9
1904997 (1 of 9) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.advmat.de COMMUNICATION Tumor-Targeted Drug and CpG Delivery System for Phototherapy and Docetaxel-Enhanced Immunotherapy with Polarization toward M1-Type Macrophages on Triple Negative Breast Cancers Lv Chen, Lulu Zhou, Chunhui Wang, Yi Han, Yonglin Lu, Jie Liu, Xiaochun Hu, Tianming Yao, Yun Lin, Shujing Liang, Shuo Shi,* and Chunyan Dong* L. Chen, Y. Han, Y. Lu, J. Liu, Dr. Y. Lin, Dr. S. Liang, Prof. C. Dong Breast Cancer Center Shanghai East Hospital Tongji University Shanghai 200120, P. R. China E-mail: [email protected] L. Zhou, C. Wang, X. Hu, Prof. T. Yao, Prof. S. Shi Shanghai Key Laboratory of Chemical Assessment and Sustainability School of Chemical Science and Engineering Tongji University Shanghai 200092, P. R. China E-mail: [email protected] The ORCID identification number(s) for the author(s) of this article can be found under https://doi.org/10.1002/adma.201904997. DOI: 10.1002/adma.201904997 females. [1] Traditional tumor-therapeutic modalities widely used in clinic, such as surgical resection, chemotherapy, and radiotherapy, often cause severe side effects to normal tissues and the immune system after a period of treatment. [2] In recent years, owing to the approval of the cytotoxic T-lymphocyte-associated pro- tein-4 antibody ipilimumab, [3] the pro- grammed cell death protein 1 antibody nivolumnab [4] and its ligand (programmed cell death-ligand 1 (PD-L1)) antibody ate- zolizumab, [5] cancer immunotherapy has brought a revolution in cancer therapy. Immunotherapy by activating cytotoxic T lymphocytes (CTLs) from dysfunction and exhaustion, as well as inhibiting immune- suppressive regulatory T cells, further activating host immune system to rec- ognize, attack, and eradicate tumor cells, has been regarded as a promising and effective strategy in clinic treatment. [6] As an immunoadjuvant, unmethylated cyto- sine–phosphate–guanine oligonucleotides (CpG) can specifically bind with Toll-like receptor 9 in plasmacytoid antigen pre- senting cells (APCs) (including dendritic cells and macrophages), [7] thus stimulate an immune response to produce helper T cell 1 (Th1) and a proinflammatory cytokine (such as tumor necrosis factor alpha, TNF-α; IL-12 (where IL stands for interleukin)). [8] IL-12, as one of most effec- tive stimulator of CTLs, could promote maturity of CTLs and infiltration in tumor sites. Simultaneously, activated CTLs could secrete interferon gamma (IFN-γ) to kill tumor cells. Compared with the excellent effects of immunotherapy in melanoma, [9] non-small-cell lung cancer [10] and other malignant tumors, [11] immunotherapy works less effectively on breast cancer, mainly because of the efficient filtration of lymphocytes [12] and mye- loid-derived suppressor cells (MDSCs). [13] MDSCs, as a group of heterogeneous and immature cells, express myeloid lin- eage marker Gr-1 and CD11b, induce T cells dysfunction and further reduce proliferation, then increase T cells apoptosis of antigen-specific CD8+ T cells, leading to immune suppres- sion. [14] Tumor-related MDSCs can be divided into two subsets: Cancer immunotherapy has achieved promising clinical responses in recent years owing to the potential of controlling metastatic disease. However, there is a limited research to prove the superior therapeutic efficacy of immuno- therapy on breast cancer compared with melanoma and non-small-cell lung cancer because of its limited expression of PD-L1, low infiltration of cytotoxic T lymphocytes (CTLs), and high level of myeloid-derived suppressor cells (MDSCs). Herein, a multifunctional nanoplatform (FA-CuS/DTX@PEI-PpIX- CpG nanocomposites, denoted as FA-CD@PP-CpG) for synergistic photo- therapy (photodynamic therapy (PDT), photothermal therapy (PTT) included) and docetaxel (DTX)-enhanced immunotherapy is successfully developed. The nanocomposites exhibit excellent PDT efficacy and photothermal conver- sion capability under 650 and 808 nm irradiation, respectively. More signifi- cantly, FA-CD@PP-CpG with no obvious side effects can remarkably inhibit the tumor growth in vivo based on a 4T1-tumor-bearing mice modal. A low dosage of loaded DTX in FA-CD@PP-CpG can promote infiltration of CTLs to improve efficacy of anti-PD-L1 antibody (aPD-L1), suppress MDSCs, and effectively polarize MDSCs toward M1 phenotype to reduce tumor burden, further to enhance the antitumor efficacy. Taken together, FA-CD@PP-CpG nanocomposites offer an efficient synergistic therapeutic modality in doce- taxel-enhanced immunotherapy for clinical application of breast cancer. Currently, breast cancer is the most common malignant tumor and has become a significant cause of cancer-related deaths in Adv. Mater. 2019, 1904997

Tumor‐Targeted Drug and CpG Delivery System for ... · In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Tumor‐Targeted Drug and CpG Delivery System for ... · In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced

1904997 (1 of 9) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

www.advmat.de

COMMUNICATION

Tumor-Targeted Drug and CpG Delivery System for Phototherapy and Docetaxel-Enhanced Immunotherapy with Polarization toward M1-Type Macrophages on Triple Negative Breast Cancers

Lv Chen, Lulu Zhou, Chunhui Wang, Yi Han, Yonglin Lu, Jie Liu, Xiaochun Hu, Tianming Yao, Yun Lin, Shujing Liang, Shuo Shi,* and Chunyan Dong*

L. Chen, Y. Han, Y. Lu, J. Liu, Dr. Y. Lin, Dr. S. Liang, Prof. C. DongBreast Cancer CenterShanghai East HospitalTongji UniversityShanghai 200120, P. R. ChinaE-mail: [email protected]. Zhou, C. Wang, X. Hu, Prof. T. Yao, Prof. S. ShiShanghai Key Laboratory of Chemical Assessment and SustainabilitySchool of Chemical Science and EngineeringTongji UniversityShanghai 200092, P. R. ChinaE-mail: [email protected]

The ORCID identification number(s) for the author(s) of this article can be found under https://doi.org/10.1002/adma.201904997.

DOI: 10.1002/adma.201904997

females.[1] Traditional tumor-therapeutic modalities widely used in clinic, such as surgical resection, chemotherapy, and radiotherapy, often cause severe side effects to normal tissues and the immune system after a period of treatment.[2] In recent years, owing to the approval of the cytotoxic T-lymphocyte-associated pro-tein-4 antibody ipilimumab,[3] the pro-grammed cell death protein 1 antibody nivolumnab[4] and its ligand (programmed cell death-ligand 1 (PD-L1)) antibody ate-zolizumab,[5] cancer immunotherapy has brought a revolution in cancer therapy. Immunotherapy by activating cytotoxic T lymphocytes (CTLs) from dysfunction and exhaustion, as well as inhibiting immune-suppressive regulatory T cells, further activating host immune system to rec-ognize, attack, and eradicate tumor cells, has been regarded as a promising and effective strategy in clinic treatment.[6] As an immunoadjuvant, unmethylated cyto-sine–phosphate–guanine oligonucleotides (CpG) can specifically bind with Toll-like receptor 9 in plasmacytoid antigen pre-senting cells (APCs) (including dendritic

cells and macrophages),[7] thus stimulate an immune response to produce helper T cell 1 (Th1) and a proinflammatory cytokine (such as tumor necrosis factor alpha, TNF-α; IL-12 (where IL stands for interleukin)).[8] IL-12, as one of most effec-tive stimulator of CTLs, could promote maturity of CTLs and infiltration in tumor sites. Simultaneously, activated CTLs could secrete interferon gamma (IFN-γ) to kill tumor cells. Compared with the excellent effects of immunotherapy in melanoma,[9] non-small-cell lung cancer[10] and other malignant tumors,[11] immunotherapy works less effectively on breast cancer, mainly because of the efficient filtration of lymphocytes[12] and mye-loid-derived suppressor cells (MDSCs).[13] MDSCs, as a group of heterogeneous and immature cells, express myeloid lin-eage marker Gr-1 and CD11b, induce T cells dysfunction and further reduce proliferation, then increase T cells apoptosis of antigen-specific CD8+ T cells, leading to immune suppres-sion.[14] Tumor-related MDSCs can be divided into two subsets:

Cancer immunotherapy has achieved promising clinical responses in recent years owing to the potential of controlling metastatic disease. However, there is a limited research to prove the superior therapeutic efficacy of immuno-therapy on breast cancer compared with melanoma and non-small-cell lung cancer because of its limited expression of PD-L1, low infiltration of cytotoxic T lymphocytes (CTLs), and high level of myeloid-derived suppressor cells (MDSCs). Herein, a multifunctional nanoplatform (FA-CuS/DTX@PEI-PpIX-CpG nanocomposites, denoted as FA-CD@PP-CpG) for synergistic photo-therapy (photodynamic therapy (PDT), photothermal therapy (PTT) included) and docetaxel (DTX)-enhanced immunotherapy is successfully developed. The nanocomposites exhibit excellent PDT efficacy and photothermal conver-sion capability under 650 and 808 nm irradiation, respectively. More signifi-cantly, FA-CD@PP-CpG with no obvious side effects can remarkably inhibit the tumor growth in vivo based on a 4T1-tumor-bearing mice modal. A low dosage of loaded DTX in FA-CD@PP-CpG can promote infiltration of CTLs to improve efficacy of anti-PD-L1 antibody (aPD-L1), suppress MDSCs, and effectively polarize MDSCs toward M1 phenotype to reduce tumor burden, further to enhance the antitumor efficacy. Taken together, FA-CD@PP-CpG nanocomposites offer an efficient synergistic therapeutic modality in doce-taxel-enhanced immunotherapy for clinical application of breast cancer.

Currently, breast cancer is the most common malignant tumor and has become a significant cause of cancer-related deaths in

Adv. Mater. 2019, 1904997

Page 2: Tumor‐Targeted Drug and CpG Delivery System for ... · In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced

© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim1904997 (2 of 9)

www.advmat.dewww.advancedsciencenews.com

M1 [marker: CCR7] and M2 [marker: mannose receptor (MR) and IL-10], which also named as M2 tumor-associated mac-rophages (TAMs).[15] As we know, M1-type macrophages are generally recognized as potent effector cells, which produce various pro-inflammatory cytokines (e.g., IL-12 and TNF-α) and inducible nitric oxide synthase (iNOS), then kill tumor cells. In contrast, M2-Type macrophages can secrete IL-10, IL-4, and IL-13 to promote tumor angiogenesis.[16] So, the key to enhance immunotherapy is to promote the infiltration of CTLs, reduce MDSCs and the ratio of TAM in MDSCs.

In traditional view, chemotherapeutics causes a suppressive effect on immune system for patients. Recent research revealed that low dosage of traditional chemotherapeutics could induce immunogenic death of tumor cells or engage immune effector mechanisms to stimulate tumor-specific immune responses.[17] Especially, docetaxel (DTX) can decrease MDSCs proportion and induce MDSCs polarizing toward an M1-like phenotype, not M2-like phenotype, and work not only on animal models[17b,18a,b] but also human patients.[18c] However, there is a challenge to design a smart nanosystem to combine low dosage chemotherapy and immunotherapy.

Recently, photodynamic therapy (PDT) and photothermal therapy (PTT) as minimally invasive tumor-therapeutic modal-ities have attracted increasing attention. During PDT, photo-sensitizers can transform surrounding oxygen molecules into cytotoxic reactive oxygen species (ROS) under light irradiation, especially singlet oxygen (1O2), leading to irreversibly protein or DNA damage, subsequently causing apoptosis and necrosis of tumor cells.[19] As for photothermal therapy, PTT usually involves light absorbing agents with a strong near-infrared (NIR) absorbance to generate heat energy and cause thermal ablation of tumor cells after photoirradiation.[20] Although PDT and PTT are promising tumor-therapeutics modalities, there inherent shortcomings, such as nonspecific accumulation at health tissues for nontargeted photosensitizers and hardness to decompose in living systems with potential toxicity prob-lems severely limit their extensive applications.[21] So rational design of smart nanomaterial that could combine multi-modal therapy and overcome their own inherent limitations is of great interest and significance toward successful cancer treatment.

In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced immunotherapy. Mesoporous CuS nanoparti-cles (NPs) were chosen as the drug nanocarrier and PTT agents owing to the high photothermal conversation efficiency and excellent biocompability.[22] Then, the surface modification with tumor target ligand folic acid (FA) was favorable for improving active delivery of nanoparticles, further enhancing the transport efficiency of DTX at tumor sites through targeting effect of FA to overexpressed FA receptors on the tumor cells.[23] To improve the water solubility of nanoparticles and further realize CpG delivery, PEI-PpIX (polyethylenimine–protoporphyrin IX) conjugates (Figure S1, Supporting Information) and CpG were anchored alternatively to fabricate FA-CuS/DTX@PEI-PpIX-CpG (denoted as FA-CD@PP-CpG) nanocomposite (Scheme 1). On the basis of scanning electron microscopy (SEM) and transmission electron microscopy (TEM) images, the as-synthesized CuS NPs showed monodisperse spherical

shape and uniform size about 100 nm (Figure 1a,d). Next, after conjugation of FA and DTX loading, there was negligible change of the morphology compared to CuS (Figure 1b,e). The average size of FA-CD@PP-CpG nanocomposite increased to 117 nm; furthermore, there was a thin polymer shell wrapped around the FA-CuS/DTX core, which might result from the anchor of PEI-PpIX conjugate (Figure 1c,f). The average diam-eter of nanoparticles could also be determined by dynamic light scattering (DLS) analysis, which showed the size increased from 102.9 ± 2.7 nm of CuS to 115.6 ± 2.3 nm of FA-CD@PP-CpG (Figure S2, Supporting Information). Energy disper-sive spectrometer (EDS) and X-ray photoelectron spectros-copy (XPS) revealed that Cu, S, O, C, N were presented in the sample, indicating the successful preparation of FA-CD@PP-CpG nanocomposite (Figures S3a,b and S5a, Supporting Information). And the content of polymer measured by ther-mogravimetric analysis (TGA) was confirmed to be 20.15% (Figure S3c, Supporting Information). Also, FA-CD@PP-CpG nanocomposite dispersed well in water, phosphate buffer saline (PBS), Dulbecco’s modified Eagle medium (DMEM), and fetal bovine serum (FBS) (inset of Figure 1f), which was verified by the hydrodynamic size and zeta-potential of FA-CD@PP-CpG in different solutions (Figure S4a, Supporting Information). Moreover, the stability of the particles was monitored for ten days through DLS measurement. No obvious changes about the size of FA-CD@PP-CpG were observed during the tracking period, demonstrating the good stability of the nanocomposite in water and physiological solution (Figure S4b, Supporting Information).

X-ray powder diffraction (XRD) pattern (Figure S5b, Sup-porting Information) demonstrated a covellite CuS phase (JCPDS No. 06–0464). The XRD data of FA-CD@PP-CpG nano-composite showed that the peaks intensity decreased, which may be attributed to the filling of pores with the drug and the high degree of disorder.[24] The final FA-CD@PP-CpG nano-composite showed a decrease in potential to −16 mV compared to the positive potential of FA-CD@PP (+ 20.2 mV), indicating the successful anchor of CpG due to the negative charge of DNA (Figure S5c, Supporting Information). From UV–vis–NIR absorption spectra, FA-CD@PP exhibited new absorption peak of FA at near 280 nm, as well as the characteristic peak of PEI-PpIX (Figure S6a,b, Supporting Information). The results were also validated from the change of the color before and after functionalization (inset of Figure S6b, Supporting Informa-tion). Then we studied the loading efficiency of DTX with high-performance liquid chromatography (HPLC). According to the standard curve of DTX at 227 nm (Figure S6c, Supporting Information), the drug loading efficiency was determined to be about 23%. Such high loading capacity for the hydrophobic drug suggested that CuS NPs held great potential as a prom-ising nanocarrier for drug delivery.

Subsequently, diphenylisobenzofuran (DPBF) was utilized to evaluate the singlet oxygen production ability of FA-CD@PP-CpG under 650 nm irradiation (Figure 1g). In the pres-ence of FA-CD@PP-CpG, DPBF absorption decayed gradually along with the irradiation time, although the 1O2 generation ability was weaker than that of free PpIX and PEI-PpIX (con-taining equal amount of PpIX) owing to the irradiation light absorbed partly by the carrier CuS.[25] The photostability of free

Adv. Mater. 2019, 1904997

Page 3: Tumor‐Targeted Drug and CpG Delivery System for ... · In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced

© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim1904997 (3 of 9)

www.advmat.dewww.advancedsciencenews.com

PpIX, PEI-PpIX, and FA-CD@PP-CpG after laser irradiation was then investigated (Figure S7a, Supporting Information). Considering the optical adsorption of CuS in the NIR region, the photothermal response of FA-CD@PP-CpG was evaluated under 808 nm irradiation. In marked contrast to PBS and PEI-PpIX with negligible temperature variation, CuS and FA-CD@PP-CpG solutions showed considerable temperature rise of 35.8 and 34.4 °C, at a concentration of 100 µg mL−1, respec-tively (Figure 1h). The temperature rise of FA-CD@PP-CpG was found to be concentration- and irradiation-time-dependent (Figure 1i,j). Besides the excellent photothermal conversion ability, FA-CD@PP-CpG also exhibited high photostability and good reproducibility even after three laser on/off cycles (Figure 1k and Figure S7b, Supporting Information). There-fore, FA-CD@PP-CpG could serve as an efficient photothermal agent for potential application in cancer PTT.

Next, the cumulative release profiles of DTX in PBS at pH 7.4 (the physiological environment with neutral pH) and pH 5.0 (the acidic condition of endosome/lysosome in tumor cell) were investigated. FA-CD@PP-CpG nanocomposite at pH 7.4 and pH 5.0 both exhibited the apparent sustained release

behaviors for DTX, especially in the early stage with a rela-tively fast release (Figure 1l). Additionally, FA-CD@PP-CpG presented faster release at pH 5.0 compared with that at pH 7.4 condition for DTX, which might be due to the faster breakage of amide carbonyl between PEI and PpIX in the acidic condi-tion, confirmed by the change of Fourier transform infrared (FTIR) spectroscopy in Figure S8 (Supporting Information). Specifically, the accumulative release amount of DTX from the drug carrier reached 90.0% after being immersed at pH 5.0 condition for 48 h. So the FA-CD@PP-CpG nanocomposite possessed pH-responsive release behavior for DTX, resulting in the enhanced immunization.

A high biocompatibility and phagocytosis of nanomate-rials is required as a precondition for cancer applications. Compared with CuS nanoparticles, there was no obvious dif-ference in the viability (> 85% cell viability) on 4T1 cancer cells treated with FA-CuS@PP-CpG, whereas FA-CD@PP-CpG presented enhanced cytotoxicity than FA-CuS@PP-CpG at the high concentration ranges (Figure S9, Supporting Informa-tion, and Figure 2a), which is consistent that high dosage of chemotherapeutics can kill tumor cells but have little effects

Adv. Mater. 2019, 1904997

Scheme 1. Rational design and synthesis of FA-CD@PP-CpG nanocomposites (top), its application in cancer treatment (left), and illustration of FA-CD@PP-CpG for docetaxel-enhanced immunotherapy (right).

Page 4: Tumor‐Targeted Drug and CpG Delivery System for ... · In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced

© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim1904997 (4 of 9)

www.advmat.dewww.advancedsciencenews.com

at low concentration. And FA-CD@PP-CpG exhibited negli-gible toxicity toward HBL-100 normal cells within the range of concentrations tested (Figure S10, Supporting Information). Then the targeting efficiency of FA-CD@PP-CpG on 4T1 cells was evaluated by confocal laser scanning microscopy (CLSM) images. FA-CD@PP-CpG exhibited red fluorescence in the

cytoplasm of 4T1 cells after incubation for 4 h, while only weak red fluorescence could be observed in A549 lung cancer cells or in 4T1 cells treated with CD@PP-CpG without conju-gation of FA (Figure 2b and Figure S11, Supporting Informa-tion). Next, the cellular uptake efficiency of FA-CD@PP-CpG was assessed. Both flow cytometry results and CLSM images

Adv. Mater. 2019, 1904997

Figure 1. a–f) Representative SEM and TEM images of CuS (a,d), FA-CuS/DTX (b,e), and FA-CD@PP-CpG (c,f) dispersed in water. g) Decay curves of DPBF absorption at 410 nm in different solutions. h) Temperature increase curves of different solutions with the same amount of CuS or PpIX upon the laser irradiation. i) Temperature changes of FA-CD@PP-CpG at various concentrations under irradiation. j) Plot of the temperature elevation during 5 min versus the concentration of FA-CD@PP-CpG. k) Temperature change curves of FA-CD@PP-CpG (100 µg mL−1) over three laser irradiation on/off cycles. l) Release profiles of DTX from FA-CD@PP-CpG under different release medium.

Page 5: Tumor‐Targeted Drug and CpG Delivery System for ... · In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced

© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim1904997 (5 of 9)

www.advmat.dewww.advancedsciencenews.com

revealed that the red fluorescence intensity inside the cells increased remarkably with the prolonged time (Figure 2c,d and Figure S12, Supporting information). All these results dem-onstrated that FA-CD@PP-CpG with excellent biocompability could be internalized efficiently by 4T1 cells via FA-receptor mediated endocytosis.

We then investigated the generation of ROS in 4T1 cells treated with FA-CD@PP-CpG under 650 nm laser irradiation. 4T1 cells exhibited obvious green fluorescence, and the fluores-cence signals were strengthened with an increase in the con-centration of FA-CD@PP-CpG (Figure 2e). Next, we studied the combination therapy with different composites. Compared to PDT alone with CpG-free nanocomposites (FA-CD@PP), or PTT alone with FA-CD@PP, the combined treatment offered the most effective cancer cell killing. Moreover, the cell survival ratio of the group treated with FA-CD@PP-CpG for PDT, PTT, and immunotherapy exhibited negligible change compared to that of FA-CD@PP, which was due to the incapability of CpG to induce the apoptosis of 4T1 cells without invoking the antigen presenting cells[8] (Figure 2f). The antitumor efficiency in vitro was also assessed by live/dead viability (Figure 2g and Figure S13, Supporting Information). For the group treated with free CpG, no apparent dead cells could be observed because of nontoxicity of CpG. And there was significant cell death in the group subjected to dual laser irradiation com-pared with DTX, PDT, PTT, indicating the significant inhibi-tory effect of FA-CD@PP-CpG nanocomposite on tumor cells in vitro.

Afterward, photothermal imaging and fluorescence imaging in vivo was recorded to assess the feasibility of FA-CD@PP-CpG inside the tumor. All animal procedures conformed to the Guide for the Care and Use of Laboratory Animals and the ethical approval number was TJLAC-019-137. The temperature in the tumor site increased to 44 °C after irradiation for 10 min, which was high enough to kill tumor cells. Comparatively, the temperature of the tumor presented a feeble increase in the PBS group (Figure 3a,b). Weak fluorescence was observed in the tumor site after 6 h intravenous injection. And the fluo-rescence signal enhanced continuously with an increase in the housing time, reaching a maximum at 12 h. Subsequently, the local fluorescence began to fade from 12 to 24 h, suggesting the rapid clearance of FA-CD@PP-CpG in body. Moreover, the fluorescence observed in the livers and tumors, more signifi-cantly, no fluorescence observed in other tissues or organs fur-ther demonstrated FA-CD@PP-CpG highly targeted on tumors (Figure 3c–e). Importantly, few uptake of FA-CD@PP-CpG by macrophages compared with 4T1 cells suggested that the accumulation in liver was not due to the uptake by the retic-uloendothelial system, but the temporary retention because of the liver’s unique anatomical and physiological structure. (Figure S14, Supporting Information).

Next, we carried out the combination therapy in vivo using FA-CD@PP-CpG on 4T1-tumor-bearing mice. Compared with the rapid increase of tumor in PBS group, there was negligible antitumor activity in the group treated with CpG and aPD-L1 as well as group injected with free DTX. Importantly, the

Adv. Mater. 2019, 1904997

Figure 2. a) Viability of 4T1 cells treated with FA-CuS@PP-CpG and FA-CD@PP-CpG without laser irradiation. b) CLSM images of 4T1 cells and A549 cells treated with FA-CD@PP-CpG for 4 h (left) and corresponding quantitative fluorescence curves (right). c) Flow cytometric assay and d) CLSM images of 4T1 cells incubated with FA-CD@PP-CpG for different time periods. 2-(4-amidinophenyl)-6-indolecarbamidine dihydrochloride (DAPI) for nuclei staining (blue) and PpIX fluorescence (red) were recorded. e) Intracellular ROS detection in 4T1 cells incubated with various concentrations of FA-CD@PP-CpG under 650 nm irradiation. f) Viabilities and g) corresponding fluorescence images of 4T1 cells constrained with calcein AM (live cells, green) and propidium iodide (dead cells, red) after being treated with different conditions. Scale bar: 100 µm. The error bars are based on the SD of three wells.

Page 6: Tumor‐Targeted Drug and CpG Delivery System for ... · In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced

© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim1904997 (6 of 9)

www.advmat.dewww.advancedsciencenews.com

group treated with FA-CD@PP-CpG + aPD-L1 + PDT + PTT (abbreviated as aPD-L1 + PDT + PTT) exhibited the remark-able tumor growth inhibition effect compared to various con-trol groups (Figure 3f–h). Survival rate of different groups fur-ther demonstrated that aPD-L1 + PDT + PTT group presented the best antitumor efficacy (Figure 3i). Hematoxylin and eosin (H&E) staining revealed that tumor tissues in the aPD-L1 + PDT + PTT group were severely damaged in comparison

with that of PBS group (Figure S15, Supporting Information). Simultaneously, all mice in the seven groups displayed no significant body weights difference, indicating negligible sys-temic side effect of FA-CD@PP-CpG (Figure 3j), which was also confirmed by images of H&E stained major organs (heart, liver, spleen, lung, and kidney) and the corresponding blinded scores of major tissues (Figure S15 and Table S1, Supporting Information).

Adv. Mater. 2019, 1904997

Figure 3. a) The in vivo thermal images of the mice after intravenous injection of PBS and FA-CD@PP-CpG under 808 nm irradiation. b) Temperature change curve of tumor sites as a function of irradiation time. c) Fluorescence images of the mice after intravenous injection of FA-CD@PP-CpG and d) ex vivo imaging of tumor and major organs after 24 h postinjection. e) Quantitative fluorescence curve of tumor tissues. f) Tumor growth curves in different groups. g) The weight and h) representative photographs of tumor tissue in different groups obtained on day 14. i) The survival rate and j) the body weight changes in different groups. k–m) Physiological function assessment of liver and kidney toxicity from healthy, control, and aPD-L1 + PDT + PTT treated mice. The error bars are based on the SD of five mice. *p < 0.05, **p < 0.01, and ***p < 0.01.

Page 7: Tumor‐Targeted Drug and CpG Delivery System for ... · In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced

© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim1904997 (7 of 9)

www.advmat.dewww.advancedsciencenews.com

Adv. Mater. 2019, 1904997

In addition, all blood biochemical values, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin (ALB), blood urea nitrogen (BUN), creatinine (CREA), and uric acid (UA), were not statistically different between mice treated with aPD-L1 + PDT + PTT and healthy mice, which suggested that no hepatic dysfunctions and abnormal renal functions were caused by FA-CD@PP-CpG (Figure 3k–m and Figure S16, Supporting Information).

We then investigated the specificity of FA-CD@PP-CpG on the CTLs generation. A ratio of 4.8:1 carboxyfluorescein succin-imidyl ester (CFSE) losing on FA-CD@PP-CpG versus control group showed an average > fivefold multiplication (5 genera-tions), higher than CpG + aPD-L1 (4 generations) and DTX (3.5 generations) (Figure 4a). To further explore the differentiation of MDSCs, we studied the presence of M1 (CCR7) or M2 (MR) markers on MDSCs after different treatments. Figure 4b showed that the level of CCR7 was upregulated and correspondingly, the

level of MR was downregulated significantly by aPD-L1 + PDT + PTT compared with DTX group, which revealed the polarization of MDSCs population to an M1-like phenotype via DTX trans-ported by nanocomposite. Subsequently, the result of cytokine secretion in MDSCs supernatants showed that MDSCs treated with aPD-L1 + PDT + PTT had significant upregulation of IL-12 but downregulation of IL-10 production compared with other three groups (Figure 4c,d). Next, we acquired various macrophage cell surface markers of different groups (CD40, CD80, CD86, MHC class II, and CD11c) (Figure 4e). For aPD-L1 + PDT + PTT group, MDSCs were found to express highest levels of all above mentioned cell surface markers compared with CpG + aPD-L1 treated, DTX-treated and the control group. The results showed that DTX transported by FA-CD@PP-CpG could polarize MDSCs toward an M1-like phenotype by expressing high levels of CCR7, increasing the generation of IL-12 and further kill cancer cells effectively.

Figure 4. a) The proliferation of CD3 + CD8 + T cells assessed by CFSE dilution, with unstimulated CD3 + CD8 + T cells as the control (gray shade) after 48 h incubation (left) and quantitative fluorescence curve of T cells (right). b) Histogram representation of CCR7 and MR levels in DTX and aPD-L1 + PDT + PTT treated tumors. c,d) Cytokine levels of IL-12 (c) and IL-10 (d) in MDSC supernatant from mice. e) Flow cytometric assay of CD40, CD80, CD86, MHC class II, and CD11c expression from CpG + aPD-L1, DTX, and aPD-L1 + PDT + PTT treated tumors. p values:*p < 0.05, **p < 0.01, and ***p < 0.01.

Page 8: Tumor‐Targeted Drug and CpG Delivery System for ... · In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced

© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim1904997 (8 of 9)

www.advmat.dewww.advancedsciencenews.com

Adv. Mater. 2019, 1904997

The significant inhibition of tumors in the group treated with aPD-L1 + PDT + PTT may contribute to the increased infiltration of CTLs in tumor sites. Flow cytometry result revealed that group treated with CpG +aPD-L1 or DTX had an improved level on T-cell infiltration compared with PBS group with limited T-cell infiltration. Comparatively, tumors treated with aPD-L1 + PDT + PTT were remarkably infiltrated by CTLs (Figure 5a,b). Additionally, for aPD-L1 + PDT + PTT group, tumor presented a highest level of TNF-α, IL-12 and IFN-γ because of the synergetic effect of CpG and DTX compared to other control groups (Figure 5c–e). The same outcomes of CTLs infiltration in tumor site were observed through immu-nofluorescence (Figure 5f and Figure S17, Supporting informa-tion). Tumors from aPD-L1 + PDT + PTT treated mice were remarkably infiltrated by both CD4+ and CD8+ T cells, and the percentage of CD8+ T cells was threefold of that in the CpG +  aPD-L1 group, twofold of that in the DTX group and 1.5-fold of that in the other groups (Figure 5g).

In summary, we report herein the tumor-targeted DTX and CpG delivery system as a smart platform for cancer PDT, PTT, and docetaxel-enhanced-immunotherapy in 4T1 breast cancer

cells bearing mice. FA-CD@PP-CpG nanocomposite presented negligible toxicity to normal tissues, but could cause remarkable damage to tumors in vivo when combined with aPD-L1 and dual laser irradiation (650 and 808 nm). Combination of FA-CD@PP-CpG with aPD-L1 enhanced the infiltration of CTLs, which was favorable of aPD-L1 to block the PD-1/PD-L1 checkpoint-blockade and further prevent CTLs from dysfunction and exhaustion, thus kill cancer cells efficiently. Furthermore, DTX transported by FA-CD@PP-CpG to tumor cells could directly induce an phenotype polarization of MDSCs contributes to more generation of proinflammatory cytokines, thus resulting in enhanced immunotherapy efficacy. Taken together, our study presented a novel strategy for highly superior antitumor efficiency by combining PDT, PTT, and docetaxel-enhanced immunotherapy, which might open new avenues in breast cancer immunotherapy for clinical translation.

Supporting InformationSupporting Information is available from the Wiley Online Library or from the author.

Figure 5. a) Flow cytometric data and b) corresponding proportion of CTLs’ infiltration in tumors. c–e) Cytokine levels of TNF-α (c), IL-12 (d), and IFN-γ (e) in serum from mice. f) Representative immunofluorescence images of tumor tissues from PBS and aPD-L1 + PDT + PTT group. Blue, DAPI-labeled nuclei; green, anti-CD4+ antibody-labeled T cells; red, anti-CD8+ antibody-labeled T cells. Scale bar:100 µm. g) Proportion of tumor-infiltrating CD8 + T cells according to (f) and Figure S16 (Supporting Information). The error bars are based on the SD of five mice. p values: *p < 0.05, **p < 0.01, and ***p < 0.01.

Page 9: Tumor‐Targeted Drug and CpG Delivery System for ... · In this work, we constructed a tumor-targeted drug and CpG delivery system for near-infrared induced phototherapy and doc-etaxel-enhanced

© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim1904997 (9 of 9)

www.advmat.dewww.advancedsciencenews.com

Adv. Mater. 2019, 1904997

AcknowledgementsL.C. and L.Z. contributed equally to this work. This work was supported by the Fundamental Research Funds for the Central Universities, Key Cross-cutting Projects of Central Universities (1507219075), the Project of Key Disciplines Group Construction of Pudong Health Bureau of Shanghai (PWZxq2017D-10), the National Natural Science Foundation of China (Nos. 21877084, 21671150, 8186110475, 81573008, and 81860547), Science and Technology Commission of Shanghai Municipality (No.14DZ2261100), and Project of Distinguished Young Physicians of Shanghai Municipality (jcqnrc201901). Thanks for the help by associate professor Xiaojuan Xu (Tongji University School of Medicine, Department of Pathology) in the blinded scores of all organs. All animal procedures conformed to the Guide for the Care and Use of Laboratory Animals. The 4T1 murine breast cancer cell line was obtained from the Cell Library at Tongji University (Shanghai, China). These cells were cultured from cells originally purchased from Chinese Academy of Sciences Cell Library, ATCC number ATCC CRL-2539 (CRL-2539 and ATCC are trademarks of American Type Culture Collection).

Conflict of InterestThe authors declare no conflict of interest.

KeywordsCpG, docetaxel, immunotherapy, PD-L1/anti PD-L1, phototherapy

Received: August 2, 2019Revised: October 4, 2019

Published online:

[1] a) F. Bray, J. Ferlay, I. Soerjomataram, R. L. Siegel, L. A. Torre, A. Jemal, Ca-Cancer J. Clin. 2018, 68, 394; b) W. Chen, R. Zheng, P. D. Baade, S. Zhang, H. Zeng, F. Bray, A. Jemal, X. Q. Yu, J. He, Ca-Cancer J. Clin. 2016, 66, 115.

[2] a) Y. Tian, X. Jiang, X. Chen, Z. Shao, W. Yang, Adv. Mater. 2014, 26, 7393; b) S. Mura, J. Nicolas, P. Couvreur, Nat. Mater. 2013, 12, 991; c) A. R. Kirtane, S. M. Kalscheuer, J. Panyam, Adv. Drug Delivery Rev. 2013, 65, 1731.

[3] H. Ledford, Nature 2015, 526, 622.[4] B. Fleisher, S. Ait-Oudhia, OncoTargets Ther. 2018, 11, 113.[5] H. O. Alsaab, S. Sau, R. Alzhrani, K. Tatiparti, K. Bhise,

S. K. Kashaw, A. K. Iyer, Front. Pharmacol. 2017, 8, 561.[6] a) X. P. Duan, C. Chan, W. B. Lin, Angew. Chem., Int. Ed. 2019, 58,

670; b) K. D. Lu, C. B. He, N. N. Guo, C. Chan, K. Y. Ni, G. X. Lan, H. D. Tang, C. Pelizzari, Y. X. Fu, M. T. Spiotto, R. R. Weichselbaum, W. B. Lin, Nat. Biomed. Eng. 2018, 2, 600; c) M. E. Marmarelis, M. R. Davis, N. S. Sethi, K. M. Krajewksi, R. R. McKay, T. K. Choueiri, P. A. Ott, J. ImmunoTher. Cancer 2016, 4, 26.

[7] a) A. M. Krieg, Oncogene 2008, 27, 161; b) J. Vollmer, A. M. Krieg, Adv. Drug Delivery Rev. 2009, 61, 195; c) K. Wang, S. M. Wen, L. H. He, A. Li, Y. Li, H. Q. Dong, W. Li, T. B. Ren, D. L. Shi, Y. Y. Li, ACS Nano 2018, 12, 6398; d) A. V. Kroll, R. H. Fang, Y. Jiang, J. Zhou, X. Wei, C. L. Yu, J. Gao, B. T. Luk, D. Dehaini, W. Gao, L. Zhang, Adv. Mater. 2017, 29, 1703969.

[8] J. C. Yang, Y. Shang, Y. H. Li, Y. Cui, X. B. Yin, Chem. Sci. 2018, 9, 7210.[9] a) R. J. Sullivan, K. T. Flaherty, Nat. Rev. Clin. Oncol. 2015, 21, 2424;

b) W. P. Zou, J. D. Wolchok, L. P. Chen, Sci. Transl. Med. 2016, 8, 328rv4.

[10] a) M. Reck, D. Rodriguez-Abreu, A. G. Robinson, R. Hui, T. Csoszi, A. Fulop, M. Gottfried, N. Peled, A. Tafreshi, S. Cuffe, M. O’Brien,

S. Rao, K. Hotta, M. A. Leiby, G. M. Lubiniecki, Y. Shentu, R. Rangwala, J. R. Brahmer, K. Investigators, N. Engl. J. Med. 2016, 375, 1823; b) A. Rittmeyer, F. Barlesi, D. Waterkamp, Lancet 2017, 389, 255.

[11] T. J. Daskivich, A. Belldegrun, Eur. Urol. 2015, 67, 816.[12] a) S. D. Meng, L. Li, M. L. Zhou, W. J. Jiang, H. Niu, K. X. Yang,

Mol. Med. Rep. 2018, 18, 4247; b) H. R. Ali, E. Provenzano, S. J. Dawson, F. M. Blows, B. Liu, M. Shah, H. M. Earl, C. J. Poole, L. Hiller, J. A. Dunn, S. J. Bowden, C. Twelves, J. M. S. Bartlett, S. M. A. Mahmoud, E. Rakha, I. O. Ellis, S. Liu, D. Gao, Tn. Nielsen, P. D. P. Pharoah, C. Caldas, Ann. Oncol. 2014, 25, 1536. c) J. Stagg, B. Allard, Ther. Adv. Med. Oncol. 2013, 5, 169.

[13] F. Hossain, A. A. Al-Khami, D. Wyczechowska, C. Hernandez, L. Q. Zheng, K. Reiss, L. Del Valle, J. Trillo-Tinoco, T. Maj, W. P. Zou, P. C. Rodriguez, A. C. Ochoa, Cancer Immunol. Res. 2015, 3, 1236.

[14] E. Safari, S. Ghorghanlu, H. Ahmadi-khiavi, S. Mehranfar, R. Rezaei, M. Motallebnezhad, J. Cell. Physiol. 2019, 234, 9966.

[15] N. Umemura, M. Saio, T. Suwa, Y. Kitoh, J. Bai, K. Nonaka, G. F. Ouyang, M. Okada, M. Balazs, R. Adany, T. Shibata, T. Takami, J. Leukocyte Biol. 2008, 83, 1136.

[16] a) A. Mantovani, A. Sica, S. Sozzani, P. Allavena, A. Vecchi, M. Locati, Trends Immunol. 2004, 25, 677; b) A. Mantovani, S. Sozzani, M. Locati, P. Allavena, A. Sica, Trends Immunol. 2002, 23, 549.

[17] a) T. J. Haggerty, I. S. Dunn, L. B. Rose, E. E. Newton, S. Martin, J. L. Riley, J. T. Kurnick, Cancer Immunol. Immunother. 2011, 60, 133; b) K. N. Kodumudi, K. Woan, D. L. Gilvary, E. Sahakian, S. Wei, J. Y. Djeu, Clin. Cancer Res. 2010, 16, 4583; c) W. J. Lesterhuis, C. J. A. Punt, S. V. Hato, D. Eleveld-Trancikova, B. J. H. Jansen, S. Nierkens, G. Schreibelt, A. de Boer, C. M. L. Van Herpen, J. H. Kaanders, J. H. J. M. van Krieken, G. J. Adema, C. G. Figdor, I. J. M. de Vries, J. Clin. Invest. 2011, 121, 3100; d) W. M. Liu, D. W. Fowler, P. Smith, A. G. Dalgleish, Br. J. Cancer 2010, 102, 115; e) B. L. Mundy-Bosse, G. B. Lesinski, A. C. Jaime-Ramirez, K. Benninger, M. Khan, P. Kuppusamy, K. Guenterberg, S. V. Kondadasula, A. R. Chaudhury, K. M. La Perle, M. Kreiner, G. Young, D. C. Guttridge, W. E. Carson, Cancer Res. 2011, 71, 5101; f) A. K. Nowak, R. A. Lake, A. L. Marzo, B. Scott, W. R. Heath, E. J. Collins, J. A. Frelinger, B. W. S. Robinson, J. Immunol. 2003, 170, 4905; g) A. K. Nowak, B. W. S. Robinson, R. A. Lake, Cancer Res. 2003, 63, 4490; h) D. Z. Qian, B. L. S. Rademacher, J. Pittsenbarger, C. Y. Huang, A. Myrthue, C. S. Higano, M. Garzotto, P. S. Nelson, T. M. Beer, Prostate 2010, 70, 433; i) J. Vincent, G. Mignot, F. Chalmin, S. Ladoire, M. Bruchard, A. Chevriaux, F. Martin, L. Apetoh, C. Rebe, F. Ghiringhelli, Cancer Res. 2010, 70, 3052.

[18] a) A. Sica, A. Mantovani, J. Clin. Invest. 2012, 122, 787; b) A. Sevko, T. Michels, M. Vrohlings, L. Umansky, P. Beckhove, M. Kato, G. V. Shurin, M. R. Shurin, V. Umansky, J. Immunol. 2013, 190, 2464; c) C. R. Millrud, M. Mehmeti, K. Leandersson, Exp. Cell Res. 2018, 362, 525.

[19] S. S. Lucky, K. C. Soo, Y. Zhang, Chem. Rev. 2015, 115, 1990.[20] J. Liu, X. P. Zheng, L. Yan, L. J. Zhou, G. Tian, W. Y. Yin, L. M. Wang,

Y. Liu, Z. B. Hu, Z. J. Gu, C. Y. Chen, Y. L. Zhao, ACS Nano 2015, 9, 696.[21] a) W. Guo, Z. Y. Qiu, C. S. Guo, D. D. Ding, T. C. Li, F. Wang,

J. Z. Sun, N. N. Zheng, S. Q. Liu, ACS Appl. Mater. Interfaces 2017, 9, 9348; b) G. Feng, Y. Fang, J. Liu, J. Geng, D. Ding, B. Liu, Small 2017, 13, 1602807.

[22] a) H. Shi, R. Yan, L. Wu, Y. Sun, S. Liu, Z. Zhou, J. He, D. Ye, Acta Biomater. 2018, 72, 256; b) S. Liang, Z. X. Xie, Y. Wei, Z. Y. Cheng, Y. Q. Han, J. Lin, Dalton Trans. 2018, 47, 7916.

[23] W. Jiang, H. Zhang, J. Wu, G. Zhai, Z. Li, Y. Luan, S. Garg, ACS Appl. Mater. Interfaces 2018, 10, 34513.

[24] H. Y. Zhang, W. Jiang, R. L. Liu, J. Zhang, D. Zhang, Z. H. Li, Y. X. Luan, ACS Appl. Mater. Interfaces 2017, 9, 19687.

[25] Z. Zhao, S. Shi, Y. Huang, S. Tang, X. Chen, ACS Appl. Mater. Interfaces 2014, 6, 8878.