transmission model shows that VOC 202012/01 will lead to large resurgences of COVID-19 cases.
Related Papers (5)
Frequently Asked Questions (1)
Q1. What contributions have the authors mentioned in the paper "Emerging sars-cov-2 variants of concern evade humoral immune responses from infection and vaccination" ?

These VOC include B. 1. 1. 7 54 ( 20I/N501Y. V1, first detected in the United Kingdom ), B. 1. 351 ( 20H/N501Y. V2, first detected in South 55 Africa ) and B. 1. 1. 28. P1 ( P. 1, 20J/N501Y. V3, first detected in Brazil ), which have since spread globally and 56 as of April 2021, cases with these variants have been reported in 132, 82 and 52 countries, respectively 57 ( 2–5 ). ( which was not certified by peer review ) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.