Amphiphilic PEGylated Lanthanide-Doped Upconversion

Jul 25, 2017 - Preparation of the HIV Attachment Inhibitor BMS-663068. Part 9. Active Pharmaceutical Ingredient Process Development and Powder Propert...
30 downloads 14 Views 2MB Size
Article pubs.acs.org/journal/abseba

Amphiphilic PEGylated Lanthanide-Doped Upconversion Nanoparticles for Significantly Passive Accumulation in the Peritoneal Metastatic Carcinomatosis Models Following Intraperitoneal Administration Yilin Gao,† Lang Liu,‡ Bin Shen,† Xiaofeng Chen,§ Li Wang,§ Liya Wang,‡ Wei Feng,† Chunhui Huang,† and Fuyou Li*,† †

Department of Chemistry & Institute of Biomedicine Science & State Key Laboratory of Molecular Engineering of polymers, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China ‡ College of Chemical and Pharmaceutical Engineering, Nanyang Normal University, Nanyang 473061, P.R. China § Center of Analysis and Measurement, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China S Supporting Information *

ABSTRACT: Inorganic nanoparticles have emerged as attractive materials for cancer research, because of their exceptional physical properties and multifunctional engineering. However, inorganic nanoparticle accumulation in the tumors located in the abdominal cavity after intravenous (IV) administration is confined because of the peritoneum−plasma barrier. To improve this situation, we developed lanthanide-doped upconversion nanoparticles (UCNPs), coated by amphiphilic polyethylene glycol (PPEG), serving as a representative of inorganic nanoparticles. Following intraperitoneal (IP) administration into the peritoneal metastatic carcinomatosis models, UCNPs coated by P-PEG (P-PEG-UCNPs) passively accumulated in the cancerous tissues at a larger amount than that in the main normal organs. On the basis of spatial proximity, PPEG-UCNPs administrated via the IP route exhibited higher passive accumulation in the tumors in the abdominal cavity compared to that via the IV route. It is suggested that IP administration could be a promising strategy for inorganic nanoparticles to be efficaciously applied in peritoneal cancer research. KEYWORDS: lanthanide-doped upconversion nanoparticles, amphiphilic polyethylene glycol, peritoneal metastatic carcinomatosis, intraperitoneal administration

1. INTRODUCTION Inorganic nanoparticles have become considerably attractive as imaging and therapeutic agents in oncology due to superior optical, magnetic and X-ray attenuation properties, shapedependent tunabe absorbance/scattering/fluorescence properties and surface modification.1−3 The efficacy of inorganic nanoparticle application in cancer research largely depends on administrating routes. Different administration can bring about significantly different odyssey of inorganic nanoparticles from administration site to site of action.4,5 In spite of various administration routes of agents in clinical, the common way of inorganic nanoparticles delivered into body is intravenous (IV) injection so far.6,7 As reported, under the normal flow condition in the blood vessels, inorganic nanoparticles following IV injection mainly accumulate in the liver and the spleen.8,9 This leads to inadequate accumulation of particles in the cancerous tissues located in the abdominal cavity, due to the peritoneumplasma barrier. In comparison, intraperitoneal (IP) administration appears to be advantageous for peritoneal cancer because of direct delivery of agents into the abdominal cavity.10−12 The peritoneal tumors can therefore be exposed to high agent concentrations based on © XXXX American Chemical Society

spatial proximity. Several clinical trials have demonstrated that IP administration of theranostic agents in nanoparticle formulation, using organic molecules as building blocks, could yield longer overall survival time in peritoneal cancer research, compared to IV administration.13,14 Although inorganic nanoparticles are attractive alternatives to organic nanoparticles in medicine field, based on their unique physicochemical properties, their accumulation dynamics in peritoneal cancer have not been studied quantitatively or qualitatively. Therefore, it is necessary to carry out this research to explore the potential of inorganic nanoparticles following IP administration in peritoneal cancer research. Among various inorganic nanoparticles, lanthanide-doped upconversion nanoparticles (UCNPs) represent upconversion luminescence (UCL). This optical property endows UCNPs extraordinary advantages in biomedical research, such as lack of autofluorescence from biosamples, large penetration depth, low photobleaching, and multiplex upconversion.15−21 Accordingly, Received: June 27, 2017 Accepted: July 25, 2017 Published: July 25, 2017 A

DOI: 10.1021/acsbiomaterials.7b00416 ACS Biomater. Sci. Eng. XXXX, XXX, XXX−XXX

Article

ACS Biomaterials Science & Engineering

NaYF4:Yb,Tm,Er@NaLuF4 was obtained subsequently. Excessive ethanol was added into the cooling solution. It was centrifugated at 14 000 rpm for 10 min three times. The precipitate was collected and dispersed in cyclohexane for subsequent experiments. Cyclohexane dispersing the core−shell nanoparticles was dropped into dichloromethane containing 10 mg P-PEG, and the mixture was stirred overnight at 30 °C. The products were centrifugated at 14 000 rpm for 10 min three times. The precipitate was redispersed in deionized water for utilization. 2.3. Characterization of NaYF4:Yb,Tm,Er@NaLuF4 Nanoparticles Coated by P-PEG. Nanoparticles were dispersed in cyclohexane and dropped on the surface of copper grids for transmission electron microscope (TEM), high-resolution transmission electron microscope (HRTEM) and energy-dispersive X-ray analysis (EDXA), performed by a JEOL JEM-2010F low- to highresolution transmission electron microscope at 200 kV. The nanoparticle size from three different batches of particles was measured via TEM. X-ray power diffraction (XRD) measurement was fulfilled with a Bruker D4X-ray diffractometer (Cu Kα radiation, λ 0.15406 nm). Fourier-transform infrared (FTIR) spectroscopy was carried out with an IR Prestige-21 spectrometer (Shimadzu) from samples in KBr pellets. UCL spectra of nanoparticles were achieved on an Edinburgh FLS-920 spectrometer, with an external 0−3 W adjustable continuous-wave (CW) laser at 980 nm (Connet Fiber Optics, China) instead of the xenon lamp as excitation source. 2.4. Cell Culture. To establish peritoneal metastatic carcinomatosis models, we selected human liver hepatocellular carcinoma cells, HepG2, because of their risk of spread to the peritoneal cavity. The cells were cultured according to the reported protocols.34 Specifically, the cell line was cultured in Gibco RPMI 1640 containing 10% fetal bovine serum and 1% antibiotic at 37 °C in 5% (V/V) CO2 atmosphere. 2.5. Animal Protocols. All animal experiments were performed in compliance with the guidelines of National Institute for Food and Drug Control, China. These experiments were approved by the Institutional Animal Care and Use Committee, School of Pharmacy, Fudan University. The experimental goal was to study the different tumoral accumulation in peritoneal metastatic carcinomatosis models following IP administration, in comparison with that following IV administration. To ensure accuracy of the experiment data, the other parameters, except the administrating ways, were kept the same. Herein, male athymic Balb/C nude mice (4 week-old) were used for the tumoral accumulation experiments. As for the biosafety evaluation, healthy Kunming male mice were used. These mice were purchased from the Second Military Medical University (Shanghai, China). A week after introduced, the nude mice were injected intraperitoneally with 1 × 106 HepG2 cells suspended in 200 μL PBS, to establish peritoneal carcinomatosis models.35 After about 10 days of cells inoculation, the nude mice could represent obviously distended abdomen, indicating that peritoneal metastatic carcinomatosis models were established successfully. These tumor-bearing mice were divided into two groups at random: the IP group and the IV group, which were single injected at the same dosage of P-PEG-UCNPs. 2.6. Distribution of P-PEG-UCNPs in the Peritoneal Metastatic Carcinomatosis Models after IP or IV Injection. Before animal experiments, UCL intensity of the synthesized P-PEGUCNPs dispersed in aqueous solution was detected with the imaging system (In Vivo Xtreme). Upon excitation at 980 nm by a CW laser, UCL signals of the nanoparticles at 800 ± 12 nm were analyzed with Kodak Molecular Imaging Software. The concentration of P-PEGUCNPs could be recommended for bioimaging, when UCL counts were about 3 000. P-PEG-UCNPs at such concentration in 50 μL were administrated into mice. In this study, 40 mg/kg wt was the injecting dosage for P-PEG-UCNPs to perform bioimaging. After IP injection, the mice were euthanized and sacrificed at 1, 6, 12, 24, and 72 h. The peritoneal solid tumors, the heart, the liver, the spleen, the intestines and the kidneys were collected for qualitative detection with Imaging Software. The tissues were digested in HNO3 for 12 h at 60 °C and diluted with deionized water. Inductively coupled plasma-atomic emission spectrum (ICP-AES) assay for Lu3+

UCNPs can be selected to mimic the physiological behavior of the common inorganic nanoparticles in vivo. Nanoparticle design parameters involving size, shape, and surface ligand, should be optimizedT to overcome delivery barriers.22−26 Blanco suggested that spherical particles averaging less than 50 nm in size could avoid nonspecial uptake by resident macrophages of the mononuclear phagocyte system as much as possible.24 Besides, amphiphilic polyethylene glycol (P-PEG) remains the most widely used ligand, because of its excellent biocompatibility and poor immunogenicity.27−29 Hence, in the present study, ytterbium (Yb), thulium (Tm), and erbium (Er) ion-doped sodium yttrium (Y) fluoride nanoparticles (NaYF4 : 20%Yb, 0.4%Tm, 1.6%Er) were synthesized, following NaLuF4 shell coating. The core−shell nanoparticles, NaYF4: 20%Yb, 0.4%Tm, 1.6%Er@NaLuF4, served as the representative inorganic nanoparticles. Such spherical nanomaterials with oleic acids (OA) were coated by P-PEG. The developed nanoparticles in the average size of less than 50 nm were intraperitoneally or intravenously administrated into the peritoneal metastatic carcinomatosis models, arising from several gastrointestinal and pancreatic cancer in clinical. P-PEG-UCNP distribution in cancer models after IP or IV administration were analyzed quantitatively and qualitatively. Additionally, the biosafety of P-PEG-UCNPs administrated via the IP route at the dosage used for UCL imaging in vivo was evaluated. This study acts as a fundamental verification that P-PEG-UCNPs following IP injection can be effectively applied in peritoneal metastatic carcinomatosis research.

2. EXPERIMENTAL SECTION 2.1. Materials. Deionized water was employed in this experiment. Rare earth oxides with purity of 99.999%, including Y2O3, Yb2O3, Tm2O3, Er2O3, were purchased from Shanghai Yuelong New Materials Co., Ltd. The corresponding chlorides were prepared according to the method reported by Li et al.30 OA and 1-octadecane (ODE, 90%) were purchased from Alfa Aesar Ltd. Poly(maleic anhydride-alt-1octadecene) and 1-ethyl-3-(3-(dimethylamino)propyl) carbodiimide hydrochloride (EDC·HCl) were purchased from J&K Scientific Ltd. Chloroform, absolute ethanol and cyclohexane were purchased from Sinopharm Chemical Reagent Co. Ltd. Poly(ethylene glycol) methyl ether was prepared into amphiphilic block-polymer poly(maleic anhydride-alt-1-octadecene)-PEG, based on the literature stated by Prenciple et al.31 Roswell Park Memorial Institute’s medium (RPMI) 1640 culture solution was bought from Hangzhou Jinuo Biomedical Co., Ltd. 2.2. Synthesis of NaYF4:Yb,Tm,Er@NaLuF4 Nanoparticles Coated by P-PEG. OA-NaYF4: Yb, Tm, Er nanoparticles were prepared by a modified solvothermal process in accordance with the literature.32 0.78 mmol YCl3, 0.20 mmol YbCl3, 0.004 mmol TmCl3, and 0.016 mmol ErCl3 were mixed with 8.0 mL OA and 15 mL ODE. The reactive solution was heated at 140 °C, and then cooled; 2.5 mmol of NaOH and 4.0 mmol of NH4F, dissolved in 15 mL of methanol, were stirred for 30 min at 90 °C. The solution was later adjusted to 300 °C, maintaining for 1 h at argon atmosphere. The solution was cooled to ambient temperature, and ethanol at an excessive amount was added. The resultant mixture was centrifuged at 14 000 rpm for 10 min. The precipitate was collected and washed with ethanol twice again. The as-prepared NaYF4:Yb,Tm,Er particles were dispersed in cyclohexane, serving as the core. The core/shell UCNPs were prepared with an epitaxial growth method;33 1.0 mmol of LuCl3 was added into the compound of 6.0 mL of OA and 15 mL of ODE, and stirred at 140 °C. After 30 min, the transparent solution was dropped into NaYF4:Yb,Tm,Er. The solution was constantly stirred for 30 min at 90 °C. 4.0 mmol of NH4F and 2.5 mmol of NaOH were added. The solution was heated at 90 °C for 30 min and later adjusted to 300 °C, keeping for 1 h in an argon atmosphere. B

DOI: 10.1021/acsbiomaterials.7b00416 ACS Biomater. Sci. Eng. XXXX, XXX, XXX−XXX

Article

ACS Biomaterials Science & Engineering concentration, offered by Research Centre for Analysis & Measurement, Fudan University, was performed on a P-4010 inductively coupled plasma atomic emission spectrometer (Hitachi Limited, Japan), with a set of calibration samples. Passive accumulating effectiveness of P-PEG-UCNPs in the tumors was evaluated by mass accumulations (%ID) of Lu3+, or unit mass accumulations (%ID/g) of Lu3+. The former was calculated by comparing the amounts of Lu3+ in certain tissues with initial injected dose (ID),36 and the later was calculated through comparison of amounts in certain tissues with initial injected dosage per gram.37 The corresponding values were evaluated as mean ± standard deviation of Lu3+ for three mice per time-point. As the control, the IV group was performed and analyzed in the same way. 2.7. UCL Confocal Imaging of Tumors after IP or IV Injection. The mice injected with P-PEG-UCNPs were sacrificed and their solid tumors in the abdominal cavity were undertaken sequential process, involving being excised, fixed in 4% paraformaldehyde, embedded in paraffin and sectioned into 5 μm thickness.38 The treated sections were observed under laser scanning UCL microscopy with an Olympus FV1000 scanning unit. UCL signals in the channel of 600−700 nm were collected at excitation of 980 nm. 2.8. Biosafety Evaluation of P-PEG-UCNPs Following IP Administration at the Dosage Used for UCL Imaging in Vivo. Kunming male mice were intraperitoneally injected with P-PEGUCNPs at a dose of 40 mg/kg wt. This dose was used for the above UCL imaging. Mice receiving the injection solution without P-PEGUCNPs were set as the control. Administrated mice, of which blood was collected for serology analysis at 24 h and 60 d, were sacrificed. Utilizing the collected blood, routine hematological assay was carried out to confirm the difference between the experimental group and the control group. The indices included white blood cell (WBC), neutrophil granulocyte (NEUT), mononuclear macrophage (MONO), eosinophile granulocyte (EO), lymphocyte (LYMPH) and red blood cell (RBC).39 The sera were separated from blood samples so as to study the influence of nanoparticles via IP injection on the liver and the renal function in the normal organism. The indices contained aspartate transaminase (AST), alanine transaminase (ALT), alkaline phosphatase (ALP), globulin (GLOB), albumin (ALB), total protein (TP), creatinine (CRE), uric acid (UA), and blood urea nitrogen (BUN).40 Furthermore, to analyze the possible injury after injection, we prepared the organs into sections with 4 μm thickness, and then stained then with hematoxylin and eosin for pathological examination.41 2.9. Statistical Analysis. The quantitative data were averages ± SD of at least three separate experiments. Statistical significance was assessed with an unpaired Student’s t-test, in which p values of less than 5% were considered statistically significant, as well as less than 1%, high significance.

Figure 1. (a) TEM image of NaYF4:Yb,Tm,Er. (b) TEM image of NaYF4:Yb,Tm,Er@NaLuF4 coated with OA. (c) TEM image of PPEG-NaYF4:Yb,Tm,Er@NaLuF4. (d) HRTEM image of P-PEGNaYF4:Yb,Tm,Er@NaLuF4.

3. RESULTS AND DISCUSSION 3.1. Characterization of NaYF4:Yb,Tm,Er@NaLuF4 Nanoparticles Coated by P-PEG. The as-prepared OANaYF4:Yb,Tm,Er and OA-NaYF4:Yb,Tm,Er@NaLuF4 were measured to be 24.8 ± 0.07 nm and 30.0 ± 0.06 nm in diameter, respectively (Figure 1a, b). P-PEG-UCNPs were measured to be approximately 30.3 ± 0.02 nm (Figure 1c, d). As illustrated (Figure 2a), XRD patterns of core and core−shell of nanocrystals were in accordance with hexagonal-phase NaYF 4 and NaLuF 4 (JCPSD No.16−0334 and 27− 0726).42−44 Ligand modification was verified with FTIR spectroscopy (Figure S1). The bands of P-PEG-UCNPs at 2926 and 2862 cm−1 were respectively ascribed to asymmetric and symmetric C−H stretching, suggesting the existence of OA on the surface of UCNPs.45 Compared with OA-UCNPs, PPEG-UCNPs presented a new peak at 1105 cm−1. This corresponded to the stretching vibrations of C−O−C group which was confirmed by the peak at 1108 cm−1 in P-PEG FTIR spectrum.46 Thus, it was proved that P-PEG was successfully

Figure 2. (a) XRD patterns of as-prepared OA-NaYF4:Yb,Tm,Er, NaYF4:Yb,Tm,Er@NaLuF4, and standard data of hexagonal-phase βNaYF4 (JCPDS No.16−0334) and β-NaLuF4 (JCPDS No.27−0726). (b) Luminescence spectra of NaYF4:Yb,Tm,Er and NaYF4:Yb,Tm,Er@ NaLuF4 upon excitation at 980 nm.

coated on the surface of UCNPs. Lu elements were present in OA-NaYF4:Yb,Tm,Er@NaLuF4, compared with OA-NaYF4:Yb,Tm,Er nanoparticles (Figure S2). Upon excitation from a CW 980 nm laser, NaYF4:Yb,Tm,Er and NaYF4:Yb,Tm,Er@ C

DOI: 10.1021/acsbiomaterials.7b00416 ACS Biomater. Sci. Eng. XXXX, XXX, XXX−XXX

Article

ACS Biomaterials Science & Engineering NaLuF4, both dispersed in cyclohexane, represented five characteristic UCL emission bands at 474 nm, 540 nm, 645 nm, 695 and 803 nm, separately assigned to 1G4 → 3H6 of Tm3+, 4S3/2 → 4I15/2 of Er3+, 4F9/2 → 4I15/2 of Er3+, 3F3 → 3H6 of Tm3+, and 3H4 → 3H6 of Tm3+ (Figure 2b).47−49 3.2. Distribution of P-PEG-UCNPs in the Peritoneal Carcinomatosis Models after IP or IV Injection. To examine distribution of nanoparticles in tumor-bearing mice, PPEG-UCNPs dispersed in physiological saline were injected via the IP route, while P-PEG-UCNPs that were injected via the IV route served as the control. After IP administration, the amounts of P-PEG-UCNPs accumulating in the tumors were higher than that in the liver, the heart, the spleen, the intestines and the kidneys within 24 h (Figure 3a). It was suggested that

passive accumulation in the tumors in the peritoneal cancer models. Moreover, based on the comparison of P-PEG-UCNP accumulation in the tumors in the IP group with that in the IV group (Figure 4), passive tumor-targeting effectiveness of P-

Figure 4. Comparison of unit mass accumulation (%ID/g) of Lu3+ in the tumors of the cancer models following IP or IV injection of PPEG-NaYF4:Yb,Tm,Er@NaLuF4 at different time points (1, 6, 12, 24, and 72 h) (n = 3).

PEG-UCNPs following IP injection was higher than that following IV injection, as %ID/g of Lu3+ in the IP group was more 5.0−6.5 times than that in the IV group within 24 h. Particularly, in the IP group, accumulating amounts of P-PEGUCNPs in the tumors at 12 h was the highest. Hence, these results demonstrated that IP injection was favorable for P-PEGUCNPs to efficaciously accumulate in the tumors located in the abdominal cavity. Imaging was also performed to validate the quantitative results above. Marked UCL signals were observed in the tumors in the mesentery, the liver, the pancreas, the epididymis and the diaphragm within 24 h after IP administration (Figure 5 and Figure S3). In contrast, no positive signals were captured in the tumors within 24 h in the IV group, except marked luminescence signals in the tumors in the diaphragm within 6 h due to the systemic circulation and the lymphatic drainage. It is indicated that P-PEG-UCNPs following IP injection yielded faster and more prolonged accumulation in the peritoneal tumors, in comparison with that following IV injection. This result is in accordance with the conclusion verified by Colby.51 Generally, based on the extensive study of nanoparticle accumulation in the tumors, it has been demonstrated that nanoparticles with different size, shape, and surface characteristics mainly depend on passive or active mechanism of accumulation in the cancerous tissues.52 Specifically, the passive accumulation of nanoparticles relies on enhanced permeability and retention (EPR) effect involving extended retention due to increased leakiness of neovascularization and impaired lymphatic drainage in the tumors. On the other hand, active mechanism of nanoparticles in the tumors may be through the combination between biological targeting ligands of the particles and the special target antigens on the surface of cancer cells. 3.3. UCL Confocal Imaging of Tumors after IP or IV Injection. To further examine the difference of nanoparticle distribution in the substructure of the solid tumors between the IP and the IV groups, the tumors in the abdominal cavity of the mice at 12 h after IP or IV injection were excised and fabricated

Figure 3. Mass accumulation (% ID) of Lu3+ in the main normal organs (the heart, the liver, the spleen, the intestines and the kidneys) and the tumors of the cancer models at different time points (1, 6, 12, 24, and 72 h) after (a) IP injection or (b) IV injection of P-PEGNaYF4:Yb,Tm,Er@NaLuF4 (n = 3).

passive targeting efficiency of P-PEG-UCNPs in the peritoneal tumors was higher than that in the major normal organs through IP administration. Besides, in the control group, PPEG-UCNPs administrated via the IV route were dominantly absorbed in the liver and the spleen, only a few in the tumors within 24 h (Figure 3b). Specifically, at 6 h, Lu3+ of P-PEGUCNPs was detected in the liver at 55.8%ID, more 12 times than that in the tumors (4.3%ID). Lu et al. reported that IV administration resulted in poor tumoral delivery with less than 1% of the injected dose accumulating in the tumors, contributing to systemic distribution and rapid first-pass clearance.50 These results further indicated that P-PEGUCNPs administered via the IP route resulted in significantly D

DOI: 10.1021/acsbiomaterials.7b00416 ACS Biomater. Sci. Eng. XXXX, XXX, XXX−XXX

Article

ACS Biomaterials Science & Engineering

Figure 5. Ex vivo images of the cancer models within 72 h post (a1−a5) IP or (b1−b5) IV injection. UCL signals were collected at 800 ± 12 nm under excitation with a CW 980 nm laser. Note: MT, tumor in mesentery; ET, tumor in epididymis; LT, tumor in liver; PT, tumor in pancreas.

Figure 6. NIR fluorescence distribution in slices of the disseminated solid tumors in the cancer models injected via (a) IP or (b) IV injection at 12 h. UCL signals were collected at 600−700 nm under excitation with a CW 980 nm laser. Scale bars represented 200 μm.

into sections with 5 μm thickness and then observed by laser scanning UCL microscopy. Upon excitation at 980 nm, UCL signals at 645 nm were observed in the tumors in the mesentery, the epididymis, the liver, the pancreas and the diaphragm in the IP group, whereas scattered signals were observed only in the tumors in the liver and the diaphragm in the IV group (Figure 6). Furthermore, UCL signals were positively detected in the exterior of the tumors in the IP group, as well as in the interior of the tumors in the IV group. It demonstrated that tumoral accumulation of P-PEG-UCNPs in the cancer models following IP administration was different from that following IV administration. 3.4. Biosafety Evaluation of P-PEG-UCNPs Following IP Administration at the Dosage Used for UCL Imaging in Vivo. To investigate physiological influence of P-PEGUCNPs following IP injection at the dosage used for

bioimaging, hematological and histological tests were performed. Routine blood indices in the experimental mice at 24 h and 60 d after IP injection were not significantly different from those in the controls receiving intraperitoneal injection solution without P-PEG-UCNPs (Figure 7). However, following biochemical detection, alkaline phosphatase (ALP) and uric acid (UA) levels in the 60 d group were significantly different from the controls. It is suggested that P-PEG-UCNPs via IP injection at the dosage used for bioimaging were biosafe in a short period, but could cause renal function damage over a long period.53,54 In addition, the major organs were taken out and examined through hematoxylin and eosin staining. The results showed that the main tissues were not physiologically damaged after IP injection with P-PEG-UCNPs, except the kidneys at 60 d, according to collections of amyloid in glomerular vascular plexus (Figure 8). It also indicated the renal function injury.55 E

DOI: 10.1021/acsbiomaterials.7b00416 ACS Biomater. Sci. Eng. XXXX, XXX, XXX−XXX

Article

ACS Biomaterials Science & Engineering

Figure 7. (a, b) Blood routine indices of the normal male mice at 24 h and 60 d treated with P-PEG-NaYF4:Yb,Tm,Er@NaLuF4 via IP injection. (c, d) Indexes of liver and renal function of the normal male mice at 24 h and 60 days after IP injection with P-PEG-NaYF4:Yb,Tm,Er@NaLuF4. The mice receiving the injection solution without P-PEG-NaYF4:Yb,Tm,Er@NaLuF4 served as the control. The data were based on six mice per group. **p < 0.01 versus the control.

Figure 8. Histological analysis of the tissues in the normal male mice at 24 h and 60 days after IP injection of P-PEG-NaYF4:Yb,Tm,Er@NaLuF4. The tissues were harvested from the liver, spleen, lung, kidney, and pancreas. Scale bars represented 50 μm.

and so on.56 As reported, size dependent toxicity of superparamagnetic iron oxide nanoparticles was observed in male Sprague−Dawley rats.57 Toxicity of quantum dots likely resulted from liberation of metal ions from the core−shell.58 Repeated intraperitoneal injection of citrate-coated gold nanoparticles in mice within 8 days exhibited most administrated dosage accumulated in the liver followed by the kidneys

The histological results were in agreement with the hematological results. Thus, the working concentration of PPEG-UCNPs in vivo should be adjusted to biosafe level in the further study. According to the present study of nanoparticle toxicology, the degree of long-term toxicity of such materials varies with the instinct physical characteristics, size, shape, surface groups, F

DOI: 10.1021/acsbiomaterials.7b00416 ACS Biomater. Sci. Eng. XXXX, XXX, XXX−XXX

Article

ACS Biomaterials Science & Engineering

(5) Feliu, N. D.; Heine, M.; del Pino, P.; Ashraf, S.; Kolosnjaj-Tabi, J.; Macchiarini, P.; Nielsen, P.; Alloyeau, D.; Gazeau, F.; Stauber, R. H.; Parak, W. J. In vivo degeneration and the fate of inorganic nanoparticles. Chem. Soc. Rev. 2016, 45 (9), 2440−2457. (6) Jia, H. Z.; Wang, W.; Zheng, D. W.; Wang, X.; Yu, W. Y.; Li, S. Y.; Zhuo, R. X.; Zhao, Y. F.; Feng, J.; Zhang, X. Z. Multifunctional nanotherapeutics with all-in-one nanoentrapment of drug/gene/ inorganic nanoparticle. ACS Appl. Mater. Interfaces 2016, 8 (11), 6784−6789. (7) Bednarski, M.; Dudek, M.; Knutelska, J.; Nowinski, L.; Sapa, J.; Zygmunt, M.; Nowak, G.; Luty-Blocho, M.; Wojnicki, M.; Fitzner, K.; Tesiorowski, M. The influence of the route of administration of gold nanoparticles on their tissue distribution and basic biochemical parameters: in vivo studies. Pharmacol. Rep. 2015, 67 (3), 405−409. (8) Yu, J.; Yin, W. Y.; Peng, T.; Chang, Y. N.; Zu, Y.; Li, J.; He, X.; Ma, X. Y.; Gu, Z. J.; Zhao, Y. L. Biodistribution, excretion, and toxicity of polyethyleneimine modified NaYF4:Yb,Er upconversion nanoparticles in mice via different administration routes. Nanoscale 2017, 9 (13), 4497−4507. (9) Fraga, S.; Brandao, A.; Soares, M. E.; Morais, T.; Duarte, J. A.; Pereira, L.; Soares, L.; Neves, C.; Pereira, E.; Bastos, M. d. L.; Carmo, H. Short- and long-term distribution and toxicity of gold nanoparticles in the rat after a single-dose intravenous administration. Nanomedicine 2014, 10 (8), 1757−1766. (10) Dakwar, G. R.; Shariati, M.; Willaert, W.; Ceelen, W.; De Smedt, S. C.; Remaut, K. Nanomedicine-based intraperitoneal therapy for the treatment of peritoneal carcinomatosis - Mission possible? Adv. Drug Delivery Rev. 2017, 108, 13−24. (11) Hasovits, C.; Clarke, S. Pharmacokinetics and pharmacodynamics of intraperitoneal cancer chemotherapeutics. Clin. Pharmacokinet. 2012, 51 (4), 203−224. (12) Tsai, M.; Lu, Z.; Wang, J.; Yeh, T. K.; Wientjes, M. G.; Au, J. L. Effects of carrier on disposition and antitumor activity of intraperitoneal paclitaxel. Pharm. Res. 2007, 24 (9), 1691−1701. (13) Kinoshita, J.; Fushida, S.; Tsukada, T.; Oyama, K.; Watanabe, T.; Shoji, M.; Okamoto, K.; Nakanuma, S.; Sakai, S.; Makino, I.; Furukawa, H.; Hayashi, H.; Nakamura, K.; Inokuchi, M.; Nakagawara, H.; Miyashita, T.; Tajima, H.; Takamura, H.; Ninomiya, I.; Fujimura, T.; Masakazu, Y.; Hirakawa, K.; Ohta, T. Comparative study of the antitumor activity of nab-paclitaxel and intraperitoneal solvent-based paclitaxel regarding peritoneal metastasis in gastric cancer. Oncol. Rep. 2014, 32 (1), 89−96. (14) Fujiwara, K.; Armstrong, D.; Morgan, M.; Markman, M. Principles and practice of intraperitoneal chemotherapy for ovarian cancer. Int. J. Gynecol Cancer 2007, 17 (1), 1−20. (15) Sheng, Y.; Liao, L. D.; Bandla, A.; Liu, Y. H.; Thakor, N.; Tan, M. C. Size and shell effects on the photoacoustic and luminescence properties of dual modal rare-earth-doped nanoparticles for infrared photoacoustic imaging. ACS Biomater. Sci. Eng. 2016, 2 (5), 809−817. (16) Jeyaraman, J.; Shukla, A.; Sivakumar, S. Targeted stealth polymer capsules encapsulating Ln3+-doped LaVO4 nanoparticles for bioimaging applications. ACS Biomater. Sci. Eng. 2016, 2 (8), 1330− 1340. (17) Nadort, A.; Zhao, J.; Goldys, E. M. Lanthanide upconversion luminescence at the nanoscale: fundamentals and optical properties. Nanoscale 2016, 8 (27), 13099−13130. (18) Zhou, J.; Liu, Q.; Feng, W.; Sun, Y.; Li, F. Upconversion luminescent materials: advances and applications. Chem. Rev. 2015, 115 (1), 395−465. (19) Zhou, A.; Wei, Y.; Chen, Q.; Xing, D. In vivo near-infrared photodynamic therapy based on targeted upconversion nanoparticles. J. Biomed. Nanotechnol. 2015, 11 (11), 2003−2010. (20) Chen, D.; Tao, R.; Tao, K.; Chen, B.; Choi, S.; Tian, Q.; Xu, Y.; Zhou, G.; Sun, K. Efficacy dependence of photodynamic therapy mediated by upconversion nanoparticles: subcellular positioning and irradiation productivity. Small 2017, 13, 1−11. (21) Yue, C.; Zhang, C.; Alfranca, G.; Yang, Y.; Jiang, X.; Yang, Y.; Pan, F.; Fuente, J.; Cui, D. Near-infrared light triggered ROS-activated theranostic platform based on Ce6-CPT-UCNPs for simultaneous

and spleen, resulting in renal tubules with cloudy inflammation and vacuolar degeneration of nephron.59 Upconversion nanoparticles coated by polyacrylic acids injected at 15 mg/kg wt via the intravenous route did not cause any toxicity in mice for more than 4 months in biochemical and histopathological analysis.43 Surface area played an important role in the biological toxicity of graphenes. The higher surface areas of graphenes increased reactive oxygen species production, leading to worse damage in vivo. Yet their administration into mice at a dosage of 20 mg/kg wt after PEG functionalization, no significant toxicity was observed.60

4. CONCLUSION Here, we fabricated P-PEG-NaYF4:Yb,Tm,Er@NaLuF4, with favorable biocompatibility in a physiological environment. In the peritoneal metastatic carcinomatosis models, P-PEGUCNPs administrated via the IP route accumulated more in the cancerous tissues in the abdominal cavity, compared to that in the normal tissues. Moreover, passive tumoral accumulation of UCNPs following IP injection was higher efficacious than that following IV injection, according to the quantitative results. Meanwhile, UCL confocal imaging showed that absorption of P-PEG-UCNPs in the tumors in the IP group was overtly different from that in the IV groups. In conclusion, P-PEGUCNPs administrated via the IP route at a proper dose could be an excellent platform for peritoneal metastatic carcinomatosis research.



ASSOCIATED CONTENT

S Supporting Information *

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsbiomaterials.7b00416. Figures S1−S3 (PDF)



AUTHOR INFORMATION

Corresponding Author

*E-mail: [email protected]. ORCID

Liya Wang: 0000-0001-9125-859X Wei Feng: 0000-0002-8096-2212 Fuyou Li: 0000-0001-8729-1979 Notes

The authors declare no competing financial interest.

■ ■

ACKNOWLEDGMENTS The authors acknowledge funding support from the National Natural Science Foundation of China (21231004). REFERENCES

(1) Nam, J.; Won, N.; Bang, J.; Jin, H.; Park, J.; Jung, S.; Jung, S.; Park, Y.; Kim, S. Surface engineering of inorganic nanoparticles for imaging and therapy. Adv. Drug Delivery Rev. 2013, 65 (5), 622−648. (2) Kim, D.; Lee, N.; Park, Y. I.; Hyeon, T. Recent advances in inorganic nanoparticle-based NIR luminescence imaging: semiconductor nanoparticles and lanthanide nanoparticles. Bioconjugate Chem. 2017, 28 (1), 115−123. (3) Smith, B. R.; Gambhir, S. S. Nanomaterials for in vivo imaging. Chem. Rev. 2017, 117 (3), 901−986. (4) Nichols, J. W.; Bae, Y. H. Odyssey of a cancer nanoparticle: from injection site to site of action. Nano Today 2012, 7 (6), 606−618. G

DOI: 10.1021/acsbiomaterials.7b00416 ACS Biomater. Sci. Eng. XXXX, XXX, XXX−XXX

Article

ACS Biomaterials Science & Engineering fluorescence imaging and chemo-photodynamic combined therapy. Theranostics 2016, 6 (4), 456−469. (22) Williams, D. F. Biocompatibility pathways: biomaterials-induced sterile inflammation, mechanotransduction, and principles of biocompatibility control. ACS Biomater. Sci. Eng. 2017, 3 (1), 2−35. (23) Moser, B. A.; Steinhardt, R. C.; Esser-Kahn, A. P. Surface coating of nanoparticles reduces background inflammatory activity while increasing particle uptake and delivery. ACS Biomater. Sci. Eng. 2017, 3 (2), 206−213. (24) Blanco, E.; Shen, H.; Ferrari, M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 2015, 33 (9), 941−951. (25) Zhang, W.; Cui, T.; Liu, L.; Wu, Q.; Sun, L.; Li, L.; Wang, N.; Gong, C. Improving anti-tumor activity of curcumin by polymeric micelles in thermosensitive hydrogel system in colorectal peritoneal carcinomatosis model. J. Biomed. Nanotechnol. 2015, 11, 1173−1182. (26) Feuser, P.; Bubniak, L.; Bodack, C.; Valério, A.; Silva, M. C.; Ricci-Júnior, E.; Sayer, C.; Araújo, P. In vitro cytotoxicity of poly (methyl methacrylate) nanoparticles and nanocapsules obtained by miniemulsion polymerization for drug delivery application. J. Nanosci. Nanotechnol. 2016, 16, 7669−7676. (27) Lu, H. D.; Wilson, B. K.; Lim, T. L.; Heinmiller, A.; Prud’homme, R. K. Real-time and multiplexed photoacoustic imaging of internally normalized mixed-targeted nanoparticles. ACS Biomater. Sci. Eng. 2017, 3 (3), 443−451. (28) Gal, N.; Lassenberger, A.; Herrero-Nogareda, L.; Scheberl, A.; Charwat, V.; Kasper, C.; Reimhult, E. Interaction of size-tailored PEGylated iron oxide nanoparticles with lipid membranes and cells. ACS Biomater. Sci. Eng. 2017, 3 (3), 249−259. (29) Maurizi, L.; Papa, A.; Dumont, L.; Bouyer, F.; Walker, P.; Vandroux, D.; Millot, N. Influence of surface charge and polymer coating on internalization and biodistribution of polyethylene glycolmodified iron oxide nanoparticles. J. Biomed. Nanotechnol. 2015, 11, 126−136. (30) Li, Z. Q.; Wang, L. M.; Wang, Z. Y.; Liu, X. H.; Xiong, Y. J. Modification of NaYF4:Yb,Er@SiO2 nanoparticles with gold nanocrystals for tunable green-to-red upconversion emissions. J. Phys. Chem. C 2011, 115 (8), 3291−3296. (31) Prencipe, G.; Tabakman, S. M.; Welsher, K.; Liu, Z.; Goodwin, A. P.; Zhang, L.; Henry, J.; Dai, H. PEG branched polymer for functionalization of nanomaterials with ultralong blood circulation. J. Am. Chem. Soc. 2009, 131 (13), 4783−4787. (32) Liu, Q.; Chen, M.; Sun, Y.; Chen, G.; Yang, T.; Gao, Y.; Zhang, X.; Li, F. Multifunctional rare-earth self-assembled nanosystem for trimodal upconversion luminescence/fluorescence /positron emission tomography imaging. Biomaterials 2011, 32 (32), 8243−8253. (33) Zhao, L. Z.; Peng, J. J.; Huang, Q.; Li, C. Y.; Chen, M.; Sun, Y.; Lin, Q.; Zhu, L. Y.; Li, F. Y. Near-infrared photoregulated drug release in living tumor tissue via yolk-shell upconversion nanocages. Adv. Funct. Mater. 2014, 24 (3), 363−371. (34) Medina, S. H.; Tiruchinapally, G.; Chevliakov, M. V.; Durmaz, Y. Y.; Stender, R. N.; Ensminger, W. D.; Shewach, D. S.; Elsayed, M. E. Targeting hepatic cancer cells with PEGylated dendrimers displaying N-acetylgalactosamine and SP94 peptide ligands. Adv. Healthcare Mater. 2013, 2 (10), 1337−1350. (35) Aoki, H.; Aoki, M.; Katsuta, E.; Ramanathan, R.; Idowu, M. O.; Spiegel, S.; Takabe, K. Host sphingosine kinase 1 worsens pancreatic cancer peritoneal carcinomatosis. J. Surg. Res. 2016, 205 (2), 510−517. (36) Merian, J.; De Souza, R.; Dou, Y.; Ekdawi, S. N.; Ravenelle, F.; Allen, C. Development of a liposome formulation for improved biodistribution and tumor accumulation of pentamidine for oncology applications. Int. J. Pharm. 2015, 488 (1−2), 154−164. (37) Zhang, C.; Zhou, L.; Zhang, J.; Fu, Y. Y.; Zhang, X.; Yu, C.; Sun, S. K.; Yan, X. P. Green and facile synthesis of a theranostic nanoprobe with intrinsic biosafety and targeting abilities. Nanoscale 2016, 8 (36), 16204−16211. (38) Simon-Gracia, L.; Hunt, H.; Scodeller, P.; Gaitzsch, J.; Kotamraju, V. R.; Sugahara, K. N.; Tammik, O.; Ruoslahti, E.; Battaglia, G.; Teesalu, T. iRGD peptide conjugation potentiates

intraperitoneal tumor delivery of paclitaxel with polymersomes. Biomaterials 2016, 104, 247−257. (39) Zhang, X. D.; Wu, H. Y.; Wu, D.; Wang, Y. Y.; Chang, J. H.; Zhai, Z. B.; Meng, A. M.; Liu, P. X.; Zhang, L. A.; Fan, F. Y. Toxicologic effects of gold nanoparticles in vivo by different administration routes. Int. J. Nanomed. 2010, 5, 771−781. (40) Xu, S.; Fan, H.; Yin, L.; Zhang, J.; Dong, A.; Deng, L.; Tang, H. Thermosensitive hydrogel system assembled by PTX-loaded copolymer nanoparticles for sustained intraperitoneal chemotherapy of peritoneal carcinomatosis. Eur. J. Pharm. Biopharm. 2016, 104, 251− 259. (41) Tian, B.; Wang, Q.; Su, Q.; Feng, W.; Li, F. In vivo biodistribution and toxicity assessment of triplet-triplet annihilationbased upconversion nanocapsules. Biomaterials 2017, 112, 10−19. (42) Cao, T. Y.; Yang, T.; Gao, Y.; Yang, Y.; Hu, H.; Li, F. Y. Watersoluble NaYF4:Yb/Er upconversion nanophosphors: synthesis, characteristics and application in bioimaging. Inorg. Chem. Commun. 2010, 13 (3), 392−394. (43) Xiong, L.; Yang, T.; Yang, Y.; Xu, C.; Li, F. Long-term in vivo biodistribution imaging and toxicity of polyacrylic acid-coated upconversion nanophosphors. Biomaterials 2010, 31 (27), 7078−7085. (44) Liu, Q.; Sun, Y.; Yang, T.; Feng, W.; Li, C.; Li, F. Sub-10 nm hexagonal lanthanide-doped NaLuF4 upconversion nanocrystals for sensitive bioimaging in vivo. J. Am. Chem. Soc. 2011, 133 (43), 17122− 17125. (45) Cao, T.; Yang, Y.; Sun, Y.; Wu, Y.; Gao, Y.; Feng, W.; Li, F. Biodistribution of sub-10 nm PEG-modified radioactive/upconversion nanoparticles. Biomaterials 2013, 34 (29), 7127−7134. (46) Zhu, X.; Da Silva, B.; Zou, X. M.; Shen, B.; Sun, Y.; Feng, W.; Li, F. Y. Intra-arterial infusion of PEGylated upconversion nanophosphors to improve the initial uptake by tumors in vivo. RSC Adv. 2014, 4 (45), 23580−23584. (47) Zhu, X.; Zhou, J.; Chen, M.; Shi, M.; Feng, W.; Li, F. Core-shell Fe3O4@NaLuF4:Yb,Er/Tm nanostructure for MRI, CT and upconversion luminescence tri-modality imaging. Biomaterials 2012, 33 (18), 4618−4627. (48) Hu, S. G.; Yu, Y.; Wu, X. F.; Hu, P.; Cao, H. Y.; Wu, Q. Y.; Tang, Z. J.; Guo, Y. J.; Liu, Y. X. Upconversion luminescence turning of NaREF4 (RE = 0.4Y+0.4La+0.2 (Yb, Er, Tm)) nanoparticles and their applications for detecting Rhodamine B in shrimp. J. Rare Earths 2017, 35 (2), 120−126. (49) Ehlert, O.; Thomann, R.; Darbandi, M.; Nann, T. A four-color colloidal multiplexing nanoparticle system. ACS Nano 2008, 2 (1), 120−124. (50) Lu, Z.; Wang, J.; Wientjes, M. G.; Au, J. L. Intraperitoneal therapy for peritoneal cancer. Future Oncol. 2010, 6 (10), 1625−1641. (51) Colby, A. H.; Oberlies, N. H.; Pearce, C. J.; Herrera, V. L.; Colson, Y. L.; Grinstaff, M. W. Nanoparticle drug-delivery systems for peritoneal cancers: a case study of the design, characterization and development of the expansile nanoparticle. Wiley interdisciplinary reviews. Nanomedicine and nanobiotechnology 2017, 9 (3), e1451. (52) Bertrand, N.; Wu, J.; Xu, X.; Kamaly, N.; Farokhzad, O. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv. Drug Delivery Rev. 2014, 66, 2−25. (53) Ozmen, M.; Yurekli, M. Subacute toxicity of uranyl acetate in Swiss-Albino mice. Environ. Toxicol. Pharmacol. 1998, 6, 111−115. (54) Tokumoto, T.; Tanabe, K.; Ishida, H.; Shimmura, H.; Ishikawa, N.; Goya, N.; Akiba, T.; Toma, H. Impact of fluvastatin on hyperlipidemia after renal transplantation. Transplant. Proc. 2004, 36 (7), 2141−2144. (55) Sethi, S.; Dasari, S.; Amin, M. S.; Vrana, J. A.; Theis, J. D.; Alexander, M. P.; Kurtin, P. J. Clinical, biopsy, and mass spectrometry findings of renal gelsolin amyloidosis. Kidney Int. 2017, 91 (4), 964− 971. (56) Ahmad, M.; Abdel-Wahab, B.; Alam, A.; Zafar, S.; Ahmad, J.; Ahmad, F.; Midoux, P.; Pichon, C.; Akhter, S. Toxicity of inorganic nanoparticles used in targeted drug delivery and other biomedical application: an update account on concern of biomedical nanotoxicology. J. Nanosci. Nanotechnol. 2016, 16, 7873−7897. H

DOI: 10.1021/acsbiomaterials.7b00416 ACS Biomater. Sci. Eng. XXXX, XXX, XXX−XXX

Article

ACS Biomaterials Science & Engineering (57) Zhu, M.; Wang, B.; Wang, Y.; Yuan, L.; Wang, H.; Wang, M.; Ouyang, H.; Chai, Z.; Feng, W.; Zhao, Y. Endothelial dysfunction and inflammation induced by iron oxide nanoparticle exposure: Risk factors for early atherosclerosis. Toxicol. Lett. 2011, 203 (2), 162−171. (58) Zhang, T.; Gerion, D.; Stilwell, J.; Ding, L.; Elboudwarej, O.; Cooke, P.; Gray, J.; Alivisatos, A.; Chen, F. Cellular effect of high doses of silica-coated Qdot profiled with high throughput gene expression analysis and high content cellomics measurements. Nano Lett. 2006, 6, 800. (59) Lasagna-Reeves, C.; Gonzalez-Romero, D.; Barria, M.; Olmedo, I.; Clos, A.; Sadagopa Ramanujam, V. M.; Urayama, A.; Vergara, L.; Kogan, M.; Soto, C. Bioaccumulation and toxicity of gold nanoparticles after repeated administration in mice. Biochem. Biophys. Res. Commun. 2010, 393, 649−655. (60) Yang, K.; Wan, J.; Zhang, S.; Zhang, Y.; Lee, S.-T.; Liu, Z. In vivo pharmacokinetics, long-term biodistribution, and toxicology of PEGylated graphene in mice. ACS Nano 2011, 5, 516−522.

I

DOI: 10.1021/acsbiomaterials.7b00416 ACS Biomater. Sci. Eng. XXXX, XXX, XXX−XXX