scispace - formally typeset
Open AccessPosted ContentDOI

Naturally enhanced neutralizing breadth to SARS-CoV-2 after one year

TLDR
In the absence of vaccination antibody reactivity to the receptor binding domain (RBD) of SARS-CoV-2, neutralizing activity and the number of RBD-specific memory B cells remain relatively stable from 6 to 12 months as discussed by the authors.
Abstract
Over one year after its inception, the coronavirus disease-2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) remains difficult to control despite the availability of several excellent vaccines. Progress in controlling the pandemic is slowed by the emergence of variants that appear to be more transmissible and more resistant to antibodies 1,2 . Here we report on a cohort of 63 COVID-19-convalescent individuals assessed at 1.3, 6.2 and 12 months after infection, 41% of whom also received mRNA vaccines 3,4 . In the absence of vaccination antibody reactivity to the receptor binding domain (RBD) of SARS-CoV-2, neutralizing activity and the number of RBD-specific memory B cells remain relatively stable from 6 to 12 months. Vaccination increases all components of the humoral response, and as expected, results in serum neutralizing activities against variants of concern that are comparable to or greater than neutralizing activity against the original Wuhan Hu-1 achieved by vaccination of naive individuals 2,5-8 . The mechanism underlying these broad-based responses involves ongoing antibody somatic mutation, memory B cell clonal turnover, and development of monoclonal antibodies that are exceptionally resistant to SARS-CoV-2 RBD mutations, including those found in variants of concern 4,9 . In addition, B cell clones expressing broad and potent antibodies are selectively retained in the repertoire over time and expand dramatically after vaccination. The data suggest that immunity in convalescent individuals will be very long lasting and that convalescent individuals who receive available mRNA vaccines will produce antibodies and memory B cells that should be protective against circulating SARS-CoV-2 variants.

read more

Content maybe subject to copyright    Report

Naturally enhanced neutralizing breadth to SARS-CoV-2 after one year
1
2
3
Zijun Wang
1,*
, Frauke Muecksch
2,*
, Dennis Schaefer-Babajew
1,*
, Shlomo Finkin
1,*
, Charlotte
4
Viant
1,*
, Christian Gaebler
1,*
, Hans- Heinrich Hoffmann
3
, Christopher O. Barnes
4
, Melissa
5
Cipolla
1
, Victor Ramos
1
, Thiago Y. Oliveira
1
, Alice Cho
1
, Fabian Schmidt
2
, Justin da Silva
2
, Eva
6
Bednarski
2
, Lauren Aguado
3
, Jim Yee
5
, Mridushi Daga
1
, Martina Turroja
1
, Katrina G. Millard
1
,
7
Mila Jankovic
1
, Anna Gazumyan
1, 6
, Zhen Zhao
5
, Charles M. Rice
3
, Paul D. Bieniasz
2, 6
, Marina
8
Caskey
1
, Theodora Hatziioannou
2
, Michel C. Nussenzweig
1, 6
9
10
11
12
1
Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
13
2
Laboratory of Retrovirology, The Rockefeller University, New York, NY 10065, USA
14
3
Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY,
15
10065, USA.
16
4
Division of Biology and Biological Engineering, California Institute of Technology, Pasadena,
17
CA, USA.
18
5
Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY,
19
10065, USA  
20
6
Howard Hughes Medical Institute
21
22
*equal contribution
23
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted June 2, 2021. ; https://doi.org/10.1101/2021.05.07.443175doi: bioRxiv preprint

Address correspondence to: Paul D. Bieniasz, pbieniasz@rockefeller.edu; Marina Caskey,
24
mcaskey@rockefeller.edu; Theodora Hatziioannou, thatziio@rockefeller.edu; or Michel C.
25
Nussenzweig, nussen@rockefeller.edu.
26
27
28
Over one year after its inception, the coronavirus disease-2019 (COVID-19) pandemic
29
caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) remains
30
difficult to control despite the availability of several excellent vaccines. Progress in
31
controlling the pandemic is slowed by the emergence of variants that appear to be more
32
transmissible and more resistant to antibodies
1,2
. Here we report on a cohort of 63 COVID-
33
19-convalescent individuals assessed at 1.3, 6.2 and 12 months after infection, 41% of
34
whom also received mRNA vaccines
3,4
. In the absence of vaccination antibody reactivity to
35
the receptor binding domain (RBD) of SARS-CoV-2, neutralizing activity and the number
36
of RBD-specific memory B cells remain relatively stable from 6 to 12 months. Vaccination
37
increases all components of the humoral response, and as expected, results in serum
38
neutralizing activities against variants of concern that are comparable to or greater than
39
neutralizing activity against the original Wuhan Hu-1 achieved by vaccination of naïve
40
individuals
2,5-8
. The mechanism underlying these broad-based responses involves ongoing
41
antibody somatic mutation, memory B cell clonal turnover, and development of
42
monoclonal antibodies that are exceptionally resistant to SARS-CoV-2 RBD mutations,
43
including those found in variants of concern
4,9
. In addition, B cell clones expressing broad
44
and potent antibodies are selectively retained in the repertoire over time and expand
45
dramatically after vaccination. The data suggest that immunity in convalescent individuals
46
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted June 2, 2021. ; https://doi.org/10.1101/2021.05.07.443175doi: bioRxiv preprint

will be very long lasting and that convalescent individuals who receive available mRNA
47
vaccines will produce antibodies and memory B cells that should be protective against
48
circulating SARS-CoV-2 variants.
49
50
We initially characterized immune responses to SARS-CoV-2 in a cohort of convalescent
51
individuals 1.3 and 6.2 months after infection
3,4
. Between February 8 and March 26, 2021, 63
52
participants between the ages of 26 and 73 years old (median 47 years) returned for a 12-month
53
follow-up visit. Among those, 26 (41%) had received at least one dose of either the Moderna
54
(mRNA-1273) or Pfizer-BioNTech (BNT162b2) vaccines, on average 40 days (range 2-82 days)
55
before their study visit and 311 days (range 272-373 days) after onset of acute illness
56
(Supplementary Table 1). Participants were was almost evenly split between sexes (43% female)
57
and of the individuals that returned for a 12-month follow-up, only 10% had been hospitalized
58
and the remainder had experienced relatively mild initial infections. Only 14% of the individuals
59
reported persistent long-term symptoms after 12 months, reduced from 44% at the 6-month time
60
point
4
. Symptom persistence was not associated with the duration and severity of acute disease
61
or with vaccination status (Extended Data Fig. 1 a-c). All participants tested negative for active
62
infection at the 12-month time point as measured by a saliva-based PCR assay
4
. The
63
demographics and clinical characteristics of the participants are shown in Supplementary Tables
64
1 and 2.
65
66
Plasma SARS-CoV-2 Antibody Reactivity
67
Antibody reactivity in plasma to the RBD and nucleoprotein (N) were measured by enzyme-
68
linked immunosorbent assay (ELISA)
3
. We limited our analysis to RBD because plasma anti-
69
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted June 2, 2021. ; https://doi.org/10.1101/2021.05.07.443175doi: bioRxiv preprint

RBD antibodies are strongly correlated with neutralizing activity
3,10-12
. Convalescent
70
participants who had not been vaccinated maintained most of their anti-RBD IgM (103%), IgG
71
(82%), and IgA (72%) titers between 6 and 12 months (Fig. 1a and Extended Data Fig. 2a-k).
72
Consistent with previous reports
5-8
, vaccination increased the anti-RBD plasma antibody levels,
73
with IgG titers increasing by nearly 30-fold compared to unvaccinated individuals (Fig. 1a right).
74
The 2 individuals who did not show an increase had been vaccinated only 2 days before sample
75
collection. In contrast to anti-RBD antibody titers that were relatively stable, anti-N antibody
76
titers decreased significantly between 6 and 12 months in this assay irrespective of vaccination
77
(Fig. 1b and Extended Data Fig. 2l-n).
78
79
Plasma neutralizing activity in 63 participants was measured using an HIV-1 pseudotyped with
80
the SARS-CoV-2 spike protein
3,4,13
(Fig. 1c-d and Extended Data Fig. 2o). Twelve-months after
81
infection, the geometric mean half-maximal neutralizing titer (NT
50
) for the 37 individuals that
82
had not been vaccinated was 75, which was not significantly different from the same individuals
83
at 6.2 months (Fig. 1c). In contrast, the vaccinated individuals showed a geometric mean NT
50
of
84
3,684, which was nearly 50-fold greater than unvaccinated individuals and slightly better
85
compared to the 30-fold increase in anti-RBD IgG antibodies (Fig. 1a, c, and d). Neutralizing
86
activity was directly correlated with IgG anti-RBD (Extended Data Fig.2p) but not with anti-N
87
titers (Extended Data Fig.2r). We conclude that neutralizing titers remain relatively unchanged
88
between 6 to 12 months after SARS-CoV-2 infection, and that vaccination further boosts this
89
activity by nearly 50-fold.
90
91
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted June 2, 2021. ; https://doi.org/10.1101/2021.05.07.443175doi: bioRxiv preprint

To determine the neutralizing activity against circulating variants of concern/interest, we
92
performed neutralization assays on HIV-1 virus pseudotyped with the S protein of the following
93
SARS-CoV-2 variants of concern/interest: B.1.1.7, B.1.351, B.1.526 and P.1
1,14,15
. Twelve-
94
months after infection neutralizing activity against the variants was generally lower than against
95
wild-type SARS-CoV-2 virus in the same assay with the greatest loss of activity against B.1.351
96
(Fig. 1e). After vaccination the geometric mean NT
50
rose to 11,493, 48,341, 22,109 and 26,553
97
against B.1.351, B.1.1.7, B.1.526 and P.1, respectively. These titers are an order of magnitude
98
higher than the neutralizing titers we and others have reported against the wild-type SARS-CoV-
99
2 at the peak of the initial response in infected individuals and in naïve individuals receiving both
100
doses of mRNA vaccines (Fig. 1d and
2-8
). Similar results were also obtained using authentic
101
SARS-CoV-2 WA1/2020 and B.1.351 (Extended Data Fig.2s).
102
103
Memory B cells
104
The memory B cell compartment serves as an immune reservoir that contains a diverse collection
105
of antibodies
16,17
. Although antibodies to the N-terminal domain and other parts of S can also be
106
neutralizing, we limited our analysis to memory B cells that produce anti-RBD antibodies
107
because they are the most numerous and potent
18,19
. To enumerate RBD-specific memory B
108
cells, we performed flow cytometry using a biotin-labeled RBD
3
(Fig. 2a and Extended Data Fig.
109
3a and b). In the absence of vaccination, the number of RBD specific memory B cells present at
110
12 months was only 1.35-fold lower than the earlier timepoint (p= 0.027, Fig. 2a). In contrast
111
and consistent with previous reports
5,8,20
, convalescent individuals that received mRNA vaccines
112
showed an average 8.6-fold increase in the number of circulating RBD specific memory B cells
113
(Fig. 2a). B cells expressing antibodies that bound to both wild-type and K417N/E484K/N501Y
114
was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprint (whichthis version posted June 2, 2021. ; https://doi.org/10.1101/2021.05.07.443175doi: bioRxiv preprint

Citations
More filters
Journal ArticleDOI

Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics

TL;DR: In this paper, a suite of recently developed techniques that localize RNA within tissue, including multiplexed in situ hybridization and in situ sequencing (here defined as high-plex RNA imaging) and spatial barcoding, can help address this issue.
Journal ArticleDOI

Long-term persistence of SARS-CoV-2 neutralizing antibody responses after infection and estimates of the duration of protection.

TL;DR: There will be relatively long-lived protection from re-infection following symptomatic COVID-19 disease, and there was a high level of correlation between PRNT50 antibody titers and the % of inhibition in surrogate virus neutralization tests.
Posted ContentDOI

Convergent epitope-specific T cell responses after SARS-CoV-2 infection and vaccination

TL;DR: In this paper, the authors compared epitope-specific T cells elicited after natural SARS-CoV-2 infection, and vaccination of both naive and recovered individuals, and found that T cells responding to spike-derived epitopes show similar magnitudes of response, memory phenotypes, TCR repertoire diversity, and αβTCR sequence motifs, demonstrating the potency of this vaccination platform.
References
More filters
Journal ArticleDOI

Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction

TL;DR: A new method of total RNA isolation by a single extraction with an acid guanidinium thiocyanate-phenol-chloroform mixture is described, providing a pure preparation of undegraded RNA in high yield and can be completed within 4 h.
Journal ArticleDOI

A new coronavirus associated with human respiratory disease in China.

TL;DR: Phylogenetic and metagenomic analyses of the complete viral genome of a new coronavirus from the family Coronaviridae reveal that the virus is closely related to a group of SARS-like coronaviruses found in bats in China.
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)
Trending Questions (1)
What do B cells do after vaccination?

In addition, B cell clones expressing broad and potent antibodies are selectively retained in the repertoire over time and expand dramatically after vaccination.