CT imaging-guided multimodal therapy of cancer

CT imaging-guided multimodal therapy of cancer

Accepted Manuscript PEGylated mesoporous Bi2S3 Nanostars loaded with Chlorin e6 and doxorubicin for fluorescence/CT imaging-guided multimodal therapy ...

12MB Sizes 0 Downloads 17 Views

Accepted Manuscript PEGylated mesoporous Bi2S3 Nanostars loaded with Chlorin e6 and doxorubicin for fluorescence/CT imaging-guided multimodal therapy of Cancer

Lihong Sun, Mengmeng Hou, Lei Zhang, Dongxiang Qian, Qingquan Yang, Zhigang Xu, Yuejun Kang, Peng Xue PII: DOI: Reference:

S1549-9634(19)30006-1 https://doi.org/10.1016/j.nano.2018.12.013 NANO 1927

To appear in:

Nanomedicine: Nanotechnology, Biology, and Medicine

Revised date:

20 December 2018

Please cite this article as: Lihong Sun, Mengmeng Hou, Lei Zhang, Dongxiang Qian, Qingquan Yang, Zhigang Xu, Yuejun Kang, Peng Xue , PEGylated mesoporous Bi2S3 Nanostars loaded with Chlorin e6 and doxorubicin for fluorescence/CT imaging-guided multimodal therapy of Cancer. Nano (2019), https://doi.org/10.1016/j.nano.2018.12.013

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT PEGylated Mesoporous Bi2S3 Nanostars Loaded with Chlorin e6 and Doxorubicin for Fluorescence/CT Imaging-Guided Multimodal Therapy of Cancer

T

Lihong Suna, b, Mengmeng Houa, b, Lei Zhangc, Dongxiang Qiand, Qingquan Yange,

Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest

CR

a

IP

Zhigang Xua, b, Yuejun Kanga, b*, Peng Xuea, b*

University), Ministry of Education, Faculty of Materials and Energy, Southwest

Chongqing Engineering Research Center for Micro-

AN

b

US

University, Chongqing 400715, China.

Nano Biomedical Materials and Devices, Chongqing 400715, China. Institute of Sericulture and System Biology, Southwest University, Chongqing 400716,

M

c

d

ED

China.

The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150,

The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110002, China.

CE

e

PT

China.

Corresponding authors:

AC

[email protected] (P. Xue), phone: +86-23-68253792 [email protected] (Y. Kang), phone: +86-23-68254056 Abstract: 142 words Manuscript: 4996 words References: 52 references Number of figures: 8

1

ACCEPTED MANUSCRIPT Funding: This study is supported by Technology Innovation and Application Demonstration Grant of Chongqing (cstc2018jscx-msybX0078), financial support from Fundamental Research Funds for Central Universities (XDJK2017C001, XDJK2016A010), and National Natural

IP

T

Science Foundation of China (51703186, 31671037).

CR

Conflict of interest:

AC

CE

PT

ED

M

AN

US

There is no conflict of interest.

2

ACCEPTED MANUSCRIPT Abstract Taking advantage of the mesoporous structure of bismuth sulfide nanostars (Bi2S3 NSs), a chemotherapeutic drug of doxorubicin (DOX) and a photosensitizer of chlorin e6 (Ce6) were concurrently loaded in the PEGylated Bi2S3 NSs to formulate a multifunctional

T

nanocomplex (BPDC NSs) for tumor theranostics. BPDC NSs have excellent

IP

photothermal conversion efficiency and a capacity of yielding reactive oxygen species

CR

(ROS) upon laser irradiation, and can realize on-demand drug release by either pH-

US

activation or thermal induction. Accumulation of the nanodrug could be monitored in real-time by infrared thermal imaging, fluorescence imaging and computed tomography

AN

(CT). More importantly, the combination effects of photothermal therapy (PTT), photodynamic therapy (PDT) and chemotherapy was demonstrated to dramatically

M

suppress solid tumors without recurrence in vivo. Featured by the low systemic toxicity

ED

and high biocompatibility, this nanoplatform could be a promising derivative of Bi2S3

AC

Keywords

CE

PT

NSs for imaging-guided theranostics of cancer.

Bismuth (Bi) chalcogenides; Doxorubicin; Chlorin e6; Controlled drug release; Cancer theranostics.

3

ACCEPTED MANUSCRIPT Background Imaging-guided therapeutics, providing precise and personalized intervention in cancer therapy, has attracted considerable interests of researchers searching for efficient strategies against malignant tumors

1, 2

. The integration of imaging capability with

T

various therapeutics not only provides the exact location of tumors for localized treatment

IP

with minimal impact on normal tissues, but also allows real-time monitoring the

CR

biodistribution and physiological effects of therapeutic agents

3-9

. Although simply

US

combining multiple agents may compensate the intrinsic limitations of each individual treatment modality, there could be serious issues associated with their stability,

applications

10, 11

AN

compatibility and potential side effects, which may seriously constrain their clinical . Therefore, development of a single agent with multifunctional

M

treatment modality becomes particularly necessary for efficacious tumor theranostics.

ED

In the past decade, photo-triggered tumor therapy becomes popular in the 12

. Among

PT

development of “smart” nanoplatforms to achieve unique clinical benefits

these phototherapeutics, near-infrared (NIR) light-responsive agents provide distinct

CE

ability in converting light energy into local hyperthermia for photothermal therapy (PTT)

AC

of cancer13-15. Recently, bismuth (Bi) chalcogenides have become increasingly prevalent not only as topological insulators but also as NIR-sensitive photothermal agents for biomedical applications 16-20. As a powerful clinical diagnostics, CT is featured with high spatiotemporal resolution and non-invasiveness

21, 22

. However, traditional CT suffers

from the limitations of poor contrast in soft tissues and ionizing radiation, as well as the pharmacokinetic problems of commercial CT contrast agents

23

. Usually, CT contrast

agents with higher atomic number (Z) may contribute to sharper image contrast at an

4

ACCEPTED MANUSCRIPT equivalent mass concentration

24

. Given a typical high Z-element, Bi elementary

substance and its compounds with a corresponding high X-ray attenuation coefficient are promising candidates as CT contrast agents for biomedical applications

25

.Particularly,

bismuth sulfide (Bi2S3)-based nanostructures are a very appealing class of materials 26-28

. In

T

because of their biocompatibility, long blood circulation and cost effectiveness

28

. Nevertheless, current Bi2S3-based biomedical materials are mainly

CR

and drug delivery

IP

addition, highly porous Bi2S3 nanostructures have demonstrated their capability for PTT

based on solitary therapeutic modalities, which is usually not sufficient for efficacious

US

tumor eradication.

AN

Photodynamic therapy (PDT), as a minimally-invasive therapeutic modality for tumor ablation, induces cellular damage in tumoral tissues by producing cytotoxic 29-32

M

reactive oxygen species (ROS), e.g., singlet oxygen

. As one of the most prevalently

ED

used photosensitizers, chlorin e6 (Ce6) has remarkable quantum yield of singlet oxygen and NIR fluorescence emission for PDT

33, 34

. However, Ce6 molecules are usually

PT

unstable and prone to aggregating in an aqueous environment, resulting in a short half-

CE

life of circulation due to rapid degradation 35. Furthermore, Ce6 aggregates in the form of large crystals also hinder the endocytotic cellular uptake

36

. Alternatively, Ce6 can also

AC

be delivered into cancerous tissues and tumor cells intracellularly facilitated by a variety of nanocarriers

37-39

. More interestingly, Ce6 molecules can be activated specifically

within tumor region in the presence of either exogenous or endogenous stimuli, significantly reducing adverse systemic side effects 40, 41. Based on the unique characteristics of these materials above, a class of PEGylated Bi2S3 nanostars (NSs) were designed and synthesized in this study, aiming to coordinate

5

ACCEPTED MANUSCRIPT both diagnostic and therapeutic functionalities by encapsulating a chemotherapeutic drug of doxorubicin (DOX) and a photosensitizer of chlorin e6 (Ce6) (Figure 1). Basically, the Bi2S3 NSs were derived from Bi2O3 nanospheres as a precursor and template after a typical hydrothermal process. Then, the surface of Bi2S3 NSs were functionalized with

T

polyethylene glycol (PEG) to improve the biocompatibility and physiological stability of

IP

the drug carrier. The resultant Bi2S3@PEG NSs exhibited a highly porous structure for

CR

loading DOX and Ce6 to form Bi2S3@PEG/DOX/Ce6 (BPDC) NSs. After intravenous injection, as-synthesized nanodrugs accumulated in tumor region because of the

US

enhanced permeability and retention (EPR) effect. Subsequently, DOX release was

AN

accelerated in more acidic tumor microenvironment. 808 nm NIR light is the mostly used for inducing local hyperthermia mediated by photothermal agents, owing to its good

M

tissue penetration depth and the least absorption by water and hemoglobin13,

15

.

rendering PDT

33, 34

ED

Meanwhile, Ce6 can be activated by a 660 nm laser to produce singlet oxygen (1O2) for . By combining dual laser irradiation (808 nm and 660 nm), PTT-

PT

induced hyperthermia and PDT-induced ROS generation worked concurrently for

CE

ablation of tumorous tissue. Meanwhile, DOX release was further promoted under elevated local temperature. Furthermore, BPDC NSs were able to couple fluorescence/CT

AC

dual-modal imaging for cancer diagnosis, providing more precise information of the lesion and thus guiding the therapeutics.

Methods Preparation of BPDC NSs

6

ACCEPTED MANUSCRIPT The BPDC NSs were synthesized following a one-step nanoprecipitation reaction method. Firstly, 10 mg of Bi2S3@PEG NSs were dispersed into 18 mL of 1×PBS (pH = 7.4). Afterwards, 10 mg of Ce6 and 2 mL of DOX·HCl (5 mg·mL-1) were added into the above dispersion under stirring, followed by introducing 80 µL of NaOH solution (1

T

mol·L-1). After further stirring for 24 h at room temperature in darkness, precipitates

IP

obtained through centrifugation were washed with DI water to remove free DOX and Ce6,

CR

and BPDC NSs were finally synthesized. To calculate the loading content and encapsulation efficiency of DOX and Ce6, 0.5 mg of BPDC NSs were dissolved in 10

US

mL DMSO under sonication. After centrifugation, the supernatant was collected to

AN

determine the amount of encapsulated DOX and Ce6 using the corresponding standard calibration curve measured from their fluorescence spectra. To assess the DOX release in

M

vitro, 2 mg of BPDC NSs dispersed in 1 mL of 1×PBS was introduced into a dialysis bag,

ED

which was further submerged into 80 mL of saline buffer (pH=7.4 or 5.0) as the releasing medium at 37oC. At predesignated time points, 1 mL of releasing medium was withdrawn

PT

from the system and replaced by fresh saline buffer of the same volume. Then, the DOX

CE

and Ce6 release kinetics was determined using a calibration curve based on fluorescence

AC

spectrophotometry (Ex/Em: 485/535 nm for DOX, Ex/Em: 405/660 nm for Ce6).

Results

Synthesis and characterizations of BPDC NSs The procedures of fabricating BPDC NSs is illustrated in Figure 1. As revealed by the SEM and TEM imaging, the Bi2O3 NPs exhibited a spherical mesoporous structure, and their hydrodynamic size was measured as ~186.2 nm using DLS (Figure S1, S2).

7

ACCEPTED MANUSCRIPT Originated from Bi2O3 NPs, Bi2S3 nanostructure showed a distinct star-like morphology with a hydrodynamic diameter of ~257.8 nm (Figure 2a, b). The TEM image of BPDCs with DOX and Ce6 encapsulation exhibited a well-dispersed star-like nanostructure with a hydrodynamic size of 272.8 nm (Figure S3). For a comparative study, the

T

hydrodynamic size and polydispersity index (PDI) of all the intermediate products and

IP

BPDC NSs measured by DLS were listed in Table S1. The zeta potentials of Bi2S3@PEG

CR

NSs and BPDC NSs were measured as -29.7 mV and -14.1 mV, respectively (Figure 2c), and the negative surface charge may benefit their long circulation half-lives in blood

42

.

US

Furthermore, the elemental composition and chemical valence of Bi2O3 NPs and

AN

Bi2S3 NSs were investigated using X-ray diffraction (XRD) and X-ray photoelectron spectroscopy (XPS) analysis. All the diffraction peaks of Bi2O3 NPs were ascribed to a

M

typical crystal form (JCPDS 898964) as shown in Figure S4a. The XPS survey spectrum

ED

of Bi2O3 NPs implied the co-existence of Bi and O elements, which was consistent with their chemical composition (Figure S4b-d). For Bi2S3 NSs, the full scan survey spectrum

PT

revealed that the sample was primarily composed of Bi, S, O and C elements (Figure 2d).

CE

The peak of O1s could be attributed to the absorbed oxygen species present on the particle surface, which is commonly observed in the sample exposed to the atmosphere. The peak

AC

representing C element was attributed to the substrate carbon film required for XPS measurement. Particularly, two strong peaks at ~157.6 and ~162.9 eV were corresponding to Bi4f7/2 and Bi4f5/2, respectively (Figure 2e)

43

. The binding energy

locating at ~224.8 eV could be assigned to S2s (Figure 2f) 44. The crystallization phase of as-prepared Bi2S3 NSs was further explored by XRD. The pattern of Bi2S3 NSs showed a typical amorphous phase and all the diffraction peaks could be completely indexed to

8

ACCEPTED MANUSCRIPT orthorhombic Bi2S3 (JCPDS 898964), indicating the high purity of Bi2S3 NSs (Figure 2g). Moreover, the porosity of Bi2S3 NPs was calculated based on nitrogen adsorption– desorption isotherms using Brunauer–Emmett–Teller (BET) analysis (Figure 2h, i). The BET surface area and total pore volume of Bi2S3 NSs were calculated as 28.493 m2·g−1

T

and 0.149 cm3·g−1, respectively. Furthermore, the pore size distribution of Bi2S3 NSs

IP

indicated an average pore size of 6 nm.

CR

The UV-vis-NIR absorbance spectrum of Bi2S3 NSs, DOX, Ce6 and BPDC NSs

US

were investigated in the range of 300-900 nm (Figure 3a). Strong optical absorbance of BPDC NSs was observed in the near infrared range of 700-900 nm, indicating a

AN

prominent potential as a PTT agent. Although there was a strong and wide absorbance

M

peak attributed to the Bi2S3@PEG component, the characteristic peaks at 488 nm and 400/660 nm corresponding to DOX and Ce6, respectively, were still discernible from the

ED

absorbance spectrum of BPDC NSs particularly under higher concentrations (Figure 3b),

PT

implying the successful encapsulation of these two therapeutic molecules. Meanwhile, the absorbance intensity of BPDC NSs increased proportionally with NS concentration,

CE

suggesting their good dispersity in aqueous condition (Figure 3b, S5a). Furthermore, no

AC

obvious aggregation of BPDC NSs was observed in the dispersions in DI water, 1×PBS or DMEM (with 10% FBS), showing good aqueous stability (Figure S5b). Next, the hydrodynamic size of BPDC NSs dispersed in above media was continuously monitored for two weeks. There was no considerable size change or observable aggregation of BPDC NSs in any of these three solution systems over 14 days (Figure S6). Therefore, these results indicated the long-term stability of BPDC NSs in aqueous conditions. The encapsulation efficiency (EE) for DOX and Ce6 were calculated as 15.3% and 9.5%, and

9

ACCEPTED MANUSCRIPT the drug loading content (DLC) for DOX and Ce6 were determined as 13.3% and 8.7%, respectively.

Photothermal effect of BPDC NSs in vitro

T

The photothermal response of BPDC NSs was evaluated by exposing sample-laden

IP

quartz cuvettes to a NIR laser (808 nm, 2 W·cm-2). Compared to DI water, remarkable

CR

temperature elevation over time was observed in the dispersions of both Bi2S3 NSs and

US

BPDC NSs under laser irradiation, suggesting a strong photo-induced hyperthermia effect (Figure 3c). Additionally, the temperature increase of BPDC NP suspension exhibited a

AN

typical concentration- and irradiation power-dependent profile (Figure 3d, e, g and Figure

M

S7). The photothermal response of BPDC NS dispersions was also monitored in real-time by infrared imaging (Figure 3g), which revealed similar results as recorded by digital

ED

thermometer. To assess the photothermal stability of BPDC NSs, the dispersions were

PT

subject to periodic laser irradiation and cooling process for four cycles (Figure 3f)45. In

CE

vitro drug release results are provided in Supplementary data (Figure S8).

AC

Cellular uptake of BPDC NSs Intracellular internalization of BPDC NSs was investigated using HeLa cells (a human cervical cancer cell line) and 4T1 cells (a murine mammary carcinoma cell line). Confocal laser scanning microscopy (CLSM) was used to visualize the cellular uptake of NPs. As shown in the confocal images, the fluorescence signals of both Ce6 and DOX intensified over the time of incubation, indicating effective internalization of BPDC NSs by HeLa cells (Figure 4a, S9). Particularly, Ce6 signal was mainly located in the

10

ACCEPTED MANUSCRIPT cytoplasm, while DOX signal was both observed in the nuclei and cytoplasm. In addition, flow cytometry analysis was applied to quantify the cellular uptake efficiency of BPDC NSs. The fluorescence intensity of single cell emission measured in flow analysis was corresponding to the amount of NSs internalized in each cell. The peak and mean

T

fluorescence intensity shifted to higher levels after prolonged incubation time (Figure 4b,

IP

c), suggesting the increased cellular uptake of NSs by HeLa cells. Specifically, the

CR

percentage of cells taking in detectable amount of NSs increased from 38.4% in 0.5 h to 97.4% in 6 h. Similar results were also found for 4T1 cell line (Figure S10, S11). Results

M

PTT/PDT-induced cytotoxicity in vitro

AN

US

of intracellular ROS generation are provided in Supplementary data (Figure S12, 13).

BPC NSs and some intermediate products were examined to investigate the PTT/PDT-

ED

induced toxicity on tumor cells (Figure S14). Obviously, strong orange fluorescence of

PT

JC-1 aggregates was observed in the group of cells incubated with BPC NSs without laser irradiation, similar to the control groups treated with only PBS or laser exposure and

CE

corresponding to the normal state of mitochondria without MMP depolarization.

AC

Meanwhile, very weak green fluorescence denoting JC-1 monomers appeared in the cells treated with BP NSs or Ce6 under laser irradiation, indicating the occurrence of mitochondrial damage due to individual PTT or PDT, respectively. In contrast, strong green fluorescence of JC-1 monomers was observed in the cells incubated with BPC NSs under laser irradiation, implying a severe level of mitochondrial damage. A similar effect mediated by BPC NPs was also observed in 4T1 tumor cells in vitro (Figure S15).

11

ACCEPTED MANUSCRIPT To further assess the PTT/PDT effect mediated by BPC NSs, LIVE/DEAD cell viability assays were conducted on HeLa cells after various treatments (Figure 6a). No observable cell death was found in the cells treated solely with BPC NSs or laser irradiation compared to the blank control. However, cell death became pronounced when

T

the cells were subject to Bi2S3@PEG NSs or Ce6 under laser irradiation as evidenced by

IP

the red fluorescence inside the irradiation spot, suggesting strong PTT- or PDT-induced

CR

damage of tumor cells, respectively. More importantly, the highly intensive red fluorescence of the cells treated with BPC NSs under laser irradiation indicated the most

US

severe cell destruction ascribed to a combined PTT/PDT effect. To evaluate the

AN

biocompatibility of the nanocarrier in vitro, the cytotoxicity to somatic cells, including human umbilical vein endothelial cells (HUVECs) and L929 mouse fibroblasts (L929s),

M

was studied in a wide range of concentrations using MTT cell viability assays (Figure 6b,

ED

S16). The cell viability was maintained at above 80% even when the cells were incubated with Bi2S3@PEG NSs at a concentration as high as 200 µg·mL-1, suggesting acceptable

CE

PT

biocompatibility of the carrier material.

In vitro cytotoxicity

AC

The combined PTT/PDT/chemotherapy effect mediated by BPDC NSs was investigated on HeLa cells after various treatments using MTT assays. In the non-irradiated group, negligible cell death was found in those treated with Bi2S3@PEG NSs or free Ce6 in 12 h of incubation (Figure 6c). In contrast, free DOX or BPDC NSs resulted in a notable reduction of HeLa cell viability due to the chemotherapeutic effect of DOX, particularly when the drug concentration was higher than 1 g·mL-1. After applying laser irradiation,

12

ACCEPTED MANUSCRIPT the cytotoxicity of Bi2S3@PEG NSs and Ce6 under higher drug concentrations was induced by PTT and PDT effects, respectively (Figure 6d). Meanwhile, the most significant damage occurred in the cells subject to combination PTT/PDT/chemotherapy mediated by BPDC NS, under which the cell viability decreased dramatically to 6.26 ±

T

0.21% at an equivalent DOX concentration of 5 µg·mL-1. For the other tumor cell line we

IP

investigated, 4T1 cells showed similar responses to the applied recipes of drugs with

CR

various dosages (Figure S17). Furthermore, we discovered that the photo-induced

AN

both 808 nm and 660 nm lasers (Table S2, S3).

US

cytotoxicity of BPDC NSs was also positively correlated to the laser power densities of

M

Potential of BPDC NSs as a CT contrast agent

ED

The CT contrast of BPDC NSs was evaluated by acquiring the phantom images of aqueous NS dispersion at various concentrations in vitro (Figure 7a, b). It was noted that

PT

brightness of the CT phantom images increased linearly with the concentration of BPDC NSs, indicating the positive correlation between the CT signal intensity and nanodrug

CE

concentration. As a high-Z (high atomic number) element with larger X-ray interaction

AC

cross section, bismuth and its analogues are a very promising type of contrast agent for enhanced CT imaging 46, 47.

Pharmacokinetics and biodistribution Effective accumulation of NPs in tumorous tissue through enhanced permeability and retention (EPR) effect is a prerequisite for combination cancer therapy. Therefore, the

13

ACCEPTED MANUSCRIPT pharmacokinetics of BPDC NSs in KM mice was firstly investigated by examining the blood circulation of DOX through fluorescence spectrometry. At 24 h post-injection, ~10.63 ID·g-1 (equivalent DOX dose per gram of tissue) of BPDC NPs was found in blood circulation, which was considerably higher than ~4.12 ID·g-1 of free DOX (Figure

T

7c). The biodistribution of BPDC NSs in 4T1 tumor bearing Balb/c mice was similarly

IP

assessed by examining the content of DOX in various organs and tumors. At 24 h post-

CR

injection, the mice were euthanized, and the tumors as well as major organs were excised for analysis of drug biodistribution (Figure 7d, e). The BPDC NSs exhibited a notable

US

uptaking rate of ~8.14 ID·g-1 in excised tumors at 24 h, which was considerably higher

AN

than ~1.8% ID·g-1 for free DOX.

The fluorescence imaging capacity of BPDC NSs was investigated in 4T1 tumor

M

bearing BALB/c mice intravenously injected with Bi2S3 NSs, Ce6 and BPDC NSs

ED

(Figure 7f, S18). For the group treated with BPDC NSs, strong fluorescence was

PT

observed from the solid tumor region at 24 h post-injection (Figure 7f). However, for the mice treated with free Ce6, much weaker fluorescence was observed in the solid tumor

CE

region while much stronger fluorescence was detected in the liver (Figure S18), attributed

AC

to the rapid clearance of free small molecule drugs and a typical uptake by the reticuloendothelial system (RES). Distinct from the groups treated with Ce6 or BPDC NSs, only very weak background fluorescence could be observed in the group treated with Bi2Se3@PEG NSs. Collectively, these results suggested the tumor-specific accumulation and retention of BPDC NSs under the EPR effect and prolonged blood circulation.

14

ACCEPTED MANUSCRIPT In vivo antitumor efficiency and biosafety of BPDC NSs When tumor volume reached ~200 mm3, all the mice were randomly divided into six groups subject to various treatments. Firstly, the temperature profiles of the mouse body shell at 24 h post-injection were recorded using an infrared thermal camera during

IP

T

NIR laser irradiation on the tumor sites (Figure 8a, S19). As compared to control groups, the temperature of tumor sites treated with Bi2S3@PEG NSs and BPDC NSs rapidly

CR

increased to 55.9°C and 56.1°C within 5 min of NIR laser irradiation, respectively,

US

indicating a remarkable photothermal effect. Furthermore, we explored the antitumor effect of BPDC NSs in vivo by monitoring the tumor volume variation in each group

AN

within 14 days after various treatments (Figure 8c). Similar to the saline group, groups of

M

(3), (4) and (5) indicated insufficient levels of tumor suppression under the individual effects of PTT, chemotherapy and PDT, respectively. However, laser-activated BPDC

ED

NSs in group (6) suppressed the growth of 4T1 tumor evidently with an impressive

PT

inhibition rate of 93.29% without recurrence. On the other hand, non-activated BPDC NSs could not produce a satisfactory tumor suppression effect in group (2) with an

CE

inhibition rate of merely 20.9%, indicating that laser irradiation was an imperative switch

AC

to achieve optimum therapeutic outcome. At day 14 post-treatment, all the mice were euthanized and the tumors were excised and weighted, as shown in Figure 8b. The average weight of tumors in group (6) was the lowest among all groups (Figure 8d), consistent with the terminal tumor volume measured in vivo (Figure 8c). Moreover, there was no obvious loss of body weight in all groups, suggesting a minimum systemic toxicity of BPDC NSs (Figure 8e). Finally, we examined the excised tumors in various groups histologically by hematoxylin and eosin (H&E) staining and terminal

15

ACCEPTED MANUSCRIPT deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. H&E staining indicated that most tumor cells retained their regular morphology with intact nuclei in group (1) treated by saline. Compared to other control groups of (2)-(5), severe pyknosis, karyorrhexis and karyolysis of nuclei were observed in the tumoral tissues treated with

T

laser-activated BPDC NSs in group (6), indicating predominant tumor cell necrosis

IP

resulted from combination therapy (Figure 8f). Furthermore, TUNEL assays revealed

CR

serious tumor cell apoptosis in group (6) as evidenced by the prevalent green

US

fluorescence (Figure 8g).

Potential BPDC-induced hemolytic effect was investigated on red blood cells

AN

(RBCs). RBCs resuspended in DI water and 1×PBS served as the positive and negative

M

controls, respectively. Owing to the imbalanced osmotic pressure in DI water, RBCs tend to lyse and thus release hemoglobin into surrounding medium. However, 1×PBS provides

ED

an isotonic physiological environment that maintains the RBC structure integrity. As

PT

shown in Figure S20, the maximum hemolytic rate of the RBCs was only 1.92% in 4 h after treatment with BPDC NSs at a concentration of 200 µg·mL−1, suggesting the good

CE

hemocompatibility of BPDC NSs. Moreover, routine hematology examination on the

AC

peripheral blood was carried out for up to a week after the mice were administered with BPDC NSs. All the key blood components showed insignificant variation within the provided reference ranges of healthy mice (Figure S21, Table S1). Next, the major organs of treated mice were excised at day 14 for histopathological analysis by H&E staining (Figure S22). There was no apparent inflammation or lesion occurred at either cellular or tissue level among all treated groups.

16

ACCEPTED MANUSCRIPT Discussion Excellent photothermal conversion capacity is always an essential feature of a mediator for efficacious PTT. A terminal temperature of 47.1oC could be achieved within 10 min of irradiation at the BPDC NS concentration of 60 ppm (Figure 3d), exceeding the

T

threshold temperature of 43oC for tumor ablation. Moreover, there was no observable

IP

alternation in the peak temperature of each cycle during the repeated photothermal

CR

responses, confirming a good photothermal stability of BPDC NSs (Figure 3f). In another

US

aspect, the acidity-promoted drug release could be attributed to the protonation of the amino groups in DOX molecules, which facilitated drug dissolution in the release 48

. Given the more acidic tumor microenvironment, as-synthesized BPDC NSs

AN

medium

could potentially realize effective tumor-specific DOX delivery (Figure S8). Moreover, a

M

photo-sensitive drug release behaviour can be attributed to the highly responsive

ED

photothermal capability of Bi2S3, which weakened the carrier-drug interaction upon laser

PT

activation and resulted in an enhanced release rate as also reported in prior literature 49, 50. Efficient cellular uptake of therapeutic agents is critical to ensure the sufficient

CE

bioavailability of drugs at tumor site and thus the efficacy of treatment. Both Ce6 and

AC

DOX can produce fluorescence emission under specific photo-excitation, which can be utilized to trace the internalized BPDC NPs without additional fluorescence markers. Results of both confocal imaging and flow cytometry indicated a remarkable cellular uptake efficiency of BPDC NSs (Figure 4). Reactive oxygen species (ROS) play a key role in cell signalling and homeostasis, and are a facilitator for photodynamic therapy (PDT). In the present study, the production of intracellular ROS was characterized in vitro using DCFH-DA, which can be hydrolysed into DCFH intracellularly and further

17

ACCEPTED MANUSCRIPT oxidized into fluorescent dichlorofluorescein (DCF) under oxidative stress. Since the fluorescence spectra of DOX and DCF overlapped, DOX-free Bi2S3@PEG/Ce6 (BPC) NSs were utilized to avoid the interference with ROS detection. Cells treated with free Ce6 were concurrently examined for comparison. The results verified that BPC provided

T

a potent nanocarrier for more efficient delivery of the photosensitizer Ce6 than its free

IP

molecular forms (Figure 5).

CR

Apoptosis is closely related to the dysfunction of mitochondria, which can be

conversion process

51, 52

US

induced by the oxidative stress from ROS and hyperthermia during photothermal . Mitochondrial membrane potential (MMP) was a critical

AN

indicator of the physiological status of mitochondria, which can be monitored using a JC-

M

1 fluorescence staining kit. JC-1 is a cationic carbocyanine dye, which tends to aggregate in the matrix of normal mitochondrial and produces orange fluorescence (emission peak:

ED

590 nm). However, the MMP of damaged mitochondria decreases and causes conversion

PT

of JC-1 from aggregated state into monomers, which diffuse into cytoplasm and emit green fluorescence (emission peak: 520 nm). Both of HeLa cells and 4T1 cells presented

CE

severe mitochondrial damage mediated by BPC NPs, suggesting a strong combined

AC

PTT/PDT effect (Figure S14, S15). When evaluating the BPC NSs cytotoxicity using Live/Dead cell viability assay, the results were well in accordance with those obtained via JC-1 staining. Cytotoxicity of BPDC NSs from MTT assay quantitatively demonstrated a capable and highly responsive nanoplatform to realize the combined treatment of PTT/PDT/chemotherapy. A relatively longer circulation half-life of BPDC NSs compared to free DOX favors effective drug accumulation in tumors via the EPR effect (Figure 7c). Biodistribution of free DOX in tumor site further confirmed an enhanced drug retention

18

ACCEPTED MANUSCRIPT mediated by BPDC nanocarrier (Figure 7d). Fluorescence and CT properties of BPDC NSs could potentially realize multimodal imaging for cancer diagnosis and staging for clinical

usages.

Encouraged

by

the

promising

effect

of

combined

PTT/PDT/chemotherapy mediated by BPDC NSs in vitro, we proceeded to an in vivo

T

investigation using BALB/c mice bearing 4T1 tumors. The results demonstrated a

IP

remarkable therapeutic efficacy for ablating solid tumors under the combinatorial effect

CR

of local hyperthermia, intracellular ROS and chemotherapeutic toxicity mediated by BPDC NSs (Figure 8). The potential toxicity of BPDC NSs were assessed in terms of

US

hemocompatibility and histocompatibility. The results provided a clear evidence that

AN

BPDC NPs caused minimum systemic toxicity or damage on major organs, which could

M

favor the prolonged circulation in the body after intravenous administration. In summary, PEGylated mesoporous Bi2S3 NSs were synthesized and

ED

concurrently loaded with a photosensitizer (Ce6) and a chemotherapeutic drug (DOX) as

PT

a robust nanoagent for imaging-guided multimodal tumor therapy. At the in vitro level, on-demand drug release was demonstrated to be precisely regulated by the endogenous

CE

factor of pH condition as well as the exogenous stimuli of laser irradiation. The strong

AC

local hyperthermia generation and the yield of ROS mediated by BPDC NSs were simply realized by laser irradiation. After systemic administration in tumor bearing mice, BPDC NSs exhibited rapid accumulation inside the tumor as evidenced by tracing the fluorescence signal of Ce6. A remarkable antitumor efficacy was achieved with the aid of BPDC NSs upon laser irradiation, resulted from the combined PTT/PDT/chemotherapy effect. Benefited from the good biocompatibility, no obvious hepatotoxicity or systemic toxicity was observed during BPDC NS-mediated tumor therapy. This capable

19

ACCEPTED MANUSCRIPT theranostic nanoagent has a good potential to coordinate multiple bioimaging modalities, including infrared thermal imaging, fluorescence imaging and high contrast CT. Therefore, this proof-of-concept work may enlighten further explorations of inorganic metal chalcogenides for applications in multimodal imaging-guided tumor combination

IP

T

therapy.

CR

References

US

1. Jiang S, Gnanasammandhan MK, Zhang Y. Optical imaging-guided cancer therapy

AN

with fluorescent nanoparticles. J R Soc Interface 2010;7(42):3-18. 2. Huang YR, He S, Cao WP, Cai KY, Liang XJ. Biomedical nanomaterials for imaging-

M

guided cancer therapy. Nanoscale 2012;4(20):6135-6149.

ED

3. Ahmed M, Radiology SI. Image-Guided Tumor Ablation: Standardization of Terminology and Reporting Criteria-A 10-Year Update: Supplement to the Consensus

PT

Document. J Vasc Interv Radiol 2014;25(11):1706-1708.

CE

4. Keereweer S, Van Driel PBAA, Snoeks TJA, Kerrebijn JDF, de Jong RJB, Vahrmeijer AL, et al. Optical Image-Guided Cancer Surgery: Challenges and Limitations. Clin

AC

Cancer Res 2013;19(14):3745-3754. 5. Kim DH. Image-Guided Cancer Nanomedicine. J Imaging Sci 2018;4(1). 6. Xie J, Lee S, Chen XY. Nanoparticle-based theranostic agents. Adv Drug Delivery Rev 2010;62(11):1064-1079. 7. Ma XW, Zhao YL, Liang XJ. Theranostic Nanoparticles Engineered for Clinic and Pharmaceutics. Acc Chem Res 2011;44(10):1114-1122.

20

ACCEPTED MANUSCRIPT 8. Cole JT, Holland NB. Multifunctional nanoparticles for use in theranostic applications. Drug Delivery Transl Res 2015;5(3):295-309. 9. Arranja AG, Pathak V, Lammers T, Shi Y. Tumor-targeted nanomedicines for cancer theranostics. Pharmacol Res 2017;115:87-95.

T

10. Fan WP, Yung B, Huang P, Chen XY. Nanotechnology for Multimodal Synergistic

IP

Cancer Therapy. Chem Rev 2017;117(22):13566-13638.

CR

11. Jang B, Kwon H, Katila P, Lee SJ, Lee H. Dual delivery of biological therapeutics for multimodal and synergistic cancer therapies. Adv Drug Delivery Rev 2016;98:113-133.

US

12. Rai P, Mallidi S, Zheng X, Rahmanzadeh R, Mir Y, Elrington S, et al. Development

AN

and applications of photo-triggered theranostic agents. Adv Drug Delivery Rev 2010;62(11):1094-1124.

M

13. Zou LL, Wang H, He B, Zeng LJ, Tan T, Cao HQ, et al. Current Approaches of

ED

Photothermal Therapy in Treating Cancer Metastasis with Nanotherapeutics. Theranostics 2016;6(6):762-772.

therapy:

current

status

and

future

perspective.

Nanomedicine

CE

photothermal

PT

14. Hwang S, Nam J, Jung S, Song J, Doh H, Kim S. Gold nanoparticle-mediated

2014;9(13):2003-2022.

AC

15. Jaque D, Maestro LM, del Rosal B, Haro-Gonzalez P, Benayas A, Plaza JL, et al. Nanoparticles for photothermal therapies. Nanoscale 2014;6(16):9494-9530. 16. Kuroda K, Ye M, Kimura A, Eremeev SV, Krasovskii EE, Chulkov EV, et al. Experimental Realization of a Three-Dimensional Topological Insulator Phase in Ternary Chalcogenide TlBiSe2. Phys Rev Lett 2010;105(14).

21

ACCEPTED MANUSCRIPT 17. Hsieh D, Qian D, Wray L, Xia Y, Hor YS, Cava RJ, et al. A topological Dirac insulator in a quantum spin Hall phase. Nature 2008;452(7190):970-U5. 18. Xiao‐Dong Z, Jie C, Yuho M, Bae PG, Xiu S, Sha‐Sha S, et al. Metabolizable Bi2Se3 Nanoplates: Biodistribution, Toxicity, and Uses for Cancer Radiation Therapy

T

and Imaging. Adv Funct Mater 2014;24(12):1718-1729.

IP

19. Song GS, Liang C, Yi X, Zhao Q, Cheng L, Yang K, et al. Perfluorocarbon-Loaded

CR

Hollow Bi2Se3 Nanoparticles for Timely Supply of Oxygen under Near-Infrared Light to

US

Enhance the Radiotherapy of Cancer. Adv Mater 2016;28(14):2716-2723. 20. Xie HH, Li ZB, Sun ZB, Shao JD, Yu XF, Guo ZN, et al. Metabolizable Ultrathin

AN

Bi2Se3 Nanosheets in Imaging-Guided Photothermal Therapy. Small 2016;12(30):41364145.

M

21. Liu JH, Han JG, Kang ZC, Golamaully R, Xu NN, Li HP, et al. In vivo near-infrared

ED

photothermal therapy and computed tomography imaging of cancer cells using novel

PT

tungsten-based theranostic probe. Nanoscale 2014;6(11):5770-5776. 22. Brenner DJ, Hall EJ. Current concepts - Computed tomography - An increasing

CE

source of radiation exposure. N Engl J Med 2007;357(22):2277-2284.

AC

23. Ke H, Yue X, Wang J, Xing S, Zhang Q, Dai Z, et al. Gold nanoshelled liquid perfluorocarbon nanocapsules for combined dual modal ultrasound/CT imaging and photothermal therapy of cancer. Small 2014;10(6):1220-7. 24. FitzGerald PF, Colborn RE, Edic PM, Lambert JW, Torres AS, Bonitatibus PJ, et al. CT Image Contrast of High-Z Elements: Phantom Imaging Studies and Clinical Implications. Radiology 2016;278(3):723-33.

22

ACCEPTED MANUSCRIPT 25. Kandanapitiye MS, Gao M, Molter J, Flask CA, Huang SPD. Synthesis, Characterization, and X-ray Attenuation Properties of Ultrasmall BiOI Nanoparticles: Toward

Renal

Clearable

Particulate

CT

Contrast

Agents.

Inorg

Chem

2014;53(19):10189-10194.

T

26. Dou RX, Du Z, Bao T, Dong XH, Zheng XP, Yu M, et al. The polyvinylpyrrolidone

IP

functionalized rGO/Bi2S3 nanocomposite as a near-infrared light-responsive nanovehicle

CR

for chemo-photothermal therapy of cancer. Nanoscale 2016;8(22):11531-11542. 27. Liu J, Zheng XP, Gu ZJ, Chen CY, Zhao YL. Bismuth sulfide nanorods as a precision

US

nanomedicine for in vivo multimodal imaging-guided photothermal therapy of tumor.

AN

Nanomedicine (N. Y., NY, U. S.) 2016;12(2):486-487.

28. Li ZL, Hu Y, Chan ML, Howard KA, Fan XL, Sun Y, et al. Highly porous PEGylated

M

Bi2S3 nano-urchins as a versatile platform for in vivo triple-modal imaging, photothermal

ED

therapy and drug delivery. Nanoscale 2016;8(35):16005-16016. 29. Hong EJ, Choi DG, Shim MS. Targeted and effective photodynamic therapy for

PT

cancer using functionalized nanomaterials. Acta Pharm Sin B 2016;6(4):297-307.

CE

30. Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, et al. Photodynamic Therapy of Cancer: An Update. Ca-Cancer J Clin 2011;61(4):250-281.

AC

31. Juarranz A, Jaen P, Sanz-Rodriguez F, Cuevas J, Gonzalez S. Photodynamic therapy of cancer. Basic principles and applications. Clin Transl Oncol 2008;10(3):148-54. 32. Selvestrel F, Moret F, Segat D, Woodhams JH, Fracasso G, Echevarria IMR, et al. Targeted delivery of photosensitizers: efficacy and selectivity issues revealed by multifunctional ORMOSIL nanovectors in cellular systems. Nanoscale 2013;5(13):61066116.

23

ACCEPTED MANUSCRIPT 33. Chin WWL, Heng PWS, Thong PSP, Bhuvaneswari R, Hirt W, Kuenzel S, et al. Improved formulation of photosensitizer chlorin e6 polyvinylpyrrolidone for fluorescence diagnostic imaging and photodynamic therapy of human cancer. Eur J Pharm Biopharm 2008;69(3):1083-1093.

T

34. Mojzisova H, Bonneau S, Vever-Bizet C, Brault D. Cellular uptake and subcellular

CR

lipoproteins. Bba-Biomembranes 2007;1768(11):2748-2756.

IP

distribution of chlorin e6 as functions of pH and interactions with membranes and

35. Kostryukova LV, Prozorovskiy VN, Medvedeva NV, Ipatova OM. Comparison of a

AN

free form. FEBS Open Bio 2018;8(2):201-210.

US

new nanoform of the photosensitizer chlorin e6, based on plant phospholipids, with its

36. Yin T, Zhang Q, Wu HG, Gao G, Shapter JG, Shen YL, et al. In vivo high-efficiency

M

targeted photodynamic therapy of ultra-small Fe3O4@polymer-NPO/PEG-Glc@Ce6

ED

nanoprobes based on small size effect. NPG Asia Mater 2017;9: e383. 37. Xu J, Xu LG, Wang CY, Yang R, Zhuang Q, Han X, et al. Near-Infrared-Triggered

PT

Photodynamic Therapy with Multitasking Upconversion Nanoparticles in Combination

CE

with Checkpoint Blockade for Immunotherapy of Colorectal Cancer. ACS Nano 2017;11(5):4463-4474.

AC

38. Yang GB, Xu LG, Chao Y, Xu J, Sun XQ, Wu YF, et al. Hollow MnO2 as a tumormicroenvironment-responsive biodegradable nano-platform for combination therapy favoring antitumor immune responses. Nat Commun 2017;8:902. 39. Wang C, Cheng L, Liu Z. Upconversion Nanoparticles for Photodynamic Therapy and Other Cancer Therapeutics. Theranostics 2013;3(5):317-330.

24

ACCEPTED MANUSCRIPT 40. Fan F, Yu Y, Zhong F, Gao M, Sun TM, Liu JX, et al. Design of Tumor AcidityResponsive Sheddable Nanoparticles for Fluorescence/Magnetic Resonance ImagingGuided Photodynamic Therapy. Theranostics 2017;7(5):1290-1302. 41. Zhang N, Zhao FF, Zou QL, Li YX, Ma GH, Yan XH. Multitriggered Tumor-

T

Responsive Drug Delivery Vehicles Based on Protein and Polypeptide Coassembly for

IP

Enhanced Photodynamic Tumor Ablation. Small 2016;12(43):5936-5943.

CR

42. Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol 2015;33(9):941-951.

using

bismuth

trisxanthate single source precursors.

Chem

Mater

AN

nanowire

US

43. Koh YW, Lai CS, Du AY, Tiekink ERT, Loh KP. Growth of bismuth sulfide

2003;15(24):4544-4554.

M

44. Chen Z, Liu XW, Liu YH, Gunsel S, Luo JB. Ultrathin MoS2 Nanosheets with

ED

Superior Extreme Pressure Property as Boundary Lubricants. Sci Rep 2015;5:12869. 45.Lin Z, Liu Y, Ma XM, Hu SY, Zhang JW, Wu Q, et al. Photothermal ablation of bone

CE

2015;5: 11709.

PT

metastasis of breast cancer using PEGylated multi-walled carbon nanotubes. Sci Rep

46. Yu X, Li A, Zhao C, Chen X, Li W. Ultrasmall Semimetal Nanoparticles of Bismuth

AC

for Dual-Modal Computed Tomography/Photoacoustic Imaging and Synergistic Thermoradiotherapy. ACS Nano. 2017;11:3990-4001. 47. Kinsella JM, Jimenez RE, Karmali PP, Rush AM, Kotamraju VR, Ruoslahti E, et al. X-Ray Computed Tomography Imaging of Breast Cancer by using Targeted PeptideLabeled Bismuth Sulfide Nanoparticles. Angew Chem Int Ed. 2011;50:12308-12311.

25

ACCEPTED MANUSCRIPT 48. Mohan P, Rapoport N. Doxorubicin as a Molecular Nanotheranostic Agent: Effect of Doxorubicin Encapsulation in Micelles or Nanoemulsions on the Ultrasound-Mediated Intracellular Delivery and Nuclear Trafficking. Mol Pharmaceutics 2010;7(6):1959-1973. 49. Chen KJ, Chaung EY, Wey SP, Lin KJ, Cheng F, Lin CC, et al. Hyperthermia-

IP

Specific Chemotherapy. ACS Nano 2014;8(5):5105-5115.

T

Mediated Local Drug Delivery by a Bubble-Generating Liposomal System for Tumor-

CR

50. Ponce AM, Vujaskovic Z, Yuan F, Needham D, Dewhirst MW. Hyperthermia mediated liposomal drug delivery. Int J Hyperthermia 2006;22(3):205-213.

US

51. Ahmed K, Tabuchi Y, Kondo T. Hyperthermia: an effective strategy to induce

AN

apoptosis in cancer cells. Apoptosis 2015;20(11):1411-1419. 52. Redza-Dutordoir M, Averill-Bates DA. Activation of apoptosis signalling pathways

M

by reactive oxygen species. Biochim Biophys Acta Mol Cell Res 2016;1863(12):2977-

AC

CE

PT

ED

2992.

26

ACCEPTED MANUSCRIPT Figure legends Figure 1. Schematic illustration of the synthetic procedure of PEGylated mesoporous Bi2S3 nanostars encapsulating DOX and Ce6 (BPDC NSs) as a multifunctional platform

T

for cancer theranostics.

IP

Figure 2. Physicochemical characterizations: (a) a high-resolution TEM image of Bi2S3

CR

NSs (scale bar: 200 nm); (b) hydrodynamic diameter of Bi2S3 NSs measured by DLS; (c) zeta potentials of intermediate and final products; (d-f) XPS survey spectra of Bi2S3 NSs,

US

including (d) full scan XPS survey spectrum of Bi2S3 NSs and a core level XPS spectrum

AN

corresponding to (e) Bi4f and (f) S2S; (g) XRD patterns of Bi2S3 NSs; (h) N2 adsorption– desorption isotherm and (i) the corresponding pore size distribution of Bi2S3 NSs.

M

Figure 3. Photothermal characterizations: (a) UV-vis-NIR absorption spectra of

ED

Bi2S3@PEG NSs, free DOX, free Ce6 and BPDC NSs; (b) UV-vis-NIR absorption spectra of BPDC NSs dispersed in DI water at various concentrations; (c) temperature

PT

variation in the dispersions of BPDC NSs (100 ppm) , BP NSs (100 ppm) and DI water

CE

under NIR laser irradiation for 10 min (808 nm, 2 W·cm-2); (d) temperature variation in BPDC NS dispersions at various concentrations (0 to 100 ppm) under NIR laser

AC

irradiation for 10 min (808 nm, 2 W·cm-2); (e) temperature variation of BPDC NS dispersion (100 ppm) under NIR laser irradiations with different power intensity for 10 min; (f) temperature variation of BPDC NS dispersion (100 ppm) under cyclic laser exposures (10 min of laser irradiation per cycle); (g) thermographic imaging of sampleladen cuvettes corresponding to (c), (d) and (e).

27

ACCEPTED MANUSCRIPT Figure 4. Characterizations of cellular uptake of BPDC NSs: (a) Confocal fluorescence images of HeLa cells incubated with BPDC NSs for 1 h or 4 h. The fluorescence signals of DOX and Ce6 are displayed in green and red colors, respectively (scale bars: 20 µm). Images in 2nd and 4th rows denote the magnified areas enclosed in dashed box in 1st and

T

3rd rows, respectively. (b) Flow cytometry analysis of cellular uptake of BPDC NSs in

IP

HeLa cells after different incubation time. (c) Mean intensity of fluorescence emission

CR

from the cells corresponding to (b). (d) Flow cytometry dot plots with BPDC uptaking

US

rate of the cells corresponding to (b).

Figure 5. Characterization of intracellular ROS generation using DCFH-DA probe:

AN

confocal fluorescence images of HeLa cells subject to Ce6 or BPC NSs with or without

M

660 nm laser irradiation (scale bars: 20 µm). Images in 2nd, 4th, 6th and 8th rows are the

ED

magnified areas enclosed in dashed box in 1st, 3rd, 5th and 7th rows, respectively. Figure 6. (a) Live/Dead cell viability assays on HeLa cells under various treatments. The

PT

dashed circles denote laser irradiation spots (scale bars: 200 µm, 0.232 mm2 per spot); (b) Viability of HUVECs and L929 cells after treatment with Bi2S3@PEG NSs at various

CE

concentrations (0 to 200 µg·mL-1); Viability of HeLa cells after treatment with

AC

Bi2S3@PEG NSs, free Ce6, DOX or BPDC NSs without (c) or with (d) laser irradiation. Figure 7. (a) In vitro CT images of BPDC NSs; (b) CT values of the aqueous dispersions of BPDC NSs at various concentrations corresponding to (a); (c) In vivo pharmacokinetic curves over 24 h after intravenous injection of free DOX or BPDC NSs; (d) Biodistribution of free DOX or BPDC NSs at 24 h post-injection; (e) The fluorescence image of excised organs and tumors at 24 h post-injection; (f) Fluorescence images of

28

ACCEPTED MANUSCRIPT BALB/c mice bearing 4T1 tumors before and after the administration of BPDC NSs (dashed circles denote the location of solid tumor). Figure 8. Treatment of tumors in vivo with BPDC NSs: (a) infrared thermal images of tumors after intravenously injection with various agents upon NIR laser irradiation; (b)

IP

T

photographs of the excised tumors at day 14 after various treatments; (c) tumor volume variation after various treatments; (d) weight of the excised tumors at day 14 after various

CR

treatments; (e) change in mouse body weight after various treatments; histological

US

analysis of tumor sections using (f) H&E staining (black dashed boxes denote magnified

AC

CE

PT

ED

M

AN

sections) and (g) TUNEL staining.

29

ACCEPTED MANUSCRIPT Graphical abstracts

Highly biocompatible PEGylated mesoporous bismuth sulfide nanostars encapsulating

T

doxorubicin and chlorin e6 (BPDC NSs) were facilely synthesized for tumor theranostics.

IP

BPDC NSs, as a potent contrast agent, also enable specific multimodal imaging of tumors,

CR

including infrared thermal imaging, fluorescence imaging and computed tomography (CT). Moreover, aided by the passive targeting effect, the accumulation of BPDC NSs

US

contributed to the combination effects of photothermal therapy, photodynamic therapy

AN

and chemotherapy of solid tumors. Collectively, BPDC NPs may serve as a promising nanomedicine for highly efficacious tumor suppression and eradication with imaging

AC

CE

PT

ED

M

guidance.

30

Figure 1

Figure 2

Figure 3

Figure 4

Figure 5

Figure 6

Figure 7

Figure 8