MOT 2020 - Transcriptional Profiles of Adjuvanted Hepatitis B Vaccines Display Variable Interindividual Homogeneity But A Shared Core Signature

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

VACCINES Copyright © 2020


The Authors, some
Transcriptional profiles of adjuvanted hepatitis B rights reserved;
exclusive licensee
vaccines display variable interindividual homogeneity American Association
for the Advancement
but a shared core signature of Science. No claim
to original U.S.
Government Works
Laurane De Mot1*†, Viviane Bechtold1*, Vanesa Bol1, Andrea Callegaro1, Margherita Coccia1,
Ahmed Essaghir1, Dicle Hasdemir2,3, Fernando Ulloa-Montoya1, Emilio Siena4, Age Smilde3,
Robert A. van den Berg5, Arnaud M. Didierlaurent1‡§, Wivine Burny1‡, Robbert G. van der Most1‡||

The current routine use of adjuvants in human vaccines provides a strong incentive to increase our understanding
of how adjuvants differ in their ability to stimulate innate immunity and consequently enhance vaccine immuno-
genicity. Here, we evaluated gene expression profiles in cells from whole blood elicited in naive subjects receiving

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


the hepatitis B surface antigen formulated with different adjuvants. We identified a core innate gene signature
emerging 1 day after the second vaccination and that was shared by the recipients of vaccines formulated
with adjuvant systems AS01B, AS01E, or AS03. This core signature associated with the magnitude of the hepatitis
B surface-specific antibody response and was characterized by positive regulation of genes associated with
interferon-related responses or the innate cell compartment and by negative regulation of natural killer cell–­
associated genes. Analysis at the individual subject level revealed that the higher immunogenicity of AS01B-­
adjuvanted vaccine was linked to its ability to induce this signature in most vaccinees even after the first vaccination.
Therefore, our data suggest that adjuvanticity is not strictly defined by the nature of the receptors or signaling
pathways it activates but by the ability of the adjuvant to consistently induce a core inflammatory signature
across individuals.

INTRODUCTION quantities. AS03 contains -tocopherol in an oil-in-water emulsion,


Adjuvants in human vaccines can potentiate the immunogenicity of and AS04 contains MPL adsorbed on aluminum salt (Alum). Mecha-
vaccine antigens, particularly those derived from purified proteins, nistic studies in mice treated with AS-containing vaccines have iden-
through activation of a complex network of innate immune re- tified key mechanisms leading to efficient antigen presentation to
sponses. This activation is thought to be a prerequisite for eliciting adaptive immune cells in the lymph node (10–13). Links between these
adaptive immune responses (1, 2). Human system biology studies mechanisms identified in mice and the peripheral blood signatures
have contributed to unraveling the transcriptional pathways acti- detected in nonhuman primates have been proposed (13–15).
vated by adjuvanted vaccines (3–8), but such studies have yet to AS-containing vaccines have demonstrated immunogenicity and
include a direct head-to-head comparison of different adjuvants. In efficacy in diverse human populations, irrespective of age [reviewed in
particular, the understanding of how the nature of these transcrip- (2, 9, 16)]. These populations include older or immunocompromised
tional pathways and the intensity of their activation can influence adults (the licensed AS01B-adjuvanted herpes zoster and AS04-­
interindividual variability in the immune response to adjuvanted adjuvanted HBV vaccines), adolescents (the licensed AS04-adjuvanted
vaccines remains elusive. HPV-16/18 vaccine), and children (the AS01E-adjuvanted vaccines
Here, we evaluated gene expression profiles in cells in whole against malaria and tuberculosis). Furthermore, several age ranges
blood elicited in naive subjects receiving two doses of vaccines are targeted by AS03-adjuvanted influenza vaccines. Although age
containing the prototypic hepatitis B virus (HBV) surface antigen is an important determinant for an individual’s vaccine responsive-
(HBs) formulated with AS01B, AS01E, AS03, or AS04. These four ness, a host of other, mostly nonheritable, intrinsic factors are also
adjuvant systems (AS) were designed to combine various proin- relevant (17). Their cumulative effect causes substantial variation in
flammatory immunostimulants, each with their own distinctive fea- prevaccination (baseline) immune status and vaccine responses be-
tures (9). Their components include 3-O-desacyl-4′-monophosphoryl tween individuals, even within a population spanning the same age
lipid A (MPL) and saponin fraction 21 extracted from Quillaja segment (18, 19). For instance, baseline frequencies of monocytes and
saponaria Molina (QS-21), both present in AS01B and AS01E. AS01E natural killer (NK) cells in healthy adults can vary from negligible to
is composed of a half-dose of AS01B with respect to MPL and QS-21 constituting about half of the total immune cell population (17, 20).
Accepted population-based thresholds of vaccine-induced protec-
tive immunity, such as those established for the hemagglutination
1
GSK, 1330 Rixensart, Belgium. 2Bioinformatics Laboratory, University of Amsterdam, inhibition antibody titers used in influenza vaccine evaluations, do
1012 WX Amsterdam, Netherlands. 3Biosystems Data Analysis Group, University
of Amsterdam, 1012 WX Amsterdam, Netherlands. 4GSK, 53100 Siena, Italy. 5GSK,
not account for such variability. A better understanding of inter-
Rockville, MD 20850, USA. individual variation in vaccine responses could enable tailoring
*These authors contributed equally to this work as co-first authors. these responses to specific needs, particularly with respect to low-­
†Present address: Clarivate Analytics, 2600 Berchem, Belgium. responding or at-risk individuals.
‡These authors contributed equally to this work as co-senior authors.
§Present address: Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland. In this study, we built on previous analyses of the innate and
||Corresponding author. Email: robbert.x.van-der-most@gsk.com adaptive responses detected (21, 22) by performing a deeper analysis

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 1 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

of the blood transcriptional responses at various time points. We


focused on the heterogeneity of the early response across individuals
who received the same adjuvanted vaccine. Using this global gene
profiling approach, we expanded the targeted gene expression data
(22) by identifying additional functional pathways. Using the Alum-­
adjuvanted HBs vaccine as the comparator, previous studies reported
the most potent enhancement of adaptive and innate responses for
the AS01B-containing HBs vaccine, and an overall ranking (AS01B ≥
AS01E > AS03 > AS04 > Alum) emerged from these studies (21, 22).
We also previously detected rapid (within 3 to 6 or 24 hours) re-
sponses of inflammatory markers in recipients of any of these HBs/
AS vaccines, monocytes and neutrophils in recipients of the AS01B
or AS01E (AS01B/E) formulations, and cytokines implicated in innate
immunity in recipients of the AS01B/E or AS03 formulations (22).
Interferon (IFN)–related responses and increased C-reactive pro-

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


tein (CRP) and interleukin-6 (IL-6) concentrations correlated with
adaptive responses and these markers were shared in recipients of
the AS01- or AS03-adjuvanted vaccines (22), despite their distinct
modes of action in mice (11, 12). This raised the question of whether
these commonalities reflected truly convergent response profiles
or a core inflammatory signature, with more nuanced distinctions
remaining undetected.
Here, we identified a distinct core gene expression signature
induced by hepatitis B vaccine adjuvants, irrespective of their com-
position, and investigated the association of this signature with
antibody response. Through an innovative data analysis approach to
study the interindividual heterogenicity of the response, we found
that this core signature was more consistently induced by some ad-
juvants, that is, some adjuvants induced this core response in most
recipients. The current study provides criteria to consider in adju-
vant selection for the development of future human vaccines.

RESULTS
Expression profile after one AS01B dose resembles those Fig. 1. Kinetics and clustering profiles of gene expression in cells from whole
seen after two AS01E or AS03 doses blood induced by adjuvanted vaccines. Baseline-corrected gene expression was
determined for the primary gene set (fig. S1) at 3 to 6 hours and 1, 14, and 30 days
To assess the dynamic regulation of gene expression, we used
after the first dose (D0+, D1, D14, and D30) and 3 to 6 hours and 1, 3, and 7 days
linear-mixed models and moderated t tests on datasets normalized
after the second dose (D30+, D31, D33, and D37) of hepatitis B virus surface antigen
by GCRMA (guanine cytosine robust multi-array analysis) (fig. S1). (HBs) vaccines adjuvanted with AS01B, AS01E, AS03, AS04, or aluminum hydroxide
When applying a false discovery rate (FDR)–adjusted P value of (Alum), administered to HBs-naive adults. (A) Effects on the whole blood transcrip-
<0.05 for the gene expression observed for at least one adjuvant and tome, expressed for each adjuvant group as numbers of differentially expressed
on at least one time point, we retained a primary gene set of 1791 genes [DEG versus baseline; false discovery rate (FDR) P value of <0.05], are pre-
unique, differentially expressed genes (DEGs). Consistent with pre- sented. Increased expression and decreased expression (up and down) were de-
vious findings (22), transcriptomic response (in terms of numbers fined by the sign of the moderated t statistics (>0, up and <0, down, indicating
of DEGs) across all time points were highest for the AS01B, AS01E, increased and decreased gene expression, respectively). (B) The dendrogram above
and AS03 groups (≤1050 DEGs); modest for the AS04 (≤102 DEGs) the heatmap presents the global similarities among the timed gene expression
profiles per adjuvant group and was obtained by hierarchical clustering of the
group; and low for the Alum (≤4 DEGs) group (Fig. 1A). This was
baseline-corrected gene expression presented in the heatmap (red/blue: up-/
in line with the more complex innate immune stimulation provided
down-regulated genes represented by positive/negative moderated t statistics)
by AS in comparison to Alum (22). for each group and time point (among D1, D31, D33, and D37).
After the first vaccination, responses for the AS01B, AS01E, and
AS03 groups (in numbers of DEGs) were negligible after 3 to 6 hours
[day 0+ (D0+)] but prominent on D1 (24 hours after administration) and D37 following the hierarchy observed after the first dose:
with the largest D1 response detected in the AS01B group. These AS01B > AS01E > AS03. Responses in the AS04 group peaked at D14
responses had decreased by D14 after the first vaccination and were after dose 1, but the response kinetics after dose 2 were comparable
low in the AS01B group and absent in the AS01E and AS03 groups to that seen for the other AS groups.
at 1 month after vaccination (D30). A greater number of DEGs for To extend this analysis, we identified adjuvant and time point
these three groups was seen after dose 2. In contrast to the responses conditions with similar expression profiles, focusing on the tran-
at D1, these three groups had similar numbers of DEGs at D31 scriptionally most active time points. Hierarchical clustering by
(1 day after the second dose) with the subsequent responses at D33 adjuvant and time point revealed a dichotomy between “early” (D1

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 2 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

and D31) and “late” (D33 and D37) DEG responses (Fig. 1B). The AS01E group, and more strongly down-regulated at D31 in the
predominant clustering patterns were mostly time point based for AS01B/E and AS03 groups. Overall, the changes functional responses
the vaccines containing AS01B, AS01E, or AS03 but treatment-based represented by C1, C2, and C3 were transient: Responses decreased
for the AS04- or Alum-containing vaccines. The D1 response for to undetectable or balanced (similar numbers of DEGs showing
the AS01B group clustered with the D31 response for the AS01B, up-regulation as those showing down-regulation) at either D14 for
AS01E, or AS03 groups. This early branch also contained the D1, AS01B/E and AS03 or D30 for AS04 after the first dose and at D33
D31, and D33 responses for the AS04 group. The late branch con- for the decrease in the NK cell response (C3) or D37 for the innate
tained mostly responses to the second dose (that is, D33 responses and IFN pathway responses (C1 and C2) after the second dose. The
for AS01B/E and AS03 and D37 responses for all four AS), as well as exception was the inverted response in C1 for AS01B at D37.
a separate cluster for all time points in the Alum group. In contrast to the early response clusters (C1 and C2), the late
Thus, similarities were detected in the D1 responses for AS01B response clusters (C4 and C5) in the AS01B/E and AS03 groups
and the D31 responses for AS01B, AS01E, and AS03. The different generally persisted at D37. The C4 response exhibited complex dy-
clustering patterns—temporal for AS01 or AS03 versus treatment-­ namics. Responses related to plasma cells and B and T lymphocytes
based for AS04 or Alum—possibly signified different modes of (C4) were mostly low and reduced at D1 (a low percentage of DEGs
action for the three sets of adjuvants, which could be grouped as in the cluster and most of those were down-regulated). However, at

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


AS01B/E/AS03, AS04, and Alum on the basis of this analysis. D30+ (3 to 6 hours after the second dose) in the AS01B and AS03
groups, the adaptive response cluster (C4) was activated, possibly
AS01B, AS01E, and AS03 induce similar functional patterns associated with the proliferation of these cells. This was followed at
after two doses D31 by primary repression in the AS01B/E groups or bidirectional
We characterized the functional pathways perturbed by each adju- regulation in the AS03 group and at D37 by uniform up-regulation
vant. To identify gene sets with similar expression kinetics, we per- in these three groups, with AS01B > AS01E ≈ AS03. A reason for the
formed clustering analyses of the primary gene set of 1791 DEGs. greater response in the AS01B group at D37 was that several T cell–
We clustered the DEGs based on the time profiles in each adjuvant related modules (M83, M97, M7.4, M19, and M7.0) were activated
group (see fig. S1 and Materials and Methods for analysis details). only in this group.
Using these gene clusters, we performed enrichment analysis using Up-regulation of metabolism-related functions (C5) was weak at
blood transcriptional modules (BTMs) as defined in (23) and ob- D1 in the AS01B/E groups and modest at D14 in the AS01B group
tained 88 modules (FDR q value of <0.05). We performed an addi- but stronger after the second dose in the AS01B/E and AS03 groups,
tional clustering step, which yielded five functional clusters, referred persisting with a high percentage of DEGs up-regulated through
to as C1 to C5 (Fig. 2, table S1, and data file S1). Of these clusters, D37 in the AS01B/E groups. This is consistent with proliferation of
two were related to regulation of early response parameters: C1 in- cycling adaptive cells and aligns with the trends observed for other
cluded 35 BTMs, many of which related to innate immune cells, and AS01-adjuvanted antigens or oil-in-water emulsion-adjuvanted
C2 included 8 BTMs, many of which related to the IFN pathway antigens (6, 8).
and antiviral sensing. C3 included 10 BTMs, which corresponded Together, responses to HBs/AS01B/E and HBs/AS03 were char-
principally to NK cells. Late response clusters pertained to the acti- acterized by a core signature comprising the activation of genes
vation of adaptive immune cells and cell division (C4 with 21 BTMs) regulating innate cellular or IFN responses or both, consistent with
or to metabolism and involved cell cycling, heme biosynthesis, and previous data (22), and down-regulation of NK cell–associated genes.
platelet and erythrocyte functions (C5 with 14 BTMs). Aligned with The responses followed an intergroup gradient (AS01B > AS01E > AS03)
the hierarchy between groups seen with the condition-based clus- after the primary vaccination, which disappeared after the secondary
tering (Fig. 1B), the AS01B/E and AS03 groups had DEGs indicating vaccination, because these three AS shared similar modular response
responses for each cluster, whereas the AS04 group had DEGs asso- levels at peak (D31). In contrast, HBs/AS04 triggered exclusively
ciated with responses for just C1, and the Alum group had no DEGs innate cell–related responses, which peaked at D14, rather than D1,
enriched in any cluster. as was observed for the other AS-adjuvanted vaccines.
With these clustered data, we noted different kinetics of the
functional responses. Innate cell–related responses (C1) were mark- Interindividual variability in the early response depends
edly up-regulated in the AS01B/E and AS03 groups 1 day after each on AS adjuvanticity
vaccination. In line with the trends seen in DEG responses across We asked whether all individuals receiving the same adjuvant would
the groups (Fig. 1A), the strongest up-regulation at D1 occurred in respond in the same fashion. Two approaches were followed to
the AS01B group. Responses in this group were mostly repressed at define the interindividual variability in the transcriptional response.
D37. In the AS04 group, responses were moderate at D14 and ab- First, individual response patterns were visualized using cumulative
sent at D30, but responses at D31 (1 day after the second dose) were distribution curves for each of the functional clusters. Second, syn-
increased when compared to the response 1 day after the first dose. chronization of gene expression at the group level was approached
IFN-related responses (C2) in the AS01B/E and AS03 groups were by principal components (PC) analysis (PCA).
robustly up-regulated after each dose. The magnitudes at D1, in terms In the first approach, the number of genes that were significantly
of percentages of DEGs in this functional cluster, were AS01B ≈ up- or down-regulated in each BTM cluster was counted for each
AS01E > AS03, but the magnitudes were similar among these groups individual. We set the threshold absolute fold change of >1.5 in gene
after dose 2. These kinetics resembled those of the STAT1 up-­ expression over its expression at D0. To quantify the homogeneity
regulation for these groups previously identified by quantitative in the numbers of perturbed genes across all subjects receiving the
polymerase chain reaction (qPCR) (22). NK cell gene signatures same vaccine, the number of significantly up- or down-regulated genes
(C3) were slightly down-regulated at D1, most prominently for the in each BTM cluster was then plotted as cumulative distribution

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 3 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

Fig. 2. Functional charac-


terization of the transcrip-
tomic vaccine responses.
The figure shows the amount
of activity in the 88 blood
transcriptional modules
(BTMs) enriched (FDR q
value of <0.05) using DEGs
induced by the indicated
adjuvanted vaccine. BTMs
were grouped in five data-­
driven clusters (C1 to C5)
using k-means clustering

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


analyses. Gene sets included
in the modules have been
described (23). Sphere size
indicates the percentage
of the DEGs in the BTM,
calculated as the number
of DEGs (FDR P < 0.05)
among all genes in the
BTM × 100, over the total
number of genes present
in the microarray chip for
the given BTM. Sphere color
indicates the change in
activity of the BTM based
on the ratio of up-regulated
to number of DEGs in the
BTM (as determined by
moderated t statistics >0
for up-regulated and <0 for
down-regulated). Changes
in activity were considered
positive (red) if more DEGs
were up-regulated than
down-regulated, no change
(0) if the amount of up-
and down-regulated DEGs
were equal, or negative
(blue) if more DEGs were
down-regulated than up-­
regulated. Dashed vertical
lines separate the first and
second administrations of
the same adjuvanted vac-
cine; solid vertical lines
separate the different ad-
juvanted vaccines. Day of
sample collection is indi-
cated along the bottom
from early (D0+ to D37)
for each.

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 4 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

curve for each vaccine group (Fig. 3A). This workflow was adapted interindividual heterogeneity between the adjuvant groups, with
from (24). Cumulative distribution curves of early (D1 and D31) patterns varying both by time point and by functional cluster
responses in the individual subjects revealed different degrees of (Fig. 3B). At D1, individual responses of genes related to the innate
response, IFN pathway, and NK cells
(C1 to C3) displayed a hierarchy among
the adjuvants of AS01B > AS01E > AS03,
except for NK cells in which AS01E >
AS01B. For instance, for expression of
genes related to the IFN pathway (C2),
changes in ≥25 genes were detected in
~70% of AS01B and 55% of AS01E re-
cipients but in only ~25% of the AS03
recipients. As expected, none of the AS04
or Alum recipients reached this high of
a response.

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


At D31, a clear separation between
the groups emerged with respect to
innate cell responses (C1), adaptive
immune cell responses (C4), and IFN-­
mediated responses (C2). At this time
point, we detected relatively homogeneous
individual responses for AS01 recipients
for these clusters. For example, for C1,
>80% of individuals have between 160
and 180 genes for AS01 differentially
expressed at D31, whereas this was more
spread out for the other AS vaccines.
Heterogeneity among individual re-
sponses was higher for AS03 recipients
than for the AS01 recipients; however,
the amount of heterogeneity at D31 in
the AS03 group was closer to that of the
AS01 groups when compared to the
heterogeneity at D1. The most hetero-
geneous expression at D31 for innate
cell responses, adaptive immune cell re-
sponses, and IFN-mediated responses
was observed for the AS04 or Alum re-
cipients. Responses for NK cell–related
functions (C3) were stronger in terms of
numbers of genes perturbed for AS01B
recipients and more modest for AS01E
or AS03 recipients. The responses in
the metabolism-related cluster (C5) were
heterogeneous at both D1 and D31 with
a slight separation in the curves between
the Alum and AS04 recipients and the
AS01B/E and AS03 recipients.
In the second approach, we quantified
the interindividual synchronization of
each gene’s expression in subjects of the
same adjuvant group. For each gene
and each adjuvant group, we performed
Fig. 3. Relationship between the adjuvants and individual transcriptional responses 1 day after each vaccina-
PCA on datasets that contained expres-
tion. (A) Schematic overview of the workflow followed to generate the reverse cumulative distribution plots. (B) Reverse
cumulative distribution plots representing the numbers of responsive genes 1 day after primary (D1) or secondary
sion values over all time points and all of
(D31) vaccination (│FCD1/D0│ > 1.5 and │FCD31/D0│ > 1.5, respectively, where FC refers to fold change), detected in the group’s subjects (39,200 probe sets;
cells from whole blood from individuals who received two doses of vaccine with the indicated adjuvants. Results are fig. S1). We then used the percentage of
stratified by the five identified BTM clusters (C1 to C5; see Fig. 2). Genes included at D1 or D31 were those of the variation captured by PC1 as a metric to
primary gene set (fig. S1). quantify the amount of synchronization

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 5 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

(Fig. 4A). The percent synchronization


was slightly higher in AS01B recipients
than in all other recipients (Fig. 4B).
We used the percent of synchronization
for the highly synchronized outlying
genes to compare the synchronization
among different groups. Using the max-
imum value of the outlying genes, we
found that the groups followed the hier-
archy we observed in other analyses:
~85% in the AS01B/E groups, ~70% in
the AS03 group, and ~55% in the AS04
and Alum groups (Fig. 4B).
To provide biological context, we clus-
tered the highly synchronized genes,

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


those exhibiting percent of synchroni-
zation of >55%, using synchronization
metric values over five adjuvants. This
cluster analysis of the highly synchro-
nized genes resulted in three main func-
tional synchronization clusters (SC1, SC2,
and SC3) (Fig. 4C). We observed that
both SC1 and SC3 displayed a highly
synchronized expression for the AS01B/E
or AS03 groups. Synchronization levels
in SC1 (of which the gene shown in
Fig. 4A is a representative) displayed
the intergroup hierarchy as expected
from the preceding analyses, whereas
SC3 displayed similar levels for both
AS01 groups. We functionally annotated
the SCs using BTM enrichment analy-
ses (see fig. S1). SC3 contained mainly
IFN-inducible genes, whereas the genes
in SC1 were not significantly enriched
in any BTM (table S2). SC2, by contrast,
displayed only a highly synchronized ex-
pression for AS01B recipients and in-
cluded genes mediating T cell activation,
inflammation, or antigen presentation.
Collectively, the results from analy-
ses at the individual level indicated that
the differences observed in group response
magnitudes in terms of numbers of DEGs
(Figs. 1A and 2) between the AS01B/E
and AS03 groups at D1 appeared linked
to differences in the proportions of re-
sponding subjects rather than to func-
tional differences (Fig. 3B). Vaccination
with HBs/AS01 B led to a high inter-
subject homogeneity even after the
primary vaccination, and this initial homo­ Fig. 4. Synchronization analysis workflow and results. Figure shows a schematic presentation of the workflow
followed to assess the synchronization of gene expression among subjects. (A) Plots represent the kinetics of the
geneous response was particularly evident
standardized expression profiles of a representative gene (for example, ApoL6, encoding apolipoprotein L6) over the
for the genes comprising the core signa-
nine time points. The values in each plot represent the percentages of variance captured by the first principal com-
ture of IFN-inducible genes and genes ponent (PC1). PCA, principal components analysis. (B) Percentages of variance captured by PC1 among subjects of
regulating innate and NK cell–related func- the same adjuvant group are presented. Boxplots represent the medians, and the lower and upper whiskers present
tions (Fig. 3B). This core signature was the first quartile − [1.5 × the interquartile range (IQR)] and the third quartile + (1.5 × IQR), respectively, based on a
identified on the basis of the group re- default option in the R program. (C) The percentage of variance explained by PC1 is presented for each functional
sponses to the AS01B/E- or AS03-adjuvanted synchronization cluster (SC) and for each adjuvant group. BTMs included in each SC are presented in table S2.

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 6 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

vaccines (Fig. 2). The difference in heterogeneity between AS01B/E


and AS03 observed at D1 persisted after the second dose (D31)
(Fig. 3B), despite the apparent similarity of gene expression patterns,
especially those associated with the IFN pathway, identified at the
adjuvant-group level (Fig. 2).

Early activation of specific innate pathways is associated


with the adaptive response
The observation of the apparent convergence of the blood tran-
scriptomic signatures identified for AS01B/E and AS03 recipients
prompted us to assess whether these gene responses were associated
with the HBs-specific adaptive response at D44, as reported previ-
ously (21). To that aim, we performed PC regression of the log2 fold
change in the expression values of genes within the primary gene set
and used the adjuvant group, the first three PCs, and the technical

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


scaling factor as predictors. Expression at D1 or D31 versus D0 and
expression at D31 versus D30 were analyzed.
The gene loadings summarized in PC1 were associated with the
scaling factor values. Gene loadings of PC2 for D31 versus D30 were
associated with the antibody response at D44 (P = 0.001) but not
with any of the CD4+ T cell response parameters (Fig. 5, A and B).
As identified by pathway analysis, genes contributing to this associ-
ation (that is, those with the highest ranking PC2 loadings) were Fig. 5. PCA results of the transcriptomic response at D31 compared to D30.
mostly implicated in regulation or activation, or both, of the IFN (A) Statistical associations between the (log 10) adaptive response parameters
pathway (Fig. 5C). Previously, we identified an association between (antibodies and CD4 T cells) at 14 days after dose 2 (see table S5) and the transcrip-
early responses after the second dose—in particular, increased tional response at D31/D30 [represented by the scores of the second principal
serum IL-6 and CRP concentrations and IFN pathway activation— component (PC2) for the 1791 genes of the primary gene set; fig. S1] were calculated
and the humoral or cellular responses to these vaccines at D44 (22). using linear regression models. Note that the PC1 was not used because it was
Here, consistent with the previous analysis (22), we found a strong strongly associated with the microarray scaling factor. PCA was performed on the
association (P < 0.001) between the PC2 that summarizes these early log2 FCs in gene expression at D1 and D31 over baseline expression (D1/D0 and
D31/D0, respectively) or at D31 over expression on D30 (day of the second dose)
responses for the current cohort and the PC2 for the D31 versus
(D31/D30). P values were not adjusted for multiplicity; statistical significance
D30 transcriptomic data using linear regression analysis (fig. S2). (P < 0.01) is indicated in bold. Polyfunctionality indices were calculated on
To integrate the outcomes of the different analyses, we subjected CD4 + T cells positive for at least CD40L alone or with an additional one, two,
the 300 genes most highly ranked in PC2 loadings and therefore or three markers among interleukin-2 (IL-2), tumor necrosis factor– (TNF-), or
most likely to be associated with the antibody titers to cluster analysis interferon- (IFN-) (21). (B) D44 antibody (Ab) concentrations (in log10) are plotted
using the five functional clusters defined in Fig. 2 (Fig. 6). These against the PC2 scores (D31/D30). Each point represents an individual subject,
genes were annotated mostly in the three clusters regulating early color-coded by the adjuvant treatment. (C) Plots of loadings of expression values
and NK cell responses (C1, C2, and C3; see Fig. 2), with a small (PC2 and PC3) (FC D31/D30) for the primary gene set are shown. For ease of
fraction of the genes belonging in the late response clusters (C4 and C5). representation of the data, an arbitrary cutoff of 0.070 was applied for the PC2
However, ~60% (184 of 300) of the genes in the analysis were not loadings. Genes with loadings below this cutoff are shown as gray dots; genes with
a PC2 loading of ≥0.070 are labeled, red indicates an association with the IFN pathway,
associated with any of the 88 BTMs contained in the five functional
and black indicates no association.
clusters. When we subjected these 184 genes to Ingenuity Pathway
Analysis, we found that only few canonical pathways were signifi-
cantly enriched, of which four were related to the already identified persisted after the second dose of AS03, regardless of the apparent
innate (TREM1, FcRIIB, CCR5, and salvage pathways of pyrimidine similarity in responses to the AS01B/E- or AS03-adjvuvanted vaccines
ribonucleotides) and cell cycle responses (table S3). at the group level.
The cluster analysis also visualized the individual heterogeneity To assess whether these findings were unique to the HBs/AS
within the same treatment group. We demonstrated that the adjuvant vaccines, we evaluated these same 300 genes and five BTM clusters
groups differed with respect to the numbers of “responders” after the in 15 recipients of the malaria vaccine RTS,S/AS01E (4, 25) and ob-
second dose (where responders are individuals expressing the specific served a similar pattern to the transcriptomic responses 1 day after
transcriptome signature identified as being associated with antibody the second vaccination (D29 versus D28 data) (Fig. 6).
titers). Most AS01B or AS01E recipients responded, but only about Collectively, our results showed that the transcriptomic responses
half of the AS03 recipients responded, with a marked separation after immunization with HBs antigen formulated with AS01 B,
between nonresponders and responders. Proportions of responders AS01E, or AS03 were described by five clusters of BTMs. The sta-
were low among AS04 and Alum recipients, although some of these tistical association between transcripts relating to innate immunity
individuals mounted modest responses for specific genes. These re- at D31 (comprising transcriptomic responses of innate-related,
sults support our results from the reverse cumulative distribution IFN-related, and NK cell–related genes) and the antibody titers
analysis (Fig. 3) that, compared to the response induced by two doses at D44 is consistent with the previously identified association (22).
of AS01B, substantial heterogeneity among individual responses Responses to two doses of AS01B/E or AS03 shared a core signature;

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 7 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

sponses (C3), which was followed by an


increase in adaptive cell-related responses
(C4). The last cluster related to metabo-
lism (C5) was most likely linked to both
innate and adaptive responses. C5 also
contained BTMs related to platelet acti-
vation, which may provide interesting
avenues for further research. The de-
scriptive analysis of the transcriptional
data revealed a dichotomy between the
AS01 B-, AS01 E-, or AS03-formulated
vaccines, which induced shared tran-
scriptional responses, and vaccines for-
mulated with AS04 or Alum, for which
responses were weaker (AS04) or nearly

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


absent (Alum). Second, transcriptional
responses for several clusters, C3 and
C4 for AS01B/E and C1 to C4 for AS03,
were stronger after the second dose than
after the first dose. Third, the nature of
the early response was similar between
the groups receiving AS01B/E and the
group receiving AS03, although these AS
contain different adjuvants with no
common immunostimulants. We de-
termined that these three AS shared a
core signature that was characterized by
a strong IFN-related (C2) response, which
is consistent with previous data (22), and
by innate responses and down-regulation
of NK cell–associated responses. Despite
Fig. 6. Interindividual heterogeneity in transcriptomic responses linked to antibody responses. Linear regres- this overall similarity in the responses
sion models were used to assess the association between the transcriptomic response at D31 and the antibody re- induced by these three AS, compared to
sponse at D44. Data of the five leftmost heatmaps represent the 300 genes (data file S1) (presented in rows) with the the first dose of AS01E or AS03, the first
highest loadings (absolute values) in PC2 from the PCA performed on the log2 FC D31/D30 matrix (see Fig. 5). The dose of AS01 B appeared to induce a
vertical colored bar shows the corresponding BTM cluster number(s) for the genes (see Fig. 2). Each column corre-
strong innate signal in a higher number
sponds to an individual subject. The rightmost heatmap represents the data generated for these 300 genes, using
samples from 15 healthy malaria-naive participants in a clinical trial evaluating the AS01E-adjuvanted RTS,S malaria
of individuals, coinciding with a lower
vaccine (4, 25). The rightmost heatmap was generated from a Pearson correlation–based hierarchical clustering anal- degree of interindividual heterogeneity.
ysis performed on the log2 FC D29/D28 data, with D29 corresponding to D1 after the second dose of RTS,S/AS01E. Although this apparent difference be-
tween the three groups was reduced
after the second dose, a considerable
however, the responses differed in the amount of interindividual degree of heterogeneity persisted in the AS03 cohort. Last, the pre-
heterogeneity. viously identified association between early transcriptional responses
and D44 antibody titers (22) was confirmed for the full transcrip-
tomic dataset.
DISCUSSION Collectively, the data painted a diverse picture of functional
A better understanding of the mechanisms underpinning adjuvant-­ responses, which were quantitatively or qualitatively different be-
induced innate immunity, including its interaction with adaptive tween the two doses and sometimes both quantitatively and qualita-
immunity, should inform the development and licensure of novel tively different. Activation of BTMs C1 to C3—linked to myeloid
vaccines and adjuvants for various human populations. For the four cells, dendritic cells (DCs), Toll-like receptor (TLR) and IFN signal-
AS evaluated here, we previously unraveled mechanisms of action ing (through a positive regulation), and to NK cells (through a
in animal models (10–13) and characterized the innate response in negative regulation)—was already detected after the first injection
human whole blood (22). To further our understanding, here, we of the AS01B/E- and AS03-formulated vaccines. This reflected a
used a systems biology approach and performed transcriptomics data direct activation of innate immunity, which also occurs in animal
analyses, both at the group and individual subject level. models (11–13). BTMs linked to adaptive responses (C4), compris-
Several conclusions can be drawn. First, four of the five identi- ing genes associated with B cells and CD4+ T cells, were observed
fied BTM clusters collectively captured the evolution from innate to later, at D37, in line with the higher HBs-specific responses mea-
adaptive immunity—early stimulation of innate responses and the sured after a second dose. The low and heterogeneous transcrip-
IFN pathway (C1 and C2), along with suppression of NK cell re- tomic response to the vaccine adjuvanted with AS04 suggested that,

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 8 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

in humans, this AS is a less potent stimulator of innate immunity is likely not mediated by TLR stimulation. Instead, the adjuvant
than are AS01B/E or AS03, with the caveats that (i) the described property of AS03 is, at least partially, mediated by inducing endo-
responses are systemic, thus not fully reflecting any local responses; plasmic reticulum stress (12, 33). Data from mice show that adju-
and (ii) the absence of the identified core transcriptomic signature vants, irrespective of their composition and the innate pathways that
does not necessarily imply the absence of involvement of other they trigger, can induce a shared inflammatory response characterized
pathways. We detected activation of modules in the C1 cluster reg- by common antigen presentation and cytokine-related pathways
ulating cells of the myeloid, but not lymphoid, lineages after each (34, 35). It is therefore conceivable that the innate pathways activated
dose of HBs/AS04, aligning with the changes in the frequencies of by AS01B/E and AS03 in humans also eventually converge toward a
innate cells and the increased IL-6 concentrations previously reported shared innate response that is detectable in blood.
in recipients of this adjuvanted vaccine (22). A potential concern However, AS01B/E- and AS03-induced responses also exhibited
with our analysis is the timing of data collection, which could have differences, particularly after the first dose. Compared to HBs/AS03,
missed peak responses. However, in mice, a TLR4-dependent, tran- the magnitude of the response (the number of DEGs) induced by
sient inflammatory response and increased numbers of activated the first dose was notably higher for HBs/AS01B, with an overall
antigen-loaded monocytes and DCs were observed within 1 day expression profile (Fig. 1B) that resembled those induced by two
after administration of AS04 (10), aligning with the activation of doses of AS01E- or AS03-containing vaccines. Furthermore, our

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


human DCs in response to MPL observed ex vivo (26). Thus, we data indicated that AS03 differs from AS01B/E after the second dose
think it unlikely that our selected time points missed detection of in terms of interindividual response heterogeneity. The differences
the peak response. Overall, the consistency in our gene-based and in numbers of DEG and degrees of heterogeneity might reflect dif-
BTM-based analyses (IFN-related signals and cell cycle–associated ferences not only in the composition but also in the type of interac-
and lymphocyte proliferation–associated responses) with previous tions between components of the AS01 and AS03 adjuvants. The
observations in human vaccinees (22) and in animal studies (13) interaction between squalene and tocopherol in AS03 results mainly
supports the ability of our study to further translational research in additive effects, whereas MPL and QS-21 in AS01 show molecular
efforts linking human and animal model data. and cellular synergy, resulting in stronger activation of the innate
Another key observation was the difference between the BTM immune response (12, 13). One key feature of this synergy is IFN-
responses measured after the first and second doses of AS01B/E- or production by lymph node–resident lymphoid cells, such as NK
AS03-adjuvanted vaccines. Responses in the clusters containing innate cells, as well as an overall increase in innate responses. This may
and IFN-associated BTMs (C1 and C2) were more prominent after explain the more prominent activation of innate (C1)– and IFN
the second dose, particularly for AS03. Moreover, the down-regulation (C2)–associated BTMs that we detected after the first dose of AS01B/E.
of NK responses (C3) was stronger at D31 compared to that at D1 The higher level of innate stimulation by AS01B/E then resulted in a
for the AS01B/E or AS03 groups. Similar observations were made for more homogeneous response than was observed for AS03. Although
AS01E-adjuvanted malaria or tuberculosis vaccines (4, 6). Because interindividual variability in baseline gene expression was not as-
the current subjects were naive before immunization, an obvious sessed, we postulate that the stronger innate response mitigated the
difference between the first and second dose responses was the effects of any baseline variability on the immune response. This hy-
presence of immunological memory at the time of the second dose. pothesis is supported by data showing that the inflammatory immune
As proposed (22), and given the key role of early IFN- in the mode status before vaccination can negatively influence the response to
of action of adjuvants (13, 27), a mechanism could be envisaged by an Alum-adjuvanted HBs vaccine, presumably due to the weaker
which CD4+ T cell memory elicited after the first dose enhances the adjuvanticity elicited by this vaccine (36).
subsequent down-regulation of NK cell responses and up-regulation Using PCA, we detected a correlation between antibody re-
of IFN- responses, possibly through IL-2–mediated NK cell activation sponses and 300 genes captured in the PC2 of genes regulated at
and migration (28, 29), which then leads to an enhanced innate re- D31 versus D30. From the PC2 loadings, we concluded that the
sponse. Consistent with such a mechanism are the peripheral IFN-­– up-regulation of transcripts related to IFN signaling and innate
producing NK cells observed in human recipients of AS01-adjuvanted cells observed 1 day after dose 2 correlated with the adaptive re-
tuberculosis and malaria vaccines (29, 30) and the increased serum sponses 13 days later (D44) and that there was an inverse correla-
IFN- concentrations in human or nonhuman primate recipients of tion between these adapt­ive responses and expression of transcripts
AS01-adjuvanted HBs or malaria vaccines (4, 13, 22, 31), which were related to NK cells. The positive correlation is consistent with
all detected mostly after the second dose. In contrast, AS03-adjuvanted the previously described correlation of antibody titers with D31
H1N1 or H5N1 split influenza vaccines induce up-regulation of the interferon-gamma (IFN-) and induced protein 10 (IP-10, also
IFN signaling pathway after the primary vaccination (3, 8, 32). A pos- known as CXCL 10) production (22). The presence of this positive
sible explanation for this response to the influenza vaccines is that correlation is also consistent with several observations made in hu-
primed participants, who had received influenza vaccines or experienced mans and mice: in human or murine recipients of AS01-adjuvanted
influenza infection previously, had cross-reactive nucleoprotein-­ vaccines (4, 5, 13), in human recipients of a single dose of either
specific memory CD4+ T cells that provided the innate-stimulating AS03- or MF59-adjuvanted H5N1 or H1N1 vaccines (3, 8, 32, 37)
memory signal at the time of the first dose. Alternatively, differences or nonadjuvanted seasonal influenza vaccine (38), and in virus-­
in early transcriptomics responses could reflect trained innate infected mice (39). To investigate this further, we analyzed the ex-
immunity (22). pression profiles of these 300 genes in blood from participants of a
The similarities in gene signatures in cells in whole blood be- clinical malaria study collected 1 day after the second dose of RTS,S/
tween the vaccines adjuvanted with AS01B/E or AS03 is remarkable, AS01E vaccine (4, 25). The gene profile induced by this vaccine was
given that these AS are thought to have different mechanisms of similar to the one observed in specific individuals in the AS01B/E or
action. In the absence of the TLR4 ligand MPL, AS03 adjuvanticity AS03 groups. These results confirmed the individual variability of

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 9 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

the response to another AS01E-adjuvanted vaccine in a different by all institutional Ethics Committees and conducted in accordance
clinical setting. with the Helsinki Declaration and Good Clinical Practice guidelines
Together, the human blood cell gene expression signatures seen (21, 22). Written informed consent was obtained from each subject
after two doses of the HBs/AS01B/E or HBs/AS01 vaccines studied before trial participation. Healthy HBV-naive men or women 18 to
here or other AS01- or AS03-adjuvanted vaccines (3–6, 22), com- 45 years of age received two intramuscular injections, 1 month apart,
bined with the local immune cell recruitment and pathway activa- of vaccines containing HBs antigen (20 g of dose) adjuvanted with
tion inferred from preclinical models (10–13), support our models AS01B, AS01E, AS03A, or with AS04 (Fendrix, GSK) or with Alum as
for the modes of action of AS01 and AS03 adjuvanticity. At D31, the Al(OH)3 (Engerix-B, GSK).
peak in up-regulation of innate cell– and IFN-associated functions Evaluations of adaptive and innate immunogenicity (primary and
coincided with depletion of signatures regulated to NK cell pheno- secondary endpoints, respectively) were previously performed for
types, other lymphoid cells, such as nonspecific B and T cells. This their respective according-to-protocol (ATP) cohorts (21, 22). The
depletion may indicate local recruitment of the circulating cells out current analyses were conducted for the 112 subjects included in
of the primary lymphoid organs and blood, into the peripheral lym- the adaptive immunogenicity ATP cohort, as well as in subset 1 or 2
phoid organs, as occurs in human recipients of AS01-adjuvanted of the innate immunogenicity ATP cohort recruited at the Immune
malaria and tuberculosis vaccines (4, 6). Early cytokines produced Health Centre, La Louvière, Belgium (n = 18 for AS01B, 23 for

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


in the muscle and draining lymph node could enter the bloodstream AS01E, 28 for AS03, 22 for AS04, and 21 for Alum). The demo-
and instigate the activation of, among others, IFN-inducible genes graphic profile of subjects across groups was balanced in terms of
(15, 40). At D37, HBs-specific T and B cells would then migrate to age (mean age, 33.9 years; SD, 6.6 years) and gender (49.1% females),
the bloodstream, aligning with the observed up-regulation of corre- and most subjects (97.3%) were white Caucasian (table S4). For the
sponding BTMs, while blood monocyte and neutrophil cell counts current study cohort, an overview of the innate and adaptive im-
have returned to steady-state levels, as evident from previous data mune response features (serum concentrations of CRP and key cyto-
(22). Rather than an absolute decrease of innate cell counts, the re- kines, frequencies of different CD4+ T cell phenotypes, and antibody
pression of innate cell transcripts seen for HBs/AS01B at D37 may concentrations) is presented in data file S2.
reflect changes in the relative proportions of innate and adaptive Whole blood samples for microarray analyses were collected be-
cells. Correspondingly, in human recipients of AS03-adjuvanted fore vaccination (D0 and D30), 3 to 6 hours after each dose (D0+
vaccine, frequencies of blood innate cells were significantly reduced and D30+) or 1 day after each dose (D1 and D31), 14 days after dose 1
by D7 (32). However, because blood-derived transcriptional signa- (D14), and 3 and 7 days after dose 2 (D33 and D37). Adaptive re-
tures reflect an aggregate of changes expressed in discrete cell popula- sponses were previously evaluated at 2 weeks after dose 2 (D44) (22).
tions and are affected by the relative cell type proportions and absolute
cell counts, verification of these concepts in humans requires a cell- RNA extraction, microarray hybridization,
based systems approach and investigation of local tissue responses, and data preprocessing
including those of muscle and peripheral lymphoid organs. Total RNA was extracted and purified as described (22). RNA quality
This study is limited by a small sample size. The previous study was assessed using a Bioanalyzer-2100 (Agilent Technologies); 50 ng
of recipients of the same vaccine preparations identified an associa- of total RNA was used for complementary DNA (cDNA) amplifica-
tion between the CD4+ T cell response and the IFN-signaling path- tion, fragmentation, and labeling using the Ovation Kit (NuGEN).
way (22). We did not detect this association here possibly because Fragmented cDNA was hybridized using hgu133 Plus2.0 GeneChip
the previous study had less parameters for testing as compared to (Affymetrix; 54,675 probe sets). After screening of arrays by standard
the current study. A limitation of the data analysis is that, because quality control (QC), 84 of 1008 samples were missing or excluded.
our interpretation of BTM regulation was based on the numbers of The data were then GCRMA-normalized, log2-transformed, and fil-
perturbed genes within the BTMs, rather than on fold changes in tered by interquartile range (IQR > 0.75), retaining 3770 probe sets.
expression, functional differences between the adjuvants with re- Data were submitted to the National Center for Biotechnology Infor-
spect to the intensity of the regulation were not considered in that mation Gene Expression Omnibus (GEO) (GSE116975).
evaluation. Probe set annotation was performed using Bioconductor package
Last, it will be of interest to extend the current transcriptomics hgu133plus2.db. Probe sets with no available annotation were re-
analysis of adjuvanted HBs vaccines in antigen-naive young adults moved. Data for genes mapping to multiple probe sets were collapsed
to analysis of other AS-containing vaccines in different populations. using PCA, without centering or scaling, of a matrix containing all
Of interest are AS01-adjuvanted herpes zoster or tuberculosis vac- data corresponding to the gene. The probe set with the highest
cines in populations of older adults or primed individuals (41, 42), Pearson correlation with PC1 was used; this probe set interrogated
respectively, or AS03-adjuvanted H5N1 influenza vaccines in influenza-­ 2194 unique gene symbols.
primed adults (43). The recent consideration of AS03 as an adjuvant
for COVID-19 (coronavirus disease 2019) vaccine development (44) Per-gene analysis
further emphasizes the relevance of in-depth analysis of the mode A linear-mixed model [R package; limma (45, 46)] was fitted to the
of action of vaccine adjuvants in clinical research. entire transcriptomic dataset. Moderated t statistics of the expres-
sion relative to D0 (with up- or down-regulation defined as values
>0 or <0, respectively) were calculated per gene, per time point after
MATERIALS AND METHODS vaccination, and per adjuvant group, along with the corresponding
Study design P values and Benjamini-Hochberg FDR-adjusted P values. In total,
The protocol for the previously reported, observer-blinded, multi- 1791 genes were identified as differentially expressed (FDR P < 0.05)
center, randomized controlled trial (NCT00805389) was approved at ≥1 time point and in ≥1 adjuvant group (the “primary gene set”;

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 10 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

fig. S1 and data file S1). For selected time points of this gene set (D1, for the log10 values of either HBs antigen; antibody concentrations;
D31, D33, and D37), the moderated t statistics were organized in a frequencies of HBsAg-specific CD4+ T cells expressing at least one
matrix, with DEG in rows and adjuvant group and time point con- of IL-2, CD40L, or IFN- and TNF- (tumor necrosis factor–), as
ditions in columns, and subjected to hierarchical clustering analysis measured by flow cytometry; or polyfunctional HBsAg-specific CD4+
based on Euclidean distance. Data were scaled by dividing each data T cells on the CD4+ T cell polyfunctionality index (Fig. 5A) (21, 48).
point by the maximal value within the same matrix row. Ai was modeled as a function of regression coefficients  of the scores
of the first three PCs and the treatment effect for each AS group
BTM enrichment analysis relative to that of the Alum group, corrected for the technical scal-
BTM enrichment analysis was performed for the primary gene set ing factor, and with  of the Alum group intercept (0) considered
using previously annotated BTMs (23). Partitioning clustering analysis as baseline
was performed by adjuvant group on a matrix containing moderated
t statistics, with the genes (differentially expressed in ≥1 time point ​​A​ i​​  = ​​  0​​  + ​​  1​​ × adjuvant group + ​​  2​​  × PC1 [ ​EX​  Dx/Dy​​ ] + ​​  3​​  ×
for the same adjuvant) in rows and time points in columns, using PC2 [ ​EX​  Dx/Dy​​ ] + ​​  4​​ × PC3 [ ​EX​  Dx/Dy​​ ] + ​​  5​​ × ​SF​  Dx/Dy​​​
the k-means function (Rcmdr package, R) with ≤100 iterations and
10 seeds. The appropriate number of clusters (k = 6 for AS01B, 5 for Generated P values were not adjusted for multiplicity. This ex-

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


AS01E, 5 for AS03, and 4 for AS04) was determined using a graphical ploratory study was not powered to evaluate multiple regression
test (a standard statistical approach referred to as the “elbow” method). models. PC2 loadings were inverted to keep the correlated genes
BTM enrichment (FDR q value of <0.05) was assessed by cluster, and the positive correlation with the antibody titers on the right-
using hypergeometric tests (t-mod package, R), with the unfiltered hand side of the figures (Fig. 5, B and C). Similar linear regression
list of genes of the microarray chip set as background. The 88 BTMs analyses were performed to assess the association between gene ex-
enriched in ≥1 cluster and in ≥1 group (see “primary BTM set”; fig. S1) pression and previously assessed (22) innate immune response data
were grouped using k-means partitioning clustering (k = 5; elbow after the second dose. Input parameters included the PC2 of hema-
method) of a matrix containing the percentages of DEG (with their tological parameters, CRP and cytokines, and the PC2 of the gene
sign depending on the behavior of the majority of genes), with BTMs expression data, both measured after the second dose and expressed
in rows and adjuvant group and time point conditions in columns. in fold changes of the amounts at D30+, D31, D33, or D37 over
The amount of BTM activation was visualized in a bubble chart as D30. Innate parameters included CRP and the cytokines IL-1, IL-6,
described in the legend of Fig. 2. TNF-, IL-5, IL-10, IP-10, IFN-, and MCP-1. Innate hemato-
logical parameters included the cell subsets lymphocytes, monocytes,
Transcriptome synchronization analyses eosinophils, basophils, neutrophils, and white blood cells.
The GCRMA-normalized transcriptomics dataset was filtered to To generate Fig. 6, the above models were used to identify the
remove subjects not adhering to cohort criteria and then subjected 300 genes with the highest absolute loadings in the PC2 from the
to within subject autoscaling. Samples from subjects with probe sets PCA performed on the log2 fold change D31 over D30 matrix.
exhibiting no change in expression over time and probe sets for Where possible, the genes were assigned to the BTM clusters (C1 to
which ≥40% of the data (considering all time points and subjects) C5). Genes that could not be assigned in one of the five BTM clus-
were absent were excluded, leaving 39,200 probe sets in the analysis. ters were analyzed using the Ingenuity Pathway Analysis algorithm
Data from each probe set were stored by the adjuvant group in a (QIAGEN).
matrix containing all nine time points (rows) and subjects (columns). We also compared the core gene expression profile obtained for
Missing data were imputed using the missMDA (47) package in R. the subjects from the current study with that obtained for subjects
The amount of gene expression synchronization, across subjects and from a previously described observer-blind, randomized phase 2a
time points, was expressed as the percentage of variance captured study of the RTS,S/AS01E malaria vaccine conducted in healthy,
by the first PC (VARPC1) of a PCA performed on each matrix. On malaria-naive adults (4, 25). One cohort in this study received three
the basis of the data for the AS04 and Alum groups, only highly doses of RTS,S/AS01E at 28-day intervals (“RRR” group; N = 21; %
synchronized data points (VARPC1 of >55% in ≥1 group) were re- female/male ratio, 37/63; mean age, 30 years). Transcriptional
tained, leaving 1529 probe sets in the analysis. After excluding responses to vaccination as compared to prevaccination baseline
probe sets not mapping to any gene and additional probe sets map- (D0) were described previously (4). To enable comparison with the
ping to the same gene, 922 probe sets were retained. Hierarchical current dataset generated for fold changes in gene expression of
clustering analysis based on Euclidean distance was performed on D31 over D30, we analyzed the samples from 15 subjects of the RRR
the VARPC1 data, yielding seven SCs. Three SCs included a signifi- group collected at D29, 1 day after their second vaccination. We
cant number of genes and were subjected to BTM (23) enrichment then analyzed the log2-transformed fold changes in the expression
analysis (FDR q value of <0.05) using a hypergeometric test developed of the 300 genes identified in PC2 by comparing D29 to D28. We
in-house. performed a Pearson correlation–based hierarchical clustering
analysis to generate a heatmap and assigned the genes to the five
PCA and regression models BTM clusters.
For genes in the primary gene set, PCA was performed on a matrix
containing the individual unscaled log2-transformed fold-change
data for D1 over D0, D31 over D0, and D31 over D30, with subjects SUPPLEMENTARY MATERIALS
stm.sciencemag.org/cgi/content/full/12/569/eaay8618/DC1
in rows and genes in columns, using the prcomp function in R. Linear Fig. S1. Analysis pipelines.
regression models were used to assess the association between the Fig. S2. Early cytokine and hematology responses are associated with gene expression after
PC scores and the adaptive immune parameter per individual (Ai), dose 2.

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 11 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

Fig. S3. Temporal regulation of responses after the second dose of HBs/AS01B or HBs/AS03. 16. A. M. Didierlaurent, B. Laupèze, A. Di Pasquale, N. Hergli, C. Collignon, N. Garçon,
Table S1. Activated BTMs listed by cluster. Adjuvant system AS01: Helping to overcome the challenges of modern vaccines. Expert
Table S2. BTM enrichment in clusters of highly synchronized genes. Rev. Vaccines 16, 55–63 (2017).
Table S3. Canonical pathway enrichment analysis for genes not annotated in the BTMs. 17. P. Brodin, V. Jojic, T. Gao, S. Bhattacharya, C. J. L. Angel, D. Furman, S. Shen-Orr,
Table S4. Demography of the study cohort. C. L. Dekker, G. E. Swan, A. J. Butte, H. T. Maecker, M. M. Davis, Variation in the human
Data file S1. List of DEGs. immune system is largely driven by non-heritable influences. Cell 160, 37–47 (2015).
Data file S2. Summary of immune response parameters. 18. P. Brodin, M. M. Davis, Human immune system variation. Nat. Rev. Immunol. 17, 21–29
View/request a protocol for this paper from Bio-protocol. (2017).
19. G. A. Poland, I. G. Ovsyannikova, R. B. Kennedy, N. D. Lambert, J. L. Kirkland, A systems
biology approach to the effect of aging, immunosenescence and vaccine response.
REFERENCES AND NOTES Curr. Opin. Immunol. 29, 62–68 (2014).
1. A. Iwasaki, R. Medzhitov, Control of adaptive immunity by the innate immune system. 20. S. S. Shen-Orr, D. Furman, B. A. Kidd, F. Hadad, P. Lovelace, Y.-W. Huang, Y. Rosenberg-Hasson,
Nat. Immunol. 16, 343–353 (2015). S. Mackey, F. A. G. Grisar, Y. Pickman, H. T. Maecker, Y.-H. Chien, C. L. Dekker, J. C. Wu,
2. G. Del Giudice, R. Rappuoli, A. M. Didierlaurent, Correlates of adjuvanticity: A review A. J. Butte, M. M. Davis, Defective signaling in the JAK-STAT pathway tracks with chronic
on adjuvants in licensed vaccines. Semin. Immunol. 39, 14–21 (2018). inflammation and cardiovascular risk in aging humans. Cell Syst. 3, 374–384.e4 (2016).
3. O. Sobolev, E. Binda, S. O’Farrell, A. Lorenc, J. Pradines, Y. Huang, J. Duffner, R. Schulz, 21. G. Leroux-Roels, A. Marchant, J. Levy, P. Van Damme, T. F. Schwarz, Y. Horsmans, W. Jilg,
J. Cason, M. Zambon, M. H. Malim, M. Peakman, A. Cope, I. Capila, G. V. Kaundinya, P. G. Kremsner, E. Haelterman, F. Clément, J. J. Gabor, M. Esen, A. Hens, I. Carletti,
A. C. Hayday, Adjuvanted influenza-H1N1 vaccination reveals lymphoid signatures L. Fissette, F. T. Da Silva, W. Burny, M. Janssens, P. Moris, A. M. Didierlaurent,

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


of age-dependent early responses and of clinical adverse events. Nat. Immunol. 17, R. Van Der Most, N. Garçon, P. Van Belle, M. Van Mechelen, Impact of adjuvants on CD4+
204–213 (2016). T cell and B cell responses to a protein antigen vaccine: Results from a phase II,
4. D. Kazmin, H. I. Nakaya, E. K. Lee, M. J. Johnson, R. van der Most, R. A. van den Berg, randomized, multicenter trial. Clin. Immunol. 169, 16–27 (2016).
W. R. Ballou, E. Jongert, U. Wille-Reece, C. Ockenhouse, A. Aderem, D. E. Zak, J. Sadoff, 22. W. Burny, A. Callegaro, V. Bechtold, F. Clement, S. Delhaye, L. Fissette, M. Janssens,
J. Hendriks, J. Wrammert, R. Ahmed, B. Pulendran, Systems analysis of protective immune G. Leroux-Roels, A. Marchant, R. A. van den Berg, N. Garçon, R. van der Most,
responses to RTS,S malaria vaccination in humans. Proc. Natl. Acad. Sci. U.S.A. 114, A. M. Didierlaurent; ECR-002 Study Group, Different adjuvants induce common innate
2425–2430 (2017). pathways that are associated with enhanced adaptive responses against a model antigen
5. R. A. van den Berg, M. Coccia, W. R. Ballou, K. E. Kester, C. F. Ockenhouse, J. Vekemans, in humans. Front. Immunol. 8, 943 (2017).
E. Jongert, A. M. Didierlaurent, R. G. van der Most, Predicting RTS,S vaccine-mediated 23. S. Li, N. Rouphael, S. Duraisingham, S. Romero-Steiner, S. Presnell, C. Davis, D. S. Schmidt,
protection from transcriptomes in a malaria-challenge clinical trial. Front. Immunol. 8, 557 S. E. Johnson, A. Milton, G. Rajam, S. Kasturi, G. M. Carlone, C. Quinn, D. Chaussabel,
(2017). A. K. Palucka, M. J. Mulligan, R. Ahmed, D. S. Stephens, H. I. Nakaya, B. Pulendran,
6. R. A. van den Berg, L. De Mot, G. Leroux-Roels, V. Bechtold, F. Clement, M. Coccia, Molecular signatures of antibody responses derived from a systems biology study of five
E. Jongert, T. G. Evans, P. Gillard, R. G. van der Most, Adjuvant-associated peripheral blood human vaccines. Nat. Immunol. 15, 195–204 (2014).
mrna profiles and kinetics induced by the adjuvanted recombinant protein candidate 24. J. Menche, E. Guney, A. Sharma, P. J. Branigan, M. J. Loza, F. Baribaud, R. Dobrin,
tuberculosis vaccine M72/AS01 in bacillus calmette-guérin-vaccinated adults. A.-L. Barabási, Integrating personalized gene expression profiles into predictive
Front. Immunol. 9, 564 (2018). disease-associated gene pools. NPJ Syst. Biol. Appl. 3, 10 (2017).
7. J. Anderson, T. A. Olafsdottir, S. Kratochvil, P. F. McKay, M. Östensson, J. Persson, 25. C. F. Ockenhouse, J. Regules, D. Tosh, J. Cowden, A. Kathcart, J. Cummings, K. Paolino,
R. J. Shattock, A. M. Harandi, Molecular signatures of a TLR4 agonist-adjuvanted HIV-1 J. Moon, J. Komisar, E. Kamau, T. Oliver, A. Chhoeu, J. Murphy, K. Lyke, M. Laurens,
vaccine candidate in humans. Front. Immunol. 9, 301 (2018). A. Birkett, C. Lee, R. Weltzin, U. Wille-Reece, M. Sedegah, J. Hendriks, I. Versteege,
8. H. I. Nakaya, E. Clutterbuck, D. Kazmin, L. Wang, M. Cortese, S. E. Bosinger, N. B. Patel, M. G. Pau, J. Sadoff, Y. Vanloubbeeck, M. Lievens, D. Heerwegh, P. Moris, Y. G. Mendoza,
D. E. Zak, A. Aderem, T. Dong, G. D. Giudice, R. Rappuoli, V. Cerundolo, A. J. Pollard, E. Jongert, J. Cohen, G. Voss, W. R. Ballou, J. Vekemans, Ad35.CS.01-RTS,S/AS01
B. Pulendran, C.-A. Siegrist, Systems biology of immunity to MF59-adjuvanted versus heterologous prime boost vaccine efficacy against sporozoite challenge in healthy
nonadjuvanted trivalent seasonal influenza vaccines in early childhood. Proc. Natl. Acad. malaria-naive adults. PLOS ONE 10, e0131571 (2015).
Sci. U.S.A. 113, 1853–1858 (2016). 26. J. Ismaili, J. Rennesson, E. Aksoy, J. Vekemans, B. Vincart, Z. Amraoui, F. Van Laethem,
9. N. Garçon, A. Di Pasquale, From discovery to licensure, the adjuvant system story. Hum. M. Goldman, P. M. Dubois, Monophosphoryl lipid A activates both human dendritic cells
Vaccin. Immunother. 13, 19–33 (2017). and T cells. J. Immunol. 168, 926–932 (2002).
10. A. M. Didierlaurent, S. Morel, L. Lockman, S. L. Giannini, M. Bisteau, H. Carlsen, A. Kielland, 27. A. L. Desbien, S. J. Reed, H. R. Bailor, N. Dubois Cauwelaert, J. D. Laurance, M. T. Orr,
O. Vosters, N. Vanderheyde, F. Schiavetti, D. Larocque, M. Van Mechelen, N. Garçon, AS04, C. B. Fox, D. Carter, S. G. Reed, M. S. Duthie, Squalene emulsion potentiates the adjuvant
an aluminum salt- and TLR4 agonist-based adjuvant system, induces a transient localized activity of the TLR4 agonist, GLA, via inflammatory caspases, IL-18, and IFN-. Eur.
innate immune response leading to enhanced adaptive immunity. J. Immunol. 183, J. Immunol. 45, 407–417 (2015).
6186–6197 (2009). 28. T. A. Fehniger, M. A. Cooper, G. J. Nuovo, M. Cella, F. Facchetti, M. Colonna, M. A. Caligiuri,
11. A. M. Didierlaurent, C. Collignon, P. Bourguignon, S. Wouters, K. Fierens, M. Fochesato, CD56bright natural killer cells are present in human lymph nodes and are activated by
N. Dendouga, C. Langlet, B. Malissen, B. N. Lambrecht, N. Garçon, M. Van Mechelen, T cell-derived IL-2: A potential new link between adaptive and innate immunity. Blood
S. Morel, Enhancement of adaptive immunity by the human vaccine adjuvant AS01 101, 3052–3057 (2003).
depends on activated dendritic cells. J. Immunol. 193, 1920–1930 (2014). 29. A. Horowitz, J. C. R. Hafalla, E. King, J. Lusingu, D. Dekker, A. Leach, P. Moris, J. Cohen,
12. S. Morel, A. Didierlaurent, P. Bourguignon, S. Delhaye, B. Baras, V. Jacob, C. Planty, J. Vekemans, T. Villafana, P. H. Corran, P. Bejon, C. J. Drakeley, L. von Seidlein, E. M. Riley,
A. Elouahabi, P. Harvengt, H. Carlsen, A. Kielland, P. Chomez, N. Garçon, M. Van Mechelen, Antigen-specific IL-2 secretion correlates with NK cell responses after immunization
Adjuvant system AS03 containing -tocopherol modulates innate immune response of Tanzanian children with the RTS,S/AS01 malaria vaccine. J. Immunol. 188, 5054–5062
and leads to improved adaptive immunity. Vaccine 29, 2461–2473 (2011). (2012).
13. M. Coccia, C. Collignon, C. Hervé, A. Chalon, I. Welsby, S. Detienne, M. J. van Helden, 30. A. Penn-Nicholson, H. Geldenhuys, W. Burny, R. van der Most, C. L. Day, E. Jongert,
S. Dutta, C. J. Genito, N. C. Waters, K. Van Deun, A. K. Smilde, R. A. van den Berg, D. Franco, P. Moris, M. Hatherill, O. Ofori-Anyinam, W. Hanekom, V. S. Team, A. Bollaerts,
P. Bourguignon, S. Morel, N. Garçon, B. N. Lambrecht, S. Goriely, R. van der Most, M.-A. Demoitie, A. K. K. Luabeya, E. De Ruymaeker, M. Tameris, D. Lapierre, T. J. Scriba,
A. M. Didierlaurent, Cellular and molecular synergy in AS01-adjuvanted vaccines results Safety and immunogenicity of candidate vaccine M72/AS01E in adolescents in a TB
in an early IFN response promoting vaccine immunogenicity. NPJ Vaccines 2, 25 (2017). endemic setting. Vaccine 33, 4025–4034 (2015).
14. J. R. Francica, D. E. Zak, C. Linde, E. Siena, C. Johnson, M. Juraska, N. L. Yates, B. Gunn, 31. K. E. Kester, J. F. Cummings, O. Ofori-Anyinam, C. F. Ockenhouse, U. Krzych, P. Moris,
E. De Gregorio, B. J. Flynn, N. M. Valiante, P. Malyala, S. W. Barnett, P. Sarkar, M. Singh, R. Schwenk, R. A. Nielsen, Z. Debebe, E. Pinelis, L. Juompan, J. Williams, M. Dowler,
S. Jain, M. Ackerman, M. Alam, G. Ferrari, A. Salazar, G. D. Tomaras, D. T. O’Hagan, V. A. Stewart, R. A. Wirtz, M. C. Dubois, M. Lievens, J. Cohen, W. R. Ballou, D. G. Heppner Jr.,
A. Aderem, G. Alter, R. A. Seder, Innate transcriptional effects by adjuvants Randomized, double-blind, phase 2a trial of falciparum malaria vaccines RTS,S/AS01B
on the magnitude, quality, and durability of HIV envelope responses in NHPs. Blood Adv. and RTS,S/AS02A in malaria-naive adults: Safety, efficacy, and immunologic associates
1, 2329–2342 (2017). of protection. J. Infect. Dis. 200, 337–346 (2009).
15. F. Liang, G. Lindgren, K. J. Sandgren, E. A. Thompson, J. R. Francica, A. Seubert, 32. L. M. Howard, K. L. Hoek, J. B. Goll, P. Samir, A. Galassie, T. M. Allos, X. Niu, L. E. Gordy,
E. De Gregorio, S. Barnett, D. T. O’Hagan, N. J. Sullivan, R. A. Koup, R. A. Seder, K. Loré, C. B. Creech, N. Prasad, T. L. Jensen, H. Hill, S. E. Levy, S. Joyce, A. J. Link, K. M. Edwards,
Vaccine priming is restricted to draining lymph nodes and controlled by adjuvant- Cell-based systems biology analysis of human AS03-adjuvanted H5N1 avian influenza
mediated antigen uptake. Sci. Transl. Med. 9, eaal2094 (2017). vaccine responses: A phase I randomized controlled trial. PLOS ONE 12, e0167488 (2017).

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 12 of 13
SCIENCE TRANSLATIONAL MEDICINE | RESEARCH RESOURCE

33. C. Givord, I. Welsby, S. Detienne, S. Thomas, A. Assabban, V. L. Silva, C. Molle, R. Gineste, 46. G. K. Smyth, J. Michaud, H. S. Scott, Use of within-array replicate spots for assessing
M. Vermeersch, D. Perez-Morga, O. Leo, C. Collignon, A. M. Didierlaurent, S. Goriely, differential expression in microarray experiments. Bioinformatics 21, 2067–2075 (2005).
Activation of the endoplasmic reticulum stress sensor IRE1 by the vaccine adjuvant 47. J. Josse, F. Husson, missMDA: A package for handling missing values in multivariate data
AS03 contributes to its immunostimulatory properties. NPJ Vaccines 3, 20 (2018). analysis. J. Stat. Softw. 70, 1–31 (2016).
34. F. Mosca, E. Tritto, A. Muzzi, E. Monaci, F. Bagnoli, C. Iavarone, D. O'Hagan, R. Rappuoli, 48. M. Larsen, D. Sauce, L. Arnaud, S. Fastenackels, V. Appay, G. Gorochov, Evaluating
E. De Gregorio, Molecular and cellular signatures of human vaccine adjuvants. Proc. Natl. cellular polyfunctionality with a novel polyfunctionality index. PLOS ONE 7, e42403
Acad. Sci. U.S.A. 105, 10501–10506 (2008). (2012).
35. T. A. Olafsdottir, M. Lindqvist, I. Nookaew, P. Andersen, J. Maertzdorf, J. Persson,
D. Christensen, Y. Zhang, J. Anderson, S. Khoomrung, P. Sen, E. M. Agger, R. Coler, Acknowledgments: We are grateful to the study participants and staff members of the
D. Carter, A. Meinke, R. Rappuoli, S. H. E. Kaufmann, S. G. Reed, A. M. Harandi, different study sites for contribution to the study. We thank N. Garçon, G. Leroux-Roels, and
Comparative systems analyses reveal molecular signatures of clinically tested vaccine A. Marchant for expert review of the data, helpful discussions, and contributions to this study.
adjuvants. Sci. Rep. 6, 39097 (2016). We also thank E. Oe (GSK) for providing scientific writing services in the manuscript’s
36. E. Bartholomeus, N. De Neuter, P. Meysman, A. Suls, N. Keersmaekers, G. Elias, H. Jansens, development and G. Drevon (GSK) and J. Ghesquière (Business & Decision Life Sciences) for
N. Hens, E. Smits, V. Van Tendeloo, P. Beutels, P. Van Damme, B. Ogunjimi, K. Laukens, editorial support and manuscript coordination. Editorial services were provided by
G. Mortier, Transcriptome profiling in blood before and after hepatitis B vaccination N. R. Gough (BioSerendipity). The ECR02 study group includes the following authors:
shows significant differences in gene expression between responders and Viviane Bechtold, Wivine Burny, Isabelle Carletti, Frédéric Clément, Arnaud M. Didierlaurent,
non-responders. Vaccine 36, 6282–6289 (2018). Meral Esen, Laurence Fissette, Julian Gabor, Nathalie Garçon, Edwige Haelterman, Caroline Hervé,
37. H. I. Nakaya, T. Hagan, S. S. Duraisingham, E. K. Lee, M. Kwissa, N. Rouphael, D. Frasca, Yves Horsmans, Michel Janssens, Peter Kremsner, Geert Leroux-Roels, Arnaud Marchant,

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


M. Gersten, A. K. Mehta, R. Gaujoux, G.-M. Li, S. Gupta, R. Ahmed, M. J. Mulligan, Philippe Moris, Tino F. Schwarz, Fernanda Tavares da Silva, Pierre Van Damme,
S. Shen-Orr, B. B. Blomberg, S. Subramaniam, B. Pulendran, Systems analysis of immunity Robert A. van den Berg, Robbert G. van der Most, and Dirk Zuchner. The Business & Decision
to influenza vaccination across multiple years and in diverse populations reveals shared Life Sciences platform provided editorial assistance and manuscript coordination, on behalf of
molecular signatures. Immunity 43, 1186–1198 (2015). GSK. Funding: GlaxoSmithKline Biologicals SA funded the study, was involved in all stages of
38. K. L. Bucasas, L. M. Franco, C. A. Shaw, M. S. Bray, J. M. Wells, D. Niño, N. Arden, J. M. Quarles, the original studies conduct and analysis (ClinicalTrials.gov Identifiers: NCT00805389), and
R. B. Couch, J. W. Belmont, Early patterns of gene expression correlate with the humoral funded the manuscript’s development. This work was supported by the Bill & Melinda Gates
immune response to influenza vaccination in humans. J. Infect. Dis. 203, 921–929 (2011). Foundation (grant number OPP1120977 to R.G.v.d.M. and R.A.v.d.B.). Author contributions:
39. C. Rydyznski, K. A. Daniels, E. P. Karmele, T. R. Brooks, S. E. Mahl, M. T. Moran, C. Li, L.D.M., V. Bechtold, F.U.-M., E.S., R.A.v.d.B., A.M.D., W.B., and R.G.v.d.M. contributed substantially
R. Sutiwisesak, R. M. Welsh, S. N. Waggoner, Generation of cellular immune memory to the conception and design of the studies. L.D.M., V. Bechtold, V. Bol, A.C., M.C., A.E., D.H.,
and B-cell immunity is impaired by natural killer cells. Nat. Commun. 6, 6375 (2015). F.U.-M., E.S., A.S., and R.A.v.d.B. participated in the collection and analysis of the data. L.D.M.,
40. F. Liang, K. Loré, Local innate immune responses in the vaccine adjuvant-injected muscle. V. Bechtold, A.M.D., W.B., and R.G.v.d.M. contributed to the interpretation of the results. All
Clin. Transl. Immunol. 5, e74 (2016). authors had full access to the data and were involved in critical revision of the manuscript for
41. A. L. Cunningham, T. C. Heineman, H. Lal, O. Godeaux, R. Chlibek, S.-J. Hwang, important intellectual content. The corresponding author was responsible for manuscript
J. E. McElhaney, T. Vesikari, C. Andrews, W. S. Choi, M. Esen, H. Ikematsu, M. K. Choma, submission. Competing interests: V. Bechtold, V. Bol, A.C., M.C., A.E., F.U.-M., E.S., R.A.v.d.B.,
K. Pauksens, S. Ravault, B. Salaun, T. F. Schwarz, J. Smetana, C. V. Abeele, P. Van den Steen, A.M.D., W.B., and R.G.v.d.M. are employees of the GSK group of companies. V. Bechtold, A.C.,
I. Vastiau, L. Y. Weckx, M. J. Levin; ZOE-50/70 Study Group, Immune responses F.U.-M., E.S., R.A.v.d.B., A.M.D., W.B., and R.G.v.d.M. hold shares in the GSK group of companies
to a recombinant glycoprotein E herpes zoster vaccine in adults aged 50 years or older. as part of their employee remuneration. A.M.D. and R.G.v.d.M. own patents related to AS01
J. Infect. Dis. 217, 1750–1760 (2018). (Methods for inducing an immune response, US10624961B2). L.D.M. was an employee of the
42. O. Van Der Meeren, M. Hatherill, V. Nduba, R. J. Wilkinson, M. Muyoyeta, E. Van Brakel, GSK group of companies at the time of the study. D.H. received a grant from GSK to conduct
H. M. Ayles, G. Henostroza, F. Thienemann, T. J. Scriba, A. Diacon, G. L. Blatner, the study. A.S. reports no conflict of interest. Fendrix and Engerix-B are trademarks owned by
M.-A. Demoitié, M. Tameris, M. Malahleha, J. C. Innes, E. Hellström, N. Martinson, T. Singh, or licensed to the GSK group of companies. Data and materials availability: Anonymized
E. J. Akite, A. K. Azam, A. Bollaerts, A. M. Ginsberg, T. G. Evans, P. Gillard, D. R. Tait, Phase individual participant data and study documents can be requested for further research at
2b controlled trial of M72/AS01E vaccine to prevent tuberculosis. N. Engl. J. Med. 379, www.clinicalstudydatarequest.com. Transcriptomic data are available from the NCBI GEO
1621–1634 (2018). (accession no. GSE116975).
43. S. Khurana, E. M. Coyle, J. Manischewitz, L. R. King, J. Gao, R. N. Germain,
P. L. Schwartzberg, J. S. Tsang, H. Golding; the CHI Consortium, AS03-adjuvanted H5N1 Submitted 24 July 2019
vaccine promotes antibody diversity and affinity maturation, NAI titers, cross-clade H5N1 Accepted 23 October 2020
neutralization, but not H1N1 cross-subtype neutralization. npj Vaccines 3, 40 (2018). Published 11 November 2020
44. T. Thanh Le, Z. Andreadakis, A. Kumar, R. Gómez Román, S. Tollefsen, M. Saville, 10.1126/scitranslmed.aay8618
S. Mayhew, The COVID-19 vaccine development landscape. Nat. Rev. Drug Discov. 19,
305–306 (2020). Citation: L. De Mot, V. Bechtold, V. Bol, A. Callegaro, M. Coccia, A. Essaghir, D. Hasdemir,
45. B. Phipson, S. Lee, I. J. Majewski, W. S. Alexander, G. K. Smyth, Robust hyperparameter F. Ulloa-Montoya, E. Siena, A. Smilde, R. A. van den Berg, A. M. Didierlaurent, W. Burny, R. G. van der Most,
estimation protects against hypervariable genes and improves power to detect Transcriptional profiles of adjuvanted hepatitis B vaccines display variable interindividual
differential expression. Ann. Appl. Stat. 10, 946–963 (2016). homogeneity but a shared core signature. Sci. Transl. Med. 12, eaay8618 (2020).

De Mot et al., Sci. Transl. Med. 12, eaay8618 (2020) 11 November 2020 13 of 13
Transcriptional profiles of adjuvanted hepatitis B vaccines display variable
interindividual homogeneity but a shared core signature
Laurane De Mot, Viviane Bechtold, Vanesa Bol, Andrea Callegaro, Margherita Coccia, Ahmed Essaghir, Dicle
Hasdemir, Fernando Ulloa-Montoya, Emilio Siena, Age Smilde, Robert A. van den Berg, Arnaud M. Didierlaurent,
Wivine Burny and Robbert G. van der Most

Sci Transl Med 12, eaay8618.

Downloaded from http://stm.sciencemag.org/ at Cambridge University on November 21, 2020


DOI: 10.1126/scitranslmed.aay8618

Patterns of adjuvanticity
Understanding how adjuvants improve vaccine-induced immune responses is important for the
development of successful vaccines. De Mot et al. performed transcriptome analysis on whole blood taken from
the participants of a previously reported clinical study that trialed different adjuvants alongside hepatitis B vaccine
antigen. Differential expression of a core gene signature related to innate immune response and natural killer cells
correlated with the antibody response observed in vaccine recipients after two doses regardless of the adjuvant
administered, suggesting that these responses are important for vaccine immunogenicity.

ARTICLE TOOLS http://stm.sciencemag.org/content/12/569/eaay8618

SUPPLEMENTARY http://stm.sciencemag.org/content/suppl/2020/11/09/12.569.eaay8618.DC1
MATERIALS

RELATED http://stm.sciencemag.org/content/scitransmed/12/550/eabc3539.full
CONTENT
http://stm.sciencemag.org/content/scitransmed/12/553/eabb4757.full
http://stm.sciencemag.org/content/scitransmed/12/543/eaay8924.full
http://stm.sciencemag.org/content/scitransmed/11/507/eaaw4236.full
REFERENCES This article cites 48 articles, 10 of which you can access for free
http://stm.sciencemag.org/content/12/569/eaay8618#BIBL

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Use of this article is subject to the Terms of Service

Science Translational Medicine (ISSN 1946-6242) is published by the American Association for the Advancement of
Science, 1200 New York Avenue NW, Washington, DC 20005. The title Science Translational Medicine is a
registered trademark of AAAS.
Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement
of Science. No claim to original U.S. Government Works

You might also like