Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Full Paper

www.afm-journal.de

Multifunctional Nanoparticle Loaded Injectable


Thermoresponsive Hydrogel as NIR Controlled Release
Platform for Local Photothermal Immunotherapy to Prevent
Breast Cancer Postoperative Recurrence and Metastases
Yan Peng Jia, Kun Shi, Fan Yang, Jin Feng Liao, Ru Xia Han, Li Ping Yuan, Ying Hao,
Meng Pan, Yao Xiao, Zhi Yong Qian,* and Xia Wei Wei*

1. Introduction
For breast cancer patients who have undergone breast-conserving surgery,
effective treatments to prevent local recurrences and metastases is very Breast cancer has been the leading cancer
diagnosed in female globally.[1,2] Surgical
essential. Here, a local injectable therapeutic platform based on a thermo-
resection, especially breast-conserving sur-
sensitive PLEL hydrogel with near-infrared (NIR)-stimulated drug release is gery (BCS) has been a preferred therapy
developed to achieve synergistic photothermal immunotherapy for preven- method for primary breast cancer with its
tion of breast cancer postoperative relapse. Self-assembled multifunctional advantage of better cosmetic results.[3,4]
nanoparticles (RIC NPs) are composed of three therapeutic components Unfortunately, the residue tumor cells
including indocyanine green, a photothermal agent; resiquimod (R848), a around the resection cavity usually lead
to quick recurrence and metastasis, which
TLR-7/8 agonist; and CPG ODNs, a TLR-9 agonist. RIC NPs are physically could result in patient death.[5–7] Worse
incorporated into the thermosensitive PLEL hydrogel. The RIC NPs encap- still, for postoperative patients, severe
sulated PLEL hydrogel (RIC NPs@PLEL) is then locally injected into the local immunosuppression associated
tumor resection cavity for local photothermal therapy to ablate residue tumor with wound healing and inflammation
tissues and produce tumor-associated antigens. At the same time, NIR also further increases the incidence of relapse
and metastases by promoting cancer cell
triggers the release of immune components CPG ODNs and R848 from ther-
invasion and suppressing the activity of
moresponsive hydrogels PLEL. The released immune components, together antitumor immune cells.[8,9] Although
with tumor-associated antigens, work as an in situ cancer vaccine for postsur- therapies including chemotherapy and
gical immunotherapy by inducing effective and sustained antitumor immune radiotherapy are often provided after sur-
effect. Overall, this work suggests that photothermal immunotherapy based gery in clinics, they bear disadvantages
on local hydrogel delivery system has great potential as a promising tool for such as undesirable side effects, treat-
ment resistance, or radiation-induced
the postsurgical management of breast cancer to prevent recurrences and damage.[10,11] Therefore, developing effec-
metastases. tive and safe strategies to address relapse
and metastases for patients undergone the
BCS are urgently needed.
Recently, immunotherapy has shown great potential to
Y. P. Jia, Dr. K. Shi, F. Yang, R. X. Han, L. P. Yuan, Dr. Y. Hao, M. Pan,
prevent tumor regression and metastasis by utilizing the
Y. Xiao, Prof. Z. Y. Qian, Prof. X. W. Wei immune system of the body to kill the tumor cells.[12–14] Alter-
State Key Laboratory of Biotherapy and Cancer Center natively, photothermal therapy (PPT) represents a noninva-
National Clinical Research Center for Geriatrics sive therapy in which heat generated by photothermal agents
West China Hospital upon near-infrared (NIR) light irradiation is applied to kill
Sichuan University
Chengdu 610041, P. R. China cancer cells. Moreover, the tumor-associated antigens gener-
E-mail: zhiyongqian@scu.edu.cn; xiaweiwei@scu.edu.cn ated in situ after photothermal tumor ablation could present
Dr. J. F. Liao immune vaccine-like functions.[15–17] Considering the immu-
State Key Laboratory of Oral Diseases nosuppressive microenvironment after surgery, strategies
West China School of Stomatology based on synergistic photothermal immunotherapy thereby
Sichuan University
Chengdu 610041, P. R. China
have great potential to control postsurgery local relapse and
metastasis.[18]
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/adfm.202001059. Although photothermal immunotherapy shows such advan-
tages, overactive immune system due to the systemic adminis-
DOI: 10.1002/adfm.202001059 tration of therapeutic agents could also lead to either adverse

Adv. Funct. Mater. 2020, 2001059 2001059 (1 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Scheme 1. Schematic illustration of a) the preparation process of RIC NPs@PLEL hydrogels and b) photothermal-immune therapy to prevent
postsurgery tumor reoccurrence.

effects or limited administration dosage.[19] In this regard, local As mentioned above, with all the advantages of photo-
drug delivery represents a desirable administration method for thermal immunotherapy, NIR controlled drug release as well
postsurgical therapy.[20–23] Injectable biodegradable hydrogels as localized hydrogel delivery system, exploring the feasibility
are among the best candidates for local drug release with the of designing an all-in-one powerful hydrogel therapeutic plat-
following merits: higher dosage at tumor site than the systemic form for effective prevention of postresection recurrence and
approach; minimized adverse effects to normal tissues; a sus- metastasis is very necessary. However, to the best of our knowl-
tained and even controlled drug release; moreover, multi­ ple edge, there are almost no previous reports proposed it. Herein,
drugs that act synergistically could be incorporated into the in this study, we developed a localized photothermal immuno-
same hydrogel together for combination therapy.[21,24–27] In therapy platform for postresection prevention of breast cancer
particular, injectable intelligent hydrogels have been widely recurrences and metastases as shown in Scheme 1. An inject-
explored as locally controlled drug delivery system with the able thermosensitive PDLLA-PEG-PDLLA (PLEL) hydrogel
unique property of sol–gel phase transition when triggered by incorporating three-component nanoparticles RIC NPs was
stimuli such as pH, temperature, and light.[28–31] Among them, fabricated as a localized NIR-controlled drug delivery system.
temperature-responsive hydrogels represent an ideal candi- PDLLA-PEG-PDLLA was prepared as described before.[32] RIC
date for sustained and controlled local drug release. They are NPs is formed by self-assembly technology with indocyanine
sol at room temperature, which allow them to load agents and green (ICG), resiquimod (R848), and CPG ODNs. ICG is an
be injected into the tumor site, then they transformed into gel FDA-approved excellent NIR fluorescent dye for PTT with supe-
upon injection at the physiological temperature. As a result, rior photothermal effect. R848 is a small molecular agonist of
loaded drugs can be released from the hydrogel in a sustained TLR-7/8, CPG ODNs is TLR-9 agonist, they are both used to
way.[30,32] To be noted, some temperature-responsive hydrogels activate dendritic cells (DCs) via multiple TLRs and stimulate
can undergo further transformation such as gel to sol again proinflammatory cytokines release. However, the three func-
when triggered by a higher temperature, making it possible to tional components have either poor aqueous stability or easy
release drug in a controlled pattern.[32] Recently, except to be degradation issues. Here our hydrogel local delivery system
applied in photothermal therapy, NIR responsive photothermal solved these problems. Upon local injection, RIC NPs@PLEL
agents have also been reported as effective heat generators for systems changed from liquid state at room temperature to the
controlled drug release.[28,30] Therefore, for temperature-sensi- gel (above 37 °C). Upon further NIR laser irradiation, ICG gen-
tive hydrogels, developing NIR as switches to achieve locally erated high temperature (above 45 °C) promote further trans-
controlled drug release has great potential in tumor therapy formation of the gel to sol, thus triggering on-demand release
application. of R848 and CPG ODNs, together with the tumor-associated

Adv. Funct. Mater. 2020, 2001059 2001059 (2 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 1. A) TEM image and B) particle size distribution of RIC NPs. The RIC NPs were negatively stained with 1% phosphotungstic acid for TEM
imaging. C) UV–vis absorption spectra of RIC NPs. D) Near infrared imaging of blank PLEL hydrogel, free ICG, and ICG@PLEL hydrogel under 808 nm
laser irradiation. E) Temperature changes of (D) under NIR laser irradiation.

antigens generated in situ after PTT of residue tumor, they breast cancer recurrence, the excellent photothermal effect
play vaccine-like functions by inducing subsequent antitumor is expected to achieve photothermal ablation of postsurgical
immune response. residue tumor tissues and thermal-induced immunotherapy.
Therefore, we investigated the photothermal effect of free ICG
and ICG loaded PLEL hydrogels to confirm the photothermal
2. Results and Discussion effect of ICG is not weakened when incorporating into the
hydrogel, blank PLEL hydrogel was used as control. As shown
2.1. Preparation and Characterization of RIC NP Loaded in Figure 1D,E, under 808 nm laser irradiation (1 W cm−2), the
PLEL Hydrogels temperature of the hydrogel increased to almost 60 °C within
1 min, and finally reached about 70 °C over time. These results
RIC NPs are prepared by self-assembly method of three mole- verified no obvious difference of photothermal ability between
cules ICG, CPG, and R848. ICG is an NIR dye with strong photo­ free ICG and ICG@PLEL hydrogels, implying that being
thermal effect, CPG and R848 work as important immune encapsulated into hydrogel did not affect the photothermal
stimulators by activating the TLRs of DCs. To be noted, the effect of ICG.
three components we chose are of good biocompatibility, which Next, the viscoelastic behavior of RIC NP loaded PLEL
make it a promising candidate for further clinical application. hydrogels and blank PLEL hydrogels were evaluated. The
The successful preparation of RIC NPs is demonstrated by results confirmed a similar temperature response (10–60 °C)
the transmission electron microscope image which showed a between them (Figure 2A,B). For both RIC NP loaded PLEL
well-defined morphology and uniform size of about 180 nm of hydrogels and blank PLEL hydrogels, the storage modulus
the RIC NPs (Figure 1A). The mean hydrodynamic size of RIC (G′) was higher than loss modulus (G″) at a physical tempera-
NPs measured is about 280 nm as shown in Figure 1B. The ture of 37 °C, indicating the gel status of the hydrogel. When
UV–vis absorption spectrum of RIC NPs revealed the charac- the temperature reached approximately 45 °C, the hydrogel
teristic absorption peaks of ICG at about 780 nm, CPG at about remained a higher loss modulus (G″) than storage modulus
260 nm, and R848 at about 340 nm, further suggesting the RIC (G′), which revealed that the hydrogel returned to the flowing
NPs were successfully prepared (Figure 1C). The PLEL hydrogel sol state. These findings demonstrated our hydrogel system
is synthesized and characterized as described in our previous exhibited optimal photothermal effect and thermosensitive
studies.[32] The RIC NPs is incorporated into PLEL hydrogels property. We next observed the appearance changes of RIC NP
by physically mixing them with certain weight ratios. As we loaded PLEL hydrogels at different temperatures. As presented
intended to apply RIC NP loaded PLEL hydrogels for photo- in Figure 2C, the hydrogel has good liquidity at 4 °C. With
thermal enhanced immunotherapy to prevent the postoperative the increase of temperature, the hydrogel underwent sol–gel

Adv. Funct. Mater. 2020, 2001059 2001059 (3 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 2. A,B) Temperature dependence of storage (G′) and loss modulus (G″) for blank PLEL and RIC NPs@PLEL as a function of temperature.
C) Reversible sol–gel transformation photos of PLEL hydrogels and RIC NPs@PLEL hydrogels.

transformation at 37 °C, and swelling behavior was observed. 2.2. In Vitro and In Vivo Biocompatibility Assay
When the temperature came to 55 °C, hydrogel shrinkage and
gel–sol transformation occurred. To be noted, the whole trans- Although PLEL has been applied with good biocompatibility
formation process from 4 to 55 °C was reversible. This result before, it is still necessary to evaluate its biosafety as a synthe-
confirmed the thermally responsive property of RIC NP loaded sized material. As for the three components in RIC NPs, ICG
PLEL hydrogels. Furthermore, Figure 2C also implied that laser as a photothermal agent should undergo safety test to con-
irradiation could directly control the rheological status of RIC firm its safety, while for R848 and CPG ODNs, there is usu-
NP loaded PLEL hydrogel by photothermal effect. Therefore, it ally no need since they could function with small dose and
is concluded that the RIC NPs@PLEL hydrogels is a promising they work by stimulating immune cells such as DCs. The cyto-
candidate for topical photothermal cancer therapy and it may toxicity of ICG@PLEL hydrogel was thus investigated via the
serve as a local drug delivery system capable of NIR-triggered 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide
drug release. (MTT) assay. As shown in Figure 3A,B, 4T1 and RAW246.7 cell

Figure 3. In vitro cell viability of A) DC 2.4 cells and B) RAW 246.7 cells incubated with ICG@PLEL hydrogels at various concentrations. C–E) Blood
biochemistry data of mice in the PLEL group and ICG@PLEL group (ALT: alanine aminotransferase; AST: aspartate aminotransferase; ALP: alkaline
phosphatase; TP: total protein; BUN: blood urea nitrogen).

Adv. Funct. Mater. 2020, 2001059 2001059 (4 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

lines were respectively cocultured with the nanocomplexes at the results were further quantified as shown in Figure 4B. For
various concentrations for 24 h. The assays showed that ICG@ free ICG, the fluorescence intensity weakened rapidly within
PLEL exhibited little cytotoxicity in the given concentration 24 h revealing that the free ICG diffuses away or even degraded
range with the cell viability all higher than 80%. For in vivo very rapidly. In contrast, fluorescence intensity of ICG encapsu-
application, we estimated the biosafety of our PLEL hydrogel lated in PLEL hydrogel remained strong all over the monitoring
system. 14 days after injection of PLEL hydrogel or ICG@PLEL. period. These findings proved that the PLEL hydrogel could
The blood of mice in each group was collected and subjected to efficiently extend the local release of its payloads. Such drug
a blood chemical analysis. There were no detectable changes in sustainable release property of PLEL hydrogel is promising in
the concentration of the biomarkers of liver enzymes including keeping relatively stable and long-term drug concentrations in
alanine aminotransferase and aspartate aminotransferase, vivo for perdurable therapy.
kidney function (blood urea nitrogen) in the blood across treat- For NIR controlled release test, BALB/c mice bearing 4T1
ment groups (Figure 3C–E). Besides, the injection site was also tumor were injected with cy-3 labeled CPG@PLEL intratumorally
collected for histopathological analysis and no inflammation and divided into two groups of CPG@PLEL hydrogel and CPG@
was observed (Figure S2, Supporting Information). PLEL hydrogel/NIR. For the CPG@PLEL hydrogel/NIR group,
NIR irradiation was performed 1 h before IVIS imaging. Mice
in both groups were then monitored at different time points by
2.3. Extended and Controlled Release Behavior of RIC NP fluorescence IVIS imaging (Figure 4C,D). It is found that the flu-
Loaded PLEL Hydrogels orescence signal of CPG@PLEL group without laser treatment
still retained intense fluorescence signal for 48 h. In contrast, for
In recent years, smart drug release behavior triggered by multi- the CPG@PLEL hydrogel/NIR group, the fluorescence intensity
stimuli such as NIR laser, pH, temperature, and enzymes have decreased a lot compared with CPG@PLEL without NIR. Similar
showed enormous advantages in drug delivery application.[32] results were observed when the laser treatment was repeated for
Since our PLEL hydrogel has thermal sensitive property, we another two cycles, suggesting that the release rate of CPG could
then studied the NIR laser triggered drug release behaviors be regulated intelligently by switching on or off of NIR laser.
of PLEL hydrogel to see if it could serve as an ideal scaffold Thus, these data demonstrated that PLEL hydrogel can serve as a
for NIR controlled drug release. As well known, hydrogel has stable, biodegradable depot with the release of anticancer drugs
the advantage of sustained drug release property. First, we controlled by the stimuli of NIR irradiation.
confirmed that our hydrogel PLEL could extend the release of
its payloads in vivo. As a model drug, ICG was used as free
ICG solution or ICG@PLEL hydrogel and both were injected 2.4. DC Maturation Assay In Vitro
locally to the side of healthy female BALB/c mice. Then, the
mice were monitored at different time points through fluores- DCs, the most important antigen-presenting cells (APCs), play
cence in vivo imaging system (IVIS) imaging (Figure 4A), and crucial roles in antitumor immunity. Immature DCs (iDCs)

Figure 4. A) In vivo extended release of ICG from ICG@PLEL hydrogel recorded by IVIS and B) quantitative analysis. C) In vivo controlled release of
CPG from PLEL hydrogel recorded by IVIS and D) quantitative analysis.

Adv. Funct. Mater. 2020, 2001059 2001059 (5 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

collect antigens from the surrounding fluid and then process promoting intratumoral infiltration and proliferation of effector
them into peptides during migration to nearby draining lymph CTLs. Although in all the rest groups, tumor recurrence hap-
nodes, where they present the peptide major histocompatibility pened on the whole mice, there are still differences in the delay
complex (MHC) to T cell receptors (TCR) for T cell activation. of recurrence. The quickest recurrence occurred in the PLEL
The maturation of DCs could be confirmed by co-stimulatory group. Figure 6E analyzed the tumor volume among time after
molecules CD80 and CD86, the typical markers indicating therapy, the results further confirmed the most effective pre-
maturation of DCs. After bone marrow-derived dendritic cells vention of recurrence in RIC NPs@PLEL/NIR group. However,
(BMDCs) were treated in various formulations for 24 h, the due to the sensitivity problems of luciferase during biolumines-
percentage of DCs maturation (CD11c+CD80+CD86+) was cence images, the IVIS results in Figure 7A existed deviations
then analyzed by flow cytometry (Figure 5A–E). It turned out from the actual results. Even so, the IVIS results still revealed
that DCs maturation in the RIC NPs@PLEL/NIR group was up the recurrence rate varies among different groups. In the PLEL
to 32%, the highest among all groups. In contrast, the same group, recurrence happened at an earlier time among all the
amount of RIC NPs@PLEL without NIR induced a little less mice when compared with other therapeutic groups. On the
DC maturation of 25%. To be noted, ICG@PLEL/NIR group whole, RIC NPs@PLEL/NIR group presented the best result.
alone also promoted DC maturation three times higher than Besides, representative mice photos of tumor site were also
the blank group suggesting the photothermal effect did trigger recorded to show the tumor recurrence in Figure 7B. Hema-
immune response. Since matured DC secretes immune-related toxylin and eosin (H&E) staining of tissues including heart,
cytokines such as tumor necrosis factor α (TNF-α), we meas- liver, spleen, and kidney in various groups indicated that there
ured the concentration of cytokines in the medium supernatant is no visible tissue damage (Figure 7C). In addition, except for
by ELISA kit. It was found that the secretion levels of TNF-α the modest decrease after surgery, no obvious body weight
and IL-6 totally enhanced after RIC NPs@PLEL/NIR treatment loss was observed across all groups during the whole therapy
(Figure 5F–G). These results implied that tumor-associated period (Figure 7D), implying the high biocompatibility of our
antigens liberated from PPT-triggered tumor cell lysis, together hydrogel system. Lung metastasis is another malignant clin-
with TLR agonists R848 and CPG ODNs, they induced DC ical problem for breast cancer patients undergone operation.
maturation in vitro efficiently. Evaluation of lung metastasis is also a very important factor
for therapy effectiveness assessments. As shown in Figure 8A,
the photos of lung tissues clearly revealed that for lung tissue
2.5. In Vivo Inhibition of Breast Cancer Recurrence in the untreated group severe metastases occurred with the
by Photothermal Immunotherapy most lung nodules found. For the RIC@PLEL hydrogels/NIR
group, the lung tissue kept health with no metastasis nodules
The superior photothermal effect, relatively precise NIR con- found. HE analysis of lung tissues further confirmed these
trolled local drug release, as well as strong immune effect of results. Besides, the lung weight and the number of pulmonary
RIC NPs@PLEL hydrogel motivated us to further explore its nodules among all the groups were also analyzed. Compared
potential application in anticancer photothermal immuno- with the untreated group, the lung weight and nodules both
therapy. Considering hydrogel’s great advantages in local drug decreased in different therapy groups. (Figure 8B,C). Taken
delivery especially for postresection model, we expected that together, these results proved that during the in vivo therapy
our RIC@PLEL hydrogel could prevent local recurrence and process, the RIC NPs@PLEL/NIR hydrogels mediated photo-
metastases of breast cancer after surgery. The design of our in thermal ablation, as well as photothermally triggered release of
vivo experiment was shown in Figure 6A. Female BALB/c mice RIC NPs in resection site, effectively elicits systemic immunity
with orthotopic 4T1-Luc breast cancer was used. Half an hour response against both tumors recurrence and metastasis.
after resection of primary tumor as shown in Figure S1 in the To understand the fundamental immune mechanisms behind
Supporting Information, the hydrogel-based therapeutic formu- the tumor inhibition effects of RIC@PLEL hydrogels/NIR, we
lations were injected into the surgical bed. For NIR laser irra- determined the DCs and T cells proliferation and activation by
diation groups, the temperature of injection site rose to about flow cytometry. For in vivo DC maturation analysis, 4T1-bearing
60 °C rapidly under 808 nm laser irradiation, it is high enough mice were intratumorally administrated with various PLEL-based
to kill the residue tumor tissues (Figure 6B). This result also formulations, then sacrificed 3 days later to harvest the draining
demonstrated the excellent photothermal effect of ICG@PLEL LNs for flow cytometry analysis of DCs maturation. As shown
hydrogel for in vivo application. The growth of the postresec- in Figure 9A,C, RIC NPs@PLEL/NIR remarkably activated 24%
tion residue tumors in each group was monitored every other DC maturation while for RIC NPs@PLEL without NIR irradia-
day for one mouth. As shown in Figure 6C, the survival rates tion the percentage was a little lower. In comparison, bare NIR
in all the therapeutic groups are encouraging 100%, except 80% irradiation in ICG@PLEL group also promoted DC maturation
in ICG@PLEL/NIR group, suggesting that PTT alone could slightly, which could be attributed to NIR-triggered liberation
destroy the tumor residues and induce antitumor immune of tumor antigens. Furthermore, the serum levels of TNF-α
response to some extent. However, when talking about recur- also showed significant up-regulation in mice after RIC NPs@
rence rate, we found that only in RIC NPs@PLEL/NIR group, PLEL/NIR treatments (Figure 9E). Overall, the in vivo findings
80% of the mice were completely cured without recurrence verified that our RIC NPs@PLEL/NIR could efficiently induce
(Figure 6D). This could be attributed to the antitumor immune DC maturation and may act as an in situ vaccine to initiate
response since NIR laser irradiation triggering tumor antigen the immune responses. As well known, CD8+T cells, called
generation as well as R848 and CPG ODN release, which cytotoxic T lymphocytes (CTL), could directly recognize and

Adv. Funct. Mater. 2020, 2001059 2001059 (6 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 5. A) Expression levels of surface molecules (CD80 and CD86) on BMDCs after various treatments in transwell system. B) Schematic illustra-
tion of transwell system for DCs/4T1 tumor cells coculture. C–E) Quantification of expression levels of CD80 and CD86 on the surface of BMDCs.
F,G) TNF-α and IL-6 secreted in the BMDC cell culture supernatant after different treatments. **p < 0.01 and ***p < 0.001.

Adv. Funct. Mater. 2020, 2001059 2001059 (7 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 6. A) Schematic diagram of the postsurgery breast cancer model and postoperative therapy of the tumor. B) IR thermal images of postsurgery
4T1 residue tumor-bearing mice with 808 nm laser irradiation. C) Survival curves of mice after various treatments. D) Recurrence-free rate of mice after
various treatments. E) Tumor volume curves after various treatments.

Figure 7. A) In vivo bioluminescence images to evaluate the recurrence of 4T1-Luc tumor after various treatments. B) Typical photographs of mice to
evaluate the recurrence of 4T1-Luc tumor after various treatments. C) H&E staining of mice tissues after various treatments. D) Body weight curves
after various treatments. The scale bar indicated 100 µm.

Adv. Funct. Mater. 2020, 2001059 2001059 (8 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 8. A) Typical photographs and H&E staining of 4T1 metastatic foci for mice after different treatments. The scale bar indicated 100 µm. B) Lung
weight at the end of different treatments. C) Lung metastatic nodules at the end of different treatments. **p < 0.01 and ***p < 0.001.

kill cancer cells. On the other hand, CD4+T cells play impor- unlike free CpG ODNs or R848, nanotechnology has further
tant roles in the regulation of adaptive immunities. There- improved the in vivo stability and DCS uptake efficiency of
fore, we collected spleens of mice in all groups after different these adjuvants. Collectively, the basic mechanism of hydrogel
treatments to test the CD8+ (CD3+CD4−CD8+) and CD4+ system’s therapeutic effect is the activation of DCs, and sub-
(CD3+CD4+CD8−) T cells by flow cytometry (Figure 9B,D). The sequent exposure of the activated DCs to tumor antigens in
percentage of CD8+T cells in RIC NPs@PLEL/NIR group was vivo so that a tumor-specific T cell response is induced. RIC
dramatically increased by about twofold compared to the con- NPs@PLEL/NIR hydrogel could thus be considered an in situ
trol. This result confirmed that the combination of RIC NPs@ autologous cancer vaccine that utilizes whole tumor cells as the
PLEL vaccination and laser irradiation significantly activated source of tumor antigens.
the antitumor immune response. In contrast, no significant It is reported that in the process of photothermal ablation
enhancement of CD4+T cell was observed in therapy groups. of cancer, the increased temperature, as an exogenous cellular
To be noted, with the thermal sensitive property, our PLEL stress, killed tumor cell mostly through necrosis. Necrotic cells
hydrogel is injectable at room temperature. Once injected into released tumor associated antigens as well as damage asso-
the surgical bed where the temperature is 37 °C, the hydrogel ciated molecular pattern molecules (DAMPs). DAMPs are
transformed into gel condition with no mobility as shown in inflammatory mediators including ATP, nucleic acids and intra-
Figure S2 in the Supporting Information, which is advanta- cellular proteins such as heat shock proteins (Hsp70) and high-
geous for local administration and extended release of thera- mobility group box 1 (HMGB-1). [33] DAMPs working as danger
peutic components. The efficient inhibition of local breast signals have attracted much attention in recent years in cancer
cancer recurrence by the RIC NPs@PLEL hydrogel was due research. These inflammation molecules with immunostimu-
to the synergetic effects of photothermal therapy and immu- latory properties are normally existed within live cells. Once
notherapy. First, the in situ injection of hydrogel system can secreted by damaged tumor cells, they are found to improve
effectively eliminate residual tumor tissues by photothermal adaptive antitumor immunity by promoting maturation of
ablation. Second, PPT itself can initiate moderate immunolog- antigen-presenting cells and production of cytokines. [34] These
ical responses by triggering tumor associated antigens release DAMPs are reported to bind specific pattern-recognition recep-
from tumor lysis and some “danger signals” such as HSP tors (PRRs) such as Toll-like receptors (TLRs) and trigger their
70.[33] Third, RIC NPs, containing typically used immunoadju- activation. [35] As well known, PRRs are important costimula-
vants R848 and CPG ODNs, were released from hydrogel upon tory molecules expressed by natural immune cells and play an
NIR irradiation to further enhance immune response. Fourth, important role in regulating innate immune response. Notably,

Adv. Funct. Mater. 2020, 2001059 2001059 (9 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Figure 9. Representative flow cytometry plots showing A) matured DCs in lymph node and B) cytotoxic CD3 + CD8 + T cells and CD3 + CD4 + T cells
in spleens after various treatments. C,D) Quantitation of A) the matured DCs and B) CD8 + T cells and CD4+T cells in the spleen. E) TNF-α levels
detected by ELISA assay in the serum of mice after different treatments. *p < 0.05 and **p < 0.01.

the immune effect of photothermal ablation during tumor simple fabrication procedure of RIC NPs@PLEL hydrogel,
treatment deserves more exploration. as well as its ability to simultaneously induce a photothermal
enhanced immune response, holds great potential for clinical
prevention of postsurgical recurrence of breast cancer.
3. Conclusion
In summary, we developed a local injectable therapeutic platform 4. Experimental Section
by loading multifunctional nanoparticles RIC NPs (composed
of photothermal agent ICG, immunoadjuvants R848, and CPG Materials: Poly (ethylene glycol) (PEG, Mn = 1500, 1000, and 2000,
respectively), stannous octoate (Sn (Oct)2, 95%), ε-caprolactone
ODNs) into thermoresponsive hydrogel PLEL. The hybrid RIC (ε-CL), and MTT were obtained from Sigma-Aldrich (USA). Penicillin–
NPs@PLEL hydrogel system exhibited excellent biocompat- streptomycin, DMEM medium, RMPI 1640 medium, and fetal bovine
ibility both in vitro and in vivo. In the orthotopic breast cancer serum (FBS) were all purchased from Gibco (USA). CpG ODNs
model undergone breast-conserving surgery, the RIC NPs@ (5′-TCC ATG ACG TTC CTG ATG C-3′) and cy3-labeled CpG ODNs were
PLEL system successfully ablated residue tumors through NIR- provided by Sangon Biotech (Shanghai, China). R848 was purchased
induced photothermal ablation. Furthermore, the photothermal from InvivoGen (France). ICG was purchased from melonepharma
(Dalian, China). TNF-α, IL-6 ELISA kits were purchased from Invivogen.
ablation, together with the loaded immunoadjuvant, triggered a
Fluorochrome-labeled antibodies were all purchased from eBioscience
strong immune response to inhibit lung metastasis and induced (USA). Recombinant mouse GM-CSF and IL-4 were purchased from
a strong antitumor immunity to prevent locoregional tumor Peprotech. All other chemical reagents used in this study were of
recurrence. Taken together, our strategy demonstrated that the analytical grade and used without further purification.

Adv. Funct. Mater. 2020, 2001059 2001059 (10 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Preparation and Characterization of RIC NPs@PLEL Hydrogel: supplemented with 10 ng mL−1 mouse granulocyte macrophage colony
RIC nanoparticles were prepared by the self-assembly method of stimulating factor (GM-CSF) and 1 ng mL−1 mouse IL-4. Culture media
CpG ODNs, R848, and ICG solution. In brief, a certain amount of were refreshed every 2 days. After 7 days incubation, the nonadherent
CpG ODNs, R848, and ICG was weighted and dissolved by deionized and loosely adherent cells were harvested as dendritic cells. DC
(DI) water, and then mixed and stirred for half an hour to form RIC purity was evaluated by flow cytometry (ACEA Biosciences, Inc.). The
nanoparticles. The dynamic nanoparticle size and zeta potential of percentage of CD11c positive cells in the obtained BMDC was more than
resultant RIC NPs were determined by Nano ZS (Malvern Instruments, 70%.
UK), and the UV–vis absorption spectra were measured by the UV–vis BMDC Maturation Assay: For in vitro DC stimulation experiments,
absorption spectrometry (PE, USA). Transmission electron microscope a transwell system was used in which the upper chamber was saline,
(TEM) (JEM-2100F, JEOL Ltd., Japan) was also used to confirm the visual PLEL hydrogel, ICG@PLEL/NIR, RIC NPs@PLEL, and RIC NPs@PLEL/
morphology of RIC NPs. PLEL thermal-sensitive hydrogel was prepared NIR, respectively, while the lower chamber was seeded with immature
and characterized by the group as described previously. [32] RIC NPs DCs. After various treatments for 24 h, DCs in all groups were collected,
incorporated PLEL hydrogel (RIC NPs@PLEL) was prepared by simple washed, and stained with antibodies of PE antimouse CD11c, APC
physical mixing with a certain ratio. The rheology of PLEL hydrogel and antimouse CD86 and FITC antimouse CD80, and then matured DCs
RIC NPs@PLEL hydrogel were measured by using an HAAKE Rheostress were analyzed by flow cytometry (ACEA Biosciences, Inc.). Meanwhile,
6000 rheometer (Thermo Scientific, USA). The storage modulus (G′) and the culture medium in the transwell system was also collected for
loss modulus (G″) were measured as functions of temperature between measurement of proinflammatory cytokines TNF-α and IL-6 with
10 to 60 °C. In addition, the appearance changes of RIC NP loaded PLEL cytokine ELISA assay.
hydrogels at different temperatures were recorded by taking photos. In Vivo DC Maturation Assay: For in vivo DC maturation experiment,
In Vitro Photothermal Effect: For in vitro evaluation of photothermal when 4T1 tumors inoculated on BALB/c mice reached 100 mm3, saline,
property of ICG, free ICG, PLEL hydrogel, and ICG@PLEL hydrogel PLEL hydrogel, ICG@PLEL/NIR, RIC NPs@PLEL, and RIC NPs@PLEL/
were added into EP tubes and irradiated with an 808 nm NIR laser at a NIR were injected intratumorally. For NIR treated group, mice were
power density of 1.0 W cm−2 for 5 min. This process was simultaneously irradiated with 808 nm NIR laser at 1.5 W cm−2 for 5 min. Three days
recorded by a Fluke Ti32 Infrared (IR) thermal camera (Fluke Ti32, USA) later, mice were then sacrificed to harvest draining lymph nodes to
In Vitro Cytotoxicity Test: Mouse breast cell line 4T1 and luciferase examine DC maturation by flow cytometry. For cytokine detection, serum
labeled 4T1 were originally from ATCC and cultured in complete RMPI samples in all groups were collected from mice at 72 h after various
1640 (supplemented with 10% fetal bovine serum, 1% penicillin, and treatments. TNF-α in the serum were analyzed with ELISA kits.
1% streptomycin). Macrophage cell line RAW246.7 and DC2.4 were In Vivo Inhibition of Breast Cancer Recurrence: Female BALB/c mice
maintained in complete Dulbecco’s Modified Eagle Medium (DMEM). were inoculated orthotopically with 1 × 105 4T1-Luc cells in the second
Cells were all incubated in a humidified incubator at 37 °C with 5% CO2. mammary fat pad. When tumor volumes reached around 100 mm3,
MTT assays were used to detect cellular viability of ICG@PLEL with mice were randomly divided into five groups. Before surgery, mice
RAW246.7 and DC2.4 cell lines. Briefly, RAW246.7 and DC2.4 cells were were anesthetized by intraperitoneal injection 4% chloralhydrate.
seeded in transwell plates and incubated for 24 h. Then, ICG@PLEL of Then, local resection was performed with a layer of surrounding skin
various concentrations were added and incubated for another 24 h. MTT left to construct the local postoperative recurrence model, and then
solution was used to determine cell viabilities. wound was sutured with medical clips and threads. Half an hour later,
Systemic Toxicity Evaluation: Female wide-type BALB/c (6–8 weeks mice in each group were injected with the following formulations in
old) were purchased from Beijing HFK Bioscience Co., Ltd. They were the resultant cavity: PLEL hydrogel, ICG@PLEL hydrogel/NIR, ICG-
housed in an SPF environment with a temperature of 25 ± 2 °C and a R848-CPG NPs@PLEL hydrogel, and ICG-R848-CPG NPs@PLEL
relative humidity of 50–60% with 12 h light–dark cycles. Free access to hydrogel/NIR. Untreated group was used as control. For the groups
food and water was allowed. The animal experiments were all approved subjected to NIR treatment, 808 nm laser irradiation (1.5 W cm−2,
by the Animal Care and Use Committee of Sichuan University (Chengdu, 5 min) was performed 24 h postsurgery. Thermal imaging was
China) and were carried out according to the approved guidelines. All captured using an infrared thermal imaging camera (Ti27, Fluke, USA)
mice were treated humanely. Female BALB/c mice (3 mice per group) simultaneously. The NIR laser irradiation was repeatedly performed on
were injected with the PLEL hydrogel and ICG@PLEL hydrogel via sc days 3 and 5 after surgery. Body weight and tumor recurrence were
injection. Two weeks later, serum samples were collected to evaluate the monitored every 2 days.
liver/kidney functions. In Vivo Bioluminescence Imaging: After treatments, mice were
In Vivo Extended and Controlled Drug Release: To evaluate the in inspected by bioluminescent IVIS imaging weekly for local tumor
vivo sustained release behaviors of PLEL hydrogel, ICG incorporated recurrence detection. In brief, substrate of Luc2 called d-luciferin was
hydrogels were injected subcutaneously into nontumor-bearing mice. As intraperitoneally injected (150 mg kg−1). 10 min later, the mice were
control, free ICG solutions were used. Fluorescence was then acquired anesthetized under 2% isoflurane and imaged by the IVIS (Perkin
at different time point using the IVIS Spectrum (Perkin Elmer). Elmer).
To further monitor in vivo NIR controlled release of CpG ODNs, At the end of the experiment, mice were sacrificed and lung and other
BALB/c mice were subcutaneously inoculated with 1 × 106 4T1 cells. tissues of each group were harvested. Furthermore, the lung weight and
When the tumor size reached 100 mm3, tumor-bearing mice were pulmonary nodule of each group were counted and recorded. Then, the
divided into two groups of cy3-CPG ODNs@PLEL and cy3-CPG samples were fixed for H&E histological analysis.
ODNs@PLEL/NIR. For mice in cy3-CPG ODNs@PLEL group, 50 µL Analysis of Immunocell In Vivo: To analyze the immune effect induced
of cy3-CPG ODNs@PLEL were intratumorally injected, and for mice by these treatments, when 4T1 tumors inoculated on BALB/c mice
in the cy3-CPG ODNs@PLEL/NIR group, cy3-CPG ODNs/ICG@PLEL reached 100 mm3, saline, PLEL hydrogel, ICG@PLEL/NIR, RIC NPs@
were intratumorally injected. One hour later, mice in the NIR group were PLEL, and RIC NPs@PLEL/NIR were injected intratumorally. For NIR
irradiated upon 808 nm NIR laser (1.50 W cm−2, 5 min). Afterward, mice treated group, mice were irradiated with 808 nm NIR laser at 1.5 W cm−2
in the two groups were imaged by the IVIS Spectrum (Perkin Elmer). At for 5 min. Six days later, the same formulations were administrated
24 and 48 h after injection, the same process was repeated. repeatedly. Mice were sacrificed on day 12 and spleens were harvested
Generation of Immature BMDCs: BMDCs were isolated from 6 week and prepared as single cell suspensions, then stained using antimouse
old C57BL/6 mice according to the established protocol.[13] Briefly, mice antibodies of anti-CD3-FITC, anti-CD8a-APC, and anti-CD4-PerCP, and
were sacrificed and the femurs and tibiae were collected to harvest cells analyzed by flow cytometry.
from bone marrow. The obtained cells were resuspended in red blood Statistical Analysis: All data in the study were indicated as mean result ±
cell lysis buffer (Beyotime) to deplete erythrocytes and then counted standard deviation (SD). The statistically difference analysis was
and resuspended to culture dish with RPMI-1640 complete medium evaluated by one-way ANOVA using Origin 2016 software.

Adv. Funct. Mater. 2020, 2001059 2001059 (11 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.afm-journal.de

Supporting Information [9] S. T. Raa, S. J. Oosterling, N. P. van der Kaaij, M. P. van den Tol,
R. H. Beelen, S. Meijer, C. H. van Eijck, J. R. van der Sijp,
Supporting Information is available from the Wiley Online Library or M. van Egmond, J. Jeekel, J. Surg. Oncol. 2005, 92, 124.
from the author. [10] J. L. Khatcheressian, A. C. Wolff, T. J. Smith, E. Grunfeld, H. B. Muss,
V. G. Vogel, F. Halberg, M. R. Somerfield, N. E. Davidson, J. Clin.
Oncol. 2006, 24, 5091.
[11] B. Fisher, N. Slack, P. Cavanaugh, B. Gardner, R. Ravdin, Ann. Surg.
Acknowledgements 1970, 172, 711.
The authors greatly appreciate the help of Dr. Shanling Wang from [12] Y. Shi, T. Lammers, Acc. Chem. Res. 2019, 52, 1543.
Analytical & Testing Center in Sichuan University for TEM analysis [13] J. Xu, L. Xu, C. Wang, R. Yang, Q. Zhuang, X. Han, Z. Dong, W. Zhu,
of the nanoparticles. This work was financially supported by the R. Peng, Z. Liu, ACS Nano 2017, 11, 4463.
National Natural Science Foundation of China (Nos. 31930067, [14] J. Peng, Q. Yang, Y. Xiao, K. Shi, Q. Liu, Y. Hao, F. Yang, R. Han,
31525009, 31800797, and 31771096), the National Key Research Z. Qian, Adv. Funct. Mater. 2019, 29, 1900004.
and Development Program of China (Nos. 2017YFC1103502 and [15] L. Luo, C. Zhu, H. Yin, M. Jiang, J. Zhang, B. Qin, Z. Luo, X. Yuan,
2016YFA0201402), the China Postdoctoral Science Foundation funded J. Yang, W. Li, Y. Du, J. You, ACS Nano 2018, 12, 7647.
project (No. 2018M631094), the Postdoctoral Innovation Talents [16] L. Guo, D. Yan, D. Yang, Y. Li, X. Wang, O. Zalewski, ACS Nano
Support Program (No. BX20180207), and 1·3·5 project for disciplines of 2014, 8, 5670.
excellence, West China Hospital, Sichuan University (ZYGD18002).
[17] Q. Chen, L. Xu, C. Liang, C. Wang, R. Peng, Z. Liu, Nat. Commun.
2016, 7, 13193.
[18] J. Shao, C. Ruan, H. Xie, Z. Li, H. Wang, P. K. Chu, X. F. Yu, Adv. Sci.
Conflict of Interest 2018, 5, 1700848.
[19] T. Yata, Y. Takahashi, M. Tan, H. Nakatsuji, S. Ohtsuki, T. Murakami,
The authors declare no conflict of interest. H. Imahori, Y. Umeki, T. Shiomi, Y. Takakura, M. Nishikawa,
Biomaterials 2017, 146, 136.
[20] A. Tabet, M. P. Jensen, C. C. Parkins, P. G. Patil, C. Watts,
O. A. Scherman, Adv. Healthcare Mater. 2019, 8, e1801391.
Keywords [21] L. L. Bu, J. Yan, Z. Wang, H. Ruan, Q. Chen, V. Gunadhi, R. B. Bell,
cancer recurrence, controlled drug release, immunotherapy, Z. Gu, Biomaterials 2019, 219, 119182.
photothermal, thermal-responsive hydrogels [22] Q. Chen, C. Wang, X. Zhang, G. Chen, Q. Hu, H. Li, J. Wang,
D. Wen, Y. Zhang, Y. Lu, G. Yang, C. Jiang, J. Wang, G. Dotti, Z. Gu,
Received: February 4, 2020 Nat. Nanotechnol. 2019, 14, 89.
Revised: March 25, 2020 [23] C. Song, H. Phuengkham, Y. S. Kim, V. V. Dinh, I. Lee, I. W. Shin,
Published online: H. S. Shin, S. M. Jin, S. H. Um, H. Lee, K. S. Hong, S. M. Jin, E. Lee,
T. H. Kang, Y. M. Park, Y. T. Lim, Nat. Commun. 2019, 10, 3745.
[24] Y. Qi, H. Min, A. Mujeeb, Y. Zhang, X. Han, X. Zhao, G. J. Anderson,
Y. Zhao, G. Nie, ACS Appl. Mater. Interfaces 2018, 10, 6972.
[1] F. Bray, J. Ferlay, I. Soerjomataram, R. L. Siegel, L. A. Torre, A. Jemal, [25] C. Bastiancich, P. Danhier, V. Preat, F. Danhier, J. Controlled Release
Ca-Cancer J. Clin. 2018, 68, 394. 2016, 243, 29.
[2] I. Soerjomataram, J. Lortet-Tieulent, D. M. Parkin, J. Ferlay, [26] M. Norouzi, B. Nazari, D. W. Miller, Drug Discovery Today 2016, 21,
C. Mathers, D. Forman, F. Bray, Lancet 2012, 380, 1840. 1835.
[3] S. Wang, X. Ma, X. Hong, Y. Cheng, Y. Tian, S. Zhao, W. Liu, Y. Tang, [27] X. Chen, M. Wang, X. Yang, Y. Wang, L. Yu, J. Sun, J. Ding,
R. Zhao, L. Song, Z. Teng, G. Lu, ACS Nano 2017, 12, 662. Theranostics 2019, 9, 6080.
[4] J. Dongen, A. Voogd, I. Fentiman, C. Legrand, R. Sylvester, D. Tong, [28] T. Wang, D. Wang, H. Yu, B. Feng, F. Zhou, H. Zhang, L. Zhou,
E. Schueren, P. Helle, K. Zijl, H. Bartelink, J. Natl. Cancer Inst. 2000, S. Jiao, Y. Li, Nat. Commun. 2018, 9, 1532.
92, 1143. [29] Q. Li, J. Wen, C. Liu, Y. Jia, Y. Wu, Y. Shan, Z. Qian, J. Liao, ACS
[5] R. Demicheli, M. W. Retsky, W. J. Hrushesky, M. Baum, I. D. Gukas, Biomater. Sci. Eng. 2019, 5, 768.
Ann. Oncol. 2008, 19, 1821. [30] Y. Qu, B. Y. Chu, J. R. Peng, J. F. Liao, T. T. Qi, K. Shi, X. N. Zhang,
[6] W. Ceelen, P. Pattyn, M. Mareel, Crit. Rev. Oncol. Hematol. 2014, 89, Y. Q. Wei, Z. Y. Qian, NPG Asia Mater. 2015, 7, e207.
16. [31] H. Wu, S. Liu, L. Xiao, X. Dong, Q. Lu, D. L. Kaplan, ACS Appl.
[7] O. Al-Sahaf, J. H. Wang, T. J. Browne, T. G. Cotter, H. P. Redmond, Mater. Interfaces 2016, 8, 17118.
Ann. Surg. 2010, 252, 1037. [32] K. Shi, Y. L. Wang, Y. Qu, J. F. Liao, B. Y. Chu, H. P. Zhang, F. Luo,
[8] A. Costa, Y. Kieffer, A. Scholer-Dahirel, F. Pelon, B. Bourachot, Z. Y. Qian, Sci. Rep. 2016, 6, 19077.
M. Cardon, P. Sirven, I. Magagna, L. Fuhrmann, C. Bernard, [33] J. Zhou, G. Wang, Y. Chen, H. Wang, Y. Hua, Z. Cai, J. Cell. Mol.
C. Bonneau, M. Kondratova, I. Kuperstein, A. Zinovyev, A. M. Givel, Med. 2019, 23, 4854.
M. C. Parrini, V. Soumelis, A. Vincent-Salomon, F. Mechta-Grigoriou, [34] T. Gong, L. Liu, W. Jiang, R. Zhou, Nat. Rev. Immunol. 2020, 20, 95.
Cancer Cell 2018, 33, 463. [35] J. Cerny, I. Striz, Clin. Sci. 2019, 133, 1549.

Adv. Funct. Mater. 2020, 2001059 2001059 (12 of 12) © 2020 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

You might also like