ArticlePDF Available

Delivery of STING Agonist Using Polypeptide‐Modified Dendrimer Nanoparticles in the Treatment of Melanoma

Wiley
Advanced NanoBioMed Research
Authors:

Abstract and Figures

The activation of stimulator of interferon genes (STING) in the cytosol by cyclic dinucleotides (CDNs) enhances antitumor immunity through the induction of pro‐inflammatory cytokines, such as type‐I interferons (IFN‐I). However, the high hydrophilicity and negative charge of CDNs hinders their delivery into cells. Here, by developing a library of cationic polypeptide‐modified dendrimers, we show that CDNs can be efficiently delivered intracellularly in vitro and in vivo. With respect to naked dendrimers, generation‐5 polyamidoamine (G5‐PAMAM) dendrimers modified with arginine or with a mixture of arginine/lysine polypeptides afforded higher CDN‐packaging capacity and led to higher activation of IFN‐I and the nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NF‐κB) pro‐inflammatory signaling pathway. In the B16‐F10 murine model of melanoma, the intratumoral administration of a synthetic CDN via arginine‐modified G5‐PAMAM dendrimers at a low dose induced strong antitumor responses and inhibited tumor growth. We also show that the combination of this therapy with immune checkpoint blockade (ICB) further improves the therapeutic outcomes. Cationic polypeptide dendrimers may be advantageous in the delivery of gene‐based immunomodulators for the treatment of solid tumors. This article is protected by copyright. All rights reserved.
This content is subject to copyright. Terms and conditions apply.
Delivery of Stimulator of Interferon Genes (STING) Agonist
Using Polypeptide-Modied Dendrimer Nanoparticles in
the Treatment of Melanoma
Pere Dosta, Alexander M. Cryer, Michaela Prado, Michelle Z. Dion, Shiran Ferber,
Santhosh Kalash, and Natalie Artzi*
1. Introduction
Using the innate immune system to
instigate an antitumor response is an
increasingly attractive approach in cancer
immunotherapy. Engagement of pathogen-
associated molecular patterns (PAMPs)
with their respective pattern recognition
receptors (PRRs) was reported to elicit robust
downstream endogenous cytokine produc-
tion and immune cell activation,
[1]
which
is responsible for the potent immune
responses generated by vaccines and against
tumors. Cyclic dinucleotides (CDNs), such
as the second messenger 2 030-cyclic guano-
sine monophosphate-adenosine monophos-
phate (cGMP-AMP or cGAMP), are a class
of PAMPs that are generated upon sensing
cytosolic DNA.
[2]
The production of cGAMP
leads to agonism of stimulator of interferon
genes (STING),
[3]
enacting a type-I inter-
feron (IFN-I)-driven proinammatory
program including the stimulation of
dendritic cells (DCs) and cross-presentation of tumor antigens
to T-cells, thereby priming them for antitumor effector functional-
ity.
[4]
This bridge between innate and adaptive antitumor immunity
positions STING as a critical regulator of immunosurveillance,
reinforced by studies in STING-decient mice highlight-
ing increased susceptibility to tumorigenesis and diminished
responsiveness to immunotherapy such as immune checkpoint
inhibitors (ICIs).
[5]
The role of STING signaling in antitumor responses as well as
insufcient endogenous agonism has prompted investigations
into exogenous cGAMP and structural analogs as therapeutics
to promote antitumor immunity.
[6]
Intratumoral (IT) injection
of CDNs has reached phase I clinical trials;
[7]
however, CDNs
are anionic and highly hydrophilic, which restricts their entry into
the cytoplasm where STING resides.
[8]
Consequently, CDNs have
transient interactions with immune cells (e.g., DCs, macrophages)
in the tumor microenvironment (TME) and are rapidly eliminated
from the tumor site.
Biomaterial-based delivery strategies can be leveraged to
improve internalization into cells, therefore augmenting the
activity of adjuvants such as STING agonists.
[9]
Indeed, it has
previously been demonstrated that using nanoparticles (NPs)
of lipidic
[10]
or polymeric
[11]
origin to encapsulate CDNs can
Dr. P. Dosta, Dr. A. M. Cryer, M. Prado, M. Z. Dion, Dr. S. Ferber,
Dr. S. Kalash, Dr. N. Artzi
Institute for Medical Engineering and Science
Massachusetts Institute of Technology
Cambridge, MA 02139, USA
E-mail: nartzi@mit.edu, nartzi@bwh.harvard.edu
Dr. P. Dosta, Dr. A. M. Cryer, M. Prado, M. Z. Dion, Dr. S. Ferber,
Dr. S. Kalash, Dr. N. Artzi
Department of Medicine
Division of Engineering in Medicine
Brigham and Womens Hospital
Harvard Medical School
Boston, MA 02115, USA
The ORCID identication number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/anbr.202100006.
© 2021 The Authors. Advanced NanoBiomed Research published by
Wiley-VCH GmbH. This is an open access article under the terms of
the Creative Commons Attribution License, which permits use,
distribution and reproduction in any medium, provided the original
work is properly cited.
DOI: 10.1002/anbr.202100006
The activation of stimulator of interferon genes (STING) in the cytosol by cyclic
dinucleotides (CDNs) enhances antitumor immunity through the induction of
proinammatory cytokines, such as type-I interferons (IFN-I). However, the high
hydrophilicity and negative charge of CDNs hinders their delivery into cells. Here,
by developing a library of cationic polypeptide-modied dendrimers, it is shown
that CDNs can be efciently delivered intracellularly in vitro and in vivo. With
respect to naked dendrimers, generation-5 polyamidoamine (G5-PAMAM) den-
drimers modied with arginine or with a mixture of arginine/lysine polypeptides
affords higher CDN-packaging capacity and leads to higher activation of IFN-I
and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)
proinammatory signaling pathway. In the B16-F10 murine model of melanoma,
the intratumoral administration of a synthetic CDN via arginine-modied G5-
PAMAM dendrimers at a low dose induces strong antitumor responses and
inhibits tumor growth. It is also shown that the combination of this therapy with
immune checkpoint blockade (ICB) further improves the therapeutic outcomes.
Cationic polypeptide dendrimers may be advantageous in the delivery of gene-
based immunomodulators for the treatment of solid tumors.
RESEARCH ARTICLE
www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (1 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
improve their antitumor activity. However, liposomes suffer
from poor packaging capacity and limited storage stability and
the encapsulation of small hydrophilic molecules within poly-
meric NPs remains challenging.
[12]
We reasoned that stable elec-
trostatic complexes could be formed between anionic CDNs and
cationic NPs, which would act to improve their stability, cytosolic
localization, and endosomal escape, resulting in greater antitu-
mor responses.
Dendrimers have an established history as drug delivery
vehicles
[13]
and have been used in immunotherapy as accessories
in antibodydrug conjugates or in vaccine formulations.
[14]
Polyamidoamine (PAMAM) dendrimers are hyperbranched
polymers consisting of tertiary amines throughout the den-
drimer core and terminal primary amines, the number of which
increases with each layer of branching, known as a generation.
[15]
This dened architecture yields cationic, low- polydispersity NPs
with many surface-reactive sites per NP. It has been noted that
highly localized cationicity is not well tolerated on a cellular
level,
[16]
with the external primary amines being the principal
drivers of this toxicity. Therefore, we leveraged the reactivity
of these amines to functionalize dendrimers with basic polypep-
tide motifs consisting of arginine, lysine, or histidine. By replac-
ing external amines with biocompatible amino acids, the
cytotoxicity is signicantly attenuated, cellular internalization
and transfection efciency are improved, and endosomal escape
can be better controlled.
[17]
In addition, it has been demonstrated
that these polypeptides can inuence the nanoparticle composi-
tion, allowing the design of cell-specic nanoparticle formula-
tions.
[18]
Endowed with these properties, we hypothesized that
polypeptide-functionalized dendrimers could effectively complex
with CDNs and be internalized by immune cells, enhancing
CDN activity in vitro and in vivo. We investigated the physico-
chemical properties of the polypeptide-modied dendrimers,
their ability to deliver the synthetic CDN ADU-S100 to immune
cells, biocompatibility, cellular uptake, and antitumor efcacy
in vivo, as well as the associated immune responses.
2. Results
2.1. Synthesis of Polypeptide-Modied Dendrimers
We developed a library of dendrimers with diverse surface mod-
ications using cationic polypeptides that can complex with CDNs
and form a wide range of NPs with distinct physicochemical
properties. Synthesis of polypeptide-conjugated dendrimers was
performed via a two-step reaction. First, the PAMAM G5 den-
drimer was modied with a bifunctional succinimidyl-[(N-malei-
midopropionamido)-diethyleneglycol]-ester linker [SM(PEG)
2
]by
conjugating the primary dendrimer amines to the succinimidyl
group. Then, the modied dendrimers were conjugated with
cationic polypeptide moieties through sulfhydrylmaleimide con-
jugation of the thiol group of cysteine-terminated polypeptides to
the maleimide group of the SM(PEG)
2
linker, obtaining around 79
peptides per dendrimer (Figure 1A and Figure S9, Supporting
Information). Polypeptide-modied dendrimers were puried by
dialysis and their molecular structure characterized by
1
H-NMR.
The chemical structure of the resultant dendrimerswas conrmed
by the presence of signals associated with the conjugated polypep-
tides (Figure S1S8, Supporting Information).
To assess the effect of the polypeptides on our dendrimer for-
mulations, size (hydrodynamic diameter) and surface charge val-
ues were obtained using dynamic light scattering (DLS) and zeta
potential measurements, respectively (Figure 1). Whereas the
unmodied dendrimers were small (5.47 0.05 nm) and posi-
tively charged (14.77 6.32 mV), the end-modied dendrimers
were larger, ranging from 7 to 10 nm, and those modied with
arginine (D-CR3), lysine (D-CK3), and a mixture of lysine/histi-
dine (D-K/H) and arginine/lysine (D-R/K) showed an increase in
surface charge compared to nonmodied dendrimers (ranging
from þ24 mV to þ31 mV). In contrast, dendrimers modied
with histidine (D-CH3) or a mixture of histidine/arginine
(D-R/H) were less positive (þ10 mV) than nonmodied
dendrimers (þ15 mV). In addition, when the dendrimer
was only modied with a bifunctional SM(PEG)
2
linker (without
polypeptides), the zeta potential was markedly more negative
(7.07 0.34 mV). The addition of different polypeptides can
be used to tune the nal nanoparticle surface charge, ranging
from neutral to positive.
2.2. Formulation and Biophysical Characterization of
Polypeptide-Modied Dendrimers
The CDN complexation efcacy of the newly synthesized
dendrimers was evaluated by agarose gel electrophoresis at dif-
ferent dendrimer/CDN ratios (w/w) (Figure 2 and Figure S10,
Supporting Information). A uorescently labeled CDN [termed
CDN-F; c-(Ap-8-Fluo-AET-Gp)] was used to assess the complexa-
tion efciency. Complexes prepared with unmodied dendrimer
and uorescent CDN (CDN-F) revealed free CDN-F at ratios
below 64:1 dendrimer:CDN-F (w/w), while at a ratio of 64:1
complete CDN-F complexation was observed. D-CR3 showed
complete CDN-F retardation at 16:1 D-CR3/CDN-F ratios, sug-
gesting a more efcient complexation of CDN-F (0.5 CDN-F mol-
ecules per unmodied dendrimer vs 2 CDN-F molecules per
D-CR3). In contrast, D-CK3 required dendrimer-to-CDN-F ratios
similar to unmodied PAMAM dendrimer to achieve full CDN-F
complexation. Moreover, D-CH3 was not able to complex the
CDN-F at any of the tested ratios (Figure S10, Supporting
Information). These results suggest that, compared to unmodi-
ed PAMAM dendrimer, D-CR3 signicantly increases the num-
ber of CDN molecules per dendrimer NP, whereas dendrimers
modied with lysine and histidine polypeptides contain a similar
or lower number of CDN molecules per dendrimer NP than the
unmodied dendrimer.
2.3. In Vitro Selection of Polypeptide-Modied Dendrimer for
Efcient ADU-S100 Delivery
Given the importance of interferon regulatory factor (IRF)3 and
nuclear factor (NF)-κBinefcient STING signaling, we assessed
the ability of functionalized dendrimers to deliver ADU-S100 and
stimulate IRF3 and NF-κB responses in human monocytic THP-
1 Dual cells (Figure 3). THP-1 Dual cells allow the simultaneous
study of the IRF3 pathway, by assessing the activity of a secreted
luciferase, and the NF-κB pathway, by monitoring
www.advancedsciencenews.com www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (2 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
Figure 1. Synthesis and characterization of polypeptide-modied dendrimers. A) Different polypeptides were used for the synthesis of a new family of
end-modied dendrimers (arginine-modied dendrimer, D-CR3; lysine-modied dendrimer, D-CK3; histidine-modied dendrimer, D-CH3; 50% arginine
50% histidine modied dendrimer, D-R/H; 50% lysine50% histidine modied dendrimer, D-K/H; and 50% arginine50% lysine modied dendrimer,
D-R/K). B) Size (determined by DLS) and (C) Z-potential measurements of end-modied dendrimers (n¼3); data shown as mean SD.
Figure 2. Agarose retardation assay of polypeptide-modied dendrimerCDN-F polyplexes. Polyplexes were formed using CDN-F and different
polypeptide-modied dendrimers at indicated w/w ratios and loaded onto an agarose gel to assess CDN-F mobility by electrophoresis for
A) nonmodied dendrimer and B) D-CR3 formulation.
www.advancedsciencenews.com www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (3 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
the activity of secreted embryonic alkaline phosphatase (SEAP).
D-CR3 and D-R/K achieved higher IRF3 activation than the
unmodied dendrimer. In contrast, D-CH3 or D-K/H showed
the lowest level of activation, similar to that of free ADU-S100.
D-CK3 or D-R/H showed high IRF3 activation at higher
ADU-S100 doses (500 n
M
). A similar trend was seen in terms
of NF-κB activation for all formulations (Figure 3B). A 16-fold
NF-κB increase was observed when ADU-S100 was delivered
using either the D-CR3 or D-R/K formulation at 250 n
M
. In con-
trast, no NF-κB activation was detected when D-CH3 or D-K/H
was used, and NF-κB expression was only detected at high
ADU-S100 doses when D-CK3 and D-R/H formulations were
used. In addition, we did not observe activation of IRF3 or
NF-κB in STING-decient THP-1 Dual cells (Figure S12,
Supporting Information).
To determine if cellular internalization could account for
the differences in IRF3 and NF-κB, uptake of polypeptide-
modied dendrimers was assessed in THP-1 cells (Figure 3C).
Dendrimers were uorescently labeled, and their uptake was
quantied by ow cytometry. We conrmed that formulations
showing high IRF3 activation exhibited higher cellular internali-
zation than those with low IRF3 activation. D-CR3, D-CK3, and
D-R/K were more efciently internalized than the unmodied
dendrimer and D-CH3 and D-R/H showed the lowest internali-
zation. More than 90% of the cells showed nanoparticle uptake
for all of the dendrimer formulations, except those of the D-CH3
formulation. Interestingly, D-CK3 was efciently internalized,
but less potent IRF3 activation was observed compared to D-
CR3 and D-R/K. This can stem from the fact that less CDN is
delivered per (Figure S10, Supporting Information). An increase
in the number of dendrimer surface amines in the case of D-CK3
can enhance its internalization. However, its lysine residues have
a lower pKa than arginine in D-CR3, which will impart weaker
electrostatic interactions between the D-CK3 and the CDN com-
pared to that with D-CR3.
To ensure the biocompatibility of functionalized dendrimers,
the viability of THP-1 cells was determined 24 h post ADU-S100
delivery using our library of dendrimer formulations
(Figure 3D). The cell viability of the polypeptide-modied den-
drimers was higher than that of unmodied dendrimers at a
CDN concentration of 500 n
M
, suggesting that the addition of
natural peptides can increase the biocompatibility of dendrimer
NPs. D-CR3 and D-R/K, which were highly efcient at delivering
ADU-S100, showed different cell viability proles. While D-CR3
Figure 3. In vitro screening of polypeptide-modied dendrimers. A,B) Potent IRF3 and NF-κB activation following CDN delivery using polypeptide-
modied dendrimers. The polypeptide-modied dendrimers were screened for A) IRF3 activation and B) NF-κB activation using THP-1 Dual cells.
Cells were treated with different concentrations of ADU-S100 prior to measuring IRF3 and NF-κB 24 h posttreatment. Values were normalized to
untreated cells. C) Cell internalization efciency of polypeptide-modied dendrimers in THP-1 Dual cells. Cells were treated with polypeptide-modied
dendrimers containing ADU-S100 at the nal concentration of 125 n
M
, and uorescence expression per cell was determined 2 h posttransfection by ow
cytometry. D) Cell viability of different concentrations of polypeptide-modied dendrimers was analyzed 24 h posttreatment. Samples were normalized to
untreated cells. Data are represented as mean SD (n¼3). Multiple comparisons among groups were determined using one-way ANOVA followed by a
Fishers LSD test. P-value: *p<0.05, **p<0.01, ***p<0.001.
www.advancedsciencenews.com www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (4 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
cell viability was greater than 80%, the D-R/K formulation
showed some toxicity at high CDN concentrations (500 n
M
).
Taking that into account, we selected the D-CR3 formulation
to deliver ADU-S100 in vivo.
2.4. IT Delivery of ADU-S100 with D-CR3 NPs Inhibits B16-10
Tumor Growth and Increases Survival
The recent approval of different checkpoint inhibitor therapies
for the treatment of immunogenic tumors has opened the door
to the treatment of different cancers; however, the development
of more efcacious therapies is still needed. Recently, it has been
demonstrated that ICIs antibodies, such as antiprogrammed cell
death protein 1 (anti-PD-1), have a synergistic effect with STING
agonist therapies.
[19]
Here, we assessed whether combination
therapy with D-CR3 NPs complexed with ADU-S100 (termed
D-CR3-ADU) and anti-PD-1 showed a signicant benet com-
pared to free ADU-S100 alone or D-CR3-ADU NPs alone in a
syngeneic subcutaneous murine melanoma model. ADU-S100
therapy (0.5 μg per dose) was initiated when the tumor volume
reached 100 mm
3
and was delivered intratumorally (IT) four
times, every 4 days, over the course of 12 days. Anti-PD-1 was
administered via intraperitoneal injection, in the combination
therapy groups, 24 h after ADU-S100 delivery (four injections,
every 4 days). Tumor size was monitored every other day
(Figure 4A). Control tumors treated with phosphate buffered
saline (PBS) injection grew rapidly, whereas tumors treated with
ADU-S100-containing formulations showed delayed tumor
growth (Figure 4B). D-CR3-ADU-treated mice showed slower
tumor growth and increased survival when compared to ADU-
S100. The combination of D-CR3-ADU and anti-PD-1 signi-
cantly inhibited tumor growth compared to ADU-S100 alone.
A similar trend was observed when free ADU-S100 was com-
bined with anti-PD-1, where reduced tumor growth and
improved survival were observed. However, the NP-based deliv-
ery of ADU-S100, D-CR3 NPs, further inhibited tumor growth
and enhanced survival compared to the free drug form.
2.5. IT Delivery of CDN Modies the Immune Prole
of the Tumor Microenvironment
To understand the mechanism by which the D-CR3 formulation
containing ADU-S100 resulted in enhanced antitumor activity,
we conducted immunohistochemical analysis of the TME 7 days
after delivery of the last IT dose (19 days after treatment
initiation) (Figure 5). We found that tumors treated with
D-CR3-ADU have less actively proliferating cells [determined by
hematoxylin and eosin (H&E) and Ki-67 staining] compared to
tumors treated with free ADU-S100, which is further reduced when
D-CR3-ADU is combined with anti-PD-1. In addition, inltration of
CD8
þ
T-cells and upregulation of the immune checkpoint mole-
cule PD-L1 on tumor cells were observed when tumors were treated
with D-CR3-ADU compared to untreated tumors or those treated
with free ADU-S100. Similar results were obtained for the combi-
nation with anti-PD-1 compared to D-CR3-ADU alone.
Next, we conducted immunophenotyping of the TME and
tumor-draining lymph node (tdLN) 3 days after a single IT injec-
tion of ADU-S100, with or without D-CR3 complexation,
Figure 4. D-CR3-CDN polyplex therapy reduces tumor growth and increases animal survival, which is further enhanced when combined with anti-PD-1.
A) Study design of subcutaneous B16-F10 model. NP administration was initiated 8 days following tumor induction, when the tumor size reached
100 mm
3
. Intraperitoneal administration of anti-PD-1 was performed 24 h post NP administration. B) Tumor growth was monitored every other
day after the rst administration. 0.5 μg of ADU-S100 delivered with D-CR3 NP, with and without anti-PD-1, resulted in statistically reduced tumor burden
compared to free ADU-S100. Data are represented as mean SD (n¼5). Multiple comparisons among groups were determined using uncorrected
Dunns test. C) KaplanMeier survival curves of mice treated with the indicated formulation using a 1000 mm
3
tumor volume or poor body condition as
the endpoint criterion. Statistical analysis (n¼5) was performed using a log-rank MantelCox test.
www.advancedsciencenews.com www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (5 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
as well as with or without anti-PD-1 combination therapy. Flow
cytometry analysis of tumoral immune inltrates (Figure 6A)
revealed an increase in the CD8
þ
/CD4
þ
T-cell ratioa common
prognosticative biomarker of responsiveness to immunother-
apy
[20]
in the D-CR3-ADU-treated groups, compared to
untreated and to free-ADU-S100-treated tumors. We also wit-
nessed a slight increase in the CD8
þ
/T
reg
(dened as CD4
þ
FOXP3
þ
) ratio in tumors treated with D-CR3-ADU and anti-
PD-1 and a trend towards an increased presence of tumor-
inltrating T-cells (Figure 6B). Remarkably, the number of NK
cells was higher in all treated groups compared to untreated
tumors, but signicantly more in the tumors treated with
D-CR3-ADU. A signicant increase in activated granulocytic cells
(CD11b
þ
Gr-1
þ
, which includes monocytes, neutrophils, eosino-
phils, and myeloid-derived suppressor cells) was seen in all treat-
ment groups (Figure 6C). Moreover, increased expression of the
DCs activation marker CD86 was observed within the TME in all
the treated groups, compared to untreated tumors (Figure 6D).
Interestingly, DC inltration in the tdLN was elevated in all the
treatment groups, with D-CR3-ADU in combination with aPD-1
having the highest DC expression (Figure 6E). Also, higher PD-1
expression was observed in CD8
þ
and CD4
þ
cells in the tdLN
when ADU-S100 was delivered with a D-CR3 NP (Figure 6FG).
3. Discussion
The generation of endogenous CDNs in response to cytoplasmic
double-stranded DNA is an evolutionarily conserved innate
immune defense mechanism, resulting in the production of
IFN-I. Traditionally recognized as a response to viral infection,
this process has been increasingly implicated in antitumor
immunity.
[21]
Indeed, the potency of CDNs spurred efforts to
produce structural analogs of the endogenous adjuvant
Figure 5. D-CR3-ADU polyplex therapy reduces cancer cell proliferation, increases CD8
þ
cytotoxic T-cell inltration and PD-L1 cancer cell expression.
Representative H&E staining (left, scale bar: 100 μm; right: scale bar: 50 μm), Ki-67 staining (scale bar: 100 μm), immunouorescence staining of
CD8
þ
T-cells (red, CD8; blue, DAPI; scale bar: 100 μm), and PD-L1
þ
cancer cells (PD-L1þ, green; scale bar: 100 μm) in tumors harvested on day 19.
www.advancedsciencenews.com www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (6 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
cGAMP for use as antitumor therapeutics, such as ADU-S100.
[6a]
Here, we designed an NP system to overcome the delivery bar-
riers intrinsic to hydrophilic, anionic molecules such as CDNs
that require cytoplasmic delivery to enable their function.
Previous efforts for IT delivery of CDNs centralized around
encapsulation-based strategies whereby the CDN was housed
inside the NP. For instance, liposomes
[19a,22]
and polymeric
NPs such as polymersomes,
[11]
poly lactic-co-glycolic acid,
[23]
and acetalated dextran
[24]
have been used for the IT administra-
tion of CDNs. These formulations typically rely on intrinsic ele-
ments of the synthesized material (e.g., pH responsiveness,
endosomal disruption) for effective delivery. Depending on
the formulation and batch, the amount of CDN may vary, espe-
cially as hydrophilic molecules are difcult to encapsulate with
complete delity, and their storage stability can be compromised.
We present a two-component dendrimer system that is avidly
Figure 6. IT delivery of the D-CR3-CDN polyplex shifts the immune TME composition. A) Representative ow cytometry dot plot and quantication of
tumor-inltrating CD4
þ
and CD8
þ
T cellsratio. B) Ratio of CD8
þ
to Tregs cells in the TME. C) Flow cytometric quantication of the number of lym-
phocytes cells (CD3; CD3
þ
), natural killer cells (NK; NK1.1
þ
), macrophages (MΦ; CD11b
þ
F4/80
þ
), granulocytic cells (Gran; CD11b
þ
Gr-1
þ
), and den-
dritic cells (DC; CD11c
þ
MHCII
þ
) per milligram of tumor. D) Quantication of CD86 expression by IT DCs. E) Quantication of DCs in tdLN. F,G)
Quantication of PD-1
þ
expression by CD8
þ
and CD4
þ
cells in tumor-draining lymph node. Data are represented as mean SEM (n¼5).
Multiple comparisons among groups were determined using one-way ANOVA followed by a posthoc test. P-value: *p<0.05, **p<0.01, ***p<0.001.
www.advancedsciencenews.com www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (7 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
internalized by cells and requires only simple mixing that is
tuned to incorporate almost all CDN molecules, which has poten-
tial for scale-up. We,
[13b]
and others,
[13a]
have previously used
PAMAM dendrimers as nucleic acid delivery systems, which
is facilitated by electrostatic interactions between the cationic
dendrimer and anionic nucleic acid. We modied dendrimers
with polypeptide moieties with a dual-purpose function to
enhance the cationic nature of the dendrimer by virtue of addi-
tional amines while simultaneously attenuating the toxicity orig-
inating from unmodied PAMAM dendrimers. This was
achieved using a heterobifunctional linker that permits amide
bond formation with the dendrimer and Michael addition with
the thiol of polypeptide motifs of lysine, arginine, histidine, or
combinations thereof. Successful conjugation was conrmed
by
1
H-NMR and reected by the increased size of all polypep-
tide-modied dendrimers and changes in surface charge
(Figure 1). Histidine-containing formulations were less posi-
tively charged than their lysine or arginine counterparts at phys-
iological pH due to the lower afnity for protons, resulting in
partial protonation. This was further reected by the greater com-
plexation of ADU-S100 using D-CR3 compared to that of unmod-
ied dendrimer (Figure 2 and Figure S10, Supporting
Information). A combination of the increased amine groups
from polypeptide-modied dendrimer and cationicity was theo-
rized to be responsible for our observations, whereby D-CR3
could electrostatically entrap the most CDN-F as arginine has
the highest pKa values (pKa of side chain ¼12.48, 10.53, and
6.00, respectively, for arginine, lysine, and histidine). This is ben-
ecial as less dendrimer is required to deliver the same dose of
CDN-F, relative to other formulations.
Differential activation of IRF3 and NF-κB was observed
when we screened our formulations in vitro (Figure 3).
Greater activation of IRF3 was seen with NP formulations com-
pared to equimolar concentrations of free ADU-S100, particu-
larly beyond 250 n
M
. Dendrimer formulations containing
arginine were found to be the most potent inducers of IRF3
and NF-κB, implying that these particles were internalized effec-
tively and successfully delivered ADU-S100 to the cytoplasm. The
activation of IRF3, which is downstream of STING, leads to tran-
scription of IFN-Is and NF-κB signaling further elicits proinam-
matory mediator activation.
[25]
Indeed, modication of
dendrimers with arginine has been described to improve the
transfection efciency while maintaining biocompatibility,
[26]
matching our observations. NP formulations were differentially
internalized into immune cells in a modication-dependent
manner (Figure 3D). D-CH3 formulations were less efciently
internalized than D-CR3 and D-CK3, which would explain the
lower IRF3 activation despite the potent endosomal escape prop-
erties of histidine.
[27]
Lysine-modied dendrimers were internal-
ized to a greater extent but were less potent activators of IRF3
compared to arginine-modied dendrimers. This may be due
to the lower number of CDN delivered per dendrimer molecule,
which may affect overall uptake and endosomal escape capaci-
ties. The mixture of arginine and lysine was well internalized
and potently activated IRF3 and NF-κB; however, this formula-
tion was more cytotoxic than the arginine alone.
Based on our in vitro data, we took forward the D-CR3 formu-
lation, featuring high CDN complexation and biocompatibility,
for in vivo evaluation. Previous reports demonstrated that high
doses of IT delivered CDN (up to 20 μg) induced tumor regres-
sion; however, the efcacy is severely diminished at lower
doses.
[28]
Here, we demonstrated that our NP formulation pre-
vented growth of a poorly immunogenic murine melanoma
model using 0.5 μg of ADU-S100, an effect that was further aug-
mented when used in combination with anti-PD-1 (Figure 4).
Interestingly, we observed that a low dose of free ADU-S100, par-
ticularly used in combination with anti-PD-1, was able to attenu-
ate tumor growth, although less effective than the D-CR3-ADU.
It is known that the intensity of STING signaling has a notable
effect on the type and magnitude of the immune response. Low-
dose IT administration of ADU-S100 was found to be more
immunogenic than ablative (over 100 μg) doses and was optimal
for combination with immune checkpoint blockade (ICB).
[19b]
Even by potentially dosing with immunogenic amounts of free
ADU-S100, our NPs were still able to improve antitumor
responses compared to ADU-S100. Notably, without anti-PD-1,
D-CR3-ADU was still able to attenuate tumor growth and to a
much greater extent than equivalent free ADU-S100 alone.
Indeed, the efcacy of our NP is comparable to that of other elec-
trostatic polymeric NP-CDN formulations
[29]
despite using lower
CDN dose.
These observations led us to dive deeper into the mechanism
underpinning antitumor efcacy. A decrease of proliferating
cells and an increase of CD8
þ
T-cell inltration was observed
when ADU-S100 was delivered using D-CR3 compared to
tumors treated with free ADU-S100, which are hallmarks of anti-
tumor immune activation and induction of an immunogenic
TME (Figure 5). This was further corroborated by the upregula-
tion of the immune checkpoint molecule PD-L1 on tumor cells,
which is known to occur in response to IFN-β
[30]
a direct down-
stream product of STING signalingor IFN-γproduced by
tumor-inltrating T-cells.
[31]
This suggests that increased
STING signaling within the TME, facilitated by the D-CR3,
resulted in greater production of IFNs such as IFN-β, leading
to greater CD8
þ
T-cell inltration and PD-L1 expression.
Indeed, the improved therapeutic outcome seen with the inclu-
sion of anti-PD-1 may be in part due to the reduced immunosup-
pressive PD-1/PD-L1 interactions that would otherwise occur
upon STING-mediated upregulation of PD-L1.
In addition, ow cytometry analysis of the tumor immune
inltrate (Figure 6) conrmed an increase in CD8
þ
/CD4
þ
T-cell
and CD8
þ
T-cell/T
reg
ratios in the D-CR3-ADU-treated groups
compared with ADU-S100-treated or untreated tumors.
Increased tumoral T
reg
presence is a negative clinical prognostic
indicator across a broad range of cancers,
[32]
and thus our
T-cell-related observations were encouraging. Interestingly, the
number of NK cells was higher in all treated groups compared
to untreated tumors, but signicantly higher (twofold change) in
the tumors treated with D-CR3-ADU. It has been shown that
mobilization of NK cells in response to STING agonism can
induce tumor regression independently of CD8
þ
T-cells and is
a direct consequence of IFN-I production.
[33]
A signicant
increase in activated granulocytic cells was evident in all treat-
ment groups, which is consistent with the notion that granulo-
cytes, particularly neutrophils, are among the rst responders
that migrate to the site of inammationin this case induced
by STING activation. Interestingly, a trend of increase in the
number of macrophages (CD11b
þ
F4/80
þ
) and DCs (CD11c
þ
www.advancedsciencenews.com www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (8 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
MHC-II
þ
) compared to untreated tumors has only been observed
in groups treated with anti-PD-1, which may be responsible for
the improved efcacy witnessed when ADU-S100 was combined
with anti-PD-1, given the role of DCs and macrophages in
STING-mediated antitumor efcacy.
[34]
Moreover, increased
expression of the activation marker CD86 was evident on DCs
within the TME treated with free ADU-S100 or complexed using
D-CR3. Taken together, these immunological data show that
D-CR3-ADU administration can remodel the TME to a more
inamed and immunoactive environment. Interestingly, we
observed an increase in the presence of DCs in the tdLN-treated
mice, which was most pronounced in D-CR3-ADU-treated mice.
This suggests that a higher level of antigen presentation may
have taken place as more DCs migrated from the TME to the
tdLN to then cross-present to and prime CD8
þ
T-cells.
4. Conclusion
In the current study, we describe a library of polypeptide-
modied dendrimers that can be used to effectively deliver
CDN in vitro and in vivo. We found that arginine-modied den-
drimers permit higher CDN complexation efciency compared
to nonmodied dendrimers. Our data show that D-CR3-ADU
formulations are efciently internalized into THP-1 cells, result-
ing in the activation of IRF3 and NF-κB, and present lower tox-
icity than nonmodied dendrimers. In vivo results demonstrated
that IT delivery of the D-CR3-ADU formulation in combination
with anti-PD-1 induced strong tumor regression in a murine
melanoma model. This polypeptide-modied dendrimer-based
system can serve as a platform for efcient delivery of STING
agonists in poorly immunogenic tumors, providing new oppor-
tunities for combination cancer therapy.
5. Experimental Section
Materials: All reagents and solvents were purchased from Sigma Aldrich
unless otherwise stated. Polypeptides (HCysArgArgArgNH
2
,
HCysLysLysLysNH
2
, and HCysHisHisHisNH
2
) were
obtained from CPC Scientic with a purity of at least 90%. Generation
5 PAMAM dendrimer was purchased from Dendritech. Fluorescent
CDN (Ap-8-Fluo-AET-Gp) was obtained from Biolog, Inc.
Fluorescent Labeling of Dendrimer: G5 PAMAM dendrimer was uores-
cently tagged with AlexaFluor 594 carboxylic acid, succinimidyl ester
(AF594) at 1:0.5 molar ratio (PAMAM dendrimer:AF594). Briey, 14.22 μL
of AF594 at 10 mg mL
1
in dimethylformamide (DMF) was mixed with
10 mg PAMAM dendrimer in 0.1
M
bicarbonate buffer (pH 8.5) and
reacted for 1 h at room temperature in the dark. Fluorescent dendrimers
were washed with PBS and recovered by centrifugal ltration (10 kDa
MWCO Amicon Ultra-0.5 mL Centrifugal Filters, Millipore) at 14 000 g
for 10 min at 4 C.
Synthesis of Polypeptide-Modied Dendrimers: PAMAM dendrimer was
modied with bifunctional SM(PEG)
2
linker (Thermo Fisher), whereby
10 mg of dendrimer was dissolved in 500 μL of 0.1
M
phosphate buffer
at pH 7.5 and 500 μL PBS containing 19 mg of SM(PEG)
2
was added drop-
wise. The mixture was allowed to react for 30 min at room temperature.
The dendrimer was then mixed with equimolar SM(PEG)
2
to polypeptide
ratio and reacted at room temperature for 3 h. The polypeptide-modied
dendrimer was puried by dialysis against PBS for 2 days at 4 ºC. For struc-
tural analysis, modied dendrimers were freeze dried, dissolved in D
2
O,
and analyzed by
1
H-NMR, and recorded using a 400 MHz Varian NMR
spectrometer (NMR Instruments, Clarendon Hills, IL).
Quantication of the Ratio of Peptides per Dendrimer by UVVis
Spectroscopy: NanoDrop 2000c (Thermo Scientic, Tewksbury, MA) was
used to characterize the polypeptide-modied dendrimer. Absorbance
at 275 nm was used to determine the ratio of peptide to dendrimer.
Because the uorescent dendrimer also absorbs at 275 nm, the equation
that follows was used to deconvolute the signals from each component
(dendrimer or peptide) and obtain the peptide-to-dendrimer ratio
(Equation (1)). The individual absorption coefcients were calculated from
the respective UVvis standard curves.
ADendF &Peptide ¼ADendFþAPeptide (1)
εDendF &Peptide cDendF &Peptide ¼εDendFcDendFþεPeptide cPeptide
(2)
cDendF & Peptide ¼cDendF(3)
cpeptide
cDendF
¼εDendF &Peptide εDendF
εPeptide
(4)
Equation (1) is the peptide-to-dendrimer ratio calculation.
Biophysical Characterization of Polypeptide-Modied Dendrimers:To
assess CDN complexation, different uorescent CDN (Ap-8-Fluo-AET-
Gp) to dendrimer ratios (w/w) between 0.5:1 and 64:1 were studied.
DendrimerCDN complexes were freshly prepared; for example, to form
100 μL of D-CR3 polyplexes, 50 μL of CDN at 0.05 mg mL
1
was mixed
with 50 μL of D-CR3 at 0.8 mg mL
1
. The dendrimer solution was added
to CDN solution, pipette mixed, and incubated at room temperature for
10 min. DendrimerCDN polyplexes were loaded in 4% E-Gel Precast
Agarose Gels (Thermo Fisher), run following the manufacturers instruc-
tions, and visualized in uorescence mode. The size and surface charge
were determined by DLS and zeta potential measurements, respectively.
Polyplexes were prepared as previously described and after 10 min of incu-
bation at room temperature, 100 μL of dendrimer was diluted with 900 μL
of PBS and analyzed using a Zetasizer Nano ZS equipped with a HeNe
laser (λ¼633 nm) at a scattering angle of 137(Malvern Instruments Ltd,
United Kingdom).
CDN Loading Efciency:DendrimerCDN complexes were freshly pre-
pared as previously reported. To form 100 μL of D-CR3-CDN polyplexes,
50 μL of CDN at 0.05 mg mL
1
was mixed with 50 μLof
D-CR3 at 0.8 mg mL
1
.Toform100μL of dendrimerCDN polyplexes,
50 μL of CDN at 0.05 mg mL
1
was mixed with 50μL of unmodied den-
drimer at 3.2 mg mL
1
. Noncomplexed CDN-F molecules were puried by
centrifugal ltration (10 kDa MWCO Amicon Ultra-0.5 mL Centrifugal
Filters, Millipore) at 14 000 gfor 10 min at 4 C and quantied by uores-
cence (λ
ex
¼490; (λ
em
¼530) using a multimodal plate reader (TECAN).
Cell Lines:Mus musculus skin melanoma (B16-F10, from ATCC) was
maintained in Dulbeccos minimum essential medium (DMEM) supple-
mented with 10% fetal bovine serum (FBS), 100 U mL
1
penicillin, and
100 μgmL
1
streptomycin, 2 m
ML
-glutamine. Human monocyte THP-1
Dual cells and THP-1 Dual KO-STING cells (InvivoGen) were maintained
in RPMI 1640 supplemented with 10% FBS, 2 m
ML
-glutamine,
25 m
M
HEPES, 100 μgmL
1
Normocin, Zeocin, 10 μgmL
1
Blasticidin
(InvivoGen), and 100 U mL
1
penicillin and 100 μgmL
1
streptomycin.
All cell lines were maintained in a humidied incubator at 37 C, 5% CO
2
.
In Vitro Evaluation of IRF and NF-κB Pathways: Human monocyte THP-1
Dual cells and THP-1 Dual KO-STING cells were seeded in 96-well plates at
110
5
cells per well and incubated with the different dendrimer NP for-
mulations or free CDN at CDN concentrations ranging from 0 to 500n
M
.
At 24 h post-treatment, IRF activity was examined using the QUANTI-Luc
reagent (InvivoGen) and NF-κB activity was determined using the QUANTI-
Blue reagent (InvivoGen) according to the manufacturers instructions.
Cell Viability: Cell viability was assessed using the MTS assay (Promega)
as instructed by the manufacturer. After 24 h treatment with varying
dendrimerCDN NP formulations, MTS reagent was added to the cells
to achieve a nal MTS concentration of 20% (v/v). Cells were incubated
at 37 C, 5% CO
2
up to 2 h and absorbance was measured at 490 nm using
a multimodal plate reader (TECAN).
www.advancedsciencenews.com www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (9 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
Cellular Uptake: THP-1 Dual cells were seeded in 12-well plates at
510
5
cells per well and treated with uorescent polypeptide-modied
dendrimer at a concentration equivalent to 125 n
M
CDN. After 2 h at
37 C, 5% CO
2
, excess NPs were removed by washing with PBS and cells
were collected by centrifugation. Following xation with 1% (w/v) parafor-
maldehyde, cells were analyzed by ow cytometry using a BD LSRFortessa
ow cytometer (BD Biosciences).
In Vivo Therapeutic Efcacy: Female C57BL/6 mice (68 weeks old) were
purchased from Charles River. All mouse procedures were conducted at
the Hale Building for Transformative Medicine and Koch Institute for
Integrative Cancer Research at the Massachusetts Institute of Technology
(MIT) under the protocol approved for this study by the Institutional
Animal Care and Use Committee (IACUC). To induce tumors, 5 10
5
B16-F10 cells in 100 μL of Hanksbalanced salt solution (HBSS) were
injected subcutaneously into the right ank of the mice. Upon reaching
50100 mm
3
, tumors were intratumorally injected with 30 μL of PBS con-
taining D-CR3-CDN polyplexes or free CDN (0.5 μg). Mice were injected
four times with treatments spaced 4 days apart. Mice receiving ICB were
injected intraperitoneally with 100 μg of anti-PD-1 (clone RMP1.14, Bio X
Cell) 24 h post IT injection. The tumor size was measured every other day
via caliper measurements, and the tumor volume was calculated using the
equation V¼(LWH)/π÷6. Body weight was measured contempora-
neously with tumor volume. Mice were euthanized when tumors reached a
volume of 1000 mm
3
or for otherwise poor body condition.
Analysis of Immune Inltrate: B16-F10 tumors were harvested,
chopped, and digested in a solution of HBSS supplemented with collage-
nase I, II, and IV (100 ng mL
1
), and DNase I (1 μgmL
1
) for 2 h at 37 C.
TdLNs were harvested and mechanically dissociated. Single-cell suspen-
sions of tumors and tdLNs were ltered through a 40 μm nylon cell
strainer. Tumor cells were further treated with ACK Lysing Buffer (Gibco).
Cells were counted and stained with uorescent antibodies at a concen-
tration of 1 10
6
cells mL
1
in 100 μL cell-staining buffer (BioLegend).
Intracellular staining was performed using a FoxP3 Staining Buffer Set
(Miltenyi) according to the manufacturers protocol. The following anti-
mouse antibodies were used for ow cytometry were purchased from
BioLegend: CD45 APC-Cy7 (clone 30-F11), NK-1.1 BV710 (clone
PK136), FOXP3 PE (clone MF-14) IFN-γBV421 (clone XMG1.2),
CD279 (PD-1) FITC (clone 29 F.1A12), CD45 BV785 (clone 30-F11),
CD11b BV421 (clone M1/70), Gr-1 APC-Cy7 (clone RB6-8C5), CD8a
BV421 (clone 53-6.7), CD86 BV510 (clone GL-1), CD206 PE (clone
C068C2), and MHCII BV605 (clone M5/114.15.2) CD11c APC (clone
N418). The following antimouse antibodies were purchased from BD
Biosciences: CD3 BB700 (clone 17A2), CD4 BUV395 (clone GK1.5),
CD8a BUV737 (clone 53-6.7), F4/80 BUV395 (clone T45-2342), CD103
BUV395 (clone M290), and CD80 BUV737 (clone 16-10A1). Live cells
were gated using LIVE/DEAD (Thermo Fisher) aqua (cat. no. L34966),
green (cat. no. L34970), or NIR (cat. no. L34976). Stained cells were
analyzed by ow cytometry using a BD LSRFortessa ow cytometer
(BD Biosciences) and all data were analyzed using FlowJo software
(Flowjo LLC).
Immunohistochemistry, Immunouorescence, and Imaging: B16-F10
tumors were resected kept in 10% (v/v) formalin for a minimum of
24 h and then in 70% (v/v) ethanol until processing. H&E as well as
expression of Ki-67, CD8, and PD-L1 protein was assessed by immunohis-
tochemistry (IHC) from histological sections (5 μm) of tumors. H&E stain-
ing was performed using standard protocols. For protein expression,
parran-embedded sections were deparranized, rehydrated, and washed
in distilled water. Antigen retrieval was then performed in citrate buffer
(pH 6.0) at 125 C for 5 min. To quench endogenous peroxidase activity,
samples were incubated with 3% (v/v) H
2
O
2
for 5 min at room tempera-
ture and washed with PBS-Tween (PBS-T). The samples were then blocked
with blocking buffer consisting of 10% (v/v) donkey serum, 1% (w/v) BSA
in PBS-T for 60 min at room temperature. Sections were then incubated
with antimouse CD8a (Thermo Fisher, cat. no. PA5-81344) or anti-mouse
PD-L1 (Proteintech, cat. no. 66248-1-Ig) overnight at 4 C in a humidied
chamber. After rinsing with PBS-T, samples were incubated with either
goat antirabbit AF594 (abcam, cat. no. ab150080) or goat antimouse
AF488 (Jackson ImmunoResearch, cat. no. 115-543003) for 2 h at room
temperature in the dark. The slides were washed with PBS-T, stained with
Hoescht 33342 (Thermo Fisher), washed again with PBS-T, and mounted
using ProLong Diamond antifade mountant (Thermo Fisher). For Ki-67
expression, sections were run using a LabVision Autostainer 360
(Thermo Fisher) following an automated protocol using a polymer-based
antibody detection system (Vector Laboratories) and visualization with
3,30-diaminobenzidine. H&E and Ki-67 images were obtained using an
Aperio AT2 slide scanner (Leica Biosystems) and uorescence images
were captured using a Nikon Ti-E microscope and were processed with
ImageJ software.
Statistical Analysis: Statistical analyses were conducted using Graph-Pad
Prism 8 (GraphPad Software). All data are reported as mean SEMs. For
in vitro experiments, a minimum of n¼3 biological replicates were used
per condition in each experiment. Pairwise comparisons were performed
using Student t-tests. Multiple comparisons among groups were deter-
mined using one-way ANOVA followed by a posthoc test. For in vivo
experiments, a minimum of n¼5 biological replicates were used per con-
dition in each experiment. Multiple comparisons among groups were
determined using KruskalWallis test with uncorrected Dunns test.
KaplanMeier survival curve statistical analysis was determined using
the two-tailed MantelCox test. No specic preprocessing of data was per-
formed prior to statistical analyses. Differences between groups were con-
sidered signicant at p-values below 0.05 (*p<0.05, **p<0.01,
***p<0.001).
Supporting Information
Supporting Information is available from the Wiley Online Library or from
the author.
Acknowledgements
The authors thank the Hale Building for Transformative Medicine for the
assistance with animal housing. The authors thank Swanson
Biotechnology Center at the Koch Institute for Integrative Cancer
Research at the Massachusetts Institute of Technology (MIT) for assis-
tance with animal experiments and facilities, especially the microscopy,
ow cytometry, and histology cores. The authors thank the Department
of Comparative Medicine at MIT. The authors thank G. Paradis for
FACS assistance with Cancer Center Support (FACS core). The authors
thank Kathleen S. Cormier and Charlene Condon for histology and immu-
nohistochemistry assistance. The authors thank Takeda Pharmaceuticals
for provision of ADU-S100.
Conict of Interest
The authors declare no conict of interest.
Data Availability Statement
Research data are not shared.
Keywords
dendrimer nanoparticles, immunotherapy, intratumoral delivery,
melanoma, stimulator of interferon genes agonist
Received: January 4, 2021
Revised: March 10, 2021
Published online: April 9, 2021
www.advancedsciencenews.com www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (10 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
[1] T. Shekarian, S. Valsesia-Wittmann, J. Brody, M. C. Michallet, S. Depil,
C. Caux, A. Marabelle, Ann. Oncol. 2017,28, 1756.
[2] a) P. Gao, M. Ascano, Y. Wu, W. Barchet, Barbara L. Gaffney,
T. Zillinger, A. A. Serganov, Y. Liu, R. A. Jones, G. Hartmann,
T. Tuschl, D. J. Patel, Cell 2013,153, 1094; b) L. Sun, J. Wu,
F. Du, X. Chen, Z. J. Chen, Science 2013,339, 786.
[3] a) X. Zhang, H. Shi, J. Wu, X. Zhang, L. Sun, C. Chen, Z. J. Chen, Mol.
Cell 2013,51, 226; b) E. J. Diner, D. L. Burdette, S. C. Wilson,
K. M. Monroe, C. A. Kellenberger, M. Hyodo, Y. Hayakawa,
M. C. Hammond, R. E. Vance, Cell Rep. 2013,3, 1355.
[4] a) S. Yum, M. Li, A. E. Frankel, Z. J. Chen, Annu. Rev. Cancer Biol. 2019,
3, 323; b) J. Kwon, S. F. Bakhoum, Cancer Discovery 2020,10,26.
[5] a) H. Ishikawa, Z. Ma, G. N. Barber, Nature 2009,461, 788;
b) S.-R. Woo, M. B. Fuertes, L. Corrales, S. Spranger, M. J. Furdyna,
M. Y. K. Leung, R. Duggan, Y. Wang, G. N. Barber, K. A. Fitzgerald,
M.-L. Alegre, T. F. Gajewski, Immunity 2014,41, 830.
[6] a) L. Corrales, L. H. Glickman, S. M. McWhirter, D. B. Kanne,
K. E. Sivick, G. E. Katibah, S. R. Woo, E. Lemmens, T. Banda,
J. J. Leong, K. Metchette, T. W. Dubensky, Jr., T. F. Gajewski, Cell
Rep. 2015,11, 1018; b) T. Ohkuri, A. Kosaka, K. Ishibashi,
T. Kumai, Y. Hirata, K. Ohara, T. Nagato, K. Oikawa, N. Aoki,
Y. Harabuchi, E. Celis, H. Kobayashi, Cancer Immunol.
Immunother. 2017,66, 705; c) B. A. Flood, E. F. Higgs, S. Li,
J. J. Luke, T. F. Gajewski, Immunol. Rev. 2019,290, 24.
[7] B. R. Huck, L. Kotzner, K. Urbahns, Angew. Chem., Int. Ed. 2018,
57, 4412.
[8] T. W. Dubensky, D. B. Kanne, M. L. Leong, Therap. Adv. Vaccines 2013,
1, 131.
[9] a) R. Zhang, M. M. Billingsley, M. J. Mitchell, J. Controll. Release 2018,
292, 256; b) D. J. Irvine, E. L. Dane, Nat. Rev. Immunol. 2020,20, 321.
[10] S. T. Koshy, A. S. Cheung, L. Gu, A. R. Graveline, D. J. Mooney, Adv.
Biosyst. 2017,1, 1600013.
[11] D. Shae, K. W. Becker, P. Christov, D. S. Yun, A. K. R. Lytton-Jean,
S. Sevimli, M. Ascano, M. Kelley, D. B. Johnson, J. M. Balko,
J. T. Wilson, Nat. Nanotechnol. 2019,14, 269.
[12] G. Bozzuto, A. Molinari, Int. J. Nanomed. 2015,10, 975.
[13] a) L. Palmerston Mendes, J. Pan, V. P. Torchilin, Molecules 2017,22,
1401; b) J. Conde, N. Oliva, M. Atilano, H. S. Song, N. Artzi, Nat.
Mater. 2016,15, 353.
[14] a) E. Vacas-Córdoba, N. Climent, F. J. De La Mata, M. Plana, R. Gómez,
M. Pion, F. García, M. Á. Mu˜noz-Fernández, Nanomedicine
2014,9, 2683; b) S. R. Bonam, A. Areti, P. Komirishetty, S. Muller,
in Pharmaceutical Applications of Dendrimers (Eds.: A. Chauhan,
H. Kulhari), Elsevier, Amsterdam 2020,p.233.
[15] F. Abedi-Gaballu, G. Dehghan, M. Ghaffari, R. Yekta, S. Abbaspour-
Ravasjani, B. Baradaran, J. E. N. Dolatabadi, M. R. Hamblin, Appl.
Mater. Today 2018,12, 177.
[16] R. Duncan, L. Izzo, Adv. Drug Deliv. Rev. 2005,57, 2215.
[17] a) N. Segovia, P. Dosta, A. Cascante, V. Ramos, S. Borrós, Acta
Biomater. 2014,10, 2147; b) P. Dosta, N. Segovia, A. Cascante,
V. Ramos, S. Borrós, Acta Biomater. 2015,20, 82; c) R. Nunez-Toldra,
P. Dosta, S. Montori, V. Ramos, M. Atari, S. Borros, Acta Biomater.
2017,53, 152.
[18] a) P. Dosta, I. Tamargo, V. Ramos, S. Kumar, D. W. Kang, S. Borros,
H. Jo, Adv. Healthcare Mater. 2021, https://doi.org/10.1002/adhm.
202001894; b) P. Dosta, C. Demos, V. Ramos, D. W. Kang,
S. Kumar, H. Jo, S. Borros, Cardiovasc. Eng. Technol. 2021,12, 114.
[19] a) S. T. Koshy, A. S. Cheung, L. Gu, A. R. Graveline, D. J. Mooney,
Adv. Biosyst. 2017,1, 1600013; b) K. E. Sivick, A. L. Desbien,
L. H. Glickman, G. L. Reiner, L. Corrales, N. H. Surh,
T. E. Hudson, U. T. Vu, B. J. Francica, T. Banda, G. E. Katibah,
D. B. Kanne, J. J. Leong, K. Metchette, J. R. Bruml,
C. O. Ndubaku, J. M. McKenna, Y. Feng, L. Zheng, S. L. Bender,
C. Y. Cho, M. L. Leong, A. van Elsas, T. W. Dubensky,
S. M. McWhirter, Cell Rep. 2018,25, 3074.
[20] N.-P. Rudqvist, K. A. Pilones, C. Lhuillier, E. Wennerberg, J.-W. Sidhom,
R. O. Emerson, H. S. Robins, J. Schneck, S. C. Formenti, S. Demaria,
Cancer Immunol. Res. 2018,6,139.
[21] Z. Cheng, T. Dai, X. He, Z. Zhang, F. Xie, S. Wang, L. Zhang, F. Zhou,
Signal Transd. Targeted Therapy 2020,5, 91.
[22] a) T. Nakamura, H. Miyabe, M. Hyodo, Y. Sato, Y. Hayakawa,
H. Harashima, J Control Release 2015,216, 149; b) N. Cheng,
R. Watkins-Schulz, R. D. Junkins, C. N. David, B. M. Johnson,
S. A. Montgomery, K. J. Peine, D. B. Darr, H. Yuan, K. P. McKinnon,
Q. Liu, L. Miao, L. Huang, E. M. Bachelder, K. M. Ainslie, J. P. Ting,
JCI Insight 2018,3, e120638.
[23] S. Chattopadhyay, Y. H. Liu, Z. S. Fang, C. L. Lin, J. C. Lin, B. Y. Yao,
C. J. Hu, Nano Lett. 2020,20, 2246.
[24] R. Watkins-Schulz, P. Tiet, M. D. Gallovic, R. D. Junkins, C. Batty,
E. M. Bachelder, K. M. Ainslie, J. P. Y. Ting, Biomaterials 2019,
205, 94.
[25] T. Abe, G. N. Barber, J. Virol. 2014,88, 5328.
[26] a) J. S. Choi, K. Nam, J.-y. Park, J.-B. Kim, J.-K. Lee, J.-s. Park,
J. Controlled Release 2004,99, 445; b) T. Okuda, A. Sugiyama,
T. Niidome, H. Aoyagi, Biomaterials 2004,25, 537.
[27] Y. Wen, Z. Guo, Z. Du, R. Fang, H. Wu, X. Zeng, C. Wang, M. Feng,
S. Pan, Biomaterials 2012,33, 8111.
[28] J. Fu, D. B. Kanne, M. Leong, L. H. Glickman, S. M. McWhirter,
E. Lemmens, K. Mechette, J. J. Leong, P. Lauer, W. Liu,
K. E. Sivick, Q. Zeng, K. C. Soares, L. Zheng, D. A. Portnoy,
J. J. Woodward, D. M. Pardoll, T. W. Dubensky, Jr., Y. Kim, Sci.
Transl. Med. 2015,7, 283ra52.
[29] D. R. Wilson, R. Sen, J. C. Sunshine, D. M. Pardoll, J. J. Green,
Y. J. Kim, Nanomed. Nanotechnol. Biol. Med. 2018,14, 237.
[30] A. Garcia-Diaz, D. S. Shin, B. H. Moreno, J. Saco, H. Escuin-Ordinas,
G. A. Rodriguez, J. M. Zaretsky, L. Sun, W. Hugo, X. Wang, G. Parisi,
C. P. Saus, D. Y. Torrejon, T. G. Graeber, B. Comin-Anduix,
S. Hu-Lieskovan, R. Damoiseaux, R. S. Lo, A. Ribas, Cell Rep.
2017,19, 1189.
[31] A. Thiem, S. Hesbacher, H. Kneitz, T. di Primio, M. V. Heppt,
H. M. Hermanns, M. Goebeler, S. Meierjohann, R. Houben,
D. Schrama, J. Exp. Clin. Cancer Res. 2019,38, 397.
[32] B. Shang, Y. Liu, S.-j. Jiang, Y. Liu, Sci. Rep. 2015,5, 15179.
[33] C. J. Nicolai, N. Wolf, I. C. Chang, G. Kirn, A. Marcus, C. O. Ndubaku,
S. M. McWhirter, D. H. Raulet, Sci. Immunol. 2020,5, eaaz2738.
[34] R. E. Vatner, E. M. Janssen, Mol. Immunol. 2019,110, 13.
www.advancedsciencenews.com www.advnanobiomedres.com
Adv. NanoBiomed Res. 2021,1, 2100006 2100006 (11 of 11) © 2021 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
... The multivalency of these dendrimer and dextran polymers facilitates the integration of therapies onto the hydrogel network, enabling their continuous release as the hydrogel degrades. Given these properties, the use of adhesive hydrogels for intracranial delivery of therapies targeting GBM, such as CDN-NPs (114) recently developed by the Artzi group to activate the cGAS-STING pathway, holds promise in blunting immunosuppression in the GBM TME. These particles reduced myeloid-mediated immunosuppression in the TME of different tumor types, including melanoma, colon and breast cancers, and increased the recruitment and activation of anti-tumor immune effector cells, such as cytotoxic T and NK cells (94). ...
... These particles reduced myeloid-mediated immunosuppression in the TME of different tumor types, including melanoma, colon and breast cancers, and increased the recruitment and activation of anti-tumor immune effector cells, such as cytotoxic T and NK cells (94). Furthermore, they have been optimized for enhanced biocompatibility and robust cytoplasmic delivery of CDNs, resulting in potent immune cell activation and extended survival in multiple tumor models ( Fig. 1 A, iii and iv) (94,114). While local polymer-based delivery systems (e.g., Gliadel™) (36) and other preclinical delivery systems have previously been explored, numerous challenges still exist, including poor control over spatiotemporal drug release and adhesive mismatch between the implant and brain tissue (37). ...
Article
Full-text available
Integrating multimodal neuro- and nanotechnology-enabled precision immunotherapies with extant systemic immunotherapies may finally provide a significant breakthrough for combatting glioblastoma (GBM). The potency of this approach lies in its ability to train the immune system to efficiently identify and eradicate cancer cells, thereby creating anti-tumor immune memory while minimizing multi-mechanistic immune suppression. A critical aspect of these therapies is the controlled, spatiotemporal delivery of structurally defined nanotherapeutics into the GBM tumor microenvironment (TME). Architectures such as spherical nucleic acids or poly(beta-amino ester)/dendrimer-based nanoparticles have shown promising results in preclinical models due to their multivalency and abilities to activate antigen-presenting cells and prime antigen-specific T cells. These nanostructures also permit systematic variation to optimize their distribution, TME accumulation, cellular uptake, and overall immunostimulatory effects. Delving deeper into the relationships between nanotherapeutic structures and their performance will accelerate nano-drug development and pave the way for the rapid clinical translation of advanced nanomedicines. In addition, the efficacy of nanotechnology-based immunotherapies may be enhanced when integrated with emerging precision surgical techniques, such as laser interstitial thermal therapy, and when combined with systemic immunotherapies, particularly inhibitors of immune-mediated checkpoints and immunosuppressive adenosine signaling. In this perspective, we highlight the potential of emerging treatment modalities, combining advances in biomedical engineering and neurotechnology development with existing immunotherapies to overcome treatment resistance and transform the management of GBM. We conclude with a call to action for researchers to leverage these technologies and accelerate their translation into the clinic.
... The stimulation of innate immune danger sensors, such as the cytosolic cyclic dinucleotide (CDN) sensor stimulator of interferon genes (STING), is a powerful therapeutic strategy to generate durable antitumour immune responses [1][2][3][4] . STING agonism enacts a type I interferon proinflammatory pathway that drives multifaceted antitumour immune functions in the tumour microenvironment (TME) and tumour-draining lymph nodes (tdLNs) by recruiting and activating immune cells 5,6 and enhancing natural killer (NK) cell 7,8 and T-cell-mediated tumour killing 9,10 . To achieve exogenous STING activation, small-molecule mimetics of CDNs have been synthesized [11][12][13] , leading to notable efficacy in preclinical models, which was further enhanced when combined with other therapeutic modalities [14][15][16] . ...
Article
Full-text available
Intravenously administered cyclic dinucleotides and other STING agonists are hampered by low cellular uptake and poor circulatory half-life. Here we report the covalent conjugation of cyclic dinucleotides to poly(β-amino ester) nanoparticles through a cathepsin-sensitive linker. This is shown to increase stability and loading, thereby expanding the therapeutic window in multiple syngeneic tumour models, enabling the study of how the long-term fate of the nanoparticles affects the immune response. In a melanoma mouse model, primary tumour clearance depends on the STING signalling by host cells—rather than cancer cells—and immune memory depends on the spleen. The cancer cells act as a depot for the nanoparticles, releasing them over time to activate nearby immune cells to control tumour growth. Collectively, this work highlights the importance of nanoparticle structure and nano-biointeractions in controlling immunotherapy efficacy.
Article
Full-text available
The cGAS-STING signaling plays an important role in the immune response in a tumor microenvironment (TME) of triple-negative breast cancer (TNBC). The acute and controlled activation of cGAS-STING signaling results in tumor suppression, while chronic activation of cGAS-STING signaling results in immune-suppressive TME that could result in tumor survival. There is a need, therefore, to develop therapeutic strategies for harnessing tumor suppressive effects of cGAS-STING signaling while minimizing the risks associated with chronic activation. Combination therapies and nanocarriers-based delivery of cGAS-STING agonists have emerged as promising strategies in immunotherapy for controlled modulation of cGAS-STING signaling in cancer. These approaches aim to optimize the tumor suppressive effects of the cGAS-STING pathway while minimizing the challenges associated with modulators of cGAS-STING signaling. In the present review, we discuss recent advancements and strategies in combination therapies and nanocarrier-based delivery systems for effectively controlling cGAS-STING signaling in cancer immunotherapy. Further, we emphasized the significance of nanocarrier-based approaches for effective targeting of the cGAS-STING signaling, tackling resistance mechanisms, and overcoming key challenges like immune suppression, tumor heterogeneity, and off-target effects. Graphical Abstract
Article
Full-text available
Biobased, DNA delivery vectors have been synthesized with a core motif composed of 2,5-bishydroxymethylfuran (BHMF) readily available from an important biomass feedstock 5-hydroxymethyl furfural (HMF). To generate the product, BHMF was first converted to 2,5-furan bishydroxymethyl diacrylate (2,5-FDA), which was later conjugated with different types of secondary amines. Rich in tertiary nitrogen, these oligomeric FDA-amino esters demonstrated stable electrostatic interactions with negatively charged plasmid DNA in an aqueous environment. We evaluated synthetic routes toward these plasmid DNA-binding amino esters (pFASTs), identified their nanoscale features, and attempted to establish their structure–property relationship in the context of the DNA delivery. Our preliminary studies show that the pFASTs formed stable complexes with the plasmid DNA. Dynamic light scattering indicated that the DNA polyplexes of pFASTs have hydrodynamic diameters within the size range of 100–150 nm with a surface charge (ζ-potential) ranging from −10 to +33 mV, depending on pFAST type. These oligomeric amino esters rich in furan motif were also found to successfully transfect the GFP-expressing plasmid DNA intracellularly. Collectively, this study establishes a new route to produce DNA transfection agents from sustainable resources that can be used for transferring genetic materials for humans, veterinary, and agrochemical purposes.
Article
The recurrent nature of glioblastoma negatively impacts conventional treatment strategies leading to a growing need for nanomedicine. Nanotherapeutics, an approach designed to deliver drugs to specific sites, is experiencing rapid growth and gaining immense popularity. Having potential in reaching the hard‐to‐reach disease sites, this field has the potential to show high efficacy in combatting glioblastoma progression. The presence of glioblastoma stem cells (GSCs) is a major factor behind the poor prognosis of glioblastoma multiforme (GBM). Stemness potential, heterogeneity, and self‐renewal capacity, are some of the properties that make GSCs invade across the distant regions of the brain. Despite advances in medical technology and MRI‐guided maximal surgical resection, not all GSCs residing in the brain can be removed, leading to recurrent disease. The aggressiveness of GBM is often correlated with immune suppression, where the T‐cells are unable to infiltrate the cancer initiating GSCs. Standard of care therapies, including surgery and chemotherapy in combination with radiation therapy, have failed to tackle all the challenges of the GSCs, making it increasingly important for researchers to develop strategies to tackle their growth and proliferation and reduce the recurrence of GBM. Here, we will focus on the advancements in the field of nanomedicine that has the potential to show positive impact in managing glioblastoma tumor microenvironment. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease
Article
Adjuvants are of critical value in vaccine development as they act on enhancing immunogenicity of antigen and inducing long‐lasting immunity. However, there are only a few adjuvants that have been approved for clinical use, which highlights the need for exploring and developing new adjuvants to meet the growing demand for vaccination. Recently, emerging evidence demonstrates that the cGAS‐STING pathway orchestrates innate and adaptive immunity by generating type I interferon responses. Many cGAS‐STING pathway agonists have been developed and tested in preclinical research for the treatment of cancer or infectious diseases with promising results. As adjuvants, cGAS‐STING agonists have demonstrated their potential to activate robust defense immunity in various diseases, including COVID‐19 infection. This review summarized the current developments in the field of cGAS‐STING agonists with a special focus on the latest applications of cGAS‐STING agonists as adjuvants in vaccination. Potential challenges were also discussed in the hope of sparking future research interests to further the development of cGAS‐STING as vaccine adjuvants.
Article
Full-text available
STING (Stimulator of Interferon Genes) agonists have emerged as promising agents in the field of cancer immunotherapy, owing to their excellent capacity to activate the innate immune response and combat tumor-induced immunosuppression. This review provides a comprehensive exploration of the strategies employed to develop effective formulations for STING agonists, with particular emphasis on versatile nano-delivery systems. The recent advancements in delivery systems based on lipids, natural/synthetic polymers, and proteins for STING agonists are summarized. The preparation methodologies of nanoprecipitation, self-assembly, and hydrogel, along with their advantages and disadvantages, are also discussed. Furthermore, the challenges and opportunities in developing next-generation STING agonist delivery systems are elaborated. This review aims to serve as a reference for researchers in designing novel and effective STING agonist delivery systems for cancer immunotherapy.
Article
Full-text available
Cytosolic DNA is an indicator of pathogen invasion or DNA damage. The cytosolic DNA sensor cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) detects DNA and then mediates downstream immune responses through the molecule stimulator of interferon genes (STING, also known as MITA, MPYS, ERIS and TMEM173). Recent studies focusing on the roles of the cGAS-STING pathway in evolutionary distant species have partly sketched how the mammalian cGAS-STING pathways are shaped and have revealed its evolutionarily conserved mechanism in combating pathogens. Both this pathway and pathogens have developed sophisticated strategies to counteract each other for their survival. Here, we summarise current knowledge on the interactions between the cGAS-STING pathway and pathogens from both evolutionary and mechanistic perspectives. Deeper insight into these interactions might enable us to clarify the pathogenesis of certain infectious diseases and better harness the cGAS-STING pathway for antimicrobial methods.
Article
Full-text available
Background: Immune checkpoint inhibition and in particular anti-PD-1 immunotherapy have revolutionized the treatment of advanced melanoma. In this regard, higher tumoral PD-L1 protein (gene name: CD274) expression is associated with better clinical response and increased survival to anti-PD-1 therapy. Moreover, there is increasing evidence that tumor suppressor proteins are involved in immune regulation and are capable of modulating the expression of immune checkpoint proteins. Here, we determined the role of p53 protein (gene name: TP53) in the regulation of PD-L1 expression in melanoma. Methods: We analyzed publicly available mRNA and protein expression data from the cancer genome/proteome atlas and performed immunohistochemistry on tumors with known TP53 status. Constitutive and IFN-ɣ-induced PD-L1 expression upon p53 knockdown in wildtype, TP53-mutated or JAK2-overexpressing melanoma cells or in cells, in which p53 was rendered transcriptionally inactive by CRISPR/Cas9, was determined by immunoblot or flow cytometry. Similarly, PD-L1 expression was investigated after overexpression of a transcriptionally-impaired p53 (L22Q, W23S) in TP53-wt or a TP53-knockout melanoma cell line. Immunoblot was applied to analyze the IFN-ɣ signaling pathway. Results: For TP53-mutated tumors, an increased CD274 mRNA expression and a higher frequency of PD-L1 positivity was observed. Interestingly, positive correlations of IFNG mRNA and PD-L1 protein in both TP53-wt and -mutated samples and of p53 and PD-L1 protein suggest a non-transcriptional mode of action of p53. Indeed, cell line experiments revealed a diminished IFN-ɣ-induced PD-L1 expression upon p53 knockdown in both wildtype and TP53-mutated melanoma cells, which was not the case when p53 wildtype protein was rendered transcriptionally inactive or by ectopic expression of p53L22Q,W23S, a transcriptionally-impaired variant, in TP53-wt cells. Accordingly, expression of p53L22Q,W23S in a TP53-knockout melanoma cell line boosted IFN-ɣ-induced PD-L1 expression. The impaired PD-L1-inducibility after p53 knockdown was associated with a reduced JAK2 expression in the cells and was almost abrogated by JAK2 overexpression. Conclusions: While having only a small impact on basal PD-L1 expression, both wildtype and mutated p53 play an important positive role for IFN-ɣ-induced PD-L1 expression in melanoma cells by supporting JAK2 expression. Future studies should address, whether p53 expression levels might influence response to anti-PD-1 immunotherapy.
Article
Full-text available
The fact that a subset of human cancers showed evidence for a spontaneous adaptive immune response as reflected by the T cell‐inflamed tumor microenvironment phenotype led to the search for candidate innate immune pathways that might be driving such endogenous responses. Preclinical studies indicated a major role for the host STING pathway, a cytosolic DNA sensing pathway, as a proximal event required for optimal type I interferon production, dendritic cell activation, and priming of CD8+ T cells against tumor‐associated antigens. STING agonists are therefore being developed as a novel cancer therapeutic, and a greater understanding of STING pathway regulation is leading to a broadened list of candidate immune regulatory targets. Early phase clinical trials of intratumoral STING agonists are already showing promise, alone and in combination with checkpoint blockade. Further advancement will derive from a deeper understanding of STING pathway biology as well as mechanisms of response vs resistance in individual cancer patients.
Article
PurposeEndothelial cell (EC) dysfunction underlies the pathology of multiple disease conditions including cardiovascular and pulmonary diseases. Dysfunctional ECs have a distinctive gene expression profile compared to healthy ECs. RNAi therapy is a powerful therapeutic approach that can be used to silence multiple genes of interests simultaneously. However, the delivery of RNAi to ECs in vivo continues to be a major challenge. Here, we optimized a polymer formulation based on poly(β-amino ester)s (pBAEs) to deliver siRNA to vascular ECs.Methods We developed a library of bioinspired oligopeptide-modified pBAE nanoparticles (NPs) with different physicochemical proprieties and screened them for cellular uptake and efficacy of RNAi delivery in vitro using ECs, vascular smooth muscle cells, and THP-1 monocytes. From the screening, the lysine-/histidine-oligopeptide modified pBAE (C6-KH) NP was selected and further tested ex vivo using mouse aorta and in mice to determine efficiency of siRNA delivery in vivo.ResultsThe in vitro screening study showed that C6-KH was most efficient in delivering siRNA to ECs. Ex vivo study showed that C6-KH nanoparticles containing siRNAs accumulated in the endothelial layer of mouse aortas. In vivo study showed that C6-KH nanoparticles carrying siICAM2 injected via tail-vein in mice significantly reduced ICAM2 level in the artery endothelium (55%), lung (52%), and kidney (31%), but not in the liver, heart, and thymus, indicating a tissue-specific delivery pattern.Conclusions We demonstrate that C6-KH pBAE can used for delivery of siRNAs to the artery endothelium and lung, while minimizing potential side or toxic effects in the liver and heart.
Article
Several immunotherapy approaches that mobilize CD8 ⁺ T cell responses stimulate tumor rejection, and some, such as checkpoint blockade, have been approved for several cancer indications and show impressive increases in patient survival. However, tumors may evade CD8 ⁺ T cell recognition via loss of MHC molecules or because they contain few or no neoantigens. Therefore, approaches are needed to combat CD8 ⁺ T cell–resistant cancers. STING-activating cyclic dinucleotides (CDNs) are a new class of immune-stimulating agents that elicit impressive CD8 ⁺ T cell–mediated tumor rejection in preclinical tumor models and are now being tested in clinical trials. Here, we demonstrate powerful CDN-induced, natural killer (NK) cell–mediated tumor rejection in numerous tumor models, independent of CD8 ⁺ T cells. CDNs enhanced NK cell activation, cytotoxicity, and antitumor effects in part by inducing type I interferon (IFN). IFN acted in part directly on NK cells in vivo and in part indirectly via the induction of IL-15 and IL-15 receptors, which were important for CDN-induced NK activation and tumor control. After in vivo administration of CDNs, dendritic cells (DCs) up-regulated IL-15Rα in an IFN-dependent manner. Mice lacking the type I IFN receptor specifically on DCs had reduced NK cell activation and tumor control. Therapeutics that activate NK cells, such as CDNs, checkpoint inhibitors, NK cell engagers, and cytokines, may represent next-generation approaches to cancer immunotherapy.
Article
Many favorable anti-cancer treatments owe their success to the induction immunogenic cell death (ICD) in cancer cells, which activates antigen presenting cells to prime anti-cancer adaptive immunity. We describe a strategy to synthetically induce ICD by delivering the agonist of stimulator of interferon genes (STING), 2′3′-cyclic guanosine monophosphate–adenosine monophosphate (cGAMP) into tumor cells using hollow polymeric nanoshells. Following intracellular delivery of exogenous adjuvant, subsequent cytotoxic treatment creates immunogenic cellular debris, by a process herein termed synthetic immunogenic cell death (sICD). sICD is indiscriminate to the type of chemotherapeutics adopted for cancer treatment and enables co-localization of exogenously administered immunologic adjuvants and tumor antigens for enhanced antigen presentation and development of anticancer adaptive immunity. In three mouse tumor models with distinctive chemotherapeutic treatments, sICD enhances therapeutic efficacy and restrains tumor progression. The present study highlights the therapeutic benefit of temporally coordinated STING agonists delivery to cancer cells, paving ways to new nanoparticle designs for enhancing anti-cancer chemo-immunotherapies.
Article
Therapeutic targeting of the immune system in cancer is now a clinical reality and marked successes have been achieved, most notably through the use of checkpoint blockade antibodies and chimeric antigen receptor T cell therapy. However, efforts to develop new immunotherapy agents or combination treatments to increase the proportion of patients who benefit have met with challenges of limited efficacy and/or significant toxicity. Nanomedicines — therapeutics composed of or formulated in carrier materials typically smaller than 100 nm — were originally developed to increase the uptake of chemotherapy agents by tumours and to reduce their off-target toxicity. Here, we discuss how nanomedicine-based treatment strategies are well suited to immunotherapy on the basis of nanomaterials’ ability to direct immunomodulators to tumours and lymphoid organs, to alter the way biologics engage with target immune cells and to accumulate in myeloid cells in tumours and systemic compartments. We also discuss early efforts towards clinical translation of nanomedicine-based immunotherapy.
Article
The recognition of DNA as an immune-stimulatory molecule is an evolutionarily conserved mechanism to initiate rapid innate immune responses against microbial pathogens. The cGAS–STING pathway was discovered as an important DNA-sensing machinery in innate immunity and viral defense. Recent advances have now expanded the roles of cGAS–STING to cancer. Highly aggressive, unstable tumors have evolved to co-opt this program to drive tumorigenic behaviors. In this review, we discuss the link between the cGAS–STING DNA-sensing pathway and antitumor immunity as well as cancer progression, genomic instability, the tumor microenvironment, and pharmacologic strategies for cancer therapy. Significance The cGAS–STING pathway is an evolutionarily conserved defense mechanism against viral infections. Given its role in activating immune surveillance, it has been assumed that this pathway primarily functions as a tumor suppressor. Yet, mounting evidence now suggests that depending on the context, cGAS–STING signaling can also have tumor and metastasis-promoting functions, and its chronic activation can paradoxically induce an immune-suppressive tumor microenvironment.
Article
Immunotherapies have significantly improved cancer patient survival, but response rates are still limited. Thus, novel formulations are needed to expand the breadth of immunotherapies. Pathogen associated molecular patterns (PAMPs) can be used to stimulate an immune response, but several pathogen recognition receptors are located within the cell, making delivery challenging. We have employed the biodegradable polymer acetalated dextran (Ace-DEX) to formulate PAMP microparticles (MPs) in order to enhance intracellular delivery. While treatment with four different PAMP MPs resulted in tumor growth inhibition, cyclic GMP-AMP (cGAMP) MPs were most effective. cGAMP MPs showed anti-tumor efficacy at doses 100-1000 fold lower than published doses of soluble cGAMP in two murine tumor models. Treatment with cGAMP MPs resulted in increased natural killer cell numbers in the tumor environment. Immune cell depletion studies confirmed that NK cells were responsible for the anti-tumor efficacy in an aggressive mouse melanoma model. NK cells and CD8 ⁺ T cells were both required for early anti-tumor function in a triple negative breast cancer model. In summary, cGAMP MP treatment results in NK and T cell-dependent anti-tumor immune response.