SlideShare a Scribd company logo
1 of 26
Download to read offline
Article
Robust T Cell Immunity in Convalescent Individuals
with Asymptomatic or Mild COVID-19
Graphical Abstract
Highlights
d Acute-phase SARS-CoV-2-specific T cells display an
activated cytotoxic phenotype
d Convalescent-phase SARS-CoV-2-specific T cells generate
broad responses
d Polyfunctional SARS-CoV-2-specific T cells also occur in
seronegative individuals
Authors
Takuya Sekine, André Perez-Potti,
Olga Rivera-Ballesteros, ...,
Hans-Gustaf Ljunggren, Soo Aleman,
Marcus Buggert
Correspondence
marcus.buggert@ki.se
In Brief
Sekine et al. provide a functional and
phenotypic map of SARS-CoV-2-specific
T cells across the full spectrum of
exposure, infection, and COVID-19
severity. They observe that SARS-CoV-2
elicits broadly directed and functionally
replete memory T cells that may protect
against recurrent episodes of severe
COVID-19.
Sekine et al., 2020, Cell 183, 158–168
October 1, 2020 ª 2020 The Author(s). Published by Elsevier Inc.
https://doi.org/10.1016/j.cell.2020.08.017 ll
Article
Robust T Cell Immunity in Convalescent Individuals
with Asymptomatic or Mild COVID-19
Takuya Sekine,1,15 André Perez-Potti,1,15 Olga Rivera-Ballesteros,1,15 Kristoffer Strålin,2,16 Jean-Baptiste Gorin,1,16
Annika Olsson,2 Sian Llewellyn-Lacey,3 Habiba Kamal,2 Gordana Bogdanovic,4,13 Sandra Muschiol,4,13
David J. Wullimann,1 Tobias Kammann,1 Johanna Emgård,1 Tiphaine Parrot,1 Elin Folkesson,2 Karolinska COVID-19
Study Group, Olav Rooyackers,5,6 Lars I. Eriksson,6,7 Jan-Inge Henter,8,14 Anders Sönnerborg,2,9 Tobias Allander,4,9,13
Jan Albert,4,9,13 Morten Nielsen,10,11 Jonas Klingström,1 Sara Gredmark-Russ,1,2 Niklas K. Björkström,1
Johan K. Sandberg,1 David A. Price,3,12 Hans-Gustaf Ljunggren,1,15 Soo Aleman,1,2,15 and Marcus Buggert1,15,17,*
1Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
2Division of Infectious Diseases, Karolinska University Hospital, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
3Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
4Division of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
5Department of Clinical Interventions and Technology, Karolinska Institutet, Stockholm, Sweden
6Function Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
7Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
8Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
9Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
10Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
11Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martı́n, San Martı́n, Argentina
12Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
13Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
14Theme of Children’s and Women’s Health, Karolinska University Hospital, Stockholm, Sweden
15These authors contributed equally
16These authors contributed equally
17Lead Contact
*Correspondence: marcus.buggert@ki.se
https://doi.org/10.1016/j.cell.2020.08.017
SUMMARY
SARS-CoV-2-specific memory T cells will likely prove critical for long-term immune protection against
COVID-19. Here, we systematically mapped the functional and phenotypic landscape of SARS-CoV-2-spe-
cific T cell responses in unexposed individuals, exposed family members, and individuals with acute or
convalescent COVID-19. Acute-phase SARS-CoV-2-specific T cells displayed a highly activated cytotoxic
phenotype that correlated with various clinical markers of disease severity, whereas convalescent-phase
SARS-CoV-2-specific T cells were polyfunctional and displayed a stem-like memory phenotype. Importantly,
SARS-CoV-2-specific T cells were detectable in antibody-seronegative exposed family members and conva-
lescent individuals with a history of asymptomatic and mild COVID-19. Our collective dataset shows that
SARS-CoV-2 elicits broadly directed and functionally replete memory T cell responses, suggesting that nat-
ural exposure or infection may prevent recurrent episodes of severe COVID-19.
INTRODUCTION
The world changed in December 2019 with the emergence of a
new zoonotic pathogen, severe acute respiratory syndrome co-
ronavirus 2 (SARS-CoV-2), which causes a variety of clinical syn-
dromes collectively termed coronavirus disease 2019 (COVID-
19). At present, there is no vaccine against SARS-CoV-2, and
the excessive inflammation associated with severe COVID-19
can lead to respiratory failure, septic shock, and ultimately,
death (Guan et al., 2020; Wolfel et al., 2020; Wu and McGoogan,
2020). The overall mortality rate is 0.5%–3.5% (Guan et al., 2020;
Wolfel et al., 2020; Wu and McGoogan, 2020). However, most
people seem to be affected less severely and remain asymptom-
atic or develop only mild symptoms during COVID-19 (He et al.,
2020b; Wei et al., 2020; Yang et al., 2020). It will therefore be crit-
ical for public health reasons to determine whether people with
milder forms of COVID-19 develop robust immunity against
SARS-CoV-2.
Global efforts are currently underway to map the determi-
nants of immune protection against SARS-CoV-2. Recent data
have shown that SARS-CoV-2 infection generates near-com-
plete protection against rechallenge in rhesus macaques
ll
OPEN ACCESS
158 Cell 183, 158–168, October 1, 2020 ª 2020 The Author(s). Published by Elsevier Inc.
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
A
0
10
20
30
40
50
%
of
memory
CD4+
T
cells
CD38
0
1
2
3
4
5
CD69
0
5
10
15
20
25
Ki-67
2020 BD MC AM AS
0
20
40
60
80
100
PD-1
0
20
40
60
80
100
CD38
%
of
memory
CD8+
T
cells
0
10
20
30
CD39
0
5
10
15
20
CD69
0
5
10
15
CTLA-4
0
10
20
30
40
50
HLA-DR
0
10
20
30
Ki-67
0
5
10
15
LAG-3
0
5
10
15
TIM-3
-6
-4
-2
0
2
4
6
PC2
-2
0
2
4
6
8
PC2
Memory CD4+ T cells Memory CD8+ T cells
Ki-67
CD38
TIM-3
HLA-DR
PD-1
CD69
CD39 RA
CTLA-4
CXCR5
CD25
CD27
CCR7
CD28
TIGIT
TCF1
CD127 LAG-3
Perforin
TOX
2B4
GzmB
CD38
CTLA-4
Ki-67CD39
LAG-3
TIM-3
HLA-DR
PD-1
CXCR5
CD69 CCR7
CD27
CD25
CD28
CD127
TIGIT
Perforin
TOX
GzmB
2B4
RA
TCF1
0
1000000
2000000
3000000
Abs count T cells
0
10
20
30
40
%CD3+CD15-CD14-
CD19-
of
CD45+
% T cells
200000
400000
600000
800000
2000000
Abs count CD8+ T cells
0
500000
1000000
1500000
2000000
Abs count CD4+ T cells
0
5
10
20
% CD8+ T cells
0
10
20
30
% CD4+ T cells
0
%CD3+CD8+
of
CD45+
%CD3+CD8+
of
CD45+
B C
2020 BD AM AS
1
1
<1
30
27
14
2020 BD AS
Gated on memory CD8+ T cells
0
20
40
60
0
10
20
30
40
0
5
10
15
20
CD38
Ki-67
HLA-DR
PD-1
CD38+PD-1+
CD38+HLA-DR+
CD38+Ki-67+
%
of
memory
CD8+
T
cells
E
2020 BD MC AM AS
UMAP2
UMAP1
D
HLA-DR
Ki-67
CD38
LAG-3
TIM-3
CD39
2B4
TIGIT
PD-1
CD27
CD127
CXCR5
GzmB
Perforin
CD95
CD28
CD45RA
CCR7
Gated on memory
CD8+ T cells
Max
Min
CTLA-4 CD25
CD69
TOX TCF1
2020 BD MC AM AS
*
Cycling
TEMRA
TEM
TCM
-2
0
2
4
6
8
PC2
(22
%)
-5 5
2.5
0
-2.5 10
7.5
PC1 (34 %)
-2
0
2
4
6
8
PC2
(22
%)
-5 5
2.5
0
-2.5 10
7.5
PC1 (34 %)
-6
-4
-2
0
2
4
PC2
(22
%)
6
4
2
0
-2 10
6
PC1 (34 %)
8
-6
-4
-2
0
2
4
PC2
(22
%)
6
4
2
0
-2 10
6
PC1 (34 %)
8
***
***
***
**
***
***
***
**
***
*
***
*** ***
*
*** *
***
**
***
***
*
**
***
***
***
***
*** **
**
***
***
***
***
***
***
**
***
***
*
*
**
**
**
***
**
***
**
**
***
***
***
**
***
*** **
***
***
***
***
***
***
**
***
***
***
***
***
***
***
Figure 1. T Cell Perturbations in COVID-19
(A) Dot plots summarizing the absolute counts and relative frequencies of CD3+
(left), CD4+
(center), and CD8+
T cells (right) in healthy blood donors from 2020
(2020 BD ) and patients with acute moderate (AM ) or acute severe COVID-19 (AS). Each dot represents one donor. Data are shown as median ± IQR. *p < 0.05,
***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons.
(B) Top: PCA plots showing the distribution and segregation of memory CD4+
and CD8+
T cells by group. Each dot represents one donor. Memory cells were
defined by exclusion of naive cells (CCR7+
CD45RA+
CD95 ). Center: PCA plots showing the corresponding trajectories of key markers that influenced the group-
(legend continued on next page)
ll
OPEN ACCESS
Cell 183, 158–168, October 1, 2020 159
Article
(Chandrashekar et al., 2020), and similarly, there is limited evi-
dence of reinfection in humans with previously documented
COVID-19 (Kirkcaldy et al., 2020). Further work is therefore
required to define the mechanisms that underlie these observa-
tions and evaluate the durability of protective immune responses
elicited by primary infection with SARS-CoV-2. Most correlative
studies of immune protection against SARS-CoV-2 have
focused on induction of neutralizing antibodies (Hotez et al.,
2020; Robbiani et al., 2020; Seydoux et al., 2020; Wang et al.,
2020). However, antibody responses are not detectable in all pa-
tients, especially those with less severe forms of COVID-19
(Long et al., 2020; Mallapaty, 2020; Woloshin et al., 2020). Previ-
ous work has also shown that memory B cell responses tend to
be short lived after infection with SARS-CoV-1 (Channappanavar
et al., 2014; Tang et al., 2011). In contrast, memory T cell re-
sponses can persist for many years (Le Bert et al., 2020; Tang
et al., 2011; Yang et al., 2006) and, in mice, protect against lethal
challenge with SARS-CoV-1 (Channappanavar et al., 2014).
SARS-CoV-2-specific T cells have been identified in humans
(Grifoni et al., 2020; Ni et al., 2020). It has nonetheless remained
unclear to what extent various features of the T cell immune
response associate with serostatus and the clinical course of
COVID-19. To address this knowledge gap, we characterized
SARS-CoV-2-specific CD4+
and CD8+
T cells in outcome-
defined cohorts of donors (total n = 206) from Sweden, which
has experienced a more open spread of COVID-19 than many
other countries in Europe (Habib, 2020).
RESULTS
T Cell Perturbations in COVID-19
Our preliminary analyses showed that the absolute numbers and
relative frequencies of CD4+
and CD8+
T cells were unphysiolog-
ically low in patients with acute moderate or severe COVID-19
(Figures 1A, S1A, and S1B). This finding has been reported pre-
viously (He et al., 2020a; Liu et al., 2020). We then used a 29-co-
lor flow cytometry panel to assess the phenotypic landscape of
these immune perturbations in direct comparisons with healthy
blood donors and individuals who had recovered from mild
COVID-19 acquired early during the pandemic (February–March
2020). Cohort demographics are described in the STAR Methods
section. None of these variables were associated with disease
severity. The following parameters were measured in each sam-
ple: viability, C-C chemokine receptor type 7 (CCR7), cluster of
differentiation 3 (CD3), CD4, CD8, CD14, CD19, CD25, CD27,
CD28, CD38, CD39, CD45RA, CD69, CD95, CD127, cytotoxic
T-lymphocyte-associated protein 4 (CTLA-4), C-X-C chemokine
receptor type 5 (CXCR5), granzyme B, human leukocyte antigen
(HLA)-DR, Ki-67, lymphocyte activation gene 3 (LAG-3), pro-
grammed cell death protein 1 (PD-1), perforin, T cell factor 1
(TCF1), T cell immunoreceptor with immunoglobulin (Ig) and
ITIM domains (TIGIT), T cell Ig and mucin domain-containing pro-
tein 3 (TIM-3), thymocyte selection-associated high-mobility
group box factor (TOX), and natural killer cell receptor 2B4.
Unbiased principal component analysis (PCA) revealed clear
segregation between memory T cells from patients with acute
moderate or severe COVID-19 and memory T cells from conva-
lescent individuals and healthy blood donors (Figure 1B), driven
largely by expression of CD38, CD69, Ki-67, and PD-1 in the
CD4+
compartment and expression of CD38, CD39, CD69,
CTLA-4, HLA-DR, Ki-67, LAG-3, and TIM-3 in the CD8+
compart-
ment (Figures 1B, 1C, and S1C).
To extend these findings, we concatenated all memory CD4+
T cells and all memory CD8+
T cells from healthy blood donors,
convalescent individuals, and patients with acute moderate or
severe COVID-19. Phenotypically related cells were identified
using the clustering algorithm PhenoGraph, and marker expres-
sion patterns were visualized using the dimensionality reduction
algorithm Uniform Manifold Approximation and Projection
(UMAP). Distinct topographical clusters were apparent in each
group (Figures 1D, S2A, and S2B). In particular, memory CD8+
T cells from patients with acute moderate or severe COVID-19
expressed a distinct cluster of markers associated with activa-
tion and the cell cycle, including CD38, HLA-DR, Ki-67, and
PD-1 (Figures 1D and S2A). A similar pattern was observed
among memory CD4+
T cells from patients with acute moderate
or severe COVID-19 (Figure S2B). These findings were
confirmed via manual gating of the flow cytometry data (Fig-
ure 1E). Correlative analyses further demonstrated that the acti-
vated/cycling phenotype was strongly associated with SARS-
CoV-2-specific IgG levels and various clinical parameters,
including age, hemoglobin concentration, platelet count, and
plasma levels of alanine aminotransferase, albumin, D-dimer,
fibrinogen, and myoglobin (Figures S2C and S2D), but less
strongly associated with plasma levels of various inflammatory
markers, including interleukin (IL)-1b, IL-10, and tumor necrosis
factor (TNF) (Table S1).
Phenotypic Characteristics of SARS-CoV-2-Specific T
Cells in Acute and Convalescent COVID-19
Unphysiologically high expression frequencies of CD38, poten-
tially driven by a highly inflammatory environment, were
defined segregation of memory CD4+
and CD8+
T cells. Bottom: dot plots showing the group-defined distribution of markers in PC2. Each dot represents one
donor. MC, individuals in the convalescent phase after mild COVID-19. **p < 0.01, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple
comparisons.
(C) Dot plots summarizing the expression frequencies of activation/cycling markers among memory CD4+
(top) and CD8+
T cells (bottom) by group. Each dot
represents one donor. Data are shown as median ± IQR. *p < 0.05, **p < 0.01, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple
comparisons.
(D) Top: UMAP plots showing the clustering of memory CD8+
T cells by group in relation to all memory CD8+
T cells (left). Bottom: UMAP plots showing the
expression of individual markers (n = 3 donors per group). UMAP plots were based on all markers distinguished in the bottom row.
(E) Left: representative flow cytometry plots showing the expression of activation/cycling markers among memory CD8+
T cells by group. Numbers indicate
percentages in the drawn gates. Right: dot plots showing the expression frequencies of activation/cycling markers among memory CD8+
T cells by group. Each
dot represents one donor. Data are shown as median ± IQR. Key as in (B) and (C). **p < 0.01, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test
for multiple comparisons.
ll
OPEN ACCESS
160 Cell 183, 158–168, October 1, 2020
Article
observed consistently among memory CD8+
T cells from pa-
tients with acute moderate or severe COVID-19 (Figures S3A
and S3B). In line with these data, we found that CD8+
T cells spe-
cific for cytomegalovirus (CMV) or Epstein-Barr virus (EBV) more
commonly expressed CD38, but not HLA-DR, Ki-67, or PD-1, in
patients with acute moderate or severe COVID-19 compared
with convalescent individuals and healthy blood donors, indi-
cating limited bystander activation and proliferation during the
early phase of infection with SARS-CoV-2 (Figures 2A, 2B, and
S3C). Of note, actively proliferating CD8+
T cells, defined by
expression of Ki-67, exhibited a predominant CCR7 CD27+
CD28+
CD45RA CD127 phenotype in patients with acute
moderate or severe COVID-19 (Figure S3D), as reported previ-
ously in the context of vaccination and other viral infections (Bug-
gert et al., 2018; Miller et al., 2008). On the basis of these find-
ings, we used overlapping peptides spanning the immunogenic
domains of the SARS-CoV-2 spike, membrane, and nucleo-
capsid proteins to stimulate peripheral blood mononuclear cells
(PBMCs) from patients with acute moderate or severe COVID-
19. A vast majority of responding CD4+
and CD8+
T cells dis-
played an activated/cycling (CD38+
HLA-DR+
Ki67+
PD-1+
)
phenotype (Figure 2C). These results were confirmed using an
activation-induced marker (AIM) assay to measure upregulation
of CD69 and 4-1BB (CD137), suggesting that most CD38+
PD-1+
CD8+
T cells were specific for SARS-CoV-2 (Figure 2D).
In further experiments, we used HLA class I tetramers as
probes to detect CD8+
T cells specific for the predicted optimal
epitopes from SARS-CoV-2 (Figure S3E; Table S2). A vast major-
ity of tetramer+
CD8+
T cells in the acute phase of infection, but
not during convalescence, displayed an activated/cycling
phenotype (Figure 2E). In general, early SARS-CoV-2-specific
CD8+
T cell populations were characterized by expression of im-
mune activation molecules (CD38, HLA-DR, and Ki-67), inhibi-
tory receptors (PD-1 and TIM-3), and cytotoxic molecules
(granzyme B and perforin), whereas convalescent-phase
SARS-CoV-2-specific CD8+
T cell populations were skewed to-
ward an early differentiated memory (CCR7+
CD127+
CD45RA /+
TCF1+
) phenotype (Figure 2F). Importantly, the expression fre-
quencies of CCR7 and CD45RA among SARS-CoV-2-specific
CD8+
T cells were positively correlated with the number of symp-
tom-free days after infection (CCR7: r = 0.79, p = 0.001; CD45RA:
r = 0.70, p = 0.008), whereas the expression frequency of gran-
zyme B among SARS-CoV-2-specific CD8+
T cells was inversely
correlated with the number of symptom-free days after infection
(r = 0.70, p = 0.007) (Figure 2G). Time from exposure was therefore
associated with emergence of stem-like memory SARS-CoV-2-
specific CD8+
T cells.
Functional Characteristics of SARS-CoV-2-Specific T
Cells in Convalescent COVID-19
On the basis of these observations, we quantified functional
SARS-CoV-2-specific memory T cell responses across five
distinct cohorts, including healthy individuals who donated
blood before or during the pandemic, family members who
shared a household with convalescent individuals and were
exposed at the time of symptomatic disease, and individuals in
the convalescent phase after mild or severe COVID-19. We de-
tected potentially cross-reactive T cell responses directed
against the spike and/or membrane proteins in 28% of healthy
individuals who donated blood before the pandemic, consistent
with previous reports (Grifoni et al., 2020; Le Bert et al., 2020),
but nucleocapsid reactivity was notably absent in this cohort
(Figures 3A, S4A, and S4B). The highest response frequencies
against any of these three proteins were observed in convales-
cent individuals who experienced severe COVID-19 (100%). Pro-
gressively lower response frequencies were observed in conva-
lescent individuals with a history of mild COVID-19 (87%),
exposed family members (67%), and healthy individuals who
donated blood during the pandemic (46%) (Figure 3A).
To assess the functional capabilities of SARS-CoV-2-specific
memory CD4+
and CD8+
T cells in convalescent individuals, we
stimulated PBMCs with the overlapping spike, membrane, and
nucleocapsid peptide sets and measured a surrogate marker
of degranulation (CD107a) along with production of interferon
(IFN)-g, IL-2, and TNF (Figures 3B and 3C). SARS-CoV-2-spe-
cific CD4+
T cells predominantly expressed IFN-g, IL-2, and
TNF (Figure 3B), whereas SARS-CoV-2-specific CD8+
T cells
predominantly expressed IFN-g and mobilized CD107a (Fig-
ure 3C). We then used the AIM assay to determine the functional
polarization of SARS-CoV-2-specific CD4+
T cells. Interestingly,
spike-specific CD4+
T cells were skewed toward a circulating T
follicular helper (cTfh) profile, suggesting a key role in the gener-
ation of potent antibody responses, whereas membrane-spe-
cific and nucleocapsid-specific CD4+
T cells were skewed
toward a Th1 or a Th1/Th17 profile (Figures 3D, S5A, and S5B).
Antibody Responses and Proliferative Capabilities of
SARS-CoV-2-Specific T Cells in Convalescent COVID-19
In a final series of experiments, we assessed the recall capabil-
ities of SARS-CoV-2-specific CD4+
and CD8+
T cells in conva-
lescent individuals, exposed family members, and healthy blood
donors. Proliferative responses were identified by tracking the
progressive dilution of a cytoplasmic dye (CellTrace Violet
[CTV]) after stimulation with the overlapping spike, membrane,
and nucleocapsid peptide sets, and functional responses to
the same antigens were evaluated 5 days later by measuring
the production of IFN-g (Blom et al., 2013; Buggert et al.,
2014). Anamnestic responses in the CD4+
and CD8+
T cell com-
partments, quantified as a function of CTVlow
IFN-g+
events (Fig-
ure 4A), were detected in most convalescent individuals (mild
COVID-19 [MC] = 96%, severe COVID-19 [SC] = 100%) and
exposed family members (92%) (Figures 4B and 4C). SARS-
CoV-2-specific CD4+
T cell responses were proportionately
larger overall than the corresponding SARS-CoV-2-specific
CD8+
T cell responses (exposed family members = 1.8-fold,
MC = 1.4-fold, SC = 1.8-fold) (Figure 4D). In addition, most
IFN-g+
SARS-CoV-2-specific CD4+
T cells produced TNF, and
most IFN-g+
SARS-CoV-2-specific CD8+
T cells expressed
granzyme B and perforin (Figure 4E).
Serological evaluations revealed a strong positive correlation
between IgG responses directed against the spike protein of
SARS-CoV-2 and IgG responses directed against the nucleo-
capsid protein of SARS-CoV-2 (r = 0.82, p < 0.001) (Figure S5C).
Moreover, SARS-CoV-2-specific CD4+
and CD8+
T cell re-
sponses were present in seronegative individuals, albeit at lower
frequencies compared with seropositive individuals (41% versus
ll
OPEN ACCESS
Cell 183, 158–168, October 1, 2020 161
Article
A
C
E
F G
D
B
Figure 2. Phenotypic Characteristics of SARS-CoV-2-Specific T Cells in Acute and Convalescent COVID-19
(A and B) Dot plots summarizing the expression frequencies of activation/cycling markers among tetramer+
CMV-specific (A) or EBV-specific CD8+
T cells (B) by
group. Each dot represents one specificity in one donor. Data are shown as median ± IQR. *p < 0.05, **p < 0.01. Kruskal-Wallis rank-sum test with Dunn’s post
hoc test for multiple comparisons.
(C) Representative flow cytometry plots (left) and bar graphs (right) showing the expression of activation/cycling markers among CD107a+
and/or IFN-g+
SARS-
CoV-2-specific CD4+
and CD8+
T cells (n = 6 donors). Numbers indicate percentages in the drawn gates. Data are shown as median ± IQR. NC, negative control.
*p < 0.05, **p < 0.01. Paired t test or Wilcoxon signed-rank test.
(legend continued on next page)
ll
OPEN ACCESS
162 Cell 183, 158–168, October 1, 2020
Article
99%, respectively) (Figure 4F). These discordant responses
were nonetheless pronounced in some convalescent individuals
with a history of mild COVID-19 (3 of 31), exposed family mem-
bers (9 of 28), and healthy individuals who donated blood during
the pandemic (5 of 31) (Figures 4F and S5D), often targeting the
internal (nucleocapsid) and surface antigens (spike and/or mem-
brane) of SARS-CoV-2 (Figure 4G). Higher frequencies of SARS-
CoV-2-specific T cells were also found in exposed seronegative
family members compared with unexposed donors (Figure S5E).
Potent memory T cell responses were therefore elicited in the
absence or presence of circulating antibodies, consistent with
a non-redundant role as key determinants of immune protection
against COVID-19 (Chandrashekar et al., 2020).
DISCUSSION
We are currently facing the biggest global health emergency in
decades, namely the devastating outbreak of COVID-19. In the
absence of a protective vaccine, it will be critical to determine
whether exposed and/or infected people, especially those with
asymptomatic or very mild forms of the disease who likely act
inadvertently as major transmitters, develop robust adaptive im-
munity against SARS-CoV-2 (Long et al., 2020).
In this study, we used a systematic approach to map cellular
and humoral immune responses against SARS-CoV-2 in patients
with acute moderate or severe COVID-19, individuals in the
convalescent phase after mild or severe COVID-19, exposed
family members, and healthy individuals who donated blood
before (2019) or during the pandemic (2020). Individuals in the
convalescent phase after mild COVID-19 were traced after re-
turning to Sweden from endemic areas (mostly northern Italy).
These donors exhibited robust memory T cell responses months
after infection, even in the absence of detectable circulating an-
tibodies specific for SARS-CoV-2, that may contribute to protec-
tion against severe COVID-19.
We found that T cell activation, characterized by expression of
CD38, was a hallmark of acute COVID-19. Similar findings have
been reported previously in the absence of specificity data
(Huang et al., 2020; Thevarajan et al., 2020; Wilk et al., 2020).
Many of these T cells also expressed HLA-DR, Ki-67, and
PD-1, indicating a combined activation/cycling phenotype,
which correlated with early SARS-CoV-2-specific IgG levels
and, to a lesser extent, plasma levels of various inflammatory
markers. Our data also showed that many activated/cycling
T cells in the acute phase were functionally replete and specific
for SARS-CoV-2. Equivalent functional profiles have been
observed early after immunization with successful vaccines
(Blom et al., 2013; Miller et al., 2008; Precopio et al., 2007).
Accordingly, the expression of multiple inhibitory receptors,
including PD-1, likely indicates early activation rather than
exhaustion (Zheng et al., 2020a, 2020b).
Virus-specific memory T cells have been shown to persist for
many years after infection with SARS-CoV-1 (Le Bert et al.,
2020; Tang et al., 2011; Yang et al., 2006). In line with these ob-
servations, we found that SARS-CoV-2-specific T cells acquired
an early differentiated memory (CCR7+
CD127+
CD45RA /+
TCF1+
) phenotype in the convalescent phase, as reported previ-
ously in the context of other viral infections and successful vac-
cines (Blom et al., 2013; Demkowicz et al., 1996; Fuertes Mar-
raco et al., 2015; Precopio et al., 2007). This phenotype has
been associated with stem-like properties (Betts et al., 2006;
Blom et al., 2013; Demkowicz et al., 1996; Fuertes Marraco
et al., 2015; Precopio et al., 2007). Accordingly, we found that
SARS-CoV-2-specific T cells generated anamnestic responses
to cognate antigens in the convalescent phase, characterized
by extensive proliferation and polyfunctionality. Of particular
note, we detected similar memory T cell responses directed
against the internal (nucleocapsid) and surface proteins (mem-
brane and/or spike) in some individuals lacking detectable circu-
lating antibodies specific for SARS-CoV-2. Indeed, almost twice
as many healthy individuals who donated blood during the
pandemic had memory T cell responses versus antibody re-
sponses, implying that seroprevalence as an indicator may un-
derestimate the extent of adaptive immune responses against
SARS-CoV-2.
Our study was cross-sectional in nature and limited in terms of
clinical follow-up and overall donor numbers in each outcome-
defined group. It therefore remains to be determined whether
robust memory T cell responses in the absence of detectable
circulating antibodies can protect against severe forms of
COVID-19. This scenario has nonetheless been inferred from
previous studies of MERS and SARS-CoV-1 (Channappanavar
et al., 2014; Li et al., 2008; Zhao et al., 2016, 2017), both of which
have been shown to induce potent memory T cell responses that
persist for years, in contrast to the corresponding antibody re-
sponses (Alshukairi et al., 2016; Shin et al., 2019; Tang et al.,
2011). Antibody responses have also been shown to wane after
infection with SARS-CoV-2 (Ibarrondo et al., 2020; Long et al.,
2020), suggesting that transient humoral immunity is a general
feature of coronavirus infections (Callow et al., 1990). However,
the fact that memory B cells (Juno et al., 2020) and memory T
cells are generated in response to SARS-CoV-2 suggests that
natural infection may elicit protection from severe COVID-19.
In line with these observations, none of the convalescent individ-
uals in this study, including those with previous mild disease,
experienced further episodes of COVID-19.
(D) Representative flow cytometry plots (left) and bar graph (right) showing the upregulation of CD69 and 4-1BB (AIM assay) among CD38+
PD-1+
SARS-CoV-2-
specific CD8+
T cells (n = 6 donors). Numbers indicate percentages in the drawn gates. S, spike; M, membrane; N, nucleocapsid.
(E) Left: representative flow cytometry plots showing the expression of activation/cycling markers among tetramer+
SARS-CoV-2-specific CD8+
T cells by group
(red) and by total frequency (black). Center: UMAP plot showing the clustering of memory CD8+
T cells. Right: UMAP plots showing the clustering of tetramer+
SARS-CoV-2-specific CD8+
T cells by group and the expression of individual markers (n = 2 donors).
(F) Dot plots summarizing the expression frequencies of all quantified markers among tetramer+
SARS-CoV-2-specific CD8+
T cells by group. Each dot rep-
resents combined specificities in one donor. Data are shown as median ± IQR.
(G) Bivariate plots showing the pairwise correlations between symptom-free days and the expression frequencies of CCR7, CD45RA, or granzyme B (GzmB).
Each dot represents combined specificities in one donor. Key as in (F). Spearman rank correlation.
ll
OPEN ACCESS
Cell 183, 158–168, October 1, 2020 163
Article
A
B
C
D
Figure 3. Functional Characteristics of SARS-CoV-2-Specific T Cells in Convalescent COVID-19
(A) Left: dot plots summarizing the frequencies of IFN-g-producing cells responding to overlapping peptides spanning the immunogenic domains of the SARS-
CoV-2 S, M, and N proteins by group (ELISpot assays). Each dot represents one donor. The dotted line indicates the cutoff for positive responses. Right: bar
graph showing the frequencies of IFN-g-producing cells responding to the internal (N) and surface antigens (M and/or S) of SARS-CoV-2 by group (ELISpot
assays). 2019 BD, healthy blood donors from 2019; Exp, exposed family members; SC, individuals in the convalescent phase after severe COVID-19; SFU, spot-
forming unit. *p < 0.05, **p < 0.01, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons.
(B) and (C) Left: representative flow cytometry plots showing the functional profiles of SARS-CoV-2-specific CD4+
(B) and CD8+
T cells (C) from a convalescent
individual (group MC). Numbers indicate percentages in the drawn gates. Right: bar graphs and pie charts summarizing the distribution of individual functions
among SARS-CoV-2-specific CD4+
(B) and CD8+
T cells (C) from convalescent individuals in groups MC (n = 12) or SC (n = 14). Data are shown as median ± IQR.
Key as in (A). *p < 0.05. Unpaired t test or Mann-Whitney U test.
(D) Left: bar graphs summarizing the functional polarization of SARS-CoV-2-specific CD4+
T cells from convalescent individuals in groups MC (n = 5) and SC (n =
6). Subsets were defined as CXCR5+
(cTfh), CCR4 CCR6 CXCR3+
CXCR5 (Th1), CCR4+
CCR6 CXCR3 CXCR5 (Th2), CCR4 CCR6+
CXCR3 CXCR5
(Th17), CCR4 CCR6+
CXCR3+
CXCR5 (Th1/17), and CCR4 CCR6 CXCR3 CXCR5 (non-Th1/2/17). Data are shown as median ± IQR. *p < 0.05. Unpaired t
test or Mann-Whitney U test. Right: line graph comparing cTfh versus Th1 polarization by specificity in convalescent individuals from groups MC and SC. Each dot
represents one donor. Key as in (A). *p < 0.05, **p < 0.01. Paired t test.
ll
OPEN ACCESS
164 Cell 183, 158–168, October 1, 2020
Article
A B
C
D
E
F G
(legend on next page)
ll
OPEN ACCESS
Cell 183, 158–168, October 1, 2020 165
Article
Of note, we detected cross-reactive T cell responses directed
against the spike and/or membrane proteins of SARS-CoV-2 in
28% of unexposed healthy blood donors, consistent with a
high degree of pre-existing immunity in the general population
(Braun et al., 2020; Grifoni et al., 2020; Le Bert et al., 2020). More-
over, these particular data were derived from cryopreserved
samples, so this figure might be considered as a lower bound es-
timate of overall prevalence (Owen et al., 2007). In this context,
our findings most likely reflect widespread exposure to seasonal
coronaviruses, which could shape the subsequent immune
response to SARS-CoV-2. As such, it remains likely that a frac-
tion of the anamnestic SARS-CoV-2-specific T cell response
was initially induced by seasonal coronaviruses in seronegative
individuals (Mateus et al., 2020). It is also tempting to speculate
that such responses may provide at least partial protection
against SARS-CoV-2, given that pre-existing T cell immunity
has been associated with beneficial outcomes after challenge
with the pandemic influenza virus strain H1N1 (Sridhar et al.,
2013; Wilkinson et al., 2012).
Collectively, our data provide a functional and phenotypic map
of SARS-CoV-2-specific T cell immunity across the full spectrum
of exposure, infection, and disease. The observation that many
individuals with asymptomatic or mild COVID-19 had highly du-
rable and functionally replete memory T cell responses, not un-
commonly in the absence of detectable humoral responses,
further suggests that natural exposure or infection could prevent
recurrent episodes of severe COVID-19.
STAR+METHODS
Detailed methods are provided in the online version of this paper
and include the following:
d KEY RESOURCES TABLE
d RESOURCE AVAILABILITY
B Lead Contact
B Material Availability
B Data and Code Availability
d EXPERIMENTAL MODEL AND SUBJECT DETAILS
B Human subjects
d METHOD DETAILS
B Flow cytometry
B Peptides
B Epitope prediction
B Tetramers
B Functional assay
B Proliferation assay
B AIM assay
B Trucount
B Principal component analysis
B UMAP
B ELISpot assay
B Serology
d QUANTIFICATION AND STATISTICAL ANALYSIS
SUPPLEMENTAL INFORMATION
Supplemental Information can be found online at https://doi.org/10.1016/j.
cell.2020.08.017.
CONSORTIA
The members of the Karolinska COVID-19 Study Group are Mira Akber, Soo
Aleman, Lena Berglin, Helena Bergsten, Niklas K. Björkström, Susanna Brigh-
enti, Demi Brownlie, Marcus Buggert, Marta Butrym, Benedict Chambers,
Puran Chen, Martin Cornillet Jeannin, Jonathan Grip, Angelica Cuapio Gomez,
Lena Dillner, Jean-Baptiste Gorin, Isabel Diaz Lozano, Majda Dzidic, Johanna
Emgård, Lars I. Eriksson, Malin Flodström Tullberg, Anna Färnert, Hedvig
Glans, Sara Gredmark Russ, Alvaro Haroun-Izquierdo, Elizabeth Henriksson,
Laura Hertwig, Habiba Kamal, Tobias Kamann, Jonas Klingstrom, Efthymia
Kokkinou, Egle Kvedaraite, Hans-Gustaf Ljunggren, Sian Llewellyn-Lacey,
Marco Loreti, Magalini Lourda, Kimia Maleki, Karl-Johan Malmberg, Nicole
Marquardt, Christopher Maucourant, Jakob Michaelsson, Jenny Mjösberg,
Kirsten Moll, Jagadees Muva, Johan Mårtensson, Pontus Nauclér, Anna
Norrby-Teglund, Annika Olsson, Laura Palma Medina, Tiphaine Parrot, Björn
Persson, André Perez-Potti, Lena Radler, Emma Ringqvist, Olga Rivera-Bal-
lesteros, Olav Rooyackers, Johan Sandberg, John Tyler Sandberg, Takuya
Sekine, Ebba Sohlberg, Tea Soini, Kristoffer Strålin, Anders Sönnerborg, Mat-
tias Svensson, Janne Tynell, Renata Varnaite, Andreas Von Kries, Christian
Unge, and David Wulliman.
ACKNOWLEDGMENTS
We express our gratitude to all donors, health care personnel, study coordina-
tors, administrators, and laboratory managers involved in this work. M.B. was
supported by the Swedish Research Council, the Karolinska Institutet, the
Swedish Society for Medical Research, the Jeansson Stiftelser, the Åke Wi-
bergs Stiftelse, the Swedish Society of Medicine, the Swedish Cancer Society,
Figure 4. Antibody Responses and Proliferation Capabilities of SARS-CoV-2-Specific T Cells in Convalescent COVID-19
(A) Representative flow cytometry plots showing the proliferation (CTV ) and functionality (IFN-g+
) of SARS-CoV-2-specific T cells from a convalescent individual
(group MC) after stimulation with overlapping peptides spanning the immunogenic domains of the SARS-CoV-2 S, M, and N proteins. Numbers indicate per-
centages in the drawn gates.
(B) and (C) Dot plots summarizing the frequencies of CTV IFN-g+
SARS-CoV-2-specific CD4+
(B) and CD8+
T cells (C) by group and specificity. Each dot
represents one donor. The dotted line indicates the cutoff for positive responses. *p < 0.05, **p < 0.01, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post
hoc test for multiple comparisons.
(D) Dot plots comparing the frequencies of CTV IFN-g+
SARS-CoV-2-specific CD4+
versus CD8+
T cells by group and specificity. Each dot represents one
donor. Data are shown as median ± IQR. Key as in (B) and (C). *p < 0.05, **p < 0.01, ***p < 0.001. Paired t test or Wilcoxon signed-rank test.
(E) Left: representative flow cytometry plots showing the production of IFN-g and TNF among CTV virus-specific CD4+
(top) and CD8+
T cells (bottom) from a
convalescent individual (group MC). Numbers indicate percentages in the drawn gates. Right: heatmaps summarizing the functional profiles of CTV IFN-g+
virus-specific CD4+
(top) and CD8+
T cells (bottom). Data are shown as mean frequencies (key).
(F) Dot plots summarizing the frequencies of CTV IFN-g+
SARS-CoV-2-specific CD4+
and CD8+
T cells by group, serostatus, and specificity. Each dot rep-
resents one donor. The dotted line indicates the cutoff for positive responses. Key as in (B) and (C). ***p < 0.001. Mann-Whitney U test.
(G) Left: bar graph summarizing percent seropositivity by group. Right: bar graph summarizing the percentage of individuals in each group with detectable T cell
responses directed against the internal (N) and surface antigens (M and/or S) of SARS-CoV-2.
ll
OPEN ACCESS
166 Cell 183, 158–168, October 1, 2020
Article
the Swedish Childhood Cancer Fund, the Magnus Bergvalls Stiftelse, the Hed-
lunds Stiftelse, the Lars Hiertas Stiftelse, the Swedish Physicians against AIDS
Foundation, the Jonas Söderquist Stiftelse, and the Clas Groschinskys Min-
nesfond. H.-G.L. and the Karolinska COVID-19 Study Group were supported
by the Alice och Knut Wallenbergs Stiftelse and Nordstjernan AB. D.A.P.
was supported by a Wellcome Trust Senior Investigator Award (100326/Z/
12/Z).
AUTHOR CONTRIBUTIONS
H.-G.L., S.A., and M.B. conceived the project. T.S., A.P.-P., O.R.-B., J.-B.G.,
S.L.-L., S.M., D.J.W., T.K., J.E., T.P., D.A.P., and M.B. designed and per-
formed experiments. T.S., A.P.-P., O.R.-B., J.-B.G., and M.B. analyzed data.
K.S., A.O., H.K., G.B., E.F., O.R., L.I.E., J.-I.H., A.S., T.A., J.A., M.N., J.K.,
S.G.-R., N.K.B., J.K.S., D.A.P., H.-G.L., S.A., and M.B. provided critical re-
sources. D.A.P. and M.B. supervised experiments. T.S., A.P.-P., O.R.-B.,
and M.B. drafted the manuscript. D.A.P. and M.B. edited the manuscript. All
authors contributed intellectually and approved the manuscript.
DECLARATION OF INTERESTS
The authors declare no competing interests.
Received: June 26, 2020
Revised: July 29, 2020
Accepted: August 11, 2020
Published: August 14, 2020
REFERENCES
Alshukairi, A.N., Khalid, I., Ahmed, W.A., Dada, A.M., Bayumi, D.T., Malic, L.S.,
Althawadi, S., Ignacio, K., Alsalmi, H.S., Al-Abdely, H.M., et al. (2016). Anti-
body Response and Disease Severity in Healthcare Worker MERS Survivors.
Emerg. Infect. Dis. 22, 1113–1115.
Beigel, J.H., Tomashek, K.M., Dodd, L.E., Mehta, A.K., Zingman, B.S., Kalil,
A.C., Hohmann, E., Chu, H.Y., Luetkemeyer, A., Kline, S., et al. (2020). Remde-
sivir for the Treatment of Covid-19— Preliminary Report. N Engl J Med. Pub-
lished online May 22, 2020. https://doi.org/10.1056/NEJMoa2007764.
Betts, M.R., Nason, M.C., West, S.M., De Rosa, S.C., Migueles, S.A.,
Abraham, J., Lederman, M.M., Benito, J.M., Goepfert, P.A., Connors, M.,
et al. (2006). HIV nonprogressors preferentially maintain highly functional
HIV-specific CD8+
T cells. Blood 107, 4781–4789.
Blom, K., Braun, M., Ivarsson, M.A., Gonzalez, V.D., Falconer, K., Moll, M.,
Ljunggren, H.G., Michaëlsson, J., and Sandberg, J.K. (2013). Temporal dy-
namics of the primary human T cell response to yellow fever virus 17D as it ma-
tures from an effector- to a memory-type response. J. Immunol. 190,
2150–2158.
Braun, J., Loyal, L., Frentsch, M., Wendisch, D., Georg, P., Kurth, F., Hippen-
stiel, S., Dingeldey, M., Kruse, B., Fauchere, F., et al. (2020). SARS-CoV-2-
reactive T cells in healthy donors and patients with COVID-19. Nature. Pub-
lished online July 29, 2020. https://doi.org/10.1038/s41586-020-2598-9.
Buggert, M., Norström, M.M., Salemi, M., Hecht, F.M., and Karlsson, A.C.
(2014). Functional avidity and IL-2/perforin production is linked to the emer-
gence of mutations within HLA-B*5701-restricted epitopes and HIV-1 disease
progression. J. Immunol. 192, 4685–4696.
Buggert, M., Nguyen, S., Salgado-Montes de Oca, G., Bengsch, B., Darko, S.,
Ransier, A., Roberts, E.R., Del Alcazar, D., Brody, I.B., Vella, L.A., et al. (2018).
Identification and characterization of HIV-specific resident memory CD8+
T cells in human lymphoid tissue. Sci. Immunol. 3, eaar4526.
Callow, K.A., Parry, H.F., Sergeant, M., and Tyrrell, D.A. (1990). The time
course of the immune response to experimental coronavirus infection of
man. Epidemiol. Infect. 105, 435–446.
Chandrashekar, A., Liu, J., Martinot, A.J., McMahan, K., Mercado, N.B., Peter,
L., Tostanoski, L.H., Yu, J., Maliga, Z., Nekorchuk, M., et al. (2020). SARS-
CoV-2 infection protects against rechallenge in rhesus macaques. Science
369, 812–817.
Channappanavar, R., Fett, C., Zhao, J., Meyerholz, D.K., and Perlman, S.
(2014). Virus-specific memory CD8 T cells provide substantial protection
from lethal severe acute respiratory syndrome coronavirus infection. J. Virol.
88, 11034–11044.
Demkowicz, W.E., Jr., Littaua, R.A., Wang, J., and Ennis, F.A. (1996). Human
cytotoxic T-cell memory: long-lived responses to vaccinia virus. J. Virol. 70,
2627–2631.
Fuertes Marraco, S.A., Soneson, C., Cagnon, L., Gannon, P.O., Allard, M.,
Abed Maillard, S., Montandon, N., Rufer, N., Waldvogel, S., Delorenzi, M.,
and Speiser, D.E. (2015). Long-lasting stem cell-like memory CD8+
T cells
with a naı̈ve-like profile upon yellow fever vaccination. Sci. Transl. Med. 7,
282ra48.
Grifoni, A., Weiskopf, D., Ramirez, S.I., Mateus, J., Dan, J.M., Moderbacher,
C.R., Rawlings, S.A., Sutherland, A., Premkumar, L., Jadi, R.S., et al. (2020).
Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with
COVID-19 Disease and Unexposed Individuals. Cell 181, 1489–1501.e15.
Guan, W.J., Ni, Z.Y., Hu, Y., Liang, W.H., Ou, C.Q., He, J.X., Liu, L., Shan, H.,
Lei, C.L., Hui, D.S.C., et al.; China Medical Treatment Expert Group for Covid-
19 (2020). Clinical Characteristics of Coronavirus Disease 2019 in China.
N. Engl. J. Med. 382, 1708–1720.
Habib, H. (2020). Has Sweden’s controversial covid-19 strategy been suc-
cessful? BMJ 369, m2376.
He, R., Lu, Z., Zhang, L., Fan, T., Xiong, R., Shen, X., Feng, H., Meng, H., Lin,
W., Jiang, W., and Geng, Q. (2020a). The clinical course and its correlated im-
mune status in COVID-19 pneumonia. J. Clin. Virol. 127, 104361.
He, X., Lau, E.H.Y., Wu, P., Deng, X., Wang, J., Hao, X., Lau, Y.C., Wong, J.Y.,
Guan, Y., Tan, X., et al. (2020b). Temporal dynamics in viral shedding and
transmissibility of COVID-19. Nat. Med. 26, 672–675.
Hotez, P.J., Corry, D.B., Strych, U., and Bottazzi, M.E. (2020). COVID-19 vac-
cines: neutralizing antibodies and the alum advantage. Nat. Rev. Immunol. 20,
399–400.
Huang, C., Wang, Y., Li, X., Ren, L., Zhao, J., Hu, Y., Zhang, L., Fan, G., Xu, J.,
Gu, X., et al. (2020). Clinical features of patients infected with 2019 novel coro-
navirus in Wuhan, China. Lancet 395, 497–506.
Ibarrondo, F.J., Fulcher, J.A., Goodman-Meza, D., Elliott, J., Hofmann, C.,
Hausner, M.A., Ferbas, K.G., Tobin, N.H., Aldrovandi, G.M., and Yang, O.O.
(2020). Rapid Decay of Anti-SARS-CoV-2 Antibodies in Persons with Mild
Covid-19. N. Engl. J. Med. Published online July 21, 2020. https://doi.org/
10.1056/NEJMc2025179.
Juno, J.A., Tan, H.X., Lee, W.S., Reynaldi, A., Kelly, H.G., Wragg, K., Esterba-
uer, R., Kent, H.E., Batten, C.J., Mordant, F.L., et al. (2020). Humoral and circu-
lating follicular helper T cell responses in recovered patients with COVID-19.
Nat. Med. Published online July 13, 2020. https://doi.org/10.1038/s41591-
020-0995-0.
Kirkcaldy, R.D., King, B.A., and Brooks, J.T. (2020). COVID-19 and Postinfec-
tion Immunity: Limited Evidence, Many Remaining Questions. JAMA. Pub-
lished online May 11, 2020. https://doi.org/10.1001/jama.2020.7869.
Le Bert, N., Tan, A.T., Kunasegaran, K., Tham, C.Y.L., Hafezi, M., Chia, A.,
Chng, M.H.Y., Lin, M., Tan, N., Linster, M., et al. (2020). SARS-CoV-2-specific
T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Na-
ture. Published online July 15, 2020. https://doi.org/10.1038/s41586-020-
2550-z.
Li, C.K., Wu, H., Yan, H., Ma, S., Wang, L., Zhang, M., Tang, X., Temperton,
N.J., Weiss, R.A., Brenchley, J.M., et al. (2008). T cell responses to whole
SARS coronavirus in humans. J. Immunol. 181, 5490–5500.
Liu, J., Li, S., Liu, J., Liang, B., Wang, X., Wang, H., Li, W., Tong, Q., Yi, J.,
Zhao, L., et al. (2020). Longitudinal characteristics of lymphocyte responses
and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients.
EBioMedicine 55, 102763.
ll
OPEN ACCESS
Cell 183, 158–168, October 1, 2020 167
Article
Long, Q.X., Tang, X.J., Shi, Q.L., Li, Q., Deng, H.J., Yuan, J., Hu, J.L., Xu, W.,
Zhang, Y., Lv, F.J., et al. (2020). Clinical and immunological assessment of
asymptomatic SARS-CoV-2 infections. Nat. Med. 26, 1200–1204.
Mallapaty, S. (2020). Will antibody tests for the coronavirus really change
everything? Nature 580, 571–572.
Mateus, J., Grifoni, A., Tarke, A., Sidney, J., Ramirez, S.I., Dan, J.M., Burger,
Z.C., Rawlings, S.A., Smith, D.M., Phillips, E., et al. (2020). Selective and
cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science.
Published online August 4, 2020. https://doi.org/10.1126/science.abd3871.
Miller, J.D., van der Most, R.G., Akondy, R.S., Glidewell, J.T., Albott, S., Maso-
pust, D., Murali-Krishna, K., Mahar, P.L., Edupuganti, S., Lalor, S., et al. (2008).
Human effector and memory CD8+
T cell responses to smallpox and yellow fe-
ver vaccines. Immunity 28, 710–722.
Ni, L., Ye, F., Cheng, M.L., Feng, Y., Deng, Y.Q., Zhao, H., Wei, P., Ge, J., Gou,
M., Li, X., et al. (2020). Detection of SARS-CoV-2-Specific Humoral and
Cellular Immunity in COVID-19 Convalescent Individuals. Immunity 52,
971–977.e3.
Owen, R.E., Sinclair, E., Emu, B., Heitman, J.W., Hirschkorn, D.F., Epling, C.L.,
Tan, Q.X., Custer, B., Harris, J.M., Jacobson, M.A., et al. (2007). Loss of T cell
responses following long-term cryopreservation. J. Immunol. Methods
326, 93–115.
Plebani, M., Padoan, A., Negrini, D., Carpinteri, B., and Sciacovelli, L. (2020).
Diagnostic performances and thresholds: The key to harmonization in serolog-
ical SARS-CoV-2 assays? Clin. Chim. Acta 509, 1–7.
Precopio, M.L., Betts, M.R., Parrino, J., Price, D.A., Gostick, E., Ambrozak,
D.R., Asher, T.E., Douek, D.C., Harari, A., Pantaleo, G., et al. (2007). Immuni-
zation with vaccinia virus induces polyfunctional and phenotypically distinctive
CD8+
T cell responses. J. Exp. Med. 204, 1405–1416.
Price, D.A., Brenchley, J.M., Ruff, L.E., Betts, M.R., Hill, B.J., Roederer, M.,
Koup, R.A., Migueles, S.A., Gostick, E., Wooldridge, L., et al. (2005). Avidity
for antigen shapes clonal dominance in CD8+
T cell populations specific for
persistent DNA viruses. J Exp Med. 202, 1349–1361.
Reynisson, B., Alvarez, B., Paul, S., Peters, B., and Nielsen, M. (2020).
NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen
presentation by concurrent motif deconvolution and integration of MS MHC
eluted ligand data. Nucleic Acids Res. 48, W449–W454.
Robbiani, D.F., Gaebler, C., Muecksch, F., Lorenzi, J.C.C., Wang, Z., Cho, A.,
Agudelo, M., Barnes, C.O., Gazumyan, A., Finkin, S., et al. (2020). Convergent
Antibody Responses to SARS-CoV-2 Infection in Convalescent Individuals.
bioRxiv. https://doi.org/10.1101/2020.05.13.092619.
Seydoux, E., Homad, L.J., MacCamy, A.J., Parks, K.R., Hurlburt, N.K., Jenne-
wein, M.F., Akins, N.R., Stuart, A.B., Wan, Y.H., Feng, J., et al. (2020). Charac-
terization of neutralizing antibodies from a SARS-CoV-2 infected individual.
bioRxiv. https://doi.org/10.1101/2020.05.12.091298.
Shin, H.S., Kim, Y., Kim, G., Lee, J.Y., Jeong, I., Joh, J.S., Kim, H., Chang, E.,
Sim, S.Y., Park, J.S., and Lim, D.G. (2019). Immune Responses to Middle East
Respiratory Syndrome Coronavirus During the Acute and Convalescent
Phases of Human Infection. Clin. Infect. Dis. 68, 984–992.
Singer, M., Deustchman, C.S., Seymour, C.W., Shankar-Hari, M., Annane, D.,
Bauer, M., Bellomo, R., Bernard, G.R., Chiche, J.D., Coopersmith, C.M., et al.
(2016). The Third International Consensus Definitions for Sepsis and Septic
Shock (Sepsis-3). JAMA 315, 801–810.
Sridhar, S., Begom, S., Bermingham, A., Hoschler, K., Adamson, W., Carman,
W., Bean, T., Barclay, W., Deeks, J.J., and Lalvani, A. (2013). Cellular immune
correlates of protection against symptomatic pandemic influenza. Nat. Med.
19, 1305–1312.
Tang, F., Quan, Y., Xin, Z.T., Wrammert, J., Ma, M.J., Lv, H., Wang, T.B., Yang,
H., Richardus, J.H., Liu, W., and Cao, W.C. (2011). Lack of peripheral memory
B cell responses in recovered patients with severe acute respiratory syn-
drome: a six-year follow-up study. J. Immunol. 186, 7264–7268.
Thevarajan, I., Nguyen, T.H.O., Koutsakos, M., Druce, J., Caly, L., van de
Sandt, C.E., Jia, X., Nicholson, S., Catton, M., Cowie, B., et al. (2020). Breadth
of concomitant immune responses prior to patient recovery: a case report of
non-severe COVID-19. Nat. Med. 26, 453–455.
Wang, C., Li, W., Drabek, D., Okba, N.M.A., van Haperen, R., Osterhaus,
A.D.M.E., van Kuppeveld, F.J.M., Haagmans, B.L., Grosveld, F., and Bosch,
B.J. (2020). A human monoclonal antibody blocking SARS-CoV-2 infection.
Nat. Commun. 11, 2251.
Wei, W.E., Li, Z., Chiew, C.J., Yong, S.E., Toh, M.P., and Lee, V.J. (2020). Pre-
symptomatic Transmission of SARS-CoV-2 — Singapore, January 23-March
16, 2020. MMWR Morb. Mortal. Wkly. Rep. 69, 411–415.
Wilk, A.J., Rustagi, A., Zhao, N.Q., Roque, J., Martinez-Colon, G.J., McKech-
nie, J.L., Ivison, G.T., Ranganath, T., Vergara, R., Hollis, T., et al. (2020). A sin-
gle-cell atlas of the peripheral immune response in patients with severe
COVID-19. Nat. Med. 26, 1070–1076.
Wilkinson, T.M., Li, C.K., Chui, C.S., Huang, A.K., Perkins, M., Liebner, J.C.,
Lambkin-Williams, R., Gilbert, A., Oxford, J., Nicholas, B., et al. (2012). Preex-
isting influenza-specific CD4+
T cells correlate with disease protection against
influenza challenge in humans. Nat. Med. 18, 274–280.
Wolfel, R., Corman, V.M., Guggemos, W., Seilmaier, M., Zange, S., Muller,
M.A., Niemeyer, D., Jones, T.C., Vollmar, P., Rothe, C., et al. (2020). Virological
assessment of hospitalized patients with COVID-2019. Nature 581, 465–469.
Woloshin, S., Patel, N., and Kesselheim, A.S. (2020). False Negative Tests for
SARS-CoV-2 Infection — Challenges and Implications. N. Engl. J. Med.
383, e38.
Wu, Z., and McGoogan, J.M. (2020). Characteristics of and Important Lessons
From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary
of a Report of 72314 Cases From the Chinese Center for Disease Control and
Prevention. JAMA. Published online February 24, 2020. https://doi.org/10.
1001/jama.2020.2648.
Yang, L.T., Peng, H., Zhu, Z.L., Li, G., Huang, Z.T., Zhao, Z.X., Koup, R.A.,
Bailer, R.T., and Wu, C.Y. (2006). Long-lived effector/central memory T-cell re-
sponses to severe acute respiratory syndrome coronavirus (SARS-CoV) S an-
tigen in recovered SARS patients. Clin. Immunol. 120, 171–178.
Yang, R., Gui, X., and Xiong, Y. (2020). Comparison of Clinical Characteristics
of Patients with Asymptomatic vs Symptomatic Coronavirus Disease 2019 in
Wuhan, China. JAMA Netw. Open 3, e2010182.
Zhao, J., Zhao, J., Mangalam, A.K., Channappanavar, R., Fett, C., Meyerholz,
D.K., Agnihothram, S., Baric, R.S., David, C.S., and Perlman, S. (2016). Airway
Memory CD4+
T Cells Mediate Protective Immunity against Emerging Respira-
tory Coronaviruses. Immunity 44, 1379–1391.
Zhao, J., Alshukairi, A.N., Baharoon, S.A., Ahmed, W.A., Bokhari, A.A., Nehdi,
A.M., Layqah, L.A., Alghamdi, M.G., Al Gethamy, M.M., Dada, A.M., et al.
(2017). Recovery from the Middle East respiratory syndrome is associated
with antibody and T-cell responses. Sci. Immunol. 2, eaan5393.
Zheng, H.Y., Zhang, M., Yang, C.X., Zhang, N., Wang, X.C., Yang, X.P., Dong,
X.Q., and Zheng, Y.T. (2020a). Elevated exhaustion levels and reduced func-
tional diversity of T cells in peripheral blood may predict severe progression
in COVID-19 patients. Cell. Mol. Immunol. 17, 541–543.
Zheng, M., Gao, Y., Wang, G., Song, G., Liu, S., Sun, D., Xu, Y., and Tian, Z.
(2020b). Functional exhaustion of antiviral lymphocytes in COVID-19 patients.
Cell. Mol. Immunol. 17, 533–535.
ll
OPEN ACCESS
168 Cell 183, 158–168, October 1, 2020
Article
STAR+METHODS
KEY RESOURCES TABLE
REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
1G1 (PE) [anti-CCR4] BD Biosciences 551120 (RRID:AB_394054)
11A9 (PE-Cy7) [anti-CCR6] BD Biosciences 560620 (RRID:AB_1727440)
RPA-T8 or UCHT1 (BUV805) [anti-CD3] BD Biosciences 565515 (RRID:AB_2739277)
RPA-T8 (BUV395) [anti-CD8] BD Biosciences 563795 (RRID:AB_2722501)
M-A251 (PE-Cy5) [anti-CD25] BD Biosciences 560988 (RRID:AB_2033955)
CD28.2 (BUV563) [anti-CD28] BD Biosciences 564438 (RRID:AB_2738808)
HIT2 (BUV496) [anti-CD38] BD Biosciences 564657 (RRID:AB_2744376)
FN50 (BV750) [anti-CD69] BD Biosciences 563835 (RRID:AB_2738442)
FN50 (BUV737) [anti-CD69] BD Biosciences 564439 (RRID:AB_2722502)
DX2 (BB630) [anti-CD95] BD Biosciences Custom conjugate
H4A3 (PE-CF594) [anti-CD107a] BD Biosciences 562628 (RRID:AB_2737686)
BNI3 (BB755) [anti-CTLA-4] BD Biosciences 560938 (RRID:AB_2033942)
RF8B2 (APC-R700) [anti-CXCR5] BD Biosciences 565191 (RRID:AB_2739103)
GB11 (BB790) [anti-granzyme B] BD Biosciences Custom conjugate
G46-6 (BUV615) [anti-HLA-DR] BD Biosciences 565073 (RRID:AB_2722500)
MQ1-17H12 (APC-R700) [anti-IL-2] BD Biosciences 565136 (RRID:AB_2739079)
B56 (BB660) [anti-Ki-67] BD Biosciences Custom conjugate
T47-530 (BUV661) [anti-LAG-3] BD Biosciences Custom conjugate
dG9 (BB700) [anti-perforin] BD Biosciences Custom conjugate
741182 (BUV737) [anti-TIGIT] BD Biosciences Custom conjugate
C1.7 (PE/Dazzle 594) [anti-2B4] BioLegend 393506 (RRID:AB_2734464)
G043H7 (APC-Cy7) [anti-CCR7] BioLegend 353211 (RRID:AB_10915272)
M5E2 (BV510) [anti-CD14] BioLegend 301842 (RRID:AB_2561946)
HIB19 (BV510) [anti-CD19] BioLegend 302242 (RRID:AB_2561668)
O323 (BV785) [anti-CD27] BioLegend 302832 (RRID:AB_2562674)
A1 (BV711) [anti-CD39] BioLegend 328227 (RRID:AB_2632893)
HI100 (BV421) [anti-CD45RA] BioLegend 304129 (RRID:AB_10900421)
HI100 (BV570) [anti-CD45RA] BioLegend 304131 (RRID:AB_10897946)
A019D5 (BV605) [anti-CD127] BioLegend 351334 (RRID:AB_2562022)
G025H7 (AF647) [anti-CXCR3] BioLegend 353712 (RRID:AB_10962948)
4S.B3 (BV785) [anti-IFN-g] BioLegend 502542 (RRID:AB_2563882)
EH12.2H7 (PE-Cy7) [anti-PD-1] BioLegend 367415 (RRID:AB_2616743)
F38-2E2 (BV650) [anti-TIM-3] BioLegend 345027 (RRID:AB_2565828)
Mab11 (BV650) [anti-TNF] BioLegend 502937 (RRID:AB_2561355)
4B41 (BV421) [anti-4-1BB] BioLegend 309820 (RRID:AB_2563830)
C63D9 (AF488) [anti-TCF1] Cell Signaling 6444 (RRID:AB_2797627)
REA473 (A647) [anti-TOX] Miltenyi Biotec 130-107-838 (RRID:AB_2654224)
S3.5 (PE-Cy5.5) [anti-CD4] Thermo Fisher Scientific MHCD0418 (RRID:AB_10376013)
eBio64DEC17 (PE) [anti-IL-17A] Thermo Fisher Scientific 12-7179-42 (RRID:AB_1724136)
Purified anti-CD28/CD49d BD Biosciences 347690 (RRID:AB_647457)
Ultra-LEAF OKT3 Purified [anti-CD3] BioLegend 317326 (RRID: AB_11150592)
LIVE/DEAD Fixable Aqua Dead Cell
Stain Kit
Thermo Fisher Scientific Cat#L34957
(Continued on next page)
ll
OPEN ACCESS
Cell 183, 158–168.e1–e5, October 1, 2020 e1
Article
RESOURCE AVAILABILITY
Lead Contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Marcus
Buggert (marcus.buggert@ki.se)
Material Availability
HLA class I tetramers can be generated and shared on a collaborative basis.
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Biological Samples
Acute moderate blood samples Karolinska Hospital N/A
Acute severe blood samples Karolinska Hospital N/A
Mild convalescent blood samples Karolinska Hospital N/A
Severe convalescent blood samples Karolinska Hospital N/A
Exposed family member blood samples Karolinska Hospital N/A
2020 blood donor samples Karolinska Universitetslaboratoriet N/A
2019 blood donor samples Karolinska Universitetslaboratoriet N/A
Chemicals, Peptides, and Recombinant Proteins
Synthetic CMV peptides Peptides & Elephants GmbH https://www.peptides.de/
Synthetic EBV peptides Peptides & Elephants GmbH https://www.peptides.de/
Synthetic overlapping SARS-CoV-2
peptides (Prot_M, N, S)
Miltenyi Biotec https://www.miltenyibiotec.com/SE-en/
Optimal SARS-CoV-2 peptides Peptides & Elephants GmbH https://www.peptides.de/
Foxp3/TF Staining Buffer Set Invitrogen Cat#00-5523-00
Staphylococcal Enterotoxin B (SEB) Sigma-Aldrich Cat#S4881
Brefeldin A BioLegend Cat#420601
BD GolgiStop (with Monensin) BD Biosciences Cat#554724
BCIP/NBT Substrate Mabtech Cat#3650-10
BL21(DE3)pLysS Competent Cells Novagen Cat#69451
Triton X-100 Buffer Sigma-Aldrich Cat#X100
8M Urea Buffer Sigma-Aldrich Cat#51457
Cysteamine Sigma-Aldrich Cat#M9768
D-(+)-Biotin Sigma-Aldrich Cat#2031
Critical Commercial Assays
iFLASH Anti-SARS-CoV-2 IgG Kit YHLO Cat#20210217
LIAISON SARS-CoV-2 ELISA IgG Kit DiaSorin Cat#311450
Human IFN-g ELISpot Kit Mabtech Cat#3420-2A
BD Multitest 6-color TBNK reagent with BD
Trucount tubes
BD Biosciences Cat#337166
CellTrace Violet Cell Proliferation Kit Thermo Fisher Scientific Cat#C34571
Software and Algorithms
NetMHCpan EL 4.1 DTU Bioinformatics http://www.cbs.dtu.dk/services/
NetMHCpan-4.1/
FlowJo 10.6.1 FlowJo https://www.flowjo.com/
UMAP plugin 2.2 FlowJo https://www.flowjo.com/
GraphPad Prism 8.4.2 GraphPad Software https://www.graphpad.com/
PhenoGraph 0.2.1 PhenoGraph https://omictools.com/phenograph-tool
RStudio RStudio https://rstudio.com/
Python (scikit-learn 0.22.1) Python https://www.python.org/
ll
OPEN ACCESS
e2 Cell 183, 158–168.e1–e5, October 1, 2020
Article
Data and Code Availability
The published article includes all data generated during this study. All codes are freely available at source.
EXPERIMENTAL MODEL AND SUBJECT DETAILS
Human subjects
Maximal disease severity was assessed using the NIH Ordinal Scale and Sequential Organ Failure Assessment (SOFA) (Beigel et al.,
2020; Singer et al., 2016). The NIH Ordinal Scale was defined as follows: (1) not hospitalized with no limitation of activities; (2) not
hospitalized with limitation of activities and/or home oxygen requirement; (3) hospitalized but not requiring supplemental oxygen
and no longer requiring ongoing medical care; (4) hospitalized and not requiring supplemental oxygen but requiring ongoing medical
care; (5) hospitalized requiring supplemental oxygen; (6) hospitalized requiring non-invasive ventilation or the use of high-flow oxygen
devices; (7) hospitalized receiving invasive mechanical ventilation or extracorporeal membrane oxygenation; and (8) death.
Donors were assigned to one of seven groups for the purposes of this study. AS: patients with acute severe disease requiring
hospitalization in the high dependency or intensive care unit, with low-flow oxygen support (< 10 L/min), high-flow oxygen support,
or invasive mechanical ventilation (n = 17). These patients had a median NIH Ordinal Scale score of 7 (IQR 6–7) and a median SOFA
score of 6 (IQR 3–6) at the time of sampling 12–17 days after disease onset (47% were viremic, and 82% were antibody-seropositive
for SARS-CoV-2). AM: patients with acute moderate disease requiring hospitalization and low-flow oxygen support (0–3 L/min;
n = 10). These patients had a median NIH Ordinal Scale score of 5 (IQR 5–5) and a median SOFA score of 1 (IQR 1–1) at the time
of sampling 11–14 days after disease onset (40% were viremic, and 50% were antibody-seropositive for SARS-CoV-2). SC: individ-
uals in the convalescent phase after severe disease (n = 26). Samples were collected 42–58 days after disease onset, corresponding
to 3–21 days after resolution of symptoms (100% were antibody-seropositive for SARS-CoV-2). MC: individuals in the convalescent
phase after mild disease (n = 40). Samples were collected 49–64 days after disease onset, corresponding to 25–53 days after
resolution of symptoms (85% were antibody-seropositive for SARS-CoV-2). Exp: family members who shared a household with
donors in groups MC or SC (n = 30). These individuals were exposed at the time of symptomatic disease (21% remained asymptom-
atic, and 63% were antibody-seropositive for SARS-CoV-2). 2020 BD: individuals who donated blood at the Karolinska University
Hospital in May 2020 (during the pandemic; n = 55). 2019 BD: individuals who donated blood at the Karolinska University Hospital
between July and September 2019 (before the pandemic; n = 28).
PBMCs were isolated from heparin-coated tubes (groups AS, AM, SC, MC, Exp, and 2020 BD) or citrate-anticoagulated buffy
coats (group 2019 BD). A separate serum separator tube was collected from each donor. All donors provided written informed
consent in accordance with the Declaration of Helsinki. The study was approved by the Swedish Ethical Review Authority. Donor
characteristics are summarized in Table S3, and immunological assay breakdowns are summarized in Table S4.
METHOD DETAILS
Flow cytometry
PBMCs were isolated from venous blood samples via standard density gradient centrifugation and used immediately (groups AS,
AM, SC, MC, Exp, and 2020 BD) or after cryopreservation in liquid nitrogen (group 2019 BD). Cells were washed in phosphate-buff-
ered saline (PBS) supplemented with 2% fetal bovine serum (FBS) and 2 mM EDTA (FACS buffer) and stained with PE-conjugated or
BV421-conjugated HLA class I tetramers and/or anti-CCR7–APC-Cy7 (clone G043H7; BioLegend) for 10 min at 37
C. Other surface
markers were detected via the subsequent addition of directly conjugated antibodies at pre-titrated concentrations for 20 min at
room temperature, and viable cells were identified by exclusion using a LIVE/DEAD Fixable Aqua Dead Cell Stain Kit (Thermo Fisher
Scientific). Cells were then washed again in FACS buffer and fixed/permeabilized using a FoxP3 / Transcription Factor Staining Buffer
Set (eBioscience). Intracellular markers were detected via the addition of directly conjugated antibodies at pre-titrated concentra-
tions for 1 hr at 4
C. Stained cells were fixed in PBS containing 1% paraformaldehyde (PFA; Biotium) and stored at 4
C. Samples
were acquired using a FACSymphony A5 (BD Biosciences). Data were analyzed with FlowJo software version 10.6.1 (FlowJo
LLC). Gating strategies were described previously (Buggert et al., 2014, 2018). Core gates included singlet isolation (FSC-H versus
FSC-A), live CD3 selection (CD3 versus Aqua, CD14, and CD19), lymphocyte enrichment (SSC-A versus FSC-A), CD4 or CD8 selec-
tion (CD4 versus CD8). Detailed gating strategies for individual markers are depicted in the relevant figures.
Peptides
Peptides corresponding to known optimal epitopes derived from CMV (pp65) and EBV (BZLF1 and EBNA-1), overlapping peptides
spanning the immunogenic domains of the SARS-CoV-2 spike (Prot_S), membrane (Prot_M), and nucleocapsid proteins (Prot_N),
and optimal peptides for the manufacture of HLA class I tetramers were synthesized at  95% purity. Lyophilized peptides were re-
constituted at a stock concentration of 10 mg/mL in DMSO and further diluted to 100 mg/mL in PBS.
Epitope prediction
Peptides were selected from full-length SARS-CoV-2 sequences spanning 82 different strains from 13 countries (National Center for
Biotechnology Information). The predicted binding affinities of conserved 9-mer peptides for HLA-A*0201 and HLA-B*0702 were
ll
OPEN ACCESS
Cell 183, 158–168.e1–e5, October 1, 2020 e3
Article
determined using NetMHCpan version 4.1 (Reynisson et al., 2020). Binders were defined by a threshold IC50 value of 500 nM. Strong
binders were defined by a % Rank  0.5, and weak binders were defined by a % Rank  2 (Table S2). A total of 13 strong binders were
identified for tetramer generation (Table S2).
Tetramers
HLA class I tetramers were generated as described previously (Price et al., 2005). Briefly, biotin-tagged HLA-A*0201 and HLA-B*0702
heavy chains were expressed under the control of a T7 promoter as insoluble inclusion bodies in Escherichia coli strain BL21(DE3)
pLysS (Novagen). IPTG-induced Escherichia coli were lyzed via repeated freeze/thaw cycles to release inclusion bodies that were
subsequently purified by washing in 0.5% Triton X-100 buffer (Sigma-Aldrich). Heavy chain and b2 m inclusion body preparations
were denatured separately in 8 M urea buffer (Sigma-Aldrich) and mixed at a 1:1 molar ratio. Each monomeric protein was refolded
in 2-mercaptoethylamine/cystamine redox buffer (Sigma-Aldrich) with the appropriate synthetic peptide (Peptides  Elephants
GmbH). Refolded monomers were purified via anion exchange after buffer replacement (10 mM Tris, pH 8.1). Purified monomers
were biotinylated using d-biotin and BirA (Sigma-Aldrich). Excess biotin was removed via gel filtration. Biotinylated monomers
were then tetramerized via the addition of fluorochrome-conjugated streptavidin at a 4:1 molar ratio, respectively. The following
specificities were used in this study: CMV A*0201 NV9 (NLVPMVATV), EBV A*0201 GL9 (GLCTLVAML), SARS-CoV-2 A*0201 AV9
(ALSKGVHFV), SARS-CoV-2 A*0201 HI9 (HLVDFQVTI), SARS-CoV-2 A*0201 KV9 (KLLEQWNLV), SARS-CoV-2 A*0201 LL9
(LLLDRLNQL), SARS-CoV-2 A*0201 LLY (LLYDANYFL), SARS-CoV-2 A*0201 SV9 (SLVKPSFYV), SARS-CoV-2 A*0201 TL9
(TLDSKTQSL), SARS-CoV-2 A*0201 VL9 (VLNDILSRL), SARS-CoV-2 A*0201 YL9 (YLQPRTFLL), SARS-CoV-2 B*0702 FI9
(FPRGQGVPI), SARS-CoV-2 B*0702 KT9 (KPRQKRTAT), SARS-CoV-2 B*0702 SA9 (SPRRARSVA), and SARS-CoV-2 B*0702 SL9
(SPRWYFYYL).
Functional assay
PBMCs were resuspended in complete medium (RPMI 1640 supplemented with 10% FBS, 1% L-glutamine, and 1% penicillin/strep-
tomycin) at 1 3 107
cells/mL and cultured at 1 3 106
cells/well in 96-well V-bottom plates (Corning) with the relevant peptides (each at
0.5 mg/mL) for 30 min prior to the addition of anti-CD28/CD49d (3 mL/mL; clone L293/L25; BD Biosciences), brefeldin A (1 mL/mL;
Sigma-Aldrich), monensin (0.7 mL/mL; BD Biosciences), and anti-CD107a–PE-CF594 (clone H4A3; BD Biosciences). Negative con-
trol wells lacked peptides, and positive control wells included staphylococcal enterotoxin B (SEB; 0.5 mg/mL; Sigma-Aldrich) or plate-
bound anti-CD3 (1 mg/mL; clone OKT3; BioLegend). Cells were analyzed by flow cytometry after incubation for 8 hr at 37
C.
Proliferation assay
PBMCs were labeled with CTV (0.5 mM; Thermo Fisher Scientific), resuspended in complete medium at 1 3 107
cells/mL, and
cultured at 1 3 106
cells/well in 96-well U-bottom plates (Corning) with the relevant peptides (each at 1 mg/mL) in the presence of
anti-CD28/CD49d (3 mL/mL; clone L293/L25; BD Biosciences) and IL-2 (10 IU/mL; PeproTech). Negative control wells lacked
peptides, and positive control wells included SEB (0.5 mg/mL; Sigma-Aldrich) or plate-bound anti-CD3 (1 mg/mL; clone OKT3;
BioLegend). Functional assays were performed as described above after incubation for 5 days at 37
C.
AIM assay
PBMCs were resuspended in complete medium at 1 3 107
cells/mL and cultured at 1 3 106
cells/well in 96-well U-bottom plates
(Corning) with the relevant peptides (each at 1 mg/mL) in the presence of anti-CD28/CD49d (3 mL/mL; clone L293/L25; BD
Biosciences). Cells were analyzed by flow cytometry after incubation for 24 hr at 37
C. The following directly conjugated monoclonal
antibodies were used to detect activation markers: anti-CD69–BUV737 (clone FN50; BD Biosciences) and anti-4-1BB–BV421 (clone
4B41; BioLegend).
Trucount
Absolute counts were obtained using Multitest 6-color TBNK reagent with Trucount tubes (BD Biosciences). Samples were fixed with
2% PFA for 2 hr prior to acquisition. Absolute CD3+
cell counts were calculated using the following formula: (# CD3+
events acquired x
total # beads x 1000) / (# beads acquired x volume of whole blood stained in mL). CD4+
and CD8+
cell counts were computed from the
respective frequencies relative to CD3+
cells.
Principal component analysis
Analyses were performed using scikit-learn version 0.22.1 in Python. Data were normalized using sklearn.preprocessing.
StandardScaler in the same package to generate z-scores for PCA.
UMAP
FCS 3.0 data files were imported into FlowJo software version 10.6.0 (FlowJo LLC). All samples were compensated electronically.
Dimensionality reduction was performed using the FlowJo plugin UMAP version 2.2 (FlowJo LLC). The downsample version 3.0.0
plugin and concatenation tool was used to visualize multiparametric data from up to a total of 120,000 CD8+
T cells (n = 3 donors
per group). Representative donors were selected from the 50th
percentile for all markers. The following parameters were used in
ll
OPEN ACCESS
e4 Cell 183, 158–168.e1–e5, October 1, 2020
Article
these analyses: metric = euclidean, nearest neighbors = 30, and minimum distance = 0.5. Clusters of phenotypically related cells
were detected using PhenoGraph version 0.2.1. The following markers were included in the cluster analysis: CCR7, CD27, CD28,
CD38, CD39, CD45RA, CD95, CD127, CTLA-4, CXCR5, granzyme B, Ki-67, LAG-3, PD-1, perforin, TCF1, TIGIT, TIM-3, TOX, and
2B4. Plots were generated using Prism version 8.2.0 (GraphPad Software Inc.).
ELISpot assay
PBMCs were rested overnight in complete medium and seeded at 2 3 105
cells/well in MultiScreen HTS Filter Plates (Merck Millipore)
pre-coated with anti-IFN-g (15 mg/mL; clone 1-D1K; Mabtech). Test wells were supplemented with overlapping peptides spanning
Prot_S, Prot_M, and Prot_N (each at 2 mg/mL; Miltenyi Biotec) or peptides corresponding to known optimal epitopes derived from
CMV (pp65) or EBV (BZLF1 and EBNA-1) (each at 2 mg/mL; Peptides  Elephants GmbH). Negative control wells lacked peptides, and
positive control wells included SEB (0.5 mg/mL; Sigma-Aldrich). Assays were incubated for 24 hr at 37
C. Plates were then washed six
times with PBS (Sigma-Aldrich) and incubated for 2 hr at room temperature with biotinylated anti-IFN-g (1 mg/mL; clone mAb-7B6-1;
Mabtech). After six further washes, a 1:1,000 dilution of alkaline phosphatase-conjugated streptavidin (Mabtech) was added for 1 hr
at room temperature. Plates were then washed a further six times and developed for 20 min with BCIP/NBT Substrate (Mabtech). All
assays were performed in duplicate. Mean values from duplicate wells were used for data representation. Spots were counted using
an automated ELISpot Reader System (Autoimmun Diagnostika GmbH).
Serology
SARS-CoV-2-specific antibodies were detected in serum using both the iFLASH Anti-SARS-CoV-2 IgG chemiluminescent micropar-
ticle immunoassay against the nucleocapsid and spike proteins (Shenzhen YHLO Biotech Co. Ltd.) as well as the LIAISON
SARS-CoV-2 IgG fully automated indirect chemiluminescent immunoassay against the S1 and S2 (spike) proteins (DiaSorin). These
assays produced highly concordant results and have been shown to perform adequately as diagnostic tools (Plebani et al., 2020). An
individual was considered seropositive if one of the two methods generated a positive result. All assays were performed by trained
employees at the clinical laboratory according to standard procedures.
QUANTIFICATION AND STATISTICAL ANALYSIS
Statistical analyses were performed using R studio or Prism version 7.0 (GraphPad Software Inc.). Differences between unmatched
groups were compared using an unpaired t test, the Mann-Whitney U test, or the Kruskal-Wallis rank-sum test with Dunn’s post hoc
test for multiple comparisons, and differences between matched groups were compared using a paired t test or the Wilcoxon signed-
rank test. Correlations were assessed using the Spearman rank correlation. Non-parametric tests were used if the data were not
distributed normally according to the Shapiro-Wilk normality test. Phenotypic relationships within multivariate datasets were
visualized using FlowJo software version 10.6.1 (FlowJo LLC).
ll
OPEN ACCESS
Cell 183, 158–168.e1–e5, October 1, 2020 e5
Article
Supplemental Figures
A
2020 BD AM AS
0
400000
800000
1000000
2000000
2020 BD AM AS
0
200000
400000
600000
800000
1000000
1500000
2020 BD AM AS
0
5
10
20
2020 BD AM AS
0
4
8
12
16
CD14
CD15
Time
SSC-A
CD45
CD3
CD45
SSC-A
FSC-A
FSC-H
CD19
CD3
Beads
0.34
3.33
95.4
33.9
75.1
Gating strategy of total T cells from SARS-CoV-2+ acute patient
B
CD95
CCR7
CD95
CCR7
Gating strategy for the identification of memory CD4+ and CD8+ T cells
Gated on CD3+CD8+
Gated on CD3+CD4+
C
CD8
CD4
%
of
memory
CD4+
T
cells
CCR7
0
20
40
60
80
100
0
5
10
15
20
25
CD25 CD27
20
40
60
80
100
CD28
0
20
40
60
80
100
0
8
16
24
32
40
CD39
0
20
40
60
80
100
CD127
CTLA4
0
4
8
12
16
20
LAG-3
0
15
30
45
60
CXCR5
0
8
16
24
32
40
HLA-DR
GzmB
0
16
32
48
64
80
0
8
16
24
32
40
0
Perforin
0
5
10
15
20
25
CD45RA
0
5
10
15
20
25
0
20
40
60
80
100
TCF-1
0
15
30
45
60
TIGIT TIM-3
TOX
%
of
memory
CD8+
T
cells
0
20
40
60
80
100
2B4
0
20
40
60
80
100
CCR7
0
10
20
30
40
50
CD25
0
20
40
60
80
100
CD27
0
20
40
60
80
100
CD28
0
20
40
60
80
100
CD127 CXCR5
0
6
12
18
24
30
0
20
40
60
80
100
GzmB
PD-1
0
20
40
60
80
100
0
20
40
60
80
100
Perforin
0
20
40
60
80
100
CD45RA
TCF-1
0
20
40
60
80
100
0
20
40
60
80
100
TIGIT
0
20
40
60
80
100
TOX
2020 BD MC AM AS
2020 BD MC AM AS
0
2
4
6
8
0
10
20
30
40
50
Abs count memory CD4+ T cells
Abs count memory CD8+ T cells
% memory CD4+ T cells
% memory CD8+ T cells
(legend continued on next page)
ll
OPEN ACCESS
Article
Figure S1. Quantification and Characterization of CD4+
and CD8+
T Cells in COVID-19, Related to Figure 1
(A) Flow cytometric gating strategy for the identification and quantification of CD4+
and CD8+
T cells. (B) Left: flow cytometric gating strategy for the identification
and quantification of memory CD4+
and CD8+
T cells. Right: dot plots summarizing the absolute numbers and relative frequencies of memory CD4+
and CD8+
T cells by group. Each dot represents one donor. Data are shown as median ± IQR. 2020 BD: healthy blood donors from 2020 (n = 18). AM: patients with acute
moderate COVID-19 (n = 11). AS: patients with acute severe COVID-19 (n = 17). (C) Dot plots summarizing the expression frequencies of phenotypic markers
among memory CD4+
and CD8+
T cells by group. Each dot represents one donor. Bars indicate median values. 2020 BD: healthy blood donors from 2020 (n = 18).
MC: individuals in the convalescent phase after mild COVID-19 (n = 31). AM: patients with acute moderate COVID-19 (n = 11). AS: patients with acute severe
COVID-19 (n = 17).
ll
OPEN ACCESS Article
***
UMAP2
UMAP1
HLA-DR
Ki-67
CD38
LAG-3
TIM-3
CD39
2B4
TIGIT
PD-1
CD27
CD127
CXCR5
GzmB
Perforin
CD95
CD28
CD45RA
CCR7
Gated on memory
CD4+ T cells
Max
Min
CTLA4 CD25
CD69
TOX TCF-1
2020 BD MC AM AS
B
CD38+
Ki67+
CD4+
CD38+
HLA-DR+
CD4+
CD38+
PD-1+
CD4+
CD38+
Ki67+
CD8+
CD38+
HLA-DR+
CD8+
CD38+
PD-1+
CD8+
CD38
+
PD-1
+
CD8
+
CD38
+
HLA-DR
+
CD4
+
CD38
+
PD-1
+
CD4
+
CD38
+
Ki67
+
CD8
+
CD38
+
HLA-DR
+
CD8
+
8
3
D
C
+
1
-
D
P
+
8
D
C
+
d
e
e
n
n
e
g
y
x
o
k
a
e
P
e
g
A
I
M
B
n
o
i
s
s
i
m
d
a
o
t
t
u
b
e
d
m
o
t
p
m
y
s
s
y
a
D
g
n
i
l
p
m
a
s
t
u
b
e
d
m
o
t
p
m
y
s
s
y
a
D
y
a
t
s
U
C
I
n
o
i
t
a
r
u
D
n
o
i
t
a
b
u
t
n
i
U
C
I
n
o
i
t
a
r
u
D
t
n
e
m
t
a
e
r
t
n
e
g
y
x
o
n
o
i
t
a
r
u
D
)
l
/
g
m
3

f
e
r
(
h
4
2
-
/
+
P
R
C
t
s
e
h
g
i
H
P
R
C
t
s
e
h
g
i
H
)
l
/
g
µ
5
.
0

f
e
r
(
h
4
2
-
/
+
T
C
P
t
s
e
h
g
i
H
Highest
PCT
Lowest
Hb
+/-
24h
(ref
117–153
g/l)
Highest
leukocyte
count
+/-
24h
(ref
3.5–8.8
x
10
9
c/l)
Lowest
leukocyte
count
+/-
24h
Highest
neutrophil
count
+/-
24h
(ref
1.6–5.9
x
10
9
c/l)
Lowest
neutrophil
count
Highest
ferritin
+/-
24h
(ref
10–150
µg/l)
Highest
ferritin
Highest
troponin
T
+/-
24h
(ref
15
ng/l)
Highest
troponin
T
Highest
D-dimer
+/-
24h
(ref
0.56
mg/l)
Highest
D-dimer
Highest
PK-INR
+/-
24h
(ref
1.2
INR)
Highest
PK-INR
Highest
fibrinogen
+/-24h
(ref
2–
4.2
g/l)
Highest
fibrinogen
Lowest
thrombocytes
+/-
24h
(ref
156–387
x
10
9
c/l)
Lowest
thrombocytes
Highest
thrombocytes
Highest
bilirubin
+/-
24h
(ref

26
µg/l)
Highest
bilirubin
Highest
ALT
(ref

0.76
µkat/l)
Highest
LD
(ref

3.5
µkat/l)
Lowest
P-albumin
+/-
24h
(ref

73
µg/l)
Highest
myoglobin
(ref

73
µg/l)
Highest
IL-6
+/-
5
days
(ref

7
ng/l)
Highest
IL-1
+/-
5
days
(ref

5
ng/l)
Highest
IL-10
+/-
5
days
(ref

5
ngl)
Highest
TNF
+/-
5
days
(ref

12
ng/l)
CMV-IgG
(U/ml),
Diasonin
Liason
XL
SARS-CoV-2-IgG
(AU/ml),
iFlash
YHLO
Lowest
lymphocyte
count
+/-
24h
(ref
1.1–3.5
x
10
9
c/l)
Lowest
lymphocyte
count
Highest
creatinine
+/-
24h
(ref

90
µg/l)
Highest
creatinine
1
0
-1
C
D
Cycling
20
10
5
15
0
CD38+PD-1+
CD38+HLA-DR+
CD38+Ki-67+
%
of
memory
CD4+
T
cells
***
***
***
***
25
40
20
10
30
0
50
20
10
5
15
0
***
*
***
***
***
***
2020 BD MC
AM AS
***
A
3
15
16
18
26
27
17
29
25
22
19
23
21
28
20
13
14
12
9
8
7
6
5
4
1
2
11
10 30
24
Phenograph clusters from
memory CD8+ T cells
C25 C29
TOX
CCR7
CXCR5
TCF1
CD95
Ki67
Perforin
CTLA4
Granzyme
CD38
CD28
HLA-DR
LAG3
TIGIT
CD45RA
CD127
Tim3
CD39
CD69
CD27
2B4
CD25
PD-1
Log2 Expression
25
29
0 50
% coontribution to each cluster
100
Healthy
Control
Convalescent Mild Severe
(legend on next page)
ll
OPEN ACCESS
Article
Figure S2. Phenograph and UMAP Clustering of Memory CD4+
and CD8+
T Cells with Correlative Analyses of Immune Activation Phenotypes
versus Clinical Parameters in Acute COVID-19, Related to Figure 1
(A) Phenograph plots showing the clustering of memory CD8+
T cells and heatmap highlighting clusters 20 and 29. (B) Top left: UMAP plots showing the clustering
of memory CD4+
T cells by group in relation to all memory CD4+
T cells (left). Bottom left: UMAP plots showing the expression of individual markers (n = 3 donors
per group). Right: dot plots summarizing the expression frequencies of activation/cycling markers among memory CD4+
T cells by group. Each dot represents
one donor. Data are shown as median ± IQR. 2020 BD: healthy blood donors from 2020 (n = 18). MC: individuals in the convalescent phase after mild COVID-19
(n = 31). AM: patients with acute moderate COVID-19 (n = 11). AS: patients with acute severe COVID-19 (n = 17). *p  0.05, **p  0.01, ***p  0.001. Kruskal-Wallis
rank-sum test with Dunn’s post hoc test for multiple comparisons. (C) and (D) Heatmaps summarizing the pairwise correlations between phenotypically defined
subpopulations of memory CD4+
or CD8+
T cells and various clinical parameters in patients with acute COVID-19. *p  0.05, **p  0.01, ***p  0.001. Spearman
rank correlation.
ll
OPEN ACCESS Article
CCR7
96 4
Acute
Healthy
CD127
89 11
TIGIT
90 10
TCF-1
96 4
Perforin
83
17
Granzyme B
94
6
CD45RA
98 2
CD28
73
26
CD27
88
12
1
1
14
3
86
97
99
99
CD38
Ki-67
Gated on memory CD8+ T cells
C D
A
E
CD38
UMAP
Y-axis
UMAP X-axis
CD38 HLA-DR
Ki-67 PD-1
26.0 2.9
12.5 1.63
29.8 22.1
B
0
20
40
60
80
2020
BD
AM
AS
%
CD8+CD38+HLA-DR−
Low
High
***
***
Ki-67
A*02:01
NV9
CM
V
HLA-DR
Gated on memory CD8+ T cells
CD38+HLA-DR+ donor CD38+HLA-DR− donor CD38+HLA-DR+ donor CD38+HLA-DR− donor
Ki-67
PD-1
E
0.0
0.1
0.2
0.3
0.4
0
-10
3
10
3
10
4
10
5
0
10
3
10
4
10
5
0
-10
3
10
3
10
4
10
5
Comp-PE-A :: B*0702 SL9
0
10
3
10
4
10
5
0.34
0.09
Representative FACS plots for 2 MC subjects
SC MC
A*0201 TL9 B*0702 SL9
A*0201
YL9
B*0702
FI9
Gated on CD8+ T cells
CCR7
CD95
%
SARS-Cov-2
tetramer+
cells
Figure S3. Immune Activation Patterns in Acute COVID-19, Related to Figure 2
(A) Representative flow cytometry plots showing the expression of activation/cycling markers among memory CD8+
T cells in patients with acute severe COVID-
19. Numbers indicate percentages in the drawn gates. (B) Top left: UMAP plots showing the clustering of memory CD8+
T cells by phenotype in relation to all
memory CD8+
T cells (left). Bottom left: UMAP plots showing the expression of individual markers (n = 1 donor per group). Right: dot plot summarizing the
frequencies of CD38+
HLA-DR memory CD8+
T cells by group. Each dot represents one donor. Data are shown as median ± IQR. 2020 BD: healthy blood donors
from 2020 (n = 18). AM: patients with acute moderate COVID-19 (n = 11). AS: patients with acute severe COVID-19 (n = 17). ***p  0.001. Kruskal-Wallis rank-sum
test with Dunn’s post hoc test for multiple comparisons. (C) Representative flow cytometry plots showing the expression of activation/cycling markers among
(legend continued on next page)
ll
OPEN ACCESS
Article
CMV-specific memory CD8+
T cells in a healthy control and a patient with acute severe COVID-19. Numbers indicate percentages in the drawn gates. (D)
Representative flow cytometry plots showing the phenotype of Ki-67+
memory CD8+
T cells in a patient with acute severe COVID-19. Numbers indicate per-
centages in the drawn gates. (E) Left: representative flow cytometry plots showing SARS-CoV-2-specific tetramer+
CD8+
T cells in two donors from group MC.
Right: dot plot summarizing the frequencies of SARS-CoV-2-specific tetramer+
CD8+
T cells in donors from groups MC (n = 10) and SC (n = 2).
ll
OPEN ACCESS Article
A
B
Figure S4. Quantification of Functional T Cell Reactivity in COVID-19, Related to Figure 3
(A) Representative images showing the detection of IFN-g-producing cells responding to overlapping peptides spanning the immunogenic domains of the SARS-
CoV-2 spike (S), membrane (M), and nucleocapsid proteins (N) by group (ELISpot assays). NC: negative control. EBV: Epstein-Barr virus. CMV: cytomegalovirus.
SEB: staphylococcal enterotoxin B. (B) Dot plots summarizing the frequencies of IFN-g-producing cells responding to optimal peptide epitopes derived from EBV
BZLF1 and EBNA-1 (left) or CMV pp65 (right) by group (ELISpot assays). Each dot represents the mean of combined specificities in one donor. Bars indicate
median values. No significant differences were detected among groups for any specificity. 2019 BD: healthy blood donors from 2019 (n = 25). 2020 BD: healthy
blood donors from 2020 (n = 24). Exp: exposed family members (n = 30). MC: individuals in the convalescent phase after mild COVID-19 (n = 31). SC: individuals in
the convalescent phase after severe COVID-19 (n = 22). SFU: spot-forming unit.
ll
OPEN ACCESS
Article
A 38
62 38
62
14.3
37.1
32.9 15.7
78.3 21.7
CXCR5
CXCR3
cTfh
Th1
CXCR5
CXCR3
Th1/17
Th17
Th2
c
T
f
h
1
c
T
f
h
2
c
T
f
h
1
7
c
T
f
h
1
/
1
7
c
T
f
h
1
c
T
f
h
2
c
T
f
h
1
7
c
T
f
h
1
/
1
7
0
20
40
60
80
c
T
f
h
1
c
T
f
h
2
c
T
f
h
1
7
c
T
f
h
1
/
1
7
c
T
f
h
1
c
T
f
h
2
c
T
f
h
1
7
c
T
f
h
1
/
1
7
c
T
f
h
1
c
T
f
h
2
c
T
f
h
1
7
c
T
f
h
1
/
1
7
c
T
f
h
1
c
T
f
h
2
c
T
f
h
1
7
c
T
f
h
1
/
1
7
cTfh
polarization
(based
on
CD69+4-1BB+)
Spike (S) Membrane (M) Nucleocapsid (N)
B
CCR4
CXCR3
CD69
4-1BB
Gated on memory CD4+ T cells
78 22
62 38
Th1
Th17
Th1/17
Th2
33
37
16
14
y
C
0.10 0.02
0.08
0.99 0.54
0.24
0.34 0.29
0.27
0.47 0.35
0.27
D
IFN-
TNF
Gated on memory CD4+ T cells
NC S M N
0 50 100 150 200
0
100
200
300
400
500 r = 0.82
p  0.001
anti-N IgG
anti-S
IgG
S
e
r
o
+
S
e
r
o
−
0.1
1
10
CD4+ S
S
e
r
o
+
S
e
r
o
−
CD4+ M
S
e
r
o
+
S
e
r
o
−
CD4+ N
S
e
r
o
+
S
e
r
o
−
CD8+ S
S
e
r
o
+
S
e
r
o
−
CD8+ M
S
e
r
o
+
S
e
r
o
−
CD8+ N
S
e
r
o
+
S
e
r
o
−
0.1
1
10
CD4+ S
S
e
r
o
+
S
e
r
o
−
CD4+ M
S
e
r
o
+
S
e
r
o
−
CD4+ N
S
e
r
o
+
S
e
r
o
−
CD8+ S
S
e
r
o
+
S
e
r
o
−
CD8+ M
S
e
r
o
+
S
e
r
o
−
CD8+ N
2020 BD MC SC
Exp
%
CTV
lo
IFN-
+
* *
E
F
2
0
1
9
B
D
S
e
r
o
−
2
0
1
9
B
D
S
e
r
o
−
CD4+ S
***
CD4+ M
***
CD8+ S
***
CD8+ M
***
CD8+ N
***
CD4+ N
***
0.1
1
10
2
0
1
9
B
D
S
e
r
o
−
2
0
1
9
B
D
S
e
r
o
−
2
0
1
9
B
D
S
e
r
o
−
2
0
1
9
B
D
S
e
r
o
−
Figure S5. Functional Polarization of SARS-CoV-2-Specific Memory CD4+
T Cells, Antibody Correlations, and Comparative Analyses of
SARS-CoV-2-Specific CD4+
and CD8+
T Cell Responses versus Serostatus in COVID-19, Related to Figures 3 and 4
(A) Representative flow cytometry plots showing the identification of memory CD4+
T cells responding to overlapping peptides spanning the immunogenic
domains of the SARS-CoV-2 nucleocapsid protein by subset (AIM assay). Subsets were defined as CXCR5+
(cTfh), CCR4 CCR6 CXCR3+
CXCR5 (Th1),
CCR4+
CCR6 CXCR3 CXCR5 (Th2), CCR4 CCR6+
CXCR3 CXCR5 (Th17), CCR4 CCR6+
CXCR3+
CXCR5 (Th1/17), and CCR4 CCR6 CXCR3
CXCR5 (non-Th1/2/17). (B) Bar graphs summarizing the functional polarization of memory CXCR5+
(cTfh) CD4+
T cells responding to overlapping peptides
spanning the immunogenic domains of the SARS-CoV-2 spike (S), membrane (M), and nucleocapsid proteins (N). Data are shown as median ± IQR. Key as in C.
(C) Correlation between anti-spike (S) and anti-nucleocapsid (N) IgG levels. Each dot represents one donor. 2020 BD: healthy blood donors from 2020 (n = 31).
Exp: exposed family members (n = 28). MC: individuals in the convalescent phase after mild COVID-19 (n = 31). SC: individuals in the convalescent phase after
severe COVID-19 (n = 23). Spearman rank correlation. (D) Representative flow cytometry plots showing functional SARS-CoV-2-specific memory CD4+
T cell
responses in a seronegative convalescent donor (group MC). Numbers indicate percentages in the drawn gates. NC: negative control. S: spike. M: membrane. N:
(legend continued on next page)
ll
OPEN ACCESS Article
nucleocapsid. (E) Dot plots summarizing SARS-CoV-2-specific CD4+
and CD8+
T cell responses versus serostatus in exposed family members (left) and in-
dividuals in the convalescent phase after mild COVID-19 (right). Each dot represents one donor. Data are shown as median ± IQR. S: spike. M: membrane. N:
nucleocapsid. *p  0.05. Mann-Whitney U test. (F) Dot plots summarizing SARS-CoV-2-specific CD4+
and CD8+
T cell responses in exposed seronegative family
members and unexposed healthy blood donors (group 2019 BD). Each dot represents one donor. Data are shown as median ± IQR. *p  0.05. Mann-Whitney
U test.
ll
OPEN ACCESS
Article

More Related Content

What's hot

疫苗研發之產學研合作經驗分享 以冠狀病毒奈米疫苗為例
疫苗研發之產學研合作經驗分享以冠狀病毒奈米疫苗為例疫苗研發之產學研合作經驗分享以冠狀病毒奈米疫苗為例
疫苗研發之產學研合作經驗分享 以冠狀病毒奈米疫苗為例hprc_tmu
 
an mrna vaccine aganist sars cov 2
an mrna vaccine aganist sars cov 2 an mrna vaccine aganist sars cov 2
an mrna vaccine aganist sars cov 2 antomathew8
 
FINAL POSTER BIOTECH
FINAL POSTER BIOTECHFINAL POSTER BIOTECH
FINAL POSTER BIOTECHEmine Taytas
 
Patil S et al 2014 Virology
Patil S et al 2014 VirologyPatil S et al 2014 Virology
Patil S et al 2014 VirologySHILPA PATIL
 
Paracrine TGF_IL2 paper
Paracrine TGF_IL2 paperParacrine TGF_IL2 paper
Paracrine TGF_IL2 paperMichael McHugh
 
Angiotensin converting enzyme 2 as COV-2 receptor
Angiotensin converting enzyme 2 as COV-2 receptorAngiotensin converting enzyme 2 as COV-2 receptor
Angiotensin converting enzyme 2 as COV-2 receptorKevin KF Ng
 
Revised B cell covid vaccine review paper
Revised B cell covid vaccine review paperRevised B cell covid vaccine review paper
Revised B cell covid vaccine review paperBartsMSBlog
 
Dr Susan E Caldwell Publication Samples
Dr Susan E Caldwell Publication SamplesDr Susan E Caldwell Publication Samples
Dr Susan E Caldwell Publication SamplesSusan E Caldwell, PhD
 
J. Virol.-2016-Patil-JVI.03090-15
J. Virol.-2016-Patil-JVI.03090-15J. Virol.-2016-Patil-JVI.03090-15
J. Virol.-2016-Patil-JVI.03090-15SHILPA PATIL
 
El medicamento aprobado por la FDA ivermectina
El medicamento aprobado por la FDA ivermectinaEl medicamento aprobado por la FDA ivermectina
El medicamento aprobado por la FDA ivermectinaDanielErnestoEscobar
 
COVID 19 Búsqueda - Dr. Freddy Flores Malpartida
COVID 19 Búsqueda - Dr. Freddy Flores MalpartidaCOVID 19 Búsqueda - Dr. Freddy Flores Malpartida
COVID 19 Búsqueda - Dr. Freddy Flores MalpartidaFreddy Flores Malpartida
 
HMH402 Alexia Bosancic Final thesis pdf
HMH402 Alexia Bosancic Final thesis pdfHMH402 Alexia Bosancic Final thesis pdf
HMH402 Alexia Bosancic Final thesis pdfAlexia Bosancic
 
COVID-19 Pandemic: The Antibody Response
COVID-19 Pandemic: The Antibody ResponseCOVID-19 Pandemic: The Antibody Response
COVID-19 Pandemic: The Antibody ResponseKevin KF Ng
 

What's hot (19)

疫苗研發之產學研合作經驗分享 以冠狀病毒奈米疫苗為例
疫苗研發之產學研合作經驗分享以冠狀病毒奈米疫苗為例疫苗研發之產學研合作經驗分享以冠狀病毒奈米疫苗為例
疫苗研發之產學研合作經驗分享 以冠狀病毒奈米疫苗為例
 
Coronavirus disease
Coronavirus disease Coronavirus disease
Coronavirus disease
 
The Yan Report
The Yan ReportThe Yan Report
The Yan Report
 
OA09.01, Cox
OA09.01, CoxOA09.01, Cox
OA09.01, Cox
 
an mrna vaccine aganist sars cov 2
an mrna vaccine aganist sars cov 2 an mrna vaccine aganist sars cov 2
an mrna vaccine aganist sars cov 2
 
JVI Paper -Shilpa Patilet al 2016
JVI Paper -Shilpa Patilet al 2016JVI Paper -Shilpa Patilet al 2016
JVI Paper -Shilpa Patilet al 2016
 
FINAL POSTER BIOTECH
FINAL POSTER BIOTECHFINAL POSTER BIOTECH
FINAL POSTER BIOTECH
 
ANA498Y_Final
ANA498Y_FinalANA498Y_Final
ANA498Y_Final
 
Patil S et al 2014 Virology
Patil S et al 2014 VirologyPatil S et al 2014 Virology
Patil S et al 2014 Virology
 
Paracrine TGF_IL2 paper
Paracrine TGF_IL2 paperParacrine TGF_IL2 paper
Paracrine TGF_IL2 paper
 
Angiotensin converting enzyme 2 as COV-2 receptor
Angiotensin converting enzyme 2 as COV-2 receptorAngiotensin converting enzyme 2 as COV-2 receptor
Angiotensin converting enzyme 2 as COV-2 receptor
 
Revised B cell covid vaccine review paper
Revised B cell covid vaccine review paperRevised B cell covid vaccine review paper
Revised B cell covid vaccine review paper
 
Dr Susan E Caldwell Publication Samples
Dr Susan E Caldwell Publication SamplesDr Susan E Caldwell Publication Samples
Dr Susan E Caldwell Publication Samples
 
Quiz (retroviruses)
Quiz (retroviruses)Quiz (retroviruses)
Quiz (retroviruses)
 
J. Virol.-2016-Patil-JVI.03090-15
J. Virol.-2016-Patil-JVI.03090-15J. Virol.-2016-Patil-JVI.03090-15
J. Virol.-2016-Patil-JVI.03090-15
 
El medicamento aprobado por la FDA ivermectina
El medicamento aprobado por la FDA ivermectinaEl medicamento aprobado por la FDA ivermectina
El medicamento aprobado por la FDA ivermectina
 
COVID 19 Búsqueda - Dr. Freddy Flores Malpartida
COVID 19 Búsqueda - Dr. Freddy Flores MalpartidaCOVID 19 Búsqueda - Dr. Freddy Flores Malpartida
COVID 19 Búsqueda - Dr. Freddy Flores Malpartida
 
HMH402 Alexia Bosancic Final thesis pdf
HMH402 Alexia Bosancic Final thesis pdfHMH402 Alexia Bosancic Final thesis pdf
HMH402 Alexia Bosancic Final thesis pdf
 
COVID-19 Pandemic: The Antibody Response
COVID-19 Pandemic: The Antibody ResponseCOVID-19 Pandemic: The Antibody Response
COVID-19 Pandemic: The Antibody Response
 

Similar to Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19

Immunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfImmunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfstudy help
 
Immunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfImmunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfstudy help
 
Sars cov 2 spike mutations paper los alamos
Sars cov 2 spike mutations paper los alamosSars cov 2 spike mutations paper los alamos
Sars cov 2 spike mutations paper los alamosJauru Freitas
 
COVID-19; Updates on Pathophysiology and Laboratory Investigations
COVID-19; Updates on Pathophysiology and Laboratory InvestigationsCOVID-19; Updates on Pathophysiology and Laboratory Investigations
COVID-19; Updates on Pathophysiology and Laboratory InvestigationsAbdulsalam Al-Ani
 
Monitoring Cellular Immune Response in Real Time with Next Generation Immunoa...
Monitoring Cellular Immune Response in Real Time with Next Generation Immunoa...Monitoring Cellular Immune Response in Real Time with Next Generation Immunoa...
Monitoring Cellular Immune Response in Real Time with Next Generation Immunoa...InsideScientific
 
Annals of Clinical and Medical Case Reports - Acmcasereport
Annals of Clinical and Medical Case Reports - AcmcasereportAnnals of Clinical and Medical Case Reports - Acmcasereport
Annals of Clinical and Medical Case Reports - Acmcasereportsemualkaira
 
COVID-19 IMMUNOPETHOGENESIS & UPCOMING IMMUNO THERAPIES
COVID-19 IMMUNOPETHOGENESIS & UPCOMING IMMUNO THERAPIESCOVID-19 IMMUNOPETHOGENESIS & UPCOMING IMMUNO THERAPIES
COVID-19 IMMUNOPETHOGENESIS & UPCOMING IMMUNO THERAPIESDharm Desai
 
Corona update 6 CORONA INDIA IN JULY
Corona update 6 CORONA INDIA IN JULYCorona update 6 CORONA INDIA IN JULY
Corona update 6 CORONA INDIA IN JULYNARENDRA MALHOTRA
 
Research and Treatment of COVID-19 - EUCYT
Research and Treatment of COVID-19 - EUCYTResearch and Treatment of COVID-19 - EUCYT
Research and Treatment of COVID-19 - EUCYTJensonAlbert
 
Immune Responses To The Pandemic New Coronavirus (COVID-19)by Prof. Mohamed L...
Immune Responses To The Pandemic New Coronavirus (COVID-19)by Prof. Mohamed L...Immune Responses To The Pandemic New Coronavirus (COVID-19)by Prof. Mohamed L...
Immune Responses To The Pandemic New Coronavirus (COVID-19)by Prof. Mohamed L...Prof. Mohamed Labib Salem
 
Awareness Talk on Novel Coronavirus, COVID-19 by Shambaditya Goswami, NIMS Un...
Awareness Talk on Novel Coronavirus, COVID-19 by Shambaditya Goswami, NIMS Un...Awareness Talk on Novel Coronavirus, COVID-19 by Shambaditya Goswami, NIMS Un...
Awareness Talk on Novel Coronavirus, COVID-19 by Shambaditya Goswami, NIMS Un...Shambaditya Goswami
 
Vaksinasi Booster COVID-19 di Indonesia 27 Juli 2022.pptx
Vaksinasi Booster COVID-19 di Indonesia 27 Juli 2022.pptxVaksinasi Booster COVID-19 di Indonesia 27 Juli 2022.pptx
Vaksinasi Booster COVID-19 di Indonesia 27 Juli 2022.pptxNuryNusdwinuringtyas1
 
Covid19 emerging data and guidance
Covid19 emerging data and guidanceCovid19 emerging data and guidance
Covid19 emerging data and guidanceAhmed Al Montasir
 
CURCUMET CAPSULES: A Natural Immunomodulator Helps to Prevent Covid-19 Infection
CURCUMET CAPSULES: A Natural Immunomodulator Helps to Prevent Covid-19 InfectionCURCUMET CAPSULES: A Natural Immunomodulator Helps to Prevent Covid-19 Infection
CURCUMET CAPSULES: A Natural Immunomodulator Helps to Prevent Covid-19 InfectionAssociate Professor in VSB Coimbatore
 
Triglytza: Counter-Regulation of RAAS and COVID-19 Treatment
Triglytza: Counter-Regulation of RAAS and COVID-19 TreatmentTriglytza: Counter-Regulation of RAAS and COVID-19 Treatment
Triglytza: Counter-Regulation of RAAS and COVID-19 TreatmentRavi Kumar, Ph.D.
 
S41421 020-0156-0
S41421 020-0156-0S41421 020-0156-0
S41421 020-0156-0gisa_legal
 
Simulated Sunlight Rapidly Inactivates SARS-CoV-2 on Surfaces
Simulated Sunlight Rapidly Inactivates SARS-CoV-2 on SurfacesSimulated Sunlight Rapidly Inactivates SARS-CoV-2 on Surfaces
Simulated Sunlight Rapidly Inactivates SARS-CoV-2 on SurfacesValentina Corona
 
Lymphocytopenia and COVID19 A Literature Review
Lymphocytopenia and COVID19 A Literature ReviewLymphocytopenia and COVID19 A Literature Review
Lymphocytopenia and COVID19 A Literature Reviewijtsrd
 

Similar to Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19 (20)

Immunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfImmunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdf
 
Immunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdfImmunobiology Abstract and Figure writing instructions.pdf
Immunobiology Abstract and Figure writing instructions.pdf
 
Sars cov 2 spike mutations paper los alamos
Sars cov 2 spike mutations paper los alamosSars cov 2 spike mutations paper los alamos
Sars cov 2 spike mutations paper los alamos
 
COVID-19; Updates on Pathophysiology and Laboratory Investigations
COVID-19; Updates on Pathophysiology and Laboratory InvestigationsCOVID-19; Updates on Pathophysiology and Laboratory Investigations
COVID-19; Updates on Pathophysiology and Laboratory Investigations
 
Monitoring Cellular Immune Response in Real Time with Next Generation Immunoa...
Monitoring Cellular Immune Response in Real Time with Next Generation Immunoa...Monitoring Cellular Immune Response in Real Time with Next Generation Immunoa...
Monitoring Cellular Immune Response in Real Time with Next Generation Immunoa...
 
Ms academy
Ms academyMs academy
Ms academy
 
Annals of Clinical and Medical Case Reports - Acmcasereport
Annals of Clinical and Medical Case Reports - AcmcasereportAnnals of Clinical and Medical Case Reports - Acmcasereport
Annals of Clinical and Medical Case Reports - Acmcasereport
 
COVID-19 IMMUNOPETHOGENESIS & UPCOMING IMMUNO THERAPIES
COVID-19 IMMUNOPETHOGENESIS & UPCOMING IMMUNO THERAPIESCOVID-19 IMMUNOPETHOGENESIS & UPCOMING IMMUNO THERAPIES
COVID-19 IMMUNOPETHOGENESIS & UPCOMING IMMUNO THERAPIES
 
Corona update 6 CORONA INDIA IN JULY
Corona update 6 CORONA INDIA IN JULYCorona update 6 CORONA INDIA IN JULY
Corona update 6 CORONA INDIA IN JULY
 
Protocol for Medicine and Technology for COVID-19 - A Mini Review
Protocol for Medicine and Technology for COVID-19 - A Mini ReviewProtocol for Medicine and Technology for COVID-19 - A Mini Review
Protocol for Medicine and Technology for COVID-19 - A Mini Review
 
Research and Treatment of COVID-19 - EUCYT
Research and Treatment of COVID-19 - EUCYTResearch and Treatment of COVID-19 - EUCYT
Research and Treatment of COVID-19 - EUCYT
 
Immune Responses To The Pandemic New Coronavirus (COVID-19)by Prof. Mohamed L...
Immune Responses To The Pandemic New Coronavirus (COVID-19)by Prof. Mohamed L...Immune Responses To The Pandemic New Coronavirus (COVID-19)by Prof. Mohamed L...
Immune Responses To The Pandemic New Coronavirus (COVID-19)by Prof. Mohamed L...
 
Awareness Talk on Novel Coronavirus, COVID-19 by Shambaditya Goswami, NIMS Un...
Awareness Talk on Novel Coronavirus, COVID-19 by Shambaditya Goswami, NIMS Un...Awareness Talk on Novel Coronavirus, COVID-19 by Shambaditya Goswami, NIMS Un...
Awareness Talk on Novel Coronavirus, COVID-19 by Shambaditya Goswami, NIMS Un...
 
Vaksinasi Booster COVID-19 di Indonesia 27 Juli 2022.pptx
Vaksinasi Booster COVID-19 di Indonesia 27 Juli 2022.pptxVaksinasi Booster COVID-19 di Indonesia 27 Juli 2022.pptx
Vaksinasi Booster COVID-19 di Indonesia 27 Juli 2022.pptx
 
Covid19 emerging data and guidance
Covid19 emerging data and guidanceCovid19 emerging data and guidance
Covid19 emerging data and guidance
 
CURCUMET CAPSULES: A Natural Immunomodulator Helps to Prevent Covid-19 Infection
CURCUMET CAPSULES: A Natural Immunomodulator Helps to Prevent Covid-19 InfectionCURCUMET CAPSULES: A Natural Immunomodulator Helps to Prevent Covid-19 Infection
CURCUMET CAPSULES: A Natural Immunomodulator Helps to Prevent Covid-19 Infection
 
Triglytza: Counter-Regulation of RAAS and COVID-19 Treatment
Triglytza: Counter-Regulation of RAAS and COVID-19 TreatmentTriglytza: Counter-Regulation of RAAS and COVID-19 Treatment
Triglytza: Counter-Regulation of RAAS and COVID-19 Treatment
 
S41421 020-0156-0
S41421 020-0156-0S41421 020-0156-0
S41421 020-0156-0
 
Simulated Sunlight Rapidly Inactivates SARS-CoV-2 on Surfaces
Simulated Sunlight Rapidly Inactivates SARS-CoV-2 on SurfacesSimulated Sunlight Rapidly Inactivates SARS-CoV-2 on Surfaces
Simulated Sunlight Rapidly Inactivates SARS-CoV-2 on Surfaces
 
Lymphocytopenia and COVID19 A Literature Review
Lymphocytopenia and COVID19 A Literature ReviewLymphocytopenia and COVID19 A Literature Review
Lymphocytopenia and COVID19 A Literature Review
 

More from Guy Boulianne

FBI - Document anti-catholique
FBI - Document anti-catholiqueFBI - Document anti-catholique
FBI - Document anti-catholiqueGuy Boulianne
 
Tableau de la coercition de Biderman
Tableau de la coercition de BidermanTableau de la coercition de Biderman
Tableau de la coercition de BidermanGuy Boulianne
 
Krakivski Visti and the Jews, 1943
Krakivski Visti and the Jews, 1943Krakivski Visti and the Jews, 1943
Krakivski Visti and the Jews, 1943Guy Boulianne
 
John F. Kennedy - Address Before the Canadian Parliament in Ottawa (May 17, 1...
John F. Kennedy - Address Before the Canadian Parliament in Ottawa (May 17, 1...John F. Kennedy - Address Before the Canadian Parliament in Ottawa (May 17, 1...
John F. Kennedy - Address Before the Canadian Parliament in Ottawa (May 17, 1...Guy Boulianne
 
John F. Kennedy -Press copy of The President and the Press, 27 April 1961
John F. Kennedy -Press copy of The President and the Press, 27 April 1961John F. Kennedy -Press copy of The President and the Press, 27 April 1961
John F. Kennedy -Press copy of The President and the Press, 27 April 1961Guy Boulianne
 
Programme de l'Union Nationale (1966)
Programme de l'Union Nationale (1966)Programme de l'Union Nationale (1966)
Programme de l'Union Nationale (1966)Guy Boulianne
 
DEFCON-21 - How to Hack Your Mini Cooper, by Jason Staggs
DEFCON-21 - How to Hack Your Mini Cooper, by Jason StaggsDEFCON-21 - How to Hack Your Mini Cooper, by Jason Staggs
DEFCON-21 - How to Hack Your Mini Cooper, by Jason StaggsGuy Boulianne
 
How to Hack Your Mini Cooper. Reverse Engineering CAN Messages on Passenger A...
How to Hack Your Mini Cooper. Reverse Engineering CAN Messages on Passenger A...How to Hack Your Mini Cooper. Reverse Engineering CAN Messages on Passenger A...
How to Hack Your Mini Cooper. Reverse Engineering CAN Messages on Passenger A...Guy Boulianne
 
Big Brother NSA and Its "Little Brothers"
Big Brother NSA and Its "Little Brothers"Big Brother NSA and Its "Little Brothers"
Big Brother NSA and Its "Little Brothers"Guy Boulianne
 
Décision disciplinaire de l’Ordre des médecins du Québec - No. 24-2018-01040
Décision disciplinaire de l’Ordre des médecins du Québec - No. 24-2018-01040Décision disciplinaire de l’Ordre des médecins du Québec - No. 24-2018-01040
Décision disciplinaire de l’Ordre des médecins du Québec - No. 24-2018-01040Guy Boulianne
 
Conspirators Hierarchy: The Story of the Committee of 300, by John Coleman
Conspirators Hierarchy: The Story of the Committee of 300, by John ColemanConspirators Hierarchy: The Story of the Committee of 300, by John Coleman
Conspirators Hierarchy: The Story of the Committee of 300, by John ColemanGuy Boulianne
 
Ordonnance du tribunal de Pesaro d’analyser le contenu des vaccins ARNm - Rés...
Ordonnance du tribunal de Pesaro d’analyser le contenu des vaccins ARNm - Rés...Ordonnance du tribunal de Pesaro d’analyser le contenu des vaccins ARNm - Rés...
Ordonnance du tribunal de Pesaro d’analyser le contenu des vaccins ARNm - Rés...Guy Boulianne
 
COVID-Period Mass Vaccination Campaign and Public Health Disaster in the USA
COVID-Period Mass Vaccination Campaign and Public Health Disaster in the USACOVID-Period Mass Vaccination Campaign and Public Health Disaster in the USA
COVID-Period Mass Vaccination Campaign and Public Health Disaster in the USAGuy Boulianne
 
The Truth about mRNA Vaccines, by Raffaele Ansovini
The Truth about mRNA Vaccines, by Raffaele AnsoviniThe Truth about mRNA Vaccines, by Raffaele Ansovini
The Truth about mRNA Vaccines, by Raffaele AnsoviniGuy Boulianne
 
Functionality and Clinical Effects of Anti-Cov2 Vaccines (Aka Mrna) And Integ...
Functionality and Clinical Effects of Anti-Cov2 Vaccines (Aka Mrna) And Integ...Functionality and Clinical Effects of Anti-Cov2 Vaccines (Aka Mrna) And Integ...
Functionality and Clinical Effects of Anti-Cov2 Vaccines (Aka Mrna) And Integ...Guy Boulianne
 
Evidence for a connection between coronavirus disease-19 and exposure to radi...
Evidence for a connection between coronavirus disease-19 and exposure to radi...Evidence for a connection between coronavirus disease-19 and exposure to radi...
Evidence for a connection between coronavirus disease-19 and exposure to radi...Guy Boulianne
 
Le mouvement conspirationniste au Québec
Le mouvement conspirationniste au QuébecLe mouvement conspirationniste au Québec
Le mouvement conspirationniste au QuébecGuy Boulianne
 
Rapport de mort du vaccin, par Dr Vladimir Zelenko
Rapport de mort du vaccin, par Dr Vladimir ZelenkoRapport de mort du vaccin, par Dr Vladimir Zelenko
Rapport de mort du vaccin, par Dr Vladimir ZelenkoGuy Boulianne
 
Memorandum of conversation between Mikhail Gorbachev and James Baker in Moscow
Memorandum of conversation between Mikhail Gorbachev and James Baker in MoscowMemorandum of conversation between Mikhail Gorbachev and James Baker in Moscow
Memorandum of conversation between Mikhail Gorbachev and James Baker in MoscowGuy Boulianne
 
MindWar, by Michael A. Aquino
MindWar, by Michael A. AquinoMindWar, by Michael A. Aquino
MindWar, by Michael A. AquinoGuy Boulianne
 

More from Guy Boulianne (20)

FBI - Document anti-catholique
FBI - Document anti-catholiqueFBI - Document anti-catholique
FBI - Document anti-catholique
 
Tableau de la coercition de Biderman
Tableau de la coercition de BidermanTableau de la coercition de Biderman
Tableau de la coercition de Biderman
 
Krakivski Visti and the Jews, 1943
Krakivski Visti and the Jews, 1943Krakivski Visti and the Jews, 1943
Krakivski Visti and the Jews, 1943
 
John F. Kennedy - Address Before the Canadian Parliament in Ottawa (May 17, 1...
John F. Kennedy - Address Before the Canadian Parliament in Ottawa (May 17, 1...John F. Kennedy - Address Before the Canadian Parliament in Ottawa (May 17, 1...
John F. Kennedy - Address Before the Canadian Parliament in Ottawa (May 17, 1...
 
John F. Kennedy -Press copy of The President and the Press, 27 April 1961
John F. Kennedy -Press copy of The President and the Press, 27 April 1961John F. Kennedy -Press copy of The President and the Press, 27 April 1961
John F. Kennedy -Press copy of The President and the Press, 27 April 1961
 
Programme de l'Union Nationale (1966)
Programme de l'Union Nationale (1966)Programme de l'Union Nationale (1966)
Programme de l'Union Nationale (1966)
 
DEFCON-21 - How to Hack Your Mini Cooper, by Jason Staggs
DEFCON-21 - How to Hack Your Mini Cooper, by Jason StaggsDEFCON-21 - How to Hack Your Mini Cooper, by Jason Staggs
DEFCON-21 - How to Hack Your Mini Cooper, by Jason Staggs
 
How to Hack Your Mini Cooper. Reverse Engineering CAN Messages on Passenger A...
How to Hack Your Mini Cooper. Reverse Engineering CAN Messages on Passenger A...How to Hack Your Mini Cooper. Reverse Engineering CAN Messages on Passenger A...
How to Hack Your Mini Cooper. Reverse Engineering CAN Messages on Passenger A...
 
Big Brother NSA and Its "Little Brothers"
Big Brother NSA and Its "Little Brothers"Big Brother NSA and Its "Little Brothers"
Big Brother NSA and Its "Little Brothers"
 
Décision disciplinaire de l’Ordre des médecins du Québec - No. 24-2018-01040
Décision disciplinaire de l’Ordre des médecins du Québec - No. 24-2018-01040Décision disciplinaire de l’Ordre des médecins du Québec - No. 24-2018-01040
Décision disciplinaire de l’Ordre des médecins du Québec - No. 24-2018-01040
 
Conspirators Hierarchy: The Story of the Committee of 300, by John Coleman
Conspirators Hierarchy: The Story of the Committee of 300, by John ColemanConspirators Hierarchy: The Story of the Committee of 300, by John Coleman
Conspirators Hierarchy: The Story of the Committee of 300, by John Coleman
 
Ordonnance du tribunal de Pesaro d’analyser le contenu des vaccins ARNm - Rés...
Ordonnance du tribunal de Pesaro d’analyser le contenu des vaccins ARNm - Rés...Ordonnance du tribunal de Pesaro d’analyser le contenu des vaccins ARNm - Rés...
Ordonnance du tribunal de Pesaro d’analyser le contenu des vaccins ARNm - Rés...
 
COVID-Period Mass Vaccination Campaign and Public Health Disaster in the USA
COVID-Period Mass Vaccination Campaign and Public Health Disaster in the USACOVID-Period Mass Vaccination Campaign and Public Health Disaster in the USA
COVID-Period Mass Vaccination Campaign and Public Health Disaster in the USA
 
The Truth about mRNA Vaccines, by Raffaele Ansovini
The Truth about mRNA Vaccines, by Raffaele AnsoviniThe Truth about mRNA Vaccines, by Raffaele Ansovini
The Truth about mRNA Vaccines, by Raffaele Ansovini
 
Functionality and Clinical Effects of Anti-Cov2 Vaccines (Aka Mrna) And Integ...
Functionality and Clinical Effects of Anti-Cov2 Vaccines (Aka Mrna) And Integ...Functionality and Clinical Effects of Anti-Cov2 Vaccines (Aka Mrna) And Integ...
Functionality and Clinical Effects of Anti-Cov2 Vaccines (Aka Mrna) And Integ...
 
Evidence for a connection between coronavirus disease-19 and exposure to radi...
Evidence for a connection between coronavirus disease-19 and exposure to radi...Evidence for a connection between coronavirus disease-19 and exposure to radi...
Evidence for a connection between coronavirus disease-19 and exposure to radi...
 
Le mouvement conspirationniste au Québec
Le mouvement conspirationniste au QuébecLe mouvement conspirationniste au Québec
Le mouvement conspirationniste au Québec
 
Rapport de mort du vaccin, par Dr Vladimir Zelenko
Rapport de mort du vaccin, par Dr Vladimir ZelenkoRapport de mort du vaccin, par Dr Vladimir Zelenko
Rapport de mort du vaccin, par Dr Vladimir Zelenko
 
Memorandum of conversation between Mikhail Gorbachev and James Baker in Moscow
Memorandum of conversation between Mikhail Gorbachev and James Baker in MoscowMemorandum of conversation between Mikhail Gorbachev and James Baker in Moscow
Memorandum of conversation between Mikhail Gorbachev and James Baker in Moscow
 
MindWar, by Michael A. Aquino
MindWar, by Michael A. AquinoMindWar, by Michael A. Aquino
MindWar, by Michael A. Aquino
 

Recently uploaded

Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?Patrick Diehl
 
Evidences of Evolution General Biology 2
Evidences of Evolution General Biology 2Evidences of Evolution General Biology 2
Evidences of Evolution General Biology 2John Carlo Rollon
 
Behavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdfBehavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdfSELF-EXPLANATORY
 
TOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physicsTOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physicsssuserddc89b
 
Temporomandibular joint Muscles of Mastication
Temporomandibular joint Muscles of MasticationTemporomandibular joint Muscles of Mastication
Temporomandibular joint Muscles of Masticationvidulajaib
 
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝soniya singh
 
Solution chemistry, Moral and Normal solutions
Solution chemistry, Moral and Normal solutionsSolution chemistry, Moral and Normal solutions
Solution chemistry, Moral and Normal solutionsHajira Mahmood
 
Transposable elements in prokaryotes.ppt
Transposable elements in prokaryotes.pptTransposable elements in prokaryotes.ppt
Transposable elements in prokaryotes.pptArshadWarsi13
 
Dashanga agada a formulation of Agada tantra dealt in 3 Rd year bams agada tanta
Dashanga agada a formulation of Agada tantra dealt in 3 Rd year bams agada tantaDashanga agada a formulation of Agada tantra dealt in 3 Rd year bams agada tanta
Dashanga agada a formulation of Agada tantra dealt in 3 Rd year bams agada tantaPraksha3
 
‏‏VIRUS - 123455555555555555555555555555555555555555
‏‏VIRUS -  123455555555555555555555555555555555555555‏‏VIRUS -  123455555555555555555555555555555555555555
‏‏VIRUS - 123455555555555555555555555555555555555555kikilily0909
 
Analytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptxAnalytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptxSwapnil Therkar
 
zoogeography of pakistan.pptx fauna of Pakistan
zoogeography of pakistan.pptx fauna of Pakistanzoogeography of pakistan.pptx fauna of Pakistan
zoogeography of pakistan.pptx fauna of Pakistanzohaibmir069
 
LIGHT-PHENOMENA-BY-CABUALDIONALDOPANOGANCADIENTE-CONDEZA (1).pptx
LIGHT-PHENOMENA-BY-CABUALDIONALDOPANOGANCADIENTE-CONDEZA (1).pptxLIGHT-PHENOMENA-BY-CABUALDIONALDOPANOGANCADIENTE-CONDEZA (1).pptx
LIGHT-PHENOMENA-BY-CABUALDIONALDOPANOGANCADIENTE-CONDEZA (1).pptxmalonesandreagweneth
 
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptxTHE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptxNandakishor Bhaurao Deshmukh
 
Twin's paradox experiment is a meassurement of the extra dimensions.pptx
Twin's paradox experiment is a meassurement of the extra dimensions.pptxTwin's paradox experiment is a meassurement of the extra dimensions.pptx
Twin's paradox experiment is a meassurement of the extra dimensions.pptxEran Akiva Sinbar
 
insect anatomy and insect body wall and their physiology
insect anatomy and insect body wall and their  physiologyinsect anatomy and insect body wall and their  physiology
insect anatomy and insect body wall and their physiologyDrAnita Sharma
 
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...lizamodels9
 
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfAnalytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfSwapnil Therkar
 
Manassas R - Parkside Middle School 🌎🏫
Manassas R - Parkside Middle School 🌎🏫Manassas R - Parkside Middle School 🌎🏫
Manassas R - Parkside Middle School 🌎🏫qfactory1
 

Recently uploaded (20)

Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?
 
Evidences of Evolution General Biology 2
Evidences of Evolution General Biology 2Evidences of Evolution General Biology 2
Evidences of Evolution General Biology 2
 
Behavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdfBehavioral Disorder: Schizophrenia & it's Case Study.pdf
Behavioral Disorder: Schizophrenia & it's Case Study.pdf
 
TOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physicsTOPIC 8 Temperature and Heat.pdf physics
TOPIC 8 Temperature and Heat.pdf physics
 
Temporomandibular joint Muscles of Mastication
Temporomandibular joint Muscles of MasticationTemporomandibular joint Muscles of Mastication
Temporomandibular joint Muscles of Mastication
 
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
 
Solution chemistry, Moral and Normal solutions
Solution chemistry, Moral and Normal solutionsSolution chemistry, Moral and Normal solutions
Solution chemistry, Moral and Normal solutions
 
Transposable elements in prokaryotes.ppt
Transposable elements in prokaryotes.pptTransposable elements in prokaryotes.ppt
Transposable elements in prokaryotes.ppt
 
Dashanga agada a formulation of Agada tantra dealt in 3 Rd year bams agada tanta
Dashanga agada a formulation of Agada tantra dealt in 3 Rd year bams agada tantaDashanga agada a formulation of Agada tantra dealt in 3 Rd year bams agada tanta
Dashanga agada a formulation of Agada tantra dealt in 3 Rd year bams agada tanta
 
‏‏VIRUS - 123455555555555555555555555555555555555555
‏‏VIRUS -  123455555555555555555555555555555555555555‏‏VIRUS -  123455555555555555555555555555555555555555
‏‏VIRUS - 123455555555555555555555555555555555555555
 
Analytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptxAnalytical Profile of Coleus Forskohlii | Forskolin .pptx
Analytical Profile of Coleus Forskohlii | Forskolin .pptx
 
zoogeography of pakistan.pptx fauna of Pakistan
zoogeography of pakistan.pptx fauna of Pakistanzoogeography of pakistan.pptx fauna of Pakistan
zoogeography of pakistan.pptx fauna of Pakistan
 
LIGHT-PHENOMENA-BY-CABUALDIONALDOPANOGANCADIENTE-CONDEZA (1).pptx
LIGHT-PHENOMENA-BY-CABUALDIONALDOPANOGANCADIENTE-CONDEZA (1).pptxLIGHT-PHENOMENA-BY-CABUALDIONALDOPANOGANCADIENTE-CONDEZA (1).pptx
LIGHT-PHENOMENA-BY-CABUALDIONALDOPANOGANCADIENTE-CONDEZA (1).pptx
 
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptxTHE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
 
Twin's paradox experiment is a meassurement of the extra dimensions.pptx
Twin's paradox experiment is a meassurement of the extra dimensions.pptxTwin's paradox experiment is a meassurement of the extra dimensions.pptx
Twin's paradox experiment is a meassurement of the extra dimensions.pptx
 
insect anatomy and insect body wall and their physiology
insect anatomy and insect body wall and their  physiologyinsect anatomy and insect body wall and their  physiology
insect anatomy and insect body wall and their physiology
 
Hot Sexy call girls in Moti Nagar,🔝 9953056974 🔝 escort Service
Hot Sexy call girls in  Moti Nagar,🔝 9953056974 🔝 escort ServiceHot Sexy call girls in  Moti Nagar,🔝 9953056974 🔝 escort Service
Hot Sexy call girls in Moti Nagar,🔝 9953056974 🔝 escort Service
 
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...
Best Call Girls In Sector 29 Gurgaon❤️8860477959 EscorTs Service In 24/7 Delh...
 
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdfAnalytical Profile of Coleus Forskohlii | Forskolin .pdf
Analytical Profile of Coleus Forskohlii | Forskolin .pdf
 
Manassas R - Parkside Middle School 🌎🏫
Manassas R - Parkside Middle School 🌎🏫Manassas R - Parkside Middle School 🌎🏫
Manassas R - Parkside Middle School 🌎🏫
 

Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19

  • 1. Article Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19 Graphical Abstract Highlights d Acute-phase SARS-CoV-2-specific T cells display an activated cytotoxic phenotype d Convalescent-phase SARS-CoV-2-specific T cells generate broad responses d Polyfunctional SARS-CoV-2-specific T cells also occur in seronegative individuals Authors Takuya Sekine, André Perez-Potti, Olga Rivera-Ballesteros, ..., Hans-Gustaf Ljunggren, Soo Aleman, Marcus Buggert Correspondence marcus.buggert@ki.se In Brief Sekine et al. provide a functional and phenotypic map of SARS-CoV-2-specific T cells across the full spectrum of exposure, infection, and COVID-19 severity. They observe that SARS-CoV-2 elicits broadly directed and functionally replete memory T cells that may protect against recurrent episodes of severe COVID-19. Sekine et al., 2020, Cell 183, 158–168 October 1, 2020 ª 2020 The Author(s). Published by Elsevier Inc. https://doi.org/10.1016/j.cell.2020.08.017 ll
  • 2. Article Robust T Cell Immunity in Convalescent Individuals with Asymptomatic or Mild COVID-19 Takuya Sekine,1,15 André Perez-Potti,1,15 Olga Rivera-Ballesteros,1,15 Kristoffer Strålin,2,16 Jean-Baptiste Gorin,1,16 Annika Olsson,2 Sian Llewellyn-Lacey,3 Habiba Kamal,2 Gordana Bogdanovic,4,13 Sandra Muschiol,4,13 David J. Wullimann,1 Tobias Kammann,1 Johanna Emgård,1 Tiphaine Parrot,1 Elin Folkesson,2 Karolinska COVID-19 Study Group, Olav Rooyackers,5,6 Lars I. Eriksson,6,7 Jan-Inge Henter,8,14 Anders Sönnerborg,2,9 Tobias Allander,4,9,13 Jan Albert,4,9,13 Morten Nielsen,10,11 Jonas Klingström,1 Sara Gredmark-Russ,1,2 Niklas K. Björkström,1 Johan K. Sandberg,1 David A. Price,3,12 Hans-Gustaf Ljunggren,1,15 Soo Aleman,1,2,15 and Marcus Buggert1,15,17,* 1Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden 2Division of Infectious Diseases, Karolinska University Hospital, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden 3Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK 4Division of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden 5Department of Clinical Interventions and Technology, Karolinska Institutet, Stockholm, Sweden 6Function Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden 7Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden 8Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden 9Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden 10Department of Health Technology, Technical University of Denmark, Lyngby, Denmark 11Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martı́n, San Martı́n, Argentina 12Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK 13Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden 14Theme of Children’s and Women’s Health, Karolinska University Hospital, Stockholm, Sweden 15These authors contributed equally 16These authors contributed equally 17Lead Contact *Correspondence: marcus.buggert@ki.se https://doi.org/10.1016/j.cell.2020.08.017 SUMMARY SARS-CoV-2-specific memory T cells will likely prove critical for long-term immune protection against COVID-19. Here, we systematically mapped the functional and phenotypic landscape of SARS-CoV-2-spe- cific T cell responses in unexposed individuals, exposed family members, and individuals with acute or convalescent COVID-19. Acute-phase SARS-CoV-2-specific T cells displayed a highly activated cytotoxic phenotype that correlated with various clinical markers of disease severity, whereas convalescent-phase SARS-CoV-2-specific T cells were polyfunctional and displayed a stem-like memory phenotype. Importantly, SARS-CoV-2-specific T cells were detectable in antibody-seronegative exposed family members and conva- lescent individuals with a history of asymptomatic and mild COVID-19. Our collective dataset shows that SARS-CoV-2 elicits broadly directed and functionally replete memory T cell responses, suggesting that nat- ural exposure or infection may prevent recurrent episodes of severe COVID-19. INTRODUCTION The world changed in December 2019 with the emergence of a new zoonotic pathogen, severe acute respiratory syndrome co- ronavirus 2 (SARS-CoV-2), which causes a variety of clinical syn- dromes collectively termed coronavirus disease 2019 (COVID- 19). At present, there is no vaccine against SARS-CoV-2, and the excessive inflammation associated with severe COVID-19 can lead to respiratory failure, septic shock, and ultimately, death (Guan et al., 2020; Wolfel et al., 2020; Wu and McGoogan, 2020). The overall mortality rate is 0.5%–3.5% (Guan et al., 2020; Wolfel et al., 2020; Wu and McGoogan, 2020). However, most people seem to be affected less severely and remain asymptom- atic or develop only mild symptoms during COVID-19 (He et al., 2020b; Wei et al., 2020; Yang et al., 2020). It will therefore be crit- ical for public health reasons to determine whether people with milder forms of COVID-19 develop robust immunity against SARS-CoV-2. Global efforts are currently underway to map the determi- nants of immune protection against SARS-CoV-2. Recent data have shown that SARS-CoV-2 infection generates near-com- plete protection against rechallenge in rhesus macaques ll OPEN ACCESS 158 Cell 183, 158–168, October 1, 2020 ª 2020 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
  • 3. A 0 10 20 30 40 50 % of memory CD4+ T cells CD38 0 1 2 3 4 5 CD69 0 5 10 15 20 25 Ki-67 2020 BD MC AM AS 0 20 40 60 80 100 PD-1 0 20 40 60 80 100 CD38 % of memory CD8+ T cells 0 10 20 30 CD39 0 5 10 15 20 CD69 0 5 10 15 CTLA-4 0 10 20 30 40 50 HLA-DR 0 10 20 30 Ki-67 0 5 10 15 LAG-3 0 5 10 15 TIM-3 -6 -4 -2 0 2 4 6 PC2 -2 0 2 4 6 8 PC2 Memory CD4+ T cells Memory CD8+ T cells Ki-67 CD38 TIM-3 HLA-DR PD-1 CD69 CD39 RA CTLA-4 CXCR5 CD25 CD27 CCR7 CD28 TIGIT TCF1 CD127 LAG-3 Perforin TOX 2B4 GzmB CD38 CTLA-4 Ki-67CD39 LAG-3 TIM-3 HLA-DR PD-1 CXCR5 CD69 CCR7 CD27 CD25 CD28 CD127 TIGIT Perforin TOX GzmB 2B4 RA TCF1 0 1000000 2000000 3000000 Abs count T cells 0 10 20 30 40 %CD3+CD15-CD14- CD19- of CD45+ % T cells 200000 400000 600000 800000 2000000 Abs count CD8+ T cells 0 500000 1000000 1500000 2000000 Abs count CD4+ T cells 0 5 10 20 % CD8+ T cells 0 10 20 30 % CD4+ T cells 0 %CD3+CD8+ of CD45+ %CD3+CD8+ of CD45+ B C 2020 BD AM AS 1 1 <1 30 27 14 2020 BD AS Gated on memory CD8+ T cells 0 20 40 60 0 10 20 30 40 0 5 10 15 20 CD38 Ki-67 HLA-DR PD-1 CD38+PD-1+ CD38+HLA-DR+ CD38+Ki-67+ % of memory CD8+ T cells E 2020 BD MC AM AS UMAP2 UMAP1 D HLA-DR Ki-67 CD38 LAG-3 TIM-3 CD39 2B4 TIGIT PD-1 CD27 CD127 CXCR5 GzmB Perforin CD95 CD28 CD45RA CCR7 Gated on memory CD8+ T cells Max Min CTLA-4 CD25 CD69 TOX TCF1 2020 BD MC AM AS * Cycling TEMRA TEM TCM -2 0 2 4 6 8 PC2 (22 %) -5 5 2.5 0 -2.5 10 7.5 PC1 (34 %) -2 0 2 4 6 8 PC2 (22 %) -5 5 2.5 0 -2.5 10 7.5 PC1 (34 %) -6 -4 -2 0 2 4 PC2 (22 %) 6 4 2 0 -2 10 6 PC1 (34 %) 8 -6 -4 -2 0 2 4 PC2 (22 %) 6 4 2 0 -2 10 6 PC1 (34 %) 8 *** *** *** ** *** *** *** ** *** * *** *** *** * *** * *** ** *** *** * ** *** *** *** *** *** ** ** *** *** *** *** *** *** ** *** *** * * ** ** ** *** ** *** ** ** *** *** *** ** *** *** ** *** *** *** *** *** *** ** *** *** *** *** *** *** *** Figure 1. T Cell Perturbations in COVID-19 (A) Dot plots summarizing the absolute counts and relative frequencies of CD3+ (left), CD4+ (center), and CD8+ T cells (right) in healthy blood donors from 2020 (2020 BD ) and patients with acute moderate (AM ) or acute severe COVID-19 (AS). Each dot represents one donor. Data are shown as median ± IQR. *p < 0.05, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons. (B) Top: PCA plots showing the distribution and segregation of memory CD4+ and CD8+ T cells by group. Each dot represents one donor. Memory cells were defined by exclusion of naive cells (CCR7+ CD45RA+ CD95 ). Center: PCA plots showing the corresponding trajectories of key markers that influenced the group- (legend continued on next page) ll OPEN ACCESS Cell 183, 158–168, October 1, 2020 159 Article
  • 4. (Chandrashekar et al., 2020), and similarly, there is limited evi- dence of reinfection in humans with previously documented COVID-19 (Kirkcaldy et al., 2020). Further work is therefore required to define the mechanisms that underlie these observa- tions and evaluate the durability of protective immune responses elicited by primary infection with SARS-CoV-2. Most correlative studies of immune protection against SARS-CoV-2 have focused on induction of neutralizing antibodies (Hotez et al., 2020; Robbiani et al., 2020; Seydoux et al., 2020; Wang et al., 2020). However, antibody responses are not detectable in all pa- tients, especially those with less severe forms of COVID-19 (Long et al., 2020; Mallapaty, 2020; Woloshin et al., 2020). Previ- ous work has also shown that memory B cell responses tend to be short lived after infection with SARS-CoV-1 (Channappanavar et al., 2014; Tang et al., 2011). In contrast, memory T cell re- sponses can persist for many years (Le Bert et al., 2020; Tang et al., 2011; Yang et al., 2006) and, in mice, protect against lethal challenge with SARS-CoV-1 (Channappanavar et al., 2014). SARS-CoV-2-specific T cells have been identified in humans (Grifoni et al., 2020; Ni et al., 2020). It has nonetheless remained unclear to what extent various features of the T cell immune response associate with serostatus and the clinical course of COVID-19. To address this knowledge gap, we characterized SARS-CoV-2-specific CD4+ and CD8+ T cells in outcome- defined cohorts of donors (total n = 206) from Sweden, which has experienced a more open spread of COVID-19 than many other countries in Europe (Habib, 2020). RESULTS T Cell Perturbations in COVID-19 Our preliminary analyses showed that the absolute numbers and relative frequencies of CD4+ and CD8+ T cells were unphysiolog- ically low in patients with acute moderate or severe COVID-19 (Figures 1A, S1A, and S1B). This finding has been reported pre- viously (He et al., 2020a; Liu et al., 2020). We then used a 29-co- lor flow cytometry panel to assess the phenotypic landscape of these immune perturbations in direct comparisons with healthy blood donors and individuals who had recovered from mild COVID-19 acquired early during the pandemic (February–March 2020). Cohort demographics are described in the STAR Methods section. None of these variables were associated with disease severity. The following parameters were measured in each sam- ple: viability, C-C chemokine receptor type 7 (CCR7), cluster of differentiation 3 (CD3), CD4, CD8, CD14, CD19, CD25, CD27, CD28, CD38, CD39, CD45RA, CD69, CD95, CD127, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), C-X-C chemokine receptor type 5 (CXCR5), granzyme B, human leukocyte antigen (HLA)-DR, Ki-67, lymphocyte activation gene 3 (LAG-3), pro- grammed cell death protein 1 (PD-1), perforin, T cell factor 1 (TCF1), T cell immunoreceptor with immunoglobulin (Ig) and ITIM domains (TIGIT), T cell Ig and mucin domain-containing pro- tein 3 (TIM-3), thymocyte selection-associated high-mobility group box factor (TOX), and natural killer cell receptor 2B4. Unbiased principal component analysis (PCA) revealed clear segregation between memory T cells from patients with acute moderate or severe COVID-19 and memory T cells from conva- lescent individuals and healthy blood donors (Figure 1B), driven largely by expression of CD38, CD69, Ki-67, and PD-1 in the CD4+ compartment and expression of CD38, CD39, CD69, CTLA-4, HLA-DR, Ki-67, LAG-3, and TIM-3 in the CD8+ compart- ment (Figures 1B, 1C, and S1C). To extend these findings, we concatenated all memory CD4+ T cells and all memory CD8+ T cells from healthy blood donors, convalescent individuals, and patients with acute moderate or severe COVID-19. Phenotypically related cells were identified using the clustering algorithm PhenoGraph, and marker expres- sion patterns were visualized using the dimensionality reduction algorithm Uniform Manifold Approximation and Projection (UMAP). Distinct topographical clusters were apparent in each group (Figures 1D, S2A, and S2B). In particular, memory CD8+ T cells from patients with acute moderate or severe COVID-19 expressed a distinct cluster of markers associated with activa- tion and the cell cycle, including CD38, HLA-DR, Ki-67, and PD-1 (Figures 1D and S2A). A similar pattern was observed among memory CD4+ T cells from patients with acute moderate or severe COVID-19 (Figure S2B). These findings were confirmed via manual gating of the flow cytometry data (Fig- ure 1E). Correlative analyses further demonstrated that the acti- vated/cycling phenotype was strongly associated with SARS- CoV-2-specific IgG levels and various clinical parameters, including age, hemoglobin concentration, platelet count, and plasma levels of alanine aminotransferase, albumin, D-dimer, fibrinogen, and myoglobin (Figures S2C and S2D), but less strongly associated with plasma levels of various inflammatory markers, including interleukin (IL)-1b, IL-10, and tumor necrosis factor (TNF) (Table S1). Phenotypic Characteristics of SARS-CoV-2-Specific T Cells in Acute and Convalescent COVID-19 Unphysiologically high expression frequencies of CD38, poten- tially driven by a highly inflammatory environment, were defined segregation of memory CD4+ and CD8+ T cells. Bottom: dot plots showing the group-defined distribution of markers in PC2. Each dot represents one donor. MC, individuals in the convalescent phase after mild COVID-19. **p < 0.01, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons. (C) Dot plots summarizing the expression frequencies of activation/cycling markers among memory CD4+ (top) and CD8+ T cells (bottom) by group. Each dot represents one donor. Data are shown as median ± IQR. *p < 0.05, **p < 0.01, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons. (D) Top: UMAP plots showing the clustering of memory CD8+ T cells by group in relation to all memory CD8+ T cells (left). Bottom: UMAP plots showing the expression of individual markers (n = 3 donors per group). UMAP plots were based on all markers distinguished in the bottom row. (E) Left: representative flow cytometry plots showing the expression of activation/cycling markers among memory CD8+ T cells by group. Numbers indicate percentages in the drawn gates. Right: dot plots showing the expression frequencies of activation/cycling markers among memory CD8+ T cells by group. Each dot represents one donor. Data are shown as median ± IQR. Key as in (B) and (C). **p < 0.01, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons. ll OPEN ACCESS 160 Cell 183, 158–168, October 1, 2020 Article
  • 5. observed consistently among memory CD8+ T cells from pa- tients with acute moderate or severe COVID-19 (Figures S3A and S3B). In line with these data, we found that CD8+ T cells spe- cific for cytomegalovirus (CMV) or Epstein-Barr virus (EBV) more commonly expressed CD38, but not HLA-DR, Ki-67, or PD-1, in patients with acute moderate or severe COVID-19 compared with convalescent individuals and healthy blood donors, indi- cating limited bystander activation and proliferation during the early phase of infection with SARS-CoV-2 (Figures 2A, 2B, and S3C). Of note, actively proliferating CD8+ T cells, defined by expression of Ki-67, exhibited a predominant CCR7 CD27+ CD28+ CD45RA CD127 phenotype in patients with acute moderate or severe COVID-19 (Figure S3D), as reported previ- ously in the context of vaccination and other viral infections (Bug- gert et al., 2018; Miller et al., 2008). On the basis of these find- ings, we used overlapping peptides spanning the immunogenic domains of the SARS-CoV-2 spike, membrane, and nucleo- capsid proteins to stimulate peripheral blood mononuclear cells (PBMCs) from patients with acute moderate or severe COVID- 19. A vast majority of responding CD4+ and CD8+ T cells dis- played an activated/cycling (CD38+ HLA-DR+ Ki67+ PD-1+ ) phenotype (Figure 2C). These results were confirmed using an activation-induced marker (AIM) assay to measure upregulation of CD69 and 4-1BB (CD137), suggesting that most CD38+ PD-1+ CD8+ T cells were specific for SARS-CoV-2 (Figure 2D). In further experiments, we used HLA class I tetramers as probes to detect CD8+ T cells specific for the predicted optimal epitopes from SARS-CoV-2 (Figure S3E; Table S2). A vast major- ity of tetramer+ CD8+ T cells in the acute phase of infection, but not during convalescence, displayed an activated/cycling phenotype (Figure 2E). In general, early SARS-CoV-2-specific CD8+ T cell populations were characterized by expression of im- mune activation molecules (CD38, HLA-DR, and Ki-67), inhibi- tory receptors (PD-1 and TIM-3), and cytotoxic molecules (granzyme B and perforin), whereas convalescent-phase SARS-CoV-2-specific CD8+ T cell populations were skewed to- ward an early differentiated memory (CCR7+ CD127+ CD45RA /+ TCF1+ ) phenotype (Figure 2F). Importantly, the expression fre- quencies of CCR7 and CD45RA among SARS-CoV-2-specific CD8+ T cells were positively correlated with the number of symp- tom-free days after infection (CCR7: r = 0.79, p = 0.001; CD45RA: r = 0.70, p = 0.008), whereas the expression frequency of gran- zyme B among SARS-CoV-2-specific CD8+ T cells was inversely correlated with the number of symptom-free days after infection (r = 0.70, p = 0.007) (Figure 2G). Time from exposure was therefore associated with emergence of stem-like memory SARS-CoV-2- specific CD8+ T cells. Functional Characteristics of SARS-CoV-2-Specific T Cells in Convalescent COVID-19 On the basis of these observations, we quantified functional SARS-CoV-2-specific memory T cell responses across five distinct cohorts, including healthy individuals who donated blood before or during the pandemic, family members who shared a household with convalescent individuals and were exposed at the time of symptomatic disease, and individuals in the convalescent phase after mild or severe COVID-19. We de- tected potentially cross-reactive T cell responses directed against the spike and/or membrane proteins in 28% of healthy individuals who donated blood before the pandemic, consistent with previous reports (Grifoni et al., 2020; Le Bert et al., 2020), but nucleocapsid reactivity was notably absent in this cohort (Figures 3A, S4A, and S4B). The highest response frequencies against any of these three proteins were observed in convales- cent individuals who experienced severe COVID-19 (100%). Pro- gressively lower response frequencies were observed in conva- lescent individuals with a history of mild COVID-19 (87%), exposed family members (67%), and healthy individuals who donated blood during the pandemic (46%) (Figure 3A). To assess the functional capabilities of SARS-CoV-2-specific memory CD4+ and CD8+ T cells in convalescent individuals, we stimulated PBMCs with the overlapping spike, membrane, and nucleocapsid peptide sets and measured a surrogate marker of degranulation (CD107a) along with production of interferon (IFN)-g, IL-2, and TNF (Figures 3B and 3C). SARS-CoV-2-spe- cific CD4+ T cells predominantly expressed IFN-g, IL-2, and TNF (Figure 3B), whereas SARS-CoV-2-specific CD8+ T cells predominantly expressed IFN-g and mobilized CD107a (Fig- ure 3C). We then used the AIM assay to determine the functional polarization of SARS-CoV-2-specific CD4+ T cells. Interestingly, spike-specific CD4+ T cells were skewed toward a circulating T follicular helper (cTfh) profile, suggesting a key role in the gener- ation of potent antibody responses, whereas membrane-spe- cific and nucleocapsid-specific CD4+ T cells were skewed toward a Th1 or a Th1/Th17 profile (Figures 3D, S5A, and S5B). Antibody Responses and Proliferative Capabilities of SARS-CoV-2-Specific T Cells in Convalescent COVID-19 In a final series of experiments, we assessed the recall capabil- ities of SARS-CoV-2-specific CD4+ and CD8+ T cells in conva- lescent individuals, exposed family members, and healthy blood donors. Proliferative responses were identified by tracking the progressive dilution of a cytoplasmic dye (CellTrace Violet [CTV]) after stimulation with the overlapping spike, membrane, and nucleocapsid peptide sets, and functional responses to the same antigens were evaluated 5 days later by measuring the production of IFN-g (Blom et al., 2013; Buggert et al., 2014). Anamnestic responses in the CD4+ and CD8+ T cell com- partments, quantified as a function of CTVlow IFN-g+ events (Fig- ure 4A), were detected in most convalescent individuals (mild COVID-19 [MC] = 96%, severe COVID-19 [SC] = 100%) and exposed family members (92%) (Figures 4B and 4C). SARS- CoV-2-specific CD4+ T cell responses were proportionately larger overall than the corresponding SARS-CoV-2-specific CD8+ T cell responses (exposed family members = 1.8-fold, MC = 1.4-fold, SC = 1.8-fold) (Figure 4D). In addition, most IFN-g+ SARS-CoV-2-specific CD4+ T cells produced TNF, and most IFN-g+ SARS-CoV-2-specific CD8+ T cells expressed granzyme B and perforin (Figure 4E). Serological evaluations revealed a strong positive correlation between IgG responses directed against the spike protein of SARS-CoV-2 and IgG responses directed against the nucleo- capsid protein of SARS-CoV-2 (r = 0.82, p < 0.001) (Figure S5C). Moreover, SARS-CoV-2-specific CD4+ and CD8+ T cell re- sponses were present in seronegative individuals, albeit at lower frequencies compared with seropositive individuals (41% versus ll OPEN ACCESS Cell 183, 158–168, October 1, 2020 161 Article
  • 6. A C E F G D B Figure 2. Phenotypic Characteristics of SARS-CoV-2-Specific T Cells in Acute and Convalescent COVID-19 (A and B) Dot plots summarizing the expression frequencies of activation/cycling markers among tetramer+ CMV-specific (A) or EBV-specific CD8+ T cells (B) by group. Each dot represents one specificity in one donor. Data are shown as median ± IQR. *p < 0.05, **p < 0.01. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons. (C) Representative flow cytometry plots (left) and bar graphs (right) showing the expression of activation/cycling markers among CD107a+ and/or IFN-g+ SARS- CoV-2-specific CD4+ and CD8+ T cells (n = 6 donors). Numbers indicate percentages in the drawn gates. Data are shown as median ± IQR. NC, negative control. *p < 0.05, **p < 0.01. Paired t test or Wilcoxon signed-rank test. (legend continued on next page) ll OPEN ACCESS 162 Cell 183, 158–168, October 1, 2020 Article
  • 7. 99%, respectively) (Figure 4F). These discordant responses were nonetheless pronounced in some convalescent individuals with a history of mild COVID-19 (3 of 31), exposed family mem- bers (9 of 28), and healthy individuals who donated blood during the pandemic (5 of 31) (Figures 4F and S5D), often targeting the internal (nucleocapsid) and surface antigens (spike and/or mem- brane) of SARS-CoV-2 (Figure 4G). Higher frequencies of SARS- CoV-2-specific T cells were also found in exposed seronegative family members compared with unexposed donors (Figure S5E). Potent memory T cell responses were therefore elicited in the absence or presence of circulating antibodies, consistent with a non-redundant role as key determinants of immune protection against COVID-19 (Chandrashekar et al., 2020). DISCUSSION We are currently facing the biggest global health emergency in decades, namely the devastating outbreak of COVID-19. In the absence of a protective vaccine, it will be critical to determine whether exposed and/or infected people, especially those with asymptomatic or very mild forms of the disease who likely act inadvertently as major transmitters, develop robust adaptive im- munity against SARS-CoV-2 (Long et al., 2020). In this study, we used a systematic approach to map cellular and humoral immune responses against SARS-CoV-2 in patients with acute moderate or severe COVID-19, individuals in the convalescent phase after mild or severe COVID-19, exposed family members, and healthy individuals who donated blood before (2019) or during the pandemic (2020). Individuals in the convalescent phase after mild COVID-19 were traced after re- turning to Sweden from endemic areas (mostly northern Italy). These donors exhibited robust memory T cell responses months after infection, even in the absence of detectable circulating an- tibodies specific for SARS-CoV-2, that may contribute to protec- tion against severe COVID-19. We found that T cell activation, characterized by expression of CD38, was a hallmark of acute COVID-19. Similar findings have been reported previously in the absence of specificity data (Huang et al., 2020; Thevarajan et al., 2020; Wilk et al., 2020). Many of these T cells also expressed HLA-DR, Ki-67, and PD-1, indicating a combined activation/cycling phenotype, which correlated with early SARS-CoV-2-specific IgG levels and, to a lesser extent, plasma levels of various inflammatory markers. Our data also showed that many activated/cycling T cells in the acute phase were functionally replete and specific for SARS-CoV-2. Equivalent functional profiles have been observed early after immunization with successful vaccines (Blom et al., 2013; Miller et al., 2008; Precopio et al., 2007). Accordingly, the expression of multiple inhibitory receptors, including PD-1, likely indicates early activation rather than exhaustion (Zheng et al., 2020a, 2020b). Virus-specific memory T cells have been shown to persist for many years after infection with SARS-CoV-1 (Le Bert et al., 2020; Tang et al., 2011; Yang et al., 2006). In line with these ob- servations, we found that SARS-CoV-2-specific T cells acquired an early differentiated memory (CCR7+ CD127+ CD45RA /+ TCF1+ ) phenotype in the convalescent phase, as reported previ- ously in the context of other viral infections and successful vac- cines (Blom et al., 2013; Demkowicz et al., 1996; Fuertes Mar- raco et al., 2015; Precopio et al., 2007). This phenotype has been associated with stem-like properties (Betts et al., 2006; Blom et al., 2013; Demkowicz et al., 1996; Fuertes Marraco et al., 2015; Precopio et al., 2007). Accordingly, we found that SARS-CoV-2-specific T cells generated anamnestic responses to cognate antigens in the convalescent phase, characterized by extensive proliferation and polyfunctionality. Of particular note, we detected similar memory T cell responses directed against the internal (nucleocapsid) and surface proteins (mem- brane and/or spike) in some individuals lacking detectable circu- lating antibodies specific for SARS-CoV-2. Indeed, almost twice as many healthy individuals who donated blood during the pandemic had memory T cell responses versus antibody re- sponses, implying that seroprevalence as an indicator may un- derestimate the extent of adaptive immune responses against SARS-CoV-2. Our study was cross-sectional in nature and limited in terms of clinical follow-up and overall donor numbers in each outcome- defined group. It therefore remains to be determined whether robust memory T cell responses in the absence of detectable circulating antibodies can protect against severe forms of COVID-19. This scenario has nonetheless been inferred from previous studies of MERS and SARS-CoV-1 (Channappanavar et al., 2014; Li et al., 2008; Zhao et al., 2016, 2017), both of which have been shown to induce potent memory T cell responses that persist for years, in contrast to the corresponding antibody re- sponses (Alshukairi et al., 2016; Shin et al., 2019; Tang et al., 2011). Antibody responses have also been shown to wane after infection with SARS-CoV-2 (Ibarrondo et al., 2020; Long et al., 2020), suggesting that transient humoral immunity is a general feature of coronavirus infections (Callow et al., 1990). However, the fact that memory B cells (Juno et al., 2020) and memory T cells are generated in response to SARS-CoV-2 suggests that natural infection may elicit protection from severe COVID-19. In line with these observations, none of the convalescent individ- uals in this study, including those with previous mild disease, experienced further episodes of COVID-19. (D) Representative flow cytometry plots (left) and bar graph (right) showing the upregulation of CD69 and 4-1BB (AIM assay) among CD38+ PD-1+ SARS-CoV-2- specific CD8+ T cells (n = 6 donors). Numbers indicate percentages in the drawn gates. S, spike; M, membrane; N, nucleocapsid. (E) Left: representative flow cytometry plots showing the expression of activation/cycling markers among tetramer+ SARS-CoV-2-specific CD8+ T cells by group (red) and by total frequency (black). Center: UMAP plot showing the clustering of memory CD8+ T cells. Right: UMAP plots showing the clustering of tetramer+ SARS-CoV-2-specific CD8+ T cells by group and the expression of individual markers (n = 2 donors). (F) Dot plots summarizing the expression frequencies of all quantified markers among tetramer+ SARS-CoV-2-specific CD8+ T cells by group. Each dot rep- resents combined specificities in one donor. Data are shown as median ± IQR. (G) Bivariate plots showing the pairwise correlations between symptom-free days and the expression frequencies of CCR7, CD45RA, or granzyme B (GzmB). Each dot represents combined specificities in one donor. Key as in (F). Spearman rank correlation. ll OPEN ACCESS Cell 183, 158–168, October 1, 2020 163 Article
  • 8. A B C D Figure 3. Functional Characteristics of SARS-CoV-2-Specific T Cells in Convalescent COVID-19 (A) Left: dot plots summarizing the frequencies of IFN-g-producing cells responding to overlapping peptides spanning the immunogenic domains of the SARS- CoV-2 S, M, and N proteins by group (ELISpot assays). Each dot represents one donor. The dotted line indicates the cutoff for positive responses. Right: bar graph showing the frequencies of IFN-g-producing cells responding to the internal (N) and surface antigens (M and/or S) of SARS-CoV-2 by group (ELISpot assays). 2019 BD, healthy blood donors from 2019; Exp, exposed family members; SC, individuals in the convalescent phase after severe COVID-19; SFU, spot- forming unit. *p < 0.05, **p < 0.01, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons. (B) and (C) Left: representative flow cytometry plots showing the functional profiles of SARS-CoV-2-specific CD4+ (B) and CD8+ T cells (C) from a convalescent individual (group MC). Numbers indicate percentages in the drawn gates. Right: bar graphs and pie charts summarizing the distribution of individual functions among SARS-CoV-2-specific CD4+ (B) and CD8+ T cells (C) from convalescent individuals in groups MC (n = 12) or SC (n = 14). Data are shown as median ± IQR. Key as in (A). *p < 0.05. Unpaired t test or Mann-Whitney U test. (D) Left: bar graphs summarizing the functional polarization of SARS-CoV-2-specific CD4+ T cells from convalescent individuals in groups MC (n = 5) and SC (n = 6). Subsets were defined as CXCR5+ (cTfh), CCR4 CCR6 CXCR3+ CXCR5 (Th1), CCR4+ CCR6 CXCR3 CXCR5 (Th2), CCR4 CCR6+ CXCR3 CXCR5 (Th17), CCR4 CCR6+ CXCR3+ CXCR5 (Th1/17), and CCR4 CCR6 CXCR3 CXCR5 (non-Th1/2/17). Data are shown as median ± IQR. *p < 0.05. Unpaired t test or Mann-Whitney U test. Right: line graph comparing cTfh versus Th1 polarization by specificity in convalescent individuals from groups MC and SC. Each dot represents one donor. Key as in (A). *p < 0.05, **p < 0.01. Paired t test. ll OPEN ACCESS 164 Cell 183, 158–168, October 1, 2020 Article
  • 9. A B C D E F G (legend on next page) ll OPEN ACCESS Cell 183, 158–168, October 1, 2020 165 Article
  • 10. Of note, we detected cross-reactive T cell responses directed against the spike and/or membrane proteins of SARS-CoV-2 in 28% of unexposed healthy blood donors, consistent with a high degree of pre-existing immunity in the general population (Braun et al., 2020; Grifoni et al., 2020; Le Bert et al., 2020). More- over, these particular data were derived from cryopreserved samples, so this figure might be considered as a lower bound es- timate of overall prevalence (Owen et al., 2007). In this context, our findings most likely reflect widespread exposure to seasonal coronaviruses, which could shape the subsequent immune response to SARS-CoV-2. As such, it remains likely that a frac- tion of the anamnestic SARS-CoV-2-specific T cell response was initially induced by seasonal coronaviruses in seronegative individuals (Mateus et al., 2020). It is also tempting to speculate that such responses may provide at least partial protection against SARS-CoV-2, given that pre-existing T cell immunity has been associated with beneficial outcomes after challenge with the pandemic influenza virus strain H1N1 (Sridhar et al., 2013; Wilkinson et al., 2012). Collectively, our data provide a functional and phenotypic map of SARS-CoV-2-specific T cell immunity across the full spectrum of exposure, infection, and disease. The observation that many individuals with asymptomatic or mild COVID-19 had highly du- rable and functionally replete memory T cell responses, not un- commonly in the absence of detectable humoral responses, further suggests that natural exposure or infection could prevent recurrent episodes of severe COVID-19. STAR+METHODS Detailed methods are provided in the online version of this paper and include the following: d KEY RESOURCES TABLE d RESOURCE AVAILABILITY B Lead Contact B Material Availability B Data and Code Availability d EXPERIMENTAL MODEL AND SUBJECT DETAILS B Human subjects d METHOD DETAILS B Flow cytometry B Peptides B Epitope prediction B Tetramers B Functional assay B Proliferation assay B AIM assay B Trucount B Principal component analysis B UMAP B ELISpot assay B Serology d QUANTIFICATION AND STATISTICAL ANALYSIS SUPPLEMENTAL INFORMATION Supplemental Information can be found online at https://doi.org/10.1016/j. cell.2020.08.017. CONSORTIA The members of the Karolinska COVID-19 Study Group are Mira Akber, Soo Aleman, Lena Berglin, Helena Bergsten, Niklas K. Björkström, Susanna Brigh- enti, Demi Brownlie, Marcus Buggert, Marta Butrym, Benedict Chambers, Puran Chen, Martin Cornillet Jeannin, Jonathan Grip, Angelica Cuapio Gomez, Lena Dillner, Jean-Baptiste Gorin, Isabel Diaz Lozano, Majda Dzidic, Johanna Emgård, Lars I. Eriksson, Malin Flodström Tullberg, Anna Färnert, Hedvig Glans, Sara Gredmark Russ, Alvaro Haroun-Izquierdo, Elizabeth Henriksson, Laura Hertwig, Habiba Kamal, Tobias Kamann, Jonas Klingstrom, Efthymia Kokkinou, Egle Kvedaraite, Hans-Gustaf Ljunggren, Sian Llewellyn-Lacey, Marco Loreti, Magalini Lourda, Kimia Maleki, Karl-Johan Malmberg, Nicole Marquardt, Christopher Maucourant, Jakob Michaelsson, Jenny Mjösberg, Kirsten Moll, Jagadees Muva, Johan Mårtensson, Pontus Nauclér, Anna Norrby-Teglund, Annika Olsson, Laura Palma Medina, Tiphaine Parrot, Björn Persson, André Perez-Potti, Lena Radler, Emma Ringqvist, Olga Rivera-Bal- lesteros, Olav Rooyackers, Johan Sandberg, John Tyler Sandberg, Takuya Sekine, Ebba Sohlberg, Tea Soini, Kristoffer Strålin, Anders Sönnerborg, Mat- tias Svensson, Janne Tynell, Renata Varnaite, Andreas Von Kries, Christian Unge, and David Wulliman. ACKNOWLEDGMENTS We express our gratitude to all donors, health care personnel, study coordina- tors, administrators, and laboratory managers involved in this work. M.B. was supported by the Swedish Research Council, the Karolinska Institutet, the Swedish Society for Medical Research, the Jeansson Stiftelser, the Åke Wi- bergs Stiftelse, the Swedish Society of Medicine, the Swedish Cancer Society, Figure 4. Antibody Responses and Proliferation Capabilities of SARS-CoV-2-Specific T Cells in Convalescent COVID-19 (A) Representative flow cytometry plots showing the proliferation (CTV ) and functionality (IFN-g+ ) of SARS-CoV-2-specific T cells from a convalescent individual (group MC) after stimulation with overlapping peptides spanning the immunogenic domains of the SARS-CoV-2 S, M, and N proteins. Numbers indicate per- centages in the drawn gates. (B) and (C) Dot plots summarizing the frequencies of CTV IFN-g+ SARS-CoV-2-specific CD4+ (B) and CD8+ T cells (C) by group and specificity. Each dot represents one donor. The dotted line indicates the cutoff for positive responses. *p < 0.05, **p < 0.01, ***p < 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons. (D) Dot plots comparing the frequencies of CTV IFN-g+ SARS-CoV-2-specific CD4+ versus CD8+ T cells by group and specificity. Each dot represents one donor. Data are shown as median ± IQR. Key as in (B) and (C). *p < 0.05, **p < 0.01, ***p < 0.001. Paired t test or Wilcoxon signed-rank test. (E) Left: representative flow cytometry plots showing the production of IFN-g and TNF among CTV virus-specific CD4+ (top) and CD8+ T cells (bottom) from a convalescent individual (group MC). Numbers indicate percentages in the drawn gates. Right: heatmaps summarizing the functional profiles of CTV IFN-g+ virus-specific CD4+ (top) and CD8+ T cells (bottom). Data are shown as mean frequencies (key). (F) Dot plots summarizing the frequencies of CTV IFN-g+ SARS-CoV-2-specific CD4+ and CD8+ T cells by group, serostatus, and specificity. Each dot rep- resents one donor. The dotted line indicates the cutoff for positive responses. Key as in (B) and (C). ***p < 0.001. Mann-Whitney U test. (G) Left: bar graph summarizing percent seropositivity by group. Right: bar graph summarizing the percentage of individuals in each group with detectable T cell responses directed against the internal (N) and surface antigens (M and/or S) of SARS-CoV-2. ll OPEN ACCESS 166 Cell 183, 158–168, October 1, 2020 Article
  • 11. the Swedish Childhood Cancer Fund, the Magnus Bergvalls Stiftelse, the Hed- lunds Stiftelse, the Lars Hiertas Stiftelse, the Swedish Physicians against AIDS Foundation, the Jonas Söderquist Stiftelse, and the Clas Groschinskys Min- nesfond. H.-G.L. and the Karolinska COVID-19 Study Group were supported by the Alice och Knut Wallenbergs Stiftelse and Nordstjernan AB. D.A.P. was supported by a Wellcome Trust Senior Investigator Award (100326/Z/ 12/Z). AUTHOR CONTRIBUTIONS H.-G.L., S.A., and M.B. conceived the project. T.S., A.P.-P., O.R.-B., J.-B.G., S.L.-L., S.M., D.J.W., T.K., J.E., T.P., D.A.P., and M.B. designed and per- formed experiments. T.S., A.P.-P., O.R.-B., J.-B.G., and M.B. analyzed data. K.S., A.O., H.K., G.B., E.F., O.R., L.I.E., J.-I.H., A.S., T.A., J.A., M.N., J.K., S.G.-R., N.K.B., J.K.S., D.A.P., H.-G.L., S.A., and M.B. provided critical re- sources. D.A.P. and M.B. supervised experiments. T.S., A.P.-P., O.R.-B., and M.B. drafted the manuscript. D.A.P. and M.B. edited the manuscript. All authors contributed intellectually and approved the manuscript. DECLARATION OF INTERESTS The authors declare no competing interests. Received: June 26, 2020 Revised: July 29, 2020 Accepted: August 11, 2020 Published: August 14, 2020 REFERENCES Alshukairi, A.N., Khalid, I., Ahmed, W.A., Dada, A.M., Bayumi, D.T., Malic, L.S., Althawadi, S., Ignacio, K., Alsalmi, H.S., Al-Abdely, H.M., et al. (2016). Anti- body Response and Disease Severity in Healthcare Worker MERS Survivors. Emerg. Infect. Dis. 22, 1113–1115. Beigel, J.H., Tomashek, K.M., Dodd, L.E., Mehta, A.K., Zingman, B.S., Kalil, A.C., Hohmann, E., Chu, H.Y., Luetkemeyer, A., Kline, S., et al. (2020). Remde- sivir for the Treatment of Covid-19— Preliminary Report. N Engl J Med. Pub- lished online May 22, 2020. https://doi.org/10.1056/NEJMoa2007764. Betts, M.R., Nason, M.C., West, S.M., De Rosa, S.C., Migueles, S.A., Abraham, J., Lederman, M.M., Benito, J.M., Goepfert, P.A., Connors, M., et al. (2006). HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood 107, 4781–4789. Blom, K., Braun, M., Ivarsson, M.A., Gonzalez, V.D., Falconer, K., Moll, M., Ljunggren, H.G., Michaëlsson, J., and Sandberg, J.K. (2013). Temporal dy- namics of the primary human T cell response to yellow fever virus 17D as it ma- tures from an effector- to a memory-type response. J. Immunol. 190, 2150–2158. Braun, J., Loyal, L., Frentsch, M., Wendisch, D., Georg, P., Kurth, F., Hippen- stiel, S., Dingeldey, M., Kruse, B., Fauchere, F., et al. (2020). SARS-CoV-2- reactive T cells in healthy donors and patients with COVID-19. Nature. Pub- lished online July 29, 2020. https://doi.org/10.1038/s41586-020-2598-9. Buggert, M., Norström, M.M., Salemi, M., Hecht, F.M., and Karlsson, A.C. (2014). Functional avidity and IL-2/perforin production is linked to the emer- gence of mutations within HLA-B*5701-restricted epitopes and HIV-1 disease progression. J. Immunol. 192, 4685–4696. Buggert, M., Nguyen, S., Salgado-Montes de Oca, G., Bengsch, B., Darko, S., Ransier, A., Roberts, E.R., Del Alcazar, D., Brody, I.B., Vella, L.A., et al. (2018). Identification and characterization of HIV-specific resident memory CD8+ T cells in human lymphoid tissue. Sci. Immunol. 3, eaar4526. Callow, K.A., Parry, H.F., Sergeant, M., and Tyrrell, D.A. (1990). The time course of the immune response to experimental coronavirus infection of man. Epidemiol. Infect. 105, 435–446. Chandrashekar, A., Liu, J., Martinot, A.J., McMahan, K., Mercado, N.B., Peter, L., Tostanoski, L.H., Yu, J., Maliga, Z., Nekorchuk, M., et al. (2020). SARS- CoV-2 infection protects against rechallenge in rhesus macaques. Science 369, 812–817. Channappanavar, R., Fett, C., Zhao, J., Meyerholz, D.K., and Perlman, S. (2014). Virus-specific memory CD8 T cells provide substantial protection from lethal severe acute respiratory syndrome coronavirus infection. J. Virol. 88, 11034–11044. Demkowicz, W.E., Jr., Littaua, R.A., Wang, J., and Ennis, F.A. (1996). Human cytotoxic T-cell memory: long-lived responses to vaccinia virus. J. Virol. 70, 2627–2631. Fuertes Marraco, S.A., Soneson, C., Cagnon, L., Gannon, P.O., Allard, M., Abed Maillard, S., Montandon, N., Rufer, N., Waldvogel, S., Delorenzi, M., and Speiser, D.E. (2015). Long-lasting stem cell-like memory CD8+ T cells with a naı̈ve-like profile upon yellow fever vaccination. Sci. Transl. Med. 7, 282ra48. Grifoni, A., Weiskopf, D., Ramirez, S.I., Mateus, J., Dan, J.M., Moderbacher, C.R., Rawlings, S.A., Sutherland, A., Premkumar, L., Jadi, R.S., et al. (2020). Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Disease and Unexposed Individuals. Cell 181, 1489–1501.e15. Guan, W.J., Ni, Z.Y., Hu, Y., Liang, W.H., Ou, C.Q., He, J.X., Liu, L., Shan, H., Lei, C.L., Hui, D.S.C., et al.; China Medical Treatment Expert Group for Covid- 19 (2020). Clinical Characteristics of Coronavirus Disease 2019 in China. N. Engl. J. Med. 382, 1708–1720. Habib, H. (2020). Has Sweden’s controversial covid-19 strategy been suc- cessful? BMJ 369, m2376. He, R., Lu, Z., Zhang, L., Fan, T., Xiong, R., Shen, X., Feng, H., Meng, H., Lin, W., Jiang, W., and Geng, Q. (2020a). The clinical course and its correlated im- mune status in COVID-19 pneumonia. J. Clin. Virol. 127, 104361. He, X., Lau, E.H.Y., Wu, P., Deng, X., Wang, J., Hao, X., Lau, Y.C., Wong, J.Y., Guan, Y., Tan, X., et al. (2020b). Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat. Med. 26, 672–675. Hotez, P.J., Corry, D.B., Strych, U., and Bottazzi, M.E. (2020). COVID-19 vac- cines: neutralizing antibodies and the alum advantage. Nat. Rev. Immunol. 20, 399–400. Huang, C., Wang, Y., Li, X., Ren, L., Zhao, J., Hu, Y., Zhang, L., Fan, G., Xu, J., Gu, X., et al. (2020). Clinical features of patients infected with 2019 novel coro- navirus in Wuhan, China. Lancet 395, 497–506. Ibarrondo, F.J., Fulcher, J.A., Goodman-Meza, D., Elliott, J., Hofmann, C., Hausner, M.A., Ferbas, K.G., Tobin, N.H., Aldrovandi, G.M., and Yang, O.O. (2020). Rapid Decay of Anti-SARS-CoV-2 Antibodies in Persons with Mild Covid-19. N. Engl. J. Med. Published online July 21, 2020. https://doi.org/ 10.1056/NEJMc2025179. Juno, J.A., Tan, H.X., Lee, W.S., Reynaldi, A., Kelly, H.G., Wragg, K., Esterba- uer, R., Kent, H.E., Batten, C.J., Mordant, F.L., et al. (2020). Humoral and circu- lating follicular helper T cell responses in recovered patients with COVID-19. Nat. Med. Published online July 13, 2020. https://doi.org/10.1038/s41591- 020-0995-0. Kirkcaldy, R.D., King, B.A., and Brooks, J.T. (2020). COVID-19 and Postinfec- tion Immunity: Limited Evidence, Many Remaining Questions. JAMA. Pub- lished online May 11, 2020. https://doi.org/10.1001/jama.2020.7869. Le Bert, N., Tan, A.T., Kunasegaran, K., Tham, C.Y.L., Hafezi, M., Chia, A., Chng, M.H.Y., Lin, M., Tan, N., Linster, M., et al. (2020). SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Na- ture. Published online July 15, 2020. https://doi.org/10.1038/s41586-020- 2550-z. Li, C.K., Wu, H., Yan, H., Ma, S., Wang, L., Zhang, M., Tang, X., Temperton, N.J., Weiss, R.A., Brenchley, J.M., et al. (2008). T cell responses to whole SARS coronavirus in humans. J. Immunol. 181, 5490–5500. Liu, J., Li, S., Liu, J., Liang, B., Wang, X., Wang, H., Li, W., Tong, Q., Yi, J., Zhao, L., et al. (2020). Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine 55, 102763. ll OPEN ACCESS Cell 183, 158–168, October 1, 2020 167 Article
  • 12. Long, Q.X., Tang, X.J., Shi, Q.L., Li, Q., Deng, H.J., Yuan, J., Hu, J.L., Xu, W., Zhang, Y., Lv, F.J., et al. (2020). Clinical and immunological assessment of asymptomatic SARS-CoV-2 infections. Nat. Med. 26, 1200–1204. Mallapaty, S. (2020). Will antibody tests for the coronavirus really change everything? Nature 580, 571–572. Mateus, J., Grifoni, A., Tarke, A., Sidney, J., Ramirez, S.I., Dan, J.M., Burger, Z.C., Rawlings, S.A., Smith, D.M., Phillips, E., et al. (2020). Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science. Published online August 4, 2020. https://doi.org/10.1126/science.abd3871. Miller, J.D., van der Most, R.G., Akondy, R.S., Glidewell, J.T., Albott, S., Maso- pust, D., Murali-Krishna, K., Mahar, P.L., Edupuganti, S., Lalor, S., et al. (2008). Human effector and memory CD8+ T cell responses to smallpox and yellow fe- ver vaccines. Immunity 28, 710–722. Ni, L., Ye, F., Cheng, M.L., Feng, Y., Deng, Y.Q., Zhao, H., Wei, P., Ge, J., Gou, M., Li, X., et al. (2020). Detection of SARS-CoV-2-Specific Humoral and Cellular Immunity in COVID-19 Convalescent Individuals. Immunity 52, 971–977.e3. Owen, R.E., Sinclair, E., Emu, B., Heitman, J.W., Hirschkorn, D.F., Epling, C.L., Tan, Q.X., Custer, B., Harris, J.M., Jacobson, M.A., et al. (2007). Loss of T cell responses following long-term cryopreservation. J. Immunol. Methods 326, 93–115. Plebani, M., Padoan, A., Negrini, D., Carpinteri, B., and Sciacovelli, L. (2020). Diagnostic performances and thresholds: The key to harmonization in serolog- ical SARS-CoV-2 assays? Clin. Chim. Acta 509, 1–7. Precopio, M.L., Betts, M.R., Parrino, J., Price, D.A., Gostick, E., Ambrozak, D.R., Asher, T.E., Douek, D.C., Harari, A., Pantaleo, G., et al. (2007). Immuni- zation with vaccinia virus induces polyfunctional and phenotypically distinctive CD8+ T cell responses. J. Exp. Med. 204, 1405–1416. Price, D.A., Brenchley, J.M., Ruff, L.E., Betts, M.R., Hill, B.J., Roederer, M., Koup, R.A., Migueles, S.A., Gostick, E., Wooldridge, L., et al. (2005). Avidity for antigen shapes clonal dominance in CD8+ T cell populations specific for persistent DNA viruses. J Exp Med. 202, 1349–1361. Reynisson, B., Alvarez, B., Paul, S., Peters, B., and Nielsen, M. (2020). NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data. Nucleic Acids Res. 48, W449–W454. Robbiani, D.F., Gaebler, C., Muecksch, F., Lorenzi, J.C.C., Wang, Z., Cho, A., Agudelo, M., Barnes, C.O., Gazumyan, A., Finkin, S., et al. (2020). Convergent Antibody Responses to SARS-CoV-2 Infection in Convalescent Individuals. bioRxiv. https://doi.org/10.1101/2020.05.13.092619. Seydoux, E., Homad, L.J., MacCamy, A.J., Parks, K.R., Hurlburt, N.K., Jenne- wein, M.F., Akins, N.R., Stuart, A.B., Wan, Y.H., Feng, J., et al. (2020). Charac- terization of neutralizing antibodies from a SARS-CoV-2 infected individual. bioRxiv. https://doi.org/10.1101/2020.05.12.091298. Shin, H.S., Kim, Y., Kim, G., Lee, J.Y., Jeong, I., Joh, J.S., Kim, H., Chang, E., Sim, S.Y., Park, J.S., and Lim, D.G. (2019). Immune Responses to Middle East Respiratory Syndrome Coronavirus During the Acute and Convalescent Phases of Human Infection. Clin. Infect. Dis. 68, 984–992. Singer, M., Deustchman, C.S., Seymour, C.W., Shankar-Hari, M., Annane, D., Bauer, M., Bellomo, R., Bernard, G.R., Chiche, J.D., Coopersmith, C.M., et al. (2016). The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 315, 801–810. Sridhar, S., Begom, S., Bermingham, A., Hoschler, K., Adamson, W., Carman, W., Bean, T., Barclay, W., Deeks, J.J., and Lalvani, A. (2013). Cellular immune correlates of protection against symptomatic pandemic influenza. Nat. Med. 19, 1305–1312. Tang, F., Quan, Y., Xin, Z.T., Wrammert, J., Ma, M.J., Lv, H., Wang, T.B., Yang, H., Richardus, J.H., Liu, W., and Cao, W.C. (2011). Lack of peripheral memory B cell responses in recovered patients with severe acute respiratory syn- drome: a six-year follow-up study. J. Immunol. 186, 7264–7268. Thevarajan, I., Nguyen, T.H.O., Koutsakos, M., Druce, J., Caly, L., van de Sandt, C.E., Jia, X., Nicholson, S., Catton, M., Cowie, B., et al. (2020). Breadth of concomitant immune responses prior to patient recovery: a case report of non-severe COVID-19. Nat. Med. 26, 453–455. Wang, C., Li, W., Drabek, D., Okba, N.M.A., van Haperen, R., Osterhaus, A.D.M.E., van Kuppeveld, F.J.M., Haagmans, B.L., Grosveld, F., and Bosch, B.J. (2020). A human monoclonal antibody blocking SARS-CoV-2 infection. Nat. Commun. 11, 2251. Wei, W.E., Li, Z., Chiew, C.J., Yong, S.E., Toh, M.P., and Lee, V.J. (2020). Pre- symptomatic Transmission of SARS-CoV-2 — Singapore, January 23-March 16, 2020. MMWR Morb. Mortal. Wkly. Rep. 69, 411–415. Wilk, A.J., Rustagi, A., Zhao, N.Q., Roque, J., Martinez-Colon, G.J., McKech- nie, J.L., Ivison, G.T., Ranganath, T., Vergara, R., Hollis, T., et al. (2020). A sin- gle-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 26, 1070–1076. Wilkinson, T.M., Li, C.K., Chui, C.S., Huang, A.K., Perkins, M., Liebner, J.C., Lambkin-Williams, R., Gilbert, A., Oxford, J., Nicholas, B., et al. (2012). Preex- isting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat. Med. 18, 274–280. Wolfel, R., Corman, V.M., Guggemos, W., Seilmaier, M., Zange, S., Muller, M.A., Niemeyer, D., Jones, T.C., Vollmar, P., Rothe, C., et al. (2020). Virological assessment of hospitalized patients with COVID-2019. Nature 581, 465–469. Woloshin, S., Patel, N., and Kesselheim, A.S. (2020). False Negative Tests for SARS-CoV-2 Infection — Challenges and Implications. N. Engl. J. Med. 383, e38. Wu, Z., and McGoogan, J.M. (2020). Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72314 Cases From the Chinese Center for Disease Control and Prevention. JAMA. Published online February 24, 2020. https://doi.org/10. 1001/jama.2020.2648. Yang, L.T., Peng, H., Zhu, Z.L., Li, G., Huang, Z.T., Zhao, Z.X., Koup, R.A., Bailer, R.T., and Wu, C.Y. (2006). Long-lived effector/central memory T-cell re- sponses to severe acute respiratory syndrome coronavirus (SARS-CoV) S an- tigen in recovered SARS patients. Clin. Immunol. 120, 171–178. Yang, R., Gui, X., and Xiong, Y. (2020). Comparison of Clinical Characteristics of Patients with Asymptomatic vs Symptomatic Coronavirus Disease 2019 in Wuhan, China. JAMA Netw. Open 3, e2010182. Zhao, J., Zhao, J., Mangalam, A.K., Channappanavar, R., Fett, C., Meyerholz, D.K., Agnihothram, S., Baric, R.S., David, C.S., and Perlman, S. (2016). Airway Memory CD4+ T Cells Mediate Protective Immunity against Emerging Respira- tory Coronaviruses. Immunity 44, 1379–1391. Zhao, J., Alshukairi, A.N., Baharoon, S.A., Ahmed, W.A., Bokhari, A.A., Nehdi, A.M., Layqah, L.A., Alghamdi, M.G., Al Gethamy, M.M., Dada, A.M., et al. (2017). Recovery from the Middle East respiratory syndrome is associated with antibody and T-cell responses. Sci. Immunol. 2, eaan5393. Zheng, H.Y., Zhang, M., Yang, C.X., Zhang, N., Wang, X.C., Yang, X.P., Dong, X.Q., and Zheng, Y.T. (2020a). Elevated exhaustion levels and reduced func- tional diversity of T cells in peripheral blood may predict severe progression in COVID-19 patients. Cell. Mol. Immunol. 17, 541–543. Zheng, M., Gao, Y., Wang, G., Song, G., Liu, S., Sun, D., Xu, Y., and Tian, Z. (2020b). Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell. Mol. Immunol. 17, 533–535. ll OPEN ACCESS 168 Cell 183, 158–168, October 1, 2020 Article
  • 13. STAR+METHODS KEY RESOURCES TABLE REAGENT or RESOURCE SOURCE IDENTIFIER Antibodies 1G1 (PE) [anti-CCR4] BD Biosciences 551120 (RRID:AB_394054) 11A9 (PE-Cy7) [anti-CCR6] BD Biosciences 560620 (RRID:AB_1727440) RPA-T8 or UCHT1 (BUV805) [anti-CD3] BD Biosciences 565515 (RRID:AB_2739277) RPA-T8 (BUV395) [anti-CD8] BD Biosciences 563795 (RRID:AB_2722501) M-A251 (PE-Cy5) [anti-CD25] BD Biosciences 560988 (RRID:AB_2033955) CD28.2 (BUV563) [anti-CD28] BD Biosciences 564438 (RRID:AB_2738808) HIT2 (BUV496) [anti-CD38] BD Biosciences 564657 (RRID:AB_2744376) FN50 (BV750) [anti-CD69] BD Biosciences 563835 (RRID:AB_2738442) FN50 (BUV737) [anti-CD69] BD Biosciences 564439 (RRID:AB_2722502) DX2 (BB630) [anti-CD95] BD Biosciences Custom conjugate H4A3 (PE-CF594) [anti-CD107a] BD Biosciences 562628 (RRID:AB_2737686) BNI3 (BB755) [anti-CTLA-4] BD Biosciences 560938 (RRID:AB_2033942) RF8B2 (APC-R700) [anti-CXCR5] BD Biosciences 565191 (RRID:AB_2739103) GB11 (BB790) [anti-granzyme B] BD Biosciences Custom conjugate G46-6 (BUV615) [anti-HLA-DR] BD Biosciences 565073 (RRID:AB_2722500) MQ1-17H12 (APC-R700) [anti-IL-2] BD Biosciences 565136 (RRID:AB_2739079) B56 (BB660) [anti-Ki-67] BD Biosciences Custom conjugate T47-530 (BUV661) [anti-LAG-3] BD Biosciences Custom conjugate dG9 (BB700) [anti-perforin] BD Biosciences Custom conjugate 741182 (BUV737) [anti-TIGIT] BD Biosciences Custom conjugate C1.7 (PE/Dazzle 594) [anti-2B4] BioLegend 393506 (RRID:AB_2734464) G043H7 (APC-Cy7) [anti-CCR7] BioLegend 353211 (RRID:AB_10915272) M5E2 (BV510) [anti-CD14] BioLegend 301842 (RRID:AB_2561946) HIB19 (BV510) [anti-CD19] BioLegend 302242 (RRID:AB_2561668) O323 (BV785) [anti-CD27] BioLegend 302832 (RRID:AB_2562674) A1 (BV711) [anti-CD39] BioLegend 328227 (RRID:AB_2632893) HI100 (BV421) [anti-CD45RA] BioLegend 304129 (RRID:AB_10900421) HI100 (BV570) [anti-CD45RA] BioLegend 304131 (RRID:AB_10897946) A019D5 (BV605) [anti-CD127] BioLegend 351334 (RRID:AB_2562022) G025H7 (AF647) [anti-CXCR3] BioLegend 353712 (RRID:AB_10962948) 4S.B3 (BV785) [anti-IFN-g] BioLegend 502542 (RRID:AB_2563882) EH12.2H7 (PE-Cy7) [anti-PD-1] BioLegend 367415 (RRID:AB_2616743) F38-2E2 (BV650) [anti-TIM-3] BioLegend 345027 (RRID:AB_2565828) Mab11 (BV650) [anti-TNF] BioLegend 502937 (RRID:AB_2561355) 4B41 (BV421) [anti-4-1BB] BioLegend 309820 (RRID:AB_2563830) C63D9 (AF488) [anti-TCF1] Cell Signaling 6444 (RRID:AB_2797627) REA473 (A647) [anti-TOX] Miltenyi Biotec 130-107-838 (RRID:AB_2654224) S3.5 (PE-Cy5.5) [anti-CD4] Thermo Fisher Scientific MHCD0418 (RRID:AB_10376013) eBio64DEC17 (PE) [anti-IL-17A] Thermo Fisher Scientific 12-7179-42 (RRID:AB_1724136) Purified anti-CD28/CD49d BD Biosciences 347690 (RRID:AB_647457) Ultra-LEAF OKT3 Purified [anti-CD3] BioLegend 317326 (RRID: AB_11150592) LIVE/DEAD Fixable Aqua Dead Cell Stain Kit Thermo Fisher Scientific Cat#L34957 (Continued on next page) ll OPEN ACCESS Cell 183, 158–168.e1–e5, October 1, 2020 e1 Article
  • 14. RESOURCE AVAILABILITY Lead Contact Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Marcus Buggert (marcus.buggert@ki.se) Material Availability HLA class I tetramers can be generated and shared on a collaborative basis. Continued REAGENT or RESOURCE SOURCE IDENTIFIER Biological Samples Acute moderate blood samples Karolinska Hospital N/A Acute severe blood samples Karolinska Hospital N/A Mild convalescent blood samples Karolinska Hospital N/A Severe convalescent blood samples Karolinska Hospital N/A Exposed family member blood samples Karolinska Hospital N/A 2020 blood donor samples Karolinska Universitetslaboratoriet N/A 2019 blood donor samples Karolinska Universitetslaboratoriet N/A Chemicals, Peptides, and Recombinant Proteins Synthetic CMV peptides Peptides & Elephants GmbH https://www.peptides.de/ Synthetic EBV peptides Peptides & Elephants GmbH https://www.peptides.de/ Synthetic overlapping SARS-CoV-2 peptides (Prot_M, N, S) Miltenyi Biotec https://www.miltenyibiotec.com/SE-en/ Optimal SARS-CoV-2 peptides Peptides & Elephants GmbH https://www.peptides.de/ Foxp3/TF Staining Buffer Set Invitrogen Cat#00-5523-00 Staphylococcal Enterotoxin B (SEB) Sigma-Aldrich Cat#S4881 Brefeldin A BioLegend Cat#420601 BD GolgiStop (with Monensin) BD Biosciences Cat#554724 BCIP/NBT Substrate Mabtech Cat#3650-10 BL21(DE3)pLysS Competent Cells Novagen Cat#69451 Triton X-100 Buffer Sigma-Aldrich Cat#X100 8M Urea Buffer Sigma-Aldrich Cat#51457 Cysteamine Sigma-Aldrich Cat#M9768 D-(+)-Biotin Sigma-Aldrich Cat#2031 Critical Commercial Assays iFLASH Anti-SARS-CoV-2 IgG Kit YHLO Cat#20210217 LIAISON SARS-CoV-2 ELISA IgG Kit DiaSorin Cat#311450 Human IFN-g ELISpot Kit Mabtech Cat#3420-2A BD Multitest 6-color TBNK reagent with BD Trucount tubes BD Biosciences Cat#337166 CellTrace Violet Cell Proliferation Kit Thermo Fisher Scientific Cat#C34571 Software and Algorithms NetMHCpan EL 4.1 DTU Bioinformatics http://www.cbs.dtu.dk/services/ NetMHCpan-4.1/ FlowJo 10.6.1 FlowJo https://www.flowjo.com/ UMAP plugin 2.2 FlowJo https://www.flowjo.com/ GraphPad Prism 8.4.2 GraphPad Software https://www.graphpad.com/ PhenoGraph 0.2.1 PhenoGraph https://omictools.com/phenograph-tool RStudio RStudio https://rstudio.com/ Python (scikit-learn 0.22.1) Python https://www.python.org/ ll OPEN ACCESS e2 Cell 183, 158–168.e1–e5, October 1, 2020 Article
  • 15. Data and Code Availability The published article includes all data generated during this study. All codes are freely available at source. EXPERIMENTAL MODEL AND SUBJECT DETAILS Human subjects Maximal disease severity was assessed using the NIH Ordinal Scale and Sequential Organ Failure Assessment (SOFA) (Beigel et al., 2020; Singer et al., 2016). The NIH Ordinal Scale was defined as follows: (1) not hospitalized with no limitation of activities; (2) not hospitalized with limitation of activities and/or home oxygen requirement; (3) hospitalized but not requiring supplemental oxygen and no longer requiring ongoing medical care; (4) hospitalized and not requiring supplemental oxygen but requiring ongoing medical care; (5) hospitalized requiring supplemental oxygen; (6) hospitalized requiring non-invasive ventilation or the use of high-flow oxygen devices; (7) hospitalized receiving invasive mechanical ventilation or extracorporeal membrane oxygenation; and (8) death. Donors were assigned to one of seven groups for the purposes of this study. AS: patients with acute severe disease requiring hospitalization in the high dependency or intensive care unit, with low-flow oxygen support (< 10 L/min), high-flow oxygen support, or invasive mechanical ventilation (n = 17). These patients had a median NIH Ordinal Scale score of 7 (IQR 6–7) and a median SOFA score of 6 (IQR 3–6) at the time of sampling 12–17 days after disease onset (47% were viremic, and 82% were antibody-seropositive for SARS-CoV-2). AM: patients with acute moderate disease requiring hospitalization and low-flow oxygen support (0–3 L/min; n = 10). These patients had a median NIH Ordinal Scale score of 5 (IQR 5–5) and a median SOFA score of 1 (IQR 1–1) at the time of sampling 11–14 days after disease onset (40% were viremic, and 50% were antibody-seropositive for SARS-CoV-2). SC: individ- uals in the convalescent phase after severe disease (n = 26). Samples were collected 42–58 days after disease onset, corresponding to 3–21 days after resolution of symptoms (100% were antibody-seropositive for SARS-CoV-2). MC: individuals in the convalescent phase after mild disease (n = 40). Samples were collected 49–64 days after disease onset, corresponding to 25–53 days after resolution of symptoms (85% were antibody-seropositive for SARS-CoV-2). Exp: family members who shared a household with donors in groups MC or SC (n = 30). These individuals were exposed at the time of symptomatic disease (21% remained asymptom- atic, and 63% were antibody-seropositive for SARS-CoV-2). 2020 BD: individuals who donated blood at the Karolinska University Hospital in May 2020 (during the pandemic; n = 55). 2019 BD: individuals who donated blood at the Karolinska University Hospital between July and September 2019 (before the pandemic; n = 28). PBMCs were isolated from heparin-coated tubes (groups AS, AM, SC, MC, Exp, and 2020 BD) or citrate-anticoagulated buffy coats (group 2019 BD). A separate serum separator tube was collected from each donor. All donors provided written informed consent in accordance with the Declaration of Helsinki. The study was approved by the Swedish Ethical Review Authority. Donor characteristics are summarized in Table S3, and immunological assay breakdowns are summarized in Table S4. METHOD DETAILS Flow cytometry PBMCs were isolated from venous blood samples via standard density gradient centrifugation and used immediately (groups AS, AM, SC, MC, Exp, and 2020 BD) or after cryopreservation in liquid nitrogen (group 2019 BD). Cells were washed in phosphate-buff- ered saline (PBS) supplemented with 2% fetal bovine serum (FBS) and 2 mM EDTA (FACS buffer) and stained with PE-conjugated or BV421-conjugated HLA class I tetramers and/or anti-CCR7–APC-Cy7 (clone G043H7; BioLegend) for 10 min at 37 C. Other surface markers were detected via the subsequent addition of directly conjugated antibodies at pre-titrated concentrations for 20 min at room temperature, and viable cells were identified by exclusion using a LIVE/DEAD Fixable Aqua Dead Cell Stain Kit (Thermo Fisher Scientific). Cells were then washed again in FACS buffer and fixed/permeabilized using a FoxP3 / Transcription Factor Staining Buffer Set (eBioscience). Intracellular markers were detected via the addition of directly conjugated antibodies at pre-titrated concentra- tions for 1 hr at 4 C. Stained cells were fixed in PBS containing 1% paraformaldehyde (PFA; Biotium) and stored at 4 C. Samples were acquired using a FACSymphony A5 (BD Biosciences). Data were analyzed with FlowJo software version 10.6.1 (FlowJo LLC). Gating strategies were described previously (Buggert et al., 2014, 2018). Core gates included singlet isolation (FSC-H versus FSC-A), live CD3 selection (CD3 versus Aqua, CD14, and CD19), lymphocyte enrichment (SSC-A versus FSC-A), CD4 or CD8 selec- tion (CD4 versus CD8). Detailed gating strategies for individual markers are depicted in the relevant figures. Peptides Peptides corresponding to known optimal epitopes derived from CMV (pp65) and EBV (BZLF1 and EBNA-1), overlapping peptides spanning the immunogenic domains of the SARS-CoV-2 spike (Prot_S), membrane (Prot_M), and nucleocapsid proteins (Prot_N), and optimal peptides for the manufacture of HLA class I tetramers were synthesized at 95% purity. Lyophilized peptides were re- constituted at a stock concentration of 10 mg/mL in DMSO and further diluted to 100 mg/mL in PBS. Epitope prediction Peptides were selected from full-length SARS-CoV-2 sequences spanning 82 different strains from 13 countries (National Center for Biotechnology Information). The predicted binding affinities of conserved 9-mer peptides for HLA-A*0201 and HLA-B*0702 were ll OPEN ACCESS Cell 183, 158–168.e1–e5, October 1, 2020 e3 Article
  • 16. determined using NetMHCpan version 4.1 (Reynisson et al., 2020). Binders were defined by a threshold IC50 value of 500 nM. Strong binders were defined by a % Rank 0.5, and weak binders were defined by a % Rank 2 (Table S2). A total of 13 strong binders were identified for tetramer generation (Table S2). Tetramers HLA class I tetramers were generated as described previously (Price et al., 2005). Briefly, biotin-tagged HLA-A*0201 and HLA-B*0702 heavy chains were expressed under the control of a T7 promoter as insoluble inclusion bodies in Escherichia coli strain BL21(DE3) pLysS (Novagen). IPTG-induced Escherichia coli were lyzed via repeated freeze/thaw cycles to release inclusion bodies that were subsequently purified by washing in 0.5% Triton X-100 buffer (Sigma-Aldrich). Heavy chain and b2 m inclusion body preparations were denatured separately in 8 M urea buffer (Sigma-Aldrich) and mixed at a 1:1 molar ratio. Each monomeric protein was refolded in 2-mercaptoethylamine/cystamine redox buffer (Sigma-Aldrich) with the appropriate synthetic peptide (Peptides Elephants GmbH). Refolded monomers were purified via anion exchange after buffer replacement (10 mM Tris, pH 8.1). Purified monomers were biotinylated using d-biotin and BirA (Sigma-Aldrich). Excess biotin was removed via gel filtration. Biotinylated monomers were then tetramerized via the addition of fluorochrome-conjugated streptavidin at a 4:1 molar ratio, respectively. The following specificities were used in this study: CMV A*0201 NV9 (NLVPMVATV), EBV A*0201 GL9 (GLCTLVAML), SARS-CoV-2 A*0201 AV9 (ALSKGVHFV), SARS-CoV-2 A*0201 HI9 (HLVDFQVTI), SARS-CoV-2 A*0201 KV9 (KLLEQWNLV), SARS-CoV-2 A*0201 LL9 (LLLDRLNQL), SARS-CoV-2 A*0201 LLY (LLYDANYFL), SARS-CoV-2 A*0201 SV9 (SLVKPSFYV), SARS-CoV-2 A*0201 TL9 (TLDSKTQSL), SARS-CoV-2 A*0201 VL9 (VLNDILSRL), SARS-CoV-2 A*0201 YL9 (YLQPRTFLL), SARS-CoV-2 B*0702 FI9 (FPRGQGVPI), SARS-CoV-2 B*0702 KT9 (KPRQKRTAT), SARS-CoV-2 B*0702 SA9 (SPRRARSVA), and SARS-CoV-2 B*0702 SL9 (SPRWYFYYL). Functional assay PBMCs were resuspended in complete medium (RPMI 1640 supplemented with 10% FBS, 1% L-glutamine, and 1% penicillin/strep- tomycin) at 1 3 107 cells/mL and cultured at 1 3 106 cells/well in 96-well V-bottom plates (Corning) with the relevant peptides (each at 0.5 mg/mL) for 30 min prior to the addition of anti-CD28/CD49d (3 mL/mL; clone L293/L25; BD Biosciences), brefeldin A (1 mL/mL; Sigma-Aldrich), monensin (0.7 mL/mL; BD Biosciences), and anti-CD107a–PE-CF594 (clone H4A3; BD Biosciences). Negative con- trol wells lacked peptides, and positive control wells included staphylococcal enterotoxin B (SEB; 0.5 mg/mL; Sigma-Aldrich) or plate- bound anti-CD3 (1 mg/mL; clone OKT3; BioLegend). Cells were analyzed by flow cytometry after incubation for 8 hr at 37 C. Proliferation assay PBMCs were labeled with CTV (0.5 mM; Thermo Fisher Scientific), resuspended in complete medium at 1 3 107 cells/mL, and cultured at 1 3 106 cells/well in 96-well U-bottom plates (Corning) with the relevant peptides (each at 1 mg/mL) in the presence of anti-CD28/CD49d (3 mL/mL; clone L293/L25; BD Biosciences) and IL-2 (10 IU/mL; PeproTech). Negative control wells lacked peptides, and positive control wells included SEB (0.5 mg/mL; Sigma-Aldrich) or plate-bound anti-CD3 (1 mg/mL; clone OKT3; BioLegend). Functional assays were performed as described above after incubation for 5 days at 37 C. AIM assay PBMCs were resuspended in complete medium at 1 3 107 cells/mL and cultured at 1 3 106 cells/well in 96-well U-bottom plates (Corning) with the relevant peptides (each at 1 mg/mL) in the presence of anti-CD28/CD49d (3 mL/mL; clone L293/L25; BD Biosciences). Cells were analyzed by flow cytometry after incubation for 24 hr at 37 C. The following directly conjugated monoclonal antibodies were used to detect activation markers: anti-CD69–BUV737 (clone FN50; BD Biosciences) and anti-4-1BB–BV421 (clone 4B41; BioLegend). Trucount Absolute counts were obtained using Multitest 6-color TBNK reagent with Trucount tubes (BD Biosciences). Samples were fixed with 2% PFA for 2 hr prior to acquisition. Absolute CD3+ cell counts were calculated using the following formula: (# CD3+ events acquired x total # beads x 1000) / (# beads acquired x volume of whole blood stained in mL). CD4+ and CD8+ cell counts were computed from the respective frequencies relative to CD3+ cells. Principal component analysis Analyses were performed using scikit-learn version 0.22.1 in Python. Data were normalized using sklearn.preprocessing. StandardScaler in the same package to generate z-scores for PCA. UMAP FCS 3.0 data files were imported into FlowJo software version 10.6.0 (FlowJo LLC). All samples were compensated electronically. Dimensionality reduction was performed using the FlowJo plugin UMAP version 2.2 (FlowJo LLC). The downsample version 3.0.0 plugin and concatenation tool was used to visualize multiparametric data from up to a total of 120,000 CD8+ T cells (n = 3 donors per group). Representative donors were selected from the 50th percentile for all markers. The following parameters were used in ll OPEN ACCESS e4 Cell 183, 158–168.e1–e5, October 1, 2020 Article
  • 17. these analyses: metric = euclidean, nearest neighbors = 30, and minimum distance = 0.5. Clusters of phenotypically related cells were detected using PhenoGraph version 0.2.1. The following markers were included in the cluster analysis: CCR7, CD27, CD28, CD38, CD39, CD45RA, CD95, CD127, CTLA-4, CXCR5, granzyme B, Ki-67, LAG-3, PD-1, perforin, TCF1, TIGIT, TIM-3, TOX, and 2B4. Plots were generated using Prism version 8.2.0 (GraphPad Software Inc.). ELISpot assay PBMCs were rested overnight in complete medium and seeded at 2 3 105 cells/well in MultiScreen HTS Filter Plates (Merck Millipore) pre-coated with anti-IFN-g (15 mg/mL; clone 1-D1K; Mabtech). Test wells were supplemented with overlapping peptides spanning Prot_S, Prot_M, and Prot_N (each at 2 mg/mL; Miltenyi Biotec) or peptides corresponding to known optimal epitopes derived from CMV (pp65) or EBV (BZLF1 and EBNA-1) (each at 2 mg/mL; Peptides Elephants GmbH). Negative control wells lacked peptides, and positive control wells included SEB (0.5 mg/mL; Sigma-Aldrich). Assays were incubated for 24 hr at 37 C. Plates were then washed six times with PBS (Sigma-Aldrich) and incubated for 2 hr at room temperature with biotinylated anti-IFN-g (1 mg/mL; clone mAb-7B6-1; Mabtech). After six further washes, a 1:1,000 dilution of alkaline phosphatase-conjugated streptavidin (Mabtech) was added for 1 hr at room temperature. Plates were then washed a further six times and developed for 20 min with BCIP/NBT Substrate (Mabtech). All assays were performed in duplicate. Mean values from duplicate wells were used for data representation. Spots were counted using an automated ELISpot Reader System (Autoimmun Diagnostika GmbH). Serology SARS-CoV-2-specific antibodies were detected in serum using both the iFLASH Anti-SARS-CoV-2 IgG chemiluminescent micropar- ticle immunoassay against the nucleocapsid and spike proteins (Shenzhen YHLO Biotech Co. Ltd.) as well as the LIAISON SARS-CoV-2 IgG fully automated indirect chemiluminescent immunoassay against the S1 and S2 (spike) proteins (DiaSorin). These assays produced highly concordant results and have been shown to perform adequately as diagnostic tools (Plebani et al., 2020). An individual was considered seropositive if one of the two methods generated a positive result. All assays were performed by trained employees at the clinical laboratory according to standard procedures. QUANTIFICATION AND STATISTICAL ANALYSIS Statistical analyses were performed using R studio or Prism version 7.0 (GraphPad Software Inc.). Differences between unmatched groups were compared using an unpaired t test, the Mann-Whitney U test, or the Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons, and differences between matched groups were compared using a paired t test or the Wilcoxon signed- rank test. Correlations were assessed using the Spearman rank correlation. Non-parametric tests were used if the data were not distributed normally according to the Shapiro-Wilk normality test. Phenotypic relationships within multivariate datasets were visualized using FlowJo software version 10.6.1 (FlowJo LLC). ll OPEN ACCESS Cell 183, 158–168.e1–e5, October 1, 2020 e5 Article
  • 18. Supplemental Figures A 2020 BD AM AS 0 400000 800000 1000000 2000000 2020 BD AM AS 0 200000 400000 600000 800000 1000000 1500000 2020 BD AM AS 0 5 10 20 2020 BD AM AS 0 4 8 12 16 CD14 CD15 Time SSC-A CD45 CD3 CD45 SSC-A FSC-A FSC-H CD19 CD3 Beads 0.34 3.33 95.4 33.9 75.1 Gating strategy of total T cells from SARS-CoV-2+ acute patient B CD95 CCR7 CD95 CCR7 Gating strategy for the identification of memory CD4+ and CD8+ T cells Gated on CD3+CD8+ Gated on CD3+CD4+ C CD8 CD4 % of memory CD4+ T cells CCR7 0 20 40 60 80 100 0 5 10 15 20 25 CD25 CD27 20 40 60 80 100 CD28 0 20 40 60 80 100 0 8 16 24 32 40 CD39 0 20 40 60 80 100 CD127 CTLA4 0 4 8 12 16 20 LAG-3 0 15 30 45 60 CXCR5 0 8 16 24 32 40 HLA-DR GzmB 0 16 32 48 64 80 0 8 16 24 32 40 0 Perforin 0 5 10 15 20 25 CD45RA 0 5 10 15 20 25 0 20 40 60 80 100 TCF-1 0 15 30 45 60 TIGIT TIM-3 TOX % of memory CD8+ T cells 0 20 40 60 80 100 2B4 0 20 40 60 80 100 CCR7 0 10 20 30 40 50 CD25 0 20 40 60 80 100 CD27 0 20 40 60 80 100 CD28 0 20 40 60 80 100 CD127 CXCR5 0 6 12 18 24 30 0 20 40 60 80 100 GzmB PD-1 0 20 40 60 80 100 0 20 40 60 80 100 Perforin 0 20 40 60 80 100 CD45RA TCF-1 0 20 40 60 80 100 0 20 40 60 80 100 TIGIT 0 20 40 60 80 100 TOX 2020 BD MC AM AS 2020 BD MC AM AS 0 2 4 6 8 0 10 20 30 40 50 Abs count memory CD4+ T cells Abs count memory CD8+ T cells % memory CD4+ T cells % memory CD8+ T cells (legend continued on next page) ll OPEN ACCESS Article
  • 19. Figure S1. Quantification and Characterization of CD4+ and CD8+ T Cells in COVID-19, Related to Figure 1 (A) Flow cytometric gating strategy for the identification and quantification of CD4+ and CD8+ T cells. (B) Left: flow cytometric gating strategy for the identification and quantification of memory CD4+ and CD8+ T cells. Right: dot plots summarizing the absolute numbers and relative frequencies of memory CD4+ and CD8+ T cells by group. Each dot represents one donor. Data are shown as median ± IQR. 2020 BD: healthy blood donors from 2020 (n = 18). AM: patients with acute moderate COVID-19 (n = 11). AS: patients with acute severe COVID-19 (n = 17). (C) Dot plots summarizing the expression frequencies of phenotypic markers among memory CD4+ and CD8+ T cells by group. Each dot represents one donor. Bars indicate median values. 2020 BD: healthy blood donors from 2020 (n = 18). MC: individuals in the convalescent phase after mild COVID-19 (n = 31). AM: patients with acute moderate COVID-19 (n = 11). AS: patients with acute severe COVID-19 (n = 17). ll OPEN ACCESS Article
  • 20. *** UMAP2 UMAP1 HLA-DR Ki-67 CD38 LAG-3 TIM-3 CD39 2B4 TIGIT PD-1 CD27 CD127 CXCR5 GzmB Perforin CD95 CD28 CD45RA CCR7 Gated on memory CD4+ T cells Max Min CTLA4 CD25 CD69 TOX TCF-1 2020 BD MC AM AS B CD38+ Ki67+ CD4+ CD38+ HLA-DR+ CD4+ CD38+ PD-1+ CD4+ CD38+ Ki67+ CD8+ CD38+ HLA-DR+ CD8+ CD38+ PD-1+ CD8+ CD38 + PD-1 + CD8 + CD38 + HLA-DR + CD4 + CD38 + PD-1 + CD4 + CD38 + Ki67 + CD8 + CD38 + HLA-DR + CD8 + 8 3 D C + 1 - D P + 8 D C + d e e n n e g y x o k a e P e g A I M B n o i s s i m d a o t t u b e d m o t p m y s s y a D g n i l p m a s t u b e d m o t p m y s s y a D y a t s U C I n o i t a r u D n o i t a b u t n i U C I n o i t a r u D t n e m t a e r t n e g y x o n o i t a r u D ) l / g m 3 f e r ( h 4 2 - / + P R C t s e h g i H P R C t s e h g i H ) l / g µ 5 . 0 f e r ( h 4 2 - / + T C P t s e h g i H Highest PCT Lowest Hb +/- 24h (ref 117–153 g/l) Highest leukocyte count +/- 24h (ref 3.5–8.8 x 10 9 c/l) Lowest leukocyte count +/- 24h Highest neutrophil count +/- 24h (ref 1.6–5.9 x 10 9 c/l) Lowest neutrophil count Highest ferritin +/- 24h (ref 10–150 µg/l) Highest ferritin Highest troponin T +/- 24h (ref 15 ng/l) Highest troponin T Highest D-dimer +/- 24h (ref 0.56 mg/l) Highest D-dimer Highest PK-INR +/- 24h (ref 1.2 INR) Highest PK-INR Highest fibrinogen +/-24h (ref 2– 4.2 g/l) Highest fibrinogen Lowest thrombocytes +/- 24h (ref 156–387 x 10 9 c/l) Lowest thrombocytes Highest thrombocytes Highest bilirubin +/- 24h (ref 26 µg/l) Highest bilirubin Highest ALT (ref 0.76 µkat/l) Highest LD (ref 3.5 µkat/l) Lowest P-albumin +/- 24h (ref 73 µg/l) Highest myoglobin (ref 73 µg/l) Highest IL-6 +/- 5 days (ref 7 ng/l) Highest IL-1 +/- 5 days (ref 5 ng/l) Highest IL-10 +/- 5 days (ref 5 ngl) Highest TNF +/- 5 days (ref 12 ng/l) CMV-IgG (U/ml), Diasonin Liason XL SARS-CoV-2-IgG (AU/ml), iFlash YHLO Lowest lymphocyte count +/- 24h (ref 1.1–3.5 x 10 9 c/l) Lowest lymphocyte count Highest creatinine +/- 24h (ref 90 µg/l) Highest creatinine 1 0 -1 C D Cycling 20 10 5 15 0 CD38+PD-1+ CD38+HLA-DR+ CD38+Ki-67+ % of memory CD4+ T cells *** *** *** *** 25 40 20 10 30 0 50 20 10 5 15 0 *** * *** *** *** *** 2020 BD MC AM AS *** A 3 15 16 18 26 27 17 29 25 22 19 23 21 28 20 13 14 12 9 8 7 6 5 4 1 2 11 10 30 24 Phenograph clusters from memory CD8+ T cells C25 C29 TOX CCR7 CXCR5 TCF1 CD95 Ki67 Perforin CTLA4 Granzyme CD38 CD28 HLA-DR LAG3 TIGIT CD45RA CD127 Tim3 CD39 CD69 CD27 2B4 CD25 PD-1 Log2 Expression 25 29 0 50 % coontribution to each cluster 100 Healthy Control Convalescent Mild Severe (legend on next page) ll OPEN ACCESS Article
  • 21. Figure S2. Phenograph and UMAP Clustering of Memory CD4+ and CD8+ T Cells with Correlative Analyses of Immune Activation Phenotypes versus Clinical Parameters in Acute COVID-19, Related to Figure 1 (A) Phenograph plots showing the clustering of memory CD8+ T cells and heatmap highlighting clusters 20 and 29. (B) Top left: UMAP plots showing the clustering of memory CD4+ T cells by group in relation to all memory CD4+ T cells (left). Bottom left: UMAP plots showing the expression of individual markers (n = 3 donors per group). Right: dot plots summarizing the expression frequencies of activation/cycling markers among memory CD4+ T cells by group. Each dot represents one donor. Data are shown as median ± IQR. 2020 BD: healthy blood donors from 2020 (n = 18). MC: individuals in the convalescent phase after mild COVID-19 (n = 31). AM: patients with acute moderate COVID-19 (n = 11). AS: patients with acute severe COVID-19 (n = 17). *p 0.05, **p 0.01, ***p 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons. (C) and (D) Heatmaps summarizing the pairwise correlations between phenotypically defined subpopulations of memory CD4+ or CD8+ T cells and various clinical parameters in patients with acute COVID-19. *p 0.05, **p 0.01, ***p 0.001. Spearman rank correlation. ll OPEN ACCESS Article
  • 22. CCR7 96 4 Acute Healthy CD127 89 11 TIGIT 90 10 TCF-1 96 4 Perforin 83 17 Granzyme B 94 6 CD45RA 98 2 CD28 73 26 CD27 88 12 1 1 14 3 86 97 99 99 CD38 Ki-67 Gated on memory CD8+ T cells C D A E CD38 UMAP Y-axis UMAP X-axis CD38 HLA-DR Ki-67 PD-1 26.0 2.9 12.5 1.63 29.8 22.1 B 0 20 40 60 80 2020 BD AM AS % CD8+CD38+HLA-DR− Low High *** *** Ki-67 A*02:01 NV9 CM V HLA-DR Gated on memory CD8+ T cells CD38+HLA-DR+ donor CD38+HLA-DR− donor CD38+HLA-DR+ donor CD38+HLA-DR− donor Ki-67 PD-1 E 0.0 0.1 0.2 0.3 0.4 0 -10 3 10 3 10 4 10 5 0 10 3 10 4 10 5 0 -10 3 10 3 10 4 10 5 Comp-PE-A :: B*0702 SL9 0 10 3 10 4 10 5 0.34 0.09 Representative FACS plots for 2 MC subjects SC MC A*0201 TL9 B*0702 SL9 A*0201 YL9 B*0702 FI9 Gated on CD8+ T cells CCR7 CD95 % SARS-Cov-2 tetramer+ cells Figure S3. Immune Activation Patterns in Acute COVID-19, Related to Figure 2 (A) Representative flow cytometry plots showing the expression of activation/cycling markers among memory CD8+ T cells in patients with acute severe COVID- 19. Numbers indicate percentages in the drawn gates. (B) Top left: UMAP plots showing the clustering of memory CD8+ T cells by phenotype in relation to all memory CD8+ T cells (left). Bottom left: UMAP plots showing the expression of individual markers (n = 1 donor per group). Right: dot plot summarizing the frequencies of CD38+ HLA-DR memory CD8+ T cells by group. Each dot represents one donor. Data are shown as median ± IQR. 2020 BD: healthy blood donors from 2020 (n = 18). AM: patients with acute moderate COVID-19 (n = 11). AS: patients with acute severe COVID-19 (n = 17). ***p 0.001. Kruskal-Wallis rank-sum test with Dunn’s post hoc test for multiple comparisons. (C) Representative flow cytometry plots showing the expression of activation/cycling markers among (legend continued on next page) ll OPEN ACCESS Article
  • 23. CMV-specific memory CD8+ T cells in a healthy control and a patient with acute severe COVID-19. Numbers indicate percentages in the drawn gates. (D) Representative flow cytometry plots showing the phenotype of Ki-67+ memory CD8+ T cells in a patient with acute severe COVID-19. Numbers indicate per- centages in the drawn gates. (E) Left: representative flow cytometry plots showing SARS-CoV-2-specific tetramer+ CD8+ T cells in two donors from group MC. Right: dot plot summarizing the frequencies of SARS-CoV-2-specific tetramer+ CD8+ T cells in donors from groups MC (n = 10) and SC (n = 2). ll OPEN ACCESS Article
  • 24. A B Figure S4. Quantification of Functional T Cell Reactivity in COVID-19, Related to Figure 3 (A) Representative images showing the detection of IFN-g-producing cells responding to overlapping peptides spanning the immunogenic domains of the SARS- CoV-2 spike (S), membrane (M), and nucleocapsid proteins (N) by group (ELISpot assays). NC: negative control. EBV: Epstein-Barr virus. CMV: cytomegalovirus. SEB: staphylococcal enterotoxin B. (B) Dot plots summarizing the frequencies of IFN-g-producing cells responding to optimal peptide epitopes derived from EBV BZLF1 and EBNA-1 (left) or CMV pp65 (right) by group (ELISpot assays). Each dot represents the mean of combined specificities in one donor. Bars indicate median values. No significant differences were detected among groups for any specificity. 2019 BD: healthy blood donors from 2019 (n = 25). 2020 BD: healthy blood donors from 2020 (n = 24). Exp: exposed family members (n = 30). MC: individuals in the convalescent phase after mild COVID-19 (n = 31). SC: individuals in the convalescent phase after severe COVID-19 (n = 22). SFU: spot-forming unit. ll OPEN ACCESS Article
  • 25. A 38 62 38 62 14.3 37.1 32.9 15.7 78.3 21.7 CXCR5 CXCR3 cTfh Th1 CXCR5 CXCR3 Th1/17 Th17 Th2 c T f h 1 c T f h 2 c T f h 1 7 c T f h 1 / 1 7 c T f h 1 c T f h 2 c T f h 1 7 c T f h 1 / 1 7 0 20 40 60 80 c T f h 1 c T f h 2 c T f h 1 7 c T f h 1 / 1 7 c T f h 1 c T f h 2 c T f h 1 7 c T f h 1 / 1 7 c T f h 1 c T f h 2 c T f h 1 7 c T f h 1 / 1 7 c T f h 1 c T f h 2 c T f h 1 7 c T f h 1 / 1 7 cTfh polarization (based on CD69+4-1BB+) Spike (S) Membrane (M) Nucleocapsid (N) B CCR4 CXCR3 CD69 4-1BB Gated on memory CD4+ T cells 78 22 62 38 Th1 Th17 Th1/17 Th2 33 37 16 14 y C 0.10 0.02 0.08 0.99 0.54 0.24 0.34 0.29 0.27 0.47 0.35 0.27 D IFN- TNF Gated on memory CD4+ T cells NC S M N 0 50 100 150 200 0 100 200 300 400 500 r = 0.82 p 0.001 anti-N IgG anti-S IgG S e r o + S e r o − 0.1 1 10 CD4+ S S e r o + S e r o − CD4+ M S e r o + S e r o − CD4+ N S e r o + S e r o − CD8+ S S e r o + S e r o − CD8+ M S e r o + S e r o − CD8+ N S e r o + S e r o − 0.1 1 10 CD4+ S S e r o + S e r o − CD4+ M S e r o + S e r o − CD4+ N S e r o + S e r o − CD8+ S S e r o + S e r o − CD8+ M S e r o + S e r o − CD8+ N 2020 BD MC SC Exp % CTV lo IFN- + * * E F 2 0 1 9 B D S e r o − 2 0 1 9 B D S e r o − CD4+ S *** CD4+ M *** CD8+ S *** CD8+ M *** CD8+ N *** CD4+ N *** 0.1 1 10 2 0 1 9 B D S e r o − 2 0 1 9 B D S e r o − 2 0 1 9 B D S e r o − 2 0 1 9 B D S e r o − Figure S5. Functional Polarization of SARS-CoV-2-Specific Memory CD4+ T Cells, Antibody Correlations, and Comparative Analyses of SARS-CoV-2-Specific CD4+ and CD8+ T Cell Responses versus Serostatus in COVID-19, Related to Figures 3 and 4 (A) Representative flow cytometry plots showing the identification of memory CD4+ T cells responding to overlapping peptides spanning the immunogenic domains of the SARS-CoV-2 nucleocapsid protein by subset (AIM assay). Subsets were defined as CXCR5+ (cTfh), CCR4 CCR6 CXCR3+ CXCR5 (Th1), CCR4+ CCR6 CXCR3 CXCR5 (Th2), CCR4 CCR6+ CXCR3 CXCR5 (Th17), CCR4 CCR6+ CXCR3+ CXCR5 (Th1/17), and CCR4 CCR6 CXCR3 CXCR5 (non-Th1/2/17). (B) Bar graphs summarizing the functional polarization of memory CXCR5+ (cTfh) CD4+ T cells responding to overlapping peptides spanning the immunogenic domains of the SARS-CoV-2 spike (S), membrane (M), and nucleocapsid proteins (N). Data are shown as median ± IQR. Key as in C. (C) Correlation between anti-spike (S) and anti-nucleocapsid (N) IgG levels. Each dot represents one donor. 2020 BD: healthy blood donors from 2020 (n = 31). Exp: exposed family members (n = 28). MC: individuals in the convalescent phase after mild COVID-19 (n = 31). SC: individuals in the convalescent phase after severe COVID-19 (n = 23). Spearman rank correlation. (D) Representative flow cytometry plots showing functional SARS-CoV-2-specific memory CD4+ T cell responses in a seronegative convalescent donor (group MC). Numbers indicate percentages in the drawn gates. NC: negative control. S: spike. M: membrane. N: (legend continued on next page) ll OPEN ACCESS Article
  • 26. nucleocapsid. (E) Dot plots summarizing SARS-CoV-2-specific CD4+ and CD8+ T cell responses versus serostatus in exposed family members (left) and in- dividuals in the convalescent phase after mild COVID-19 (right). Each dot represents one donor. Data are shown as median ± IQR. S: spike. M: membrane. N: nucleocapsid. *p 0.05. Mann-Whitney U test. (F) Dot plots summarizing SARS-CoV-2-specific CD4+ and CD8+ T cell responses in exposed seronegative family members and unexposed healthy blood donors (group 2019 BD). Each dot represents one donor. Data are shown as median ± IQR. *p 0.05. Mann-Whitney U test. ll OPEN ACCESS Article