CuS Nanodot-Loaded Thermosensitive Hydrogel for Anticancer

Sep 4, 2018 - Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital of Guangzho...
0 downloads 0 Views 2MB Size
Subscriber access provided by Kaohsiung Medical University

Article

CuS nanodots loaded thermosensitive hydrogel for anti-cancer photothermal therapy Jijun Fu, Jian-ye Zhang, Songpei Li, Lingmin Zhang, Zhongxiao Lin, Lu Liang, Aiping Qin, and Xiyong Yu Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.8b00624 • Publication Date (Web): 04 Sep 2018 Downloaded from http://pubs.acs.org on September 6, 2018

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 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 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.

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 22 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

Molecular Pharmaceutics

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

CuS nanodots loaded thermosensitive hydrogel for anti-cancer photothermal therapy Ji-Jun Fu 1 2, Jian-Ye Zhang 1 2, Song-Pei Li 1, Ling-Min Zhang 1, Zhong-Xiao Lin 1, Lu Liang 1, Ai-Ping Qin 1 #, Xi-Yong Yu 1 2 # 1 Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China, 511436 2 The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China, 510700 # Address correspondence to [email protected], [email protected] Abstract: Purpose: To establish an injectable hydrogel encapsulating the copper sulfide (CuS) nanodots for photothermal therapy against cancer. Methods: The CuS nanodots were prepared by one-pot synthesis and, the thermosensitive Pluronic F127 was used as the hydrogel matrix. The CuS nanodots and the hydrogel were characterized by morphous, particle size, serum stability, photothermal performance upon repeated 808 nm laser irradiation as well as the rheology features. The effects of the CuS nanodots and the hydrogel were evaluated qualitatively and quantitatively in 4T1 mouse breast cancer cells. The retention, photothermal efficacy, therapeutic effects and systemic toxicity of the hydrogel were assessed in tumor bearing mouse model. Results: The CuS nanodots with a diameter of about 8 nm exhibited satisfying serum stability, photo-heat conversion ability and repeated laser exposure stability. The hydrogel encapsulation did not negatively influence the above features of the photothermal agent. The nanodots loaded hydrogel shows a phase transition at body temperature and, as a result, a long retention in vivo. The photothermal agent embedded hydrogel played a promising photothermal therapeutic effects in tumor bearing mouse model with low systemic toxicity after peritumoral administration. Keywords: CuS; nanodots; hydrogel; thermosensitive; injectable; in-situ; photothermal

Introduction In the past few years, the world has witnessed many new methods in treating cancers [1-10], such as monoclonal antibodies, gene therapy [4, 10], photodynamic therapy (PDT), sonodynamic therapy, photothermal therapy (PTT) [8]. However, cancer therapy is still a big challenge, with the mean survival length of cancer patients only several years. As a traditional dosage form, hydrogel has been extensively investigated in many research fields, especially in regeneration medicine. For example, thermosensitive carbon nanotube-functionalized hydrogel was proposed to support long-term survival of cardiomyocytes and promote its alignment and proliferation [11]. Hydrogel made of hyaluronic acid and gelatin was used to encapsulate stem cell-derived exosomes, and the hydrogel could be used as a novel cell-free scaffold material for articular cartilage regeneration [12]. Injection of microporous annealing particle hydrogels in the stroke cavity was found to reduce gliosis and inflammation, as well as promote neural progenitor cell migration to the lesion [13]. CaSO4 and FGF-18 delivered by chitinpoly(D,L-lactic acid-co-glycolic acid) (PLGA) hydrogel was found to be able to enhance bone regeneration [14]. Application of the hydrogels in treating 1

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89

cancers is also a newly emerging research field. Lin et al prepared the paclitaxel hydrogels with three different dispersing states of the compound, molecules, nanocrystals and microcrystals. The hydrogel with paclitaxel nanocrystals exhibited the most outstanding long-term stability, retention in tumor tissue and anti-cancer performance [15, 16]. Nowadays, there has been an increasingly widespread interest in PTT. The photothermal agents (PTA) are able to convert laser energy, especially the near infrared light (NIR) which could penetrate deep into tissues, into heat energy. Up to now, PTT is often employed to treat cancers. Sun et al [17] proposed core-shell TiO2 nanoparticles as an excellent PTA. Jiu et al [18] reported an ultrasmall iron-gallic acid nanoparticles as photoacoustic imaging (PAI) guided PTA. MoO3-x nanostructures could also be used as PAI guided photothermal agent [19, 20]. Based on the fact of the elevated negative charges on cancer cell membrane compared to healthy cells, a positively charged superparamagnetic Fe3O4 nanoparticles was proposed to achieve tumor targeting and destruction upon NIR irradiation [21]. Zhan et al [22] fabricated special liposomes encapsulating tetrodotoxin and dexmedetomidine. The liposomes were decorated by gold nanorods (GNR) which could initiate the release of the anesthetics to achieve repeatable and on-demand nerve block upon NIR irradiation for short duration and low irridance. The authors further discovered that encapsulation of the photosensitizer 1,4,8,11,15,18,22,25-octabutoxyphthalocyaninatopalladium(II) (PdPC) in liposomes enhanced the on-demand release of the anesthetics upon NIR irradiation [23]. Normally, the PTA are intravenous administered to play the therapeutic effects. Despite great advances in PTT, the toxicity of the intravenous administered PTA remains a big concern as most of the PTA are made of heavy metal elements, such as gold and bismuth, that are difficult to tackle with. In addition, despite the enhanced penetration and retention effects (EPR) and other targeting methods, the nano-carriers finally distributing in tumor tissues account for only a small proportion of the dose administered, as a result, the unsatisfactory distribution retards superior therapeutic effects and increases systemic toxicity. In this study, an injectable Pluronic F127 hydrogel embedding copper sulfide (CuS) nanodots for peritumoural administration is proposed (Fig. 1). Pluronic F127 is widely used in constructing thermosensitive hydrogel and, is often employed in drug delivery systems for i.v. administration [24-26]. As the hydrogel is locally administered and has a long retention time, it is believed that this system will exhibit better photothermal therapeutic effects and reduced systemic toxicity. As far as we know, PTA loaded hydrogel for cancer therapy was rarely reported.

2

ACS Paragon Plus Environment

Page 2 of 22

Page 3 of 22 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

Molecular Pharmaceutics

90 91 92 93

Fig. 1 Schematic illustration

Materials and methods Preparation and characterization of CuS nanodots

95

21 mg of CuCl2•2H2O and 60 mg of polyvinylpyrrolidone (M.W. 24000) were added into 5 ml of pure water, the mixture was stirred to get clear solution.

96

Then, 30 mg of Na2S•9H2O was added with stirring. Then, the solution was

97

heated to 90 °C for 15 min with stirring to get dark green nanodispersions. After dialysis in pure water, CuS nanodots was obtained. Appearance of the nanodot was pictured by transmission electron microscopy (300 kV) and, particle size was measured by particle sizing system (Malvern). To investigate stability of the CuS nanodots, serum was added to CuS nanodispersions for a final concentration of 10%, and CuS nanodots according

94

98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123

to CuS was 0.5 mg/ml. The sample was placed in water bath at 37 °C for 6 d, visible absorption curves of the sample as well as particle size of the nanodots before and after storage were measured. To evaluate the photothermal performance of CuS nanodots, the nanodispersions with different concentrations (0, 20, 50, 100, 200, 500 µg/ml) were exposed to 808 nm laser at 1 W/cm2. Temperatures of the samples at different time points were recorded by infrared imaging camera (Fotric). To further characterize the CuS nanodots, the sample with 0.5 mg/ml CuS was exposed to NIR irradiation for five cycles. After each irradiation for 5 min at 1 W/cm2, the sample was cooled to room temperature naturally, and then exposed to 808 nm laser again. Temperatures of the sample were monitored. Particle size and visible absorption curves of the sample before and after 5-cycle irradiation were measured. Preparation and characterization of CuS nanodots embedded Pluronic F127 hydrogel Pluronic F127 granules were simply added to CuS nanodispersions with stirring in ice bath to get CuS nanodots loaded hydrogel. The concentration of Pluronic F127 was 0.2 g/ml and, the concentration of CuS nanodots was 0.1 mg/ml. Hydrogel loaded with CuS nanodots was frozen in liquid nitrogen overnight, then the frozen hydrogel was lyophilized. Morphous of the final 3

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

124 125 126 127 128 129 130 131

sample was viewed by scanning electron microscopy (SEM, 3.0 kV). Blank Pluronic F127 hydrogel was used as a control. To investigate the photothermal performance of CuS nanodots loaded hydrogel, the hydrogel was exposed to 808 nm laser at 1 W/cm2. Temperatures of the hydrogel at different time points were monitored. Blank hydrogel was used as a control. To further characterize the CuS nanodots loaded hydrogel, the rheology features of the samples were measured. The frequency was 1 Hz and, the

132

heating rate was 1 °C /min. The blank hydrogel was used as a control. To study the stability of CuS nanodots loaded hydrogel, the sample was

133 135 136 137 138 139

placed at 37 °C for days. The hydrogel sample before and after storage was diluted by water for 10 times, and then measured by particle sizing system to reflect the size of the CuS nanodots. Moreover, the heating curves upon NIR irradiation (808 nm, 1 W/cm2) were gathered before and after different storage time. The similarity between two curves was evaluated by f2 factor according to the following equation and, f2 > 50 means highly similar.

140 141 142 143

Rt and Tt mean temperature of the sample before and after storage upon NIR irradiation at t minute. Assessment of in vitro photothermal therapeutic effects

144

4T1 mouse breast cancer cells (1ⅹ104) were seeded in 96-plate and

145

incubated overnight (37 °C, 5% CO2). Then, CuS nanodispersion was added

146 147 148 149 150

to make final concentration of the nanodots 0, 2, 5, 10, 20 µg/ml, respectively. The cells were incubated for another 24 h. Then, the cells were exposed to 808 nm laser (1 W/cm2) for 1 min and, the cells free from NIR treatment were used as control. After incubation for another 4 h, the viability of the cells was measured by MTT method.

151

4T1 mouse breast cancer cells (5ⅹ104) were seeded in 24-plate and

152 153 154 155 156 157 158 159 160 161 162

incubated overnight (37 °C, 5% CO2). Then, CuS nanodispersion was added to make final concentration of the nanodots 20 µg/ml. Then, the cells were exposed to 808 nm laser (1 W/cm2) for 3 min and, the cells treated by PBS and the cells free from NIR treatment were used as controls. After incubation for another 4 h, propidium iodide (final concentration 30 µg/ml) was added and incubated for 30 min to stain the dead cells. Then, the culture media was discarded and the cells were washed by PBS for several times. 4% paraformaldehyde solution was used to fix the cells. After washing by PBS for several times, Hoechst 33342 solution was used to stain the nucleus of all the cells. At last, the cells were photographed by fluorescence microscope to view the photothermal therapeutic effects of the nanodots.

163

4T1 mouse breast cancer cells (5ⅹ104) were seeded in 24-plate in 1 ml of

164

culture media. 200 µl of the CuS nanodots embedded hydrogel or blank

134

4

ACS Paragon Plus Environment

Page 4 of 22

Page 5 of 22 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

Molecular Pharmaceutics

165

hydrogel was added to the upper transwell (8 µm). The cells were incubated at

166 167 168 169 170 171 172 173

37 °C for 24 h. Then, the hydrogels encapsulated transwells were exposed to 808 nm laser (1 W/cm2) for 3 min and, the cells treated by blank hydrogel and the cells free from NIR treatment were used as controls. Finally, the cells were treated and pictured as the above section. Retention of the hydrogels around tumor tissue The study was approved by Ethics Committee of Guangzhou Medical University. The near infrared fluorescent dye DiR labeled hydrogel was prepared by

174

adding DiR ethanol solution (100 µg/ml) to the blank hydrogel with stirring in

175

ice bath to make the final concentration of 2 µg/ml. The control DiR solution

176

was prepared by adding DiR ethanol solution (100 µg/ml) to PBS with stirring

177

to make the same final concentration of 2 µg/ml before use At day 0, Balb/c mouse (female, 6-8 week old) was subcutaneously administered 1 million of 4T1 cancer cells to establish armpit tumor bearing

178 179 181

mouse model. At day 10, 500 µl of DiR labeled hydrogel was peritumoral injected. To ensure homogenous distribution of the hydrogel surrounding the

182

tumor tissue, the needle was inserted at five different points and, 100 µl of the

183 184 185 186 187 188 189 190

hydrogel was injected at each point. Mice received 500 µl of the DiR solution were used as control. Then at different time intervals, the DiR signals were captured at 740/820 nm to image the retention of the hydrogels around tumor tissue (Berthold). In vivo anti-cancer performance At day 0, Balb/c mouse (female, 6-8 week old) was subcutaneously administered 1 million of 4T1 cancer cells to establish armpit tumor bearing mouse model. At day 7, the volume of the tumor reached approximately 200

191

mm3 according to the following equation: volume=lengthⅹ(width)2/2. Then, 500

192

µl of the CuS nanodots loaded hydrogel was administered by peritumoral

193

injection. Mice received 500 µl of the blank hydrogel were used as control. To ensure homogenous distribution of the hydrogel surrounding the tumor tissue,

180

194 195 196 197 198 199 200 201 202 203 204 205 206 207

the needle was inserted at five different points and, 100 µl of the hydrogel was injected at each point. After 2 h, the tumors were exposed to 808 nm laser at 1 W/cm2 for 5 min after the mice were anesthetized. Temperatures of the tumor tissues were recorded by infrared imaging camera. NIR treatment was carried out every day from then on. At day 15, the mice were sacrificed and, the tumors of the test and control group were photographed and weighed. Then, the tumor, heart, liver, spleen, lung and kidney samples were made into paraffin sections and H&E stained. Body weight of the mice was measured everyday. Statistical analysis All values were expressed as mean ± standard deviation (SD). All comparisons were performed by the two-tailed Student’s t test. A p-value less than 0.05 was supposed to be statistically significant and, p-value less than 5

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

208 209 210 211 212 213 214 215

0.01 was considered to be highly significant.

Results and discussions Characteristics of the CuS nanodots The CuS nanoplatform was prepared by a simple one-pot synthesis. PVP was used as the stabilizer of the nanodispersions due to its favourable hydrophilicity. The final product appeared to be a dark green solution and, the concentration of the nanoplatform could be simply adjusted by dilution through adding pure water.

6

ACS Paragon Plus Environment

Page 6 of 22

Page 7 of 22 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

Molecular Pharmaceutics

216 217 218 219

Fig. 2 In vitro characterization of the CuS nanodots. A) TEM image of the CuS nanodots; B) Particle sizes of the CuS nanodots before (red) and after 6-day incubation in serum at 37 °C (green); C) The visible absorption curves of CuS nanodots before and after 7

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239

incubation in serum for 3 and 6 days at 37 °C; D) and E) The thermographs and corresponding heating curves of the CuS nanodots at varied concentrations upon 808 nm laser exposure (1 W/cm2); F) The heating curves of the CuS nanodots upon repeated 808 nm irradiation (1 W/cm2); G) Particle sizes of the CuS nanodots before (red) and after (green) 5-cycle 808 nm irradiation (1 W/cm2); H) The visible absorption profiles of the CuS nanodots before and after 5-cycle 808 nm irradiation (1 W/cm2).

240

nanodots exposed to 10% blood serum for 3 and 6 days at 37 °C was compared to that of the original sample to assess the stability of the PTA. The sample after serum exposure still exhibited an increasing visible absorption curve, which was highly comparable to the original one (Fig. 2C). This indicated the outstanding stability of the CuS nanodots in body fluids and, promising photothermal performance in vivo. The stability of the CuS nanodots in the body fluids was also evaluated in the term of particle size, Fig. 2B shows a slight decrease in diameter from 11 nm to 10 nm after serum exposure for 6

241 242 243 244 245 246 247 248 249 250 251 252 253

Fig. 2A shows that CuS existed as irregular nanodots in the solution, with a diameter of approximately 8 nm. Particle size of the nanodots was further validated by particle sizing system (Fig. 2B), which shows a single peak around 11 nm. The appearance and size of the CuS nanodots were in accordance with other reports [27]. The NIR is considered to be biologically compatible due to its deep tissue penetration and negligible influence on tissues. PTAs are able to convert the light to heat to play anti-cancer effects by generating intolerable high temperature microenvironment for malignant cells. The light-heat conversion ability of the PTAs can be indicated by visible absorption curve. The visible absorption curve in Fig. 2C saw a steady increase from 600 nm to 1000 nm, implying the encouraging NIR absorption capacity of the CuS nanodots. The stability of PTA in the body fluids is critical for its performance in vivo. To assess the stability of the PTA, the visible absorption curve of the CuS

days at 37 °C, and the data further supported the above results. Fig. 2D and Fig. 2E show the thermo-photographs and heating curves of the CuS nanodots with a serial concentrations of 20, 50, 100, 200, 500 µg/ml upon 808 nm laser exposure. The CuS nanodots exhibited dramatic increase in temperature upon NIR irradiation at any concentration. Moreover, the temperature rises along with the increase of concentration, with the final

255 256 257 258 259 260 261 262 263 264 265 266

temperature of the five concentrations reaching 35.5, 41.8, 47.0, 55.8, 60.6 °C, respectively. Meanwhile, the NIR exposure had little influence on the pure water control, with the control showing negligible temperature changes. The above results indicated the favorable photothermal effects of the CuS nanodots. Photothermal stability is one of the most important index to assess PTA. Indocyanine green (ICG), a typical PTA belonging to the cypates, shows severe light-bleach and reduced photothermal efficacy after repeated NIR exposure [28, 29]. Despite its excellent photo-heat conversion ability, the photothermal instability hampered the practical use of ICG as PTA. As the CuS nanodots loaded hydrogel would be locally administered, it was hoped that the PTA in the formed hydrogel would be able to exert long-term therapeutic effects. Fig. 2F shows the heating curve of the CuS nanodots upon repeated

267

NIR irradiation. The nanodispersion could be re-heated to about 56 °C after 5

254

8

ACS Paragon Plus Environment

Page 8 of 22

Page 9 of 22 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

Molecular Pharmaceutics

268 269

min laser exposure once the sample was naturally cooled to room temperature. In other words, the heating curve of each cycle was identical to the other ones,

270

with the highest temperature of each cycle nearly 56 °C, and the heating curve showed negligible changes after repeated laser exposure. These data indicated that the CuS nanodots displayed satisfying photothermal stability. To further validate this feature, the particle size and visible absorption curve of the nanodots before and after 5-cycle NIR irradiation were measured. Fig. 2G shows that the particle size of the sample post-NIR treatment was about 10 nm, slightly smaller than the diameter of 11 nm before NIR treatment. Fig. 2H shows that the two visible absorption curves of the nanodots were obviously identical, with minor variations. Both graphs further support the encouraging photothermal stability of the CuS nanodots. Based on these results, it is estimated that the PTA would maintain the therapeutic effects throughout the whole retention period in vivo and would not be destroyed by body fluids or repeated laser exposure. Features of the CuS nanodots loaded Pluronic F127 hydrogel In order to reduce the systemic toxicity of intravenously administered PTA and overcom the possible problem of unsatisfying photothermal therapeutic effects due to the limited enrichment of the PTA in tumor tissues, thermosensitive in-situ hydrogel was employed to encapsulate the CuS nanodots, which would be peritumorally injected. It was believed that the injectable hydrogel would solidify locally and, form semisolid hydrogel surrounding the tumor tissue and, might exhibit a long retention time. Meanwhile, the local administration would decrease systemic toxicity and increase therapeutic outcomes. In this section, the in vitro characteristics associated with the above aims would be discussed in detail.

271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293

9

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

294 295 296 297 298 299

Fig. 3 In vitro characterization of the CuS hydrogel. A) SEM images of the blank Pluronic F127 hydrogel and the CuS nanodots loaded hydrogel; B) and C) The thermographs and corresponding heating curves of the CuS hydrogel and blank hydrogel upon 808 nm irradiation (1 W/cm2); D) Pictures of the blank hydrogel and CuS hydrogel at 4 and 37 °C; E) The complex viscosity and loss modulus curves of the blank hydrogel and CuS 10

ACS Paragon Plus Environment

Page 10 of 22

Page 11 of 22 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

Molecular Pharmaceutics

300 301

hydrogel; F) Particle size of the CuS nanodots encapsulated in the hydrogel after different storage time at 37 °C; G) The heating profiles of the CuS hydrogel upon 808 nm laser

302

exposure (1 W/cm2) after different storage time at 37 °C.

303 304 305 306

The CuS nanodots embedded hydrogel was prepared by a simple one-pot method of dissolving Pluronic F127 in the nanodispersions directly. Fig. 3A shows the morphous of the blank hydrogel and the PTA encapsulated hydrogel. Both hydrogels had a highly macroporous network structure, with the pores

307

about 50-500 µm in diameter. In detail, the blank hydrogel shows more but smaller pores than the CuS nanodots embedded hydrogel. With 10000-fold magnification, both the blank hydrogel and the CuS nanodots embedded hydrogel displayed a smooth surface. Although the CuS nanodots were supposed to be evenly dispersed in the hydrogel matrix, the nanodots were too small to be detected by SEM. Ideally, the CuS nanodots embedded hydrogel would exhibit pronouncing photothermal performance. Fig. 3B and Fig. 3C show the temperature changes of the PTA encapsulated hydrogel and the blank control hydrogel upon 808 nm laser exposure. The temperature of CuS hydrogel increased

308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330

dramatically to 54.9 °C upon NIR irradiation. The heating curve of the CuS hydrogel shown in Fig. 3C was comparable to that of the nanodispersions illustrated in Fig. 2E. Moreover, the laser exposure had little influence on the blank hydrogel, with negligible temperature changes throughout the whole period. These data indicate that the encapsulation in hydrogel did not negatively influence the photothermal performance of the CuS nanodots and, the nanodots loaded hydrogel was supposed to display excellent photo-heat conversion transition capacity in vivo. To elevate the compliance of the patients, non-invasive treatment is highly required nowadays. Injectable hydrogel is one of the most widely used non-invasive administration route. Pluronic F127 is a commercially available thermosensitive hydrogel constructing material. Fig. 3D shows that both the blank hydrogel and the CuS hydrogel are liquid at 4°C and, the two hydrogels changed into semisolid at body temperature. In other words, the formed in-situ

332 333 334 335 336 337 338 339

hydrogel maintained its fluidity at low temperature (< 20 °C) and, the fluidity assigned the injectable feature of the hydrogel. At the same time, the in-situ hydrogel solidify at body temperature and, the semisolid state would promise a long retention time in vivo. To explain the phase transition of the hydrogels in detail, the rheology characteristics of the hydrogels were studied. Fig. 3E shows the rheological characteristics of the blank hydrogel and the CuS hydrogel. Both the complex viscosity curves exhibited a sharp increase in the complex viscosity in a narrow temperature range. In detail, the complex viscosity profile of the blank hydrogel showed a phase transition temperature

340

of 30 °C, while the encapsulation of the CuS nanodots resulted in a decrease

341

in phase transition temperature to 26 °C. The phase transition temperatures of both hydrogels indicated by the loss modulus curves were in accordance with the above numbers. Normally, loading water-soluble substances will increase the phase transition temperature of the hydrogel, while embedding water-insoluble agents will decrease the phase transition temperature. Despite

331

342 343 344 345

11

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

346 347 348 349 350 351 352 353 354 355 356 357 358 359

the water-soluble PVP in the nanodots, the CuS core was water-insoluble and hydrophobic. As a result, there was a reduction in the phase transition temperature of the hydrogel after the CuS nanodots encapsulation. Anyhow, these data indicated the CuS hydrogel would undergo a phase transition process after being locally administered, assigning the hydrogel a long retention time in vivo. One of the advantage of the injectable hydrogel in treating cancers is the sustained therapeutic efficacy. To achieve this, the PTA in the hydrogels must be stable for a considerable period. In this research, the particle size and the heating curve of the CuS nanodots after being encapsulated were measured. Fig. 3F shows that the nanodots in the hydrogel maintained a nearly constant diameter around 7.5 nm, comparable to the original particle size before being loaded. The result indicated that the CuS nanodots remained stable in the hydrogel in the term of particle size. Fig. 3G shows the heating profiles of the

360

CuS hydrogel before and after storage at 37 °C for several days. It can be seen that all the heating curves were nearly identical, with the temperature

361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378

reaching about 55 ° C at the end of laser exposure. It seems that the encapsulation and storage at body temperature had little influence on the photothermal performance of the CuS nanodots. Moreover, as all the heating profiles were obtained by recording the thermograph of the same hydrogel sample upon NIR irradiation at different time intervals, it can be concluded that the CuS nanodots remained stable in photo-heat transition ability after repeated 808 nm laser exposure. Statistically, the similarity factor f2 between the initial heating curve and that of the 4th, 6th, 8th, 11th, 13th day were 88.3, 82.3, 88.1, 78.2 and 83.4, respectively. All the similarity factor f2 is over 50, which represented that all the heating profiles were highly comparable. In a word, both results supported that encapsulation in the hydrogels and storage at body temperature would not negatively influence the CuS nanodots and, implied that the PTA would perform sustained therapeutic effects in vivo. In vitro anti-cancer photothermal therapy In this section, the photothermal therapeutic effects of the CuS nanodots and the PTA loaded hydrogel against the 4T1 mouse breast cancer cells were evaluated.

12

ACS Paragon Plus Environment

Page 12 of 22

Page 13 of 22 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

Molecular Pharmaceutics

379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408

Fig. 4 The anti-cancer effects of the CuS nanodots and the hydrogel against 4T1 mouse breast cancer cells. A) The fluorescence images of the 4T1 cells after being treated by the CuS nanodots differently, the red color represents the dead cells stained by propidium iodide and the blue color represents the nucleus of all the cells, the white line is the boundary of the 808 nm laser; B) The survival rates of the 4T1 cells measured by MTT test after being treated by the CuS nanodots differently, ** represents statistically high significance (p < 0.01); C) The fluorescence images of the lower 4T1 cells after being treated by the blank hydrogel and CuS hydrogel in the upper transwell differently, the red color represents the dead cells stained by propidium iodide and the blue color represents the nucleus of all the cells.

Fig. 4A illustrates the fluorescence images of the 4T1 cells, with all the cells blue stained and the dead cells red stained. As anticipated, the PBS treatment alone resulted in few cell death, with negligible number of the cells red stained. Moreover, the combination of PBS treatment and 808 nm laser exposure also led to insignificant cell death, with only a few cells red stained. The results indicated that the NIR light did not make detrimental effects on the cell survival and, the NIR light could be regarded as bio-safe to some extents. Furthermore, the CuS nanodots at the concentration of 20 µg/ml did not exert obvious toxicity to cells, as implied by the comparable effects on cell death compared to the PBS control group. The result clued the encouraging biocompatibility of the CuS nanodots. Based on the observation, it was believed that the PTA would produce tolerable side effects to healthy tissues without NIR irradiation. As anticipated, the combination of CuS nanodots treatment and 808 nm laser exposure resulted in significant cell deaths, with almost all the cells under irradiation failed to survive. Furthermore, the overlay image shows a clear dividing line separating the alive and dead cells. Actually, the dividing line was the boundary of the laser light, merely the cells covered by the laser light were sentenced to death. The existence of the dividing line pointed out that the photothermal therapy could be carried out accurately and 13

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

409 410 411 412 413 414 415

precisely. To analyze the toxicity and therapeutic effects of the CuS nanodots quantitatively, MTT test was carried out. Fig. 4B shows the 4T1 cancer cell survival rate after being differently treated. When the cells were free from the laser exposure and treated by the CuS nanodots for 24 h only, the viability of the cells showed a decreasing trend along with the PTA concentration increase, with the survival rate declining to 91.9%, 87.8% and 71.6% at the CuS

416

nanodots concentration of 5, 10, 20 µg/ml, respectively. Once the cells treated by the CuS nanodots at the same concentrations were exposed to laser light, the viability of the cells further declined to 70.6%, 44.4% and 23.5%. The NIR exposure made statistically significant difference to the survival rate of the CuS nanodots treated cells when the PTA concentration reached 5, 10 and 20

417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457

µg/ml. Above results suggested that the CuS nanodots treatment plus the NIR irradiation would kill cancer cells and, the malignant cells could be totally wiped out at a high PTA concentration. As a result, it can be anticipated that the enrichment of the CuS nanodots surrounding the tumor tissues by local administration of the PTA encapsulated hydrogel would bring in promising therapeutic effects. In this research, transwell was employed to examine the therapeutic effects of the CuS hydrogel against the 4T1 cancer cells. The transwell membrane separated the upper PTA embedded hydrogel and the lower cells. Fig. 4C shows the fluorescence images of the cells, with all the cells blue stained and the dead cells red stained. It can be seen that the blank hydrogel alone did not induce cell death, indicating the outstanding compatibility of the Pluronic F127 hydrogel matrix. Moreover, the blank hydrogel plus the laser exposure did not produce cell death either. As discussed in the above section, the NIR irradiation did not make any changes to the hydrogel. The absence of cell death during the combination of blank hydrogel and the laser exposure further proved this point. Fig. 4C also shows that the CuS hydrogel alone was not harmful to the cells, with negligible number of cells red stained. It seems that, despite the same incubation time, the cells treated by the PTA loaded hydrogel exhibited much higher survival rate than the cells treated by the same amount of the nanodots directly, as indicated in Fig. 4B. The difference implied that the encapsulation in the hydrogel reduced the undesirable adverse effects of the CuS nanodots. It was supposed that the hydrogel matrix relieved the unwanted toxicity of the chemicals by “prisoning” them and, played the therapeutic effects through photo-heat conversion only. The capacity of relieving the direct toxicity caused by the metal chemicals renders the hydrogel an ideal candidate to decrease the systemic toxicity of the PTA. Fig. 4C also shows that the cancer cells were almost completely wiped out when the cells were exposed to the CuS hydrogel together with the NIR irradiation. Obviously, the temperature rise caused by the CuS hydrogel under the laser exposure killed the malignant cells. It is worth noting that, despite the same irradiation time and strength as well as the PTA dose, the CuS hydrogel produced much bigger cell death area than the free nanodots, as indicated in Fig. 4A. In other words, concentrated PTA played more promising photothermal therapy than the evenly dispersed PTA of the same dose. That is to say, the hydrogel not only reduced the direct chemical toxicity of the CuS nanodots, but also enhanced the photothermal therapeutic effects. The results further indicated 14

ACS Paragon Plus Environment

Page 14 of 22

Page 15 of 22 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

Molecular Pharmaceutics

458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482

the superiority of the hydrogel in achieving more pronouncing photothermal therapy. Local retention of the CuS nanodots loaded hydrogel In order to reduce the systemic toxicity and enhance the photothermal therapeutic effects, the CuS hydrogel was peritumorally administered. It was supposed that the thermosensitive hydrogel would change into semisolid hydrogel locally, and then the encapsulated CuS nanodots would have a long retention surrounding the tumor tissues and, as a result, play sustained therapeutic effects after one non-invasive administration. The prerequisite of the sustained therapeutic effects is the long retention in vivo. To image the retention of the hydrogel in vivo, near-infrared imaging technique was employed. The near-infrared fluorescence dye DiR labeled hydrogel was used, with the DiR solution used as a control group. Fig. 5A shows the fluorescence images of the breast tumor bearing mice after being peritumorally treated by DiR hydrogel or free DiR solution. The free DiR treated mice showed a rapid signal decrease, with weak fluorescence signal at day 5 and day 10 and almost undetectable fluorescence signal at day 15. The fast fluorescence signal decrease meant the rapid loss of the agent from the tumor site. At the same time, the DiR hydrogel treated mice saw a nearly constant fluorescence signal, with the signal strength at day 10 comparable to the original signal strength at day 0 and only a slight signal decrease at day 15. The stable fluorescence strength indicated little loss of the agent from the tumor site. In other words, the in-situ hydrogel guaranteed a promising retention of the treating agents surrounding the tumor site and, as a result, provided the basis of sustained therapeutic effects.

15

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

16

483

ACS Paragon Plus Environment

Page 16 of 22

Page 17 of 22 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

Molecular Pharmaceutics

484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503

Fig. 5 In vivo performances of the CuS hydrogel in tumor bearing mouse model. A) The retention of the DiR labeled hydrogel and free DiR solution after peritumoral administration (Ex/Em: 740/820 nm); B) and C) The thermographs and the corresponding heating curves of the tumor sites after peritumoral administration of the blank hydrogel or CuS hydrogel plus 808 nm irradiation (1 W/cm2); D) Pictures of the tumor tissues after photothermal therapy (1 W/cm2, 5 min) in eight sequential days, the red circle represents visibly undetectable tumor sample; E) Statistic graph of the tumor weights, * represents statistical significance (p < 0.05); F) Body weight curves of the tumor bearing mice throughout the whole experiment period; G) Hematoxylin & Eosin staining of the tumor tissues; H) Hematoxylin & Eosin staining of the heart, liver, spleen, lung and kidney of the CuS hydrogel treated mouse.

504

temperature reaching 45.9 °C at the end of irradiation. The temperature increase was probably caused by the direct tissue heating effect of the NIR. Meantime, the CuS hydrogel treated mouse displayed a sharp temperature increase at the tumor site upon the laser exposure, with the temperature

505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533

In vivo anti-cancer performance In this section, the anti-cancer performance of the CuS nanodots embedded hydrogel was evaluated in a breast cancer bearing mouse model, with the blank hydrogel as a control. Fig. 5B shows the thermographs of the CuS hydrogel or blank hydrogel peritumorally treated tumor bearing mice upon the 808 nm laser irradiation. Fig. 5C illustrated the temperatures of the tumor tissue at varied time points. It can be seen that the blank hydrogel treated mouse exhibited a moderate temperature increase at the tumor site upon the NIR exposure, with the

approaching 90 °C at the end of NIR irradiation. The results implied the outstanding photo-heat transition ability of the PTA loaded hydrogel in vivo. The satisfying photo-heat transition capacity was supposed to provide a promising photothermal therapeutic effect against cancer. After the NIR irradiation in eight sequential days, the tumor tissues of the test and control groups were collected, as shown in Fig. 5D. Despite the same laser strength and lasting time, the tumor samples of the blank hydrogel treated mice were obviously bigger than that of the CuS hydrogel treated mice. Specially, one mouse peritumorally treated by the CuS hydrogel plus the NIR irradiation was totally cured, with the tumor tissue visibly undetectable at the end of the experiment. The tumor weights between the test and control group were statistically different (p < 0.05), with the mean tumor weight 0.35 g and 0.76 g respectively, as illustrated in Fig. 5E. The above results indicated the excellent photothermal therapeutic effects of the CuS nanodots encapsulated hydrogel against cancer. To evaluate the systemic toxicity of the treatments, the body weights of the mice were measured. Fig. 5F shows that the mice of both groups had a nearly constant body weight, with a small initial fluctuation probably due to the inadaptation to anesthesia in the first few days. In order to further evaluate the systemic toxicity of the CuS hydrogel, main organs of the mice after the repeated NIR treatment were sliced and H & E stained. Heart, liver, spleen, lung and kidney all displayed normal morphology, as shown in Fig. 5H. The relatively stable body weight and normal morphology of the main organs indicated that the non-invasive administration of the thermosensitive CuS hydrogel plus the laser irradiation induced negligible systemic toxicity. As shown in Fig. 5G, it is worth noting that, comparable to the control group, the necrosis was absent in the tumor tissue of the test group. It was 17

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582

estimated that the heat generated could not penetrate into the core of the tumor tissue to induce cell death, as a result, the necrosis was undetectable in the remaining tumor tissue. The phenomenon clued that the CuS hydrogel played the therapeutic effects through “burning” the cancer cells layer by layer. In summary, it is clear that the injectable and temperature-responsive in-situ CuS hydrogel is a promising PTA for cancer treatment with low systemic toxicity.

Conclusions In this research, an injectable and thermosensitive hydrogel encapsulating CuS nanoparticles for photothermal therapy against cancer was reported. The CuS particles show nanodots structure with a diameter of about 8 nm. The nanodots evenly distributed in the hydrogel matrix and, maintained its particle size unchanged. The PTA loaded hydrogel shows excellent photo-heat conversion ability and outstanding repeated irradiation stability. The hydrogel not only exhibited thermo-responsive in-situ gel-forming capacity at body temperature, but also reduced the chemical toxicity of the CuS by “prisoning” the nanodots in the matrix. Moreover, the concentrated PTA in the hydrogel exhibited more promising therapeutic effects against cancer cells than the nanodots solution. At last, the CuS nanodots embedded hydrogel displayed pronouncing tumor curing effects after peritumoral administration due to the enhanced retention in vivo and elevated local temperature surrounding the tumor tissue upon the NIR exposure. Meantime, the preparation shows insignificant systemic toxicity.

Acknowledgements The research was supported by the Science and Technology Programs of Guangzhou (Grant No. 201604010087), National Natural Science Foundation (Grant No. 81700382), Guangdong Natural Science Foundation (Grant No. 2015A030310428) and, the funding from Guangzhou Medical University (Grant No. B185004204).

Disclosure The authors report no conflicts of interest in this work.

References [1] Tian X, Yang F, Yang C, Peng Y, Chen D, Zhu J, He F, Li L, Chen X. Toxicity evaluation of Gd2O3 @ SiO2 nanoparticles prepared by laser ablation in liquid as MRI contrast agents in vivo. International Journal of Nanomedicine 2014; 9: 4043-4053. [2] Luo CF, Hou N, Tian J, Yuan M, Liu SM, Xiong LG, Luo JD, Chen MS. Metabolic profile of puerarin in rats after intragastric administration of puerarin solid lipid nanoparticles. International Journal of Nanomedicine 2013; 8: 933-940. [3] Lin M, Tang S, Zhang C, Chen H, Huang W, Liu Y, Zhang J. Euphorbia factor L2 induces apoptosis in A549 cells through the mitochondrial pathway. Acta Pharmaceutica Sinica B. 2017; 7(1): 59-64. [4] Xia Y, Xu T, Wang C, Li Y, Lin Z, Zhao M, Zhu B. Novel functionalized nanoparticles for tumor-targeting co-delivery of doxorubicin and siRNA to enhance cancer therapy. International Journal of Nanomedicine 2018; 13: 143-159. [5] Li Y, Lin Z, Guo M, Xia Y, Zhao M, Wang C, Xu T, Chen T, Zhu B. Inhibitory activity of selenium nanoparticles functionalized with oseltamivir on HINI 18

ACS Paragon Plus Environment

Page 18 of 22

Page 19 of 22 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

Molecular Pharmaceutics

583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632

influenza virus. International Journal of Nanomedicine 2017; 12: 5733-5743. [6] Li Y, Guo M, Lin Z, Zhao M, Xiao M, Wang C, Xu T, Chen T, Zhu B. Polyethylenimine-functionalized silver nanoparticle-based co-delivery of paclitaxel to induce HepG2 cell apoptosis. International Journal of Nanomedicine 2016; 11: 6693-6702. [7] Wang H, Yin H, Yan F, Sun M, Du L, Peng W, Li Q, Feng Y, Zhou Y. Folate-mediated mitochondrial targeting with doxorubicin-polyrotaxane nanoparticles overcomes multidrug resistance. Oncotarget 2015; 6 (5): 2827-2842. [8] Yang R-M, Fu C-P, Fang J-Z, Xu X-D, Wei X-H, Tang W-J, Jiang X-Q, Zhang L-M. Hyaluronan-modified superparamagnetic iron oxide nanoparticles for bimodal breast cancer imaging and photothermal therapy. International Journal of Nanomedicine 2017; 12: 197-206. [9] Zhang R, Lin Z, Lui C-H, Wong K-Y, Tam K-H, Lee P, Lok C-N, Lamb J, Chen Y, Xia H. Silver nanoparticle treatment ameliorates biliary atresia syndrome in rhesus rotavirus inoculated mice. Nanomedicine 2017; 13 (3): 1041-1050. [10] Lin L, Cai M, Deng S, Huang W, Huang J, Huang X, Huang M, Wang Y, Shuai X, Zhu K. Amelioration of cirrhotic portal hypertension by targeted cyclooxygenase-1 siRNA delivery to liver sinusoidal endothelium with polyethylenimine grafted hyaluronic acid. Nanomedicine 2017; 13 (7): 2329-2339. [11] Brisa Pen, Susanna Bosi, Brian A. Aguado, Daniele Borin, Nikki L. Farnsworth, Evgenia Dobrinskikh, Teisha J. Rowland, Valentina Martinelli, Mark Jeong, Matthew R. G. Taylor, Carlin S. Long, Robin Shandas, Orfeo Sbaizero, Maurizio Prato, Kristi S. Anseth, Daewon Park, Luisa Mestroni. Injectable carbon nanotube-functionalized reverse thermal gel promotes cardiomyocytes survival and maturation. ACS Applied Materials & Interfaces 2017; 9 (37): 31645-31656. [12] Xiaolin Liu, Yunlong Yang, Yan Li, Xin Niu, Bizeng Zhao, Yang Wang, Chunyan Bao, Zongping Xie, Qiuning Lin, Linyong Zhu. Integration of stem cell-derived exosomes with in situ hydrogel glue as a promising tissue patch for articular cartilage regeneration. Nanoscale 2017; 9 (13): 4430-4438. [13] Lina R Nih, Elias Sideris, S. Thomas Carmichael, Tatiana Segura. Injection of microporous annealing particle (MAP) hydrogels in the stroke cavity reduces gliosis and inflammation and promotes NPC migration to the lesion. Advanced Materials. 2017; 29 (32): 1606471. [14] A. Sivashanmugam, Pornkawee Charoenlarp, S. Deepthi, Arunkumar Rajendran, Shantikumar V. Nair, Sachiko Iseki, R. Jayakumar. Injectable shear thinning CaSO4/FGF-18 incorporated chitin-PLGA hydrogels enhances bone regeneration in mice cranial bone defect model. ACS Applied Materials & Interfaces 2017; 9 (49): 42639-42652. [15] Zhiqiang Lin, Dong Mei, Meiwan Chen, Yitao Wang, Xianhui Chen, Zhaoyang Wang, Bing He, Hua Zhang, Xueqing Wang, Wenbing Dai, Yuxin Yin, Qiang Zhang. A comparative study of thermos-sensitive hydrogels with water-insoluble paclitaxel in molecule, nanocrystal and microcrystal dispersions. Nanoscale 2015; 7 (36): 14838-14847. [16] Zhiqiang Lin, Wei Gao, Hongxiang Hu, Kun Ma, Bing He, Wenbing Dai, Xueqing Wang, Jiancheng Wang, Xuan Zhang, Qiang Zhang. Novel thermo-sensitive hydrogel system with nanocrystals: high drug-loading, 19

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682

sustained release and extended local retention guaranteeing better efficacy and lower toxicity. Journal of Controlled Release 2014; 174: 161-170. [17] Lei Sun, Zhenglin Li, Zhuo Li, Ying Hu, Chong Chen, Chenhui Yang, Baosheng Du, Ye Sun, Flemming Besenbacher, Miao Yu. Design and mechanism of core-shell TiO2 nanoparticles as a high-performance photothermal agent. Nanoscale 2017; 9 (42): 16183-16192. [18] Qiutong Jiu, Wenjun Zhu, Dawei Jiang, Rui Zhang, Christopher J. Kutyreff, Jonathan W. Engle, Peng Huang, Weibo Cai, Zhuang Liu, Liang Cheng. Ultra-small iron-gallic acid coordination polymer nanoparticles for chelator-free labeling of 64Cu and multimodal imaging-guided photothermal therapy. Nanoscale 2017; 9 (34): 12609-12617. [19] Bo Li, Xin Wang, Xiaoyu Wu, Guanjie He, Ruoyu Xu, Xinwu Lu, Feng Ryan Wang, Ivan P. Parkin. Phase and morphological control of MoO3-x nanostructures for efficient cancer theranosis therapy. Nanoscale 2017; 9 (31): 11012-11016. [20] Dandan Ding, Wei Guo, Chongshen Guo, Jianzhe Sun, Nannan Zheng, Fei Wang, Mei Yan, Shaoqin Liu. MoO3-x quantum dot for photoacoustic imaging guided photothermal/photodynamic cancer treatment. Nanoscale 2017; 9 (5): 2020-2029. [21] Xiao Han, Zicheng Deng, Zi Yang, Yilong Wang, Huanhuan Zhu, Bingdi Chen, Zheng Cui, Rodney C. Ewing, Donglu Shi. Biomarkerless targeting and photothermal cancer cell killing by surface-electrically-charged superparamagnetic Fe3O4 composite nanoparticles. Nanoscale 2017; 9(4): 1457-1465.. [22] Changyou Zhan, Weiping Wang, Claudia Santamaria, Bruce Wang, Alina Rwei, Brian P. Timko, Daniel S. Kohane. Ultrasensitive phototriggered local anesthesia. Nano Letters 2017; 17 (2): 660-665. [23] Alina Y. Rwei, Bruce Y. Wang, Tianjiao Ji, Changyou Zhan, Daniel S. Kohane. Enhanced triggering of local anesthetic particles by photosensitization and photothermal effect using a common wavelength. Nano Letters 2017; 17 (11): 7138-7145. [24] Hu H, Lin Z, He B, Dai W, Wang X, Wang J, Zhang X, Zhang H, Zhang Q. A novel localized co-delivery system with lapatinib microparticles and paclitaxel nanoparticles in a peritumorally injectable in situ hydrogel. Journal of Controlled Release 2015; 220: 189-200. [25] Lin Z, Mei D, Chen M, Wang Y, Chen X, Wang Z, He B, Zhang H, Wang X, Dai W, Yin Y, Zhang Q. A comparative study of thermo-sensitive hydrogels with water-insoluble paclitaxel in molecule, nanocrystal and microcrystal dispersions. Nanoscale 2015; 7: 14838-14847. [26] Lin Z, Gao W, Hu H, Ma K, He B, Dai W, Wang X, Wang J, Zhang X, Zhang Q. Novel thermo-sensitive hydrogel system with paclitaxel nanocrystals: High drug-loading, sustained drug release and extended local retention guaranteeing better efficacy and lower toxicity. Journal of Controlled Release 2014; 174: 161-170. [27] Min Zhou, Junjie Li, Su Liang, Anil K. Sood, Dong Liang, Chun Li. CuS nanodots with ultrahigh efficient renal clearance for positron emission tomography imaging and image-guided photothermal therapy. ACS Nano 2015; 9 (7): 7085-7096. [28] Abhishek Sahu, Jong Hyun Lee, Hye Gyeong Lee, Yong Yeon Jeong, Giyoong Tae. Prussian blue/serum albumin/indocyanine green as a 20

ACS Paragon Plus Environment

Page 20 of 22

Page 21 of 22 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

Molecular Pharmaceutics

683 684 685 686 687 688 689

multifunctional nanotheranostic agent for bimodal imaging guided laser mediated combinatorial phototherapy. Journal of Controlled Release 2016; 236: 90-99. [29] Liang Cheng, Hua Gong, Wenwen Zhu, Jingjing Liu, Xiaoyong Wang, Gang Liu, Zhuang Liu. PEGylated prussian blue nanocubes as a theranostic agent for simultaneous cancer imaging and photothermal therapy. Biomaterials 2014; 35: 9844-9852.

21

ACS Paragon Plus Environment

Molecular Pharmaceutics 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

57x36mm (299 x 299 DPI)

ACS Paragon Plus Environment

Page 22 of 22