MoS2 Quantum Dot@Polyaniline Inorganic ... - ACS Publications

Sep 6, 2016 - tomography (CT) signal but also perform efficient radiotherapy ... tumor therapeutic strategy, radiation therapy (RT) usually employs th...
0 downloads 0 Views 2MB Size
Subscriber access provided by Northern Illinois University

Letter 2

MoS Quantum Dots@Polyaniline Inorganic-Organic Nanohybrids for In Vivo Dual-Modal Imaging Guided Synergistic Photothermal/Radiation Therapy Jinping Wang, Xiaoxiao Tan, Xiaojuan Pang, Li Liu, Fengping Tan, and Nan Li ACS Appl. Mater. Interfaces, Just Accepted Manuscript • DOI: 10.1021/acsami.6b08391 • Publication Date (Web): 06 Sep 2016 Downloaded from http://pubs.acs.org on September 7, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Applied Materials & Interfaces is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

MoS2 Quantum Dots@Polyaniline Inorganic-Organic Nanohybrids for In Vivo Dual-Modal Imaging Guided Synergistic Photothermal/Radiation Therapy Jinping Wang, Xiaoxiao Tan, Xiaojuan Pang, Li Liu, Fengping Tan*, Nan Li* Tianjin Key Laboratory of Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China.

*Corresponding author at: School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China. Tel.:+86-022-27404986 E-mail address: [email protected] *Co-corresponding author at: School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, PR China. E-mail address: [email protected] Tel.:+86-022-27405160

1 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 25

Abstract In this study, we introduce a versatile nanomaterial based on MoS2 quantum dot @Polyaniline (MoS2@PANI) inorganic-organic nanohybrids, which exhibit good potential to not only enhance photoaccoustic (PA) imaging/X-ray computed tomography (CT) signal but also perform efficient radiotherapy (RT)/photothermal therapy (PTT) of cancer. Upon the intravenous injection of MoS2@PANI hybrid nanoparticles, the in vivo tumor could be precisely positioned and thoroughly eliminated under the PA/CT image-guided combination therapy of PTT/RT. This versatile nanohybrid could show good potential to facilitate simultaneously dual-modal imaging and synergetic PTT/RT to realize better anticancer efficiency.

Keywords: MoS2 quantum dots, Polyaniline, Inorganic-organic nanohybrids, Photothermal/radiation therapy, Dual-modal CT/photoacoustic imaging

2 ACS Paragon Plus Environment

Page 3 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

Photothermal therapy (PTT), which uses near-infrared (NIR) light-absorbing materials to turn optical energy into heat to thermally ablate tumor cells, has been extensively explored because of its many superiorities such as fewer side effects, high specificity, and minimal trauma to normal tissues.1-3 However, PTT alone is difficult to completely eradicate tumors, especially for the deep-located tumors because that the NIR laser intensity will occur an inevitable depth-dependent decrease, which is an intrinsic drawback of optical therapy. On account of this limitation, the combination therapy is usually regarded as a promising strategy to enhance the therapeutic efficacy.4-6 As one of the most widely used tumor therapeutic strategy, radiation therapy (RT) usually employs the ionizing radiation (e.g., γ-ray, X-ray) to locally apply on the tumor to generate oxygen-centered radicals produced by the ionization of surrounding water to cause the DNA damage and a following tumor cells death with no depth restriction.7-9 However, the level of cellular oxygenation heavily influences the degree of cellular damage which is caused by ionizing radiation. Thus, the hypoxic tumor microenvironment is considered as one of the major obstacle for RT.10 Fortunately, an proper level of hyperthermia induced by PTT could increase intratumoral blood stream and subsequently enhance oxygen condition in the tumor microenvironment, which may cause the cells to be more sensitive to RT.11-13 Therefore, combining PTT with RT is preferable for improving the therapeutic outcomes of cancer. In addition, to make the therapic process more accurate, the imaging-guided treatment has been suggested as an encouraging strategy to realize precise cancer therapy.14 As a result, theranostic nanomaterials which have highly

3 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 25

integrated multifunctions and can be fabricated using relatively reliable methods would be of great interest. Recently, many nanostructures have been employed as the NIR adsorbing agents or radiosensitizers for enhancing the efficacy of PTT or RT, which can be divided into two

major

types:

the

inorganic

polymers-based nanomaterials.11,

15

materials-based

and

organic

conjugated

To date, a variety of inorganic nanomaterials

including gold nanomaterials,16 MoS2,17 WS2,13 have been widely studied as PTT nanoagents or radiosensitizer. However, these inorganic nanoscale materials can’t be biodegraded and usually remains in the body for a long time, causing a potential long-term toxicity. Thus, more biodegradable and nontoxic organic materials such as polyaniline (PANI),18 polypyrrole (PPy)15 have acquired wide attention. Nevertheless, due to the relatively low background contrast and poor tumor uptake, the organic conjugated polymers alone have limited applications in the tumor diagnosis and treatment.19 Thus, as a candidate, inorganic-organic hybrid nanomaterial has attracted much interest as a diagnostic agent and therapeutic platform since it could take advantage of both and avoid the shortcomings of each. On one hand, the inorganic materials can enhance the diagnosibility of hybrids due to their excellent optical properties, on the other hand, the medicative organic polymers can decrease the therapeutic concentration of the inorganic materials and subsequently decline their amounts retained in the body. In this work, we fabricated MoS2 quantum dot @Polyaniline (MoS2@PANI) nanohybrids and explored them as both photothermal adsorbing agents and

4 ACS Paragon Plus Environment

Page 5 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

radiosensitizers for combined PTT and RT, which has not been investigated up to now. Nanoscaled transition metal dichalcogenides, such as MoS217 and WS2,13 have been studied as new photothermal agents for PTT. Moreover, MoS2 is anticipated to be a qualified candidate for use as radiosensitizers for RT due to its high Z number. The as-prepared MoS2 nanoquantum dots (MoS2 QDs) with a mean size of 5 nm could also produce the strong fluorescence and thus could use as the potential probes for in vitro and in vivo imaging.20 The obtained MoS2 QDs core was mixed in the presence of polyvinylpyrrolidone (PVP), sequentially sealed in a PANI shell through in situ polymerization of anline by dispersing the as-grown cores in an acid aniline solution and mildly stiring at room temperature to form MoS2@PANI. Interestingly, PANI is a promising photothermal agent due to their great photothermal conversion efficiency, remarkable biocompatibility, and good photostability.21 The prepared MoS2@PANI nanohybrids could be extended to use as the contrast agents for both X-ray computed tomography (CT) and photoaccoustic (PA) imaging of tumors due to their strong X-ray attenuation and high NIR absorption efficiency. As a consequence, the MoS2@PANI nanohybrid provides a new possibility in exploring the versatile inorganic-organic nanoplatform for simultaneous biomedical imaging and combined PTT and RT of tumors. The MoS2 QDs were prepared by a simple solvothermal method20 in N-methyl-2-pyrrolidone (NMP), as shown in Figure 1a. The prepared products were centrifuged for several minutes. Afterwords, the transparent light yellow solution was obtained. In order to remove the solvent, the obtained solution was evaporated under

5 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 25

vacuum at a certain temperature. Then the obtained precipitate was redispersed in ethyl acetate to acquire the MoS2 QDs powder by filtration and followed vacuum volatilization. The MoS2 QDs powder was dispersed in water in the presence of PVP (10 kDa), which was used as a dispersing and stabilizing reagent to control the size of MoS2 QDs. Then the above solution and the mixture of aniline and 3-aminobenzoic acid (aniline-COOH) with a certain molar ratio was introduced into the acidic aqueous dispersion of PVP, ammonium persulfate (APS), and sodium dodecylsulfate (SDS). Due to the electrostatic forces, the aniline molecules would be absorbed onto the surface of MoS2 QDs to form the MoS2@PANI nanoparticles with -COOH groups on their surface (Figure S1a, Supporting Information). Then, the PEG-NH2 was conjugated to the surface of MoS2@PANI-COOH nanoparticles by the stable amide bonds formation with coupling agents NHS and EDC, obtaining the soluble and stable MoS2@PANI nanohybrids (Figure 1a). Transmission electron microscope (TEM) images shown the uniform MoS2 QDs (Figure 1b). The size distribution displayed the mean size of MoS2 QDs was about 5.3 nm (Figure 1d). In addition, TEM images clearly revealed the core-shell structure of MoS2@PANI nanoparticles (Figure 1c). The PANI nanoshells, as the efficient NIR light absorber, showed ring-like shell structures with a thickness about 15 nm. The average diameter of MoS2@PANI nanohybrids was 21.5 ± 3.9 nm (polydispersity index = 0.203), which was an excellent size range for efficient endocytosis and deep tissue penetration (Figure 1d).22 Copolymerization of aniline and aniline-COOH improved the solubility and colloidal stability of the MoS2@PANI nanoparticles. As illustrated in Figure S1b (Supporting

6 ACS Paragon Plus Environment

Page 7 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

Information), MoS2@PANI made from polymerization of pure aniline aggregated in phosphate buffered saline (PBS) and precipitated out of solution in a few hours, whereas MoS2@PANI made from copolymerization of aniline and aniline-COOH remained stable. The PANI nanoshell formation was further confirmed by the UV-vis-NIR absorption spectrum. Strong light extinction was observed around 405 nm, indicating formation of the polymer shell. Compared to the typical bipolaron absorption peak of pure PANI centered at 415 nm (Figure S2), the slight spectral blue-shift was likely due to the incorporation of aniline-COOH (10 % by molar ratio) in the copolymer. In addition, the inorganic-organic nanohybrids showed a broad absorption profile in the NIR region comparing with the original MoS2 QDs, (Figure 1f), which is highly desirable for absorption-based applications such as PA imaging and PTT. The fluorescence spectra of MoS2@PANI nanoparticles showed the strongest emission peak at 483 nm when the excitation wavelength was 390 nm (Figure 1e). When the excitation wavelength ranged from 350 to 490 nm, the fluorescence emission maximum of MoS2@PANI nanoparticles shifted to longer wavelength from 474 to 570 nm, which revealed the possible application of these nanohybrids as cell-imaging agents. An important feature of MoS2@PANI nanoparticles is their NIR light-evoked photothermal effect, which is essential for PTT. As shown in Figure 1g, the temperature changes of MoS2@PANI nanoparticles aqueous solution, pure PANI, MoS2 QDs dispersion, and pure water. Hardly any temperature change was observed when the pure water was irradiated with 808 nm laser (1.5 W cm−2) for 6 min. By

7 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 25

contrast, the temperature of MoS2@PANI nanoparticles solution (100 µg mL−1) increased from 25 to 54.8 ℃, which was sufficient to cause an irreversible damage to tumor cells.23 In addition, MoS2@PANI nanoparticles still remained an excellent capability of temperature increasing after four cycles of the laser exposure, suggesting the satisfied photostability of MoS2@PANI nanohybrids to perform repeated PTT treatment (Figure S3c, Supporting Information). Afterwards, the photothermal conversion ability of the MoS2@PANI nanoparticles was further investigated. We monitored the temperature changes of the MoS2@PANI nanoparticles (100 µg mL−1) under the NIR laser irradiation (808 nm, 1.5 W cm−2) for 300s (Figure S3a and b, Supporting Information). Based on the reported method and acquired data, the photothermal conversion efficiency (η) of MoS2@PANI nanoparticles could reach 31.6 %. Before exploring their in vivo performance, we evaluated the biocompatibility of MoS2 QDs and MoS2@PANI nanoparticles using the methyl thiazolyl tetrazolium (MTT) assay. Our results demonstrated that MoS2 QDs and MoS2@PANI nanoparticles exhibited negligible toxicity to 4T1 cells (murine breast cancer cells) and HUVECs (human umbilical vein endothelial cells) when the tested concentrations ranged from 0 to 200 µg mL−1 (Figure S4a and b, Supporting Information). The 4T1 cells and HUVECs were obtained from Wuhan Procell Life Science and Technology Co., Ltd. Next, we next assessed the cell cytotoxicity, cellular uptake and synergistic PTT/RT efficacy of MoS2@PANI nanohybrids using 4T1 cells. As shown in Figure 2a, the MoS2 + PTT group leaded to 20.5% cell mortality rate, while the toxicity of MoS2

8 ACS Paragon Plus Environment

Page 9 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

+ RT group was about 44.8%. After treated with MoS2@PANI nanohybrids, single PTT or RT caused 62.3 or 51.1% of cell dead, respectively. The simultaneous PTT/RT treatments, however, induced cell death up to 78.4%. Then, the confocal fluorescence images (Figure 2b) of propidium iodide (PI) and calcein AM costained cells further evaluated the X-ray and NIR laser induced simultaneous PTT/RT efficiency of MoS2@PANI nanoparticles. Barely dead cell could be observed in PBS control group. Besides, only a few cells were killed and exhibited red fluorescence in the single PTT or RT treated groups. Conversely, in the MoS2@PANI + PTT + RT group, almost all of the cells were destroyed and displayed red fluorescence, demonstrating that the enhanced therapeutic efficiency for the simultaneous PTT/RT effect of MoS2@PANI nanoparticles. The cellular uptake behavior of free MoS2 QDs and MoS2@PANI nanoparticles was investigated using the 4T1 cells by confocal microscopy (Figure 2c). After 6 h incubation, weak red fluorescent signals of free MoS2 QDs were observed in the cell cytoplasm. In addition, considerably enhanced intracellular MoS2 QDs fluorescence was visual in MoS2@PANI nanoparticles-incubated cells after 5 min NIR irradiation in comparison with those without irradiation. This phenomenon could be attributed to the higher cellular metabolism and membrane permeability by the laser-induced hyperthermia, thereby enhancing the nanoparticles accumulation inside cancer cells.24 To further confirm the potential radio-sensitization capability, MoS2@PANI nanoparticles-enhanced X-ray radiation therapy of 4T1 cells was evaluated by γ-H2AX immunofluorescence to investigate the RT-induced DNA damage.25 As

9 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 25

shown in Figure 2d, immunofluorescent images revealed a very low level of γ-H2AX signal in the PBS treated group, as well as only MoS2@PANI nanohybrids treated group. By contrast, higher levels of γ-H2AX foci, which is a marker of double-strand DNA breaks, were observed within cells nuclei of MoS2@PANI + PTT + RT treated group than those of MoS2@PANI + RT alone treated group, which further confirmed the PTT-enhanced RT effect of cancer cells. Ideal nanosystems used as the nanomedicine should possess multimodality in both diagnosis and therapy. Precise spatial- and temporal-specific monitoring in vivo is particularly demanded as it potentially opens a novel avenue in guiding the therapeutic process, monitoring the therapeutic response, and avoiding the damage induced by external irradiation to surrounding normal tissue. Encouraged by the strong NIR optical/X-ray absorption of MoS2@PANI nanohybrids, X-ray CT and PA imaging were used for tracking nanoparticles’ in vivo biodistribution (Figure 3a). In our experiments, MoS2@PANI nanohybrids (200 μL, 2 mg/mL) were intravenously injected into Balb/c nude mice bearing a 4T1 tumor through the tail vein, followed by PA images at different time points (Figure 3b). Before injection, the PA signal in the tumor was very weak, while the signal remarkably increased after i.v. injection over time, indicating the effective tumor retention of the MoS2@PANI nanohybrids by the enhanced permeability and retention (EPR) effect.26 After administration of MoS2@PANI nanohybrids for 8 h, the PA signal reached ∼535 au, which was 5 times higher than that before injection (∼105 au) in the tumor and lasted for 24 h (Figure 3e). Moreover, in view of high X-ray absorption coefficient of the molybdenum, these

10 ACS Paragon Plus Environment

Page 11 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

MoS2@PANI nanohybrids could also be used as CT imaging contrast agents. CT imaging as a more mature bioimaging strategy offers the high-resolution 3D detailed structure of the whole body, while PA imaging owning much higher spatial resolution illustrates the detailed distribution of nanoparticles in the tumor site. As the result, the combination of CT and PA imaging could provides the whole-body imaging with high resolution without depth limitation and thus be useful for careful therapeutic schedule.27 In order to confirm the distribution of MoS2@PANI nanohybrids and to examine their CT contrast performance, MoS2@PANI nanohybrids (200 μL, 8 mg/mL) were intravenously injected into the Balb/c nude mice bearing a 4T1 tumor through the tail vein for CT detection of the whole body 8 h post injection (Figure 3c and d). Compared to the pre-injection image, remarkably tumor CT contrast signals were found after 8 h postinjection of MoS2@PANI nanohybrids, indicating the adequate accumulation of nanohybrids in tumor sites. In addition, the HU value which represented CT contrast effect increased from 135.0 ± 23.1 before injection to 243.7 ± 19.2 after i.v. administration of MoS2@PANI nanohybrids (Figure 3f) which was in agreement with the PA results. Thus, MoS2@PANI nanohybrids experienced remarkable PA/CT imaging capabilities and obvious tumor-homing effects. Next, the in vivo blood circulation of MoS2@PANI nanohybrids was examined after intravenously injected into the normal mice to confirm the above imaging results by measuring the Mo4+ concentration of the blood samples using inductively coupled plasma mass spectrometry (ICP-MS). The results in Figure S5a and b revealed the prolonged blood circulation time of MoS2@PANI nanohybrids. The MoS2@PANI

11 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 25

nanohybrids displayed a much longer half-time (5.02 ± 1.37 h) than MoS2 QDs (0.85 ± 0.51 h). During the blood circulation, PEG and PANI covering on the surface of MoS2 QDs might delay the MoS2@PANI nanoparticles macrophage clearance and enhance the tumor accumulation of nanoparticles by the EPR effect. Following the in vivo dual-model imaging, simultaneous PTT/RT treatments were introduced on the 4T1 tumor model. Tumor-bearing mice (the volume of the tumors were about 80 mm3 ) were divided into six groups: PBS group, MoS2@PANI group, MoS2 plus PTT group, MoS2@PANI plus PTT group, MoS2@PANI plus RT group, and MoS2@PANI plus PTT/RT group. In this experiment, PTT was carried out with the 808 nm laser irradiation (1.5 W cm-2, 5 min), while the RT was introduced at the X-ray radiation dose of 6 Gy. Based on the observations from CT and PA imaging, PTT was conducted after 8 h postinjection of the samples. Upon the localized 808 nm laser irradiation with a power density of 1.5 W cm−2 for 5 min, the temperature change of tumor for mice treated with PBS, MoS2 or MoS2@PANI was monitored with the infrared (IR) thermal camera (Figure 4a). It could be found that the tumors temperature increased from 35.1 to 51.4 ℃ within 5 min for mice treated with MoS2@PANI nanohybrids, whereas tumors of PBS and MoS2 treated group showed only ∼3 ℃and ∼5 ℃ increase under the same irradiation, respectively. Then the tumor sizes in different groups were monitored (Figure 4b). It was found that tumors growth in the MoS2@PANI plus PTT/RT group was remarkably inhibited during 14 days. On the other hand, although the MoS2@PANI nanohybrids could enhance the efficacy of X-ray-induced RT treatment or NIR laser-induced PTT treatment, tumors

12 ACS Paragon Plus Environment

Page 13 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

in those groups (MoS2@PANI plus PTT group and MoS2@PANI plus RT group) still kept a certain growth rate. In contrast, tumors in other control groups, including PBS injection, MoS2 injection with PTT treatment, as well as MoS2@PANI injection, all showed rapid growth. In contrast to control groups, excellent therapeutic effectiveness of the MoS2@PANI plus PTT/RT group could be obviously found, leaving noticeable black scars at the original tumor sites (Figure S6, Supporting Information). To further confirm the antitumor efficacy of different treatments, hexatoxylin and eosin (H&E) staining was studied. Compared with the PBS group, the combination PTT/RT treatment group could lead to severe destruction of tumor cells, as evidenced by increased vacuoles, changed cell shapes, and condensed nuclei observed from micrographs of H&E-stained tumor slices (Figure 4e). Simultaneously, no noticeable sign of toxicity of the combined PTT and RT treatment with MoS2@PANI nanoparticles was observed in the micrographs of H&E-stained slices from MoS2@PANI treated mice organs collected after 14 days post-treatment. Hypoxia, as a common characteristic of the tumor microenvironment, negatively affects the clinical prognosis for all sorts of solid tumors because of its vital function in tumor progression, metastasis, and resistance to mainstream cancer treatment strategies such as RT.28 Comparing with treatment of hypoxic tumors, the ionization radiation effect could be enhanced in the presence of oxygen.29 In order to understand the mechanism of synergistic effect of combined PTT/RT with MoS2@PANI, immunofluorescence staining experiments was carried out by staining cell nuclei, blood

vessels,

and

tumor

hypoxic

areas

with

13 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 25

2-(4-amidinophenyl)-6-indolecarbamidine dihydrochloride (DAPI) (blue), anti-CD31 antibody (red), and anti-pimonidazole antibody (green), respectively (Figure 4c, d). It was found that the group treated with MoS2@PANI plus NIR displayed a smaller tumor hypoxia area than the control group (MoS2@PANI), especially for cells around the blood vessels, indicating that the tumor hypoxia could be efficiently reduced through the appropriate hyperthermia. Thus, due to the enhanced tumor blood flow under hyperthermia,28 the overall tumor oxygenation status was improved immediately after PTT. This phenomenon may subsequently help to overcome the hypoxia-associated radio-resistance in combined PTT/RT therapy. In

summary,

multifunctional

nanotheranostics

based

on

MoS2@PANI

inorganic-organic nanohybrids have been successfully fabricated to achieve simultaneous CT/PA imaging and synergistic PTT/RT combination therapy for cancer. It was found that MoS2@PANI nanohybrids demonstrated good compatibility in physiological environments and excellent biocompatibility in both in vitro and in vivo studies. With the strong X-ray attenuation and high NIR absorption efficiency, the prepared MoS2@PANI nanohybrids could used for dual-modal CT and PA imaging, respectively. Bimodal CT/PA images demonstrated the efficient tumor accumulation of these MoS2@PANI nanohybrids after systemic administration into tumor-bearing mice. Significantly, due to the enhanced oxygenation in the tumor under mild PTT, a remarkable synergistic cancer treatment effect was observed in our PTT/RT combination therapy. Our results not only provide a versatile nanomaterial for dual-modal image-guided PTT/RT therapy, but also encourage to explore other

14 ACS Paragon Plus Environment

Page 15 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

multifunctional nanomedicine for tumor diagnosis and therapy. ASSOCIATED CONTENT Supporting Information. Experimental on materials, preparation of MoS2 QDs and MoS2@PANI nanohybrids, characterization, cellular experiments, tumor model and in vivo cancer treatment, in vivo biodistribution studies, blood circulation measurement, immunohistochemistry, and statistical analysis; construction and photograph of the prepared nanoparticle, absorbance spectra of pure PANI, photothermal results of the MoS2@PANI nanohybrids, cell viability results by MTT assay, blood circulation results of different samples, and photos of mice before and after treatment. Acknowledgments National Natural Science Foundation of China (81503016), the Application Foundation and Cutting-edge Technologies Research Project of Tianjin (Young Program) (15JCQNJC13800), and the National Basic Research Project (973 Program) of China (2014CB932200) are acknowledged for financial support. References (1) Liu, H. Y.; Chen, D.; Li, L. L.; Liu, T. L.; Tan, L. F.; Wu, X. L.; Tang, F. Q. Multifunctional Gold nanoshells on Silica Nanorattles: a Platform for the Combination of Photothermal Therapy and Chemotherapy with Low Systemic Toxicity. Angew. Chem., Int. Ed. 2011, 50, 921-925. (2) Dong, W. J.; Li, Y. S.; Niu, D. C.; Ma, Z.; Gu, J. L.; Chen, Y.; Zhao, W. R.; Liu, X. H.; Liu, C. S.; Shi, J. L. Facile Synthesis of Monodisperse Superparamagnetic Fe3O4 Core@hybrid@Au Shell Nanocomposite for Bimodal Imaging and Photothermal

15 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 25

Therapy. Adv. Mater. 2011, 2, 5392-5397. (3) Zhang, Z. J.; Wang, L. M.; Wang, J.; Jiang, X. M.; Li, X. H.; Hu, Z. J.; Ji, Y. L.; Wu, X. C.; Chen, C. Y. Mesoporous Silica-coated Gold Nanorods as a Light-mediated Multifunctional Theranostic Platform for Cancer Treatment. Adv. Mater. 2012, 24, 1418-1423. (4) Wang, C.; Xu, L.; Liang, C.; Xiang, J.; Peng, R.; Liu, Z. Immunological Responses Triggered by Photothermal Therapy with Carbon Nanotubes in Combination with Anti-CTLA-4 Therapy to Inhibit Cancer Metastasis. Adv. Mater. 2014, 26, 8154-8162. (5) Song, X.; Wang, X.; Yu, S.; Cao, J.; Li, S.; Li, J.; Liu, G.; Yang, H.; Chen, X. Co9Se8 Nanoplates as a New Theranostic Platform for Photoacoustic/Magnetic Resonance Dual-Modal-Imaging-Guided Chemo-Photothermal Combination Therapy. Adv. Mater. 2015, 27, 3285-3291. (6) Xiao, Q.; Zheng, X.; Bu, W.; Ge, W.; Zhang, S.; Chen, F.; Xing, H.; Ren, Q.; Fan, W.; Zhao, K.; Hua, Y.; Shi, J. A Core/Satellite Multifunctional Nanotheranostic for In Vivo Imaging and Tumor Eradication by Radiation/Photothermal Synergistic Therapy. J. Am. Chem. Soc. 2013, 135, 13041-13048. (7) Begg, A. C.; Stewart, F. A.; Vens, C. Strategies to Improve Radiotherapy with Targeted Drugs. Nat. Rev. Cancer 2011, 11, 239-253. (8) Ma, M.; Huang, Y.; Chen, H.; Jia, X.; Wang, S.; Wang, Z.; Shi, J. Bi2S3-Embedded Mesoporous Silica Nanoparticles for Efficient Drug Delivery and Interstitial Radiotherapy Sensitization. Biomaterials 2015, 37, 447-455.

16 ACS Paragon Plus Environment

Page 17 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

(9) Huang, Y.; Luo, Y.; Zheng, W.; Chen, T. Rational Design of Cancer-Targeted BSA Protein Nanoparticles as Radiosen-sitizer to Overcome Cancer Radioresistance. ACS Appl.Mater. Interfaces 2014, 6, 19217-19228. (10) Wardman, P. Chemical Radiosensitizers for Use in Radiotherapy. Clin. Oncol. UK 2007, 19, 397-417. (11) Cheng, L.; Yuan, C.; Shen, S.; Yi, X.; Gong, H.; Yang, K.; Liu, Z. Bottom-Up Synthesis of Metal-Ion-Doped WS2 Nanoflakes for Cancer Theranostics. ACS nano, 2015, 9, 11090-11101. (12) Chen, L.; Zhong, X.; Yi, X.; Huang, M.; Ning, P.; Liu, T.; Ge, C.; Chai, Z.; Liu, Z.; Yang, K. Radionuclide 131I Labeled Reduced Graphene Oxide for Nuclear Imaging Guided Combined Radio- and Photothermal Therapy of Cancer. Biomaterials 2015, 66, 21-28. (13) Yong, Y.; Cheng, X.; Bao, T.; Zu, M.; Yan, L.; Yin, W.; Ge, C.; Wang, D.; Gu, Z.; Zhao, Y. Tungsten Sulfide Quantum Dots as Multifunctional Nanotheranostics for In Vivo Dual-Modal Image-Guided Photothermal/Radiotherapy Synergistic Therapy. ACS nano, 2015, 9, 12451-12463. (14) Fan, W.; Shen, B.; Bu, W.; Chen, F.; He, Q.; Zhao, K.; Zhang, S.; Zhou, L.; Peng, W.; Xiao, Q.; Ni, D.; Liu, J.; Shi, J. A Smart Upconversion-Based Mesoporous Silica Nanotheranostic System for Synergetic Chemo-/Radio-/Photodynamic Therapy and Simultaneous MR/UCL Imaging. Biomaterials, 2014, 35, 8992-9002. (15) Yang, K.; Xu, H.; Cheng, L.; et al. In Vitro and In Vivo Near-Infrared Photothermal Therapy of Cancer Using Polypyrrole Organic Nanoparticles. Adv.

17 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 25

Mater. 2012, 24, 5586-5592. (16) Su, Y.; Wei, X.; Peng, F.; Zhong, Y.; Lu, Y.; Su, S.; Xu, T.; Lee, S.; He, Y. Gold Nanoparticles-Decorated Silicon Nanowires as Highly Efficient Near-Infrared Hyperthermia Agents for Cancer Cells Destruction. Nano Lett. 2012, 12, 1845-1850. (17) Yin, W.; Yan, L.; Yu, J.; Tian, G.; Zhou, L.; Zheng, X.; Zhang, X.; Yong, Y.; Li, J.; Gu, Z.; Zhao, Y. High-Throughput Synthesis of Single-Layer MoS2 Nanosheets as a Near-infrared Photothermal-Triggered Drug Delivery for Effective Cancer Therapy. ACS nano 2014, 8, 6922-6933. (18) Wang, J.; Yan, R.; Guo, F.; Yu, M.; Tan, F.; Li, N. Targeted Lipid-Polyaniline Hybrid Nanoparticles for Photoacoustic Imaging Guided Photothermal Therapy of Cancer. Nanotechnology 2016, 27, 285102. (19) Zhou, J.; Lu, Z.; Zhu, X.; Wang, X.; Liao, Y.; Ma, Z.; Li, F. NIR Photothermal Therapy Using Polyaniline Nanoparticles. Biomaterials 2013, 34, 9584-9592. (20) Xu, S.; Li, D.; Wu, P. One-pot, Facile, and Versatile Synthesis of Monolayer MoS2/WS2 Quantum Dots as Bioimaging Probes and Efficient Electrocatalysts for Hydrogen Evolution Reaction. Adv. Funct. Mater. 2015, 25, 1127-1136. (21) Hsiao, C. W.; Chuang, E. Y.; Chen, H. L.; Wan, D.; Korupalli, C.; Liao, Z. X.; Chiu, Y. L.; Chia, W. T.; Liu, K. J.; Sung, H. W. Photothermal Tumor Ablation in Mice with Repeated Therapy Sessions Using NIR-absorbing Micellar Hydrogels Formed in situ. Biomaterials, 2015, 56, 26-35. (22) Cabral, H.; Matsumoto, Y.; Mizuno, K.; Chen, Q.; Murakami, M.; Kimura, M.; Terada, Y.; Kano, M. R.; Miyazono, K.; Uesaka, M.; Nishiyama, N.; Kataoka, K.

18 ACS Paragon Plus Environment

Page 19 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

Accumulation of Sub-100 nm Polymeric Micelles in Poorly Permeable Tumours Depends on Size. Nat. Nanotechol. 2011, 6, 815-823. (23) Goldberg, S. N.; Gazelle, G. S.; Mueller, P. R. Thermal Ablation Therapy for Focal Malignancy: a Unified Approach to Underlying Principles, Techniques, and Diagnostic Imaging Guidance. AJR Am. J. Roentgenol. 2000, 174, 323-331. (24) Kinner, A.; Wu, W.; Staudt, C.; Iliakis, G. γ-H2AX in Recognition and Signaling of DNA Double-Strand Breaks in the Context of Chromatin. Nucleic Acids Res. 2008, 36, 5678-5694. (25) Zhang, Z.; Wang, J.; Chen, C. Near-Infrared Light-Mediated Nanoplatforms for Cancer Thermo-Chemotherapy and Optical Imaging. Adv. Mater. 2013, 25, 3869-3880. (26) Choi, H. S. Nanoparticle Assembly: Building Blocks for tumour Delivery. Nat Nano. 2014, 9, 93-94. (27) Cheng, L.; Liu, J.; Gu, X.; Gong, H.; Shi, X.; Liu, T.; Wang, C.; Wang, X.; Liu, G.; Xing, H.; Bu, W.; Sun, B.; Liu, Z. PEGylated WS2 Nanosheets as a Multifunctional Theranostic Agent for in vivo Dual-Modal CT/Photoacoustic Imaging Guided Photothermal Therapy. Adv. Mater. 2014, 26, 1886-1893. (28) Gordijo, C. R.; Abbasi, A. Z.; Amini, M. A.; Lip, H. Y.; Maeda, A.; Cai, P.; O'Brien, P. J.; DaCosta, R. S.; Rauth, A. M.; Wu, X. Y. Design of Hybrid MnO2-Polymer-Lipid Nanoparticles with Tunable Oxygen Generation Rates and Tumor Accumulation for Cancer Treatment. Adv. Funct. Mater. 2015, 25, 1858-1872. (29) Fan, W.; Bu, W.; Shen, B.; He, Q.; Cui, Z.; Liu, Y.; Zheng, X.; Zhao, K.; Shi, J.

19 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 25

Intelligent MnO2 Nanosheets Anchored with Upconversion Nanoprobes for Concurrent pH-/H2O2-Responsive UCL Imaging and Oxygen-Elevated Synergetic Therapy. Adv. Mater. 2015, 27, 4155-4161.

20 ACS Paragon Plus Environment

Page 21 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

Figures

Figure 1. (a) Synthetic process diagram of MoS2@PANI nanohybrids. (b) TEM image of the as-made MoS2 QDs. (c) TEM image of the as-prepared MoS2@PANI nanohybrids. (d) Size distribution of MoS2 QDs and MoS2@PANI nanohybrids. (e) Fluorescence spectra of MoS2@PANI nanohybrids. (f) UV-vis-NIR absorbance spectra of MoS2 QDs and MoS2@PANI nanohybrids. (g) The temperature and photothermal images of pure water, MoS2 QDs, pure PANI, and MoS2@PANI nanohybrids (0.1 mg ml−1) solutions as a function of 808 nm laser irradiation time at the power intensity of 1.5 Wcm−2.

21 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 25

Figure 2. (a) Cell viabilities of 4T1 cells treated with PBS, MoS2, or MoS2@PANI with or without laser irradiation (808 nm, 1.5 W cm-2) and X-ray radiation (6 Gy). Results show the mean of the measurements conducted in triplicate ± standard deviation, *P < 0.05, **P < 0.01. (b) Live-dead staining images of 4T1 cells treated with PBS, MoS2@PANI + PTT, MoS2@PANI + RT, MoS2@PANI + PTT + RT, respectively.The nanoparticle concentration was 0.1mg mL−1, the NIR laser was 808 nm, 1.5 W cm-2, 5 min, and the RT irradiation dose was 6 Gy. (c) Fluorescence images of 4T1 cells incubated with MoS2 QDs, MoS2@PANI nanohybrids, and MoS2@PANI nanohybrids under 808 nm NIR irradiation.(d) Representative fluorescence images of DNA fragmentation and nuclear condensation induced by PBS or MoS2@PANI nanohybrids (0.1 mg ml−1, 2 mL) and/or 808 nm laser irradiation (1.5 W cm-2, 5 min) and/or X-ray radiation (6Gy), stained with DAPI and γ-H2AX for nuclear visualization and DNA fragmentation, respectively. 22 ACS Paragon Plus Environment

Page 23 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

Figure 3. (a) The scheme of MoS2@PANI nanohybrids used for dual modal imaging and combined PTT and RT therapy. (b) Ultrasound (US) images and PA images of 4T1 tumors after intravenously injected with MoS2@PANI nanohybrids at different time points. (c) In vivo CT images of 4T1 tumor-bearing mice before and 8 h after intravenous injection with MoS2@PANI nanohybrids. (d) In vivo CT images of tumors on mice before and 8 h after intravenous injection with MoS 2@PANI nanohybrids. (e) Corresponding intensity of the photoacoustic signal of MoS2@PANI nanohybrids in the tumor at different time points. (f) Corresponding HU value of MoS2@PANI nanohybrids in the tumor before injection and 8 h after injection. 23 ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 25

Figure 4. (a) Photothermal images of 4T1 tumor-bearing mice under the 808 nm laser irradiation (1.5 W cm-2, 5 min) after administration of PBS, MoS2 QDs and MoS2@PANI nanohybrids, respectively. (b) Tumor growth in different groups of mice after various treatments. (c) Representative immunofluorescence images of tumor slices. The nuclei, blood vessels, and hypoxic areas were stained with DAPI (blue), anti-CD31 antibody (red), and anti-pimonidazole antibody (green), respectively. (d) Quantifcation of hypoxia areas in the tumors from different groups’ mice. **P < 0.01. (e) Histological H&E staining for different organs collected from mice treated with PBS or MoS2@PANI nanohybrids under NIR laser irradiation (808 nm, 1.5 W cm-2) and X-ray radiation (6 Gy).

24 ACS Paragon Plus Environment

Page 25 of 25

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Applied Materials & Interfaces

Graphic figure

ACS Paragon Plus Environment