Do we know now whether viral vector-based vaccines exhibit this sam...
Were there any commonalities (perhaps some sort of comorbidity) bet...
Is there any explanation as to why two patients had no humoral or c...
iScience
Article
Attenuated humoral responses in HIV after SARS-
CoV-2 vaccination linked to B cell defects and
altered immune profiles
Emma Touizer,
Aljawharah
Alrubayyi,
Rosemarie Ford,
..., Emma Morris,
Dimitra Peppa,
Laura E. McCoy
d.peppa@ucl.ac.uk (D.P.)
l.mccoy@ucl.ac.uk (L.E.M.)
Highlights
PLWH have lower levels of
neutralizing anti bodies
(nAbs) after SARS-CoV-2
vaccination
Delayed pro duction o f
nAbs is associated with
greater memory B cell
(MBCs) disturbance
Additional doses increase
nAbs titers and breadth
despite persistent MBCs
perturbations
SARS-CoV-2 vaccina tion
elicits robust T cell
responses in PLWH
PLWH with spike-specific
TcellsbutnonAbshave
increased
CXCR3+CD127+C D8
+
T
cells
Touizer et al., iScience 26,
105862
January 20, 2023 ª 2023 The
Authors.
https://doi.org/10.1016/
j.isci.2022.105862
ll
OPEN ACCESS
iScience
Article
Attenuated humoral responses in HIV
after SARS-CoV-2 vaccination linked to B
cell defects and altered immune profiles
Emma Touizer,
1,8
Aljawharah Alrubayyi,
1,2,8
Rosemarie Ford,
1
Noshin Hussain,
1
Pehue
´
n Pereyra Gerber,
3
Hiu-Long Shum,
1
Chloe Rees-Spear,
1
Luke Muir,
1
Ester Gea-Mallorquı
´
,
2
Jakub Kopycinski,
2
Dylan Jankovic,
1
Anna Jeffery-Smith,
1
Christopher L. Pinder,
1
Thomas A. Fox,
1
Ian Williams,
4
Claire Mullender,
5
Irfaan Maan,
4,5
Laura Waters,
4
Margaret Johnson,
1,6
Sara Madge,
6
Michael Youle,
1,6
Tristan J. Barber,
5,6
Fiona Burns,
5,6
Sabine Kinloch,
1,6
Sarah Rowland-Jones,
2
Richard Gilson,
4,5
Nicholas J. Matheson,
3,7
Emma Morris,
1
Dimitra Peppa,
1,4,6,
*
and Laura E. McCoy
1,8,9,
*
SUMMARY
We assessed a cohort of people living with human immunodeficiency virus
(PLWH) (n = 110) and HIV negative controls (n = 64) after 1, 2 or 3 SARS-CoV-2
vaccine doses. At all timepoints, PLWH had significantly lower neutralizing anti-
bod y (nAb) titers than HIV-negative controls. We also observed a delayed devel-
opment of neutralizat ion in PLWH that was underpinned by a reduced frequency
of spike-specific memory B cells (MBCs). Improved n eutra lization breadth was
seen against the Omicron variant (BA .1) after the third vaccine dose in PLWH
but lower nAb responses persisted and were associated with global MBC
dysfunction. In contrast, SARS-CoV-2 vaccination induced robust T cell responses
that cross-recognized variants in PLWH. Strikingly, individuals with low or absent
neutralization had detectable functional T cell responses. These PLWH had
reduced numbers of circulating T follicular helper cells and an enriched population
of CXCR3
+
CD127
+
CD8
+
T cells after two doses of SARS-CoV-2 va ccination.
INTRODUCTION
People living with human immunodeficiency virus (HIV)[PLWH] appear to be at a higher risk of hospitaliza -
tion and worse clinical outcomes from COVID-19 disease, especially in the cont ext of cellu lar immunosup -
pression and unsuppressed HIV viral load.
1
Although combined antiretroviral therapy (cART) has
dramatically improved life expectancy in PLWH, t he persistence of immune dysfunction raises concerns
about the overall effectiveness and durabilit y of vaccine responses in this potentiall y more vulnerable pa-
tient group, in line with other immunocompromised groups
2,3
As a resul t, PLWH were included in either
priority group 4 (for clinically vulnerable PLWH, based on more advanced immunosuppression or co-mor-
bidities) or 6 ( all other PLWH)
4
in the UK for earlier COVID-19 vaccination than the general population. The
Joint Committee on Vaccin ation and Immunization (JCVI) advised to invite this patient g roup for an addi-
tional booster dose.
4
Previously, defects have been obser ved in serological vaccine responses i n PLWH on
cART. For example, after a full course of hepatitis B
5
or influenza vaccination
6
and long-term responses to
vaccination can b e shorter-lived in PLWH compared to the general population.
7
We and others h ave pre-
viously show n a failure to mount a ro bust antibody response follo wing COVID-19 vaccination in advanced
HIV infection with low CD4 T cell counts b elow 200 c ells/ mL.
8–12
Data on vaccine efficacy and immunogenicity in PLWH remains limited (reviewed in
13
), and although there
are some conflicting results, meta-analyses
14
and recent studies
15
have shown reduced levels of sero con-
version and neutralization after a second dose of viral vector vaccine dose in PLWH receiving cART, with
lower CD4 T cell count/viremia and older age re sulti ng in a more impaired response and more rapid break-
through infection.
16
Assessment of vaccine efficacy has been continually complicated by the ongoing
emergence of variants of concern (VOC), with the Alpha, Beta, Delta and Omicron variants being observed
to progressively evade antib odies
17,18
raised against the original Wuhan-Hu-1 strain in most vaccines. In
particular, data a fte r three v accine doses has been hard to assess because of the emerg ence o f Omicr on
1
Institute for Immunity and
Transplantation, Division of
Infection and Immunity,
University College London,
London, UK
2
Nuffield Department of
Medicine, University of
Oxford, Oxford, UK
3
Cambridge Institute of
Therapeutic Immunology and
Infectious Disease,
Department of Medicine,
University of Cambridge,
Cambridge, UK
4
Mortimer Market Centre,
Department of HIV, Central
and North West London NHS
Trust, London, UK
5
Institute for Global Health,
University College London,
London, UK
6
The Ian Charleson Day
Centre, Royal Free Hospital
NHS Foundation Trust,
London, UK
7
NHS Blood and Transplant,
Cambridge, UK
8
These authors contributed
equally
9
Lead contact
*Correspondence:
d.peppa@ucl.ac.uk (D.P.),
l.mccoy@ucl.ac.uk (L.E.M.)
https://doi.org/10.1016/j.isci.
2022.105862
iScience 26, 105862, January 20, 2023 ª 2023 The Authors.
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
1
ll
OPEN ACCESS
concurrent with vaccination, and in PLWH data on humoral and cellular responses including against Om-
icron remains limited.
19–22
However, the data available thus far suggest that the third vaccine dose provides
a strong boost to antibody responses regardless of the CD4 T cell count, including in those who had
previously not serocon vert ed and induce s robust T cell r esponses.
19,21,22
Moreover, most studies on
SARS-CoV-2 vaccine responses in PLWH to date have mostly focused on e valuating humoral responses
and ge nerat ed limited data on function al T cell responses after 2 doses
23
or 3 doses.
19,20
Therefore, it re-
mains unclear what role HIV-associated immune dysfunction plays in serological and cellular outcome after
SARS-CoV-2 vaccination.
Inferior serological responses to vaccination in PLWH (on cART or unt reate d) are most commonl y linked to
HIV-induced immune destruction of CD4 T cells and imbalance of the CD4:CD8 T cell populations.
24,25
Despite effective cART, chronic i mmune activation in HIV can lead to exhaustion of the adaptive immune
system.
26
This can translate into impaired T cell responses, likely limiting T follicular helper (T
FH
) cell help to
B cells, resulting in lower serologic al outputs. There i s also substantial e viden ce fo r dysfunction/exhaustion
in the B cell compartment during chronic infections that may limit antibody responses against the infecting
pathogen.
27,28
This B cell dysfunction persists to a variable degree after HIV viral suppression following
cART initiation,
29,30
but how these B cell defects impact serological responses to vaccination has not yet
been fully elucidated. Furthermore, there is substantial age-related decline in immune function leading
to senescenc e in both the T and B cell compartments, which may be accelerated in PLWH and c ould further
influence vaccine responses.
31
In this stu dy we h ave eval uated in a well-curat ed c ohort of PLWH on cART and HIV-negative controls
following three SARS-CoV-2 vaccine doses, the rel ation ship between humoral and functional T cel l re-
sponses. To achieve this goal, we have assessed how spike-specific memory B cell (MBC) responses, global
MBC profiles, CD4 and CD8 T cell phenotypes are linked with serological outcomes in PLWH to better un -
derstand which factors may modulat e immune responses to vacci nation.
RESULTS
Lower levels of seroconversion and neutralizing antibodies after SARS-CoV-2 immunization
in PLWH without a history of prior COVID-19 disease
Participants were recruited between January 2021 and April 2022 (n = 110 PL WH on cART and n = 64 HIV-
negative controls) as described in Table 1. Participants were sampled after 1, 2 or 3 doses of a SARS-CoV-2
vaccine and compared cross-sectionally. In addition, in 53 PLWH and 44 controls, responses were assessed
longitudinall y where sequential samples were available. Both study groups (HIV-negative and PLWH) w ere
divided according to their history of either previous SARS-CoV-2 infection (including infection prior or after
vaccination) or as SARS- CoV-2 naive. SARS-CoV-2 spike-specific IgG were tested for binding ag ain st t he S1
subunit of the SARS-CoV-2 spike protein in a semi-quantitative ELISA
32,33
to determine seropositivity.
Neutralizing antibodies (nAbs) were me asured against the ancestral vaccine-matched Wuhan Hu-1
SARS-CoV-2 (WT) strain by pseudovirus neutralization.
33
Approximately 90% of HIV-negative controls
and 80% of PLWH wi th no prior history of SARS-CoV-2 infection seroconv erted. However, alth ough over
82% of cont rols produced a neutr alizing response after o ne vacci ne d ose, only 29% of PLWH did so (Fi g-
ure 1A). As described,
34
prior history of SARS-CoV-2 infection was associated with a higher level of sero-
conversion and the development of nAbs in all individu als at every studied t imepoi nt regardless of HIV sta-
tus (Figure 1A).
Notably, PLWH had lower titers of nAbs than HIV-negative controls at all timepoints regardless of prior
SARS-CoV-2 infection (Figur es 1B and 1C). Overall, a similar trend was seen in binding responses
(Figure s S1A a nd S1B), and nAb titers correlated significantly with both bindi ng titers for S1 IgG and
nAb titers obtained from a live virus neutralization assay (Figures S1C and S1D), as previously reported.
35,36
Given t hat this observational cohort incl udes a mixture o f SARS-CoV-2 vaccine type s, it was notable that
both binding and neutralizing titers remained significantly lower in PLWH compared to controls when
only those who had received m RNA-based vaccines were co nsider ed (Figures S1E and S1F). A similar anal-
ysis for viral vector-based vaccines w as not feasible because of insufficient numbers in the control group.
Both at the pre- and post-third vaccine dose timepoints, there were more SARS-CoV-2 naive PLWH that fail
to produce nAbs (Figures 1A, 1B, and S1A) compared to the control group. Owing to the cross-sectional
nature of the analysis, at the pre-third vaccine dose timepoint additional PLWH were recruited. However,
the observed differences persisted when PLWH were stratified for co-morbidities (Figure S1G).
ll
OPEN ACCESS
2 iScience 26, 105862, January 20, 2023
iScienc
e
Article
Table 1. Co hort demographics
HIV- (n = 64) HIV+ (n = 110)
SARS-CoV-2-
(n = 27)
SARS-CoV-2+
(n = 37)
SARS-CoV-2-
(n = 65)
SARS-CoV-2+
(n = 45)
Clinical parameters
% female 76% 40% 8% 17%
% BAME 28% 24% 21% 44%
Median age (range) 33(21-65) 41(23-66) 53 (22-93) 49 (26-73)
cART 65 (100%) 45 (100%)
HIV viral load Undetectable (<50) Undetectable (<50)
Median CD4 count (range) 602 (22-1360) 560(200-1229)
Median CD4:CD8 ratio (range) 0.74 (0.13–3.05) 0.98 (0.37–2.55)
Co-morbidities
Diabetes, n 1 5 3
Hypertension/CVD, n 3 3 6
Renal disease, n 2 1
Liver disease, n 2 2
Respiratory disease, n 1 3 1
Weakened immune system
inc. cancer/transplant, n
–– 8 1
Advanced HIV/HepB
co-infection, n
–– 4
Other Splenectomy
Sarcoidosis
Splenectomy
Timepoints
Post first dose
N = 17 28 31 32
Median days post-previous
dose (range)
14 (12-74) 19 (12-60) 20 (12-102) 22 (13-82)
Vaccine (AZ |Moderna |Pfizer) 3 | 2 | 12 3 | 1 | 24 19 | 2 | 10 21 | 0 | 11
Post second dose
N = 18 25 30 24
Median days post-previous
dose (range)
39 (23-67) 26 (15-68) 20 (7-48) 21 (9-52)
Vaccine (AZ | Moderna | Pfizer) 3 | 3 | 12 2 | 1 | 23 17 | 1 | 12 12 | 0 | 12
Pre third dose
N = 21 26 39 16
Median days post-previous
dose (range)
129 (75-258) 129 (76-236) 125 (72-218) 119 (86-317)
Vaccine (AZ | Moderna | Pfizer)
Post third dose
N = 14 25 34 17
Median days post-previous
dose (range)
21 (13-43) 43 (18-129) 40 (9-149) 65 (7-140)
Vaccine (AZ |Moderna | Pfizer) 0 | 3 | 12 0 | 2 | 23 1 | 2 | 32 1 | 0 | 16
Cohort demographics, clinical characteristics and number of participants per timepoints for each group. All PLWH participants included in this study were on
cART. AZ = AZD1222; Moderna = mRNA-1273; Pfizer = BNT162b2.
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 3
iScienc
e
Article
Figure 1. Weaker post vaccination antibody responses in SARS-CoV-2 naive PLWH
(A) Percentage of individuals with detectable neutralizing antibody response, non-neutralizing but binding response, or seronegative at each timepoint as
color-coded in the key. The headings above each graph show HIV status and previous SARS-CoV-2 exposure. N numbers for each group are indicated above
each column.
(B) WT pseudovirus neutralization reciprocal 50% inhibitory titers (ID50) in PLWH (blue) compared to HIV-negative controls (gray) stratified by vaccination
timepoint (on the x axis) for individuals without prior SARS-CoV-2 infectio n. The dotted line represents the lower limit of the assay (ID50 = 1:20). Where no
neutralization was detected, samples were assigned an ID50 of <1:20 a s this was the limit of assay detection. Each data point represents the mean of n = 2
ll
OPEN ACCESS
4 iScience 26, 105862, January 20, 2023
iScienc
e
Article
Longitudinal samples fro m 53 PLWH and 44 controls we re then ev aluate d to assess binding ant ibody re-
sponses and nAb s over time after each vaccine dose. These i ncluded samples after the firs t dose and for
at least one additional timepoint, often including a baseline, post-second, pre-third and post-third sample
(Figure 1D). This analysis rev eal ed two clear t raje ctorie s of the de velopmen t of neut rali zation , firstly wh ere
nAbs were detected after a single vaccine dose,
37
defined here as ‘‘standard neutra lizati on’’, and secondly
where neutral izatio n was no t ach ieved u ntil after the second dose or later, defined as ‘delayed neutraliza-
tion’’. Most HIV- negati ve controls without prior SARS-CoV-2 infection show a standard neutralizat ion pro-
file, with only 3 individuals failing to mount a neutralizing response unti l after the second dose (Figure 1D),
and a similar effect was seenwithbindingresponses(Figures S1H–S1K). In contrast, two-thirds of SARS-
CoV-2 naive PLWH di d not make a detectable neutrali zing response until after the second dose and a sub-
stantial proportion of them lost detectable neutralizing activity before the third dose ( Figure s 1Aand1E).
However, both PLWH and HIV-negative controls with ahistoryofSARS-CoV-2infectionmadeastandard
neutralizing response (Figures 1F and 1G ). Therefore, having iden tified this delay ed neutralizati on pheno-
type in SARS-CoV-2 naive PLWH, we have evaluated its relationship with total CD4 T cell counts, which are
known to be important for SARS -CoV-2 vaccine respo nses in PLWH.
8–10,12
No significant difference was
seen in median CD4 T cell count or CD4:CD8 T cell ratio between PLWH with standard or delayed neutral-
ization profiles (Figure s 1H and 1I); or correlate either with th e rapid development of neutralizatio n (Fig-
ure S1MandS1N).
Delayed neutralization is associated with lower frequency of spike-specific MBCs and a
perturbed MBC global phenotype
Spike is the SARS-CoV-2 glycopr otein and is the sole ant igen in most vaccines. It ha s been previousl y shown
that i nfection and vaccination produce spike-specific MBCs in proportio n to serological responses.
38–42
Given that the delay in neutralization observed more frequently in PLWH was not clearly associated with
peripheral CD4 T cell counts, we next assessed the relationship with global MBCs and spike-reactive
MBC frequency and phenotype, using a previously validated flow cytometry panel, with memory B cells
defined as CD19
+
CD20
+
CD38
lo/-
IgD- (Figure S2). This analysis was performed on available PBMC samples
after the first vaccine dose, using SARS-CoV-2 naive baseline samples to determine the antigen-specific
gate (Figure 2A). We observed a significantly lower frequency of spike- spe cific MBCs in SA RS-CoV -2 nai ve
participants after the first dose as compared to those with a history of prior infection, regardless of HIV sta-
tus (Figure 2B). Moreover, a l ower freque ncy of spike-sp ecific MBCs was observed in SARS-CoV-2 naive par-
ticipants who had a delayed neutralization response, although notably there were a small number of donors
in the standard neutralization group (Figure 2C).Inlinewiththis,thepercentageofspike-specicMBCs
showed a strong correlation with the nAb titer (Figure 2D) in agreement with previous findings during
SARS-CoV-2 convalescence .
42
Subsequent gating on CD21 and C D27 expression all owed the identi fication of four popul ations of class-
switched MBCs: CD21
CD27
atypical MBCs (also known as tissue-like memory); CD21
CD27
+
activated
MBCs; CD21
+
CD27
+
classical resting MBCs and C D21
+
CD27
switched naive (also known as intermediate
memory) MBCs (Figure 2E) as previously described.
42
Global defects i n the balance of the se MBC subsets
have been identified previously in PLWH (reviewed in
30
), i nclud ing those on cART,
43
with increased
numbers of activated and atypical MBCs concurrent with a decrease in resting MBCs. This phenotype is
exemplified in (Figure 2E) for a PLWH and an HIV-negative control. We have hypothesized that these
inherent defects may have an impact on the quality of serological responses after SARS-CoV-2 vaccination.
Figure 1. Continued
biological repeats, each measured in duplicates. N numbers match those in (A). Line represents median for each group. Statistical test: Mann Whitney
U-test (MWU).
(C) Shows the equivalent data for those with prior SARS-CoV-2 infection, N numbers match those in (A).
(D) Longitu dina l ID
50
titers for HIV-negative controls without prior SARS-CoV-2 infection who provided at least two longitudinal samples, including a post
first dose sample. Samples that were neutralizing after the first dose are categorized as exhibiting a standard neutralizing response and colored gray,those
that only achieve neutralization after the second dose, exhibit a delayed neutralizing response and are color-coded in magenta. N numbers for each
category are indicated o n the graph.
(E) Shows the equivalent data for PLWH without prior SARS-CoV-2 infection.
(F) Shows the equivalent data for HIV-negative controls with prior SARS-CoV-2 infection.
(G) Shows the equivalent data for PLWH with prior SARS-CoV-2 infection.
(H) CD4 T cell counts and (I) CD4:CD8 T cell ratio for PLWH stratified by standard (gray) or delayed neutralization (magenta). N numbers are as per D-G. Line
represents median for each group. Statistical test: MWU. *p > 0.05; **p > 0.01; ***p > 0.001 and ****p > 0.0001. See also Figure S1.
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 5
iScienc
e
Article
ll
OPEN ACCESS
6 iScience 26, 105862, January 20, 2023
iScienc
e
Article
Global phenotyping of the MBC response after the first vaccine dose revealed that individuals with delayed
neutralization, consisting largely of PLWH, had significa ntly lower numbers of restin g MBCs (CD21
+
CD27
+
)
and greater numbers of both CD21
CD27
+
activated MBCs and CD21
CD27
atypical MBCs compared to
those with standard neutralization (Figure 2F). Moreover, lower frequencies of resting MBCs correlated
with lower nAb titers (Figure 2G), this a ssociation is driven by participants with standard n eutralization
and some individuals with delayed neu tral izatio n do have reasonable n umbers of resting MBCs. Similarly,
higher levels of at ypical MBCs significantly correla te d with lower nAb tit ers when considering those with
standard neutralization, although the strength of this association was relatively weak (r = 0.4867) ( Fig-
ure 2H). Frequencies of more than 10% atypical MBCs were only observed in the delayed neutralization
group. Tog ether these fin din gs suggest that the MBC subset pertu rbati ons seen in PLWH could account
for the low er serol ogical output.
Improved neutralization breadth after the thirdSARS-CoV-2doseinPLWHbutlowernAb
responses persist and are associat ed with global, but no t spike-speci fic, MBC dysfunction
To assess the breadth of nAb responses across the cohort, samples from all timepoints were tested against
an Omicron pseudovirus (BA.1 strain), which represented the dominant circulating strain at the time of
the post third vaccine dose sampling. Owing to the substantial antigenic changes in the Omicron spike,
44
in
participants with no prior infection, over 50% of HIV-negative controls and more than 90% of PLWH were
not able to neutralize Omicron after the first vaccine dose (Figure 3A). The second dose enabled most of
the control group to mount a neutralizing response whereas only a quarter of SARS-CoV-2 naive PLWH
had nAbs against Omicron. In the SARS-CoV-2 naive groups, the third dose enabled 100% of HIV-negative
controls to neutralize Omicron and increased the frequency of neutralization among PLWH to over 70%
(Figure 3A). As in the analysis of WT neutralization for individuals without prior SARS-CoV-2 infection,
median Omicron ID
50
titers were lower in SARS-CoV-2 naive PLWH compared to HIV-negative controls at all
timepoints (Figure 3B). In addition, there was no significant difference when individuals with complex co-mor-
bidities were removed from the PLWH cohort at the third vaccine dose (Figure S3C) or whether they had pre-
viously been infected with SARS-CoV-2. These data suggest that the third vaccine dose was effective in both
boosting nAb titer and broadening the response to Omicron, especially in SARS-CoV-2 naive PLWH, thus
rendering their responses closer to those of SARS-CoV-2 naive HIV-negative controls (Figures 3B and 3C).
Next, we evaluate d cross-sectionally the B cell phenotype after th e third v acci ne dose. In contrast to the
first vac cine dose, there was no significant difference between the frequency of spike-specific MBCs
when individ uals were stratified by whether th ey had been previously infected with SARS -CoV- 2 or not (Fig-
ure 3D) regardless of HIV status. However, the frequency of spike-specific MBCs after the third dose corre-
lated with Omicron titers (Fi gure 3E).Thissuggeststhatafterthreevaccinedosestheseindividualshad
mounted a specific B cell response, and that the quantity of spike-specific B cells remained linked to the
improved neutralization pote ncy and breadth obser ved (Figure s 3A–3C). Given that all individuals as-
sessed after the third dose made a robust spike-specific MBC response, we wanted to evaluate further
Figure 2. Neutralization titer is a ssociat ed wit h the frequency of spike-specific MBCs after t he first vaccine dose
(A) Spike-specific MBCs (CD19
+
CD20
+
CD38
lo/mid
IgD-excluding switched naive CD27
CD21
+
cell) according to dual positivity for spike-PE and spike-APC
to exclude non-specific binding in a representative naive pre-vaccine sample (left) or representativ e post-vaccine sample (right) after the first vaccine dose.
(B) Percentage of spike-specific MBC after the first vaccine dose stratified by prior SARS-CoV-2 infection. Line represents median for each group. Statistical
test: M-Whitney U test (MWU). Dotted lines represent lower limit of sensitivity of the assay (0.1% spike-specific MBCs, based on previous optimization).
42
(C) Percentage of spike-specific MBCs in SARS-CoV-2 naive donors after the first vaccine dose, s tratified by delayed (magenta) or standard (gray )
neutralization profile. Line represents median for each group. Statistical test: MWU. Dotted lines represent lower limit of sensitivity of the assay (0.1% spike-
specific MBCs).
(D) Correlation of the percent of spike-specific MBC with WT ID
50
titers stratified by PLWH (blue) and controls (gray) after the first dose, statistical test:
Spearman’s rank correlation coefficient.
(E) Distribution of MBCs (CD19
+
CD20
+
CD38
lo/mid
IgD-) subtypes according to CD27-BUV395 and CD21-BV711 in a representative HIV-negative donor
sample (left) or PLWH donor sample (right).
(F) Percentage of MBC subtypes (activated CD27
+
CD21
; resting CD27
+
CD21
+
; switched naive; switched naive CD27
CD21
+
and CD27
CD21
atypical)
after the first vaccine do se stratified by delayed or standard neutralization profile. Line represents median for ea ch group. Statistical test: MWU.
(G) Correlation of the percentage of resting CD27
+
CD21
+
MBCs with WT ID
50
titers stratified by delayed (magenta) or standard (gray) neutralization profile
after the first vaccine do se, statistical test: Spearman’s rank correlation coefficient.
(H) Correlation of the percent of switched naive CD27
CD21
+
MBCs with WT ID
50
titers stratified by delayed (magenta) or standard (gray) neutralization
profile after the first vaccine dose, statistical test: Spearman’s rank correlation coefficient. *p > 0.05; **p > 0.01; ***p > 0.001 and *** *p > 0 .0001.
See also Figure S2.
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 7
iScienc
e
Article
ll
OPEN ACCESS
8 iScience 26, 105862, January 20, 2023
iScienc
e
Article
whether alt erat ions in spike-specifi c MBC phenotype also contribut ed to difference s i n serum neutraliza-
tion (Figures 3A, 3B, 3F, S3A, and S3B). Spike-specific B cell s wer e found to be comparable across the
different MBC subsets in both PLWH and HIV-negative controls, except for a trend to fewer spike-specific
resting MBCs in PLWH as compared to controls (Figure 3G).ThiswasthecaseeventhoughtheglobalMBC
population for these post third vaccine dose samples showed classical anomalies in MBCs associated with
HIV infection (Figure 3H). T hese data suggest that SA RS-Co V-2 serum antibody responses are lower poten -
tially because of a global MBC disturbance thereby limiting the overall B cell response. I n line with this p ro-
posal, we anticipated that underlying global MBC disturbances wo uld also influence the efficiency of the
antigen-specific B cell response in other ways, beyond limiting the number of spike-specific MBCs, for
example by limiting class-switching. Indeed, this is supported by our data showing similar levels of IgG+
andIgM+globalMBCsinbothgroups(Figure 3I) but a significantly lower level of spi ke-speci fic IgG+
MBCs in PLWH after the third vaccine dose as c ompared to controls, and conver sely a higher frequen cy
of spike-specific IgM+ MBCs (Figur e 3J).
SARS-CoV-2 vaccination induces robust T cell responses that cross-recognize variants in
PLWH
To increase our understanding of the complementary role of cellular immunity after vaccination, we have exam-
ined T cell responses in our cohort, including their reactivity to SARS-CoV-2 variants. The magnitude of spike-
specific T cell responses was assessed cross-sectionally by IFN-g-ELISpot using overlapping peptide (OLP)
pools covering the complete sequences of the WT spike glycoprotein as previously described.
45
The majority
of PLWH had detectableSARS-CoV-2-specific T cell responsesatlevels comparable to HIV-negativeindividuals
following each vaccine dose (Figures 4A–4C). A greater magnitude of spike-specific T cells was observed in in-
dividuals with prior SARS-CoV-2 infection, irrespective of HIV status (Figures 4A–4C) in keeping with previous
reports.
34,46,47
There were no detectable T cell responses in a small number of PLWH with no prior exposure to
SARS-CoV-2 across all timepoints. These were participants with incomplete immune reconstitution on cART
and/or additional co-morbidities, such as transplant recipients on immunosuppressive therapy (Figures 4A–
4C). Next, we examined the longitudinal evolution of T cell responses in a subgroup of donors with available
PBMC samples. In SARS-CoV-2 naive individuals, spike-specific T cell responses increased following the first
vaccine dose, peaked after the second dose and were maintained after the third vaccine dose (Figure 4D). In
one HIV-positive, SARS-CoV-2-naı
¨
ve donor with advanced immunosuppression and persistently low CD4
T cell count of 100 cells/mL on cART, a third dose (mRNA) vaccine was able to elicit a T cell response despite
no evidence of neutralization (Figure 4D). A higher proportion of PLWH without prior SARS-CoV-2 infection
had detectable T cell responses at baseline compared to HIV-negative controls, which could represent the
presence of cross-reactive responses to other pathogens, probably to related coronaviruses (Figure 4D).
48–52
However, due to the small number of participants with detectable T cell responses at baseline, this study was
Figure 3. Improved neutralization against Omicron after the third vaccine dose in PLWH accompanied by minimal alteration in the spike-specific
MBC phenotype
(A) Percentage of individuals with detectable neutralizing response, non-neutralizing but binding response, or seronegative at each timepoint as co lor-
coded in the key (neutralization against Omicron pseudovirus). Headings above each graphshow the HIV status and previous SARS-CoV-2 exposure. N
numbers for each group are indicated above each column.
(B) Omicron pseudovirus neutralization ID
50
in PLWH (blue) compared to HIV-negative controls (gray) stratified by vaccination timepoint (on the x axis) for
individuals without prior SARS-CoV-2 infection. The dotted line represents the lower limit of the assay (ID
50
= 1:20). Each data point represents the mean of
n = 2 biological repeats, each measured in duplicates. Line represents median for each group. Statistical test: Mann-Whitney U test (MWU).
(C) Shows the equivalent data for those with prior SARS-CoV-2 infection, N numbers match those in (A).
(D) Percentage of spike-specific MBCs in PLWH (blue) and HIV-negative donors (gray) after the third vaccine dose stratified by SARS-CoV-2 infection. Line
represents median for each group. Statistical test: MWU.
(E) Correlation between Omicron ID
50
titers and percentage of spike-specific MBCs in PLWH (blue) and HIV-negative donors (gray) after the third vaccine
dose. Statistical test: Spearman’s rank co rrelation coefficient.
(F) Representative gating strategy to identify spike-specific MBCs subtypes.
(G) Percentage of spike-specific MBCs subtypes (activated CD27
+
CD21
; resting CD27
+
CD21
+
; switched naive CD27
CD21
+
and CD27
CD21
atypical)
after the third vaccine dose in PLWH (blue) and HIV-negative donors (gray). Line represents media n for each group. Statistical test: MWU.
(H) Percentage of MBCs subtypes (activated CD27
+
CD21
;restingCD27
+
CD21
+
; s witched naive; switched naive CD27
CD21
+
and CD27
CD21
atypical)
after the third vaccine dose in PLWH (blue) and HIV-negative donors (gray). Line represents media n for each group. Statistical test: MWU.
(I) Percentage of IgG and IgM in MBCs (excluding switched naive CD27
CD21
+
fraction) after the third vaccine dose in PLWH (blue) and HIV-negative donors
(gray). Line represents median for each group. Statist ical test: MWU.
(J) Percentage of IgG and IgM in spike-specific MBCs a fter the third vaccine dose in PLWH (blue) and HIV-negative donors (gray). Line represents median for
each group. Statistical test: M WU. *p > 0.05; **p > 0.01; ***p > 0.001 and ****p > 0.0001.
See also Figure S3.
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 9
iScienc
e
Article
esoddr3esoddn2esodts1
ED
F
1
st
d
os
H
3r
dd
o
s
e
G
2n
dd
o
s
HIV+SARS-CoV-2-
HIV+SARS-CoV-2+
A
1
10
100
1000
Spike-specific T cells
S.F.U/10
6
PBMCs
23
HIV- HIV+
Vaccine dose
2323
23
SARS-CoV-2 naive prior SARS-CoV-2
HIV- HIV+
1
10
100
1000
10000
Spike-specific T cells
S.F.U/10
6
PBMCs
0123
2-VoC-SRASroirpevian2-VoC-SRAS
+VIH-VIH HIV-HIV+
Vaccine dose
012 301 2301 23
CB
1 10 100 1000 10000
0
500
1000
1500
CD4 count (cells per µl)
r=0.5153
p=0.0239
r=0.1367
p=0.6873
SARS-CoV-2 naive
prior SARS-CoV-2
1 10 100 1000 10000
0
500
1000
1500
CD4 count (cells per µl)
r=0.4596
p=0.0121
r=-0.2642
p=0.4291
SARS-CoV-2 naive
prior SARS-CoV-2
1 10 100 1000 10000
0
500
1000
1500
CD4 count (cells per µl)
r=0.3578
p=0.0567
r=0.3164
p=0.2903
SARS-CoV-2 naive
prior SARS-CoV-2
Spike specific T cells
ΔSFU/10
6
PBMC
Spike specific T cells
ΔSFU/10
6
PBMC
Spike specific T cells
ΔSFU/10
6
PBMC
1
10
100
1000
10000
Spike-specific T cells
S.F.U/10
6
PBMCs
SARS-CoV-2 naive prior SARS-CoV-2
HIV- HIV+
HIV- HIV+
1
10
100
1000
10000
Spike-specific T cells
S.F.U/10
6
PBMCs
SARS-CoV-2 naive prior SARS-CoV-2
HIV- HIV+
HIV- HIV+
1
10
100
1000
10000
Spike-specific T cells
S.F.U/10
6
PBMCs
SARS-CoV-2 naive prior SARS-CoV-2
HIV- HIV+
HIV- HIV+
Figure 4. Comparable magnitude of s pike-speci fic T cell responses followingSARS-CoV-2vaccinationinHIV-positive and HIV-negative individuals
(A–C) Cross-sectional analysis of t he magnitude of the IFN-g-ELISpot responses to the SARS-CoV-2 spike peptide pools in HIV-negative (gray) and HIV-
positive (blue) individuals, with or without prior SARS-CoV-2 infection follow ing first dose (A) second dose (B) and third dose (C). (HIV-SARS-CoV-2-firstdose
n = 9, second dose n = 18 , third dose n = 14; HIV+ SARS-CoV-2- first dose n = 15, second dose n = 29, third dose n = 31; H IV-SARS-CoV-2+ first dose n = 23,
second dose n = 27, third dose n = 20; HIV+ SARS-CoV-2+ first dose n = 12, second dose n = 13, third dose n = 15). Statistical test: Mann-Whitney U-test
(MWU), line represents mean with SD for each group.
(D) Longitudinal analysis of the spike specific T cell responses in PLWH and HIV-negative subjects. Statistical test: Wilcoxon matched-pairs sign rank test
(WMP).
(E) Longitudinal and cross-sectional analysis of the magnitude of T cell responses to Omicron after two or three vaccine doses (n = 11 H IV-SARS-CoV-2-,n=
20 HIV + SARS-CoV-2-, n = 22 HIV-SARS-CoV-2+, n = 10 HIV + SARS-CoV-2+). Statistical test: MWU and WMP.
(F–H) Correlation between the CD4 T cell count in HIV-positive individuals and magnitude of spike-specific T cell responses after first dose (F), secon ddose
(G), and (H) third dose. Statistical test: Spearman’s rank correlation coefficient.
*p > 0.05; **p > 0.01; ***p > 0.001 and ****p > 0.00 01. See also Figure S4.
ll
OPEN ACCESS
10 iScience 26, 105862, January 20, 2023
iScienc
e
Article
F
1
10
100
1000
10000
Spike specific T cells
ΔSFU/10
6
PBMC
151-1000 >100021-151
ID
50
WT
1st dose 2nd dose
3rd dose
BA
C
D
3rd dose
SARS-CoV-2 naive HIV-
G
SARS-CoV-2- naive HIV+
3rd dose
2nd dose
SARS-CoV-2- naive HIV+
E
1 10 100 1000 10000
10
100
1000
10000
100000
Detection limit
Low level of nAb
ID
50
WT
r=0.5896
p=0.0009
r=0.6262
p=0.0001
HIV- HIV+
1 10 100 1000 10000
10
100
1000
10000
100000
Detection limit
Low level of nAb
ID
50
WT
r=0.3912
p=0.0221
r=0.3715
p=0.0110
HIV- HIV+
HIV-SARS-CoV-2-
HIV-SARS-CoV-2+
HIV+SARS-CoV-2-
HIV+SARS-CoV-2+
Spike specific T cells
ΔSFU/10
6
PBMC
Spike specific T cells
ΔSFU/10
6
PBMC
Spike specific T cells
ΔSFU/10
6
PBMC
Spike specific T cells
ΔSFU/10
6
PBMC
Spike specific T cells
ΔSFU/10
6
PBMC
Spike specific T cells
ΔSFU/10
6
PBMC
SARS-CoV-2 naive HIV-
2nd dose
1
10
100
1000
10000
151-1000
>1000
20 0001>051-12 151-1000
20 21-150 151-1000151-1000
1 10 100 1000 10000
10
100
1000
10000
100000
Detection limit
Low level of nAb
ID
50
WT
r=0.5402
p=0.0014
r=0.6755
p=0.0001
HIV- HIV+
ID
50
>1000
ID
50
20
ID
50
=21-151
ID
50
=151-1000
ID
50
>1000
ID
50
=151-1000
1
10
100
1000
10000
1
10
100
1000
10000
HIV-SARS-CoV-2-
HIV-SARS-CoV-2+
HIV+SARS-CoV-2-
HIV+SARS-CoV-2+
HIV-SARS-CoV-2-
HIV-SARS-CoV-2+
HIV+SARS-CoV-2-
HIV+SARS-CoV-2+
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 11
iScienc
e
Article
not powered to detect any association between the presence of cross-reactive T cells and magnitude of vac-
cine-induced T cell responses. In donors with prior SARS-CoV-2 infection, there was a boosting effect to
spike-specific T cells following the first vaccine dose in both study groups (Figure 4D). In parallel we have tested
T cell responses to CMV-pp65 and HIV-gag peptide stimulation within the same individuals across all time-
points. Overall, PLWH with no prior exposure to SARS-CoV-2 had robust responses to CMV-pp65 stimulation,
as expected given their higher CMV seroprevalence compared to HIV negative donors. CMV-specific re-
sponses in these individuals were higher compared to SARS-CoV-2 and Gag-specific responses following
each vaccine dose (Figures S4A–S4C). Prior SARS-CoV-2 exposure resulted in comparable SARS-CoV-2 and
CMV-pp65 T cell responses after the third vaccine dose in PLWH (Figure S4C). No significant differences
were detected between SARS-CoV-2 and CMV-specific responses in HIV-negative individuals (Figures S4A–
S4C). Overall, these results demonstrate a robust induction of T cell responses to SARS-CoV-2 vaccination in
PLWH despite attenuated antibody responses.
Previous work has demonstrated that T cell responses are largely retained against variants of concern (VOCs),
including the highly transmissible BA.1 Omicron variant, and therefore may be important when antibody levels
wane or new variants emerge that can partly escape antibody responses. To determine T cell reactivity to
VOCs, we assessed T cell responses to the mutated regions, including Omicron, in our study cohort. The
magnitude of T cell responses against B.1.1.529 was comparable between PLWH and HIV-negative donors
regardless of prior SARS-CoV-2 infection (Figure 4E). Notably, responses were further enhanced by a third vac-
cine dose in all donors, irrespective of prior SARS-CoV-2 infection or HIV status and in keeping with the bene-
ficial effect of a third vaccine dose in boosting humoral responses (Figure 4E). T cell reactivity to Omicron and
other VOCs, including Alpha, Beta and Delta, was comparable between HIV-negative and PLWH with or
without prior SARS-CoV-2 infection after three vaccine doses, and these responses were maintained against
the ancestral Wuhan Hu-1 spike peptide pool, reinforcing the relative resilience of T cell responses to spike
variation (Figures S4D and S4E). We noted that three HIV-negative and five HIV-positive individuals, regardless
of prior SARS-CoV-2 infection, had no detectable T cell responses to the Wuhan Hu-1 peptide pool, covering
only the affected regions of spike. This could be because of the VOC mutations occurring in regions that are
poorly targeted by T cell responses in some individuals.
34
Although spike-specific T cell responses were detected at similar frequencies across all groups (Figures 4A–
4C), there was variation in the magnitude of responses. To better understand the factors underlying this het-
erogeneity, we examined the role of various HIV parameters.
45
We have previously reported an association
between the CD4:CD8 T cell ratio and total SARS-CoV-2 responses, especially against the nucleocapsid
(N) and membrane (M) protein, in PLWH recovering from COVID-19 disease.
45
No correlation was observed
between the CD4:CD8 T cell ratio and spike-specific T cell responses following vaccination in our cohort
(Figures S4F–S4H). However, a positive correlation was detected between the CD4 T cell count and spike-spe-
cific T cell responses after the first vaccine dose (r = 0.5153) in SARS-CoV-2 naive PLWH (Figure 4F). This asso-
ciation was weaker after the second vaccine dose (r = 0.4596) and non-significant after the third dose
(Figures 4G and 4H). Together these observations suggest that an effective helper T cell response could drive
the induction of cellular immunity following vaccination in individuals without prior exposure to SARS-CoV-2.
However, the lack of an association between CD4 T cell counts and antibody responses further underlines the
relative importance of HIV-associated B cell defects in modulating the induction of effective humoral immunity
in addition to potential insufficient T cell priming.
A proportion of PLWH had low or absent nAbs (ID
50
< 1:150) but detectable T cell responses
following vaccination
We examined next the relationship between humoral and cellular re sponses by comparing antibo dy r e-
sponses and neutral izati on titers with T cell responses det ected by IFN-g-ELISpot following SARS-CoV-2
Figure 5. Interrelations between humoral and cellular responses following SARS-CoV-2 vaccination in HIV-positive and HIV-negative individuals
(A–C) Correlation of spike-specific T cell responses with nAb titers after rst dose (A) second dose (B) and third dose (C) of vaccine in HIV-negative andHIV-
positive donors, with or without prior SARS-CoV-2 infection (limit of detection ID
50
= 1:20, low level of nAb ID
50
= 1:150). Statistical test: Spearman’s rank
correlation coefficient.
(D and E) Hierarchy of the spike-specific T cell responses ordered by their n Ab titers in HIV-negative (D) and HIV-positive (E) SARS-CoV-2 naive donors after
two vaccine doses.
(F and G) Hierarchy of the spike-specific T cell responses after three vaccine doses in HIV negative (F) and positive (G) SARS-CoV-2 naive participants.*p>
0.05; **p > 0.01 ; ***p > 0.001 and ****p > 0.0001.
See also Figure S5.
ll
OPEN ACCESS
12 iScience 26, 105862, January 20, 2023
iScienc
e
Article
PLWH SARS-CoV- nAb
low
/
-
T
+
PLWH SARS-CoV- nAb
high
T
+
tSNE1
tSNE2
ab
- or low
ab
high
SARS-CoV-2 naive nAb
low
/
-
T
+
SARS-CoV-2 naive nAb
high
T
+
tSNE1
tSNE2
2nd dose PLWH CD4 T cells
A
0 5000 10000 15000 20000
nAb
high
T
+
nAb
-/low
T
+
0 5000 10000 15000 20000
Cluster cell count
nAb
-/low
T
+
nAb
high
T
+
Gated on CCR7
+
CD45RA
+
(naive CD8)
nAb
low
/
-
T
+
nAb
high
T
+
PLWH SARS-CoV-2 naive
CXCR3
+
EM CD4
CXCR5
+
CXCR3
+
T
FH
CD127
low
CXCR3
+
CCR7
+
CD4
CXCR3
low
CD127
+
CCR7
+
CD4
CXCR3
-
CM CD4
CD127
-
naive CD4
CD38
+
PD-1
+
CXCR3-CXCR5
+
CXCR3
+
PD-1
+
T
FH
CD127
+
CCR7
+
CD4
CXCR3
-
EM CD4
CD4
PE/Dazzle™ 594
CXCR3 PE/Cy-5CXCR5 BB515
CD4
PE/Dazzle™ 594
CD4
PE/Dazzle™ 594
CXCR3 PE/Cy-5CXCR5 BB515
CD4
PE/Dazzle™ 594
SARS-CoV-2 naive nAb
low
/
-
T
+
SARS-CoV-2 naive nAb
high
T
+
CXCR3
+
CD127
+
CD38
+
naive CD8
CXCR3
+
CD127
+
CD38
-
CM CD8
CXCR3
+
CD127
+
EM CD8
CXCR3
low
CD127
low
TEMRA CD8
CXCR3
-
CD127
-
naive CD8
CXCR3
low
CD127
+
CD38
+
CM CD8
CXCR3
+
PD-1
+
CD38
+
CCR7
low
CD8
CD127
low
CD38
low
EM CD8
CXCR3
+
CD127
+
TEMRA CD8
CD127
low
CD38
hi
EM CD8
CD127 BV650
CXCR3 PE/Cy-5
CD8 BV711
CD38 BV785
PLWH SARS-CoV-2 naive nAb
-/low
T
+
B
EDC
F
G
JIH
2nd dose PLWH CD8 T cells
10 100 1000
0
10
20
30
40
% of CXCR3
+
CD127
+
CD38
+
naive CD8 T cells
r=0.8740
p=0.0035
0 1020304050
10
100
1000
10000
% of CXCR5
+
CXCR3
+
CD4 T cells
ID
50
WT
r=0.5294
p=0.0238
Detection limit
Low level of nAb
Spike specific T cells
ΔSFU/10
6
PBMC
Cluster cell count
61.39
0-10
3
10
3
10
4
10
5
0
10
3
10
4
10
5
7.76
0 10
3
10
4
10
5
0
-10
3
10
3
10
4
10
5
39.51
0-10
3
10
3
10
4
10
5
0
10
3
10
4
10
5
32.48
0 10
3
10
4
10
5
0
-10
3
10
3
10
4
10
5
17.1281.51
0 10
3
10
4
10
5
0
10
3
10
4
10
5
15.0183.58 26.5570.93
38.8658.49
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
0
10
3
10
4
10
5
0-10
3
10
3
10
4
10
5
0
10
3
10
4
10
5
nAb
-/low
nAb
high
0
10
20
30
40
50
% of CXCR5
+
CXCR3
+
T
FH
nAb
-/low
nAb
high
0
10
20
30
40
% of CXCR3
+
CD127
+
CD38
+
naive CD8 T cells
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 13
iScienc
e
Article
vaccination. Overall, spike-specific T cells following the first, second and third vaccine doses correlated
positively with respective nAb titers in HIV-negative and PL WH. These associati ons were strong er in
PLWH after the first (r = 0.5402; p = 0.0014) and second dose o f vac cine (r = 0.5038, p = 0.0004), similarly
to HIV-negative controls (Figures 5A–5C). Similar associations were observed for S1 IgG binding titers
(Figure s S5A–S5C). One HIV-positive SARS-CoV-2 naive donor with a low CD4 T cell count of 40 cells/mL
on cAR T, and one individ ual with r elapsed lymphoma, both h ad no dete ctable humoral and cell ular re-
sponses after 2 or 3 doses of mRNA vaccine. A proportion of PLWH, i n particul ar those without prior
SARS-CoV-2 infection, had low or absent nAbs (ID
50
<1:150) but detectable T cell responses following vacci-
nation (Figures 5A–5C). To better visualize these relationships in SARS-CoV-2 naive individuals, we ranked
T cell responses after second and third doses accordin g to the magnitude of neutralizin g antibodi es
(Figure s 5E–5G). All of the HIV-negative donors had detectable cellular and neutralizing antibodies (Fig-
ure 5D). However, a prop ortion of SARS-CoV-2 naive PLWH with low or absent nAbs (n = 9 out of 10)
had measurabl e cellular responses to the spike protein after two vaccine doses (Figure 5E). These donors
were all controlled on cART with a median CD4 T cell count of 680 cells/mL and no significant underlyi ng co -
morbidity (Table S1). Although all HIV-negative individuals had both detectable n Abs and cellular re-
sponses post third dose (Figure 2F), a small number of PLWH SARS-CoV-2 naive donors ( n = 7 out of 9)
had detectable T cell responses in the absence of, or only low-level, neutralization (Figure 5G). Similarly,
these donors were all well controlled on cART with a median CD4 T cell count of 492 cells/mL. One of these
donors who presented with adv anced HIV infect ion had a persist entl y low CD4 T cell count (100 cells/ mL),
and one of the donors recruited after a third vaccine dose had a previous splenectomy. These data suggest
that in a small proportion of PLWH, serological non-responders or with evidence of low-level neutrali zation,
cellular immune responses may play an important compensatory role.
PLWH with suboptimal serological responses demonstrate an expansion of CXCR3
+
CD127
+
CD8
+
T cells after two doses of SARS-CoV-2 vaccination
The presence of detectable T cell responses in a subgroup of SARS-CoV-2 naive HIV-positive donors with
low or absent nAbs after two or three vaccine doses prompted us to furthe r evalua te the phenot ype of the
T cell compartment. We have compared T cell immune signatures in SARS-CoV-2 naive PLWH with potent
neutralization titers (>1:150) and functional T cell responses (PLWH SARS-CoV-2 naive nAb
high
T
+
,n=9),
with SARS-CoV-2 naive PLWH wi th low/absent nAbs and a functi onal T cell r esponses (PLWH SARS-CoV-
2- nAb
/low
T
+
, n = 9). Both groups were age and sex matched, well controlled on cART and with a similar
median CD4 T cell count (Table S1). We have used an unbiased approach and unsupervised high-dimen-
sional analysis, glo bal t-dis tribut ed stoc hastic nei ghbor embedd ing (t- SNE), follo wed by Flo wSOM clus-
tering, in circulating T cell populat ions in the two groups. Ten major CD4 and CD8 T cell subsets wer e
examined using a combination of various activation and differentiation markers, including CD45RA,
CCR7, CD127, CD25, CXCR3, CXCR5, PD-1, and CD38 (Figures 6A, S6A, and S6B). There was no difference
Figure 6. Phenotypic characterization of CD4 and CD8 T cells from SARS-CoV-2 naive HIV positive individua ls according to their neutralization
levels
(A) viSNE map of FlowSOM metaclusters of CD4 T cells from HIV positive SARS-CoV-2 naive subjects after two vaccine doses (nAb
/low
= no neutralization or
low level of neutralization, nAb
high
= high neutralization level; n = 9 i n each group). Each point on the high-dimensional mapping represents an individual
cell, and metaclusters are color-co ded.
(B) Cell count of each Flow SOM metaclusters out of total CD4 T cells (20,000 cells/group).
(C) Representative flow plots from a nAb
/low
and nAb
high
SARS-CoV-2 naive HIV-positive donor showing expression of CXCR5 and CXCR3 within CD4
Tcells.
(D) Sum mary analysis of the percentage of CXCR5
+
CXCR3
+
CD4 T cells (n = 9 for each group). Statistical test: Mann-Whitney U-test (MWU), line represents
mean with SD for each group.
(E) Correlation between frequency of CXCR5
+
CXCR3
+
CD4 T cells and ID
50
neutralization level in nAb
/low
and nAb
high
SARS-CoV-2 naive HIV-positive
individuals after two vaccine doses. Statistical test: Spearman’s rank correlation coefficient.
(F) viSNE map of FlowSOM metaclusters of CD8 T cells from nAb
/low
and nAb
high
HIV-positive SARS-CoV-2 naive subjects after two doses of the vaccine (n =
9ineachgroup).
(G) Cell count of each CD8 FlowSOM metaclusters out of total CD8 T cells (20,000 cells/group).
(H) Representative flow plots from a nAb
/low
and nAb
high
SARS-CoV-2 naive HIV- positive donor sho wing expression of CXCR3, CD127, and C D38 within
naive CD8 T cells.
(I) Summary analysis of the percentage of CD127
+
CXCR3
+
CD38
+
naive CD8 T cells (n = 9 for each group). Statistical test: MWU, line represents mean with SD
for each group.
(J) Correlation between proportion of CD127
+
CXCR3
+
CD38
+
naive CD8 T cells and SARS-CoV-2 specific T cell responses in nAb
/low
HIV-positive SARS-CoV-
2 naive subjects. Statistical test: Spearman’s rank correlation coefficient. *p > 0.05; **p > 0.01; ***p > 0.0 01 and * ***p > 0.0001.
See also Figure S6.
ll
OPEN ACCESS
14 iScience 26, 105862, January 20, 2023
iScienc
e
Article
in the frequencies of the main T cell subsets in the two groups (Fig ures S6C and S6D). Among CD 4 T cell s,
there w as a reduction in ci rculat ing CXCR3
+
CXCR5
+
Tfollicularhelper(T
FH
) subsets observed in HIV-pos-
itive nAb
/low
compared to nAb
high
donors (Figures 6A and 6B). The reduc ed abundance of CXCR3
+
CXCR5
+
T
FH
in nAb
/low
HIV-positive subjects was further confirmed by manual gating (Fi gures 6C, 6D,
and S3C). CXCR3
+
CXCR5
+
T
FH
cells corre late d with SARS-CoV- 2 neutralizatio n l ev els in HIV-po sitiv e
SARS-CoV-2 naive individuals (r = 0.5294 p = 0.02388) (Figur e 6E), suggesting t hat reduced availabilit y of
T
FH
cells could influence the magnitude of vaccin e-ind uced S ARS-C oV-2 antibody responses.
We have next examined the CD8 T cell compartment in the two groups. Notably, a prominent cluster delin-
eated by the expression of CXCR3
+
CD127
+
CD38
+
CCR7
+
CD45RA
+
was signi ficantl y enriched in PLWH
SARS-CoV-2- nAb
/low
T
+
(Figures 6F, 6G, and S6B). The high er abundance of CXCR3
+
CD127
+
CD38
+
CCR7
+
CD45RA
+
cells in PLWH SARS-CoV-2- n Ab
/low
T
+
was further confirmed by manual gating
(p = 0.04) (Figures 6H, 6I, and S6C). Correlation an alysis of these populations showe d a positive association
between their frequencies and SARS-CoV-2-specific T cell res ponses following two vaccine doses i n PLWH
with nAb
/low
(Figure 6J), supporting the notion that these subsets could contribute to the observed induc-
tion of T cell responses in PLWH who lacked or generated low nAb responses. Overall, our analysis of the
global T cell profile of individuals with low/absent nAbs but detectable functional T cell responses revealed
that reduced availability o f T
FH
cells could contribute to the serological defect observed in conjunction with
the previously highlighted imbalance in MBCs. Mor eover , we have i dentifi ed a subset of CD8 T cells that is
overrepresented in PLWH wi th low/absent nAbs and may enable stronger functional T ce ll responses, sup-
ported by rec ent findings showing that CXCR3
+
CD8 T cells are polyfunctional and associated with survival
in critical SARS-CoV-2 patients, and have been observed in other immunosuppressed groups.
53,54
DISCUSSION
Accumulating evidence suggests th at a br oad and we ll- coordinat ed immune response is required for
protection against se vere COVID -19 dise ase. The emergence of VOCs with inc reased abi lity t o evade
nAbs has reinforced the need for a more comprehensive assessment of adaptive immunity after vaccina-
tion, especially in more vulnerable g roups including some PLWH. Our data indicate that PLWH who are
well controlled on cART, elicited poorer humoral responses, in terms of magnitude and neutralizing ability
compared to HIV-negative donors following first, second and third doses of SARS-CoV-2 vaccine. This was
related to global B cell but not anti gen- specifi c B cell dysfunction. This suggests that t he overall distur-
bance in memory B cell homeost asis during HI V can limit the amount or quality of serum antibody pro-
duced indirectly, potentially by decreasing the total number of B cells available to participate in the anti-
gen-specific response. In contrast, the observation that antigen-specific B cells in these individuals are not
overtly dysfunctional suggests that lower serum titers are not the result of antigen-specific B cells failing to
respond fully, as has been suggested in some chronic diseases.
27
In contrast, T cell responses were com-
parable in the two groups and detectable, even in a sm all group of PLWH with very poor serological re-
sponses, suggesting a potentially important non-redundant immunological role for functional T cells.
Overall, our data reinforce the beneficial effect of an additional vaccine dose in boosting adaptive immune
responses,
20
especially against circulating VOCs in this patient group.
Weaker humoral responses were observed in PLWH compared to HIV-negative controls after each dose of
vaccine when matched by prior SARS-CoV-2 status. Although the third dose largely narrowed the gap be-
tween PLWH and controls, and enabled Omic ron neutrali zation , 13% of SARS-CoV-2 naive PLWH st ill had
no nAbs after 3 vaccin e doses. This hig hlight s t he importance of r epeated vaccination in PLWH and sug-
gests additional doses/targeted vaccines could be merited , e specially given 28% of SARS-CoV-2 naive
PLWH failed to neutr alize Omicron after the third vaccine dose. Owing to known defects in germinal center
reactions in chronic HIV infection (as reviewed in
29,30,55
recall responses following vaccination in PLWH are
likely impaired resulti ng in lower tit ers and narrowe r neutrali zation breadt h. As such, addition al vaccina-
tions to stimulate additional affinity maturation and diversification of the response are likely needed to
achieve similar outcomes to those seen in HIV-negativ e individuals. In support of this, previousl y, it has
been shown that PLWH can be nefit fr om an ad ditional vaccine dose and accelerated sc hedule s durin g hep-
atitis B immunization.
56
Moreover, in other immunocompromised groups, repeated vaccination with a third
vaccine dose resulted in similarly improved levels of seroconversion and breadth against VOCs.
57,58
Previous studies among similar cohorts of PLWH with undetectable HIV viral loads have produced mixed
results, as pr evio usly reviewed.
13
SARS-CoV-2 viral vector vaccines have shown similar magnitude and
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 15
iScienc
e
Article
durability of antibody responses to HIV-negative controls
23,59
but reduced levels of seroconversion and
neutralization have been reported after two doses in PLWH in a more recent study.
15
Furthermore, viral vec-
tor vaccines, lower CD4 T cell count/viremia and old age have been linked to lower serological responses
and breakthrough infection.
16
In terms of mRNA vaccines, both non-significant
60,61
and significant de-
creases in humoral responseshavebeenreportedinPLWH.
62–65
These differ ences may be be cause of
the size of cohorts examined and the range of immune reconstit ution in these PLWH. In contrast to previ ous
work,
8–10,12
we have found no association between the CD4 T cell count and serological outcome, which
could be beca use of insu fficient power in this stu dy to de tect differences. The recent study by
20
demon-
strated a stronger humoral response after the third dose of vaccin e in PLWH, regardless of their CD4
T cell count, which is consistent with our findings,
20
Thus, the l ow er level of nAbs observed here in
PLWH could be in part owing to potential differences in boosting of memory responses to enable breadth
against Omicron after three vaccine doses.
Serological data correlated significantly with frequency of spike-specific MBCs. The B cell phenotyping
confirmed the charac teri stic and persistent defects seen in global MBCs in the setting of HIV (reviewed
in
30
). Specifi cally, we have observed low er frequencies of resting MBCs and higher freq uenci es of atypical
and activated MBCs. This dysregulated MBC phenotype was also associated with a delay in developing
nAbs after the first dose regardless of HIV status. Further evaluation in a group of individuals after the third
vaccine dose led to the interesting observation that although the global MBC landscape is still disrupted
with lower levels of resting MBCs and higher levels of atypical and switched naive MBCs in PLWH, this is not
reflected in the antigen -specific MBCs. Spike-specific MBCs present in P LWH had a similar memory B cel l
phenotype as HIV-ne gative con tro ls, albeit fe wer rest ing MBCs. H owever , higher l evels of gl obal atyp ical
MBCs, also observed in PLWH with lower neutralization at the third vaccine dose, suggest that the excess
atypical MBCs may be effectively exhausted, as has been described.
66
Therefore, SARS-CoV-2 serum anti-
body responses may be lower not because spike-specific responses are enriched within atypical MBCs
and therefore unable to progress to an antibody secreting phenotype (as has been postu lated for
HI V/HBV
27,67
), but r ather because of global MBC disturbance. Thus, we propose that this reduced nAb
to vaccination in PLWH may not be because of an alteration in the p henotype of antigen-specific cells
but rat her limited numbers of MBCs availa ble to partici pate in the antigen-specific response vi a the canon-
ical pathway.
In cont rast to serologi cal responses, SARS -CoV-2 vaccination elicited comparable T cell responses be-
tween PLWH and HIV-negat ive controls at all sampling points, and these responses were largel y preserved
against circulating VOCs, including Omicron, following three vaccine doses. These fin dings are in line with
the recent observations showing a robust T cell response to SARS-CoV-2 after third dose in PLWH,
including to known VOCs, with either homologou s or hete rolo gous combin ation s of S ARS-CoV-2 vac-
cines.
19,20
Of interest, in a recent study despite the detection of si gnificant T cell responses post a third
dose mRNA vacc ine in PLWH wh o had completed an mRNA primary c ourse, these resp onses were re-
ported to be impaired compared to the general population.
20
These results contra st our findi ngs and cou ld
be attributed to the diffe rent st udy design/ vaccine pl atforms a nd quanti ficatio n of T cell r esponses using
whole blood assay stimulation and IFN-g quantification via EL ISA.
20
Similarly, to the scenario seen in a nti -
body responses, pr ior SARS-Co V-2 infection also resulted in higher T ce ll responses to vaccination.
34,46,47
Of interest, detectable T cell responses were noted in a proportion of SARS-CoV-2 naive indi vidual s at
baseline,
23,45
which could repr esent p re-e xi sting cr oss-re activ e T cel l c ells due to past infecti on wi th ot her
coronaviruses.
68
An association between CD4 T cell counts and the magnitude of T cell responses was
observed in SARS-CoV -2 naive PLWH fo llowi ng vacci natio n, highl ight ing the relev ance of immun e cell
reconstitution in producing effective immunity to vaccination, especially in people who lack memory re-
sponses elicited by natural infection. In this cohort, PLWH were well-controlled on cART and had undetect-
able HIV viral loads. Both PLWH with, and without, prior SARS-CoV-2 exposure had similar median CD4
T cell counts (602 and 560 cells/mL, respectively) despite different serological outcomes. However, the
full impact of HIV-related immunosuppression, i n addition to other factors, includin g age, sex and pres-
ence of co-morbidities, in dampening effective and long-lived memory responses needs to be addressed
in future larger prospective studies. It is possible that different vaccine schedules, i.e., homologous versus
heterologous vaccination, could also account for the obse rved heterogen eity in cellu lar immune re-
sponses. A h eterol ogous viral vector ed/mRNA vaccinat ion has been described to lead to increased reac-
togenicity, combining the advantages from both vaccine classes.
69
Owing to li mited numbers, it has not
been possible t o addr ess the impact of differe nt vaccine platforms in our cohort. However, previ ous
ll
OPEN ACCESS
16 iScience 26, 105862, January 20, 2023
iScienc
e
Article
work has shown that the adenovirus-based platforms induce a higher T cell response
70–72
whereas mRNA
vaccine generates a stronger antibody response.
72–74
The use of heterologous boosting strategies has
been shown to expand the quantity and the breadth of T cell immunity and improve the serological
response in PLWH.
70,75–7 7
Therefore, we speculate tha t d iffe rent vaccine plat for ms and/ or heterologous
versus homogeneous combinations could lead to different magnitude T cell responses in our cohort. How-
ever, there are insufficient data to recommend the best vaccine approach to in duce a more effective, resil-
ient, and durable r esponse in PLWH. Instead, optimization of vaccine schedules requires a randomized
controlled trial to directly compare the immunogenicity of different vaccine platforms to design the
most effective vaccination schedules.
Overall humo ral responses correlated with the magnitude of T cell responses and our findings corroborate
the importance of T
FH
cells supporting effective B cell responses after vaccination. Notably, in a small sub-
group of patients (serological non- or low-level responders), there were detectable T cell responses charac-
terized by a CXCR3+CD127+ CD8 T phenotype. This phenotype was not clearly related to HIV parameters or
presence of co-mor bidit ies. These T cell populations have been linked with i ncrease d survival in people in-
fected with SARS- CoV-2 and a re co nsistent w ith observ ation s in patie nt groups who lack B cell responses.
53
Upregulation of CXCR3 in vaccine-induc ed T cells with potential to home to lung mucosa in tuberculosis
78
suggests that the se CD8 T cells described herein could play a role in the protection against severe respira-
tory diseases su ch as SARS-CoV-2. One possibility is that in the absence of functional antibody responses,
the increased abund ance of viral antigens could drive CXCR3+ CD8
+
T cell proliferation, as these cells have
been shown to have an enhanced proliferative capacity
79
and improved effector differentiation.
80,81
These
CXCR3+ CD8
+
T cells could confer a degree of protection by localization to infected tissue compart-
ments,
79,82
and provide si te-s peci fic responses, which are know n to be important in protection against res-
piratory disease.
83
CD8
+
T cells havealsobeen shown to expand following vaccination in patients receiving B
cell depleting therapies
84
and to contribute to vaccine mediated protection against SARS-CoV-2 in rhesus
macaques.
85
However, studies in larger cohorts with breakthrough infections are necessary to clearly eval-
uate the contribution of these CD8
+
T cell popu lations in va ccine-medi ated protection.
Overall, our data support the benefit of a third SARS-CoV-2 dose in inducing nAbs against Omicron in
PLWH, as it does in the general popu lation. Mo reove r, our stu dy provi des new insight s into the reasons
why some PLWH f ail to produce effectiv e humoral responses via an in-depth assessment of B cell re-
sponses. Specifically, w e find that glob al B cell dysfuncti on is relat ed to lower ser ologi cal output in t erms
of both bindi ng and neutralizing responses. A ntibody responses take longer to de velop in individuals with
greater global B cell dysfunction, which is most commonly seen in PLWH. Although a third SARS-CoV-2 vac-
cine dose improves neutralization potency and breadth for many, lower titers and MBC disturbance are still
observed. Prospective longi tudi nal studies are now needed to assess whe ther global B cell disturb ance
fluctuates in PLWH on cART ov er time, what treatments/co-mor biditie s influence thi s and what level of B
cell dysfunction results in inferior clinical outcomes long-term with regards to infectious diseases, particu-
larly where vaccination has taken place. In parallel, CD8
+
T cell profiles and anti-viral T cell activity should be
monitored in such studies to u nderst and whether these cells do provid e the proposed immunological
compensation for defects in humoral immunity.
Limitations of the study
Our study has several limitations. These include a cross-sectional analysis, which precludes the establishment
of causal relationships. Our cohort is heterogeneous, with differences in sex, age and levels of immunosuppres-
sion that may contribute to the variability in the magnitude of responses. Moreover, the current analysis
provides an overview of responses after up to three vaccine doses, and therefore further work is required to
assess the durability and resilience of these responses against subvariants and additional vaccine doses. In
addition, we could not assess the impact of breakthrough infections on humoral and cellular immune response,
although some individuals were infected with SARS-CoV-2 after vaccination as noted above, because the
numbers of re-infections were too low across any given timepoint for meaningful analysis.
STAR+METHODS
Detailed methods are provided in the online version of this paper and include the following:
d KE Y RE SOURCES TABLE
d RE SOURCE AVAILABILITY
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 17
iScienc
e
Article
B Lead contact
B Materials availability
B Data and code availability
d EX PERIMENTAL MODEL AND SUBJ ECT DETAILS
B Ethics statement
B Patien t r ecruit ment and sampling
d ME THOD DETAILS
B PBMC isolation
B Semi-quantitative S1 ELISA
B Total IgG ELISA
B IgG purification
B Pseudovi rus production
B Pseudovi rus neutralization
B Live neutralization
B Production of bi otinyl ated prote in
B B cell phenotypic flow cy tometri c an alysis
B T cell phenotypic flow cytometric analysis
B High-dimensional data analysis
B Ex vivo IFN-g ELISpot assay
B Overlapping pepti de pool s
d QUANTIFICATION AND STATISTICAL ANALYSIS
SUPPLEMENTAL INFORMATION
Supplemental information can be found online at https://doi.org/10.1016/j.isci.2022.105862.
ACKNOWLEDGMENTS
We are grateful to all the clinic staff and participants at the Mortimer Market Center and Ian Charleson Day
Center, in particular Rebecca Matthews, Aran Dhillon, Connor McAlpine, Paulina Prymas, Marzia Fiorino,
Thomas Fernandez, Jonathan Edwards and Hemat Nargis. The authors would like to thank James E Voss
of the Scripps Research Institute for the gift of Hela ACE2 expressing cells and Peter Cherepanov of the
Francis Cri ck I nstitut e for recombinant S1 antigen.
This study was supported by UKRI MRC grant (MR/W020556/1) to LE M, DP and EM and an NIH award
(R01AI55182) to (DP). LEM receives funding from the European Research Council (ERC) under the European
Union’s Horizon 2020 research and innovation program (Grant Agreement No. 757601) and i s supported by
an MRC Career Development Award (MR/R008698/1). NJM is supported by an MRC grant (MR/T032413/1),
an NHSBT grant (WPA15-02), the Wellcome Trust ISSF (204845/Z/16/Z), Addenbrooke’s Charitable Trust,
Cambridge University Hospitals (900239) and the NIHR Cambridge BRC. E T is supported by an MRC stu-
dentship (MR/N013867/1), AA by a Saudi Ministry of Education studentship (FG-350441) and TF by a Well-
come Trust Clinical PhD Fellowship (216358/Z/19/Z). NJM is supportedby an MRC g rant (MR/T032413/1),
an NHSBT grant (WPA15-02), the Addenbrooke’s Charitable Trust, Cambridge University Hospitals
(900239) and the NIHR Cambridge BRC.
AUTHOR CONTRIBUTIONS
Conceptualizati on: L.E.M. an d D.P. Invest igation: E.T., A.A., R.F., N.H. , P.P.G. , H-L.S., C.R -S., L.M. , E.G-M. ,
J.K., D.J., A.J-S., C.L. Preexisting and TAF Analysis: E.T., A.A., P.P.G., N.J.M., D.P., and L.E.M. Patient
Recruitment: D.P., I.W. , C.M., I.M., L.W., M.J., S.M., M. Y., T.J.B., F.B., R.G., a nd S.K. Resources: L.E.M.,
D.P., E.M., and N.J.M. Writing Original Draft: E.T., A.A., D.P., and L.E.M. Writing Review and Editing:
E.T., A.A., R.F., N.H., P.P.G., H-L.S., C.R-S., L.M., E.G-M., J.K., D.J., A.J-S., C.L.P., T.A.F., I.W., C.M., I.M.,
L.W., M.J., S.M., M.Y., T.J.B., F.B., S.K., S.R-J., R.G., N.J.M., E.M., D.P., and L.E.M. Supervision: L.E.M.,
D.P., E.M., N.J.M., and S.R-J. Funding A cquis ition: L.E.M., D.P., E.M., and N.J. M.
DECLARATION OF INTERESTS
The authors declare no c ompeting interests.
ll
OPEN ACCESS
18 iScience 26, 105862, January 20, 2023
iScienc
e
Article
INCLUSION AND DIVERSITY
One or more of the authors of this paper self- iden tifies as an underreprese nted et hnic m inorit y in science.
One or more of the authors of this paper se lf-ident ified as a member of the LGBTQ + community.
Received: November 1, 2022
Revised: December 4, 20 22
Accepted: December 20, 2022
Published: J anua ry 20, 2023
REFERENCES
1. Cooper, T.J., Woodward, B.L., Alom, S., and
Harky, A. (2020). Coronavirus disease 2019
(COVID-19) outcomes in HIV/AIDS patients: a
systematic review. HIV Med. 21, 567–577.
https://doi.org/10.1111/hiv.12911.
2. Herzog Tzarfati, K., Gutwein, O., Apel, A.,
Rahimi-Levene, N., Sadovnik, M., Harel, L.,
Benveniste-Levkovitz, P., Bar Chaim, A., and
Koren-Michowitz, M. (2021). BNT162b2
COVID-19 vaccine is significantly less
effective in patients with hematologic
malignancies. Am. J. Hematol. 96, 1195–
1203. https://doi.org/10.1002/AJH.26284.
3. Kamar, N., Abravanel, F., Marion, O., Couat,
C., Izopet, J., and Del Bello, A. (2021). Three
doses of an mRNA covid-19 vaccine in solid-
organ transplant recipients. N. Engl. J. Med.
385, 661–662. https://doi.org/10.1056/
NEJMc2108861.
4. Jcvi. (2022). COVID-19 Greenbook chapter
14a. https://www.gov.uk/government/
publications/investigation-of-.
5. Cruciani, M., Mengoli, C., Serpelloni, G.,
Lanza, A., Gomma, M., Nardi, S., Rimondo,
C., Bricolo, F., Consolaro, S., Trevisan, M.,
and Bosco, O. (2009). Serologic response to
hepatitis B vaccine with high dose and
increasing number of injections in HIV
infected adult patients. Vaccine 27, 17–22.
https://doi.org/10.1016/J.VACCINE.2008.
10.040.
6. George, V.K., Pallikkuth, S., Parmigiani, A.,
Alcaide, M., Fischl, M., Arheart, K.L., and
Pahwa, S. (2015). HIV infection worsens age-
associated defects in antibody responses to
influenza vaccine. J. Infect. Dis. 211, 1959–
1968. https://doi.org/10.1093/INFDIS/
JIU840.
7. Kerne
´
is, S., Launay, O., Turbelin, C., Batteux,
F., Hanslik, T., and Boe
¨
lle, P.Y. (2014). Long-
term immune responses to vaccination in
HIV-infected patients: a systematic review
and meta-analysis. Clin. Infect. Dis. 58, 1130–
1139. https://doi.org/10.1093/cid/cit937.
8. Hassold, N., Brichler, S., Ouedraogo, E.,
Leclerc, D., Carroue, S., Gater, Y., Alloui, C.,
Carbonnelle, E., Bouchaud, O., Mechai, F.,
et al. (2022). Impaired antibody response to
COVID-19 vaccination in advanced HIV
infection. AIDS 36, F1–F5. https://doi.org/10.
1097/QAD.0000000000003166.
9. Nault, L., Marchitto, L., Goyette, G.,
Tremblay-Sher, D., Fortin, C., Martel-
Laferrie
`
re, V., Trottier, B., Richard, J., Durand,
M., Kaufmann, D., et al. (2021). Covid-19
vaccine immunogenicity in people living with
HIV-1. Preprint at bioRxiv. https://doi.org/10.
1101/2021.08.13.456258.
10. Noe, S., Ochana, N., Wiese, C., Schabaz, F.,
Von Krosigk, A., Heldwein, S., Rasshofer, R.,
Wolf, E., and Jonsson-Oldenbuettel, C.
(2022). Humoral response to SARS-CoV-2
vaccines in people living with HIV. Infection
50, 617–623. https://doi.org/10.1007/S15010-
021-01721-7
.
11. Spinelli, M.A., Peluso, M.J., Lynch, K.L., Yun,
C., Glidden, D.V., Henrich, T.J., Deeks, S.G.,
and Gandhi, M. (2021). Differences in post-
mRNA vaccination severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2)
immunoglobulin G (IgG) concentrations and
surrogate virus neutralization test response
by human immunodeficiency virus (HIV) status
and type of vaccine: a ma. Clin. Infect. Dis.
https://doi.org/10.1093/CID/CIAB1009.
12. Touizer, E., Alrubayyi, A., Rees-Spear, C.,
Fisher-Pearson, N., Griffith, S.A., Muir, L.,
Pellegrino, P., Waters, L., Burns, F., Kinloch,
S., et al. (2021). Failure to seroconvert after
two doses of BNT162b2 SARS-CoV-2 vaccine
in a patient with uncontrolled HIV. Lancet HIV
8, e317–e318. https://doi.org/10.1016/S2352-
3018(21)00099-0.
13. Mullender, C., da Costa, K.A.S., Alrubayyi, A.,
Pett, S.L., and Peppa, D. (2022). SARS-CoV-2
immunity and vaccine strategies in people
with HIV. Oxford Open Immunol. 3, iqac005.
https://doi.org/10.1093/oxfimm/iqac005.
14. Tamuzi, J.L., Muyaya, L.M., Mitra, A., and
Nyasulu, P.S. (2022). Systematic review and
meta-analysis of COVID-19 vaccines safety,
tolerability, and efficacy among HIV-infected
patients. Prepeint at. medRxiv. https://doi.
org/10.1101/2022.01.11.22269049.
15. Woldemeskel, B.A., Karaba, A.H., Garliss,
C.C., Beck, E.J., Wang, K.H., Laeyendecker,
O., Cox, A.L., and Blankson, J.N. (2022). The
BNT162b2 mRNA vaccine elicits robust
humoral and cellular immune responses in
people living with human immunodeficiency
virus (HIV). Clin. Infect. Dis. 74, 1268–1270.
https://doi.org/10.1093/cid/ciab648.
16. Sun, J., Zheng, Q., Madhira, V., Olex, A.L.,
Anzalone, A.J., Vinson, A., Singh, J.A.,
French, E., Abraham, A.G., Mathew, J., et al.
(2022). Association between immune
dysfunction and COVID-19 breakthrough
infection after SARS-CoV-2 vaccination in the
US. JAMA Intern. Med. 182, 153–162. https://
doi.org/10.1001/jamainternmed.2021.7024.
17. Planas, D., Veyer, D., Baidaliuk, A., Staropoli,
I., Guivel-Benhassine, F., Rajah, M.M.,
Planchais, C., Porrot, F., Robillard, N., Puech,
J., et al. (2021). Reduced sensitivit y of SARS-
CoV-2 variant D elta to antibody
neutralization. Nature 596, 276–280. https://
doi.org/10.1038/s41586-021-03777-9.
18. Chen, Y., Tong, P., Whiteman, N., Sanjari
Moghaddam, A., Zarghami, M., Zuiani, A.,
Habibi, S., Gautam, A., Keerti, F., Bi, C., et al.
(2022). Immune recall improves antibody
durability and breadth to SARS-CoV-2
variants. Sci. Immunol. 7, eabp8328. https://
doi.org/10.1126/sciimmunol.abp8328.
19. Fidler, S., Fox, J., Tipoe, T., Longet, S., Tipton,
T., Abeywickrema, M., Adele, S.,
Alagaratnam, J., Ali, M., Aley, P.K., et al.
(2022). Booster vaccination against SARS-
CoV-2 induces potent immune responses in
people with HIV. Clin. Infect. Dis. ciac796.
https://doi.org/10.1093/cid/ciac796.
20. Vergori, A., Cozzi Lepri, A., Cicalini, S.,
Matusali, G., Bordoni, V., Lanini, S., Meschi,
S., Iannazzo, R., Mazzotta, V., Colavita, F.,
et al. (2022). Immunogenicity to COVID-19
mRNA vaccine third dose in people living
with HIV. Nat. Commun. 13, 4922. https://doi.
org/10.1038/s41467-022-32263-7.
21. Vergori, A., Cozzi-Lepri, A., Matusali, G.,
Colavita, F., Cicalini, S., Galli, P., Garbuglia,
A.R., Fusto, M., Puro, V., Maggi, F., et al.
(2022). SARS-CoV-2 Omicron variant
neutralization after third dose vaccination in
PLWH. Viruses 14
, 81710. https://doi.org/10.
3390/v14081710.
22. Lapointe, H.R., Mwimanzi, F., Cheung, P.K.,
Sang, Y., Yaseen, F., Umviligihozo, G.,
Kalikawe, R., Speckmaier, S., Moran-Garcia,
N., Datwani, S., et al. (2022). People with HIV
receiving suppressive antiretroviral therapy
show typical antibody durability after dual
COVID-19 vaccination, and strong third dose
responses. J. Infect. Dis. jiac229. https://doi.
org/10.1093/infdis/jiac229.
23. Ogbe, A., Pace, M., Bittaye, M., Tipoe, T.,
Adele, S., Alagaratnam, J., Aley, P.K., Ansari,
M.A., Bara, A., Broadhead, S., et al. (2022).
Durability of ChAdOx1 nCoV-19 vaccination
in people living with HIV. JCI Insight 7,
e157031. https://doi.org/10.1172/jci.insight.
157031.
24. Fuster, F., Vargas, J.I., Jensen, D., Sarmiento,
V., Acun
˜
a, P., Peirano, F., Fuster, F., Arab, J.P.,
and Martı
´
nez, F.; Core-HIV Study Group
(2016). CD4/CD8 ratio as a predictor of the
response to HBV vaccination in HIV-positive
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 19
iScienc
e
Article
patients: a prospective cohort study. Vaccine
34, 1889–1895. https://doi.org/10.1016/j.
vaccine.2016.02.055.
25. Pallikkuth, S., De Armas, L.R., Pahwa, R.,
Rinaldi, S., George, V.K., Sanchez, C.M., Pan,
L., Dickinson, G., Rodriguez, A., Fischl, M.,
et al. (2018). Impact of aging and HIV infection
on serologic response to seasonal influenza
vaccination. AIDS 32, 1085–1094. https://doi.
org/10.1097/QAD.0000000000001774.
26. Fenwick, C., Joo, V., Jacquier, P., Noto, A.,
Banga, R., Perreau, M., and Pantaleo, G.
(2019). T-cell exhaustion in HIV infection.
Immunol. Rev. 292, 149–163. https://doi.org/
10.1111/imr.12823.
27. Burton, A.R., Pallett, L.J., McCoy, L.E.,
Suveizdyte, K., Amin, O.E., Swadling, L.,
Alberts, E., Davidson, B.R., Kennedy, P.T.,
Gill, U.S., et al. (2018). Circulating and
intrahepatic antiviral B cells are defective in
hepatitis B. J. Clin. Invest. 128, 4588–4603.
https://doi.org/10.1172/JCI121960.
28. Portugal, S., Tipton, C.M., Sohn, H., Kone, Y.,
Wang, J., Li, S., Skinner, J., Virtaneva, K.,
Sturdevant, D.E., Porcella, S.F., et al. (2015).
Malaria-associated atypical memory B cells
exhibit markedly reduced B cell receptor
signaling and effector function. Elife 4,
e07218. https://doi.org/10.7554/eLife.07218.
29. Moir, S., and Fauci, A.S. (2013). Insights into B
cells and hiv-specific B-cell responses in hiv-
infected individuals. Immunol. Rev. 254,
207–224. https://doi.org/10.1111/IMR.12067.
30. Moir, S., and Fauci, A.S. (2017). B-Cell
Responses to HIV Infection (Blackwell
Publishing Ltd).
31. Nasi, M., De Biasi, S., Gibellini, L., Bianchini,
E., Pecorini, S., Bacca, V., Guaraldi, G.,
Mussini, C., Pinti, M., and Cossarizza, A.
(2017). Ageing and inflammation in patients
with HIV infection. Clin. Exp. Immunol. 187,
44–52. https://doi.org/10.1111/cei.12814.
32. Ng, K.W., Faulkner, N., Cornish, G.H., Rosa,
A., Harvey, R., Hussain, S., Ulferts, R., Earl, C.,
Wrobel, A.G., Benton, D.J., et al. (2020).
Preexisting and de novo humoral immunity to
SARS-CoV-2 in humans. Science 370, 1339–
1343. https://doi.org/10.1126/science.
abe1107.
33. Rees-Spear, C., Muir, L., Griffith, S.A.,
Heaney, J., Aldon, Y., Snitselaar, J.L., Thomas,
P., Graham, C., Seow, J., Lee, N., et al. (2021).
The effect of spike mutations on SARS-CoV-2
neutralization. Cell Rep. 34, 108890. https://
doi.org/10.1016/J.CELREP.2021.108890.
34. Reynolds, C.J., Pade, C., Gibbons, J.M.,
Butler, D.K., Otter, A.D., Menacho, K.,
Fontana, M., Smit, A., Sackville-West, J.E.,
Cutino-Moguel, T., et al. (2021). Prior SARS-
CoV-2 infection rescues B and T cell
responses to variants after first vaccine dose.
Science 372, 1418–1423. https://doi.org/10.
1126/science.abh1282.
35. Brouwer, P.J.M., Caniels, T.G., van der
Straten, K., Snitselaar, J.L., Aldon, Y.,
Bangaru, S., Torres, J.L., Okba, N.M.A.,
Claireaux, M., Kerster, G., et al. (2020). Potent
neutralizing antibodies from COVID-19
patients define mu ltiple targets of
vulnerability. Science 369, 643–650. https://
doi.org/10.1126/science.abc5902.
36. Graham, C., Seow, J., Huettner, I., Khan, H.,
Kouphou, N., Acors, S., Winstone, H.,
Pickering, S., Galao, R.P., Dupont, L., et al.
(2021). Neutralization potency of monoclonal
antibodies recognizing dominant and
subdominant epitopes on SARS-CoV-2 Spike
is impacted by the B.1.1.7 variant. Immunity
54, 1276–1289.e6. https://doi.org/10.1016/j.
immuni.2021.03.023.
37. Gilbert, P.B., Montefiori, D.C., McDermott,
A.B., Fong, Y., Benkeser, D., Deng, W., Zhou,
H., Houchens, C.R., Martins, K., Jayashankar,
L., et al. (2022). Immune correlates analysis of
the mRNA-1273 COVID-19 vaccine efficacy
clinical trial. Science 375, 43–50. https://doi.
org/10.1126/science.abm3425.
38. Cohen, K.W., Linderman, S.L., Moodie, Z.,
Czartoski, J., Lai, L., Mantus, G., Norwood, C.,
Nyhoff, L.E., Edara, V.V., Floyd, K., et al.
(2021). Longitudinal analysis shows durable
and broad immune memory after SARS-CoV-
2 infection with persisting antibody
responses and memory B and T cells. Cell
Rep. Med. 2, 100354. https://doi.org/10.
1016/j.xcrm.2021.100354.
39. Dan, J.M., Mateus, J., Kato, Y., Hastie, K.M.,
Yu, E.D., Faliti, C.E., Grifoni, A., Ramirez, S.I.,
Haupt, S., Frazier, A., et al. (2020).
Immunological memory to SARS-CoV-2
assessed for up to eight months after
infection. bioRxiv. https://doi.org/10.1101/
2020.11.15.383323.
40. Terreri, S., Piano Mortari, E., Vinci, M.R.,
Russo, C., Alteri, C., Albano, C., Colavita, F.,
Gramigna, G., Agrati, C., Linardos, G., et al.
(2022). Persistent B cell memory after SARS-
CoV-2 vaccination is functional during
breakthrough infections. Cell Host Microbe
30, 400–408.e4. https://doi.org/10.1016/j.
chom.2022.01.003.
41. Goel, R.R., Painter, M.M., Apostolidis, S.A.,
Mathew, D., Meng, W., Rosenfeld, A.M.,
Lundgreen, K.A., Reynaldi, A., Khoury, D.S.,
Pattekar, A., et al. (2021). mRNA vaccines
induce durable immune memory to SARS-
CoV-2 and variants of concern. Science 374,
abm0829. https://do i.org/10.1126/science.
abm0829.
42. Jeffery-Smith, A., Burton, A.R., Lens, S., Rees-
Spear, C., Davies, J., Patel, M., Gopal, R.,
Muir, L., Aiano, F., Doores, K.J., et al. (2022).
SARS-CoV-2–specific memory B cells can
persist in the elderly who have lost detectable
neutralizing antibodies. J. Clin. Invest. 132,
e152042. https://doi.org/10.1172/JCI152042.
43. Pensieroso, S., Galli, L., Nozza, S., Ruffin, N.,
Castagna, A., Tambussi, G., Hejdeman, B.,
Misciagna, D., Riva, A., Malnati, M., et al.
(2013). B-cell subset alterations and
correlated factors in HIV-1 infection. AIDS 27,
1209–1217. https://doi.org/10.1097/QAD.
0b013e32835edc47.
44. McCallum, M., Czudnochowski, N., Rosen,
L.E., Zepeda, S.K., Bowen, J.E., Walls, A.C.,
Hauser, K., Joshi, A., Stewart, C., Dillen, J.R.,
et al. (2022). Structural basis of SARS-CoV-2
Omicron immune evasion and receptor
engagement. Science 375 , 864–868.
https://
doi.org/10.1126/science.abn8652.
45. Alrubayyi, A., Gea-Mallorquı
´
, E., Touizer, E.,
Hameiri-Bowen, D., Kopycinski, J., Charlton,
B., Fisher-Pearson, N., Muir, L., Rosa, A.,
Roustan, C., et al. (2021). Characterization of
humoral and SARS-CoV-2 specific T cell
responses in people living with HIV. Nat.
Commun. 12, 5839. https://doi.org/10.1038/
s41467-021-26137-7.
46. Lozano-Ojalvo, D., Camara, C., Lopez-
Granados, E., Nozal, P., Del Pino- Molina, L.,
Bravo-Gallego, L.Y., Paz-Artal, E., Pion, M.,
Correa-Rocha, R., Ortiz, A., et al. (2021).
Differential effects of the second SARS-CoV-2
mRNA vaccine dose on T cell immunity in
naive and COVID -19 recovered individuals.
Cell Rep. 36, 109570. https://doi.org/10.
1016/j.celrep.2021.109570.
47. Prendecki, M., Clarke, C., Brown, J., Cox, A.,
Gleeson, S., Guckian, M., Randell, P., Pria,
A.D., Lightstone, L., Xu, X.N., et al. (2021).
Effect of previous SARS-CoV-2 infection on
humoral and T-cell responses to singl e-dose
BNT162b2 vaccine. Lancet 397, 1178–11 81.
https://doi.org/10.1016/S0140-6736(21)
00502-X.
48. Braun, J., Loyal, L., Frent sch, M., Wendisch,
D., Georg, P., Kurth, F., Hippenstiel, 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
587, 270–274. https://doi.org/10.1038/
s41586-020-2598-9.
49. 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. https://doi.org/10.1016/j.cell.2020.
05.015.
50. 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. Nature 584, 457–462. https://doi.
org/10.1038/s41586-020-2550-z.
51. 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 370, 89–94. https://doi.org/10.1126/
science.abd3871.
52. Sekine, T., Perez-Potti, A., Rivera-Ballesteros,
O., Stra˚ lin, K., Gorin, J.B., Olsson, A.,
Llewellyn-Lacey, S., Kamal, H., Bogdanovic,
G., Muschiol, S., et al. (2020). Robust T cell
immunity in convalescent individuals with
asymptomatic or mild COVID-19. Cell 183,
158–168.e14. https://doi.org/10.1016/j.cell.
2020.08.017.
53. Gao, Y., Cai, C., Wullimann, D., Niessl, J.,
Rivera-Ballesteros, O., Chen, P., Lange, J.,
Cuapio, A., Blennow, O., Hansson, L., et al.
(2022). Immunodeficiency syndromes
differentially impact the functional profile of
ll
OPEN ACCESS
20 iScience 26, 105862, January 20, 2023
iScienc
e
Article
SARS-CoV-2-specific T cells elicited by mRNA
vaccination. Immunity 55, 1732–1746.e5.
https://doi.org/10.1016/j.immuni .2022.
07.005.
54. Adam, L., Rosenbaum, P., Quentric, P.,
Parizot, C., Bonduelle, O., Guillou, N.,
Corneau, A., Dorgham , K., Miyara, M., Luyt,
C.E., et al. (2021). CD8+PD-L1+CXCR3+
polyfunctional T cell abundances are
associated with survival in critical SARS-CoV-
2-infected patients. JCI Insight 6, e151571.
https://doi.org/10.1172/jci.insight.151571.
55. Thornhill, J.P., Fidler, S., Klenerman, P.,
Frater, J., and Phetsouphanh, C. (2017). The
role of CD4+ T follicular helper cells in HIV
infection: from the germinal center to the
periphery. Front. Immunol. 8, 46. https://d oi.
org/10.3389/fimmu.2017.00046.
56. Bhiva. (2015). BHIVA Guidelines on the Use of
Vaccines in HIV-Positive Adults 2015.
57. Haggenburg, S., Hofsink, Q., Lissenberg-
Witte, B.I., Broers, A.E.C., van Doesum, J.A.,
van Binnendijk, R.S., den Hartog, G.,
Bhoekhan, M.S., Haverkate, N.J.E., Burger,
J.A., et al. (2022). Antibody response in
immunocompromised patients with
hematologic cancers who received a 3-dose
mRNA-1273 vaccination schedule for COVID-
19. JAMA Oncol. 8, 1477–1483. https://doi.
org/10.1001/jamaoncol.2022.3227.
58. Tauzin, A., Beaudoin-Bussie
`
res, G., Gong,
S.Y., Chatterjee, D., Gendron-Lepage, G.,
Bourassa, C., Goyette, G., Racine, N., Khrifi,
Z., Turgeon, J., et al. (2022). Humoral immune
responses against SARS-CoV-2 Spike variants
after mRNA vaccination in solid organ
transplant recipients. iScience 25, 104990.
https://doi.org/10.1016/j.isci.2022.104990.
59. Frater, J., Ewer, K.J., Ogbe, A., Pace, M.,
Adele, S., Adland, E., Alagaratnam, J., Aley,
P.K., Ali, M., Ansari, M.A., et al. (2021). Safety
and immunogenicity of the ChAdOx1 nCoV-
19 (AZD1222) vaccine against SARS-CoV-2 in
HIV infection: a single-arm substudy of a
phase 2/3 clinical trial. Lancet HIV 396 ,
467–478. https://doi.org/10.1016/S2352-
3018(21)00103-X.
60. Heftdal, L.D., Knudsen, A.D., Hamm, S.R.,
Hansen, C.B., Møller, D.L., Pries-Heje, M.,
Fogh, K., Hasselbalch, R.B., Jarlhelt, I., Pe
´
rez-
Alo
´
s, L., et al. (2022). Humoral response to
two doses of BNT162b2 vaccination in
people with HIV. J. Intern. Med. 291, 513–5 18.
https://doi.org/10.1111/JOIM.1 3419.
61. Levy, I., Wieder-Finesod, A., Litchevsky, V.,
Biber, A., Indenbaum, V., Olmer, L., Huppert,
A., Mor, O., Goldstein, M., Levin, E.G., et al.
(2021). Immunogenicity and safety of the
BNT162b2 mRNA COVID-19 vaccine in
people living with HIV-1. Clin.Microbiol.
Infect. 27, 1851–1855. https://doi.org/10.
1016/J.CMI.2021.07.031.
62. Hensley, K.S., Jongkees, M.J., Geers, D.,
GeurtsvanKessel, C.H., Ash Dalm, V.,
Papageorgiou, G., Steggink, H., Gorska, A.,
den Hollander, J.G., Bierman, W.F.W., et al.
(2022). Immun ogenicity and reactogenicity of
SARS-CoV-2 vaccines in people living with
hiv: a nationwide prospective cohort study in
the netherlands. Preprint at medRxiv. https://
doi.org/10.1101/2022.03.31.22273221.
63. Bessen, C., Plaza-Sirvent, C., Bhat, J.,
Marheinecke, C., Urlaub, D., Bonowitz, P.,
Busse, S., Schumann, S., Vidal Blanco, E.,
Skaletz-Rorowski, A., et al. (2022). Impact of
SARS-CoV-2 vaccination on systemic immune
responses in people living with HIV. Preprint
at medRxiv. https://doi.org/10.1101/2022.04.
08.22273605.
64. Brumme, Z.L., Mwimanzi, F., Lapointe, H.R.,
Cheung, P.K., Sang, Y., Duncan, M.C.,
Yaseen, F., Agafitei, O., Ennis, S., Ng, K., et al.
(2022). Humoral immune responses to
COVID-19 vaccination in people living with
HIV receiving suppressive antiretroviral
therapy. NPJ Vaccines 7, 28. https://doi.org/
10.1038/S41541-022-00452-6.
65. Jedicke, N., Stankov, M.V., Cossmann, A.,
Dopfer-Jablonka, A., Knuth, C., Ahrenstorf,
G., Ramos, G.M., and Behrens, G.M.N. (2022).
Humoral immune response following prime
and boost BNT162b2 vaccination in people
living with HIV on antiretroviral therapy. HIV
Med. 23, 558–563. https://doi.org/10.1111/
HIV.13202.
66. Moir, S., Ho, J., Malaspina, A., Wang, W.,
DiPoto, A.C., O’Shea, M.A., Roby, G., Kottilil,
S., Arthos, J., Proschan, M.A., et al. (2008).
Evidence for HIV-associated B cell exhaustion
in a dysfunctional memory B cell
compartment in HIV-infected viremic
individuals. J. Exp. Med. 205, 1797–1805.
https://doi.org/10.1084/jem.20072683.
67. Meffre, E., Louie, A., Bannock, J., Kim, L.J.Y .,
Ho, J., Frear, C.C., Kardava, L., Wang, W.,
Buckner, C.M., Wang, Y., et al. (2016).
Maturational characteristics of HIV-specific
antibodies in viremic individuals. JCI Insight
1, e84610. https://doi.org/10.1172/jci.insight.
84610.
68. Casado, J.L., Vizcarra, P., Haemmerle, J.,
Velasco, H., Martı
´
n-Hondarza, A., Rodrı
´
guez-
Domı
´
nguez, M.J., Velasco, T., Martı
´
n, S.,
Romero-Herna
´
ndez, B., Ferna
´
ndez-
Escribano, M., and Vallejo, A. (2022). Pre-
existing T cell immunity determines the
frequency and magnitude of cellular immune
response to two doses of mRNA vaccine
against SARS-CoV-2. Vaccine X 11, 100165.
https://doi.org/10.1016/j.jvacx.2022.100165.
69. Ba
´
nki, Z., Mateus, J., Ro
¨
ssler, A., Scha
¨
fer, H.,
Bante, D., Riepler, L., Grifoni, A., Sette, A.,
Simon, V., Falkensammer, B., et al. (2022).
Heterologous ChAdOx1/BNT162b2
vaccination induces stronger immune
response than homologous ChAdOx1
vaccination: the pragmatic, multi-center,
three-arm, partially randomized HEVACC
trial. EBioMedicine 80, 104073. https://doi.
org/10.1016/j.ebiom.202 2.104073.
70. Pozzetto, B., Legros, V., Djebali, S., Barateau,
V., Guibert, N., Villard, M., Peyrot, L., Allatif,
O., Fassier, J.B., Massardier-Pilonche
´
ry, A.,
et al. (2021). Immunogenicity and efficacy of
heterologous ChAdOx1-BNT162b2
vaccination. Nature
600, 701–706. https://doi.
org/10.1038/s41586-021-04120-y.
71. Stuart, A.S.V., Shaw, R.H., Liu, X., Greenland,
M., Aley, P.K., Andrews, N.J., Cameron, J.C.,
Charlton, S., Clutterbuck, E.A., Collins, A.M.,
et al. (2022). Immunogenicity, safety, and
reactogenicity of heterologous COVID-19
primary vaccination incorporating mRNA,
viral-vector, and protein-adjuvant vaccines in
the UK (Com-COV 2): a single-blind,
randomised, phase 2, non-inferiority trial.
Lancet 399, 36–49. https://doi.org/10.1016/
S0140-6736(21)02718-5.
72. Zhang, Z., Mateus, J., Coelho, C.H., Dan,
J.M., Moderbacher, C.R., Galvez, R.I., Cortes,
F.H., Grifoni, A., Tarke, A., Chang, J., et al.
(2022). Humoral and cellular immune memory
to four covid-19 vaccines. Preprint at bioRxiv.
https://doi.org/10.1101/2022.03.18.484953.
73. Parry, H., Bruton, R., Stephens, C., Brown, K.,
Amirthalingam, G., Otter, A., Hallis, B., Zuo,
J., and Moss, P. (2021). Differential
immunogenicity of BNT162b2 or ChAdOx1
vaccines after extended-interval homologous
dual vaccination in older people. Immun.
Ageing 18, 34. https://doi.org/10.1186/
s12979-021-00246-9.
74. Parry, H., Bruton, R., Tut, G., Ali, M., Stephens,
C., Greenwood, D., Faustini, S., Hughes, S.,
Huissoon, A., Meade, R., et al. (2021).
Immunogenicity of single vaccination with
BNT162b2 or ChAdOx1 nCoV-19 at 5-
6 weeks post vaccine in participants aged 80
years or older: an exploratory analysis.
Lancet.Healthy Longev. 2, e554–e560.
https://doi.org/10.1016/S2666-7568(21)
00169-0.
75. Khoo, N.K.H., Lim, J.M.E., Gill, U.S., de Alwis,
R., Tan, N., Toh, J.Z.N., Abbott, J.E., Usai, C.,
Ooi, E.E., Low, J.G.H., et al. (2022).
Differential immunogenicity of homologous
versus heterologous boost in Ad26.COV2.S
vaccine recipients. Med (N Y) 3, 104–118.e4.
https://doi.org/10.1016/j.medj.2021.12.004.
76. Atmar, R.L., Lyke, K.E., Deming, M.E.,
Jackson, L.A., Branche, A.R., El Sahly, H.M.,
Rostad, C.A., Martin, J.M., Johnston, C.,
Rupp, R.E., et al. (2022). Homologous and
heterologous covid-19 booster vaccinations.
N. Engl. J. Med. 386, 1046–1057. https://doi.
org/10.1056/NEJMoa2116414.
77. Liu, X., Shaw, R.H., Stuart, A.S.V., Gree nland,
M., Aley, P.K., Andrews, N.J., Cameron, J.C.,
Charlton, S., Clutterbuck, E.A., Collins, A.M.,
et al. (2021). Safety and immunogenicity of
heterologous versus homologous prime-
boost schedules with an adenoviral vectored
and mRNA COVID-19 vaccine (Com-COV): a
single-blind, randomised, non-inferiority trial.
Lancet 398, 856–869. https://doi.org/10.
1016/S0140-6736(21)01694-9.
78. Jeyanathan, M., Afkhami, S., Khera, A.,
Mandur, T., Damjanovic, D., Yao, Y., Lai, R.,
Haddadi, S., Dvorkin-Gheva, A., Jor dana, M.,
et al. (2017). CXCR3 signaling is required for
restricted homing of parenteral tuberculosis
vaccine-induced T cells to both the lung
parenchyma and airway. J. Immunol. 199,
2555–2569. https://doi.org/10.4049/
jimmunol.1700382.
79. Bangs, D.J., Tsitsiklis, A., Steier, Z., Chan,
S.W., Kaminski, J., Streets, A., Yosef, N., and
Robey, E.A. (2022). CXCR3 regulates stem
and proliferative CD8+ T cells during chronic
infection by promoting interactions with DCs
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 21
iScienc
e
Article
in splenic bridging channels. Cell Rep. 38 ,
110266. https://doi.org/10.1016/j.celrep.
2021.110266.
80. De Simone, G., Mazza, E.M.C., Cassotta, A.,
Davydov, A.N., Kuka, M., Zanon, V., De Paoli,
F., Scamardella, E., Metsger, M., Roberto, A.,
et al. (2019). CXCR3 identifies human naive
CD8(+) T cells with enhanced effector
differentiation potential. J. Immunol. 203,
3179–3189. https://doi.org/10.4049/
jimmunol.1901072.
81. Kurachi, M., Kurachi, J., Suenaga, F., Tsukui,
T., Abe, J., Ueha, S., Tomura, M., Sugihara, K.,
Takamura, S., Kakimi, K., and Matsushima, K.
(2011). Chemokine receptor CXCR3 facilitates
CD8(+) T cell differentiation into short-lived
effector cells leading to memory
degeneration. J. Exp. Med. 208, 1605–1620.
https://doi.org/10.1084/jem.20102101.
82. Hickman, H.D., Reynoso, G.V., Ngudiankama,
B.F., Cush, S.S., Gibbs, J., Bennink, J.R., an d
Yewdell, J.W. (2015). CXCR3 chemokine
receptor enables local CD8(+) T cell
migration for the destruction of virus-infected
cells. Immunity 42, 524–537. https://doi.org/
10.1016/j.immuni.2015.02.009.
83. Slu
¨
tter, B., Pewe, L.L., Kaech, S.M., and Harty,
J.T. (2013). Lung airway-surveilling CXCR3(hi)
memory CD8(+) T cells are critical for
protection against influenza A virus. Immunity
39, 939–948. https://doi.org/10.1016/j.
immuni.2013.09.013.
84. Apostolidis, S.A., Kakara, M., Painter, M.M.,
Goel, R.R., Mathew, D., Lenzi, K., Rezk, A.,
Patterson, K.R., Espinoza, D.A., Kadri, J.C.,
et al. (2021). Cellular and humoral immune
responses following SARS-CoV-2 mRNA
vaccination in patients with multiple sclerosis
on anti-CD20 therapy. Nat. Med. 27, 1990–
2001. https://doi.org/10.1038/s41591-021-
01507-2.
85. Liu, J., Yu, J., McMahan, K., Jacob-Dolan, C.,
He, X., Giffin, V., Wu, C., Sciacca, M., Powers,
O., Nampanya, F., et al. (2022). CD8 T cells
contribute to vaccine protection against
SARS-CoV-2 in macaques. Sci. Immunol. 7,
eabq7647. https://doi.org/10.1126/
sciimmunol.abq7647.
86. Rogers, T.F., Zhao, F., Huang, D., Beutler, N.,
Burns, A., He, W.T., Limbo, O., Smith, C.,
Song, G., Woehl, J., et al. (2020). Isolation of
potent SARS-CoV-2 neutralizing antibodies
and protection from disease in a small animal
model. Science 369, 956–963. https://do i.
org/10.1126/science.abc7520.
87. Gerber, P.P., Duncan, L.M., Greenwood, E.J.,
Marelli, S., Naamati, A., Teixeira-Silva, A.,
Crozier, T.W., Gabaev, I., Zhan, J. R.,
Mulroney, T.E., et al. (2022). A protease-
activatable luminescent biosensor and
reporter cell line for authentic SARS-CoV-2
infection. PLoS Pathog. 18, e1010265. https://
doi.org/10.1371/journal.ppat.1010265.
88. Seow, J., Graham, C., Merrick, B., Acors, S.,
Pickering, S., Steel, K.J.A., Hemmings, O.,
O’Byrne, A., Kouphou, N., Galao, R.P., et al.
(2020). Longitudinal observation and decline
of neutralizing antibody responses in the
three months following SARS-CoV-2 infection
in humans. Nat. Microbiol. 5
, 1598–1607.
https://doi.org/10.1038/s41564-020-00813-8.
89. Zufferey, R., Nagy, D., Mandel, R.J., Naldini,
L., and Trono, D. (1997). Multiply attenuated
lentiviral vector achieves efficient gene
delivery in vivo. Nat. Biotechnol. 15, 871–875.
https://doi.org/10.1038/nbt0997-871.
90. Levin, E.G., Lustig, Y., Cohen, C., Fluss, R.,
Indenbaum, V., Amit, S., Doolman, R., Asraf,
K., Mendelson, E., Ziv, A., et al. (2021).
Waning immune humoral response to
BNT162b2 covid-19 vaccine over 6 months.
N. Engl. J. Med. 385 , e84. https://doi.org/10.
1056/NEJMOA2114583/SUPPL_FILE/
NEJMOA2114583_DATA-SHARING.PDF.
91. O’Nions, J., Muir, L., Zheng, J., Rees-Spear,
C., Rosa, A., Roustan, C., Earl, C.,
Cherepanov, P., Gupta, R., Khwaja, A., et al.
(2020). SARS-CoV-2 Antibody Responses in
Patients with Acute Leukaemia (Springer
Nature).
92. Wright, E., Temperton, N.J., Marston, D.A.,
McElhinney, L.M., Fooks, A.R., and Weiss,
R.A. (2008). Investigating antibody
neutralization of lyssaviruses using lentiviral
pseudotypes: a cross-species comparison.
J. Gen. Virol. 89, 2204–2213. https://doi.org/
10.1099/VIR.0.2008/000 349-0 .
93. Daly, J.L., Simonetti, B., Klein, K., Chen, K.E.,
Williamson, M.K., Anto
´
n-Pla
´
garo, C.,
Shoemark, D.K., Simo
´
n-Gracia, L., Bauer, M.,
Hollandi, R., et al. (2020). Neuropilin-1 is a
host factor for SARS-CoV-2 infection. Science
370, 861–865. https://doi.org/10.1126/
science.abd3072.
94. Patterson, E.I., Prince, T., Anderson, E.R.,
Casas-Sanchez, A., Smith, S.L., Cansado-
Utrilla, C., Solomon, T., Griffiths, M.J., Acosta-
Serrano, A
´
., Turtle, L., and Hughes, G.L.
(2020). Methods of inactivation of SARS-CoV-
2 for downstream biological assays. J. Infect.
Dis. 222 , 1462–1467. https://doi.org/10.1093/
infdis/jiaa507.
95. Bergamaschi, L., Mescia, F., Turner, L.,
Hanson, A.L., Kotagiri, P., Dunmore, B.J.,
Ruffieux, H., De Sa, A., Huhn, O., Morgan,
M.D., et al. (2021). Longitudinal analysis
reveals that delayed bystander CD8+ T cell
activation and early immune pathology
distinguish severe COVID-19 from mild
disease. Immunity 54, 1257–1275.e8. https://
doi.org/10.1016/j.immuni.2021.05.010.
96. van der Klaauw, A.A., Horner, E.C., Pereyra-
Gerber, P., Agrawal, U., Foster, W.S.,
Spencer, S., Vergese, B., Smith, M., Henning,
E., Ramsay, I.D., et al. (2022). Accelerated
waning of the humoral response to SARS-
CoV-2 vaccines in obesity. Preprint at
medRxiv. https://doi.org/10.1101/2022.06.
09.22276196.
97. van der Maatens, L. (2014). Accelerating t-
SNE using tree-based algorithms. J. Mach.
Learn. Res. 15, 3221–3245.
98. Van Gassen, S., Callebaut, B., Van Helden,
M.J., Lambrecht, B.N., Demeester, P.,
Dhaene, T., and Saeys, Y. (2015). FlowSOM:
using self-organizing maps for visualization
and interpretation of cytometry data.
Cytometry A. 87, 636–645. https://doi.org/10.
1002/cyto.a.22625.
ll
OPEN ACCESS
22 iScience 26, 105862, January 20, 2023
iScienc
e
Article
STAR+METHODS
KEY RESOURCES TABLE
REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
Goat anti-human F(ab)’2 Jackson ImmunoResearch Cat# 109-006-006; RRID: AB_2337553
Alkaline phosphatase-conjugated
goat anti-human IgG
Jackson ImmunoResearch Cat#109-055-098; RRID: AB_2337608
FITC Mouse Anti-Human IgG BD Biosciences Cat # 560952; RRID:AB_10563419
BV786 Mouse Anti-Human CD19 BD Biosciences Cat # 740968; RRID:AB_2740593
BUV395 Mouse Anti-Human CD27 BD Biosciences Cat # 563815; RRID:AB_2744349
PE-Cy7 Mouse Anti-Human IgD BD Biosciences Cat # 561314; RRID:AB_10642457
APC/Cyanine7 anti-human IgM Antibody BioLegend Cat # 314520; RRID:AB_10900422
Alexa Fluor 700 Mouse Anti-Human CD20 BD Biosciences Cat # 560631; RRID:AB_1727447
BV711 Mouse Anti-Human CD21 BD Biosciences Cat # 563163; RRID:AB_2738040
PE-CF594 Mouse Anti-Human CD38 BD Biosciences Cat # 562288; RRID:AB_11153122
Brilliant Violet 510 anti-human CD3 Antibody BioLegend Cat # 317332; RRID:AB_2561943
Brilliant Violet 510 anti-human CD14 Antibody BioLegend Cat # 301842; RRID:AB_2561946
APC/Cy7 anti-human CD197 (CCR7) BioLegend Cat # 353212; RRID:AB_10916390
Brilliant Violet 650 anti-human
CD127 (IL-7Ra) Antibody
BioLegend Cat # 351325; RRID:AB_11125369
Brilliant Violet 650 anti-human CD3 Antibody BioLegend Cat # 317324; RRID:AB_2563352
Brilliant Violet 711 anti-human CD27 Antibody BioLegend Cat # 302833; RRID:AB_11219201
Brilliant Violet 785 anti-human CD38 Antibody BioLegend Cat # 303530; RRID:AB_2565893
Alexa Fluor 700 anti-human CD45RA Antibody BioLegend Cat # 304120; RRID:AB_493763
Brilliant Violet 421 anti-human
CD279 (PD-1) Antibody
BioLegend Cat # 329920; RRID:AB_10960742
PE/Dazzle 594 anti-human CD4 Antibody BioLegend Cat # 300548; RRID:AB_2563566
Brilliant Violet 711 anti-human CD8a Antibody BioLegend Cat # 301044; RRID:AB_2562906
Brilliant Violet 510 anti-human CD14 Antibody BioLegend Cat # 301842; RRID:AB_2561946
Brilliant Violet 510 anti-human CD19 Antibody BioLegend Cat # 302242; RRID:AB_2561668
BB515 Rat Anti-Human CXCR5 (CD185) BD Biosciences Cat # 564624; RRID:AB_2738871
BV605 Mouse Anti-Human CD56 BD Biosciences Cat # 562780; RRID:AB_2728700
PE-Cy7 Mouse Anti-Human CD25 BD Biosciences Cat # 335824; RRID:AB_2868687
PE-Cy5 Mouse Anti-Human CD183 BD Biosciences Cat # 551128; RRID:AB_394061
PerCP-eFluor 710 Anti-Human CD3 eBioscience Cat # 46-0037-42; RRID:AB_1834395
APC Anti-Human CD19 BioLegend Cat # 302212; RRID:AB_314242
Brilliant Violet 421 Streptavidin BioLegend Cat # 405226
anti-human IFN-gamma mAb 1-D1K, purified Mabtech Cat # 3420-3-1000; RRID:AB_907282
Anti-human IFN-g mAb (7-B6-1), biotin Mabtech Cat # 3420-6-250; RRID:AB_907273
PE-Streptavidin Agilent Cat # PJRS25-1
APC-Streptavidin Agilent Cat # PJ25S
Virus strains
SARS-CoV-2 (lineage B) isolate
SARS-CoV-2/human/Liverpool/REMRQ0001/2020
Ian Goodfellow (University of Cambridge)
isolated by Lance Turtle (University of
Liverpool) and David Matthews and
Andrew Davidson (University of Bristol)
N/A
(Continued on next page)
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 23
iScienc
e
Article
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Biological samples
Donor blood samples Mortimer Market Centre and Ian
Charleson Day Centre,
RoyalFree Hospital
N/A
Chemicals, peptides, and recombinant proteins
PepTivator CMV pp65, human Miltenyi Biotec Cat # 130-093-438
PepTivator SARS-CoV-2 Prot_S Delta Miltenyi Biotec Cat # 130-128-763
PepTivator SARS-CoV-2 Prot_S Com Miltenyi Biotec Cat # 130-127-953
PepTivator SARS-C oV-2 Prot_S Omicron Miltenyi Biotec Cat # 130-129-928
Wuhan Hu-1 Alpha Biochem Shanghai Customized
Wuhan Hu-1 Beta Biochem Shanghai Customized
Alpha Mutation Pool Biochem Shanghai Customized
Beta Mutation Pool Biochem Shanghai Customized
BD Cytofix/Cytoperm Fixation/
Permeabilization Solution Kit
BD Biosciences Cat # 554714; RRID:AB_2869008
Phytohemagglutinin-L (PHA-L) Sigma-aldrich Cat # 11-249-738-001
mmPACT AMEC Red Substrate, Peroxidase 2B Scientific Cat # SK-4285
Vectastain ELite ABC PK-6100 2B Scientific Cat # PK-6100
LIVE/DEAD Fixable Blue Dead Cell Stain Invitrogen Cat #L23105
BD CompBeads anti-mouse Ig, k BD Biosciences Cat # 552843; AB_10051478
PEI-Max Polysciences, Inc Cat # 23966
Biotin Sigma-Aldrich Cat #B4639
Imidazole Sigma-Aldrich Cat #I2399
Recombinant biotinyl ated spike protein This study and
42
N/A
Recombinant biotinyl ated RBD protein This study and
42
N/A
SARS-CoV-2 spike S1 re protein Peter Cherepanov Laboratory
(The Francis Crick Institute)
32
N/A
Critical commercial assays
Bright-Glo Luciferase kit Promega Cat #E2650
Experimental models: Cell lines
HEK-293T/17 AmericanType Culture Collection ATCC CRL-11268
FreeStyle 293F Thermofischer Scientific Cat #R79007
HeLa-ACE2 James Voss Laboratory (The
Scripps Research Institute)
86
N/A
HEK-293T cells expressing Renilla luciferase
(Rluc) and SARS-CoV-2 Papain-like
protease-activatable circularly permuted
firefly luciferase (FFluc)
Nicholas Matheson Laboratory
(University of Cambridge)
87
N/A
Recombinant DNA
HIV-1 luciferase reporter vector (pCSLW) Seow et al.
88
N/A
HIV-1 packaging construct (p8.91) Zufferey et al.
89
SARS-CoV-2 spike WT (Wuhan hu-1) Katie Doores Laboratory
(King’s College London)
88
N/A
SARS-CoV-2 spike Omicron (BA.1/B.1.1.259.1) Katie Doores Laboratory
(King’s College London)
N/A
(Continued on next page)
ll
OPEN ACCESS
24 iScience 26, 105862, January 20, 2023
iScienc
e
Article
RESOURCE AVAILABILITY
Lead contact
More information and req uests for reage nts should be directed to and will be fulfilled by th e lead contact ,
Laura E McCoy (l.mc coy@ucl.ac.uk).
Materials availability
This study did not generate unique resource s or materials.
Data and code availability
Any additional information required to reanalyze the data reported in this study is available from the lead
contact upon request.
EXPERIMENTAL MODEL AND SUBJECT DETAILS
Ethics statement
The protocols and study documents for the study were approved by the local Research Ethics Committee
(REC) Berkshire (REC 16/SC/0265) and South Central - Hampshire B (REC 19/SC/0423). All subjects enrolled
into the study provided written informed consent. The study complied with all relevant ethical regulations
for work with human participants and conformed to the Helsinki declaration principles and Good Clinical
Practice (GCP) guidelines.
Patient recruitment and sampling
There were 110 HIV + par ticipan ts who were virally supp ressed and on cART and 64 HIV-n egative heal thy
controls were recruited as part of either the Jenner II or the Vaccine in Clinical Infection (VCI) cohorts.
PBMCs and plasma (o r ser um) w ere col lected at the following t imepoi nts: base line, post-first dose
(R12 day s following the first dose), post-second dose (%70 days following the second dose), pre-third
dose (R70 days following the second dose) to evaluat e wani ng response,
41,90
and post-third dose
(>7 days following the third dose). Foll owing each dose days cut-off was chosen to allo w development
of a de-novo (or recall) immune respon se foll owing vacci nati on.
88
Participants received a mix of avail able
SARS-CoV-2 v accination (Pfizer-BioNTech’s BNT162b2; Moderna’s mRNA-1273 or Astra-Zeneca’s
AZD1222) according to Joint Committee on Vaccination and Immunization, UK, guidelines.
4
Not eve ry
participant was sampled at all timepoints. At each visit, participants were asked to report any history of
SARS-CoV-2 infection.
Between vaccinations, 7 previously SARS-CoV-2 naı
¨
ve participants (3 HIV-, 4 HIV+) reported a SARS-CoV-2
infection, as such, any subsequent timepoints were moved into the ‘prior SARS-CoV-2 infection’ group for
analysis. Similarly, 4 participants with prior SARS-CoV-2 reported a further infection (3 HIV-, 1 HIV+). All par-
ticipants were recruite d at the Mor timer Market Cent re for Sexual Health and HIV Researc h and the Ian
Charleson Day Centre at the RoyalFree Hospital (London, UK) following written informed consent as
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
RBD-Avi-His tag Katie Doores Laboratory
(King’s College London)
N/A
SARS-CoV-2 Spike Avi-His tag Katie Doores Laboratory
(King’s College London)
36
N/A
BirA Addgene Cat # 20856
Software and algorithms
Flowjo 10.8.1 FlowJo, LLC https://www.flowjo.com
GraphPad Prism 9.0.0 GraphPad https://www.graphpad.com
Cytobank Beckman Coulter https://premium.cytobank.or g
Other
MSCRN-IP DURA 0.45UM CLEAR 50/PK Millipore Cat # MAIPN45 50
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 25
iScienc
e
Article
part of a study approved by the local ethics board committee. Additional information about demographic
and sampling can be found in Table S1.
METHOD DETAILS
PBMC isolation
Whole blood was colle cted i n hepari n-coated tube s. PBMC s were i solated from wh ole blood via de nsity -
gradient sedimentation. Whole blood was first spun v ia centrifugation for 5 minat 800g. Plasma was then
collected, ali quo ted and store d at 80
C for further use. Remaining blood was diluted with RPMI (Gibco,
Paisley, UK), layered over an appropriate volume of Ficoll (Cytiva, Uppsala, Sweden) and then spun via
centrifugation for 20 minat 800g without brake. The PBMC layer was collected and washed with RPMI to
be spun via centrifugation f or 10 minat 400g. PBMCs were stained with t rypan blue and counted using
Automated Cell Counter (BioRad, Hercules, Cal iforn ia, USA). PBMCs were then cryo preserve d in a cry ovial
in cell recovery freezing medium containing 10% dimethyl sulfoxide (DMSO) (Honeywell, Seetze, Germany)
and 90% heat-inactivated fetal bovine serum (FBS) and stored at 80
C in a Mr. Frosty freezing container
overnight before being tr ansferr ed into liquid ni tro gen for further stor age. If pr esent , serum s eparator
tubes were spun at 400g for 5 min to collect serum and then stored at 80
Cforfurtheruse.
Semi-quantitative S1 ELISA
This assay was set up previously by our lab.
45,91
In a 96-half-well NUNC Maxisorp
TM
plate (Nalgene, NUNC
International, Hereford, UK), three columns were coated overnight at 4
Cwith25mLofgoatanti-human
F(ab)
0
2 (Jackson ImmunoResearch, Ely, UK) (1:1000) in PBS, the o ther nine columns were coated with
25 mL of SARS-CoV-2 WT S1 protein (a kind gift from Peter Cherepanov (Ng et al., 2020), The Francis Crick
Institute) at 3 mg/mLinPBS.Thenextday,plateswerewashedwithPBS-T(0.05%TweeninPBS)andblocked
for 1 hour (h) at room temperature (RT) with assay buffer (5% milk powder PBS-T). Assay buffer was then
removed and 25 mL of patient plasma at dilutions from 1:501:10,000 in assay buffer added to the S1-
coated wells in duplicate. Serial dilutions of known concentrations of IgG were added to the F(ab)
0
2
IgG-coated we lls in triplicate to generate an internal stand ard c ur ve. Aft er 2 h of incubation at RT, plates
were washed with PBS-T and 25 mL alkaline phosphatase (AP)-conjugated goat anti-human IgG (Jackson
ImmunoResearch) at a 1:1000 dilution was add ed to each well and incubated for 1 hat RT. Plates were
then washed w ith PBS-T, and 25 mL of AP substrate (Sigma Aldrich, St Loui s, Missouri, USA) added. Optical
density (OD) was measured using a Multiskan
TM
FC (Thermo Fisher-Scientific, Horsham, UK) plate reader at
405 nm and S1-specific IgG titers were interpolated from the IgG standard curve using 4PL regression
curve-fitting on GraphPad Prism 9 (GraphPad, San Diego, California, UK).
Total IgG ELI SA
To measure total IgG levels in plasma, a 96-half-well NUNC Maxisorp plate (Nal gen e) was entirely coated
overnight at 4
Cwith25mLofgoatanti-humanF(ab)
0
2 (1:1000). As above, plates were washed in PBS-T and
blocked for 1 hat RT i n assay buffer. 25 mL of seri al dilutions of patient plasma (1:1 00 to 1:10
7
) were added in
duplicates to the plate alongside known concentrations of IgG in triplicates. As above, after 2 h of incuba-
tion at RT, p lates were w ashed with PB S-T and 25 mL AP-conjugated goat anti-human IgG was added and
then incubated for 1 hat R T. Pl ate s were washed wi th P BS-T , and 25 mL of AP substrate added. ODs were
measured using a Multiskan FC plat e read er at 405 nm and total IgG titers interpol ated f rom the IgG stan-
dard curve using 4 PL regression curve-fitting on GraphPad Prism 9.
IgG purification
As the PLWH participants in this study were on cART which can interfere with the lentivirus-based pseudo-
type neutralization assay IgG was purified from plasma using a Pierce 96-well pro tein G spin plate
(ThermoFischer Scientific). Plasma was incubated in wells containing protein G at RT for 30 min. The
captured IgG was then eluted with 0.1M Glycine (pH = 2-3) twice into 2M Tris (p H = 7.5-9) buffer. To remove
Tris/Glycine buffer from the purified IgG , the elua te was concen trated (Thermo Scientific Pierce Prote in
Concentrator PES, 50K MWCO, 0.5 mL) and washed thrice at 10000 rpm for 10 min before quantification
by measuring absorbance of 280 nm on a NanoDrop
TM
(ThermoFischer, Rockford, Illinois, UK). The entire
volume of purified IgG was then filtered sterile using a 0.22 mm PDVF hydrophilic membrane Fil trE X filter
plate (Corn ing, Corning, NY, U SA) and stored at 4
Cforfurtheruse.
ll
OPEN ACCESS
26 iScience 26, 105862, January 20, 2023
iScienc
e
Article
Pseudovirus production
In a T75 flask, 3 3 10
6
HEK-293T cells were seeded in 10 mL of complete DMEM Dulbecco’s Modified Eagle’s
Medium (Gibco) suppl emente d with 10% FBS and 50 mg/mL penicillin-streptomycin. The next day, the
following transfection mix was prepared: 1 mL of Opti-MEM
TM
(Gibco); 90 mL of PEI-max (1 mg/mL);
10 mg of p8.91 HIV-1 gag/pol packaging plasmid
89
; pCSLW HIV-1 luciferase reporte r vector p lasmid
92
and 5 mg of either S ARS-CoV-2 spike plasmid of interest, specifically WT (Wuhan-hu-1) or Omicron (BA.1/
B.1.1.529.1)
88
as indicated in the results section. The transfection mix was left to incub ate for 20 min before
being added to the cells and left to incubate at 37
C5%CO
2
for 72 h before being collected and filtered
through a 0.45 mm filter (Millipore, Cork, Ireland) and either used directly in an assay or stored at 80
C.
Pseudovirus neutralization
Neutralization assays were performed in 96-well plates by adding either duplicate serial dilutions of neat
plasma in complete Dulbecco’s Modified Eagle medium (Thermo F isher Scientific-UK) (DMEM) starting
at 1:20 dilution for HIV-negative samples or the appropriate amount of purified IgG for H IV + s amples
to give a starting dilution equivalent to 200 or 400 mg/mL of IgG as based on total IgG. These dilutions
were incub ated with the appropri ate amount of fil tered pseudotyped vi rus for 1 h at 37
C5%CO
2
before
adding 10000/mL HeLa-ACE2 cells
86
(kind gift from James Voss, The Scripps R esearch I nstitut e, USA ) in
100 mL per well. After a 72 h incubation at 37
C5%CO
2
, the supernatant was removed, and cells lysed.
Bright-G lo
TM
luciferase substrate (Promega, Madison, Wisconsin, USA) was added, and relative light
unit (RLU) values were read on a Glomax (Promega) or BioTek Synergy
TM
H1 (Agilent) plate reader.
RLU readouts were used to calculate the reciprocal inhibitory dilution at which 50% of the virus activity is
neutralized by plasma (ID
50
) for each sampl e on GraphPad Prism 9.
Live neutralization
The SARS-CoV -2 v irus used in th is st udy was the wild-t ype (l ineage B) isolate S ARS- CoV-2/ human/Li ver-
pool/REMRQ0001/2020, a kind gift from Ian Goodfellow (University of Cambridge, UK), isolated by Lance
Turtle (University of Liverpool, UK) and David Matthews and Andrew Davidson (University of Bristol,
UK)
93,94
( Plasma was heat-inactivated at 56
C for 30 mins before use, and neutralizing antibody titers at
50% inhibition (NT
50
) measured as previously described.
87,95,96
In brief, luminescent HEK293T-ACE2-30F-
PLP2 repo rter cells (clone B 7) expressing SARS-CoV-2 Papain-like protease-activatable circularly permuted
firefly luciferase (FFl uc) were seeded in flat-bottomed 96-well plates. The n ext day, S ARS-CoV- 2 viral stock
(MOI = 0.01) was pre-incubated with a 3-fold dilution series of each sample f or 2 hat 37
C, t hen added to
the cells. 16 h post-infection, cells were lysed in Bright-Glo
TM
Luciferase Buffer (Promega) diluted 1:1 with
PBS and 1% NP-40, and F Fluc activ ity measured by luminometr y. Experimen ts were conduct ed in duplicate .
To obtain NT
50
, titration curves were plotted as FFluc vs log (serum dilution), then analyzed by non-linear
regression using the Sigmoi dal, 4PL, X is log(co ncentration) function in GraphPad P rism. NT
50
were quan-
titated when (1) at least 50% inhibition was observed at the lowest serum dilution tested (1:10, or 1:20 for
pre-diluted samples), and (2) a sigmoidal curve with a good fit was generated. Samples with no detectable
neutralizing activity were assigned an arbitrary NT
50
equivalent to the lower limit of q uant ific ation.
Production of bi otinylated protein
To produce biotinylated spike and receptor binding domain (RBD) protein, HEK-293F cells were seeded at
1 3 10
6
cells/mL in Freestyle 293 Expression Medium (Gibco). The next day, a transfection mix was
prepared (for 200 mL of cells) of 72 mg of spike-Avi-His tag or RBD-Avi-His tag plasmid and 18 mgofBirA
plasmid
36,88
into 11 mL of Opti-MEM
TM
,alongside2mLofPEI-Max and 3 mL of 10 mM biotin, and left
to incubate at 37
C5%CO
2
in a shaking incu bator for 7 days beforeharvestingforpurification. The
supernatant was purified using an imidazole (Sigma-Aldrich) buffer at a final concentration o f 20 mM during
binding to the His GraviTrap (Cy tiva) column and 500 mM imidazole for el utio n. The e lu ted p ro tein was
then concentrated with a 100KD Amicon Ultra concentrator (Millipore) and washed with PBS before
quantification using a NanoDrop
TM
. Biotinylated protein was then further purified through size exclusion
chromatography using an AKTA pure system with a Superdex 200 Increase 10/300 GL column
(Sigma-Aldrich) to select for fractions c ontaining trimeric spike or RBD protein.
B cell phenotypic flow cytometric analysis
As previously described ,
42
1 mg of biotinylated spike with either streptavidin-conjugated allophycocyanin
(APC) (Agilent , Santa Clara, Cali fornia , USA) or phycoerythri n ( PE) (Ag ilen t) and 0.5 mg of biotinylated RBD
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 27
iScienc
e
Article
with BV421 (BioLegend, San Diego, California, USA) were incubated for 30 minutes in the dark to generate
fluorochrome-linked biotinylated tetramers. Previously cryopreserved aliquots of 5 3 10
6
or 10 3 10
6
cell
aliquots of PBMCs were quickly thawed in PBS, t hen stained with a panel of phenotyping antibodies and
biotinylated tetramers (see Table S2) or phenotyping antibodies only for FMO controls. PBMCs were
then washed with PBS and fixed in Cytofix /C ytoper m (BD, Franklin Lake s, New Jersey, USA) buffer.
Compensation controls we re prepar ed accordin g to manufacturer’s instructions using Anti-Mouse Ig, k
CompBeads (BD). Samples were acq uired on an LSRFortessa (BD) flow cytometer. Data was analyzed
(see Figure S2 fo r gating s trategy) on FlowJo v10 (BD). For further analysis of the phenotype of spi ke-spe -
cific MBCs, analysis was limited to samples for which at least 50 cells were acquired in the CD19
+
CD20
+
CD38lo/-IgD-MBCs(excludingCD27
CD27
+
cells) spike-PE + spike-APC +gateaspreviouslydened.
42
T cell phenotypic flow cytometric analysis
Purified cryopreserved PBMCs samples were thawed and rested for 2 hours at 37
CincompleteRPMIme-
dium (RPMI supplemented with penicillin-streptomycin, L-Glutamine, HEPES, non-essential amino acids,
2-Mercaptoethanol, and 10% FBS) (Gibco).
45
After 2-hour incubation, cel ls were washed and pl ated in a
96-round bottom plate at 0.5-1 3 10
6
per wel l and stained for chemokin e markers (CXCR3, CC R7 and
CXCR5) for 30 minutes at 37
C.Cellswerethenwashedandstainedwithsurfacemarkersat4
Cfor
20 min with different combinations of antibodies (see Table S 3) in the pre sence of fixable live/dead stain
(Invitrogen, Eugene, Oregon, USA). After 20 min of incubation, cells were washed with PBS, and fixed
with 4% paraformaldehyde for 15 minat RT. Samples were acqu ired on a LSRFortessa X-20 using
FACSDiva version 8.0 (BD) and subsequent data analysis was performed using FlowJo v10 (BD). The
gating str ategi es used for flow cytometry experiment s ar e provided in Figures 6 and S6.
High-dimensional data analysis
Visualization of high-dimensi onal single-cell data (viSNE)
97
and FlowSOM
98
analyses were performed using
the Cytobank platform (https://www.cy tobank. org ). Concatenated fil es w ere used to evaluat e overall CD4
and CD8 T cell landscape in different group s. Cell s were manually g ated for lymphocy tes, singl ets, CD14
CD19
live cells, CD3
+
and CD4
+
or CD8
+
and then subjected t o viSNE analysis. The viSNE clustering anal-
ysis was performed on 8 parameters (CCR7, CD45RA, CD127, PD-1, CD38, CXCR5, CXCR3, CD25). Equal
event sampling was selected across all samples. FlowSOM was then performed using the same markers
outlined previously for viSNE and with the following parameters: number of clusters 100, number of meta-
clusters 10; the size of clusters 15 pixels (Cytobank default).
Ex vivo IFN-g ELISpot as say
This assay was performed using cryopreserved PBMCs samples. Briefly, 96-well ELISpot plates (S5EJ044I10;
Merck Millipore, Darmstadt, Germany) pre-wetted with 30 mL of 70% ethanol for 2 min before washi ng with
200 mL of sterile PBS. Anti-IFN-g coating antibody (10 mg/mL in PBS; clone 1-D1K; Mabtech, Nacka Strand,
Sweden) was then added and the p lat es incubated overnight at 4
C. Prior to addition of cells, ELISpot
plates were washe d wit h PBS and blocked with R10 (RPMI supplemented with penicill in- strept omycin,
L-glutamine, and 10% FBS) for a minimum of 2 hat 37
C. PBMCs samples were thawed and rested for 2
hours at 37
CinR10.Cellswerethenaddedat23 10
5
cells/well, in duplicat e, and stimulated with over-
lapping pepti de po ols at 2 mg/mL for 16–18 hat 37
C. Unstimulated cells were used as a negative control
while PHA (10 mg/mL, Sigma-Aldrich) stimulated cells were used as a positive control. Plates were then
washed with 0.05% Twee n/PBS (Sigma Aldrich) and incubated for 2 h with an IFN-g dete ctio n antibody
(1 mg/mL; clone mAb-7B6-1; Mabtech) fo llowed by 1 h incubation with AP-conjugate d streptavidin
(1:1000 in PBS, Mabtech). Plates were t hen washed and visualized using the VECTASTAIN Elite ABC-
HRP kit according to the manufacturer’s instructions (Mabtech). Antigen-specific T cell responses were
quantified by subtracting the number of spots in unstimulated cells from the peptide stimulated cells.
An additional threshold of > 5 SFU/10
6
PBMCs w as used. Partici pants who lacked T cell responses to the
positive stimuli (PHA) or where antig en-specific respon ses found to be lower than two standard deviations
of negative controls were excluded from t he results.
Overlapping peptide pools
For the detection of antigen-specific T cell responses, purified cryopreserved PBMCs were stimulated with
the following peptide pools: (1) Wild-type S ARS-CoV-2 spike ; SARS-CoV -2 spike PepTiv ato r protein
pools (Miltenyi B iotec, Gladbach, Germany) were used to test T cell responses against ful l spike prot eome.
ll
OPEN ACCESS
28 iScience 26, 105862, January 20, 2023
iScienc
e
Article
(2) VOC spike-specific peptide pools; the WuhanHu-1 and variant pools containing peptides from the Wu-
han Hu-1, Alpha (B.1.1.7), Beta (B.1.351), Delta (B.1.617.2) and Omicron (BA.1/B.1.1.529.1) sequences (9, 19,
32 and 83 peptides, respecti vely) were used to define T c ell responses to mutat ed Spi ke sequences in
SARS-CoV-2 variants. Alpha and Bet a peptide pools were synt hesized by GL Bioche m Shanghai Ltd, China
and previously used in.
34
The corresponding controls to Alpha and Beta pools with Wuhan Hu-1amino acid
sequences were compar ed in parallel within the same don or. Delta and Omicron pools were obtained from
Miltenyi Biotec. (3) Non-SARS-CoV-2 antigens: Peptide pools of the pp65 prot ein of human cytomegalo-
virus (CMV) (Miltenyi Biotec), or HIV-1 Gag peptide pools (NIH AIDS Reagent Repository) were used as pos-
itive/neg ative controls.
QUANTIFICATION AND STATISTICAL ANALYSIS
All statistical analysis were carried out in GraphPad Prism 9.0 (GraphPad). All tests were two- ta iled . Mann-
Whitney U-test (MWU) was used to compare unpaired, non-parametric datawhilst Wilcoxon matched-pairs
sign rank test (WMP) was used to compare paired, non-parametric data. Non-parametric Spearman test
was used for correlat ion analysis b etween two sets of data. Where app rop riate, median for groups is
shown. Statistical significance in the figures is sho wn as p value >0.05 (*); >0.01 (**); >0.001 (***)
and >0.0001 (****).
ll
OPEN ACCESS
iScience 26, 105862, January 20, 2023 29
iScienc
e
Article
Do we know now whether viral vector-based vaccines exhibit this same trend in PLWH? Were there any commonalities (perhaps some sort of comorbidity) between these 4/5 SARS-CoV2 naive patients that produced no detectable nAb, even after 3 doses? I assume these are the same ~4 patients with co-morbidities in figure S1g? Is there any explanation as to why two patients had no humoral or cell-mediated responses after 2/3 doses of the vaccine? Is something that has been reported elsewhere?