Ultrasmall Semimetal Nanoparticles of Bismuth for Dual-Modal

Apr 10, 2017 - ... 800 Dongchuan Road, Shanghai 200240, People's Republic of China .... Muhammad Waleed Baig , Muhammad Zia , Muhammad Ali , Zabta ...
0 downloads 0 Views 2MB Size
Subscriber access provided by University of Newcastle, Australia

Article

Ultra-Small Semimetal Nanoparticles of Bismuth for Dual-Modal Computed Tomography/Photoacoustic Imaging and Synergistic Thermoradiotherapy Xujiang Yu, Ang Li, Chengzhi Zhao, Kai Yang, Xiaoyuan Chen, and Wanwan Li ACS Nano, Just Accepted Manuscript • DOI: 10.1021/acsnano.7b00476 • Publication Date (Web): 10 Apr 2017 Downloaded from http://pubs.acs.org on April 11, 2017

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 Nano 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 41

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 Nano

Ultra-Small Semimetal Nanoparticles of Bismuth for Dual-Modal Computed Tomography/Photoacoustic Imaging and Synergistic Thermoradiotherapy Xujiang Yu,1 Ang Li,1 Chengzhi Zhao,1 Kai Yang,2,* Xiaoyuan Chen3 and Wanwan Li1,* [*]

Prof. W. Li, Dr. X. Yu, A. Li

State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China E-mail: [email protected] [*]

Prof. K. Yang

School of Radiation Medicine and Protection (SRMP) and School of Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China E-mail: [email protected] Prof. X. Chen

ACS Paragon Plus Environment

1

ACS Nano

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 41

Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA

ABSTRACT: Multifunctional nanomaterials with integrated diagnostic and therapeutic functions, combination therapy to enhance treatment efficacy, as well as low toxicity have drawn tremendous attentions. Herein, we report a multifunctional theranostic agent based on peptide (LyP-1) labeled ultra-small semimetal nanoparticles of bismuth (Bi-LyP-1 NPs). Ultra-small Bi NPs (3.6 nm) were facilely synthesized using oleylamine as the reducing agent and exhibited a higher tumor accumulation after conjugated with the tumor-homing peptide (LyP-1). The abilities of absorbing both ionizing radiation and the second near-infrared (NIR-II) window laser radiation

ensured

Bi-LyP-1

NPs

being

capable

for

dual-modal

computed

tomography/photoacoustic imaging and efficient synergistic NIR-II photothermal/radiotherapy of tumors. Moreover, Bi-LyP-1 NPs could be rapidly cleared out of mice through both renal and fecal clearance, and almost completely cleared after 30 days. Such multifunctional nanoparticles as efficient cancer theranostic agents, coupled with the fast clearance and low toxicity shed light on future use of semimetal nanoparticles for biomedicine.

KEYWORDS: Semimetal nanoparticles; bismuth; the second near-infrared (NIR-II) window; multimodal imaging; thermoradiotherapy

With the advanced progress achieved in nanotechnology and biomedicine, various multifunctional nanomaterials has been exploited for uses in cancer diagnosis and therapy.1−4 The ability to embrace multiple modalities in a single nanostructure, or one particle (referred to

ACS Paragon Plus Environment

2

Page 3 of 41

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 Nano

as nanotheranostics),5,6 enables multifunctional nanomaterials to provide uses in cancer imaging, simultaneous diagnosis, and individualized treatment.7 Various multifunctional nanomaterials that have been developed based on intelligent designs to combine different functional nanoparticles8−10 or to utilize certain individual nanoparticles11,12 for multiple applications have made great progress in the field of nanomedicine. However, because there is a risk of toxicity that could be induced by the accumulation of nanomaterials in the body, the use of nanomaterials for further biomedical applications may be limited.13,14 Thus, nanomaterials with smaller size that allow for rapid clearance from the body would be of significant value for use in the clinic.15−17 Moreover, synergistic therapeutic approaches,3,18 in addition to multi-modal imaging provided by a single system19−21 are critical for the development of highly efficient cancer theranostics. Radiotherapy (RT) is an extensively used approach to treat cancer in clinical medicine. However, detrimental effects on normal tissues22 and the hypoxic nature23 of tumors remain to be the two primary issues with this treatment option that limit its efficacy and applications. Recently, nanomaterials with high Z-elements (e.g. gold,24,25 iodine,26 bismuth27 and rare earth elements28,29) have been demonstrated to function as excellent radiosensitizers that are able to concentrate higher radiation doses within tumors,30 thus enhancing RT efficacy while reducing possible side effects associated with the treatment.31 Meanwhile, approaches that can be utilized with RT to increase cancer treatment efficacy have also been explored.32−34 Interestingly, mild hyperthermia has been demonstrated to improve oxygen levels within tumors by increasing the blood flow, and thus enhancing the tumor killing ability of this treatment when used in conjunction with radiotherapy.23,35,36

ACS Paragon Plus Environment

3

ACS Nano

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 41

Photothermal therapy (PTT) is a well-known treatment used in cases of hyperthermia. This treatment works through the use of nanomaterials with high near-infrared (NIR) absorbance properties that enable them to generate heat to induce tumor ablation under laser irradiation.37 This treatment is minimally invasive and possesses a high therapeutic efficacy, enabling photothermal therapy to be applied either alone or in combination with other treatments,38,39 such as radiotherapy.35 The use of a second near-infrared (NIR-II, 1000−1350 nm) window laser, particularly within 1000−1100 nm, allows deeper penetration and higher maximum permissible exposure (MPE),40 which could dramatically increase photothermal efficacy. However, prior research has focused primarily on employing first near-infrared (NIR-I, 700−950 nm) window laser as the excitation light source.41,42 This is likely due to the strong absorption band within the NIR-I window, but negligible absorption within the NIR-II window of traditional nanomaterials (e.g. organic compounds,43 gold nanostructures,44,45 or nanocarbons46,47). Fortunately, increased attention devoted to this area has enabled the exploration of several kinds of nanomaterials as potential photothermal nanomaterials that respond to the NIR-II window, including transition metal sulfide/oxide semiconductors,48−51 noble metal nanomaterials,52−54 and polymer nanocomposites.55 By carefully tuning doping manners or adjusting stoichiometric ratios, semiconductors can exhibit very different optical bandgaps and free carrier concentrations, characteristics that strongly contribute to semiconductor absorption in the near-infrared region.56 It has recently been reported that semimetal nanomaterials of antimony exhibit both broad and strong absorption characteristics in the range of 700−1350 nm.57 This could potentially be due to their higher charge carriers compared to semiconductors, which increases their value as agents for photothermal therapy in NIR windows.

ACS Paragon Plus Environment

4

Page 5 of 41

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 Nano

Another typical semimetal element, bismuth (Bi), has attracted much interest owing to its properties (e.g. small carrier effective masses, a long Fermi wavelength, and a small band overlap energy). The recent discovery of plasmonic properties that originate from the semimetalto-semiconductor transition (also called as nano-confinement effects) in bismuth at the nanoscale58 has broadened the possible applications of Bi-based nanomaterials (traditionally used as thermoelectric materials, catalysts and sensors59) for use in biomedicine as cancer therapeutic agents.21 Moreover, research suggests that the Bi element could also function as a radiosensitizer and a contrast agent for computed tomography (CT).60,61 These potential biomedical applications motivated us to develop ultra-small bismuth nanoparticles to be used as multifunctional agents for efficient cancer theranostics. To our best knowledge, no studies concerning both of photothermal and radiation properties of Bi nanoparticles, their use in multimodal imaging, and their further development for biomedical applications have been reported so far. Herein, a theranostic agent was developed based on the semimetal nanoparticles of Bi, which exhibit strong absorption properties in the NIR-II window, suitable for both dual-modal CT/photoacoustic (PA) imaging and the synergistic PTT/RT treatment of tumors (Figure 1a). First, a facile method for ultra-small Bi nanoparticles was proposed using oleylamine (OAm) as both of the reducing and the stabilizing agent. PEGylated Bi nanoparticles (NPs) were found to exhibit a broad absorption from ultraviolet to NIR region, and were capable of acting as an excellent photothermal agent when irradiated with a 1064 nm laser, which possessed high photothermal conversion efficiency and photostability. Once labeled with a peptide (CGNKRTRGC, LyP-1), the resulting nanoparticles (Bi-LyP-1 NPs) were able to present both high cellular uptake and tumor accumulation, and also exhibited a potential for use as a CT and PA imaging contrast agent in vivo, due to the absorption ability of both X-ray and NIR light of

ACS Paragon Plus Environment

5

ACS Nano

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 41

Bi. Strong synergistic effects were also observed on tumor-bearing mice when Bi-LyP-1 NPs were used in combination with PTT and RT. It should be noted that Bi-LyP-1 NPs exhibited excellent in vivo biocompatibilities, low long-term toxicity, and were found to be rapidly cleared via both liver and renal clearance mechanisms with stable morphology. The successful demonstration of utilizing Bi nanoparticles as efficient agents for cancer diagnostics and therapy from our findings shed light on future use of Bi nanoparticles and other multifunctional nanomaterials based on semimetals for cancer theranostics. RESULTS AND DISCUSSION Synthesis and Characterization of Bi-LyP-1 NPs. Inorganic Bi nanoparticles were first synthesized through a facile organic protocol. Upon injection of the bismuth precursor into hot oleylamine under nitrogen, Bi3+ was gradually reduced by the mild reducibility of oleylamine into ultra-small nanoparticles, and were stabilized by oleylamine molecules as well. Following a 10 min incubation at 260 oC, high crystalline semimetal Bi NPs were yielded with a diameter of about 3.6 nm, and the clear lattice fringe of Bi NPs with the interplanar d spacing of 0.139 nm was revealed by high resolution transmission electron microscope (HRTEM) (Figure 1b). The characteristic X-ray power diffraction (XRD) pattern further demonstrated the presence of Bi NPs with a pure rhombohedral phase (JCPDS No. 44−1246) (Figure 1c). This facile method which used the common chemicals, bismuth acetate, oleic acid, and oleylamine exhibited great advantages over other reported methods that required precursors specially prepared through multiple steps,62,63 such as Bi[N(SiMe3)2]3. Next, the as-synthesized nanoparticles were mixed with

1,2-diastearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene

glycol)]

(Molecular weight 5000, DSPE-PEG5000) containing 15% maleimide terminal groups to yield hydrophilic PEGylated Bi NPs. In order to improve tissue-specificity, PEGylated Bi NPs were

ACS Paragon Plus Environment

6

Page 7 of 41

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 Nano

labeled with a cyclic peptide (CGNKRTRGC, LyP-1), which possesses the ability to specifically target p32 proteins expressed by 4T1 mouse breast cancer cells64 through a thioether reaction65 between the maleimide and the N-terminal cysteine moiety. The resulting nanoparticles (Bi-LyP1 NPs) were found to be conjugated with approximately 3.6 peptides on each nanoparticle (Figure S1). Bi-LyP-1 NPs showed a hydrodynamic size of about 12 nm and exhibited excellent stability in different physiological solutions (Figure S2a,b). The surface modification induced negligible change in the morphology of Bi NPs as monitored by TEM (Figure S2c). Moreover, the successful coupling of DSPE-PEG5000 and conjugation of LyP-1 peptides on Bi NPs were confirmed by the FTIR spectra and NMR spectra. The intensities of both C−H and C−O stretch bands (2896 cm−1 and 1101 cm−1) increased distinctly after coupling, which were the two intense peaks of DSPE-PEG5000 (Figure S3a,b), further studies on the NMR spectrum of PEGylated Bi NPs demonstrated the existence of malemide (6.69 ppm) and methoxy (3.37 ppm) groups on the surface of Bi NPs (Figure S4a). The appearance of a broader and stronger N−H stretch band (3351 cm−1), as well as a stronger C=O stretch band (1667 cm−1) in the FTIR spectrum of BiLyP-1 NPs was attributed to ten amino groups in each peptide (Figure S3c), indicating the conjugation of LyP-1 peptide to Bi NPs, which could also be inferred from the disappearance of maleimide peak and appearance of amino hydrogen peak at 2.85 ppm in the NMR spectrum of Bi-LyP-1 NPs (Figure S4b).66

ACS Paragon Plus Environment

7

ACS Nano

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 41

Figure 1. Scheme and characterization. a) A scheme of multifunctional Bi NPs. b) TEM images and HRTEM images of as-synthesized Bi NPs. Insert histogram shows the measured diameters of as-synthesized Bi NPs. c) XRD pattern of as-synthesized Bi NPs. d) The absorbance spectrum of PEGylated Bi NPs. e) Temperature changes of PEGylated Bi NPs at different concentrations under a 5 min irradiation from a 1064 nm laser at 1W cm−2. f) Temperature profile of PEGylated Bi NP solution (120 ppm of Bi) under heating with laser on and then cooling with laser off. g) Calculation of the time constant for heat transfer using a linear regression of the cooling profile.

ACS Paragon Plus Environment

8

Page 9 of 41

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 Nano

h) Temperature variations of PEGylated Bi NP solution (200 ppm of Bi) over 6 cycles of heating and natural cooling. In Vitro Photothermal Performance. The PEGylated Bi NPs exhibited a strong, broad absorbance band between 200 and 1100 nm, with no obvious sharp peaks (Figure 1d), and this observation prompted us to use the common 1064 nm laser as a source to trigger NIR-II photothermal therapy. Next, we evaluated the photothermal properties of PEGylated Bi NPs by testing the temperature variation of each nanoparticle solutions with different concentrations under 1064 nm laser irradiation. We found that when irradiated at a fixed power density, the photothermal heating effect of PEGylated Bi NPs was concentration-dependent (Figure 1e). The solution temperature was increased by about 17.2 oC at 200 ppm of Bi, while the temperature increase of pure water was only about 2.8 oC. Photothermal conversion efficiency (η) is an important parameter used to assess photothermal agents. The η value in our work was calculated to be 32.2% with 1064 nm laser irradiation (Figure 1f,g). This was calculated according to the method established by Roper and co-workers,67 and indicates good photothermal properties of PEGylated Bi NPs. When the concentration was fixed (200 ppm of Bi), the temperature rise was dependent on the power density of laser, with the variation of approximately 9.2 oC obtained at 0.6 W cm−2 (Figure S5a,b). Photothermal stability of Bi nanoparticles was examined further with a 5 min, 1064 nm laser irradiation of 1 mL PEGylated Bi NP solution (200 ppm of Bi) at 1 W cm−2, and then leaving it to cool down to environment temperature. The above cycle was repeated for a total of six times, and the consistency of temperature changes observed (Figure 1h), proving the good photostability properties of PEGylated Bi NPs upon irradiation. This observation was further supported by TEM images, XRD pattern, and absorption spectrum following the six-cycle irradiation of PEGylated Bi NPs (Figure S6).

ACS Paragon Plus Environment

9

ACS Nano

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 41

In Vitro Cytotoxicity Assay. Prior to the use of Bi-LyP-1 NPs for tumor targeting, and imageguided thermoradiotherapy, we investigated interactions of nanoparticles with cells. A standard cell counting kit-8 (CCK-8) assay was used to test the cytotoxicity of Bi-LyP-1 NPs after different concentrations of the Bi-LyP-1 NP solution were incubated with 4T1 cancer cells and hepatic L02 cells for 24 h, respectively (Figure 2a). No distinct cytotoxicity to both cancer and normal cells was observed after 24 h incubation with the Bi-LyP-1 NPs in our tested dose range, it is noteworthy that the relative viabilities of 4T1 cells (81%) was slightly lower than that of hepatic L02 cells (90%) at 400 µg mL−1, which could be attributed to the higher concentration of Bi-LyP-1 NPs accumulated in 4T1 cells than in L02 cells owing to the highly expressed p32 proteins (the receptor of LyP-1) of 4T1 cancer cells compared to L02 normal cells.61 The 48 h relative viabilities of 4T1 cell and L02 cells incubated Bi-LyP-1 NP solution at the same concentrations were also investigated and the similar result of low cytotoxicity was obtained (Figure S7). In Vitro Cellular Uptake. Cellular uptakes of peptide-labeled and non-labeled Bi NPs were then measured to quantify cellular uptake efficacy of NPs enhanced by LyP-1 peptides after internalized with 4T1 cancer cells for different internalization times. As shown in Figure 2b and Figure S8, the uptake efficacy of Bi-LyP-1 NPs increased gradually before reaching saturation at about 6 h, while that of non-labeled Bi kept nearly unchanged after about 5 h. The uptake efficacy of LyP-1-labeled Bi NPs was greater than that of non-labeled Bi NPs at each time point, with an approximately 2.4-fold greater cellular uptake after 6 h incubation. Such saturable and time-dependent uptake behaviors by 4T1 cells indicated the cell endocytosis of nanoparticles, and the additional cellular uptake of LyP-1-labeled Bi NPs was attributed to the homing

ACS Paragon Plus Environment

10

Page 11 of 41

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 Nano

specificity of LyP-1 peptides to the highly expressed p32 receptors at the cell surface.61 These results indicate an enhanced specificity of LyP-1-labeled Bi NPs for 4T1 cancer cells. In Vitro Radiation Therapy. Upon demonstrating the capability of Bi NPs to trigger PTT, we carried out an in vitro clonogenic survival assay to determine whether Bi NPs had the ability to enhance RT efficacy. After cultured in the presence or absence of Bi-LyP-1 NPs for 24 h, 4T1 cells were irradiated by X-ray at 0, 2, 4, 6 and 8 Gy, respectively. The results of clonogenic survival assay (Figure 2c) showed Bi-LyP-1 NPs (10 ppm of Bi) could remarkably enhance RT efficacy, and the inhibitory effect was dependent on X-ray irradiation doses. Owing to the excellent photoelectric effect and Compton scattering of Bi, the surviving fraction (SF) of cells treated with nanoparticles at 4 Gy was found to be approximately 0.107, while the surviving fraction of the untreated cells was 0.219. Survival fractions were also fitted by a single-target model formula (Table 1), with the SF2 under 2 Gy X-ray irradiation calculated to be 0.602 for untreated 4T1 cells and 0.415 for cells treated with Bi-LyP-1 NPs (10 ppm of Bi). The sensitizing enhancement ratio of SERD0 was calculated to be 1.218 for 4T1 cells treated with BiLyP-1 NPs (10 ppm of Bi), which was similar to that of the reported WS2 QDs while with a much higher concentration (100 ppm of W) (1.22).68 Moreover, the SER10 of Bi-LyP-1 NPs reached up to 1.248, and this value was higher than that of Au NPs (1.19).25,34 It is clear that BiLyP-1 NPs demonstrate a significant radiosensitization effect, even at a low concentration and a low dose of radiation.

Table 1. The calculated parameter values and fitted formula based on the radiosensitization activity of Bi-LyP-1 NPs in 4T1 cells.

ACS Paragon Plus Environment

11

ACS Nano

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 41

D0

Dq

SF2

SERD0

SER10

Fitted formula

X-ray

1.550

1.628

0.602





SF=1− (1−e−0.645*D)2.858

Bi-LyP-1+ Xray

1.273

1.230

0.415

1.218

1.248

SF=1− (1−e−0.786*D)2.637

Figure 2. Cell experiments, blood circulation and biodistribution of Bi-LyP-1 NPs in mice. a) Relative cell viabilities were evaluated with a CCK-8 assay. Both of 4T1 cells and L02 cells were incubated with different concentrations of Bi-LyP-1 NPs for 24 h. b) Cellular uptake of BiLyP-1 NPs and PEGylated Bi NPs by 4T1 cells following a 6 h incubation. c) The inhibitory effect of Bi-LyP-1 NPs (0 or 10 ppm of Bi) on 4T1 cells under various irradiation doses. d) Blood circulation of Bi-LyP-1 NPs in mice by detecting the percentage of Bi remained in blood among injected dose at different time points. e) Biodistribution of Bi-LyP-1 NPs and PEGylated Bi NPs in mice after 24 h circulation. f) Calculated ratios of Bi-LyP-1 NPs relative to PEGylated Bi NPs in the main tissues (*p < 0.05, **p < 0.01).

ACS Paragon Plus Environment

12

Page 13 of 41

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 Nano

In Vivo Blood Circulation. Motivated by these exciting in vitro experimental results, we moved forward to carry out in vivo studies. Specifically, the pharmacokinetic behavior of BiLyP-1 NPs in mice was studied in order to determine the blood circulation half-life. For these studies, mice were intravenously (i.v.) injected with Bi-LyP-1 NPs (3 mg mL−1, 200 µL), followed by collecting blood samples at nine time points in order to measure the amount of Bi by ICP-MS. The half-life of NPs circulated in blood was determined to be approximately 3.97 h (Figure 2d). The blood content of Bi-LyP-1 NPs was found to remain steady at a relatively low level following 24 h of circulation. This could be due to the ultra-small size and surface modifications of Bi NPs. In Vivo Tumor Accumulation. In addition, the biodistribution of both the peptide-labeled and non-labeled Bi NPs (3 mg mL−1, 200 µL) was studied in 4T1 tumor-bearing mice at 24 h following administration (Figure 2e). Both Bi NPs accumulated at high levels in the spleen, liver and kidneys, while at rather low levels in the lungs and heart. A similar tissue distribution pattern was observed for both nanoparticles in these organs (Figure 2f), indicating no obvious influence of the peptide modification on distribution in mice. However, Bi-LyP-1 NPs did exhibit a remarkable 1.7-fold enhancement in tumor accumulation (6.26 ± 0.33% ID/g) in contrast to that of PEGylated Bi NPs (3.66 ± 0.48% ID/g). These results correlate with those obtained in in vitro cellular experiments, demonstrating the successful modification of peptide LyP-1 on nanoparticles, and the clear enhancement of tissue specificity observed with Bi-LyP-1 NPs. In Vitro and in Vivo X-ray Computed Tomography. Prior to using Bi-LyP-1 NPs for cancer treatment, we aimed to understand their efficacy as dual-modal CT and PA imaging agents on mice. Although bismuth has been proposed as an excellent CT contrast element, and several Bibased nanoparticles have been synthesized for this reason,64,69−71 no research examining Bi

ACS Paragon Plus Environment

13

ACS Nano

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 41

nanoparticles alone has yet to be reported. We first studied the in vitro Hounsfield units (HU value) of Bi-LyP-1 NPs in PBS (pH=7.4), with iohexol used as control (Figure 3a,b). The results revealed that Bi exhibited a significantly higher slope of HU value (approximately 13.8 HU mM−1) than iohexol (approximately 4.28 HU mM−1). These results indicate the excellent CT signal enhancement capability of Bi-LyP-1 NPs. We next carried out further studies aimed at investigating in vivo CT imaging (Figure 3c). Here, CT imaging were conducted on mice with intratumoral (i.t.) injection of Bi-LyP-1 NPs (6 mg mL−1, 30 µL) both prior to and 5 min after administration. Upon injection of the nanoparticles, an obvious tumor contrast was observed, with the highest increase of HU value from 130.5 to 1193.6. This observed increase suggests that Bi-LyP-1 NPs alone could be utilized as highly effective CT contrast agents. In Vivo Photoacoustic Imaging. As a widely used, non-invasive biomedical modality, PA imaging employs laser light to acoustically visualize biological tissues. This technique is advantageous in that it can provide high imaging depth and spatial resolution.72 Because the properties of Bi nanoparticles enable a strong and broad NIR absorption, we conducted PA imaging in mice administrated with Bi-LyP-1 NPs (6 mg mL−1, 30 µL) via i.t. injection at different wavelengths. A high PA signal intensity was observed in tumors in mice following the i.t. injection, with an increase of approximately 3.3-fold when exposed to 700 nm wavelength laser irradiation (Figure 3d,e). PA imaging in mice administrated with Bi-LyP-1 NPs (6 mg mL−1, 200 µL) via i.v. injection was carried out at various time points post-administration (Figure 3f,g). From these studies, the PA signal intensity of tumors was found to be increased approximately 3.6-fold at 8 h post i.v. injection under irradiation of a 700 nm wavelength laser source. Similar results were also observed with 800 nm and 900 nm irradiation (Figure S9a-c). These results not only coincided with a high accumulation of Bi-LyP-1 NPs in tumors (Figure

ACS Paragon Plus Environment

14

Page 15 of 41

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 Nano

3h), but also indicated the great promise Bi NPs hold as highly efficient contrast agents for PA imaging guided cancer treatment under a wide range of NIR wavelengths.

Figure 3. Dual-modal CT and PA imaging both in vitro and in vivo with Bi-LyP-1 NPs. a) HU values of various concentrations of Bi-LyP-1 NPs and iohexol solutions. b) X-ray CT images of Bi-LyP-1 NPs and iohexol solutions of corresponding concentrations. c) X-ray CT images of tumors obtained prior to and 5 min after administration of Bi-LyP-1 NPs. d) PA images and e) PA signal intensities of tumors obtained before and after administration of Bi-LyP-1 NPs at 700

ACS Paragon Plus Environment

15

ACS Nano

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 41

nm. f) PA images and g) PA signal intensities of tumors obtained at each time point after administration of Bi-LyP-1 NPs at 700 nm. h) A photograph of the experimental mice for PA imaging taken at 8 h post i.v. injection. (**p < 0.01). NIR-II Photothermal and Radiotherapy in Vivo. After successfully demonstrating of the photothermal properties of NIR-II, we utilized RT enhancement and dual-modal imaging of BiLyP-1 NPs for further studies. The in vivo PTT and RT combined treatment of subcutaneous mouse tumor model was used to study six different treatment groups. Using IR imaging (Figure 4a), we first studied the surface temperature of tumors in mice with i.v. injection of Bi-LyP-1 NPs (3 mg mL−1, 200 µL) that were irradiated by the 1064 nm laser at 0.6 W cm−2. We showed that the surface temperature of these tumors rapidly increased to approximately 45 oC and then stabilized, while that of tumors injected with PBS was found to slowly increase to approximately 39.5 oC (Figure 4b). However, we found that this mild photothermal treatment alone in mice injected with Bi-LyP-1 NPs resulted only in a delay in the growth of tumors (Figure 4c). Similar results were also observed in mice exposed to X-ray treatment, with a pre-injection of either PBS or Bi-LyP-1 NPs. In these cases, tumors from both treatment groups were observed to continue to gradually grow during the rest time points due to the low dose X-ray treatment. We demonstrate that when combined together with Bi-LyP-1 NPs, X-ray irradiation was capable to inhibit the tumor growth more effectively (Figure 4c). These results clearly demonstrate the ability of Bi-LyP-1 NPs in enhancing RT efficacy. In remarkable contrast to the limited therapeutic effect observed for PTT alone (Bi-LyP-1 NPs with laser) or RT alone (Bi-LyP-1 NPs without laser), impressive results on the inhibition of tumor growth were observed when RT and PTT treatments were combined. As shown in Figures 4c,d, the growth of tumors in mice i.v. injected with Bi-LyP-1 NPs was completely inhibited following exposure to both a 1064 nm

ACS Paragon Plus Environment

16

Page 17 of 41

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 Nano

wavelength laser and X-ray irradiation. These results clearly indicate the positive synergistic effect of PTT and RT therapies on the inhibition of tumor growth.

Figure 4. Combined treatment of tumors with Bi-LyP-1 NPs. a) IR images of mice under laser irradiation after administrated with Bi-LyP-1 NPs and PBS, respectively. b) Corresponding

ACS Paragon Plus Environment

17

ACS Nano

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 41

temperature variation of tumors in (a). c) The recorded curves on tumor growth in each group. d) A photographs of tumors in each group after 14 days. e) H&E (top) and CD31 staining (bottom) of tumor sections under different treatments (**p < 0.01, ***p < 0.005). In order to better elucidate changes in the internal structures of tumors that underwent different treatment regimens, we performed hematoxylin and eosin (H&E) staining, as well as CD31 staining in tumor slices derived from these experimental groups (Figure 4e). Both significant levels of cell apoptosis and efficient suppression of angiogenesis in tumors were observed only in the PTT and RT combination treatment group. These results correlate with the effective therapeutic results observed in these treatment groups. It should also be noted that excellent therapeutic results were obtained in the case of tumors treated with only 0.6 W cm−2 for the 1064 nm laser excited PTT, and a low dose of 4 Gy for RT. This treatment regimen may lower levels of pain and other side effects throughout the course of the treatment. Moreover, all mice were observed to behave normally (Figure S10), with no death or significant body weight changes observed (Figure S11) throughout the course of treatment. The work presented here demonstrates not only the use of Bi NPs as excellent agents for NIR-II photothermal and radiotherapy treatments, but also the exhibited synergistic effects observed on the targeting of tumors when these therapies are utilized together. Blood Panels, Histology Examinations and Clearance Study in Vivo. Because both the distribution and clearance of NPs, as well as their potential long-term toxicity in vivo are crucial for their development in further biomedical applications, we investigated these properties of BiLyP-1 NPs in mice. For these studies, mice i.v. injected with Bi-LyP-1 NPs were sacrificed at 1, 7 and 30 days following administration (n=5) to measure the amount of Bi in major organs (spleen, liver, kidney, lung and heart). The major organs in which Bi was found to accumulate

ACS Paragon Plus Environment

18

Page 19 of 41

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 Nano

were the spleen and liver, followed by the kidney, while the heart and lungs showed relatively low levels of Bi accumulation (Figure 5a), which agreed with the results obtained in tumorbearing mice (Figure 2e). The levels of Bi-LyP-1 NPs remaining in these organs was found to decrease sharply at 7 d post-injection, with very low Bi levels detected after 30 days. The results from these studies indicate that Bi-LyP-1 NPs can be almost completely cleared from mice. In order to gain further insights into the clearance routes of nanoparticles, urine and feces were collected from mice that had received an i.v. injection of Bi-LyP-1 NPs (3 mg mL−1, 200 µL). From these samples, Bi levels were measured using ICP-MS. The fact that Bi was found to be present in both urine and feces (Figure 5b) suggests that these nanoparticles are cleared through fecal excretion and renal clearance mechanisms. Such clearance mechanisms would align with the observation of a high accumulation of Bi-LyP-1 NPs in the spleen, liver and kidney. Differences in the amount of Bi in feces and urine with time could suggest that the hydrodynamic size plays a role in clearance. It has been reported that nanoparticles greater than 10 nm in size have a tendency to accumulate in RES organs, while those with diameters smaller than 5.5 nm can be easily cleared by kidneys due to the fact that the effective pore size of the renal clearance barrier is approximately 8 nm.27,73 Because the hydrodynamic size of Bi-LyP-1 NPs is in the range of 8-18 nm, we can infer that nanoparticles greater than 10 nm in size are absorbed by the liver and spleen for slow, long-term excretion. Nanoparticles in the range of 810 nm in size are likely to accumulate in the kidney, where they are rapidly cleared via renal filtration. From the variation in the trend of the amount of Bi detected in urine and feces, it is likely that kidney clearance is the primary clearance mechanism on the first day, with fecal excretion gradually taking over as the primary clearance route. This could be explained by a decrease in renal clearance capacity, caused by the gradual accumulation of nanoparticles in

ACS Paragon Plus Environment

19

ACS Nano

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 41

kidney which could diminish the permeability of the merular basement membrane (GBM) and podocytes.74

Figure 5. In vivo behavior and long-term toxicity of Bi-LyP-1 NPs. a) Long-term biodistribution of Bi-LyP-1 NPs in main organs of mice at each time point after administration. b) The detected amount of Bi in feces and urine at each time point after i.v. injection. c). TEM images of feces

ACS Paragon Plus Environment

20

Page 21 of 41

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 Nano

collected at 24 h after i.v. injection. Insert shows a magnified view. d) Blood biochemistry of mice sacrificed at each time point, including alanine minotransferase (ALT), alkaline phosphatase (ALP), amino transferase (AST), blood urea nitrogen (BUN), albumin/globin ratios (A/G), and total protein (TP). e) Hematology data contained eight blood panel parameters of mice sacrificed at each time point. In addition, Bi NPs in urine and feces that were excreted after 24 h were characterized by TEM (Figure 5c and Figure S12). These studies show essentially no change in the sub-spherical morphology of Bi NPs, further indicating the excellent stability properties of Bi-LyP-1 NPs in mice. Further investigations aimed to demonstrate the low long-term toxicology of Bi-LyP-1 NPs at a 2-fold treatment dose (6 mg mL−1, 200 µL) were also carried out. These studies included serum biochemistry and a complete blood panel measurement. As depicted in Figure 5d,e, the serum biochemistry results showed the presence of liver and kidney function markers, while the parameters of complete blood tests were found to be within normal ranges at each time point after administration. Besides, H&E staining of main organs showed no visible inflammation or damage was induced to treated mice compared to untreated mice (Figure S13). With these results, we confirmed that Bi-LyP-1 NPs possess excellent in vivo biocompatibility and low long-term toxicity. CONCLUSION In summary, we show that ultra-small Bi nanoparticles with diameters of approximately 3.6 nm could be facilely synthesized using oleylamine as the stabilizing and reducing agent. As demonstrated by the strong absorbance in the second NIR window, Bi nanoparticles were found to exhibit excellent NIR-II photothermal properties, as well as superior photostability. Following

ACS Paragon Plus Environment

21

ACS Nano

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 41

the modification of the tumor-homing peptide, LyP-1, the cellular uptake efficacy and tumor accumulation of Bi-LyP-1 NPs were about 2.4-fold and 1.7-fold higher than that of non-labeled Bi NPs, respectively. These simple nanostructures (Bi-LyP-1 NPs) exhibited excellent potential as candidate agents for dual-modal CT/PA imaging. Additionally, the capability of Bi-LyP-1 NPs to absorb both ionizing radiation and NIR-II laser radiation, as well their ability to enhance RT efficacy, provides numerous beneficial properties to these nanoparticles. We show that these nanoparticles are able to produce a remarkable, effective synergistic effect of PTT and RT on tumor treatment, even irradiated at 0.6 W cm−2 and 4 Gy, while PTT or RT treatments alone were found to only partly inhibit the growth rate of tumors. Studies on the long-term toxicity and clearance behaviors of Bi-LyP-1 NPs in mice revealed that these nanoparticles are cleared through both renal and fecal clearance mechanisms, with nanoparticles almost completely cleared after 30 days, with no damage or death observed in mice. Such a multifunctional theranostic agent with excellent imaging and therapeutic properties, coupled with its fast clearance and low long-term toxicity demonstrates great promise for the use of semimetal nanoparticles in the field of biomedical science. MATERIALS AND METHODS Materials. Oleic acid and oleylamine were purchased from Aladdin. 1,2-diastearoyl-snglycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)] (Molecular weight 5000, DSPE-PEG5000) was purchased from Nanocs Incorporation. Bismuth (III) acetate was purchased from Sigma-Aldrich. Characterization. TEM images was recorded with a Tecnai G2 electron microscope (Spirit Biotwin, USA), and high resolution TEM (HRTEM) images was obtained with a JEM-2100F

ACS Paragon Plus Environment

22

Page 23 of 41

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 Nano

emission microscope (Japan). XRD pattern was performed using a SmartLab (Cu Kα, λ = 1.5418 angstrom, Japan). FTIR spectra were acquired using a Fourier transform infrared spectrometer (EQUINOX55, Germany). NMR spectra were performed with a Bruker AVANCE III 600 MHz (Germany). Inductively coupled plasma mass spectrometry (ICP-MS) analysis of Bi was conducted on an Agilent 7500a Series (USA). Optical absorbance of Bi nanoparticles was measured using an UV-visible-NIR spectrophotometer (EV300, USA). The 1064 nm laser source for photothermal experiments was generated by Laser Diode Controller (SFOLT. Co. Ltd., China), and thermal images were recorded by a thermal imaging camera E50 (FLIR, USA). Hydrodynamic diameter was tested with a Zetasizer Nano ZS instrument (Malvern, UK). PA imaging was performed using a Vevo LAZR PA system (Vevo® LAZR, VisualSonics, Canada). CT imaging was taken on the eXplore Locus (GE, USA). Synthesis of Ultra-Small Bi Nanoparticles. Typically, 1 mmol bismuth (III) acetate was dissolved in 5 mL oleic acid under vacuum at 100 oC for 1 h, followed by an additional 30 min under dry nitrogen. This precursor solution was then injected into 25 mL of hot oleylamine at 260 oC, with an entire reaction time of 10 min. Following the reaction, chloroform and ethanol were added, followed by centrifugation of the nanoparticles for three times. The final product was stored in chloroform for further use. Synthesis of Bi-LyP-1 Nanoparticles. Cyclic nine amino acid peptide with an N-terminal cysteine moiety (GL Biochem Ltd., 95% purity) was used without further purification. The assynthesized Bi nanoparticles were coated with DSPE-PEG5000 first (PEGylated Bi NPs) by mixing 1 mg Bi nanoparticles with 8.5 mg DSPE-PEG5000-methoxy and 1.5 mg DSPE-PEG5000maleimide in 3 mL chloroform prior to labeling with the LyP-1 peptide. The solution was stirred, and then evaporated under dry nitrogen, followed by centrifugation with 100 kDa ultrafiltration

ACS Paragon Plus Environment

23

ACS Nano

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 41

filters to remove the excess polymers or empty micelles. The resultant dried powder was dissolved in PBS (pH=7.4) under sonication at 70 oC for 5 min. Once the powder was redispersed, 3 mL of 200 µM LyP-1 solution was immediately added and reacted at 25 oC for 4 h with moderate agitation. The reaction product was purified through a 0.2 µm PVDF filter and a 100 kDa ultrafiltration filter and then stored for further use. Quantification of the Number of Peptides on Each Nanoparticle. To determine the density of peptides on each nanoparticle, LyP-1-(fam) peptide that was labeled with a dye of 5(6)carboxyfluorescein (fam) was purchased (95.5% purity, GL Biochem Ltd. China) and then conjugated on PEGylated Bi NPs with the same protocols used for LyP-1 peptides. The 5(6)carboxyfluorescein exhibited a characteristic absorption peak at 497 nm and could be used to quantify the peptide concentration, while the numbers of nanoparticles could be calculated with the data of average diameter from TEM, crystal parameters of XRD pattern, and the concentration of Bi measured with ICP-MS. The typical protocols for LyP-1-(fam) peptides conjugation was kept the same as that of LyP-1 peptides, 1 mg of as-synthesized Bi NPs were first reacted with 8.5 mg DSPE-PEG5000-methoxy and 1.5 mg DSPE-PEG5000-maleimide and then purified with 100 kDa ultrafiltration filters before re-dispersed in 3 mL PBS to conjugate with 3 mL of 200 µM LyP-1-(fam) peptides at 25 oC for 4 h. After purification with 0.2 µm PVDF filters to remove possible agglomerates, washing with ice-cold PBS solution and centrifugation with 100 kDa ultrafiltration filters to get rid of dissociative peptides for several times, a concentrated solution of 1 mL was obtained, with a total of 10 mL of filtrated solution contained non-labeled LyP-1-(fam) peptides. The concentration of Bi was measured with ICPMS, and the concentration of non-labeled LyP-1-(fam) peptides was measured using an UVvisible-NIR spectrophotometer.

ACS Paragon Plus Environment

24

Page 25 of 41

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 Nano

In Vitro Photothermal Performance. To measure photothermal transduction, 1 mL of PEGylated Bi NP aqueous dispersion of various concentrations was added into quartz cuvettes, and then irradiated using a 1064 nm laser source. In vitro temperature variation videos and thermal images versus time were recorded using the E50 camera. In Vitro Cytotoxicity Assay and Cellular Uptake. Both 4T1 cancer cells and hepatic L02 cells (7 × 103 cells/well) were grown in 96-well plates under standard conditions (37 oC, 5% CO2) with 90% RPMI 1640 medium and 10% fetal bovine serum (FBS, Sigma-Aldrich) for 24 h, followed by replacement with complete medium containing a series of Bi-LyP-1 NPs and incubation for another 24 h and 48 h. The cells was washed with fresh medium for three times before CCK-8 assays were performed to assess the viabilities. To determine the ability of 4T1 cells to internalize Bi nanoparticles and to compare the specific binding affinity of Bi-LyP-1 NPs versus non-labeled Bi NPs, 4T1 cells (1 × 105 cells/well) were grown in 6 well plates for 24 h. After replacing the culture medium with complete medium contained 50 ppm Bi-LyP-1 NPs, the plates were then incubated at 37 oC for a series of times (0.5, 2, 4, 6 and 12 h). Following this incubation, 4T1 cells were washed with medium three times, released with cell dissociation buffer (Invitrogen, USA), and centrifuged. The resulting pellet was then treated with concentrated aqueous HNO3 for 12 h. The amount of Bi was then analyzed using ICP-MS. Control samples were performed at the same time in both of the above experiments. Animal Experiments. Six-week old Balb/c mice were managed under protocols approved by Soochow University Laboratory Animal Center. Tumors of mice were prepared with injection of

ACS Paragon Plus Environment

25

ACS Nano

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 26 of 41

60 µL PBS containing 2 × 106 4T1 cells into the fat pad of their right legs, and tumor sizes were calculated with the equation of volume (V) = length × width2/2. In Vivo Blood Circulation. 200 µL of Bi-LyP-1 NP solution (6 mg mL−1) was i.v. injected into healthy mice (n=5), and blood samples were collected by eyeball removal at 5 min, 0.25, 0.5, 1, 2, 4, 8, 12 and 24 h post injection. These samples were treated with concentrated aqueous HNO3, and the amount of Bi was measured using ICP-MS. In Vitro Radiation Therapy. In order to evaluate the radiosensitization effect of Bi-LyP-1 NPs, 4T1 cells were initially plated into 6-well plates (100, 200, 500, 1000, and 2000 cells/well) for 24 hours, followed by replacing the culture medium with complete medium containing BiLyP-1 NPs (0 or 10 ppm) for another 24 h incubation. After irradiated with different X-ray irradiation doses (0, 2, 4, 6, 8 Gy), cells were then further incubated in fresh medium for 10 days under standard conditions. Following this 10 day period, the surviving colonies were counted (only when more than 50 cells), and each treatment group was repeated for a total of three times. The surviving fraction = (surviving colonies)/(cells seeded × plating efficiency). The sensitizer enhancement ratio SERD0 and SER10 that described the ratios of X-ray irradiation doses needed to kill 63% and 90% of the total cells were calculated by the equation of SER=D(control group)/D(experimental group), fitted formula of SF=1−(1−e−D/D0)n and parameters of mean lethal dose (D0) and quasi-threshold dose (Dq) based on practical experimental data. In Vivo Tumor Accumulation. Tumor-bearing mice and healthy mice were given an i.v. administration of 200 µL Bi-LyP-1 NP solution (3 mg mL−1) and euthanized after 24 h, respectively (n=5). Tumors and main organs were digested with concentrated HNO3 overnight in order to analyze the amount of Bi in each tissue using ICP-MS.

ACS Paragon Plus Environment

26

Page 27 of 41

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 Nano

Dual-Modal Imaging. For in vitro CT imaging, Bi-LyP-1 NP solutions and iohexol solution containing various concentrations of Bi and I were detected using the CT instrument. For in vivo CT imaging, 30 µL of Bi-LyP-1 NP solution (6 mg mL−1) was administrated into tumors in mice, and the CT imaging was performed both prior to and after administration, with the parameters used of 80 kV, 450 µA and a thickness of 45 µm. As for in vivo PA imaging, experimental mice were adminstrated with 30 µL of Bi-LyP-1 NP solution (6 mg mL−1) via i.t. injection and 200 µL of Bi-LyP-1 NP solution (6 mg mL−1) via i.v. injection, respectively. PA imaging was carried out both prior to and after each injection. Throughout the entire course, mice were anesthetized and the body temperature was maintained around 37.5 oC. NIR-II Photothermal and Radiotherapy in Vivo. Six randomly divided groups of mice were employed for therapy: (I) Control, (II) Bi-LyP-1 NPs, (III) X-ray, (IV) Bi-LyP-1 NPs + laser, (V) Bi-LyP-1 NPs + X-ray, and (VI) Bi-LyP-1 NPs + laser + X-ray. Once the tumors attained ca. 100 mm3, mice in each group were administrated with 200 µL solution of PBS (Group I and III) or Bi-LyP-1 NPs (3 mg mL−1, the other four groups) via i.v. injection, respectively. After blood circulation for 24 h, Group V was treated with laser irradiation (1064 nm, 0.6 W cm−2, 20 min), Group III and IV were treated with X-ray irradiation (4 Gy), and Group VI was treated with both laser irradiation and X-ray irradiation (1064 nm, 0.6 W cm−2, 20 min, 4 Gy). During each laser irradiation treatment, the temperature profiles were captured in real time using a thermal camera. Following each treatment, H&E (hematoxylin-eosin), CD31 staining, and tumor slice analysis was performed. The data of body weight and tumor volumes in each group over the course of 14 days were recorded.

ACS Paragon Plus Environment

27

ACS Nano

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 28 of 41

In Vivo Clearance Study. For excretion studies, five mice that i.v. administrated with 200 µL of Bi-LyP-1 NP solution (3 mg mL−1) were raised in metabolic cages for 7 days. Each day, urine and feces samples were collected for further detection of Bi with TEM and ICP-MS. For longterm biodistribution studies, healthy mice that i.v. administrated with 200 µL of Bi-LyP-1 NP solution (3 mg mL−1) were scarified at day(s) 1, 7 and 30 after administration (n=5) and main organs were collected to analyze Bi levels using ICP-MS. Blood Panels and Histology Examinations. Five healthy mice that received an i.v. administration of Bi-LyP-1 NPs at a 2-fold treatment dose (6 mg mL−1, 200 µL) were euthanatized at day(s) 1, 7, and 30 after administration. Five additional mice i.v. administrated with PBS were used as a control. Standard procedures for serum biochemistry and a complete blood panel measurement were performed with blood obtained from mice at the different time points. In addition, the main organs of heart, liver, spleen, lung and kidneys were collected for H&E staining. Images were collected using an inverted laboratory microscope (Leica DM IL LED). Statistical Analysis. The data were analyzed using a one way ANOVA statistical analysis and indicated with p-values for significance.

AUTHOR INFORMATION Corresponding Author *E-mail: [email protected]; [email protected] Notes

ACS Paragon Plus Environment

28

Page 29 of 41

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 Nano

The authors declare no competing financial interest.

ACKNOWLEDGMENT This work was financially supported by the National Natural Science Foundation of China (Project No. 81371645, 81471716, 31400861, 81671782), Science and Technology Committee of Shanghai (Project No. 15PJD020, 15441905800, 16JC1400604), the Natural Science Foundation of Jiangsu Province (BK20140320), and a Project Funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD). Medicine & Engineering Cross Research Foundation of Shanghai Jiao Tong University (Project No. YG2014MS33), and the Intramural Research Program, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH).We thank the Instrumental Analysis Center of SJTU for the assistance with TEM, XRD, XPS and ICP-MS characterizations.

ASSOCIATED CONTENT Supporting Information. The Supporting Information is available free of charge via the Internet at http://pubs.acs.org, including Figures S1−S13 (PDF).

REFERENCES (1) Melancon, M. P.; Zhou, M.; Li, C. Cancer Theranostics with Near-Infrared Light-Activatable Multimodal Nanoparticles. Acc. Chem. Res. 2011, 44, 947–956.

ACS Paragon Plus Environment

29

ACS Nano

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 30 of 41

(2) Yoo, D.; Lee, J.-H.; Shin, T.-H.; Cheon, J. Theranostic Magnetic Nanoparticles. Acc. Chem. Res. 2011, 44, 863–874. (3) Ferrari, M. Cancer Nanotechnology: Opportunities and Challenges. Nat. Rev. Cancer 2005, 5, 161–171. (4)

Lim, E.-K.; Kim, T.; Paik, S.; Haam, S.; Huh, Y.-M.; Lee, K. Nanomaterials for

Theranostics: Recent Advances and Future Challenges. Chem. Rev. 2015, 115, 327–394. (5) Lammers, T.; Aime, S.; Hennink, W. E.; Storm, G.; Kiessling, F. Theranostic Nanomedicine. Acc. Chem. Res. 2011, 44, 1029–1038. (6) Muthu, M. S.; Leong, D. T.; Mei, L.; Feng, S.-S. Nanotheranostics-Application and Further Development of Nanomedicine Strategies for Advanced Theranostics. Theranostics 2014, 4, 660–677. (7) Barreto, J. A.; O’Malley, W.; Kubeil, M.; Graham, B.; Stephan, H.; Spiccia, L. Nanomaterials: Applications in Cancer Imaging and Therapy. Adv. Mater. 2011, 23, H18–H40. (8) Ambrogio, M. W.; Thomas, C. R.; Zhao, Y.-L.; Zink, J. I.; Stoddart, J. F. Mechanized Silica Nanoparticles: A New Frontier in Theranostic Nanomedicine. Acc. Chem. Res. 2011, 44, 903– 913. (9) Ng, K. K.; Lovell, J. F.; Zheng, G. Lipoprotein-Inspired Nanoparticles for Cancer Theranostics. Acc. Chem. Res. 2011, 44, 1105–1113. (10) Song, G.; Wang, Q.; Wang, Y.; Lv, G.; Li, C.; Zou, R.; Chen, Z.; Qin, Z.; Huo, K.; Hu, R.; Hu, J. A Low-Toxic Multifunctional Nanoplatform Based on Cu9S5@mSiO2 Core-Shell

ACS Paragon Plus Environment

30

Page 31 of 41

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 Nano

Nanocomposites: Combining Photothermal- and Chemotherapies with Infrared Thermal Imaging for Cancer Treatment. Adv. Funct. Mater. 2013, 23, 4281–4292. (11) Chen, J.; Wang, D.; Xi, J.; Au L.; Siekkinen, A.; Warsen, A.; Li, Z.-Y.; Zhang, H.; Xia, Y.; Li, X. Immuno Gold Nanocages with Tailored Optical Properties for Targeted Photothermal Destruction of Cancer Cells. Nano Lett. 2007, 7, 1318–1322. (12) Zharov, V. P.; Kim, J.-W.; Curiel, D. T.; Everts, M. Self-Assembling Nanoclusters in Living Systems: Application for Integrated Photothermal Nanodiagnostics and Nanotherapy. Nanomedicine: NBM 2005, 1, 326–345. (13) Zhao, F.; Meng, H.; Yan, L.; Wang, B.; Zhao, Y. Nanosurface Chemistry and Dose Govern the Bioaccumulation and Toxicity of Carbon Nanotubes, Metal Nanomaterials and Quantum Dots in Vivo. Sci. Bull. 2015, 60, 3–20. (14) Choi, H. S.; Liu, W.; Misra, P.; Tanaka, E.; Zimmer, J. P.; Ipe, B. I.; Bawendi, M. G.; Frangioni, J. V. Renal Clearance of Quantum Dots. Nat. Biotechnol. 2007, 25, 1165–1170. (15) Perry, J. L.; Herlihy, K. P.; Napier, M. E.; Desimone, J. M. PRINT: A Novel Platform toward Shape and Size Specific Nanoparticle Theranostics. Acc. Chem. Res. 2011, 44, 990–998. (16) Lux, F.; Mignot, A.; Mowat, P.; Louis, C.; Dufort, S.; Bernhard, C.; Denat, F.; Boschetti, F.; Brunet, C.; Antoine, R.; Dugourd, P.; Laurent, S.; Elst, L. V.; Muller, R.; Sancey, L.; Josserand, V.; Coll, J.-L.; Stupar, V.; Barbier, E.; Rémy, C.; et al. Ultrasmall Rigid Particles as Multimodal Probes for Medical Applications. Angew. Chem. Int. Ed. 2011, 50, 12299–12303.

ACS Paragon Plus Environment

31

ACS Nano

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 32 of 41

(17) Jiang, W.; Kim, B. Y. S.; Rutka, J. T.; Chan, W. C. W. Nanoparticle-Mediated Cellular Response Is Size-Dependent. Nat. Nanotechnol. 2008, 3, 145–150. (18) Deng, H.; Dai, F.; Ma, G.; Zhang, X. Theranostic Gold Nanomicelles Made from Biocompatible Comb-Like Polymers for Thermochemotherapy and Multifunctional Imaging with Rapid Clearance. Adv. Mater. 2015, 27, 3645–3653. (19) Kim, J.-W.; Galanzha, E. I.; Shashkov, E. V.; Moon, H.-M.; Zharov, V. P. Golden Carbon Nanotubes as Multimodal Photoacoustic and Photothermal High-Contrast Molecular Agents. Nat. Nanotechnol. 2009, 4, 688–694. (20) Ge, J.; Jia, Q.; Liu, W.; Guo, L.; Liu, Q.; Lan, M.; Zhang, H.; Meng, X.; Wang, P. RedEmissive Carbon Dots for Fluorescent, Photoacoustic, and Thermal Theranostics in Living Mice. Adv. Mater. 2015, 27, 4169–4177. (21) Li, B.; Ye, K.; Zhang, Y.; Qin, J.; Zou, R.; Xu, K.; Huang, X.; Xiao, Z.; Zhang, W.; Lu, X.; Hu, J. Photothermal Theragnosis Synergistic Therapy Based on Bimetal Sulphide Nanocrystals Rather Than Nanocomposites. Adv. Mater. 2015, 27, 1339–1345. (22) Zhang, C.; Zhao, K.; Bu, W.; Ni, D.; Liu, Y.; Feng, J.; Shi, J. Marriage of Scintillator and Semiconductor for Synchronous Radiotherapy and Deep Photodynamic Therapy with Diminished Oxygen Dependence. Angew. Chem. Int. Ed. 2015, 127, 1790–1794. (23) Sun, X.; Li, X.-F.; Russell, J.; Xing, L.; Urano, M.; Li, G. C.; Humm, J. L.; Ling, C. C. Changes in Tumor Hypoxia Induced by Mild Temperature Hyperthermia as Assessed by DualTracer Immunohistochemistry. Radiother. Oncol. 2008, 88, 269–276.

ACS Paragon Plus Environment

32

Page 33 of 41

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 Nano

(24) Zhang, X.-D.; Luo, Z.; Chen, J.; Shen, X.; Song, S.; Sun, Y.; Fan, S.; Fan, F.; Leong, D. T.; Xie, J. Ultrasmall Au10-12(SG)10-12 Nanomolecules for High Tumor Specificity and Cancer Radiotherapy. Adv. Mater. 2014, 26, 4565–4568. (25) Jain, S.; Coulter, J. A.; Hounsell, A. R.; Butterworth, K. T.; McMahon, S. J.; Hyland, W. B.; Muir, M. F.; Dickson, G. R.; Prise, K. M.; Currell, F. J.; O’Sullivan, J. M.; Hirst, D. G. CellSpecific Radiosensitization by Gold Nanoparticles at Megavoltage Radiation Energies. Int. J. Radiat. Oncol. Biol., Phys. 2011, 79, 531–539. (26) Joh, D. Y.; Sun, L.; Stangl, M.; Zaki, A. A.; Murty, S.; Santoiemma, P. P.; Davis, J. J.; Baumann, B. C.; Alonso-Basanta, M.; Bhang, D.; Kao, G. D.; Tsourkas, A.; Dorsey, J. F. Selective Targeting of Brain Tumors with Gold Nanoparticle-Induced Radiosensitization. PLoS One 2013, 8, e62425. (27) Zhang, X.-D.; Chen, J.; Min, Y.; Park, G. B.; Shen, X.; Song, S.-S.; Sun, Y.-M.; Wang, H.; Long, W.; Xie, J.; Gao, K.; Zhang, L.; Fan, S.; Fan, F.; Jeong, U. Metabolizable Bi2Se3 Nanoplates: Biodistribution, Toxicity, and Uses for Cancer Radiation Therapy and Imaging. Adv. Funct. Mater. 2014, 24, 1718–1729. (28) 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.

ACS Paragon Plus Environment

33

ACS Nano

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 34 of 41

(29) Song, G.; Chen, Y.; Liang, C.; Yi, X.; Liu, J.; Sun, X.; Shen, S.; Yang, K.; Liu, Z. CatalaseLoaded TaOx Nanoshells as Bio-Nanoreactors Combining High-Z Element and Enzyme Delivery for Enhancing Radiotherapy. Adv. Mater. 2016, 28, 7143–7148. (30) Zaki, A. A.; Joh, D.; Cheng, Z.; Barros, A. L. B. D.; Kao, G.; Dorsey, J.; Tsourkas, A. GoldLoaded Polymeric Micelles for Computed Tomography-Guided Radiation Therapy Treatment and Radiosensitization. ACS Nano 2014, 8, 104–112. (31) Song, G.; Liang, C.; Gong, H.; Li, M.; Zheng, X.; Cheng, L.; Yang, K.; Jiang, X.; Liu, Z. Core–Shell

MnSe@Bi2Se3

Fabricated

via

A

Cation

Exchange

Method

as

Novel

Nanotheranostics for Multimodal Imaging and Synergistic Thermoradiotherapy. Adv. Mater. 2015, 27, 6110–6117. (32) Moeller, B. J.; Richardson, R. A.; Dewhirst, M. W. Hypoxia and Radiotherapy: Opportunities for Improved Outcomes in Cancer Treatment. Cancer Metastasis Rev. 2007, 26, 241–248. (33) Horsman, M. R.; Overgaard, J. Hyperthermia: A Potent Enhancer of Radiotherapy. Clin. Oncol. 2007, 19, 418–426. (34) Yu, C. Y. Y.; Xu, H.; Ji, S.; Kwok, R. T. K.; Lam, J. W. Y.; Li, X.; Krishnan, S.; Ding, D.; Tang, B. Z. Mitochondrion-Anchoring Photosensitizer with Aggregation-Induced Emission Characteristics Synergistically Boosts the Radiosensitivity of Cancer Cells to Ionizing Radiation. Adv. Mater. 2017, DOI: 10.1002/adma.201606167.

ACS Paragon Plus Environment

34

Page 35 of 41

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 Nano

(35) Sherlock, S. P.; Tabakman, S. M.; Xie, L.; Dai, H. Photothermally Enhanced Drug Delivery by Ultra-Small Multifunctional FeCo/Graphitic-Shell Nanocrystals. ACS Nano 2011, 5, 1505– 1512. (36) Liu, Y.; Liu, Y.; Bu, W.; Xiao, Q.; Sun, Y.; Zhao, K.; Fan, W.; Liu, J.; Shi, J. Radiation/Hypoxia-Induced Solid Tumor Metastasis and Regrowth Inhibited by Hypoxia-Specific Upconversion Nanoradiosensitizer. Biomaterials 2015, 49, 1–8. (37) Chu, K. F.; Dupuy, D. E. Thermal Ablation of Tumours: Biological Mechanisms and Advances in Therapy. Nat. Rev. Cancer 2014, 14, 199–208. (38) Feng, L.; Li, K.; Shi, X.; Gao, M.; Liu, J.; Liu, Z. Smart pH-Responsive Nanocarriers Based on Nano-Graphene Oxide for Combined Chemo-and Photothermal Therapy Overcoming Drug Resistance. Adv. Healthc. Mater. 2014, 3, 1261–1271. (39) Feng, L.; Yang, X.; Shi, X.; Tan, X.; Peng, R.; Wang, J.; Liu, Z. Polyethylene Glycol and Polyethylenimine Dual-Functionalized Nano-Graphene Oxide for Photothermally Enhanced Gene Delivery. Small 2013, 9, 1989–1997. (40) Bashkatov, A. N.; Genina, E. A.; Kochubey, V. I.; Tuchin, V. V. Optical Properties of Human Skin, Subcutaneous and Mucous Tissues in the Wavelength Range from 400 to 2000 nm. J. Phys. D: Appl. Phys. 2005, 38, 2543–2555. (41) Liu, J.; Zheng, X.; Yan, L.; Zhou, L.; Tian, G.; Yin, W.; Wang, L.; Liu, Y.; Hu, Z.; Gu, Z.; Chen, C.; Zhao, Y. Bismuth Sulfide Nanorods as A Precision Nanomedicine for in Vivo Multimodal Imaging-Guided Photothermal Therapy of Tumor. ACS Nano 2015, 9, 696–707.

ACS Paragon Plus Environment

35

ACS Nano

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 36 of 41

(42) Zha, Z.; Yue, X.; Ren, Q.; Dai, Z. Uniform Polypyrrole Nanoparticles with High Photothermal Conversion Efficiency for Photothermal Ablation of Cancer Cells. Adv. Mater. 2013, 25, 777–782. (43) Zhang, Z.; Wang, J.; Chen, C. Near-Infrared Light-Mediated Nanoplatforms for Cancer Thermo-Chemotherapy and Optical Imaging. Adv. Mater. 2013, 25, 3869–3880. (44) Lovell, J. F.; Jin, C. S.; Huynh, E.; Jin, H.; Kim, C.; Rubinstein, J. L.; Chan, W. C. W.; Cao, W.; Wang, L. V.; Zheng, G. Porphysome Nanovesicles Generated by Porphyrin Bilayers for Use as Multimodal Biophotonic Contrast Agents. Nat. Mater. 2011, 10, 324–332. (45) Zhang, Z.; Wang, J.; Nie, X.; Wen, T.; Ji, Y.; Wu, X.; Zhao, Y.; Chen, C. Near Infrared Laser-Induced Targeted Cancer Therapy Using Thermoresponsive Polymer Encapsulated Gold Nanorods. J. Am. Chem. Soc. 2014, 136, 7317–7326. (46) Orecchioni, M.; Cabizza, R.; Bianco, A.; Delogu, L. G. Graphene as Cancer Theranostic Tool: Progress and Future Challenges. Theranostics 2015, 5, 710–723. (47) Yang, K.; Feng, L.; Shi, X.; Liu, Z. Nano-Graphene in Biomedicine: Theranostic Applications. Chem. Soc. Rev. 2013, 42, 530–547. (48) Wu, Z.-C.; Li, W.-P.; Luo, C.-H.; Su, C.-H.; Yeh, C.-S. Rattle-Type Fe3O4@CuS Developed to Conduct Magnetically Guided Photoinduced Hyperthermia at First and Second NIR Biological Windows. Adv. Funct. Mater. 2015, 25, 6527–6537. (49) Chen, Z.; Wang, Q.; Wang, H.; Zhang, L.; Song, G.; Song, L.; Hu, J.; Wang, H.; Liu, J.; Zhu, M.; Zhao, D. Ultrathin PEGylated W18O49 Nanowires as a New 980 nm-Laser-Driven

ACS Paragon Plus Environment

36

Page 37 of 41

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 Nano

Photothermal Agent for Efficient Ablation of Cancer Cells in Vivo. Adv. Mater. 2013, 25, 2095– 2100. (50) Guo, C.; Yu, H.; Feng, B.; Gao, W.; Yan, M.; Zhang, Z.; Li, Y.; Liu, S. Highly Efficient Ablation of Metastatic Breast Cancer Using Ammonium-Tungsten-Bronze Nanocube as A Novel 1064 nm-Laser-Driven Photothermal Agent. Biomaterials 2015, 52, 407–416. (51) Li, A.; Li, X.; Yu, X.; Li, W.; Zhao, R.; An, X.; Cui, D.; Chen, X.; Li, W. Synergistic Thermoradiotherapy Based on PEGylated Cu3BiS3 Ternary Semiconductor Nanorods with Strong Absorption in the Second Near-Infrared Window. Biomaterials 2017, 112, 164–175. (52) Hu, K.-W.; Liu, T.-M.; Chung, K.-Y.; Huang, K.-S.; Hsieh, C.-T.; Sun, C.-K.; Yeh, C.-S. Efficient Near-IR Hyperthermia and Intense Nonlinear Optical Imaging Contrast on the Gold Nanorod-in-Shell Nanostructures. J. Am. Chem. Soc. 2009, 131, 14186–14187. (53) Ding, X.; Liow, C. H.; Zhang, M.; Huang, R.; Li, C.; Shen, H.; Liu, M.; Zou, Y.; Gao, N.; Zhang, Z.; Li, Y.; Wang, Q.; Li, S.; Jiang, J. Surface Plasmon Resonance Enhanced Light Absorption and Photothermal Therapy in the Second Near-Infrared Window. J. Am. Chem. Soc. 2014, 136, 15684–15693. (54) Manikandan, M.; Hasan, N.; Wu, H.-F. Platinum Nanoparticles for the Photothermal Treatment of Neuro 2A Cancer Cells. Biomaterials 2013, 34, 5833–5842. (55) Li, L.; Liu, Y.; Hao, P.; Wang, Z.; Fu, L.; Ma, Z.; Zhou, J. PEDOT Nanocomposites Mediated Dual-Modal Photodynamic and Photothermal Targeted Sterilization in Both NIR I and II Window. Biomaterials 2015, 41, 132–140.

ACS Paragon Plus Environment

37

ACS Nano

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 38 of 41

(56) Comin, A.; Manna, L. New Materials for Tunable Plasmonic Colloidal Nanocrystals. Chem. Soc. Rev. 2014, 43, 3957–3975. (57) Li, W.; Rong P.; Yang, K.; Huang, P.; Sun, K.; Chen, X. Semimetal Nanomaterials of Antimony as Highly Efficient Agent for Photoacoustic Imaging and Photothermal Therapy. Biomaterials 2015, 45, 18–26. (58) McMahon, J. M.; Schatz, G. C.; Gray, S. K. Plasmonics in the Ultraviolet with the Poor Metals Al, Ga, In, Sn, Tl, Pb, and Bi. Phys. Chem. Chem. Phys. 2013, 15, 5415–5423. (59) Tediosi, R.; Armitage, N. P.; Giannini, E.; Marel, D. V. D. Charge Carrier Interaction with A Purely Electronic Collective Mode: Plasmarons and the Infrared Response of Elemental Bismuth. Phys. Rev. Lett. 2007, 99, 016406. (60) Lusic, H.; Grinstaff, M. W. X-Ray Computed Tomography Contrast Agents. Chem. Rev. 2013, 113, 1641–1666. (61) Swy, E. R.; Schwartz-Duval, A. S.; Shuboni, D. D.; Latourette, M. T.; Mallet, C. L.; Parys, M.; Cormode, D. P.; Shapiro, E. M. Dual-Modality, Fluorescent, PLGA Encapsulated Bismuth Nanoparticles for Molecular and Cellular Fluorescence Imaging and Computed Tomography. Nanoscale 2014, 6, 13104–13112 (62) Yarema, M.; Kovalenko, M. V.; Hesser, G.; Talapin, D. V.; Heiss, W. Highly Monodisperse Bismuth Nanoparticles and Their Three-Dimensional Superlattices. J. Am. Chem. Soc. 2010, 132, 15158–15159.

ACS Paragon Plus Environment

38

Page 39 of 41

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 Nano

(63) Wang, F.; Buhro, W. E. An Easy Shortcut Synthesis of Size-Controlled Bismuth Nanoparticles and Their Use in the SLS Growth of High-Quality Colloidal Cadmium Selenide Quantum Wires. Small 2010, 6, 573–581. (64) Kinsella, J. M.; Jimenez, R. E.; Karmali, P. P.; Rush, A. M.; Kotamraju, V. R.; Gianneschi, N. C.; Ruoslahti, E.; Stupack, D.; Sailor, M. J. X-Ray Computed Tomography Imaging of Breast Cancer by Using Targeted Peptide-Labeled Bismuth Sulfide Nanoparticles. Angew. Chem. Int. Ed. 2011, 50, 12308–12311. (65) Dubertret, B.; Skourides, P.; Norris, D. J.; Noireaux, V.; Brivanlou, A. H.; Libchaber, A. In Vivo Imaging of Quantum Dots Encapsulated in Phospholipid Micelles. Science 2002, 298, 1759–1762. (66) Yan, Z.; Wang, F.; Wen, Z.; Zhan, C.; Feng, L.; Liu, Y.; Wei, X.; Xie, C.; Lu, W. LyP-1Conjugated PEGylated Liposomes: A Carrier System for Targeted Therapy of Lymphatic Metastatic Tumor. J. Control. Release 2012, 157, 118−125. (67) Roper, D. K.; Ahn, W.; Hoepfner, M. Microscale Heat Transfer Transduced by Surface Plasmon Resonant Gold Nanoparticles. J. Phys. Chem. C 2007, 111, 3636–3641. (68) 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.

ACS Paragon Plus Environment

39

ACS Nano

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 40 of 41

(69) Rabin, O.; Perez, J. M.; Grimm, J.; Wojtkiewicz, G.; Weissleder, R. An X-Ray Computed Tomography Imaging Agent Based on Long-Circulating Bismuth Sulphide Nanoparticles. Nat. Mater. 2006, 5, 118–122. (70) Liu, J.; Wang, P.; Zhang, X.; Wang, L.; Wang, D.; Gu, Z.; Tang, J.; Guo, M.; Cao, M.; Zhou, H.; Liu, Y.; Chen, C. Rapid Degradation and High Renal Clearance of Cu3BiS3 Nanodots for Efficient Cancer Diagnosis and Photothermal Therapy in Vivo. ACS Nano 2016, 10, 4587– 4598. (71) Li, Z.; Hu, Y.; Howard, K. A.; Jiang, T.; Fan, X.; Miao, Z.; Sun, Y.; Besenbacher, F.; Yu, M. Multifunctional Bismuth Selenide Nanocomposites for Antitumor Thermo-Chemotherapy and Imaging. ACS Nano 2016, 10, 984–997. (72) Ku, G.; Zhou, M.; Song, S.; Huang, Q.; Hazle, J.; Li, C. Copper Sulfide Nanoparticles as A New Class of Photoacoustic Contrast Agent for Deep Tissue Imaging at 1064 nm. ACS Nano 2012, 6, 7489–7496. (73) Zhou, C.; Long, M.; Qin, Y.; Sun, X.; Zheng, J. Luminescent Gold Nanoparticles with Efficient Renal Clearance. Angew. Chem. Int. Ed. 2011, 123, 3168–3172. (74) Balasubramanian, S. K.; Jittiwat, J.; Manikandan, J.; Ong, C.-N.; Yu, L. E.; Ong, W.-Y. Biodistribution of Gold Nanoparticles and Gene Expression Changes in the Liver and Spleen after Intravenous Administration in Rats. Biomaterials 2010, 31, 2034–2042.

SYNOPSIS

ACS Paragon Plus Environment

40

Page 41 of 41

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 Nano

Ultra-small semimetal nanoparticles of bismuth with strong absorption in second near-infrared window are fabricated. After being labeled with a tumor-homing peptide (LyP-1), Bi nanoparticles exhibited increased tumor accumulation and efficient clearance, and were successfully used as a theranostic agent with excellent dual-modal CT/PA imaging properties and synergistic effect of cancer thermoradiotherapy.

ACS Paragon Plus Environment

41