You are on page 1of 14

Research Article

Cite This: ACS Appl. Mater. Interfaces 2018, 10, 20342−20355 www.acsami.org

Photothermal-Activatable Fe3O4 Superparticle Nanodrug Carriers


with PD-L1 Immune Checkpoint Blockade for Anti-metastatic Cancer
Immunotherapy
Rui Ge,† Cangwei Liu,‡ Xue Zhang,† Wenjing Wang,† Binxi Li,† Jie Liu,‡ Yi Liu,† Hongchen Sun,‡
Daqi Zhang,*,§ Yuchuan Hou,*,∥ Hao Zhang,*,† and Bai Yang†

State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R.
China

Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun 130021, P. R. China
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

§
Department of Thyroid Surgery, China-Japan Union Hospital, Jilin University, Changchun 130033, P. R. China

Department of Urinary Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
*
S Supporting Information
Downloaded via SEOUL NATL UNIV on July 30, 2018 at 02:24:22 (UTC).

ABSTRACT: Checkpoint blockade immunotherapy has


shown great potential in clinical cancer therapy, but the
body’s systemic immune must be fully activated and generates
a positive tumor-specific immune cell response. In this work,
we demonstrate the design of the immune-adjuvant nanodrug
carriers on the basis of poly(ethylene glycol)-block-poly(lactic-
co-glycolic acid) copolymer-encapsulated Fe3O4 superparticles
(SPs), in which imiquimod (R837), a kind of Toll-like
receptor 7 agonist, is loaded. The nanodrug carriers are
defined as Fe3O4-R837 SPs. The multitasking Fe3O4-R837 SPs
can destroy the 4T1 breast tumor by photothermal therapy
(PTT) under near-infrared laser irradiation to generate the
tumor-associated antigens because of the high efficiency of tumor magnetic attraction ability and photothermal effect. The PTT
also triggers the release of R837 as the adjuvant to trigger a strong antitumor immune response. By further combining with the
checkpoint blockade adjusted by programmed death ligand 1 (PD-L1) antibody, the Fe3O4-R837 SP-involved PTT cannot only
eliminate the primary tumors but also prevent tumor metastasis to lungs/liver. Meanwhile, this synergistic therapy also shows
abscopal effects by completely inhibiting the growth of untreated distant tumors through effectively triggering the tumors
infiltrated by CD45+ leukocytes. Such findings suggest that Fe3O4-R837 SP-involved PTT can significantly potentiate the
systemic therapeutic efficiency of PD-L1 checkpoint blockade therapy by activating both innate and adaptive immune systems in
the body.
KEYWORDS: Fe3O4 superparticles, cancer immunotherapy, photothermal-activatable, checkpoint blockade therapy, anti-metastasis

■ INTRODUCTION
Cancer has become one of the major threats to human life for
apy,12−15 cytokine therapy,16,17 dendritic cell (DC)-based
immunotherapy,18,19 cancer vaccines and so forth,20−23 have
centuries.1 Because of its high metastasis and recurrence, the been established and achieved exciting results in clinical trials.
conventional treatments, such as surgery, chemotherapy and Despite cancer immunotherapy is far from being satisfactory, the
radiotherapy, are difficult to cure thoroughly.2,3 Accordingly, continuous improvements of this strategy will finally build an
massive efforts are being devoted to the development of new and alternative for cancer treatment.24,25
effective cancer treatments with low side effects, high Among the aforementioned cancer immunotherapies, check-
specificities, and anti-metastasis.4 Recently, along with the deep point-blockade therapy, which utilizes a series of antibodies that
understanding of cancer and their relationship with the immune can target to the overexpressed T-cell suppressor checkpoint
system, cancer immunotherapy by stimulating and training the signaling pathways, is a promising strategy to solve the adaptive
body’s immune system to actively attack tumor cells for the immune evasion in the tumor and has been approved by the U.S.
purpose of controlling metastatic tumor’s growth has attracted Food and Drug Administration (FDA) for cancer treatment.26,27
great attention and become one of the most promising
treatments for cancer.5−8 Several kinds of cancer immunothera- Received: April 11, 2018
pies, such as chimeric antigen receptor/T-cell receptor- Accepted: May 31, 2018
engineered T-cell therapy,9−11 checkpoint-blockade ther- Published: June 7, 2018

© 2018 American Chemical Society 20342 DOI: 10.1021/acsami.8b05876


ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

Figure 1. Schematic illustration of Fe3O4-R837 SP PTT with PD-L1 checkpoint blockade for cancer immunotherapy. With the help of tumor magnetic
attraction, Fe3O4-R837 SPs can destroy the tumors effectively under NIR irradiation. In the presence of Fe3O4-R837 SPs, the generated tumor-
associated antigens can induce strong antitumor immune responses. By further combining with PD-L1 checkpoint blockade, this method can eliminate
primary tumors, prevent the metastasis to lungs/liver, and further inhibit the distant tumor growth after PTT.

Adaptive immune evasion mechanism is a self-protecting delivery system has not been reported because of the difficulty in
behavior of tumor cells to defend against the body’s immune choosing proper NPs. In this scenario, Fe3O4 NPs have been
response.28 Programmed death 1 (PD-1) and its ligand, approved by FDA for the clinical treatment with the function of
programmed death ligand 1 (PD-L1), are two important magnetic targeting and T2-weighted magnetic resonance imaging
immune checkpoint molecules related to immune resistance.29 (MRI).49 However, individual Fe3O4 NPs show poor photo-
PD-1, expressed on T cells surface, interacts with PD-L1 thermal performance due to the low molar extinction coefficient,
expressed on tumor cells, terminating immune responses by making them useless in PTT. In our previous works, spherical
inhibiting cytotoxic/effector T-cell function and delivering anti- superparticles (SPs) of Fe3O4 NPs have been fabricated by oil-in-
apoptotic signals to tumors.30 The elimination of either one can water (O/W) microemulsion template method, which signifi-
lead to cytotoxic/effector T cells normal operation without cantly improves the photothermal performance and makes
inhibition.31 Hence, antibody-mediated PD-1/PD-L1 pathway Fe3O4 a good agent for PTT.50 Meanwhile, the SPs can also load
specific blockade can enhance the potent antitumor activity of drugs to perform controlled drug release, thus acting as
the body.32 However, the clinical trials show that if the body’s multifunctional nanodrug carriers for tumor theranostics.
immune system cannot be fully activated and produce large In this work, to utilize near-infrared (NIR)-triggered PTT to
numbers of immune cells, the durable responses generated by induce effective cancer checkpoint blockade therapy, Fe3O4-
checkpoint blockade in most kinds of tumor cells are still very based multifunctional nanodrug carriers are fabricated from three
low.33−35 Therefore, combining checkpoint blockade therapy FDA-approved agents, namely, poly(ethylene glycol)-block-
with other types of treatments that can effectively activate the poly(lactic-co-glycolic acid) copolymer (mPEG-PLGA), Fe3O4
systemic immune responses to enhance T-cell infiltration may NPs, and imiquimod (R837). The nanodrug carriers are defined
enhance the antitumor response rates and broaden the as Fe3O4-R837 SPs. Fe3O4 SPs act as the NIR photothermal,
application of immunotherapy in metastatic tumor.36,37 magnetic targeting, and T2-weighted contrast agents, and mPEG-
Photothermal therapy (PTT) is a new type of cancer PLGA is an amphiphilic copolymer for encapsulating the SPs.
treatment strategy developed in recent years and has been The loaded immune adjuvant R837, a kind of Toll-like receptor 7
proved to kill tumor cells by thermal ablation and produce the (TLR7) agonist, can promote dendritic cells (DCs), phagocytize
tumor-associated antigens from ablated tumor cell residues.38−40 tumor-associated antigens, and become mature, thus enhancing
The thermal ablation process can also activate the host immune the activation and proliferation of antigen-specific lymphocytes
system and trigger acute inflammation in the tumor area, thereby in draining lymph nodes (DLNs). The tumor retention rate of
increasing the presentation of tumor-associated antigens to T the SPs with external magnetic field is as high as 14.8%ID/g and
cells.41 A variety of inorganic nanoparticles (NPs), such as noble under NIR irradiation, the Fe3O4-R837 SPs can effectively
metals,42−45 metal chalcogenides,46,47 metal oxides, and so on,48 eliminate the primary tumors and release R837. After PTT, with
have been used for the delivery of PTT, showing obvious tumor the help of immune adjuvant R837, the released tumor-
ablation in primary animal experiments. However, the strategy associated antigens can stimulate strong systemic antitumor
for the combination of the PTT of inorganic NPs with immune response, which can be promoted by PD-L1 checkpoint
checkpoint blockade therapy by constructing a novel drug blockade therapy to prevent the lungs/liver metastasis in a 4T1
20343 DOI: 10.1021/acsami.8b05876
ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

Figure 2. Characterization of Fe3O4 and Fe3O4-R837 SPs. (a) Schematic illustration of the synthesis of Fe3O4-R837 SPs. TEM (b) and SEM (c) images
of Fe3O4-R837 SPs. Inset in (b): high-magnification TEM image of one SP. The scale bar represents 50 nm. (d) T2 relaxation rates and concentration-
dependent T2-weighted MRI at pH 7.4 under 1.5 T magnetic field (inset) for the aqueous solution of Fe3O4-R837 SPs. (e) Temperature variation of
PBS, 150 μg/mL Fe3O4 SPs, and Fe3O4-R837 SPs irradiated by a 1.0 W/cm2 808 nm laser. (f) UV−vis absorption spectra of Fe3O4 and Fe3O4-R837 SPs
at the concentration of 150 μg/mL. (g) Fourier-transform infrared (FTIR) spectra of Fe3O4 NPs, mPEG-PLGA, Fe3O4 SPs, R837, and Fe3O4-R837 SPs.

metastatic triple-negative breast cancer murine model. Besides, prepared (Figure S1a).54 Then, O/W microemulsion is
the synergistic therapy has an abscopal effect to attack and inhibit generated by mixing mPEG-PLGA chloroform solution, Fe3O4
the nonirradiated distant tumors remaining in the mouse body. NPs, R837 in dimethyl sulfoxide (DMSO), and water under
In vitro and in vivo experiments indicate that immunogenic vigorous stirring. The evaporation of chloroform generates
Fe3O4-R837 SP PTT in combination with PD-L1 blockade Fe3O4-R837 SPs. The Fe3O4 SPs are synthesized with the same
therapy has significant efficacy in PTT-activated cancer method but without R837. The obtained Fe3O4-R837 SPs are
immunotherapy (Figure 1). well dispersed in water and appear as quasispheres with good


monodispersity under transmission electron microscopy (TEM)
RESULTS AND DISCUSSION (Figure 2b). The size distribution of Fe3O4-R837 SPs is
calculated from TEM observation, which shows the mean
Preparation and Characterization of Fe3O4-R837 SPs. diameter of 149.8 nm (Figure S1b). The average number of
Fe3O4-R837 SPs are synthesized by assembling Fe3O4 NPs in O/ Fe3O4 NPs in a superparticle is 1.27 × 104 (Calculation S1). The
W microemulsion templates with the presence of mPEG-PLGA scanning electron microscopy (SEM) observation also shows the
amphiphilic block copolymer and loading with R837 (Figure homogeneous size distribution of the SPs (Figure 2c). Fe3O4-
2a).51−53 According to the conventional thermal decomposition R837 SPs exhibit an average hydrodynamic diameter of 157.3 ±
method, 5.8 nm oleic acid (OA)-stabilized Fe3O4 NPs are first 3.0 nm, a ζ potential of −26.9 ± 2.8 mV, and a polydispersity
20344 DOI: 10.1021/acsami.8b05876
ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

Figure 3. In vitro transwell coculture system experiments. (a) Schematic illustration of the design of the transwell coculture system experiments. DCs are
placed in the lower compartment, and 4T1 cells are cultured in the upper compartment. (b−h) The percentage of mature DCs (CD11c+CD80+CD86+)
is analyzed by flow cytometry after different treatments. (i) Quantification of the level of DC maturation after different treatments in the transwell system
experiments. “−” and “+” in the figure means treatment without or with irradiation, respectively. Error bars are based on standard deviations of three
parallel samples.

index (PDI) of 0.05 ± 0.016, as measured by dynamic light 55.4 °C under the irradiation of 1.0 W/cm2 808 nm laser (Figure
scattering (DLS) (Figure S1c). The structural stability of Fe3O4- 2e). The photothermal conversion efficiency of Fe3O4-R837 SPs
R837 SPs is favorable in a physiological environment, as proved is 68.2% (Figure S2 and Calculation S2). Both the UV−vis
by stable size and PDI when dispersed in phosphate-buffered absorption and Fourier-transform infrared (FTIR) spectra of
saline (PBS, pH 7.4) for up to 7 days (Figure S1d). According to Fe3O4-R837 SPs show the characteristic absorption peak of
the previous reports,55 such SPs with negative surface charge, R837, indicating the effective encapsulation of R837 in the SPs
high stability, and hydrophilic “stealth” polymer surface possess (Figure 2f,g). In water, the R837 concentration and the
prolonged blood circulation and efficiently accumulate into absorbance in 2.5−10 μg/mL maintain a linear relation (Figure
tumor tissues by enhanced permeability and retention effect, S3a). The regression equation is y = 0.0698x − 0.03415 (R2 =
potentially applicable for the living body. 0.999) (Figure S3b). Thus, the R837 loading is calculated by
Recent studies have shown that the assembly of inorganic NPs UV−vis absorption spectrum, and the relationship between
into SPs can significantly enhance MRI and the photothermal encapsulation/loading efficiency and the dosage of R837
performance.50 The self-assembly of NPs into Fe3O4-R837 SPs solution in the reaction system are shown in Figure S3c,d.
does not change the superparamagnetic behavior of Fe3O4 NPs Considering the encapsulation efficiency and safe dose in vivo
in comparison with the magnetic curve of Fe3O4 NPs (Figure (0.4 mg/kg), the optimal feeding concentration of R837 is 200
S1e). The r2 of Fe3O4-R837 SPs is 178.1 mM−1 s−1 (Figure 2d), μg/mL.57 In our system, R837 is loaded in mPEG-PLGA. Upon
which is higher than that of commercial MRI T2-weighted NIR laser irradiation, the system temperature increases due to
contrast agents (Feridex, 93 mM−1 s−1; Resovist, 143 mM−1 the photothermal effect of Fe3O4-R837 SPs. According to the
s−1).56 The high r2 allows Fe3O4-R837 SPs to act as an effective previous literature,58 the temperature increment promotes the
T2-weighted contrast, with fewer doses getting darker contrast degradation of PLGA-based NPs. So, NIR irradiation leads to
(Figure 2d inset). Compared with the final temperature of PBS rapid R837 release from Fe3O4-R837 SPs. Figure S4a shows the
solution (32.9 °C), the temperature of SP solution increases to release of R837 with and without irradiation. Without irradiation,
20345 DOI: 10.1021/acsami.8b05876
ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

Figure 4. In vivo tumor magnetic attraction experiments. (a) Schematic illustration of the design of in vivo tumor magnetic attraction. (b) A photograph
showing how the magnet is applied during magnetic attraction. (c) Blood circulation kinetics of Fe3O4-R837 SPs without magnetic attraction. (d) Blood
circulation kinetics of Fe3O4-R837 SPs with magnetic attraction. (e) Biodistribution of Fe3O4-R837 SPs in 4T1-bearing mice at 24 h p.i. administration
with or without magnetic attraction. Error bars are based on standard deviations of three parallel samples. (f) T2-weighted MRI images of mice for
control, Fe3O4-R837 SPs and Fe3O4-R837 SPs with magnetic attraction. The red dotted lined circle points to tumors.

the released R837 in water keeps a low concentration. Whereas, a necrosis more efficiently when the cells are irradiated under the
high release rate is achieved and maintained under 1 W/cm2 808 808 nm laser.
nm laser. The released percentage is nearly 34% in 28 min, The 24 h cytotoxicity of Fe3O4 SPs and Fe3O4-R837 SPs is
implying that R837 can be constantly released from Fe3O4-R837 investigated by standard methyl thiazolyltetrozolium (MTT)
SPs (Figure S4b). With higher laser power density, Fe3O4-R837 assay. Even when incubated with 400 μg/mL Fe3O4 SPs, the 4T1
SPs release more R837 (Figure S4c). cells’ viability is still maintained at a high level (>80%), (Figure
PTT-Induced Apoptosis and/or Necrosis in Vitro. 4T1 S6a). Although R837 can induce apoptosis in cancer cells, the cell
metastatic triple-negative breast cancer cells are used to test the viability is still higher (>70%). Meanwhile, the cytotoxicity of
photothermal therapy of Fe3O4-R837 SPs. Figure S5a shows that Fe3O4-R837 SPs incubated with 293 cells (human embryo
Fe3O4 SPs/Fe3O4-R837 SPs are rapidly engulfed by 4T1 cells, kidney cells) shows that the SPs also have low cytotoxicity in
with most uptake occurring within 4 h and then remaining stable normal cells. The Fe3O4-R837 SPs can effectively induce the
within 24 h. High cellular uptake and low efflux ensure the high photothermal ablation of 4T1 cells in vitro (Figure S6b). 4T1
cellular accumulation of SPs (Figure S5b). In the control group, cells are cultured with Fe3O4-R837 SPs and then irradiated with
the structure of 4T1 cells is intact and smooth. With the an 808 nm laser at different power densities for different times.
increment of incubation time, even though the cells maintain The cell viability is evaluated by MTT assay. The cell viability
structural integrity, more and more SPs are adhered and decreases with the increment of laser power and irradiation time.
internalized by the cells (Figure S5c). The high cellular When the laser power density reaches 2 W/cm2 and the
accumulation of SPs is beneficial to induce cells apoptosis/ irradiation time is 20 min, less than 5% of the 4T1 cells survive.
20346 DOI: 10.1021/acsami.8b05876
ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

Figure 5. In vivo Fe3O4-R837 SP PTT triggers DC maturation. (a) Schematic illustration showing the design of experiments to examine the immune
responses induced by Fe3O4-R837 SP PTT. (b) IR thermal images of 4T1 tumor-bearing mice performed by an IR camera. The tumors are irradiated by
a 0.33 W/cm2 808 nm laser for 5 min. “−” and “+” in the figure mean treatment without or with irradiation, respectively. (c) Quantification of the level of
DC maturation induced by control, Fe3O4 SPs, and Fe3O4-R837 SP group with or without irradiation on 4T1-bearing mice. (d−i) The percentage of DC
maturation after different treatments. After staining with CD11c, CD80, and CD86, cells in the tumor-draining lymph nodes are assessed by flow
cytometry. Error bars are based on standard deviations of three parallel samples.

Then, 4T1 cells are stained with fluorescein diacetate (FD) and SPs in vitro. To investigate the contribution of Fe3O4-R837 SPs
propidium iodide (PI), as the living cells can be stained into during the apoptosis/necrosis process, the apoptosis process on
green and apoptotic cells into red.50 Figure S6c shows the PBS, R837, Fe3O4 SPs, and Fe3O4-R837 SP group is monitored
confocal fluorescence and bright microscope images of 4T1 cells by flow cytometric analysis. As shown in Figure S7a−h, the cells
incubated with Fe3O4-R837 SPs after irradiation for 0, 5, 10, and in upper left, upper right, bottom left, and bottom right areas
20 min. The decrease in green and increase in red along with the represent necrosis, late apoptosis, viability, and early apoptosis,
irradiation time indicate the effective photothermal therapy of respectively. In comparison with the control group, R837 cannot
20347 DOI: 10.1021/acsami.8b05876
ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

cause cell necrosis distinctly but can lead to partial cell apoptosis extend the blood circulation half-life (from 1.94 ± 0.16 to 2.48 ±
(Figure S7b,f). It is consistent with the results of the MTT assay 0.23 h), the tumor retention rate of the magnetic attraction group
that the cell viability of Fe3O4-R837 SPs is a little lower than that increases almost 68% (from 8.8 to 14.8 of the injected dose per
of Fe3O4 SPs. Figure S7c,d,g,h shows that both Fe3O4 SPs and gram tissue (%ID/g)) at 24 h postinjection (p.i.) administration
Fe3O4-R837 SPs have low toxicity and can induce the early (Figure 4e). Compared with control and Fe3O4-R837 SP group,
apoptosis of 4T1 cells effectively after PTT (1 W/cm2, 10 min). T2-weighted MRI reveals a darkening effect with the magnet
The apoptosis ratios are as high as 68.7 and 71.6%, respectively. attached, also suggesting the effective accumulation of SPs in the
Fe3O4-R837 SP-Induced DC Maturation in Vitro. As one tumor. Such high tumor retention caused by magnetic attraction
of the most crucial antigen presenting cells (APCs), DCs provide can make a contribution to improve PTT efficiency.
a pivotal functional link between innate and adaptive immune On the basis of in vitro experiments, the 4T1 tumor cell
responses.18 The immature DCs can engulf and process antigens residues post NIR-induced PTT with Fe3O4-R837 SPs can
in the tissue and peripheral blood, then undergo maturation and enhance the level of DC maturation, higher than individual
migrate to lymphoid nodes where they present the antigens to Fe3O4-R837 SPs, or cell residues ablated by Fe3O4 SPs without
̈ T cells. The mature DCs can upregulate the costimulatory
naive R837. The results suggest that R837-loaded SPs serve as the
molecules such as CD80 and CD86 on the APC surface to adjuvant to induce the immune response from the tumor-
̈ CD8+ cytotoxic T cells and naive
activate both naive ̈ CD4+ helper associated antigen in the cell residues. In further in vivo
18
T cells. Hence, the immunological effects of Fe3O4-R837 SPs experiments (Figure 5a), when the 4T1 tumors grown on Balb/c
toward bone-marrow-derived DCs by analyzing the upregulation mice reach ∼200 mm3 on day 10, the mice are iv injected with
of CD80 and CD86 are first investigated. A transwell system is PBS, Fe3O4 SPs, or Fe3O4-R837 SPs and attached to a magnet.
introduced to investigate the effect at the in vitro level. The upper After 24 h on day 11, the tumors are exposed to a 0.33 W/cm2
compartment contains 4T1 cells after different treatments, 808 nm NIR laser for 30 min. The infrared (IR) thermal images
whereas the lower compartment is placed with bone-marrow- show that the temperature increment caused by Fe3O4-R837 SP
derived DCs from Balb/c mice (Figure 3a). It is found that in the PTT is sufficient to ablate the cells and prevent their malignant
absence of R837, compared with the 4T1 cells cocultured with proliferation (Figure 5b). To investigate the levels of DC
Fe3O4 SPs without irradiation (Figure 3e), the released tumor- maturation by flow analysis, the draining lymph nodes of mice are
associated antigens after PTT can notably enhance the DC removed 3 days after PTT. Compared with the PBS without
maturation from 26.3 to 39.2% (Figure 3g) and with the help of irradiation group, the percentage of matured DCs increases from
R837, Fe3O4-R837 SPs with irradiation (89.6%, Figure 3h) 6.58 to 31.7% after PTT with Fe3O4-R837 SPs (Figure 5d,i). For
further promote in vitro DC maturation even higher than free the group of Fe3O4 SPs with PTT or Fe3O4-R837 SPs without
R837 at the same dose (75.7%, Figure 3d). Note that although PTT, the percentages of DC maturation only increase to 17.6
free R837 works well for DC maturation in vitro, the low and 22.8%, respectively (Figure 5h,f). These mean that after
solubility in water greatly limits the applications for a living body. Fe3O4-R837 SP PTT treatment for tumors, more DCs are
Therefore, it must be loaded into amphiphilic drug carriers like recruited to the tumor site as APCs to engulf the tumor-
Fe3O4 SPs. Meanwhile, the DC-secreted immune related associated antigens released by apoptotic tumor cells, then
cytokines, such as tumor necrosis factor α (TNF-α) and transported to the nearby lymph nodes and matured with the
interleukin 6 (IL-6), which are also indicators of DC maturation, help of released R837 (Figure 5c).
are measured by enzyme-linked immunosorbent assay (ELISA) The secretion of cytokine is another marker of immune
kits.18 The secretion levels of TNF-α and IL-6 from DCs are activation and indicates acute inflammation, a crucial mechanism
distinctly enhanced after Fe3O4-R837 SP PTT and higher than to evoke antitumor immunity.21 Hence, the changes of various
that in the treatment with free R837 (Figure S8). Both the flow cytokines, including IL-6 (a kind of marker in the activation of
cytometry and ELISA data prove that the tumor-associated humoral immunity), TNF-α (a kind of marker in the activation of
antigens released by tumor cells after PTT, especially with the cellular immunity), and interferon γ (IFN-γ) (a kind of marker in
help of R837-loaded SPs adjuvant, can induce DC maturation the activation of innate immunity) in sera after different
effectively. treatments are analyzed by ELISA (Figure S9).21 Similarly,
In Vivo Magnetic Attraction-Enhanced Photothermal even though the individual Fe3O4-R837 SPs or PTT with Fe3O4
Tumor Ablation for Immune System Activation. The above SPs can promote the secretion of cytokine, the increment of
results indicate that Fe3O4-R837 SPs are an effective immune Fe3O4-R837 SPs PTT group is the highest, which is most
stimulating reagent in vitro. Furthermore, the PTT of Fe3O4- favorable for inducing antitumor immune responses. Such results
R837 SPs is considered to induce enhanced tumor-specific suggest that with the help of R837-loaded SPs, the tumor-
immune responses in vivo. Before this, because of the associate antigens released from tumor ablation therapy can
superparamagnetism of Fe3O4-R837 SPs, we test if adding an induce the systemic innate and adaptive immune systems
external magnetic field can effectively enhance tumor accumu- effectively in vivo.
lation of SPs, thereby increasing the treatment efficiency of PTT. In Vivo Fe3O4-R837 SP PTT with PD-L1 Blockade To
The blood circulation kinetics and biodistribution studies are Eradicate Primary Tumor and Prevent Lungs/Liver
performed on orthotopic 4T1 tumor-bearing Balb/c mice. When Metastasis. The main reason of a high cancer fatality rate is
the 4T1 tumors grown on Balb/c mice reach ∼200 mm3 on day that it is prone to metastasis, which is difficult to treat with
10, Fe3O4-R837 SPs are intravenously (iv) injected into the mice. conventional treatments once they have metastasized.2,3 Hence,
The mice are divided into two groups. The tumors of one group we investigate if PTT-activatable immunotherapy with Fe3O4-
are attached to a magnet for 24 h (Figure 4a,b). Blood samples R837 SPs can make a contribution to treat the metastatic tumor.
are collected at different time intervals and decomposed in aqua As approved by FDA for clinically used immunotherapy, PD-L1
regia solution to determine the Fe3+ concentration by inductively plays a pivotal role for tumor cells to evade the host’s immune
coupled plasma optical emission (ICP-OES). As shown in Figure system and can be blockaded by antibodies (anti-PD-L1).31
4c,d, although adding the magnetic field does not significantly Therefore, in the animal experiments, PD-L1 blockade therapy is
20348 DOI: 10.1021/acsami.8b05876
ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

Figure 6. In vivo Fe3O4-R837 SP PTT with PD-L1 blockade eliminates primary tumors and prevents lungs/liver metastasis. (a) Schematic illustration
showing the design of experiments. (b−e, j−m) Photographs showing the tumor nodules in the lungs/livers. (f−i, n−q) Representative lungs/liver
sections stained with H&E. The scale bar is 200 μm.

introduced to enhance the antitumor immunotherapy efficacies tumor nodules in lungs and large area of edge necrosis in liver.
generated by PTT with Fe3O4-R837 SPs of the primary tumor. Anti-PD-L1 blockade therapy alone shows little effect on
Figure 6a shows the experimental design. 4T1 cells are preventing lungs/liver metastasis (Figure 6c,g,k,o). Although
orthotopically injected into the right mammary fat pads, and the primary tumor elimination by PTT with Fe3O4-R837 SPs can
they can spontaneously metastasize to lungs or liver.59,60 When reduce the possibility of tumor metastasis and does not cause a
the volume of tumor reaches ∼200 mm3, the mice are i.v. injected local tissue necrosis, there are still some tumor nodules in lungs
with Fe3O4-R837 SPs and attached to a magnet every 3 days and obvious metastasis around the blood vessels in liver (Figure
three times (on day 10, 13, and 16). Twenty four hours p.i., 6d,h,l,p). Only by combining the two treatments can the tumor
tumors are exposed to a 808 nm NIR laser at 0.33 W/cm2 for 30
metastasis be effectively eliminated (Figure 6e,i,m,q). Such
min. After laser irradiation, anti-PD-L1 antibody are i.v. injected
results are consistent with the previous literature in that the
into the mice at a dose of 75 μg/mouse. As shown in Figure S10,
compared with the control group, PTT with Fe3O4-R837 SPs can individual anti-PD-L1 blockade therapy has almost no effect on
eliminate the primary tumor and PD-L1 blockade therapy alone 4T1 tumor cells and only once the immune system is activated by
fails to delay the tumor proliferation. On day 24, tumor-bearing other immunogenic therapies, the efficiency of blockade therapy
mice are all sacrificed to evaluate the extent of metastasis by can be greatly enhanced.35 Therefore, we demonstrate that the
checking the tumor nodules in the lungs and liver. The tumor-specific immunity activated by PTT with immune
dehydrated tissues are imaged by digital camera and further adjuvants can enhance the effect of checkpoint blockade therapy,
sectioned and stained with hematoxylin−eosin (H&E) staining. leading to the elimination of primary tumor and the prevention
For the control group, Figure 6b,f,j,n shows that there are many of lungs/liver metastasis.
20349 DOI: 10.1021/acsami.8b05876
ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

Figure 7. In vivo Fe3O4-R837 SP PTT with PD-L1 blockade eliminates primary 4T1 tumors and inhibits the growth of distant tumors. (a) Schematic
illustration showing the experiment’s design. 4T1 tumors are planted on both sides of the mice. The right tumors are designed as “primary tumors” for
PTT, and the left tumors are designed as “distant tumors” without PTT. (b, c) The primary and distant tumor volume growing trend for different groups.
(d, e) Tumor weights at the end point of different groups. Arrows represent the time of materials injection (black) and irradiation (red). “−” and “+”
mean treatment without or with irradiation, respectively. Error bars are based on standard deviations of five parallel samples.

Fe3O4-R837 SP PTT Plus PD-L1 Blockade To Inhibit the anti-PD-L1 antibody are i.v. injected into the mice at a dose of 75
Growth of Distant Tumors. In practical therapies, the μg/mouse three times on day 11, 14, and 17. The sizes of the
metastatic tumors may have already existed so the studies on primary and distant tumors in different groups are measured by a
the inhibition of metastatic tumor growth is necessary. To digital caliper every other day. Figure 7b,c shows that anti-PD-L1
facilitate the demonstration and further study that synergistic alone contributes minimally to the inhibition of both the primary
therapy can induce immune cells to specifically kill 4T1 tumor and the distant tumors. Fe3O4 SPs/Fe3O4-R837 SPs injection
cells and thus prevent metastasis, the larger pre-existing alone shows almost no effect on the primary or distant tumor
simulated distant tumors are constructed for the analysis of growth. PTT with Fe3O4 SPs/Fe3O4-R837 SPs in the absence of
immune cell content. anti-PD-L1 three times can eradicate the primary tumor
A bilateral orthotopic 4T1 tumor model is introduced into the effectively but does not inhibit the growth of the distant tumor
experiments to prove that the synergistic therapy cannot only significantly. The distant tumor growth of Fe3O4-R837 SP PTT
prevent the tumor metastasis but also inhibit the pre-existing group is only partially delayed because the immune system can be
metastatic tumors (Figure 7a). The first tumor is inoculated into successfully activated, as mentioned above. Only the combina-
the right mammary fat pads as the primary tumor, and the second tion of Fe3O4-R837 SP PTT and anti-PD-L1 treatment can
tumor is inoculated into the left mammary fat pads as an artificial completely eradicate the primary tumor and control the growth
mimic of distant metastatic tumor. After the SPs are systemically of the distant tumor. In addition, the average weight of mice in all
injected, the primary tumors are eliminated by PTT or not. Then, groups are monitored. As indicated in Figure S11, no significant
20350 DOI: 10.1021/acsami.8b05876
ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

Figure 8. Mechanism study of inhibiting the metastasis. (a) Schematic illustration showing the design of experiments. The distant tumors are collected
for flow analysis; the percentages of CD45+ leukocytes (CD45+PI−) (b), NK cells (CD45+CD3ε−NKp46+PI−) (c), B cells (CD45+CD3ε−B220+PI−)
(d), CD8+ T cells (CD45+CD3ε+CD8+PI−) (e), and CD4+ T cells (CD45+CD3ε+CD4+PI−) (f) in the tumors are measured. “−” and “+” in the figure
mean treatment without or with irradiation, respectively. Error bars are based on standard deviations of five parallel samples.

weight loss is observed in either the PTT treatment, anti-PD-L1 To investigate the mechanism of synergistic antitumor
alone, or the synergistic therapy of two methods, indicating the immune responses triggered by Fe3O4-R837 SP PTT plus anti-
safety of the therapies. Meanwhile, considering the full activation PD-L1 therapy, the distant 4T1 tumors are excised to test the
of the immune system may be harmful to the organs, serum immune cells on day 20 (Figure 8a). Immune cells, which can
biochemistry assay is performed in 4T1 tumor-bearing mice on also be called leukocytes, include all cells that are involved in or
day 24 after the synergistic therapy and the healthy mice (Table associated with the immune response.27 Since CD45+ molecules
S1). All parameters of the treatment group are consistent with are expressed on all leukocytes, known as leukocyte common
those of the healthy one, indicating that such a full activation of antigen, we measure the percentage of the infiltrating leukocytes
the immune system by synergistic therapy can be tolerated by the in the distant tumors. Compared to the control group (14.94 ±
mice. 1.84%), the percentage of CD45+ leukocytes increase almost 34
Mechanism Study of Synergistic Therapy To Inhibit and 46% in anti-PD-L1 therapy alone (20.03 ± 1.29%) and
the Metastasis. Before studying the mechanism of synergistic Fe3O4-R837 SP PTT alone (21.84 ± 2.18%) (Figure 8b). For the
therapy, it must be ensured that the systemic antitumor immune subspecies of leukocytes, such as NK cells, compared with the
response has been activated in the body, which makes sense for control group (6.52 ± 0.58%), the increase in anti-PD-L1 group
the immune cell test. Therefore, we evaluate the change of is about 19% (7.80 ± 0.63%) and in Fe3O4-R837 SP PTT group,
proinflammatory cytokines, including IL-6, TNF-α, and IFN-γ, about 47% (9.62 ± 0.98%) (Figure 8c). Similarly, for B cells,
in sera by ELISA kits within 7 days after a group of treatment compared with the control group (3.81 ± 0.44%), the increase in
(Figure S12). Although anti-PD-L1 alone or Fe3O4-R837 SP anti-PD-L1 group is about 14% (4.36 ± 0.25%) and in Fe3O4-
PTT alone can increase the secretion of proinflammatory R837 SP PTT group, about 58% (6.04 ± 0.15%) (Figure 8d).
cytokines, the secretions induced by the synergistic therapy are Such results indicate that compared with anti-PD-L1 alone, the
the highest, favorable for triggering antitumor immune response. percentage of NK cells and B cells is more affected by Fe3O4-
However, on day 7, after PTT treatment, all cytokine levels drop R837 SP PTT group. However, for CD8+ T cells, compared with
back to the baseline levels, indicating that the inflammation the control group (0.28 ± 0.06%), the increase in anti-PD-L1
induced by different kinds of therapy is merely an acute response. group is about 239% (0.95 ± 0.06%) and in Fe3O4-R837 SP PTT
20351 DOI: 10.1021/acsami.8b05876
ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

group, about 200% (0.84 ± 0.09%) (Figure 8e). For CD4+ T and kept at 80 °C for 20 min under vacuum and vigorous stirring.
cells, compared with the control group (0.77 ± 0.18%), the Afterward, the mixture was raised to 200 °C with the rate of 20 °C/min,
increase in anti-PD-L1 group is about 179% (2.15 ± 0.26%) and kept for 30 min at this temperature under nitrogen atmosphere, and
in Fe3O4-R837 SP PTT group, about 156% (1.97 ± 0.18%) then refluxed under 265 °C for another 30 min. Then, the reaction
solution was cooled to room temperature naturally. With the help of a
(Figure 8f). Such results suggest that anti-PD-L1 may have more magnet, Fe3O4 NPs were extracted and washed by adding hexane and
influence on promoting the accumulation of CD8+ and CD4+ T ethanol three times and finally dissolved in chloroform.
cells in the distant tumor sites. Although the previous data Preparation of mPEG-PLGA-Capped Fe3O4 SPs. mPEG-PLGA-
demonstrate that Fe3O4-R837 SP PTT can activate the systemic capped Fe3O4 SPs were synthesized by an emulsification method.51−53
immune response, the increments of CD8+ and CD4+ T cells are OA-stabilized Fe3O4 NPs and mPEG-PLGA were dissolved in
not significant. The reason is that Fe3O4-R837 SP PTT activates chloroform with the concentration of 20 and 200 mg/mL, respectively.
large numbers of negatively regulatory T cells while activating Under a nitrogen atmosphere and mechanical stirring at room
systemic immunity, resulting in a small increase in the total temperature, 0.5 mL of OA-stabilized Fe3O4 NPs, 50 μL of mPEG-
number of T cells. Therefore, we suppose only through the PLGA, and 0.5 mL of DMSO were injected dropwise into 10 mL of
deionized water. After the ultrasonic treatment, the emulsion was further
synergistic therapy, the activation of systemic immunity while
stirred to evaporate organic solvent for 4 h. The SPs were washed by
inhibiting negatively regulatory T cells, the treatment efficiency centrifugation (6000 rpm, 10 min) three times and dialyzed (molecular
can be maximized. This consideration is further proved by the weight cut-off (MWCO): 7 kDa) against deionized water for 24 h, which
experimental data. Compared to that in the control group (14.94 were defined as Fe3O4 SPs.
± 1.84%), the total percentage of CD45+ leukocytes increases Preparation of mPEG-PLGA-Capped Fe3O4-R837 SPs. The
almost 70% in the synergistic therapy group (25.47 ± 2.09%). synthesis of mPEG-PLGA-capped Fe3O4-R837 SPs was similar to that of
Similarly, the percentage of NK cells (11.24 ± 0.70%), B cells mPEG-PLGA-capped Fe3O4 SPs. R837 was dissolved in DMSO with
(6.54 ± 0.19%), CD8+ T cells (1.39 ± 0.07%), and CD4+ T cells the concentration of 4 mg/mL. Under a nitrogen atmosphere and
(2.76 ± 0.30%) significantly increase in comparison with not mechanical stirring at room temperature, 0.5 mL of OA-stabilized Fe3O4
only the control group but also the anti-PD-L1 alone or Fe3O4- NPs, 50 μL of mPEG-PLGA, and 0.5 mL of R837 were injected
dropwise into 10 mL of deionized water. After the ultrasonic treatment,
R837 SP PTT alone.


the emulsion was further stirred to evaporate the organic solvent for 4 h.
The products were washed by centrifugation (6000 rpm, 10 min) three
CONCLUSIONS times and dialyzed (MWCO: 7 kDa) against deionized water for 24 h,
In summary, immunogenic Fe3O4-R837 SPs have been designed which were defined as Fe3O4-R837 SPs.
for the effective treatment of metastatic cancer by combining R837 Loading Standard Curve and Releasing Experiments.
PTT and PD-L1 checkpoint blockade therapy. With the help of R837 solution was prepared in DMSO with the concentration of 1 mg/
mL and then diluted into 2.5, 5, 7.5, and 10 μg/mL standard solution by
an external magnetic field, Fe3O4-R837 SPs can directly destroy water. The absorbance of R837 solution with different concentrations at
the tumors upon NIR irradiation, activating the immune system 322 nm was measured by UV−vis absorption spectra. According to the
by inducing DCs’ maturation and secretion of cytokines. Further absorbance, a standard curve was obtained. The encapsulation efficiency
in combination with the PD-L1 checkpoint blockade therapy, and loading efficiency of Fe3O4-R837 SPs were calculated from the UV−
Fe3O4-R837 SPs cannot only eradicate the primary tumors vis absorption spectra against the standard curve. For R837 release, the
directly exposed to PTT but also prevent the lungs/liver SPs were irradiated under NIR laser and centrifuged to get the
metastasis and inhibit the pre-existing distant tumors left after supernatant. The released concentration and percentage can also be
PTT. Our studies show the great potential of integrating Fe3O4- calculated from the absorbance at 322 nm according to the standard
based PTT with checkpoint blockade therapy to realize a curve.
Cellular Uptake and Efflux. 4T1 cells seeded in six-well plates (2 ×
remarkable synergistic therapeutic outcome and broaden the
105 cells/well) were incubated with Fe3O4 SPs/Fe3O4-R837 SPs at a
application of cancer immunotherapy in vivo.


concentration of 150 μg/mL for 2, 4, 12, and 24 h. Then, cells were
collected and washed three times with PBS, counted with a
EXPERIMENTAL SECTION hemocytometer, and lysed with aqua regia (HCl/HNO3 = 3:1) to test
Materials. Iron acetylacetonate (Fe(acac)3, 99.9+%), benzyl ether the Fe3+ concentration with ICP-OES. The uptake level was expressed as
(99%), oleic acid (OA, 90%), oleyamine (OLA, 70%), 1,2- the amount of Fe3O4 SPs (nmol) per 104 cells.
hexadecanediol (90%), Imiquimod (R837), and fluorescein diacetate To investigate the efflux of SPs, 4T1 cells were incubated with Fe3O4
(FD) were purchased from Sigma-Aldrich. Poly(ethylene glycol)-block- SPs/Fe3O4-R837 SPs at a concentration of 150 μg/mL for 4 h. Then, the
poly(lactic-co-glycolic acid) copolymer (mPEG-PLGA, 50:50 w/w, Mw culture medium was discarded and the 4T1 cells were washed with PBS
∼ 2000:5000 Da) was purchased from Daigang Biomaterial Co., Ltd. three times. Two milliliters of fresh culture medium was added to each
Propidium iodide (PI) was purchased from Invitrogen. Dulbecco’s well, and cells were further cultured for 2, 4, 12, and 24 h. Thereafter, the
modified Eagle’s medium with fetal bovine serum and high glucose was cells were collected and washed three times with PBS, counted with a
purchased from Gibco. Chloroform and dimethyl sulfoxide (DMSO) hemocytometer, and lysed with aqua regia to test the Fe3+ concentration
were of analytical grade. Deionized water and absolute ethanol were with ICP-OES. Results were expressed as the percent of the difference
used as received. Antiprogrammed death ligand 1 used in vivo was value of SPs being retained to the 4 h cellular uptake amount.
obtained from BioXCell (α-PD-L1, clone: 10F.9G2, catalog no. The bright field images of internalization were also observed under a
BE0101). Anti-CD11c (N418), anti-CD80 (16-10A1), anti-CD86 fluorescence microscope. Fe3O4-R837 SPs were incubated with 4T1
(GL-1), anti-CD16/32 (93), anti-CD45 (30-F11), anti-CD3ε (145- cells for 2, 4, 12, and 24 h and washed with PBS three times, fixed with
2C11), anti-CD4 (RM4-5), anti-CD8a (53-6.7), anti-NKp46 (29A1.4), 4% paraformaldehyde, and observed under the fluorescence microscope.
and anti-B220 (RA3-6B2) antibodies were purchased from Biolegend. Cytotoxicity Assay in Vitro. 4T1 cells were seeded in 96-well plates
Mouse interleukin 6 (IL-6), tumor necrosis factor α (TNF-α), and at a density of 4 × 103 cells per well. Different concentrations of Fe3O4
interferon γ (IFN-γ) ELISA Kit were purchased from Dakewe biotech. SPs/Fe3O4-R837 SPs were incubated with cells for 24 h. The cell
Preparation of OA-Stabilized Fe3O4 NPs. OA-stabilized Fe3O4 viability was detected by standard MTT assay according to the
NPs were prepared by thermal decomposition route following the manufacturer’s instructions. The experiment was repeated five times.
previous literature.54 Briefly, 2 mmol Fe(acac)3, 6 mmol OLA, 6 mmol Apoptosis Analysis in Vitro. 4T1 cells seeded in six-well plates (2
OA, 5 mmol 1,2-hexadecanediol, and 20 mL of benzyl ether were mixed. × 105 cells/well) were treated with Fe3O4-R837 SPs at a concentration
The mixture was then cycled between vacuum and nitrogen three times of 150 μg/mL for 4 h and then irradiated with 808 nm NIR laser at

20352 DOI: 10.1021/acsami.8b05876


ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

different power densities for different times. The cell viability was where L and D (mm) represented the tumor lengths of long and short
evaluated by MTT assay. For fluorescence imaging, the treated cells axes, respectively. At the end of the experiments, mice were sacrificed
were fixed with 4% paraformaldehyde and stained with 1 μg/mL PI and and tumors were excised and weighed. Lungs and livers were harvested,
FD to visualize living cells (green fluorescence) and cell apoptosis (red observed for the tumor nodules, or sectioned and stained with H&E.
fluorescence). For flow cytometric analysis, treated cells were harvested, Abscopal Effect on Bilateral 4T1 Model. BALB/c mice were
washed with ice-cold PBS three times, stained with Alexa Fluor 488- injected with 1 × 106 4T1 cells into the right mammary fat pads (primary
Annexin V and PI in the dark for 20 min at room temperature, and then tumor) and 2 × 105 4T1 cells into the left mammary fat pads (distant
analyzed by flow cytometry. tumor). When the primary tumor reached ∼200 mm3, mice were
In Vitro DC Stimulation Transwell Experiments. DCs were randomly divided into eight groups (n = 5): control, anti-PD-L1, Fe3O4
isolated from the bone marrow of ∼8 week-old BALB/c mice. The mode SPs, Fe3O4 SPs PTT, Fe3O4-R837 SPs, Fe3O4-R837 SPs PTT, Fe3O4-
of transwell is the cocultivation system, and the pore diameter is 0.4 μm. R837 SPs plus anti-PD-L1, and Fe3O4-R837 SPs PTT plus anti-PD-L1.
First, 1 × 105 4T1 cells were incubated with R837, Fe3O4 SPs, or Fe3O4- The mice were i.v. injected with the materials and attached to the
R837 SPs in the upper compartment and irradiated with 808 nm NIR magnets every 3 days for a total of three injections. After 24 h of each
laser or not. The residues of 4T1 cells were added into the 5 × 105 DC injection, mice were anesthetized and irradiated with NIR laser, followed
culture in the lower compartment using the transwell system. After by i.v. injection with PD-L1 antibody at a dose of 75 μg/mouse. The
various treatments, DCs stained with anti-CD11c, anti-CD80, and anti- primary and distant tumor volumes and body weights were monitored
CD86 antibody were analyzed by flow cytometry. over 24 days. After 24 days, all mice were sacrificed and both the primary
The proinflammatory cytokines in DC medium suspensions, and distant tumors were excised and weighed. The distant tumors were
including IL-6 and TNF-α, were determined by ELISA kits following harvested, treated with 1 mg/mL collagenase I for 1 h, and ground with
standard protocols. the end of a syringe rubber. Cells were filtered, incubated in red blood
Magnetic Attraction-Enhanced Pharmacokinetics, Biodistri- cell lysis buffer on ice for 5 min, and washed with PBS three times. To
bution, and MRI. The 4T1 tumor-bearing mice were intravenously reduce nonspecific binding to fragment crystallizable receptors (FcRs),
(i.v.) administrated with Fe3O4-R837 SPs at a dose of 6 mg/kg and the single-cell suspension was incubated with anti-CD16/32.27 Cells
attached to the magnets. Animals were sacrificed (n = 3) at the indicated were further stained with fluorochrome-conjugated antibodies: anti-
time points to collect the same dose of blood from each mouse. Then, CD45, anti-CD3ε, anti-CD4, anti-CD8a, anti-NKp46, anti-B220, and PI
the blood was immediately centrifuged at 3000 rpm for 5 min to harvest and analyzed by flow cytometry. Blood was also collected, and the serum
plasma samples and dissolved in aqua regia to analyze the total amount IL-6, TNF-α, and IFN-γ were determined by using ELISA kits to
of Fe3+ with ICP-OES. Major organs and tumors were also collected evaluate the acute inflammation evoked by the treatment.
after 24 h, and the concentration of Fe3+ was measured with ICP-OES. Characterization. Transmission electron microscopy (TEM)
Before collecting the organs, the mice need cardiac perfusion with images were measured by a Hitachi H-800 electron microscope.
physiological saline and paraformaldehyde to remove the effects of Scanning electron microscopy (SEM) images were acquired with a
blood in organs. The T2-weighted MRI images were acquired after 24 h SU8020 Hitachi Company scanning electron microscope. Dynamic
p.i. using a 1.5 T human clinical scanner. The organs’ and tumors’ light scattering (DLS) measurements were measured by Zetasizer
retention rates were calculated by the formula NanoZS, Malvern Instruments. UV−vis absorption spectra were
performed with Shimadzu 3600 UV−vis−IR spectrophotometer.
ID%/g = (drug content in organs Fourier-transform infrared (FTIR) spectra were obtained by Bruker
IFS66 V instrument. T2 relaxation time was measured by a Bruker
/total amount of drug injections)/organ weight AVANCE III 500 NMR spectroscope. An 808 nm diode laser (LEO
photonics Co. Ltd.) with tunable output power densities was employed
Apoptosis, DC Stimulation, and Cytokines Release in Vivo. to study the photothermal effect. Fluorescent images of 4T1 cells were
Female BALB/c mice (6−8 weeks) were divided into groups randomly. obtained by an Olympus IX71 inverted fluorescence microscope. The
The first 4T1 tumor model was planted by an orthotopic injection of 1 × Fe3+ concentration was assessed by PerkinElmer Optima 3300DV
106 4T1 cells into the right mammary fat pads of the mice, and tumors inductively coupled plasma optical emission (ICP-OES) measurements.
were allowed to grow until ∼200 mm3 before experiments. Then, the Infrared thermal images were acquired by a Fluke infrared (IR) thermal
mice were i.v. injected with Fe3O4 SPs/Fe3O4-R837 SPs at a dose of 6 camera. Flow cytometric analysis was implemented on the BD
mg/kg and attached to the magnets. According to the previous Biosciences FACSCalibur flow cytometer. T2-weighted MRI images
literature,21,27 the laser irradiation after 24 h of injection of NPs is an were performed by GE Signa 1.5 T unit, General Electric, Milwaukee,
ideal condition that has been optimized and verified. In addition, WI. The photographs of mice and lungs/liver were taken by
macroscopic digital imaging workstation for histopathology.


according to the blood circulation kinetics of Fe3O4-R837 SPs in vivo
and 24 h tumor retention rate, it can be seen that the high drug
accumulation in the tumor area after 24 h is beneficial to maximize the ASSOCIATED CONTENT
efficiency of photothermal therapy. Therefore, 24 h after i.v. injection is *
S Supporting Information
chosen as the time point for further experiments. After 24 h, mice were
The Supporting Information is available free of charge on the
anesthetized and tumors were irradiated with an 808 nm NIR laser at
0.33 W/cm2 for 30 min. Three days later, lymph nodes were excised and ACS Publications website at DOI: 10.1021/acsami.8b05876.
analyzed by flow cytometry. At the same time, blood was collected and Additional structural characterizations, the encapsulation
the serum IL-6, TNF-α, and IFN-γ were determined by using ELISA kits and loading efficiency, cellular uptake and efflux, the
to evaluate the acute inflammation evoked by the treatment. release of cytokine, the trend body weight of mice and
Antitumor and Anti-metastatic Activity on Orthotopic 4T1 serum biochemistry (PDF)


Model. Tumor-bearing mice were randomly divided into four groups (n
= 5): control, anti-PD-L1, Fe3O4-R837 SP PTT, and Fe3O4-R837 SP
PTT plus anti-PD-L1. The mice were i.v. injected with the materials and AUTHOR INFORMATION
attached to the magnets every 3 days three times. After 24 h of each Corresponding Authors
injection, mice were anesthetized and irradiated with NIR laser, followed *E-mail: daqizhang@yeah.net (D.Z.).
by i.v. injection with PD-L1 antibody at a dose of 75 μg/mouse. Tumor *E-mail: hou63@163.com (Y.H.).
volumes were monitored and recorded over 24 days. The volume of
*E-mail: hao_zhang@jlu.edu.cn. Fax: +86 431 85193423 (H.Z.).
tumor was calculated by the equation
ORCID
1
V = L × D2 Yi Liu: 0000-0003-0548-6073
2 Hongchen Sun: 0000-0002-5572-508X
20353 DOI: 10.1021/acsami.8b05876
ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

Hao Zhang: 0000-0002-2373-1100 (16) Matsuo, Y.; Takeyama, H.; Guha, S. Cytokine Network: New
Bai Yang: 0000-0002-3873-075X Targeted Therapy for Pancreatic Cancer. Curr. Pharm. Des. 2012, 18,
2416−2419.
Author Contributions (17) Frick, S. U.; Domogalla, M. P.; Baier, G.; Wurm, F. R.; Mailänder,
H.Z. proposed and supervised the project. H.Z., R.G., X.Z., Y.L., V.; Landfester, K.; Steinbrink, K. Interleukin-2 Functionalized Nano-
H.S., D.Z., Y.H., and B.Y. designed and performed the capsules for T Cell-Based Immunotherapy. ACS Nano 2016, 10, 9216−
experiments and co-wrote the manuscript. C.L., B.L., J.L., and 9226.
W.W. participated in most experiments. All authors discussed the (18) Sabado, R. L.; Balan, S.; Bhardwaj, N. Dendritic Cell-Based
results and commented on the manuscript. Immunotherapy. Cell Res. 2017, 27, 74−95.
(19) Kim, S.-Y.; Phuengkham, H.; Noh, Y.-W.; Lee, H.-G.; Um, S. H.;
Notes Lim, Y. T. Immune Complexes Mimicking Synthetic Vaccine Nano-
The authors declare no competing financial interest. particles for Enhanced Migration and Cross-Presentation of Dendritic


Cells. Adv. Funct. Mater. 2016, 26, 8072−8082.
(20) Zhu, G.; Zhang, F.; Ni, Q.; Niu, G.; Chen, X. Efficient
ACKNOWLEDGMENTS
Nanovaccine Delivery in Cancer Immunotherapy. ACS Nano 2017,
This work was supported by NSFC (51603084 and 51425303), 11, 2387−2392.
JLU Science and Technology Innovative Research Team (21) Chen, Q.; Xu, L.; Liang, C.; Wang, C.; Peng, R.; Liu, Z.
2017TD-06, and the Special Project from MOST of China. Photothermal Therapy with Immune-Adjuvant Nanoparticles Together


with Checkpoint Blockade for Effective Cancer Immunotherapy. Nat.
Commun. 2016, 7, No. 13193.
REFERENCES (22) Carreno, B. M.; Magrini, V.; Becker-Hapak, M.; Kaabinejadian, S.;
(1) Siegel, R. L.; Miller, K. D.; Jemal, A. Cancer Statistics, 2016. CA Hundal, J.; Petti, A. A.; Ly, A.; Lie, W.-R.; Hildebrand, W. H.; Mardis, E.
Cancer J. Clin. 2016, 66, 7−30. R.; Linette, G. P. A Dendritic Cell Vaccine Increases the Breadth and
(2) Couzin-Frankel, J. Cancer Immunotherapy. Science 2013, 342, Diversity of Melanoma Neoantigen-Specific T Cells. Science 2015, 348,
1432−1433. 803−808.
(3) Gotwals, P.; Cameron, S.; Cipolletta, D.; Cremasco, V.; Crystal, A.; (23) Fang, R. H.; Hu, C.-M. J.; Luk, B. T.; Gao, W.; Copp, J. A.; Tai, Y.;
Hewes, B.; Mueller, B.; Quaratino, S.; Sabatos-Peyton, C.; Petruzzelli, O’Connor, D. E.; Zhang, L. Cancer Cell Membrane-Coated Nano-
L.; Engelman, J. A.; Dranoff, G. Prospects for Combining Targeted and particles for Anticancer Vaccination and Drug Delivery. Nano Lett. 2014,
Conventional Cancer Therapy with Immunotherapy. Nat. Rev. Cancer 14, 2181−2188.
2017, 17, 286−301. (24) Ledford, H. Immunotherapy’s Cancer Remit Widens. Nature
(4) Jiang, W.; von Roemeling, C. A.; Chen, Y.; Qie, Y.; Liu, X.; Chen, J.; 2013, 497, 544.
Kim, B. Y. S. Designing Nanomedicine for Immuno-Oncology. Nat. (25) Wang, R.-F. A Special Issue on Cancer Immunotherapy. Cell Res.
Biomed. Eng. 2017, 1, No. 0029. 2017, 27, 1−2.
(5) Shao, K.; Singha, S.; Clemente-Casares, X.; Tsai, S.; Yang, Y.; (26) Webster, R. M. The Immune Checkpoint Inhibitors: Where Are
Santamaria, P. Nanoparticle-Based Immunotherapy for Cancer. ACS We Now? Nat. Rev. Drug Discovery 2014, 13, 883−884.
Nano 2015, 9, 16−30. (27) Duan, X.; Chan, C.; Guo, N.; Han, W.; Weichselbaum, R. R.; Lin,
(6) Rosenberg, S. A.; Yang, J. C.; Restifo, N. P. Cancer W. Photodynamic Therapy Mediated by Nontoxic Core-Shell Nano-
Immunotherapy: Moving Beyond Current Vaccines. Nat. Med. 2004, particles Synergizes with Immune Checkpoint Blockade To Elicit
10, 909−915. Antitumor Immunity and Antimetastatic Effect on Breast Cancer. J. Am.
(7) Grivennikov, S. I.; Greten, F. R.; Karin, M. Immunity, Chem. Soc. 2016, 138, 16686−16695.
Inflammation, and Cancer. Cell 2010, 140, 883−899. (28) Drake, C. G.; Jaffee, E.; Pardoll, D. M. Mechanisms of Immune
(8) Irvine, D. J.; Hanson, M. C.; Rakhra, K.; Tokatlian, T. Synthetic Evasion by Tumors. Adv. Immunol. 2006, 90, 51−81.
Nanoparticles for Vaccines and Immunotherapy. Chem. Rev. 2015, 115, (29) Liu, Z.; Ravindranathan, R.; Kalinski, P.; Guo, Z. S.; Bartlett, D. L.
11109−11146. Rational Combination of Oncolytic Vaccinia Virus and PD-L1 Blockade
(9) Curran, K. J.; Brentjens, R. J. Chimeric Antigen Receptor T cells for
Works Synergistically to Enhance Therapeutic Efficacy. Nat. Commun.
Cancer Immunotherapy. J. Clin. Oncol. 2015, 33, 1703−1706.
2017, 8, No. 14754.
(10) Kalos, M.; June, C. H. Adoptive T Cell Transfer for Cancer
(30) Taube, J. M.; Klein, A.; Brahmer, J. R.; Xu, H.; Pan, X.; Kim, J. H.;
Immunotherapy in the Era of Synthetic Biology. Immunity 2013, 39,
Chen, L.; Pardoll, D. M.; Topalian, S. L.; Anders, R. A. Association of
49−60.
PD-1, PD-1 Ligands, and Other Features of the Tumor Immune
(11) Restifo, N. P.; Dudley, M. E.; Rosenberg, S. A. Adoptive
Immunotherapy for Cancer: Harnessing the T Cell Response. Nat. Rev. Microenvironment with Response to Anti-PD-1 Therapy. Clin. Cancer
Immunol. 2012, 12, 269−281. Res. 2014, 20, 5064−5074.
(12) Gubin, M. M.; Zhang, X.; Schuster, H.; Caron, E.; Ward, J. P.; (31) Errico, A. PD-1-PD-L1 Axis: Efficient Checkpoint Blockade
Noguchi, T.; Ivanova, Y.; Hundal, J.; Arthur, C. D.; Krebber, W.-J.; Against Cancer. Nat. Rev. Clin. Oncol. 2015, 12, 63.
Mulder, G. E.; Toebes, M.; Vesely, M. D.; Lam, S. S.; Korman, A. J.; (32) Topalian, S. L.; Hodi, F. S.; Brahmer, J. R.; Gettinger, S. N.; Smith,
Allison, J. P.; Freeman, G. J.; Sharpe, A. H.; Pearce, E. L.; Schumacher, T. D. C.; McDermott, D. F.; Powderly, J. D.; Carvajal, R. D.; Sosman, J. A.;
N.; Aebersold, R.; Rammensee, H. G.; Melief, C. J.; Mardis, E. R.; Atkins, M. B.; Leming, P. D.; Spigel, D. R.; Antonia, S. J.; Horn, L.;
Gillanders, W. E.; Artyomov, M. N.; Schreiber, R. D. Checkpoint Drake, C. G.; Pardoll, D. M.; Chen, L.; Sharfman, W. H.; Anders, R. A.;
Blockade Cancer Immunotherapy Targets Tumour-Specific Mutant Taube, J. M.; McMiller, T. L.; Xu, H.; Korman, A. J.; Jure-Kunkel, M.;
Antigens. Nature 2014, 515, 577−581. Agrawal, S.; McDonald, D.; Kollia, G. D.; Gupta, A.; Wigginton, J. M.;
(13) Pardoll, D. M. The Blockade of Immune Checkpoints in Cancer Sznol, M. Safety, Activity, and Immune Correlates of Anti-PD-1
Immunotherapy. Nat. Rev. Cancer 2012, 12, 252−264. Antibody in Cancer. N. Engl. J. Med. 2012, 366, 2443−2454.
(14) Wang, C.; Sun, W.; Wright, G.; Wang, A. Z.; Gu, Z. Inflammation- (33) Tumeh, P. C.; Harview, C. L.; Yearley, J. H.; Shintaku, I. P.;
Triggered Cancer Immunotherapy by Programmed Delivery of CpG Taylor, E. J.; Robert, L.; Chmielowski, B.; Spasic, M.; Henry, G.;
and Anti-PD1 Antibody. Adv. Mater. 2016, 28, 8912−8920. Ciobanu, V.; West, A. N.; Carmona, M.; Kivork, C.; Seja, E.; Cherry, G.;
(15) Wang, D.; Wang, T.; Liu, J.; Yu, H.; Jiao, S.; Feng, B.; Zhou, F.; Fu, Gutierrez, A. J.; Grogan, T. R.; Mateus, C.; Tomasic, G.; Glaspy, J. A.;
Y.; Yin, Q.; Zhang, P.; Zhang, Z.; Zhou, Z.; Li, Y. Acid-Activatable Emerson, R. O.; Robins, H.; Pierce, R. H.; Elashoff, D. A.; Robert, C.;
Versatile Micelleplexes for PD-L1 Blockade-Enhanced Cancer Photo- Ribas, A. PD-1 Blockade Induces Responses by Inhibiting Adaptive
dynamic Immunotherapy. Nano Lett. 2016, 16, 5503−5513. Immune Resistance. Nature 2014, 515, 568−571.

20354 DOI: 10.1021/acsami.8b05876


ACS Appl. Mater. Interfaces 2018, 10, 20342−20355
ACS Applied Materials & Interfaces Research Article

(34) Rizvi, N. A.; Hellmann, M. D.; Snyder, A.; Kvistborg, P.; Makarov, Feasible Approach for Clinics. J. Am. Chem. Soc. 2015, 137, 15145−
V.; Havel, J. J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T. S.; Miller, M. L.; 15151.
Rekhtman, N.; Moreira, A. L.; Ibrahim, F.; Bruggeman, C.; Gasmi, B.; (47) Zhang, S.; Sun, C.; Zeng, J.; Sun, Q.; Wang, G.; Wang, Y.; Wu, Y.;
Zappasodi, R.; Maeda, Y.; Sander, C.; Garon, E. B.; Merghoub, T.; Dou, S.; Gao, M.; Li, Z. Ambient Aqueous Synthesis of Ultrasmall
Wolchok, J. D.; Schumacher, T. N.; Chan, T. A. Mutational Landscape PEGylated Cu2‑xSe Nanoparticles as a Multifunctional Theranostic
Determines Sensitivity to PD-1 Blockade in Non-Small Cell Lung Agent for Multimodal Imaging Guided Photothermal Therapy of
Cancer. Science 2015, 348, 124−128. Cancer. Adv. Mater. 2016, 28, 8927−8936.
(35) He, C.; Duan, X.; Guo, N.; Chan, C.; Poon, C.; Weichselbaum, R. (48) Shen, S.; Wang, S.; Zheng, R.; Zhu, X.; Jiang, X.; Fu, D.; Yang, W.
R.; Lin, W. Core-Shell Nanoscale Coordination Polymers Combine Magnetic Nanoparticle Clusters for Photothermal Therapy with Near-
Chemotherapy and Photodynamic Therapy to Potentiate Checkpoint Infrared Irradiation. Biomaterials 2015, 39, 67−74.
Blockade Cancer Immunotherapy. Nat. Commun. 2016, 7, No. 12499. (49) Lin, L.-S.; Cong, Z.-X.; Cao, J.-B.; Ke, K.-M.; Peng, Q.-L.; Gao, J.;
(36) Zheng, D.-W.; Chen, J.-L.; Zhu, J.-Y.; Rong, L.; Li, B.; Lei, Q.; Fan, Yang, H.-H.; Liu, G.; Chen, X. Multifunctional Fe3O4@Polydopamine
J.-X.; Zou, M.-Z.; Li, C.; Cheng, S.-X.; Xu, Z.; Zhang, X.-Z. Highly Core-Shell Nanocomposites for Intracellular mRNA Detection and
Integrated Nano-Platform for Breaking the Barrier between Chemo- Imaging-Guided Photothermal Therapy. ACS Nano 2014, 8, 3876−
therapy and Immunotherapy. Nano Lett. 2016, 16, 4341−4347. 3883.
(37) Bauleth-Ramos, T.; Shahbazi, M.-A.; Liu, D.; Fontana, F.; Correia, (50) Ge, R.; Li, X.; Lin, M.; Wang, D.; Li, S.; Liu, S.; Tang, Q.; Liu, Y.;
A.; Figueiredo, P.; Zhang, H.; Martins, J. P.; Hirvonen, J. T.; Granja, P.; Jiang, J.; Liu, L.; Sun, H.; Zhang, H.; Yang, B. Fe3O4@Polydopamine
Sarmento, B.; Santos, H. A. Nutlin-3a and Cytokine Co-loaded Composite Theranostic Superparticles Employing Preassembled Fe3O4
Spermine-Modified Acetalated Dextran Nanoparticles for Cancer Nanoparticles as the Core. ACS Appl. Mater. Interfaces 2016, 8, 22942−
Chemo-Immunotherapy. Adv. Funct. Mater. 2017, 27, No. 1703303. 22952.
(38) Pelaz, B.; Alexiou, C.; Alvarez-Puebla, R. A.; Alves, F.; Andrews, A. (51) Xia, Y.; Tang, Z. Monodisperse Inorganic Supraparticles:
M.; Ashraf, S.; Balogh, L. P.; Ballerini, L.; Bestetti, A.; Brendel, C.; Bosi, Formation Mechanism, Properties and Applications. Chem. Commun.
S.; Carril, M.; Chan, W. C.; Chen, C.; Chen, X.; Chen, X.; Cheng, Z.; 2012, 48, 6320−6336.
Cui, D.; Du, J.; Dullin, C.; Escudero, A.; Feliu, N.; Gao, M.; George, M.; (52) Zhuang, J.; Wu, H.; Yang, Y.; Cao, Y. C. Controlling Colloidal
Gogotsi, Y.; Grunweller, A.; Gu, Z.; Halas, N. J.; Hampp, N.; Hartmann, Superparticle Growth Through Solvophobic Interactions. Angew. Chem.,
R. K.; Hersam, M. C.; Hunziker, P.; Jian, J.; Jiang, X.; Jungebluth, P.; Int. Ed. 2008, 47, 2208−2212.
Kadhiresan, P.; Kataoka, K.; Khademhosseini, A.; Kopecek, J.; Kotov, N. (53) Bai, F.; Wang, D.; Huo, Z.; Chen, W.; Liu, L.; Liang, X.; Chen, C.;
A.; Krug, H. F.; Lee, D. S.; Lehr, C. M.; Leong, K. W.; Liang, X. J.; Ling Wang, X.; Peng, Q.; Li, Y. A Versatile Bottom-up Assembly Approach to
Lim, M.; Liz-Marzan, L. M.; Ma, X.; Macchiarini, P.; Meng, H.; Colloidal Spheres from Nanocrystals. Angew. Chem., Int. Ed. 2007, 46,
Mohwald, H.; Mulvaney, P.; Nel, A. E.; Nie, S.; Nordlander, P.; Okano, 6650−6653.
T.; Oliveira, J.; Park, T. H.; Penner, R. M.; Prato, M.; Puntes, V.; Rotello, (54) Sun, S.; Zeng, H. Size-Controlled Synthesis of Magnetite
V. M.; Samarakoon, A.; Schaak, R. E.; Shen, Y.; Sjoqvist, S.; Skirtach, A. Nanoparticles. J. Am. Chem. Soc. 2002, 124, 8204−8205.
G.; Soliman, M. G.; Stevens, M. M.; Sung, H. W.; Tang, B. Z.; Tietze, R.; (55) Wang, S.; Huang, P.; Chen, X. Hierarchical Targeting Strategy for
Udugama, B. N.; VanEpps, J. S.; Weil, T.; Weiss, P. S.; Willner, I.; Wu, Y.; Enhanced Tumor Tissue Accumulation/Retention and Cellular
Yang, L.; Yue, Z.; Zhang, Q.; Zhang, Q.; Zhang, X. E.; Zhao, Y.; Zhou, X.; Internalization. Adv. Mater. 2016, 28, 7340−7364.
Parak, W. J. Diverse Applications of Nanomedicine. ACS Nano 2017, 11, (56) Rohrer, M.; Bauer, H.; Mintorovitch, J.; Requardt, M.;
2313−2381. Weinmann, H.-J. Comparison of Magnetic Properties of MRI Contrast
(39) Zhou, F.; Wu, S.; Song, S.; Chen, W. R.; Resasco, D. E.; Xing, D. Media Solutions at Different Magnetic Field Strengths. Invest. Radiol.
Antitumor Immunologically Modified Carbon Nanotubes for Photo- 2005, 40, 715−724.
thermal Therapy. Biomaterials 2012, 33, 3235−3242. (57) Miller, R. L.; Gerster, J. F.; Owens, M. L.; Slade, H. B.; Tomai, M.
(40) Tao, Y.; Ju, E.; Ren, J.; Qu, X. Immunostimulatory A. Imiquimod Applied Topically: a Novel Immune Response Modifier
Oligonucleotides-Loaded Cationic Graphene Oxide with Photo- and New Class of Drug. Int. J. Immunopharmacol. 1999, 21, 1−14.
thermally Enhanced Immunogenicity for Photothermal/Immune (58) Kim, H. J.; Lee, S.-M.; Park, K.-H.; Mun, C. H.; Park, Y.-B.; Yoo,
Cancer Therapy. Biomaterials 2014, 35, 9963−9971. K.-H. Drug-loaded Gold/Iron/Gold Plasmonic Nanoparticles for
(41) Guo, L.; Yan, D. D.; Yang, D.; Li, Y.; Wang, X.; Zalewski, O.; Yan, Magnetic Targeted Chemo-photothermal Treatment of Rheumatoid
B.; Lu, W. Combinatorial Photothermal and Immuno Cancer Therapy Arthritis. Biomaterials 2015, 61, 95−102.
Using Chitosan-Coated Hollow Copper Sulfide Nanoparticles. ACS (59) Wong, C. W.; Song, C.; Grimes, M. M.; Fu, W.; Dewhirst, M. W.;
Nano 2014, 8, 5670−5681. Muschel, R. J.; Al-Mehdi, A.-B. Intravascular Location of Breast Cancer
(42) He, J.; Huang, X.; Li, Y.-C.; Liu, Y.; Babu, T.; Aronova, M. A.; Cells after Spontaneous Metastasis to the Lung. Am. J. Pathol. 2002, 161,
Wang, S.; Lu, Z.; Chen, X.; Nie, Z. Self-Assembly of Amphiphilic 749−753.
Plasmonic Micelle-Like Nanoparticles in Selective Solvents. J. Am. (60) Aslakson, C. J.; Miller, F. R. Selective Events in the Metastatic
Process Defined by Analysis of the Sequential Dissemination of
Chem. Soc. 2013, 135, 7974−7984.
Subpopulations of a Mouse Mammary Tumor. Cancer Res. 1992, 52,
(43) Wang, J.; Liu, J.; Liu, Y.; Wang, L.; Cao, M.; Ji, Y.; Wu, X.; Xu, Y.;
1399−1405.
Bai, B.; Miao, Q.; Chen, C.; Zhao, Y. Gd-Hybridized Plasmonic Au-
Nanocomposites Enhanced Tumor-Interior Drug Permeability in
Multimodal Imaging-Guided Therapy. Adv. Mater. 2016, 28, 8950−
8958.
(44) Du, Y.; Jiang, Q.; Beziere, N.; Song, L.; Zhang, Q.; Peng, D.; Chi,
C.; Yang, X.; Guo, H.; Diot, G.; Ntziachristos, V.; Ding, B.; Tian, J.
DNA-Nanostructure-Gold-Nanorod Hybrids for Enhanced In Vivo
Optoacoustic Imaging and Photothermal Therapy. Adv. Mater. 2016, 28,
10000−10007.
(45) Li, Y.; Shi, Q.; Zhang, P.; Xiahou, Y.; Li, S.; Wang, D.; Xia, H.
Empirical Structural Design of Core@Shell Au@Ag Nanoparticles for
SERS Applications. J. Mater. Chem. C 2016, 4, 6649−6656.
(46) Riedinger, A.; Avellini, T.; Curcio, A.; Asti, M.; Xie, Y.; Tu, R.;
Marras, S.; Lorenzoni, A.; Rubagotti, S.; Iori, M.; Capponi, P. C.; Versari,
A.; Manna, L.; Seregni, E.; Pellegrino, T. Post-Synthesis Incorporation
of 64Cu in CuS Nanocrystals to Radiolabel Photothermal Probes: A

20355 DOI: 10.1021/acsami.8b05876


ACS Appl. Mater. Interfaces 2018, 10, 20342−20355

You might also like