PreprintPDF Available

Comparative Molecular, Innate, and Adaptive Impacts of Chemically Diverse STING Agonists

Authors:
Preprints and early-stage research may not have been peer reviewed yet.

Abstract

Pharmacologic activation of the innate immune response is being actively being pursued for numerous clinical purposes including enhancement of vaccine potency and potentiation of anti-cancer immunotherapy. Pattern recognition receptors (PRRs) represent especially useful targets for these efforts as their engagement by agonists can trigger signaling pathways that associate with phenotypes desirable for specific immune outcomes. Stimulator of interferon genes (STING) is an ER-resident PRR reactive to cyclic dinucleotides such as those synthesized endogenously in response to cytosolic dsDNA. STING activation leads to transient generation of type I interferon (IFN-I) and proinflammatory responses that augment immunologically relevant effects including antiviral responses, antigen presentation, immune cell trafficking, and immunogenic cell death. In recent years engineered cyclic dinucleotides and small molecules have been discovered that induce STING and safely confer clinically useful outcomes in animal models such as adjuvanticity of anti-microbial vaccines and tumor clearance. Unfortunately, clinical trials examining the efficacy of STING agonists have thus far failed to satisfactorily recapitulate these positive outcomes and this has prevented their translational advancement. A likely relevant yet perplexingly under investigated aspect of pharmacologic STING activation is the diversity of molecular and immune responses that associate with chemical properties of the agonist. Based on this, a comparative survey of these was undertaken using unrelated STING-activating molecules to characterize the molecular, innate, cellular, and immune outcomes they elicit. This was done to inform and direct future studies aimed at designing and selecting agonists appropriate for desired clinical goals. This revealed demonstrable differences between the agonists in potency, transcriptomes, cytokine secretion profiles, immune cell trafficking, and antigen-directed humoral and cell mediated immune responses. As such, this work illustrates that phenotypes deriving from activation of a protein target can be linked to chemical properties of the engaging agonist and thus heightened scrutiny is necessary when selecting molecules to generate specific in vivo effects.
1
2
3
4
Comparative Molecular, Innate, and Adaptive Impacts of Chemically Diverse STING Agonists 5
6
7
8
9
10
11
Nobuyo Mizuno1, Dylan Boehm1, Kevin Jimenez-Perez1, Jinu Abraham1, Laura Springgay1, Ian Rose1, Victor 12
R. DeFilippis1*
13
14
15
16
17
1Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States 18
of America. 19
20
*To whom correspondence should be addressed: defilipp@ohsu.edu21
22
Click here to access/download;Title Page, unmarked "clean"
version;Title_Page.docx
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
61
62
63
64
65
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
ABSTRACT
1
Pharmacologic activation of the innate immune response is being actively being pursued for numerous clinical
2
purposes including enhancement of vaccine potency and potentiation of anti-cancer immunotherapy. Pattern
3
recognition receptors (PRRs) represent especially useful targets for these efforts as their engagement by
4
agonists can trigger signaling pathways that associate with phenotypes desirable for specific immune outcomes.
5
Stimulator of interferon genes (STING) is an ER-resident PRR reactive to cyclic dinucleotides such as those
6
synthesized endogenously in response to cytosolic dsDNA. STING activation leads to transient generation of
7
type I interferon (IFN-I) and proinflammatory responses that augment immunologically relevant effects including
8
antiviral responses, antigen presentation, immune cell trafficking, and immunogenic cell death. In recent years
9
engineered cyclic dinucleotides and small molecules have been discovered that induce STING and safely confer
10
clinically useful outcomes in animal models such as adjuvanticity of anti-microbial vaccines and tumor clearance.
11
Unfortunately, clinical trials examining the efficacy of STING agonists have thus far failed to satisfactorily
12
recapitulate these positive outcomes and this has prevented their translational advancement. A likely relevant
13
yet perplexingly under investigated aspect of pharmacologic STING activation is the diversity of molecular and
14
immune responses that associate with chemical properties of the agonist. Based on this, a comparative survey
15
of these was undertaken using unrelated STING-activating molecules to characterize the molecular, innate,
16
cellular, and immune outcomes they elicit. This was done to inform and direct future studies aimed at designing
17
and selecting agonists appropriate for desired clinical goals. This revealed demonstrable differences between
18
the agonists in potency, transcriptomes, cytokine secretion profiles, immune cell trafficking, and antigen-directed
19
humoral and cell mediated immune responses. As such, this work illustrates that phenotypes deriving from
20
activation of a protein target can be linked to chemical properties of the engaging agonist and thus heightened
21
scrutiny is necessary when selecting molecules to generate specific in vivo effects.
22
23
Keywords: STING, adjuvant, cytokines, vaccines, interferon.
24
25
Click here to access/download;Abstract;Abstract.docx
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
61
62
63
64
65
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
2
ABSTRACT
23
Pharmacologic activation of the innate immune response is being actively being pursued for numerous clinical
24
purposes including enhancement of vaccine potency and potentiation of anti-cancer immunotherapy. Pattern
25
recognition receptors (PRRs) represent especially useful targets for these efforts as their engagement by
26
agonists can trigger signaling pathways that associate with phenotypes desirable for specific immune outcomes.
27
Stimulator of interferon genes (STING) is an ER-resident PRR reactive to cyclic dinucleotides such as those
28
synthesized endogenously in response to cytosolic dsDNA. STING activation leads to transient generation of
29
type I interferon (IFN-I) and proinflammatory responses that augment immunologically relevant effects including
30
antiviral responses, antigen presentation, immune cell trafficking, and immunogenic cell death. In recent years
31
engineered cyclic dinucleotides and small molecules have been discovered that induce STING and safely confer
32
clinically useful outcomes in animal models such as adjuvanticity of anti-microbial vaccines and tumor clearance.
33
Unfortunately, clinical trials examining the efficacy of STING agonists have thus far failed to satisfactorily
34
recapitulate these positive outcomes and this has prevented their translational advancement. A likely relevant
35
yet perplexingly under investigated aspect of pharmacologic STING activation is the diversity of molecular and
36
immune responses that associate with chemical properties of the agonist. Based on this, a comparative survey
37
of these was undertaken using unrelated STING-activating molecules to characterize the molecular, innate,
38
cellular, and immune outcomes they elicit. This was done to inform and direct future studies aimed at designing
39
and selecting agonists appropriate for desired clinical goals. This revealed demonstrable differences between
40
the agonists in potency, transcriptomes, cytokine secretion profiles, immune cell trafficking, and antigen-directed
41
humoral and cell mediated immune responses. As such, this work illustrates that phenotypes deriving from
42
activation of a protein target can be linked to chemical properties of the engaging agonist and thus heightened
43
scrutiny is necessary when selecting molecules to generate specific in vivo effects.
44
45
Keywords: STING, adjuvant, cytokines, vaccines, interferon.
46
47
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
3
INTRODUCTION
48
Subunit vaccine antigens when used alone do not typically exhibit levels of immunogenicity that are sufficient for
49
the establishment of protective immunity. As such, they require coadministration of factors such as adjuvants to
50
stimulate the innate immune system in a manner resembling that which occurs during exposure to molecular
51
signals of proliferative microbial danger [1]. Unfortunately, only a small number of adjuvants are approved for
52
clinical use and effective protection requires pathogen-specific immune polarization such that one adjuvant is
53
unlikely to achieve all objectives [2]. Moreover, the precise mechanisms of action of clinical and experimental
54
vaccine adjuvants are often poorly characterized [3]. The need is therefore great for pursuit and characterization
55
of new adjuvant classes, especially due to the likelihood of pathogen emergence and the number of established
56
infections for which no or suboptimal vaccines exist. A promising investigative approach to adjuvant discovery is
57
identification of pharmacologic activators of pattern recognition receptors (PRRs) [4]. Engagement of PRRs by
58
pathogen- or danger-associated molecular patterns initiate intracellular signaling processes that culminate in
59
innate immune activation. This includes proinflammatory cytokine secretion and activation of antigen presenting
60
cells (APC) that facilitate and direct antigen-directed adaptive immune responses.
61
62
Stimulator of interferon genes (STING) is an ER-resident PRR reactive to cyclic purine dinucleotides (CDNs)
63
synthesized by bacterial and eukaryotic nucleotidyl cyclases. Most prominently, cyclic GMP-AMP (cGAMP)
64
synthase (cGAS) is a vertebrate enzyme activated following contact with cytosolic DNA to synthesize 2’3’ cGAMP
65
from ATP and GTP. Activated STING leads to phosphorylation of TANK binding kinase 1 (TBK1) which in turn
66
phosphorylates transcription factors such as interferon regulatory factor 3 (IRF3) and nuclear factor B (NF-B).
67
These leads to expression of type I interferons (IFN-I) as well as other pro-inflammatory cytokines and
68
costimulatory factors. In addition, STING is expressed across a wide range of cell types and induces
69
transcription-independent processes such as autophagy, inflammasome induction, and lysosomal biogenesis
70
often in ways that are cell- and agonist-specific [58] . These collectively contribute to the innate immune
71
phenotypes conferred by activated STING and facilitate the establishment of adaptive immune responses
72
through localized recruitment and maturation of APCs, antigen loading and processing, and antigen presentation
73
in the draining lymph node (dLN). STING is therefore an increasingly attractive cellular target for development
74
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
4
of new adjuvants and its stimulation as such has shown to enhance efficacy of vaccines against diverse pathogen
75
types.
76
77
Multiple CDN classes can activate STING and enhance vaccine-mediated immune responses in animal models.
78
Furthermore, synthetic small molecules have also been identified that induce STING and adjuvant functions
79
have been described for many of these. Numerous efforts are currently underway to thus discover and
80
characterize novel STING-activating molecules for similar use. However, liabilities associated with certain
81
chemical classes can affect their in vivo suitability, potency, and performance. For instance, CDNs are
82
susceptible to degradation by phosphodiesterases and also cross cell membranes poorly due to their size and
83
hydrophilicity. More importantly, the profile of STING-mediated molecular processes is influenced by the
84
chemical structure of the ligand. This may be due to a number of causes including engagement of off-target
85
proteins, pharmacokinetics, region of binding, or molecule-specific conformational changes in STING that affect
86
its activity. As such, it is likely insufficient to simply target STING using any activating ligand since the consequent
87
immune phenotypes could vary substantially and confer outcomes inconsistent with a vaccine’s purpose.
88
89
This study aims to undertake in vitro and in vivo characterization of three well described but chemically dissimilar
90
STING agonists. Our purpose is to perform direct comparisons of molecular, innate, and adaptive immune
91
responses that they induce to identify effects that are both shared and divergent between them. We hope these
92
will inform future studies that pursue STING inducers for specifically defined clinical outcomes. This work also
93
highlights multi-level phenotype patterns correlating with STING activation that were unexpected in our models.
94
95
96
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
5
RESULTS
97
Dose-dependent activity of STING agonists in vitro
98
To compare molecular, innate, and adaptive immune phenotypes elicited by chemically dissimilar STING ligands
99
we used cyclic di-AMP analog ML-RR-S2 CDA (CDA) [9], the amidobenzimidazole diABZI [10], and 5,6-
100
dimethylxantheonone-4-acetic acid (DMXAA) [9,11,12]. We first characterized the potency, patterns of innate
101
activation, and cytotoxicity to allow identification of suitable doses for subsequent in vivo use. For this we used
102
RAW264.7 and J774 macrophage-like murine cell lines engineered to express luciferase in response to IFN-I-
103
dependent signaling [13,14]. These were exposed in duplicate to a range of concentrations of the molecules and
104
luciferase signal measured after 24 h. CDA was transfected since it is not easily cell permeable due to its size
105
and hydrophilicity. In parallel, cytotoxicity was also measured using the ATP-based Cell Titer Glo reagent. As
106
shown in Figures 1A and 1B, differences were observed between the cell types primarily at the highest
107
concentrations of each molecule with RAW264.7 displaying overall lower viability than J774 cells. Moreover,
108
J774 cells also showed greater sensitivity to the compounds as indicated by higher overall fold changes in
109
luciferase induction for all agonists. Additionally, CDA elicited the highest peak induction levels in both cell types.
110
J774 cells co-express secreted alkaline phosphatase (SEAP) in response to NF-kB activation and we therefore
111
also examined this response. As shown in Figure 1C, detectable SEAP was observed at the highest dose of
112
DMXAA and at multiple doses of CDA but was not detected at any diABZI dose. Importantly, these results
113
indicate that levels of cytotoxicity at doses eliciting peak innate induction in both cell types were not unsuitably
114
low indicating that these were appropriate for use in additional studies. Moreover, NF-kB activation was only
115
significantly detected in response to CDA, highlighting a key difference in phenotypic response to the agonists.
116
117
Activation of STING is also known to stimulate the NLRP3 inflammasome, which can greatly affect the
118
proinflammatory tissue environment through secretion of IL-1 and IL-18 [5,15]. However, the degrees to which
119
this is induced by different agonist chemical classes has not been examined. We therefore used the J774
120
monocyte cell line as has been previously described for NLRP3 inflammasome studies to assess the response
121
to these agonists [16,17]. In duplicate cells were primed for 5 h with LPS and exposed overnight to a dosing
122
range of diABZI, CDA, or DMXAA. Secretion of IL-1 was then measured using bead-based Luminex assay. As
123
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
6
shown in Figure 1D, all three agonists stimulated secretion of IL-1 although with divergent induction patterns
124
that were maximal near 0.032 µM. However, while diABZI secretion declined linearly with higher agonist doses,
125
DMXAA and CDA generated dissimilarly sigmoidal decreases with higher dosage. Based on these results we
126
conclude that the agonists collectively stimulate qualitatively different patterns of intracellular innate induction as
127
indicated by degree of dose-dependence, cytokine (IFN-I and IL-1) secretion, and NF-B activation.
128
129
STING agonist-mediated dendritic cell maturation
130
Adjuvant-enhanced adaptive immune processes require the local activation of dendritic cells (DC) to enable
131
antigen uptake and presentation on MHC-II to CD4+ helper cells in the draining lymph node (dLN) [18]. We
132
therefore compared the ability of STING ligands to induce the expression of MHC-II and costimulatory proteins
133
CD86, CD80, and CD40. For this we used nontoxic concentrations of each molecule near those that displayed
134
the highest reporter signal in in vitro assays (Figure 1). Control stimuli included LPS and Alum as respective
135
inducers of DC activation and subunit vaccine response enhancement. Immature bone marrow derived DC
136
(BMDC) from C57Bl/6 mice were treated overnight and flow cytometry used to measure surface expression of
137
the markers. As shown in Figure 2, all STING ligands as well as LPS led to significant upregulation of the four
138
markers relative to DMSO-treated cells whereas no such induction was observed for Alum. In addition, DMXAA
139
appears capable of eliciting significantly higher levels of expression of these proteins than the other STING
140
ligands and (with the exception of MHC-II) LPS. As such, these STING agonists are highly but differentially
141
effective at inducing maturation of DC in an ex vivo system, a process necessary for establishing adaptive
142
immune responses.
143
144
In vivo cytokine induction by STING agonists
145
Adjuvant mechanisms associate with stimulation of innate processes immediately following in vivo
146
administration. While administered locally, this often manifests as secretion of proinflammatory cytokines that
147
are detectable in peripheral blood [19,20]. We therefore compared this indicator of systemic innate immune
148
induction by the three molecules using secretion of peripheral cytokines as a readout. Using doses that are
149
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
7
consistent with what was found to induce near maximal IFN-I levels, we injected each compound intramuscularly
150
(IM) into C57Bl/6 mice to simulate a typical vaccination route using PBS as a negative control injection. Mice
151
were euthanized at 24 h and peripheral blood collected. Levels of cyto/chemokines were then measured in serum
152
using Luminex multiplex bead-based assay. As shown in Figure 3, three different cytokine induction patterns
153
were detected for the stimuli. Secretion of cytokines such as CCL7 (MCP-3) and IL-1 was significantly and
154
similarly induced by all three treatments relative to control mice. Cytokines such as CXCL10 and IL-18 were
155
significantly stimulated by CDA and DMXAA but not diABZI. Finally, DMXAA alone induced significant secretion
156
of CCL2, CCL3, CCL4, CCL5, CCL11, CXCL1, CXCL2, IL-6, IL-10, IL-12p70, IL-17A, IL-22, IL-27, and TNF.
157
These results suggest that while some innate pathways such as those leading to secretion of CCL7 and IL-
158
1are stimulated comparably in response to all stimuli, others can be differentially activated in an agonist-
159
specific manner. Moreover, the response to DMXAA appears unusual among the agonists in that it is uniquely
160
capable of inducing secretion of a subset of cytokines. This is underscored by the fact that no secretion of CCL3,
161
IL-10, IL-17A, IL-22, and IL-27 above the limit of detection was observed in response to other agonists for yet
162
they were significantly stimulated by DMXAA.
163
164
In vivo transcriptional impact of STING agonist administration
165
The rapid innate processes conventionally induced by adjuvants are most highly localized to the site of
166
administration and dLN where adaptive immune processes commence. We next examined this at the
167
transcriptional level by injecting mice with diABZI, CDA, DMXAA, or PBS as described above. Total RNA was
168
isolated from dLN harvested at 5 h, 24 h, or 72 h and semi-quantitative RT-PCR (qPCR) used to measure
169
adjuvant-dependent upregulation of mRNAs encoding conventional IFN-stimulated and pro-inflammatory
170
proteins. As shown in Figure 4A, transcriptional induction peaked for nearly all genes at 5 h and declined in later
171
time points although at different rates between agonists. Importantly, levels of all examined mRNAs displayed
172
significant differences between at least two stimuli at multiple time points with the most differences observed
173
between DMXAA and CDA and the fewest between DMXAA and diABZI. Moreover, DMXAA stimulation led to
174
the highest mean induction of all genes.
175
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
8
Based on these results we decided to obtain a global picture of changes occurring in whole dLN transcriptomes
176
in response to IM injection of the three agonists. For this we undertook a high content mRNA hybridization array
177
experiment encompassing >21,000 mouse transcripts. Given the degree of temporal diminishment in
178
transcriptional induction (Figure 4A), we examined only 5 and 24 h time points. The number of mRNAs showing
179
significant up- or downregulation in dLN harvested following agonist injection were determined by comparing
180
absolute signal levels of mRNA-specific probe binding between STING agonist and PBS injected mice to obtain
181
transcript fold change (Supplemental Table 1). As indicated in Figure 4B, DMXAA triggers a stronger
182
transcriptional response relative to the other molecules as indicated by the number of significantly induced and
183
repressed mRNAs at both time points. This is also consistent with what is observed by qPCR of targeted genes
184
(Figure 4A). Sample clusters obtained using principal component analysis (PCA) further indicated that the
185
transcriptomes generated in response to DMXAA at both time points were substantially more divergent than for
186
the other two stimuli (Figure 4C). Consistent with this, at 5 h, stimulation with CDA and diABZI led to upregulation
187
of fewer shared transcripts (779) than were uniquely induced by DMXAA (1642). However, at 24 h diABZI
188
activated considerably more transcripts than did CDA, a larger portion of which were uniquely shared with
189
DMXAA. Over both time points CDA displayed the response of the smallest magnitude and the fewest uniquely
190
induced genes (Figure 4D).
191
192
We next used pathway analysis to complement quantitative methods and obtain a context-oriented
193
understanding of the biological activities potentially affected by STING agonist administration (Supplemental
194
Table 2). We observed within these results pathways similarly activated by all three agonists as shown in Figure
195
4E. Intriguingly, pathways were also detected that were only induced by DMXAA at both 5 h and 24 h (Figure
196
4F). This includes RAN (Ras-related nuclear protein) signaling and Folate transformation pathways, which were
197
predicted to be uniquely induced after both 5 h and 24 h DMXAA treatment. RAN signaling is known to confer
198
inhibitory effects on T cell function largely due to impaired nuclear accumulation of AP-1 transcription factors (c-
199
Jun, c-Fos) [21]. Moreover, folate is known to be important for proliferation of CD8+ T cells and may impact
200
CD4+/CD8+ T cell ratio [22]. These results demonstrate that chemically dissimilar agonists are capable of
201
activating transcription that functionally associates with STING stimulation as well as unique pathways linked
202
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
9
with immunologically impactful processes. As such, agonists may confer differential effects on establishment of
203
T cell mediated immune responses.
204
205
Adjuvant-associated changes in cell activation and recruitment to dLN
206
Innate immune activating adjuvant administration leads to kinetic, activation, and migratory changes in cellular
207
populations at the site of injection and in the dLN [2326]. This is especially true of APCs such as DC that are
208
essential for initiating antigen-specific T and B cell activation and subsequent differentiation. Importantly, STING
209
activation has been shown to inhibit growth and even induce death of T, B, and monocytic cells [2730]. How
210
this occurs and its impacts in the context of transient pharmacologic STING activation during vaccination is not
211
known. Given the strong transcriptional response observed in dLN, we used flow cytometry to examine changes
212
in cell populations here 24 h after intramuscular administration of the antigen ovalbumin (OVA) in the presence
213
or absence of CDA, diABZI, and DMXAA using duplicate experiments (Supplemental Figure 1). Alum was used
214
as a STING- and IRF3-independent control treatment. As shown in Figure 5A, total dLN cell numbers were not
215
significantly altered in response to administration of any stimulus. However, CDA and diABZI led to significant
216
increases and DMXAA significant decreases in B (CD3-, CD19+) cell composition of dLN while Alum had no
217
effect (Figure 5B). DMXAA also led to a significant decrease in the percentage of T (CD3+, CD19-) cells in the
218
dLN while no other stimulus had an effect (Figure 5C). Interestingly, when we examined DC populations, DMXAA
219
led to significant increases in frequencies of cDC1 (CD3/B220/CD11b-, CD11c/CD8+), cDC2 (CD3/B220-,
220
CD11c/CD11b+), and pDC (CD3/CD11b/CD8-, B220/CD11c+) whereas no such changes were observed for the other
221
agonists as shown in Figure 5C-E. In addition, DMXAA was also uniquely associated with increases in dLN-
222
localized neutrophils (CD3/B220/F4/80/CD11c-, CD11b+/Ly6G+; Figure 5D) and macrophages
223
(CD3/B220/CD11c-, F4/80/CD11b+; Figure 5D). To understand the mechanistic basis of these unusual DMXAA-
224
mediated observations we asked whether IFN-I responses are required. For this we used mice lacking the IFN
225
receptor (IFNAR-/-). WT and IFNAR-/- mice were treated intramuscularly with OVA alone or admixed with DMXAA.
226
As shown in Figure 5C, functional IFN-I signaling was required for increased DMXAA-dependent dLN trafficking
227
of cDC1, cDC2, and pDC but not neutrophils or macrophages. Moreover, IFN-I signaling was also required for
228
DMXAA-dependent decreases in dLN levels of B and T cells. Based on this we conclude that DMXAA induces
229
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
10
changes in dLN cell population that are both dependent on and independent of IFN-I signaling. Given the unique
230
transcriptomic and cytokine profiles observed in response to DMXAA it is probable that other factors are also
231
highly relevant and this will require additional investigation.
232
233
STING ligand-associated enhancement of humoral responses
234
Given the obvious innate induction stimulated by STING ligands we next compared the extent to which they
235
augment elicitation of antibody responses when co-administered with protein antigen. For this we used a
236
prime/boost strategy via intramuscular injection of OVA. As shown in Figure 6A, all ligands led to titers of OVA-
237
reactive total IgG that were significantly higher than observed when OVA was injected in the absence of adjuvant.
238
Additionally, these levels did not significantly differ from those observed when Alum was used. We next examined
239
levels of IgG1 and IgG2c subtypes as an indicator of class switching and T helper (Th) polarization. As shown
240
in Figures 6B and 6C, while IgG1 levels for each adjuvant followed the same pattern as total IgG, levels of
241
IgG2c were significantly higher for the STING ligands relative to Alum. This is consistent with what is known
242
about strong Th2 biased immune phenotypes observed for Alum [31] and Th1 biased responses observed for
243
CDA [32]. These results additionally indicate that the small molecule STING ligands diABZI and DMXAA
244
generate T helper polarization that more closely resembles CDA than Alum. Surprisingly, levels of IgG1 but not
245
IgG2c induced by diABZI were significantly lower than those induced by the other STING ligands. This may
246
indicate differential induction of immune processes by the molecule that are mediated by STING or other
247
unknown cellular targets. Regulation of antibody production and affinity maturation is mediated in large part
248
through the function of T follicular helper (TFH) cells whose activity can be enhanced by the use of adjuvants [33].
249
We therefore asked whether adjuvant-specific differences are evident in TFH activation measured following ex
250
vivo stimulation of splenocytes with OVA-inclusive peptides. As shown in Figure 6C, DMXAA but not the other
251
agonists was able to significantly increase OVA-specific TFH activity following vaccination (Supplemental Figure
252
2). Whether this leads to measurable differences in generation of antibody avidity will require additional studies.
253
254
STING ligand-associated enhancement of cell mediated responses
255
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
11
Previous vaccination models have shown enhanced antigen-directed T cell activation when STING agonists are
256
used as adjuvants [32,34,35]. We therefore used IFN ELISPOT to examine whether any differences are
257
observed between the ligands with respect to OVA-directed T cell activation. In duplicate experiments animals
258
were vaccinated as described above and harvested seven days after boost. Splenocytes were then stimulated
259
ex vivo using either an OVA-inclusive peptide pool or the MHC class I immunodominant OVA peptide SIINFEKL.
260
As shown in Figure 7A, stimulation with the peptide pool led to significant expression of IFN in splenocytes
261
harvested from animals adjuvanted with CDA or DMXAA relative to naïve animals or those vaccinated with OVA
262
+ PBS. Moreover, CDA adjuvant also led to significant IFN expression relative to all other adjuvants following
263
stimulation with SIINFEKL. Expectedly Alum was also unable to elicit any observed IFN expression. We next
264
used flow cytometry of splenocytes stimulated with OVA peptide pools to distinguish polyfunctional activation in
265
CD4+ and CD8+ T cells as indicated by expression levels of IFN and TNF measured following intracellular
266
cytokine staining (ICS) (Supplemental Figure 3). As shown in Figure 7B, significant expression of TNF and
267
IFN was observed in CD4+ and CD8+ T cells only following vaccination with CDA. These results suggest that
268
chemical structure of STING ligand associates with differential enhancement of antigen-directed T cell activation.
269
270
271
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
12
DISCUSSION
272
Transient pharmacological activation of STING has been shown in animal models to confer desirable immune-
273
mediated outcomes for numerous, seemingly unrelated clinical conditions. This includes enhancement of
274
protective responses conferred by vaccines against viral [3538], bacterial [3941], and protozoal [34,42]
275
pathogens. STING agonists have also been shown to impair acute replication of diverse virus types in vivo when
276
administered before challenge [4347]. Moreover, a wide variety of STING agonist platforms have shown
277
remarkable efficacy in mediating T (reviewed in [48]) and NK [49,50] cell-mediated clearance of transplanted
278
tumors including the generation of abscopal responses [9,51,52]. Intriguingly, pharmacologic STING activation
279
has also been shown to alleviate pain (nociception) via IFN-mediated effects on sensory neurons [53] and to
280
attenuate experimental autoimmune encephalitis [54,55]. As such, STING-dependent phenotypes associate with
281
cellular and immunological processes that can be harnessed for diverse clinical benefits and thus pursuit of new
282
agonists of the protein is highly incentivized.
283
284
While largely under investigated, the use of exogenous ligands to activate STING can lead to observable
285
differences in phenotypic responses that are linked to chemical structures and protein binding properties of the
286
agonists. The most overt demonstration of this is exemplified by the human-selective agonist C53, which induces
287
activation of STING by engaging the protein’s transmembrane domain [56]. When compared with molecules
288
such as diABZI or cGAMP that activate via the cytosolic domain, C53 is unable (and indeed blocks) induction of
289
TBK1-independent STING-mediated processes such as NLRP3 inflammasome activation, autophagy, and
290
lysosomal biogenesis [7,8,57]. The collective effects of differential induction of these cellular activities on STING-
291
mediated immunological processes are virtually unknown. Additionally, while cyclic dinucleotides including
292
cGAMP, cyclic-di-AMP (CDA) and cyclic di-GMP (CDG) bind to and activate STING, they can also distinctively
293
engage STING-independent PRRs including RECON and ERAdP (CDA [58,59]), or TPL2, DDX41, and TLR7
294
(CDG [6062]). These proteins are known to elicit innate immune effects that can enhance or counteract
295
canonical STING-mediated molecular processes through transcription factors such as IRF3 or NF-B or even
296
induce STING-independent activities such as CREB-dependent transcription. Whether the various classes of
297
small molecule agonists such as those examined here are also capable of activating STING-independent PRRs
298
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
13
is unknown. In vivo, STING agonist types display differences with respect to cytokine secretion [35,63],
299
enhancement of humoral immunity to subunit vaccines [35,64,65], and cell-mediated tumor clearance [9].
300
However, an understanding of the combined mechanistic impact of differential STING engagement or induction
301
of other PRRs by STING agonists on downstream in vivo immune effects is lacking. As such, studies that
302
comparatively characterize these are necessary to inform future work whose aim is to apply a pharmacologic
303
STING agonism approach toward achieving specific clinical goals.
304
305
Work described here demonstrates measurable differences in responses to chemically different STING agonists
306
using in vitro, ex vivo, and in vivo models. This includes profound differences in transcriptional activity, cytokine
307
secretion profiles, expression of DC maturation markers, elicitation of antigen-directed humoral responses, and
308
cell mediated immune responses. Pharmacokinetic variability is certain to exist between the agonists but given
309
the differences in observed effects in vitro and ex vivo are unlikely to be solely responsible for the differences
310
detected in vivo. This could be due to engagement of STING-independent PRRs, differential induction of STING-
311
mediated processes linked to ligand-induced conformational changes, specific regions of STING binding, or
312
regulation of unknown signaling pathways. These results clearly show that DMXAA generates the most unusual
313
molecular responses despite not displaying atypical relative potency. Interestingly, DMXAA is the only agonist
314
examined here that is species selective, being inactive against primate STING orthologs [12,66]. As such it may
315
engage the protein with unique affinity or kinetics that influence activation properties. Since mammalian STING
316
evolves rapidly under positive selection, likely imposed by the fitness impacts of microbial pathogens [67],
317
species selective agonists are common [13,47,56,6870]. Moreover, cross-reactive agonists may elicit ortholog-
318
specific molecular activities and thus their use necessitates confirming activation of immunologically relevant
319
phenotypes in both species.
320
321
These results also suggest agonist-associated differences in the ability to augment antigen-directed CD4+ and
322
CD8+ T cell responses with CDA displaying more efficacy in this regard than the small molecule agonists (Figure
323
7). This has relevance for both vaccine enhancement and anti-cancer activity as T cells play key roles in both
324
pathogen clearance and tumor cell killing. Importantly, CDA was observed to induce T cell polyfunctionality as
325
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
14
indicated by their expression of both IFN and TNF. As such, better protective immune effects may be
326
demonstrable when using cyclic dinucleotides to potentiate antigen-directed immune responses. Whether this
327
can be mechanistically linked to differential molecular processes induced (or unaffected) by CDA such as
328
activation of NF-B-dependent transcription (Figure 1C), inflammasome activation (Figure 1D), or
329
transcriptomic patterns (Figure 4) requires additional inquiry but may allow discovery of precise phenotypes
330
desirable for future adjuvant development.
331
332
STING represents a valuable pharmacologic target with demonstrated beneficial applications to vaccine
333
enhancement, anti-tumor responses, antiviral protection, and pain suppression. By demonstrating agonist-
334
associated differences in molecular, cellular, and immunological responses this work provides a rationale for
335
pursuing deeper comparative characterization of STING agonists under consideration for clinical use. This
336
includes quantifying cellular processes they induce such as autophagy [6] , lysosomal biogenesis [7] , and
337
inflammasome activation [5] . Additionally, an understanding cell type-specific differences including agonist
338
sensitivity, transcriptomes, and impact on effector phenotypes will be essential to predict whether the mechanism
339
of action necessary for a desired outcome will be operational across species for a specific agonist. This will also
340
allow identification of criteria essential for validation in both animal and human models that will direct their clinical
341
use.
342
343
344
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
15
MATERIALS AND METHODS
345
Reagents and Antibodies
346
diABZI was purchased from MedChemExpress (Cat # HY-112921B). DMXAA was purchased from ApexBio (Cat
347
# A8233). Sendai virus (SeV) was purchased from Charles River Laboratories (Cat # PI-1). Endotoxin-free
348
ovalbumin was purchased from Invivogen (Cat # vac-pova-100). Steady GLO luciferase reagent was purchased
349
from Promega (Cat # E25100). Quanti-Luc luciferase detection system was purchased from Invivogen (Cat #
350
rep-qlc4lg1). Quanti-Blue SEAP detection reagent was purchased from InVivogen (Cat # rep-qbs). Cell Titer Glo
351
was purchased from Promega (Cat # G7572). Alhydrogel aluminum hydroxide was purchased from Invivogen
352
(Cat # vac-alu-250). ML-RR-S2 CDA was purchased from Invivogen (Cat # tlrl-nacda2r-05). LPS was purchased
353
from Sigma-Aldrich (Cat # L5293). J774-Dual cells were purchased from Invivogen (cat # j774d-nfis). RAW264.7-
354
ISG-Lucia cells were purchased from Invivogen (cat # rawl-isg). GM-CSF and IL-4 were obtained from Peprotech
355
(Cat # 214-14, 315-03. Antibodies used are as follows: anti-mouse CD3 (BioLegend Cat # 100355), anti-mouse
356
CD40 (BioLegend Cat # 124622), anti-mouse CD80 (BioLegend Cat # 124622), anti-mouse CD86 (BioLegend
357
Cat # 105008), anti-mouse CD11c (BioLegend Cat # 117353), anti-mouse CD8 (Thermo Fisher Cat # 45-0081-
358
82), anti-mouse CD19 (BioLegend Cat # 115540), anti-mouse CD45R (BD Bioscience Cat # 553088), anti-
359
mouse CD11b (Thermo Fisher Cat # 48-0112-82), anti-mouse Ly-6G (BD Biosciences Cat # 562700), anti-
360
mouse F4/80 (BioLegend Cat # 123118), anti-mouse IFN (BD Biosciences Cat # 554412), anti-mouse TNF
361
(BioLegend Cat # 506307), anti-mouse CD25 (BD Biosciences Cat #564458), anti-mouse PD-1 (BioLegend Cat
362
#135225), anti-mouse OX40 (BioLegend Cat #119415), anti-mouse Bcl-6 (BD Biosciences Cat # 563363), anti-
363
mouse CD16/CD32 Ab (BD Biosciences Cat #553142), anti-mouse CD45R (BioLegend Cat #103243), anti-
364
mouse CXCR5 (BioLegend Cat #145512), anti-mouse CD3 (BioLegend Cat #100353), anti-mouse CD4 (Thermo
365
Fisher Cat #48-0041-82), and anti-mouse CD8a (Thermo Fisher Cat #MA5-17597).
366
367
Mouse Experiments
368
All mouse experiments were performed at Oregon Health and Science University in ABSL2 laboratories in
369
compliance with OHSU Institutional Animal Care and Use Committee (IACUC), under protocol 0913. The OHSU
370
IACUC adheres to NIH Office of Laboratory Animal Welfare standards (OLAW welfare assurance A3304-1).
371
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
16
C57B6/J or IFNAR-/- mice (aged 5-8 weeks; Jackson Laboratories) were housed in ventilated cage units and
372
cared for under USDA guidelines for laboratory animals. For serum cytokine measurement, transcriptomic, and
373
dLN cell population experiments STING agonists and adjuvant controls were administered intramuscularly (IM)
374
in the quadriceps with DMXAA at 25 mg/kg, diABZI at 1.5 mg/kg, ML-RR-S2 at 0.5 mg/kg, aluminum hydroxide
375
Alum at 15 mg/kg, or PBS. For vaccination experiments animals were primed and identically boosted at 2 w and
376
harvested at 4 w (for antibody readout) or 5 w (for T cell readout). For transcriptomic and draining lymph node
377
experiments STING agonists and other adjuvants were admixed with 10 µg Endofit-ovalbumin (OVA). For T cell
378
proliferation experiments adjuvants were administered with 50 µg Endofit-OVA.
379
380
RAW264.7 and J774 Assays
381
RAW264.7-ISG-Lucia or J774-Dual cells were seeded to confluence in a 96-well plate overnight in DMEM + 10%
382
FCS. Cells were treated in duplicate with serial dilutions of STING ligands; diABZI and DMXAA starting at 100
383
µM concentrations. Lipofectamine 3000 (Invitrogen) was used to transfect CDA into RAW264.7 and J774 cells,
384
starting at 100 µM. Lipofectamine was added prior to serial dilutions of CDA. Cells were incubated with STING
385
ligands overnight, 18 h, in 50 uL of DMEM + 2% FBS. Quanti-Luc, Quanti-Blue, or Cell Titer Glo reagents were
386
added (1:1 [v/v]) to each well, and luminescence or absorbance measured on a Synergy plate reader (BioTek).
387
For IL-1 secretion experiments J774 cells were primed 6 h with 100 ng/mL LPS and treated overnight with
388
indicated stimuli. Media was harvested and IL-1 quantified using bead-based Luminex assay per the
389
manufacturer’s instructions (Thermo Fisher Cat # EPX01A-26002-901).
390
391
ELISA Assays
392
Serological responses specific to OVA were quantified by enzyme-linked immunosorbent assays (ELISA). Blood
393
was collected at 2 w post boost, stored for approx. 1 h at 4° C, and then centrifuged to separate sera. Sera was
394
aspirated and stored at -80 °C until analysis. High-binding microtiter plates were coated with OVA (1 µg/well)
395
(Invivogen) overnight at at 4° C. Coated plates were blocked using 5% (w/v) nonfat dry milk in PBS plus 0.05%
396
Tween 20 (v/v) (PBS-T) for 1 hour at room temperature. Mouse sera was heat-inactivated for 30 mins at 55°C,
397
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
17
then serial diluted in 5% nonfat dry milk starting at 1:50 dilution and added to microtiter plate in duplicate. After
398
2 h incubation plates were washed 3X using accuWash plate washer (Fisher Scientific). Goat anti-mouse IgG-
399
HRP (SouthernBiotech Cat # 1030-05), anti-mouse IgG1-HRP (SouthernBiotech Cat # 1070-05), or anti-mouse
400
IgG2c-HRP (SouthernBiotech Cat # 1079-05) conjugated antibodies (at 1:10,000) were added and incubated for
401
1 h at room temp. ELISA plates were washed and developed with 0.4 mg/ml o-phenylene diamine buffer (50 nM
402
citric acid, 100 mM dibasic sodium phosphate, pH 5.0). The reaction was halted with 1 N HCl and A490 was
403
measured on a BioTek Synergy plate reader. Absorbance was quantified by calculating area under the curve
404
across serial dilutions above baseline detection.
405
406
Cytokine quantification
407
Proinflammatory cytokines in the sera were examined 5 h after injection. Blood was collected, stored for approx.
408
1 h at 4°C, and then centrifuged to separate sera which was aliquoted and stored at -80 °C until analysis. Per
409
the manufacturer’s instructions cyto/chemokines from 50 µL sample of each serum aliquot was quantified using
410
a ProcartaPlex Mouse Th1/Th2 multiplex panel (ThermoFisher Cat # EPX260-26088-901).
411
412
RNA isolation and dLN RT-qPCR
413
Total RNA was isolated from inguinal draining lymph nodes at 5, 24, and 72 hours after IM injection. RNA was
414
treated on column with DNase provided in a Quick RNA miniprep kit (Zymo Research Cat # R1055), according
415
to manufacturer’s protocol. Single-stranded cDNA was generated from total RNA using a RevertAid First Strand
416
cDNA synthesis kit (Thermo Fisher Cat # 1622), using random hexamers to prime first strand synthesis. mRNA
417
expression levels between samples was performed using semiquantitative real-time revere transcription-PCR
418
(qPCR) and quantified by ΔΔCT [71] . Prevalidated Prime-Time 6-carboxyfluorescein qPCR primer/probe sets
419
were obtained from IDT and used for all genes.
420
421
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
18
Hybridization Array Analysis
422
Differential gene expression in lymph nodes of STING agonist IM-injected mice was determined using RNA
423
isolated from the inguinal dLN at 5 and 24 h. Hybridization array data were collected by the OHSU Integrated
424
Genomics Laboratory. RNA sample quantity and purity were measured by UV absorbance at 260, 280, and 230
425
nm with a NanoDrop 1000 spectrophotometer (ThermoScientific). RNA integrity and size distribution were
426
determined by running total RNA on a Nano chip instrument (Agilent Technologies). RNA was prepared for array
427
hybridization by labeling 100 ng aliquots using the 3’IVT Express kit (Affymetrix). RNA was reverse transcribed
428
to generate first-strand cDNA containing a T7 promoter sequence. A second-strand cDNA synthesis step was
429
performed that converted the single-stranded cDNA into a dsDNA template for transcription. Amplified and biotin-
430
labeled cRNA was generated during the in vitro transcription step. After a magnetic bead purification step to
431
remove enzymes, salts, and unincorporated nucleotides, the cRNA was fragmented. Labeled and fragmented
432
cRNA was combined with hybridization cocktail components and hybridization controls, and 130 µL of each
433
hybridization cocktail containing 6.5 µg of labeled target was injected into a cartridge containing the Clariom S
434
murine array (ThermoFisher cat # 902930) containing >221,900 probes interrogating over 150,000 transcripts
435
from >22,100 genes. Arrays were incubated for 18 h at 45°C, followed by washing and staining on a GeneChip
436
Fluidics Station 450 (Affymetrix) and the associated hybridization wash and stain kit. Arrays were scanned using
437
the GeneChip Scanner 3000 7G with an autoloader. Image inspection was performed manually immediately
438
following each scan. Image processing of sample .DAT files to generate probe intensity .CEL files was performed
439
using the Affymetrix GeneChip Command Console (AGCC) software. Each array file was then analyzed using
440
Transcriptome Analysis Console (TAC; Version 4.0.3) to obtain array performance metrics, calculate transcript
441
fold changes, and perform principal component analysis. To identify probe sets that were significantly regulated
442
in treated versus untreated (mock) cells, we employed a traditional unpaired one-way (single-factor) ANOVA for
443
each pair of condition groups as implemented in TAC. Probe sets were considered differentially regulated if the
444
ANOVA P value was <0.05 and fold change >2. Pathway analysis of significantly regulated mRNAs performed
445
using Ingenuity Pathway Analysis (Qiagen). Venn diagrams were constructed using BioVenn [72].
446
447
Intracellular Cytokine Staining (ICS) assay
448
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
19
Splenocytes from vaccinated mice were incubated with 1 µg/mL OVA peptide pool (#130-099-771; Miltenyi
449
Biotec) at 37oC with 5% CO2 for 6 h in the presence of Brefeldin A (#TNB-4506; Tonbo Bioscience) in 96-well U-
450
bottom plates at a concentration of 106 cells/well. Stimulation with 25 ng/mL PMA (Invivogen Cat # tlrl-pma) and
451
500 ng/mL Ionomycin (Sigma-Aldrich Cat #10634) was included as a positive control. DMSO was used as a
452
negative control. After stimulation, the cells were incubated in anti-mouse CD16/CD32 and stained for 30 min at
453
4oC with the surface-staining fluorochrome-conjugated Abs: CD3, CD4 (Clone: GK1.5; #48-0041-82; Thermo
454
Fisher Scientific), and CD8a (Clone: CT-CD8a; # MA5-17597; Thermo Fisher Scientific). Intracellular staining
455
was performed for 30 min at 4◦C with the fluorochrome-conjugated anti-mouse IFNγ and anti-mouse TNFα using
456
BD Cytofix/Cytoperm Fixation/Permeabilization kit (BD Biosciences Cat #554714), accordingly to manufacturer’s
457
instructions. Cell events were collected on BD Symphony flow cytometer, and data were analyzed using FlowJo
458
10 software.
459
460
Activation induced marker (AIM) assay
461
Splenocytes from vaccinated mice were incubated with 2 µg/mL OVA peptide pool (Miltenyi Cat # #130-099-
462
771) at 37◦C with 5% CO2 for 18h. Stimulation with 5 µg/mL Con A (InVivogen Cat # inh-cona-2) was included
463
as a positive control. After stimulation, cells were stained with LIVE/DEAD Fixable Near-IR Dead Cell Stain Kit
464
(ThermoFisher Scientific Cat # L10119) and blocked with mouse Fc block CD16/CD32 (BD Biosciences Cat
465
#553141). Cells were then stained for 30 min at 4◦C with the following fluorochrome-conjugated anti-mouse Abs:
466
CD3, CD4, CD45R/B220, CD25, PD-1, OX40, and CXCR5. Cell events were collected on LSR-II (BD
467
Biosciences) flow cytometer, and data were analyzed using FlowJo 10 software.
468
469
BMDC activation
470
Bone marrow extraction was performed by flushing RPMI media through mouse femurs. Cells were pelleted by
471
centrifugation for 5 mins at 500 x g and RBC lysed with ACK lysis (Gibco) buffer at room temp for 3 mins. After
472
centrifugation cells were resuspened in RPMI (1 mL/femur), and counted by trypan blue exclusion (Countess,
473
Invitrogen). Cells were adjusted to 5 x 106 cells per 10 cm petri dish, RPMI was supplemented with 20 ng/mL
474
GM-CSF and 10 ng/ml IL-4 for dendritic cell differentiation. Plates were incubated for 8 d with fresh supplemented
475
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
20
media added on days 3 and 6. On day 8 adherent bone marrow derived dendritic cells (BMDC) were chemically
476
lifted from petri dish (Cellstripper, Corning Cat # 25-056-Cl). BMDCs were again counted and plated into 6-well
477
plates at 1-2 x 106 cells/well. Stimuli were added to wells as follows; CDA (1.36 µM), diABZI (17.5 nM), DMXAA
478
(17.5 µM), Alum (3 µg/ml), LPS (100 ng/ml), and DMSO (0.5%). BMDCs were incubated for 20 h, then harvested
479
as before. Antibody staining was performed in FACS buffer [PBS, 0.5% BSA (w/v), 1% 0.5 M EDTA (v/v)]. Fc-
480
mediated binding was blocked with mouse FC-Block (CD16/CD32) (BD Bioscience Cat # 553141), and dead
481
cells stained with Live/Dead Near IR. BMDCs were stained for activation markers CD80, CD86, CD40, and MHC-
482
II for 45 mins on ice. Data acquisition was performed using an LSR-II (BD Biosciences) and analyzed using
483
Flowjo 10.
484
485
APC recruitment
486
Mice were injected with STING agonists and OVA as described above. Mice euthanized at 24 h time point, and
487
inguinal dLN harvested. Single-cell suspensions were blocked with Fc Block and stained with anti-CD19, anti-
488
B220, anti-CD11b, anti-CD11c, anti-Ly6-G PE-CF594, and anti-F4/80 for 45 mins on ice. Data acquisition was
489
performed using an LSR-II (BD Biosciences) and analyzed using Flowjo 10.
490
491
ELISpot Assays
492
IFN ELISpot assays were conducted as described [73]. Single-cell suspensions of splenocytes were obtained
493
following RBC lysis and added to prewashed mouse IFNγ ELISPot plates (MabTech Cat # 3321-2A).
494
Splenocytes were stimulated with SIINFEKL peptide or OVA peptide pool (10 µg/well), Concanavalin A (0.1
495
µg/well), PMA/Ion (0.8ng/30ng/well), or 0.5% DMSO. Splenocytes were incubated in ELISpot plates for 18 h.
496
ELISpots were stained according to manufacturer’s protocol. Briefly, plates were washed and incubated with
497
anti-mouse IFNγ biotin antibody for 2 h. Steptavidin-ALP secondary antibody was added for 1 h after plates were
498
washed. Spots were detected using BCIP/NPT-plus substrate, rinsed with water, and dried before counting with
499
an AID ELISpot plate reader.
500
501
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
21
FIGURE LEGENDS
502
Figure 1. Dose-dependent innate responses induced by STING agonists in murine cell lines. RAW264.7
503
(A) and J774 (B, C, D) were grown to confluence in 96 well plates and exposed in duplicate overnight to CDA,
504
DMXAA, or diABZI at indicated molarities. A. Cells were exposed to either Quanti-Luc or Cell Titer Glo lysis
505
buffer to measure IFN-I signaling induction or cell viability, respectively. Data presented are mean ± SEM relative
506
luminescence units (RLU, black) or percent viability (red) determined relative to control cells treated with DMSO-
507
or transfection reagent (CDA); C. NF-kB-dependent SEAP expression expressed as mean ± SEM absorbance
508
at OD655 for indicated treatment relative to control treated cells; D. Secretion of IL-1b from primed J774 cells
509
treated with indicated doses of DMXAA, diABZI, or CDA. Data displayed are mean ± SEM IL-1b pg/mL secreted
510
into culture media. Two-way ANOVA with Tukey’s correction for multiple comparisons was used to examine
511
statistical significance of dose-specific activation of NF-kB or IL-1b secretion between agonists (*p < 0.05, **p <
512
0.01, ***p < 0.001, ****p < 0.0001 color coded as indicated for agonist pairing).
513
514
Figure 2. Maturation of Dendritic Cells from C57Bl/6 Mice in Response to Innate Stimuli. Immature bone
515
marrow derived DC (BMDC) from C57Bl/6 mice were treated with 0.5% DMSO, 1 µM diABZI, 50 μM DMXAA, 1
516
µM CDA, 100 ng/mL LPS, or 3 µg/mL Alum as indicated. Cells were collected after 24 h and stained for
517
expression of CD40, CD80, CD86 and MHC-II and analyzed by flow cytometry. A. Mean fluorescence intensities
518
(MFI) ± SD for indicated treatment and surface marker. One way ANOVA with Tukey’s multiple comparisons test
519
was used to compare results between treatments (***p < 0.001****; p < 0.0001); B. Representative MFI
520
histograms for each marker are shown for cells exposed to indicated stimulus.
521
522
Figure 3. Induction of systemic cytokine secretion by STING agonists in vivo. C57Bl/6 mice were injected
523
IM with PBS, 30 µg diABZI, 10 µg, CDA, or 500 µg DMXAA and serum harvested at 24 h. Luminex bead based
524
multiplex assay was then used to measure absolute levels of indicated cytokines. Data presented are mean ±
525
SD pg/mL. One way ANOVA with Dunnet correction for multiple comparisons was used to determine statistical
526
significance (**p <0.01, ***p < 0.001, ***p < 0.0001).
527
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
22
528
Figure 4. STING agonist-induced transcriptomic changes in vivo. C57Bl/6 mice were injected
529
intramuscularly with PBS, CDA, diABZI or DMXAA and total RNA from draining lymph nodes harvested at
530
indicated time point. A. Levels of indicated mRNA transcript in draining lymph nodes at 5 h, 24 h, or 72 h for
531
indicated treatment as determined by qPCR. Data presented are mean ± SEM fold changes relative to PBS
532
treated control animals. Statistical significance was determined using a mixed effects ANOVA with Tukey’s
533
correction for multiple comparisons of log transformed fold changes between stimuli at each time point (*p <
534
0.05; **p < 0.01); B. Number of mRNAs significantly up or downregulated more than twofold relative to PBS
535
treatment following indicated treatment as determined by hybridization array; C. Principal component analysis of
536
transcript fold changes for indicated stimulus and time point; D. Venn diagram of transcripts significantly
537
upregulated for indicated stimulus at 5 h and 24 h; E. Signaling pathways displaying a predicted Z score > 1 for
538
all three stimuli at 5 h and 24 h as determined by Ingenuity Pathway Analysis; F. Pathways displaying a predicted
539
Z score > 1 following only DMXAA treatment at 5 h and 24 h.
540
541
Figure 5. STING agonist-induced changes in draining lymph node immune cell populations. In duplicate
542
experiments C57Bl/6 mice were injected intramuscularly with OVA in the presence of PBS, CDA, diABZI DMXAA,
543
or Alum and single cell suspensions of draining lymph nodes obtained at 24 h post treatment. Flow cytometry
544
was then used to quantify indicated cell types. A. Total number of lymph node cells following indicated treatment;
545
B. Percent of total lymph node cells represented by indicated B cells, T cells, cDC1, cDC2, pDC, neutrophils,
546
and macrophages as indicated; C. Percent of total lymph node cells represented by indicated B cells, T cells,
547
cDC1, cDC2, pDC, neutrophils, and macrophages in WT and IFNAR-/- mice as indicated. Statistical significance
548
was determined using ANOVA with Dunnet’s correction for multiple comparisons (*p < 0.05; **p < 0.01; ***p <
549
0.001; ****p < 0.0001). Individual experiments indicated by circle color.
550
551
Figure 6. Enhancement of humoral immune response to OVA by co-administered STING agonists.
552
C57Bl/6 mice were vaccinated IM with OVA in the presence of PBS, CDA, diABZI, DMXAA, or Alum, identically
553
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
23
boosted 2 w later and harvested 2 w later. A. ELISA was performed using serial dilution of harvested sera to
554
quantify levels of total IgG, IgG1, and IgG2c reactive to OVA antigen. Data presented are mean + SEM
555
absorbance (top) as well as mean + SD area under curve (AUC) of absorbance signal including individual animal
556
measurements (bottom); Total number of lymph node cells following indicated treatment; B. Mean + SD ratio of
557
IgG2c to IgG1 AUC titers; C. Mean + SD percentage of OVA-reactive (OX40/CD25+) TFH (CXCR5+PD-1high)
558
cells harvested seven days after boost with OVA + indicated adjuvant. Statistical significance was determined
559
using ANOVA with Dunnet’s correction for multiple comparisons (*p < 0.05; **p < 0.01; ***p < 0.001; ****p <
560
0.0001).
561
562
Figure 7. STING-adjuvant associated enhancement of cell mediated responses. C57Bl/6 mice were left
563
untreated or vaccinated IM using a prime/boost strategy with OVA in the presence of PBS, CDA, diABZI, DMXAA,
564
or Alum. A. Splenocytes were harvested at 7 d post boost and stimulated ex vivo with a peptide pool spanning
565
the OVA protein (left) or SIINFEKL peptide (right). ELISpot was then used to quantify the number of IFNg positive
566
cells. Data presented are mean ± SD number of IFNg positive spot forming units (SFU) per 2.5 x 105 total cells.
567
Results from individual animals are also presented with duplicate experiments represented by marker color; B.
568
Intracellular cytokine staining and flow cytometry was used to quantify OVA peptide pool-stimulated expression
569
of IFNg and TNFa from CD4+ and CD8+ T cells in splenocytes. Data presented are mean ± SD percentage of
570
CD8+ (left) or CD4+ (right) T cells that display positivity for indicated cytokine. Individual animals represented by
571
circles. One way ANOVA using Tukey’s multiple comparison was used to determine statistical significance (*p <
572
0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001).
573
574
Supplemental Table 1. STING agonist-mediated dLN transcriptomic changes. Fold changes of mapped
575
transcripts from hybridization array. Data presented are log2 transformed fold changes for mRNAs and
576
corresponding probe IDs for indicated stimulus relative to mock treated animals and -log(FDRp) values.
577
578
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
24
Supplemental Table 2. Stimulus-induced pathway regulation. Predicted Z scores of indicated biological
579
pathways based on dLN transcriptomic patterns of STING agonists.
580
581
Supplemental Figure 1. Representative flow cytometry gating plots for dLN B cells, T cells, neutrophils, pDC,
582
cDC1, cDC2, pDC, and macrophages.
583
584
Supplemental Figure 2. Representative flow cytometry gating plots for IFN+ and TNF+ CD4+ and CD8+ T
585
cells.
586
587
Supplemental Figure 3. Representative flow cytometry gating plots for activated TFH cells.
588
589
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
25
REFERENCES
590
591
[1] Coffman RL, Sher A, Seder RA. Vaccine adjuvants: putting innate immunity to work. Immunity
592
2010;33:492503. https://doi.org/10.1016/j.immuni.2010.10.002.
593
[2] Shi S, Zhu H, Xia X, Liang Z, Ma X, Sun B. Vaccine adjuvants: Understanding the structure and mechanism
594
of adjuvanticity. Vaccine 2019:112. https://doi.org/10.1016/j.vaccine.2019.04.055.
595
[3] Pulendran B, Arunachalam PS, O’Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat
596
Rev Drug Discov 2021;20:122. https://doi.org/10.1038/s41573-021-00163-y.
597
[4] Singleton KL, Joffe A, Leitner WW. Review: Current trends, challenges, and success stories in adjuvant
598
research. Front Immunol 2023;14:1105655. https://doi.org/10.3389/fimmu.2023.1105655.
599
[5] Abdul-Sater AA, Tattoli I, Jin L, Grajkowski A, Levi A, Koller BH, et al. Cyclic-di-GMP and cyclic-di-AMP
600
activate the NLRP3 inflammasome. EMBO Reports 2013;14:9006. https://doi.org/10.1038/embor.2013.132.
601
[6] Gui X, Yang H, Li T, Tan X, Shi P, Li M, et al. Autophagy induction via STING trafficking is a primordial
602
function of the cGAS pathway. Nature 2019;567:2626. https://doi.org/10.1038/s41586-019-1006-9.
603
[7] Lv B, Dion WA, Yang H, Xun J, Kim D-H, Zhu B, et al. A TBK1-independent primordial function of STING in
604
lysosomal biogenesis. Mol Cell 2024. https://doi.org/10.1016/j.molcel.2024.08.026.
605
[8] Xu Y, Wang Q, Wang J, Qian C, Wang Y, Lu S, et al. The cGAS-STING pathway activates transcription
606
factor TFEB to stimulate lysosome biogenesis and pathogen clearance. Immunity 2024.
607
https://doi.org/10.1016/j.immuni.2024.11.017.
608
[9] Corrales L, Glickman LH, McWhirter SM, Kanne DB, Sivick KE, Katibah GE, et al. Direct Activation of
609
STING in the Tumor Microenvironment Leads to Potent and Systemic Tumor Regression and Immunity. Cell
610
Reports 2015:114. https://doi.org/10.1016/j.celrep.2015.04.031.
611
[10] Ramanjulu JM, Pesiridis GS, Yang J, Concha N, Singhaus R, Zhang S-Y, et al. Design of
612
amidobenzimidazole STING receptor agonists with systemic activity. Nature 2018;564:43943.
613
https://doi.org/10.1038/s41586-018-0705-y.
614
[11] Plowman J, Narayanan VL, Dykes D, Szarvasi E, Briet P, Yoder OC, et al. Flavone acetic acid: a novel
615
agent with preclinical antitumor activity against colon adenocarcinoma 38 in mice. Cancer Treat Rep
616
1986;70:6315.
617
[12] Conlon J, Burdette DL, Sharma S, Bhat N, Thompson M, Jiang Z, et al. Mouse, but not Human STING,
618
Binds and Signals in Response to the Vascular Disrupting Agent 5,6-Dimethylxanthenone-4-Acetic Acid. J
619
Immunol 2013;190:521625. https://doi.org/10.4049/jimmunol.1300097.
620
[13] Gall B, Pryke K, Abraham J, Mizuno N, Botto S, Sali TM, et al. Emerging Alphaviruses Are Sensitive to
621
Cellular States Induced by a Novel Small-Molecule Agonist of the STING Pathway. J Virol 2018;92.
622
https://doi.org/10.1128/jvi.01913-17.
623
[14] Doel P van den, Volz A, Roose JM, Sewbalaksing VD, Pijlman GP, Middelkoop I van, et al. Recombinant
624
modified vaccinia virus Ankara expressing glycoprotein E2 of Chikungunya virus protects AG129 mice against
625
lethal challenge. PLoS Neglected Tropical Diseases 2014;8:e3101.
626
https://doi.org/10.1371/journal.pntd.0003101.
627
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
26
[15] Gaidt MM, Ebert TS, Chauhan D, Ramshorn K, Pinci F, Zuber S, et al. The DNA Inflammasome in Human
628
Myeloid Cells Is Initiated by a STING-Cell Death Program Upstream of NLRP3. Cell 2017:134.
629
https://doi.org/10.1016/j.cell.2017.09.039.
630
[16] Hoyt LR, Ather JL, Randall MJ, DePuccio DP, Landry CC, Wewers MD, et al. Ethanol and Other Short-
631
Chain Alcohols Inhibit NLRP3 Inflammasome Activation through Protein Tyrosine Phosphatase Stimulation. J
632
Immunol 2016;197:132234. https://doi.org/10.4049/jimmunol.1600406.
633
[17] Dhuna K, Helliwell R, Luca SND, Spencer SJ, Stokes L. Ginsenosides enhance P2X7-dependent cytokine
634
secretion from LPS-primed rodent macrophages. Purinergic Signal 2024;20:6571.
635
https://doi.org/10.1007/s11302-023-09935-0.
636
[18] Palucka K, Banchereau J, Mellman I. Designing Vaccines Based on Biology of Human Dendritic Cell
637
Subsets. Immunity 2010;33:46478. https://doi.org/10.1016/j.immuni.2010.10.007.
638
[19] Rosa SCD, Cohen KW, Bonaparte M, Fu B, Garg S, Gerard C, et al. Wholeblood cytokine secretion
639
assay as a highthroughput alternative for assessing the cellmediated immunity profile after two doses of an
640
adjuvanted SARSCoV2 recombinant protein vaccine candidate. Clin Transl Immunol 2022;11:e1360.
641
https://doi.org/10.1002/cti2.1360.
642
[20] Burny W, Callegaro A, Bechtold V, Clément F, Delhaye S, Fissette L, et al. Different Adjuvants Induce
643
Common Innate Pathways That Are Associated with Enhanced Adaptive Responses against a Model Antigen
644
in Humans. Frontiers in Immunology 2017;8:8017. https://doi.org/10.3389/fimmu.2017.00943.
645
[21] Qiao X, Pham DNT, Luo H, Wu J. Ran Overexpression Leads to Diminished T Cell Responses and
646
Selectively Modulates Nuclear Levels of c-Jun and c-Fos*. J Biol Chem 2010;285:548896.
647
https://doi.org/10.1074/jbc.m109.058024.
648
[22] Courtemanche C, Elson-Schwab I, Mashiyama ST, Kerry N, Ames BN. Folate Deficiency Inhibits the
649
Proliferation of Primary Human CD8+ T Lymphocytes In Vitro. J Immunol 2004;173:318692.
650
https://doi.org/10.4049/jimmunol.173.5.3186.
651
[23] Caproni E, Tritto E, Cortese M, Muzzi A, Mosca F, Monaci E, et al. MF59 and Pam3CSK4 Boost Adaptive
652
Responses to Influenza Subunit Vaccine through an IFN Type I-Independent Mechanism of Action. J Immunol
653
2012;188:308898. https://doi.org/10.4049/jimmunol.1101764.
654
[24] Agallou M, Margaroni M, Tsanaktsidou E, Badounas F, Kammona O, Kiparissides C, et al. A liposomal
655
vaccine promotes strong adaptive immune responses via dendritic cell activation in draining lymph nodes. J
656
Control Release 2023;356:386401. https://doi.org/10.1016/j.jconrel.2023.03.006.
657
[25] Heo Y, Ko E, Park S, Park S-O, Ahn B-C, Yum J-S, et al. L-PampoTM, a Novel TLR2/3 Agonist, Acts as a
658
Potent Cancer Vaccine Adjuvant by Activating Draining Lymph Node Dendritic Cells. Cancers 2023;15:3978.
659
https://doi.org/10.3390/cancers15153978.
660
[26] Nakagawa T, Tanino T, Onishi M, Tofukuji S, Kanazawa T, Ishioka Y, et al. S-540956, a CpG
661
Oligonucleotide Annealed to a Complementary Strand With an Amphiphilic Chain Unit, Acts as a Potent
662
Cancer Vaccine Adjuvant by Targeting Draining Lymph Nodes. Front Immunol 2021;12:803090.
663
https://doi.org/10.3389/fimmu.2021.803090.
664
[27] Larkin B, Ilyukha V, Sorokin M, Buzdin A, Vannier E, Poltorak A. Cutting Edge: Activation of STING in T
665
Cells Induces Type I IFN Responses and Cell Death. J Immunol 2017;199:397402.
666
https://doi.org/10.4049/jimmunol.1601999.
667
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
27
[28] Cerboni S, Jeremiah N, Gentili M, Gehrmann U, Conrad C, Stolzenberg M-C, et al. Intrinsic
668
antiproliferative activity of the innate sensor STING in T lymphocytes. J Exp Med 2017;214:176985.
669
https://doi.org/10.1084/jem.20161674.
670
[29] Walker MM, Crute BW, Cambier JC, Getahun A. B CellIntrinsic STING Signaling Triggers Cell Activation,
671
Synergizes with B Cell Receptor Signals, and Promotes Antibody Responses. J Immunol 2018;201:264153.
672
https://doi.org/10.4049/jimmunol.1701405.
673
[30] Polidarova MP, Brehova P, Dejmek M, Birkus G, Brazdova A. STING Agonist-Mediated Cytokine
674
Secretion Is Accompanied by Monocyte Apoptosis. Acs Infect Dis 2021.
675
https://doi.org/10.1021/acsinfecdis.1c00554.
676
[31] McKee AS, Munks MW, Marrack P. How Do Adjuvants Work? Important Considerations for New
677
Generation Adjuvants. Immunity 2007;27:68790. https://doi.org/10.1016/j.immuni.2007.11.003.
678
[32] Volckmar J, Knop L, Stegemann-Koniszewski S, Schulze K, Ebensen T, Guzmán CA, et al. The STING
679
activator c-di-AMP exerts superior adjuvant properties than the formulation poly(I:C)/CpG after subcutaneous
680
vaccination with soluble protein antigen or DEC-205-mediated antigen targeting to dendritic cells. Vaccine
681
2019:112. https://doi.org/10.1016/j.vaccine.2019.07.019.
682
[33] Desbien AL, Cauwelaert ND, Reed SJ, Bailor HR, Liang H, Carter D, et al. IL-18 and Subcapsular Lymph
683
Node Macrophages are Essential for Enhanced B Cell Responses with TLR4 Agonist Adjuvants. The Journal
684
of Immunology 2016;197:43519. https://doi.org/10.4049/jimmunol.1600993.
685
[34] Matos MN, Cazorla SI, Schulze K, Ebensen T, Guzmán CA, Malchiodi EL. Immunization with Tc52 or its
686
amino terminal domain adjuvanted with c-di-AMP induces Th17+Th1 specific immune responses and confers
687
protection against Trypanosoma cruzi. PLoS Neglected Tropical Diseases 2017;11:e0005300.
688
https://doi.org/10.1371/journal.pntd.0005300.
689
[35] Liu Z, Zhou J, Xu W, Deng W, Wang Y, Wang M, et al. A novel STING agonist-adjuvanted pan-
690
sarbecovirus vaccine elicits potent and durable neutralizing antibody and T cell responses in mice, rabbits and
691
NHPs. Cell Res 2022;32:26987. https://doi.org/10.1038/s41422-022-00612-2.
692
[36] Tang CK, Aoshi T, Jounai N, Ito J, Ohata K, Kobiyama K, et al. The chemotherapeutic agent DMXAA as a
693
unique IRF3-dependent type-2 vaccine adjuvant. PLoS ONE 2013;8:e60038.
694
https://doi.org/10.1371/journal.pone.0060038.
695
[37] Su J, Brunner L, Oz EA, Sacherl J, Frank G, Kerth HA, et al. Activation of CD4 T cells during prime
696
immunization determines the success of a therapeutic hepatitis B vaccine in HBV-carrier mouse models. J
697
Hepatol 2023. https://doi.org/10.1016/j.jhep.2022.12.013.
698
[38] Germanó MJ, Giai C, Cargnelutti DE, Colombo MI, Blanco S, Konigheim B, et al. Receptorbinding
699
domainbased SARSCoV2 vaccine adjuvanted with cyclic diadenosine monophosphate enhances humoral
700
and cellular immunity in mice. J Med Virol 2023;95:e28584. https://doi.org/10.1002/jmv.28584.
701
[39] Dis EV, Sogi KM, Rae CS, Sivick KE, Surh NH, Leong ML, et al. STING-Activating Adjuvants Elicit a Th17
702
Immune Response and Protect against Mycobacterium tuberculosis Infection. Cell Reports 2018;23:143547.
703
https://doi.org/10.1016/j.celrep.2018.04.003.
704
[40] Hu D-L, Narita K, Hyodo M, Hayakawa Y, Nakane A, Karaolis DKR. c-di-GMP as a vaccine adjuvant
705
enhances protection against systemic methicillin-resistant Staphylococcus aureus (MRSA) infection. Vaccine
706
2009;27:486773. https://doi.org/10.1016/j.vaccine.2009.04.053.
707
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
28
[41] Yan H, Kuolee R, Tram K, Qiu H, Zhang J, Patel GB, et al. 3’,5’-Cyclic diguanylic acid elicits mucosal
708
immunity against bacterial infection. Biochemical and Biophysical Research Communications 2009;387:5814.
709
https://doi.org/10.1016/j.bbrc.2009.07.061.
710
[42] Alberti AS, Bivona AE, Cerny N, Schulze K, Weißmann S, Ebensen T, et al. Engineered trivalent
711
immunogen adjuvanted with a STING agonist confers protection against Trypanosoma cruzi infection. Npj
712
Vaccines 2017:111. https://doi.org/10.1038/s41541-017-0010-z.
713
[43] Cerón S, North BJ, Taylor SA, Leib DA. The STING agonist 5,6-dimethylxanthenone-4-acetic acid
714
(DMXAA) stimulates an antiviral state and protects mice against herpes simplex virus-induced neurological
715
disease. Virology 2019;529:238. https://doi.org/10.1016/j.virol.2019.01.006.
716
[44] Zhu Q, Hu H, Liu H, Shen H, Yan Z, Gao L. A synthetic STING agonist inhibits the replication of human
717
parainfluenza virus 3 and rhinovirus 16 through distinct mechanisms. Antivir Res 2020;183:104933104933.
718
https://doi.org/10.1016/j.antiviral.2020.104933.
719
[45] Li M, Ferretti M, Ying B, Descamps H, Lee E, Dittmar M, et al. Pharmacological activation of STING blocks
720
SARS-CoV-2 infection. Sci Immunol 2021;6. https://doi.org/10.1126/sciimmunol.abi9007.
721
[46] Humphries F, Shmuel-Galia L, Jiang Z, Wilson R, Landis P, Ng S-L, et al. A diamidobenzimidazole STING
722
agonist protects against SARS-CoV-2 infection. Sci Immunol 2021;6:eabi9002.
723
https://doi.org/10.1126/sciimmunol.abi9002.
724
[47] Sali TM, Pryke KM, Abraham J, Liu A, Archer I, Broeckel R, et al. Characterization of a Novel Human-
725
Specific STING Agonist that Elicits Antiviral Activity Against Emerging Alphaviruses. Plos Pathog
726
2015;11:e1005324. https://doi.org/10.1371/journal.ppat.1005324.
727
[48] Wang J, Meng F, Yeo Y. Delivery of STING agonists for cancer immunotherapy. Curr Opin Biotechnol
728
2024;87:103105. https://doi.org/10.1016/j.copbio.2024.103105.
729
[49] Berger G, Knelson EH, Jimenez-Macias JL, Nowicki MO, Han S, Panagioti E, et al. STING activation
730
promotes robust immune response and NK cellmediated tumor regression in glioblastoma models. Proc
731
National Acad Sci 2022;119:e2111003119. https://doi.org/10.1073/pnas.2111003119.
732
[50] Nicolai CJ, Wolf N, Chang I-C, Kirn G, Marcus A, Ndubaku CO, et al. NK cells mediate clearance of CD8+
733
T cellresistant tumors in response to STING agonists. Sci Immunol 2020;5.
734
https://doi.org/10.1126/sciimmunol.aaz2738.
735
[51] Wang-Bishop L, Wehbe M, Shae D, James J, Hacker BC, Garland K, et al. Potent STING activation
736
stimulates immunogenic cell death to enhance antitumor immunity in neuroblastoma. J Immunother Cancer
737
2020;8:e000282. https://doi.org/10.1136/jitc-2019-000282.
738
[52] Ghaffari A, Peterson N, Khalaj K, Vitkin N, Robinson A, Francis J-A, et al. STING agonist therapy in
739
combination with PD-1 immune checkpoint blockade enhances response to carboplatin chemotherapy in high-
740
grade serous ovarian cancer. British Journal of Cancer 2018;119:4409. https://doi.org/10.1038/s41416-018-
741
0188-5.
742
[53] Donnelly CR, Jiang C, Andriessen AS, Wang K, Wang Z, Ding H, et al. STING controls nociception via
743
type I interferon signalling in sensory neurons. Nature 2021;591:27580. https://doi.org/10.1038/s41586-020-
744
03151-1.
745
[54] Lemos H, Huang L, Chandler PR, Mohamed E, Souza GR, Li L, et al. Activation of the STING adaptor
746
attenuates experimental autoimmune encephalitis. Journal of Immunology (Baltimore, Md : 1950)
747
2014;192:55718. https://doi.org/10.4049/jimmunol.1303258.
748
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
29
[55] Johnson BM, Uchimura T, Gallovic MD, Thamilarasan M, Chou W-C, Gibson SA, et al. STING Agonist
749
Mitigates Experimental Autoimmune Encephalomyelitis by Stimulating Type I IFN-Dependent and -
750
Independent Immune-Regulatory Pathways. Journal of Immunology (Baltimore, Md : 1950) 2021;206:2015–28.
751
https://doi.org/10.4049/jimmunol.2001317.
752
[56] Lu D, Shang G, Li J, Lu Y, Bai X, Zhang X. Activation of STING by targeting a pocket in the
753
transmembrane domain. Nature 2022;604:55762. https://doi.org/10.1038/s41586-022-04559-7.
754
[57] Liu B, Carlson RJ, Pires IS, Gentili M, Feng E, Hellier Q, et al. Human STING is a proton channel. Science
755
2023;381:50814. https://doi.org/10.1126/science.adf8974.
756
[58] McFarland AP, Luo S, Ahmed-Qadri F, Zuck M, Thayer EF, Goo YA, et al. Sensing of Bacterial Cyclic
757
Dinucleotides by the Oxidoreductase RECON Promotes NF-κB Activation and Shapes a Proinflammatory
758
Antibacterial State. Immunity 2017;46:43345. https://doi.org/10.1016/j.immuni.2017.02.014.
759
[59] Chen J, Hao L, Li C, Ye B, Du Y, Zhang H, et al. The endoplasmic reticulum adaptor protein ERAdP
760
initiates NK cell activation via the Ubc13-mediated NF-κB pathway. Journal of Immunology (Baltimore, Md :
761
1950) 2015;194:1292303. https://doi.org/10.4049/jimmunol.1402593.
762
[60] Zhang Z, Ohto U, Shibata T, Taoka M, Yamauchi Y, Sato R, et al. Structural Analyses of Toll-like Receptor
763
7 Reveal Detailed RNA Sequence Specificity and Recognition Mechanism of Agonistic Ligands. Cell Rep
764
2018;25:3371-3381.e5. https://doi.org/10.1016/j.celrep.2018.11.081.
765
[61] Parvatiyar K, Zhang Z, Teles RM, Ouyang S, Jiang Y, Iyer SS, et al. The helicase DDX41 recognizes the
766
bacterial secondary messengers cyclic di-GMP and cyclic di-AMP to activate a type I interferon immune
767
response. Nature Publishing Group 2012;13:115561. https://doi.org/10.1038/ni.2460.
768
[62] Wang M, Chaudhuri R, Ong WWS, Sintim HO. c-di-GMP Induces COX-2 Expression in Macrophages in a
769
STING-Independent Manner. Acs Chem Biol 2021;16:166370. https://doi.org/10.1021/acschembio.1c00342.
770
[63] Takahama S, Ishige K, Nogimori T, Yasutomi Y, Appay V, Yamamoto T. Model for predicting age-
771
dependent safety and immunomodulatory effects of STING ligands in non-human primates. Mol Ther -
772
Methods Clin Dev 2023;28:99115. https://doi.org/10.1016/j.omtm.2022.12.008.
773
[64] Rudicell RS, Garinot M, Kanekiyo M, Kamp HD, Swanson K, Chou T, et al. Comparison of adjuvants to
774
optimize influenza neutralizing antibody responses. Vaccine 2019;37:620820.
775
https://doi.org/10.1016/j.vaccine.2019.08.030.
776
[65] Blaauboer SM, Mansouri S, Tucker HR, Wang HL, Gabrielle VD, Jin L. The mucosal adjuvant cyclic di-
777
GMP enhances antigen uptake and selectively activates pinocytosis-efficient cells in vivo. ELife 2015;4:900.
778
https://doi.org/10.7554/elife.06670.
779
[66] Kim S, Li L, Maliga Z, Yin Q, Wu H, Mitchison TJ. Anticancer flavonoids are mouse-selective STING
780
agonists. ACS Chemical Biology 2013;8:1396401. https://doi.org/10.1021/cb400264n.
781
[67] Mozzi A, Pontremoli C, Forni D, Clerici M, Pozzoli U, Bresolin N, et al. OASes and STING: adaptive
782
evolution in concert. Genome Biology and Evolution 2015;7:101632. https://doi.org/10.1093/gbe/evv046.
783
[68] Abraham J, Botto S, Mizuno N, Pryke K, Gall B, Boehm D, et al. Characterization of a Novel Compound
784
That Stimulates STING-Mediated Innate Immune Activity in an Allele-Specific Manner. Front Immunol
785
2020;11:1430. https://doi.org/10.3389/fimmu.2020.01430.
786
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
30
[69] Cavlar T, Deimling T, Ablasser A, Hopfner K-P, Hornung V. Species-specific detection of the antiviral
787
small-molecule compound CMA by STING. The EMBO Journal 2013;32:144050.
788
https://doi.org/10.1038/emboj.2013.86.
789
[70] Li J, Canham SM, Wu H, Henault M, Chen L, Liu G, et al. Activation of human STING by a molecular glue-
790
like compound. Nat Chem Biol 2024;20:36572. https://doi.org/10.1038/s41589-023-01434-y.
791
[71] Livak KJ, Schmittgen TD. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR
792
and the 2−ΔΔC T Method. Methods 2001;25:402–8. https://doi.org/10.1006/meth.2001.1262.
793
[72] Hulsen T, Vlieg J de, Alkema W. BioVenn - a web application for the comparison and visualization of
794
biological lists using area-proportional Venn diagrams. BMC Genomics 2008;9:488.
795
https://doi.org/10.1186/1471-2164-9-488.
796
[73] Broeckel RM, Haese N, Ando T, Dmitriev I, Kreklywich CN, Powers J, et al. Vaccine-Induced Skewing of T
797
Cell Responses Protects Against Chikungunya Virus Disease. Front Immunol 2019;10:2563.
798
https://doi.org/10.3389/fimmu.2019.02563.
799
800
801
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
31
Funding 802
This research was supported by NIH R01 AI143660, R01 AI143660, and HHSN272201400055C. 803
CRediT authorship contribution statement 804
Victor R. DeFilippis: Writing original draft, Validation, Methodology, Formal analysis, Conceptualization, 805
Supervision, Funding acquisition. Nobuyo Mizuno: Writing Methodology, Investigation, Formal 806
analysis. Dylan Boehm: Investigation, Formal analysis. Kevin Jimenez-Perez: Investigation, Formal 807
analysis. Jinu Abraham: Investigation, Formal analysis. Laura Springgay: Investigation, Formal analysis. Ian 808
Rose: Investigation, Formal analysis. 809
Declaration of competing interest 810
The authors declare no financial interests/personal relationships which may be considered as potential 811
competing interests. 812
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
A
B
Figure 1. Dose-dependent innate responses induced by STING agonists in murine cell lines. RAW264.7 (A) and J774 (B, C, D) were grown to confluence in 96 well plates and
exposed in duplicate overnight to CDA, DMXAA, or diABZI at indicated molarities. A. Cells were exposed to either Quanti-Luc or Cell Titer Glo lysis buffer to measure IFN-I signaling
induction or cell viability, respectively. Data presented are mean ± SEM relative luminescence units (RLU, black) or percent viability (red) determined relative to control cells treated with
DMSO- or transfection reagent (CDA); C. NF-B-dependent SEAP expression expressed as mean ± SEM absorbance at OD655 for indicated treatment relative to control treated cells; D.
Secretion of IL-1from primed J774 cells treated with indicated doses of DMXAA, diABZI, or CDA. Data displayed are mean ± SEM IL-1 pg/mL secreted into culture media. Two-way
ANOVA with Tukey’s correction for multiple comparisons was used to examine statistical significance of dose-specific activation of NF-B or IL-1 secretion between agonists (*p < 0.05,
**p < 0.01, ***p < 0.001, ****p < 0.0001 color coded as indicated for agonist pairing).
C
D
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
Figure 2. Maturation of Dendritic Cells from C57Bl/6 Mice in Response to Innate Stimuli. Immature bone marrow
derived DC (BMDC) from C57Bl/6 mice were treated with 0.5% DMSO, 1 µM diABZI, 50 μM DMXAA, 1 µM CDA, 100 ng/mL
LPS, or 3 µg/mL Alum as indicated. Cells were collected after 24 h and stained for expression of CD40, CD80, CD86 and
MHC-II and analyzed by flow cytometry. A. Mean fluorescence intensities (MFI) ± SD for indicated treatment and surface
marker. One way ANOVA with Tukey’s multiple comparisons test was used to compare results between treatments (***p <
0.001****; p < 0.0001); B. Representative MFI histograms for each marker are shown for cells exposed to indicated stimulus.
B
CD40 MFI CD80 MFI CD86 MFI MHC-II MFI
DMSO
Counts
CDA
diABZI
DMXAA
LPS
Alum
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
PBS
diABZI
CDA
DMXAA
Figure 3. Induction of systemic cytokine secretion by STING agonists in vivo. C57Bl/6 mice were injected IM with PBS, 30 µg diABZI, 10 µg, CDA, or 500 µg DMXAA and serum
harvested at 24 h. Luminex bead based multiplex assay was then used to measure absolute levels of indicated cytokines. Data presented are mean ± SD pg/mL. One way ANOVA with
Dunnet correction for multiple comparisons was used to determine statistical significance (**p <0.01, ***p < 0.001, ***p < 0.0001).
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
*
*
*
**
**
*
**
*
**
*
*
*
*
**
*
**
*
*
*
**
**
*
*
*
**
*
**
*
*
*
*
**
**
*
*
*
*
**
*
*
*
**
**
Vs.
Figure 4. STING agonist-induced transcriptomic changes in vivo. C57Bl/6 mice were injected intramuscularly with PBS, CDA, diABZI or DMXAA and total RNA from draining lymph nodes
harvested at indicated time point. A. Levels of indicated mRNA transcript in draining lymph nodes at 5 h, 24 h, or 72 h for indicated treatment as determined by qPCR. Data presented are
mean ± SD fold changes relative to PBS treated control animals. Statistical significance was determined using a mixed effects ANOVA with Tukey’s correction for multiple comparisons of log
transformed fold changes between stimuli at each time point (*p < 0.05; **p < 0.01); B. Number of mRNAs significantly up or downregulated more than twofold relative to PBS treatment
following indicated treatment as determined by hybridization array; C. Principal components analysis of transcript fold changes for indicated stimulus and time point; D. Venn diagram of
transcripts significantly upregulated for indicated stimulus at 5 h and 24 h; E. Signaling pathways displaying a predicted Z score > 1 for all three stimuli at 5 h and 24 h as determined by
Ingenuity Pathway Analysis; F. Pathways displaying a predicted Z score > 1 for following DMXAA treatment only at 5 h and 24 h.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
CDA 5h
diABZI 5h
DMXAA 5h
CDA 24h
diABZI 24h
DMXAA 24h
0
1000
2000
3000
4000
5000
6000
# Transcripts
mRNA Regulation
Down
Up
CDA
diABZI
DMXAA
CDA
diABZI
DMXAA
5 h 24 h
BCD
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
5 h
24 h
E
F
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
B Cells
T Cells
cDC1
cDC2 pDC Neutrophils Macrophages
A
B
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
IFNAR-/-
C57Bl/6
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
OVA
OVA + DMXAA
C
Figure 5. STING agonist-induced changes in draining lymph node immune cell populations. In duplicate experiments C57Bl/6 mice were injected intramuscularly with OVA in the
presence of PBS, CDA, diABZI DMXAA, or Alum and single cell suspensions of draining lymph nodes obtained at 24 h post treatment. Flow cytometry was then used to quantify indicated cell
types. A. Total number of lymph node cells following indicated treatment; B. Percent of total lymph node cells represented by indicated B cells, T cells, cDC1, cDC2, pDC, neutrophils, and
macrophages as indicated; C. Percent of total lymph node cells represented by indicated B cells, T cells, cDC1, cDC2, pDC, neutrophils, and macrophages in WT and IFNAR-/- mice as
indicated. Statistical significance was determined using ANOVA with Dunnet’s correction for multiple comparisons (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001). Individual experiments
indicated by circle color.
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
PBS
CDA
diABZI
DMXAA
Anti-OVA Total IgG Anti-OVA IgG1 Anti-OVA IgG2c
Alum
Anti-OVA IgG2c
Anti-OVA IgG1
101102103104105106
0.0
0.5
1.0
1.5
2.0
Dilutions
Abs490
IgG1- Absorbance
OVA + PBS
OVA + CDA
OVA + diABZI
OVA + DMXAA
OVA + Alum
Naive
101102103104105106
0.0
0.5
1.0
1.5
2.0
2.5
3.0
Dilutions
Abs490
IgG2c- Absorbance
OVA + PBS
OVA + CDA
OVA + diABZI
OVA + DMXAA
OVA + Alum
Naive
Anti-OVA Total IgG
IgG2c:IgG1
AUC Ratio
PBS
CDA
diABZI
DMXAA
Alum
PBS
CDA
diABZI
DMXAA
Alum
PBS
CDA
diABZI
DMXAA
Alum
A
Figure 6. Enhancement of humoral immune
response to OVA by co-administered
STING agonists. C57Bl/6 mice were
vaccinated IM with OVA in the presence of
PBS, CDA, diABZI, DMXAA, or Alum,
identically boosted 2 w later and harvested 2
w later. A. Absorbance of serum antibodies
reactive to OVA as determined by ELISA. Data
presented are mean + SEM raw 490
absorbance over indicated serum dilution for
IgG, IgG1, and IgG2c; B. Antibody titers as
determined by area under the curve (AUC)
calculations for each adjuvant. Data presented
are mean + SEM AUC including individual
animal measurements; C. Mean + SD ratio of
IgG2c to IgG1 AUC titers; D. Number of OVA-
reactive TFH (CD4+CXCR5+PD-1+) splenocytes
harvested seven days after boost with OVA +
indicated adjuvant. Statistical significance was
determined using ANOVA with Dunnet’s
correction for multiple comparisons (*p < 0.05;
**p < 0.01; ***p < 0.001; ****p < 0.0001).
Log AUC
B
C
D
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
Figure 7. STING-Adjuvant Associated Enhancement of Cell Mediated Responses. C57Bl/6 mice were left untreated or vaccinated IM using a prime/boost strategy with OVA in the presence of PBS, CDA, diABZI, DMXAA, or Alum. A. Splenocytes were
harvested at 7 d post boost and stimulated ex vivo with a peptide pool spanning the OVA protein (left) or SIINFEKL peptide (right). ELISpot was then used to quantify the number of IFN positive cells. Data presented are mean ± SD number of IFN positive
spot forming units (SFU) per 2.5 x 105 total cells. Results from individual animals are also presented with duplicate experiments represented by marker color; B. Intracellular cytokine staining and flow cytometry was used to quantify OVA peptide pool-
stimulated expression of IFN and TNF from CD4+ and CD8+ T cells in splenocytes. Data presented are mean ± SD percentage of CD8+ (left) or CD4+ (right) T cells that display positivity for indicated cytokine. Individual animals represented by circles. One
way ANOVA using Tukeys multiple comparison was used to determine statistical significance (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001).
PBS
CDA
diABZI
DMXAA
Alum
Naive
PBS
CDA
diABZI
DMXAA
Alum
Naive
PBS
CDA
diABZI
DMXAA
Alum
Naive
PBS
CDA
diABZI
DMXAA
Alum
Naive
PBS
CDA
diABZI
DMXAA
Alum
Naive
PBS
CDA
diABZI
DMXAA
Alum
Naive
A
B
.CC-BY-NC-ND 4.0 International licenseavailable under a
(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made
The copyright holder for this preprintthis version posted February 27, 2025. ; https://doi.org/10.1101/2025.02.21.639458doi: bioRxiv preprint
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Stimulator of interferon genes (STING) is a dimeric transmembrane adapter protein that plays a key role in the human innate immune response to infection and has been therapeutically exploited for its antitumor activity. The activation of STING requires its high-order oligomerization, which could be induced by binding of the endogenous ligand, cGAMP, to the cytosolic ligand-binding domain. Here we report the discovery through functional screens of a class of compounds, named NVS-STGs, that activate human STING. Our cryo-EM structures show that NVS-STG2 induces the high-order oligomerization of human STING by binding to a pocket between the transmembrane domains of the neighboring STING dimers, effectively acting as a molecular glue. Our functional assays showed that NVS-STG2 could elicit potent STING-mediated immune responses in cells and antitumor activities in animal models.
Article
Full-text available
TLR agonists have emerged as an efficient cancer vaccine adjuvant system that induces robust immune responses. L-pampo™, a proprietary vaccine adjuvant of TLR2 and TLR3 agonists, promotes strong humoral and cellular immune responses against infectious diseases. In this study, we demonstrate that vaccines formulated with L-pampo™ affect the recruitment and activation of dendritic cells (DCs) in draining lymph nodes (dLNs) and leading to antigen-specific T-cell responses and anti-tumor efficacy. We analyzed DC maturation and T-cell proliferation using flow cytometry and ELISA. We determined the effect of L-pampo™ on DCs in dLNs and antigen-specific T-cell responses using flow cytometric analysis and the ELISPOT assay. We employed murine tumor models and analyzed the anti-tumor effect of L-pampo™. We found that L-pampo™ directly enhanced the maturation and cytokine production of DCs and, consequently, T-cell proliferation. OVA or OVA peptide formulated with L-pampo™ promoted DC migration into dLNs and increased activation markers and specific DC subsets within dLNs. In addition, vaccines admixed with L-pampo™ promoted antigen-specific T-cell responses and anti-tumor efficacy. Moreover, the combination of L-pampo™ with an immune checkpoint inhibitor synergistically improved the anti-tumor effect. This study suggests that L-pampo™ can be a potent cancer vaccine adjuvant and a suitable candidate for combination immunotherapy.
Article
Full-text available
Novel adjuvants are highly desired to improve immune responses of SARS-CoV-2 vaccines. This work reports the potential of the stimulator of interferon genes (STING) agonist adjuvant, the cyclic di-adenosine monophosphate (c-di-AMP), in a SARS-CoV-2 vaccine based on the receptor binding domain (RBD). Here, mice immunized with two doses of monomeric RBD adjuvanted with c-di-AMP intramuscularly were found to exhibit stronger immune responses compared to mice vaccinated with RBD adjuvanted with aluminium hydroxide (Al(OH)3) or without adjuvant. After two immunizations, consistent enhancements in the magnitude of RBD-specific IgG antibody response were observed by RBD+c-di-AMP (mean:15360) compared to RBD+Al(OH)3 (mean:3280) and RBD alone (n.d.). Analysis of IgG subtypes indicated a predominantly Th1-biased immune response (IgG2c, mean:14480; IgG2b, mean:1040, IgG1, mean:470) in mice vaccinated with RBD+c-di-AMP compared to a Th2-biased response in those vaccinated with RBD+Al(OH)3 (IgG2c, mean:60; IgG2b: n.d.; IgG1, mean:16660). In addition, the RBD+c-di-AMP group showed better neutralizing antibody responses as determined by pseudovirus neutralization assay and by plaque reduction neutralization assay with SARS-CoV-2 WT. Moreover, the RBD+c-di-AMP vaccine promoted IFN-γ secretion of spleen cells cultures after RBD stimulation. Furthermore, evaluation of IgG-antibody titers in aged-mice showed that di-AMP was able to improve RBD-immunogenicity at old-age after 3 doses (mean:4000). These data suggest that c-di-AMP improves immune responses of a SARS-CoV-2 vaccine based on RBD, and would be considered a promising option for future COVID-19 vaccines. This article is protected by copyright. All rights reserved.
Article
Full-text available
Vaccine adjuvant research is being fueled and driven by progress in the field of innate immunity that has significantly advanced in the past two decades with the discovery of countless innate immune receptors and innate immune pathways. Receptors for pathogen-associated molecules (PAMPs) or host-derived, danger-associated molecules (DAMPs), as well as molecules in the signaling pathways used by such receptors, are a rich source of potential targets for agonists that enable the tuning of innate immune responses in an unprecedented manner. Targeted modulation of immune responses is achieved not only through the choice of immunostimulator – or select combinations of adjuvants – but also through formulation and systematic modifications of the chemical structure of immunostimulatory molecules. The use of medium and high-throughput screening methods for finding immunostimulators has further accelerated the identification of promising novel adjuvants. However, despite the progress that has been made in finding new adjuvants through systematic screening campaigns, the process is far from perfect. A major bottleneck that significantly slows the process of turning confirmed or putative innate immune receptor agonists into vaccine adjuvants continues to be the lack of defined in vitro correlates of in vivo adjuvanticity. This brief review discusses recent developments, exciting trends, and notable successes in the adjuvant research field, albeit acknowledging challenges and areas for improvement.
Article
Stimulator of interferon genes (STING) is activated in many pathophysiological conditions, leading to TBK1-dependent interferon production in higher organisms. However, primordial functions of STING independent of TBK1 are poorly understood. Here, through proteomics and bioinformatics approaches, we identify lyso-somal biogenesis as an unexpected function of STING. Transcription factor EB (TFEB), an evolutionarily conserved regulator of lysosomal biogenesis and host defense, is activated by STING from multiple species, including humans, mice, and frogs. STING-mediated TFEB activation is independent of TBK1, but it requires STING trafficking and its conserved proton channel. GABARAP lipidation, stimulated by the channel of STING, is key for STING-dependent TFEB activation. STING stimulates global upregulation of TFEB-target genes, mediating lysosomal biogenesis and autophagy. TFEB supports cell survival during chronic sterile STING activation, a common condition in aging and age-related diseases. These results reveal a primordial function of STING in the biogenesis of lysosomes, essential organelles in immunity and cellular stress resistance.
Article
Proton leakage from organelles is a common signal for noncanonical light chain 3B (LC3B) lipidation and inflammasome activation, processes induced upon stimulator of interferon genes (STING) activation. On the basis of structural analysis, we hypothesized that human STING is a proton channel. Indeed, we found that STING activation induced a pH increase in the Golgi and that STING reconstituted in liposomes enabled transmembrane proton transport. Compound 53 (C53), a STING agonist that binds the putative channel interface, blocked STING-induced proton flux in the Golgi and in liposomes. STING-induced LC3B lipidation and inflammasome activation were also inhibited by C53, suggesting that STING's channel activity is critical for these two processes. Thus, STING's interferon-induction function can be decoupled from its roles in LC3B lipidation and inflammasome activation.
Article
Subunit proteins provide a safe source of antigens for vaccine development especially for intracellular infections which require the induction of strong cellular immune responses. However, those antigens are often limited by their low immunogenicity. In order to achieve effective immune responses, they should be encapsulated into a stable antigen delivery system combined with an appropriate adjuvant. As such cationic liposomes provide an efficient platform for antigen delivery. In the present study, we describe a liposomal vaccine platform for co-delivery of antigens and adjuvants able to elicit strong antigen-specific adaptive immune responses. Liposomes are composed of the cationic lipid dimethyl dioctadecylammonium bromide (DDAB), cholesterol (CHOL) and oleic acid (OA). Physicochemical characterization of the formulations showed that their size was in the range of ~250 nm with a positive zeta potential which was affected in some cases by the enviromental pH facilitating endosomal escape of potential vaccine cargo. In vitro, liposomes were effectively taken up by bone marrow dendritic cells (BMDCs) and when encapsulated IMQ they promoted BMDCs maturation and activation. Upon in vivo intramuscular administration, liposomes' active drainage to lymph nodes was mediated by DCs, B cells and macrophages. Thus, mice immunization with liposomes having encapsulated LiChimera, a previously characterized anti-leishmanial antigen, and IMQ elicited infiltration of CD11blow DCs populations in draining LNs followed by increased antigen-specific IgG, IgG2a and IgG1 levels production as well as indcution of antigen-specific CD4+ and CD8+ T cells. Collectively, the present work provides a proof-of-concept that cationic liposomes composed of DDAB, CHOL and OA adjuvanted with IMQ provide an efficient delivery platform for protein antigens able to induce strong adaptive immune responses via DCs targeting and induction of maturation.
Article
Background & Aims Therapeutic vaccination represents a promising approach to cure HBV. We employ a heterologous therapeutic vaccination scheme (TherVacB) comprising a particulate protein-prime followed by modified vaccinia-virus Ankara (MVA) vector-boost. The key determinants required to overcome HBV-specific immune tolerance, however, remained unclear. Here, we unravel the essential role of CD4 T-cell activation during the priming phase for antiviral efficacy of TherVacB. Methods Recombinant hepatitis B surface (HBsAg) and core antigen (HBcAg) particles were formulated with different liposome-based or oil-in-water emulsion combination adjuvants containing saponin QS21 and monophosphoryl lipid A (MPL) and compared them to STING-agonist c-di-AMP and conventional alum formulation. Immunogenicity and antiviral effects of protein antigen formulations and the vector boost within TherVacB was evaluated in AAV-HBV infected and HBV-transgenic mice. Results Combination adjuvant formulations preserved HBsAg and HBcAg integrity for ≥ 12 weeks, promoted human and mouse dendritic cell activation and within TherVacB elicited robust HBV-specific antibody and T-cell responses in wild-type and HBV-carrier mice. Combination adjuvants priming a balanced HBV-specific type 1 and 2 T helper response induced high-titer anti-HBs antibodies, cytotoxic T-cell responses and long-term control of HBV. Lack of a T-cell booster by the MVA vector as well selective CD8 T-cell depletion allowed for a drop of HBsAg mediated mainly by anti-HBs antibodies but resulted in a lack of HBV control. Selective CD4 T-cell depletion during the priming phase of TherVacB resulted in a complete loss of vaccine-induced immune responses and its therapeutic anti-viral effect in mice. Conclusions Our results identify CD4 T-cell activation during the priming phase as a key determinant for HBV-specific antibody and CD8 T cell immunity using TherVacB as targeted therapeutic vaccine. Impact and Implications Therapeutic vaccination is a promising, potentially curative treatment for chronic hepatitis B, but the factors essential to break immune tolerance in HBV carriers and determinants of success remain unclear. Our study provides the first direct evidence that correct priming of HBV-specific CD4 T cells preparing a booster of CD8 T-cell responses determines the success of therapeutic hepatitis B vaccination. Understanding how to fine-tune therapeutic vaccines by the selection of appropriate vaccine components proved essential for its antiviral effect in two preclinical HBV-carrier mouse models and may help to guide the clinical development of therapeutic vaccines for chronic hepatitis B. Lay summary Therapeutic vaccination is a promising, potentially curative treatment for chronic hepatitis B. Which factors are essential for breaking immune tolerance in HBV carriers and determine a successful outcome of therapeutic vaccination, however, remains unclear. Our study provides the first direct evidence that efficient priming of HBV-specific CD4 T cells determines the success of therapeutic hepatitis B vaccination in two preclinical HBV-carrier mouse models. Applying an optimal formulation of HBV antigens that allows activating CD4 and CD8 T cells during prime immunization provided the foundation for an antiviral effect of therapeutic vaccination, while depletion of CD4 T cells lead to a complete loss of vaccine-induced antiviral efficacy. Boosting CD8 T cells was important to finally control HBV in these mouse models. Our findings provide important insights into the rational design of therapeutic vaccines to cure chronic hepatitis B.