Ligand-Modified Cell Membrane Enables the Targeted Delivery of

May 9, 2019 - (Fudan University), Ministry of Education. and PLA. , Shanghai. 201203,. &. State Key Laboratory. of Medical Neurobiology and the ...
0 downloads 0 Views 7MB Size
www.acsnano.org

Ligand-Modified Cell Membrane Enables the Targeted Delivery of Drug Nanocrystals to Glioma Downloaded via BUFFALO STATE on July 18, 2019 at 14:12:07 (UTC). See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Zhilan Chai,†,‡ Danni Ran,†,‡ Linwei Lu,⊥ Changyou Zhan,†,‡,§,# Huitong Ruan,†,‡ Xuefeng Hu,†,‡ Cao Xie,†,‡ Kuan Jiang,†,‡ Jinyang Li,†,‡ Jianfen Zhou,†,‡ Jing Wang,†,‡ Yanyu Zhang,†,‡ Ronnie H. Fang,∥ Liangfang Zhang,*,∥ and Weiyue Lu*,†,‡,○ †

Department of Pharmaceutics, School of Pharmacy and Key Laboratory of Smart Drug Delivery, Ministry of Education and PLA, Fudan University, Shanghai 201203, China ‡ State Key Laboratory of Medical Neurobiology and the Collaborative Innovation Center for Brain Science and §Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China ∥ Department of Nanoengineering and Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States ⊥ The Department of Integrative Medicine, Huashan Hospital, Fudan University and The Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China # State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, China ○ Minhang Branch, Zhongshan Hospital and Institute of Fudan-Minghang Academic Health System, Minghang Hospital, Fudan University, Shanghai 201199, China S Supporting Information *

ABSTRACT: The safe and efficient delivery of chemotherapeutic agents remains critical to anticancer therapy. Herein, we report on a targeted drug delivery system based upon a modified cell membrane coating technique and drug nanocrystals (NCs). Specifically, red blood cell (RBC) membrane was modified with targeting peptides through a facile insertion method involving avidin−biotin interactions. The RBC membrane-coated drug NCs (RBC-NCs) exhibited high drug loading, long-term stability, excellent biocompatibility, and prolonged retention time, all of which make them suitable for effective drug delivery. When modified with the tumor-targeting peptide c(RGDyK), the resulting RGD-RBC-NCs showed superior tumor accumulation and therapeutic efficacy both in mice bearing a subcutaneous tumor as well as orthotropic glioma. RBC-NC therapeutics can be readily generalized to the delivery of various drugs and for the treatment of a wide range of cancers. KEYWORDS: biomimetic nanocarrier, drug delivery, tumor-targeting, drug nanocrystal, glioma

N

boost drug concentration at the targeted sites. Driven by these hypotheses, herein we report on a tumor-targeted nanotherapeutic platform consisting of a ligand-modified cell membrane shell and a drug nanocrystal (NC) core. In recent years, nature-inspired biomimetic nanocarriers have been extensively studied for the treatment of various

anoparticulate delivery systems have long been explored to achieve targeted and controlled delivery of therapeutic agents to tumor tissue and cells.1−3 However, the overall ability of the administered nanotherapeutics to efficiently reach diseased sites can still be improved.4 Hypothetically, the drug concentration at tumor sites can be increased through targeted delivery, in which the specific binding between targeting ligands on the nanoparticle surface and the receptors expressed at tumor sites can lead to preferential accumulation of drug payloads.5−9 Moreover, high drug carrying capacity and loading yield are likely to further © 2019 American Chemical Society

Received: January 25, 2019 Accepted: May 9, 2019 Published: May 9, 2019 5591

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601

Article

Cite This: ACS Nano 2019, 13, 5591−5601

Article

ACS Nano

Figure 1. Preparation and in vitro characterization of RBC-NC(DTX) formulations. (A) Schematic depicting the preparation of targeted RBC-NC. (B) Optimization of DTX mass to RBC membrane ratio by short-term serum stability study. (C−E) TEM imaging of (C) NC(DTX), (D) RBC-NC(DTX), and (E) RGD-RBC-NC(DTX). Scale bars: 50 nm. (F−H) Long-term stability study of different DTX formulations. (F) Size change was measured in PBS, and change in absorbance at 560 nm was measured in both (G) PBS and (H) 10% FBS. (I−K) In vitro drug release from different DTX formulations in (I) PBS at pH 7.4, (J) PBS at pH 6.5, or (K) 10% FBS at 37 °C. Data represents mean with SD; n = 3.

diseases.10−15 Among them, cell membrane-coated nanoparticles have been widely investigated for anticancer therapy.16−18 The proteins and glycosyl groups on the membrane surface endow the nanoparticles with prolonged systematic retention time, less reticuloendothelial system (RES) uptake, and reduced immunorecognition, hence improving drug accumulation in tumor sites due to the enhanced permeability and retention (EPR) effect.19−23 Modifying cell membranes with targeting ligands that bind to tumor sites is likely to further improve the delivery efficiency. However, achieving such surface modification is particularly difficult through conventional chemical reaction approaches, which would compromise the protein profile of the cell membrane. Herein, we developed a robust and versatile

approach to incorporating targeting moieties onto cell membrane-coated nanoparticles. However, the cell membrane-coating strategy has been widely applied on nanostructures such as PLGA nanoparticles, gold nanoparticles, nanogels, and silica nanoparticles,24−27 which generally have moderate drug loading yields. Here, we extended the cell membrane coating technique to drug NCs, which are regarded as “pure particles of drug” with extremely high drug loading yields.28 Due to their intrinsic instability, drug NCs are mostly applied by oral administration or local injection; systemic administration usually produces severe side effects, including capillary blockage and embolism.29−32 We therefore speculated that the use of ligand-modified natural cell 5592

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601

Article

ACS Nano

Figure 2. In vivo toxicity and safety studies of RBC-NC(DTX) formulations in healthy nude mice. Healthy mice were injected with PBS, TXT, NC(DTX), RBC-NC(DTX), and RGD-RBC-NC(DTX) at a single dosage of 25 milligrams of DTX per kilogram of animal body weight. (A−F) H&E-stained histological sections. (A) Heart, (B) liver, (C) spleen, (D) lung, (E) kidney, and (F) brain of mice from each group were observed. Scale bars: 100 μm. (G) Pharmacokinetic behavior of different DTX formulations. Mice were treated with TXT, RBCNC(DTX), and RGD-RBC-NC(DTX) at a dosage of 25 mg DTX/kg (n = 3). (H) Body weight over time (n = 6). (I) WBC counts over time (n = 3). (J−M) Blood biochemical indexes of mice from each group (n = 3). (J) Creatinine (CREA) levels and (K) urea nitrogen levels represent kidney function. (L) Aspartate aminotransferase (AST) levels and (M) alanine aminotransferase (ALT) levels represent liver function. Data represent mean with SD. Single asterisks indicate p < 0.05, double asterisks indicate p < 0.005, and triple asterisks indicate p < 0.001.

membrane to cloak and stabilize drug NCs could represent an effective means of enabling their intravenous administration. In this study, we prepared RBC membrane-coated drug NCs (RBC-NCs) and functionalized them with tumor-targeting ligands for anticancer drug delivery. Specifically, we chose docetaxel (DTX), an insoluble chemotherapeutic drug with severe toxicities,33 as a model payload to synthesize NC(DTX) and RBC-NC(DTX). We subsequently introduced c(RGDyK), a tumor-targeting ligand, to the surface of the RBC membrane utilizing an insertion approach involving multivalent avidin−biotin interactions. In vitro studies verified long-term phosphate-buffered saline (PBS) and serum stability, and in vivo safety evaluation demonstrated the reduced systemic toxicity of RBC-NC(DTX) and RGD-RBC-NC(DTX) compared with NC(DTX). In both anti-subcutaneous tumor- and anti-orthotropic glioma models, the RGD-modified RBC-NC(DTX) showed significantly higher drug accumu-

lation in tumor sites and enhanced antitumor efficacy with a single intravenous administration.

RESULTS AND DISCUSSION Preparation and Characterization of RGD-RBC-NC(DTX). As illustrated in Figure 1A, to prepare RBC-NC(DTX), RBC membrane was derived from RBCs and DTX NCs were prepared using a filming-rehydration method with F127 as the sole excipient. We further incorporated a stable targeting ligand, the peptide c(RGDyK), which binds to the integrins on tumor cells and neovasculature, onto the surface of the RBC membrane. Briefly, streptavidin was introduced to the RBC membrane through a lipid insertion method and then incubated with biotin-linked c(RGDyK). The resulting RBC membrane was then coated onto NC(DTX) through bath sonication at the optimal NC(DTX) to RBC membrane mass ratio of 2.0 (Figure 1B). The successful fusion of RBC membrane with NC(DTX) was confirmed by transmission 5593

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601

Article

ACS Nano

Figure 3. In vitro therapeutic efficacy of RGD-RBC-NC(DTX). (A, B) Interactions between recombinant human integrin αVβ3 and (A) c(RGDyK) peptide or (B) RGD-RVs were assessed by SPR at 25 °C in HBS-EP buffer. (C, D) Stabilization of the microtubule structure by different DTX formulations in (C) HUVECs and (D) U87 cells. All of the cells were stained with α-tubulin antibody (red) and DAPI (blue) followed by imaging using confocal fluorescence microscopy. Scale bars: 10 μm. (E, F) Cytotoxic effect of different DTX formulations on (E) HUVECs and (F) U87 cells was assessed by an MTT assay after 48 h of incubation. Nonlinear regression analysis was performed using GraphPad Prism 6.0 to calculate IC50 values. (G) Effect of different DTX formulations on HUVEC tube formation. (H) U87 cell apoptosis induced by different DTX formulations after BBTB traversal. Data represent mean with SD; n = 3. Single asterisks indicate p < 0.05, and double asterisks indicate p < 0.005.

RBC-NC(DTX), healthy nude mice were administered with PBS, Taxotere (Sanofi, DTX injection concentrate for intravenous infusion, TXT), NC(DTX), RBC-NC(DTX), or RGD-RBC-NC(DTX) at a single dosage of 25 mg DTX/kg via the tail vein. H&E staining of the major organs showed that TXT treatment caused myolysis in the heart and inflammatory infiltration in the liver; NC(DTX) treatment caused thickened alveolar walls and a great deal of inflammatory infiltration in the lungs, suggesting the possibility of pulmonary embolism. RBC-NC(DTX) and RGD-RBC-NC(DTX) treatment showed negligible organ damage in line with the PBS group (Figure 2A−F). Most of the mice in the NC(DTX) group died soon after administration (Figure S3), likely due to capillary blockage and embolism caused by the serious in vivo agglomeration of nanocrystals, as others have previously reported.30 Therefore, further animal experiments were not performed with the NC(DTX) formulation. One of the 8 mice in the TXT group died, while all of those administered with RBC-NC(DTX) or RGD-RBC-NC(DTX) survived at a dosage of 25 mg DTX/kg. A pharmacokinetics study showed that both RBC-NC(DTX) and RGD-RBC-NC(DTX) had a 6fold higher systemic retention time compared with TXT, significantly outperforming the clinically approved formulation (Figure 2G and Table S1). The study looked at the concentration of DTX because it represented a major constituent and the active pharmaceutical ingredient of each formulation. Mice of the TXT group showed obvious body weight decrease during the experiment (Figure 2H). White blood cells (WBCs) were counted 1, 3, 5, 7, 9, and 11 days

electron microscopy (TEM). NC(DTX) displayed irregular shapes (Figures 1C and S1A), while RBC-NC(DTX) and RGD-RBC-NC(DTX) particles showed a core−shell structure with an average size of 70 nm (Figures 1D,E and S1B,C). Long-term stability of different formulations was assessed in PBS and fetal bovine serum (FBS). Because FBS may influence the measurement of size, a previously reported method was used to monitor the agglomeration of particles in its presence.34 Particle size was measured by dynamic light scattering (DLS) in PBS (Figure 1F and Figure S2A), and particle aggregation was evaluated by measuring the absorbance at 560 nm in both PBS (Figure 1G) and FBS (Figure 1H). RBC-NC(DTX) and RGD-RBC-NC(DTX) were stable in both types of media for at least 48 h, while NC(DTX) agglomerated immediately, indicating that the RBC membrane coating could significantly improve the stability of NC(DTX). The surface ζ potential of RGD-RBC-NC(DTX) was slightly higher than RBC-NC(DTX), which was likely due to the positive charge of c(RGDyK) (Figure S2B). We also studied the in vitro drug release kinetics of all formulations. NC(DTX) showed a similar release behavior in PBS at both pH 7.4 (Figure 1I) and pH 6.5 (Figure 1J), while an obvious initial burst profile was observed in 10% FBS (Figure 1K). Interestingly, RBC-NC(DTX) and RGD-RBC-NC(DTX) only released quickly in PBS at pH 6.5. These results suggested the enhanced stability of NC(DTX) and controlled release behavior after RBC membrane coating. In Vivo Toxicity and Safety. To investigate the in vivo safety and biocompatibility of RBC-NC(DTX) and RGD5594

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601

Article

ACS Nano

Figure 4. In vivo antitumor efficacy using a subcutaneous tumor model. Mice were administered with PBS, Taxotere (TXT), RBC-NC(DTX), or RGD-RBC-NC(DTX) at a single dose of 25 milligrams of DTX per kilogram of body weight 7 days after subcutaneous U87 tumor implantation. (A) Biodistribution of DTX formulations. DTX accumulation in the major organs, blood, and tumor tissues of tumor-bearing mice was quantified 2 and 24 h after administration (n = 3). (B) Tumor growth curves of mice. Tumor volumes were measured every 2 days after administration (n = 6). (C) Cumulative survival of mice (n = 6). (D, E) Quantification of integral optical density (IOD) of tumor sections from each group with (D) TUNEL or (E) CD31 immunofluorescence histochemical staining. Mean density: IOD (SUM)/area (n = 3). (F) TUNEL (green) staining and (G) CD31 (green) staining of tumor sections were observed by fluorescence microscopy. Cell nuclei were stained with DAPI (blue). Scale bars: 100 μm. Data represent mean with SD. Single asterisks indicate p < 0.05, double asterisks indicate p < 0.005, and triple asterisks indicate p < 0.001.

after administration. Mice treated with TXT showed a significant decrease in WBC count (Figure 2I), suggesting the occurrence of bone marrow suppression, which is one of the main side effects of DTX. The loss of body weight and decrease in WBC count was significantly attenuated with RBCNC(DTX) and RGD-RBC-NC(DTX) treatment. Liver and kidney functions were evaluated 13 days after administration. Blood biochemistry analysis showed that the creatinine, urea, aspartate aminotransferase, and alanine aminotransferase levels of mice treated with TXT were significantly higher than those of other groups (Figure 2J−M). No significant hematological toxicity was caused by RBC-NC(DTX) or RGD-RBCNC(DTX). The histological and blood biochemistry results confirmed the lower toxicity and better in vivo safety of RBC membrane-coated NCs, possibly due to their enhanced stability, minimized RES uptake, and controlled drug release profile. In Vitro Targeting and Efficacy. With advantages in drug loading, stability, and safety, we subsequently evaluated the

tumor-targeting capability and therapeutic efficacy of the targeted RBC-NC formulation. As an initial study to evaluate the targeting ability of peptide-modified RBC membrane, a surface plasmon resonance (SPR) assay was conducted to assess the binding affinity of RGD-modified RBC vesicles (RGD-RVs) without drug payload to integrin αVβ3. It was interesting that RGD-RVs displayed a higher binding affinity than c(RGDyK) peptide alone (without RBC vesicles) because their equilibrium dissociation constants (KD) were 0.06981 and 4.719 μM, respectively (Figure 3A,B). Given that higher binding affinity can be achieved between several RGDcontaining peptides and integrin αVβ3 protein compared to a single peptide,35 the strengthened interaction of integrin αVβ3 to RGD-RVs was likely attributed to the membrane fluidity effect and the high surface density of the peptide on the RBC membrane facilitated by the multivalent binding capability of streptavidin. Unmodified RVs were taken as a control group and showed no specific binding affinity with integrin αVβ3, as 5595

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601

Article

ACS Nano

Figure 5. In vivo antitumor efficacy using an orthotropic glioma model. Mice were administered with PBS, Taxotere (TXT), RBC-NC(DTX), or RGD-RBC-NC(DTX) at a single dose of 25 mg DTX/kg body weight 10 days after orthotropic U87 glioma implantation. (A) Biodistribution of DTX formulations. DTX accumulation in the brains, blood, and other major organs of mice were quantified 2 and 24 h after administration (n = 3). (B) Kaplan−Meier survival curves of mice (n = 10). (C, D) Quantification of integral optical density (IOD) of glioma sections from each group with (C) TUNEL or (D) CD31 immunofluorescence histochemical staining. Mean density: IOD (SUM)/ area (n = 3). (E) TUNEL (green) staining and (F) CD31 (red) staining of brain (with glioma) sections were observed by fluorescence microscopy. Cell nuclei were stained with DAPI (blue). The yellow dotted lines represent the margins of the glioma and the yellow arrows point to the glioma. Scale bars: 100 μm. Data represent mean with SD. Single asterisks indicate p < 0.05, double asterisks indicate p < 0.005, triple asterisks indicate p < 0.001, and quadruple asterisks indicate p < 0.0001.

that the nontargeted particles had low amounts of nonspecific uptake. The cellular uptake of different DTX formulations was studied by measuring the DTX content in U87 cells and HUVECs (Figure S5). It was confirmed that RGD modification could significantly increase cytosolic DTX concentration in both cell lines. The effects of different DTX formulations on HUVEC tube formation were also evaluated. Cells in culture media only, with enclosed and extensive networks, served as a blank control. RGD-RBC-NC(DTX) exhibited significantly higher inhibition in HUVEC tube formation (Figure 3G), which was consistent with the cytotoxicity results. To assess the blood−brain tumor barrier (BBTB) transcytosis effect of different DTX formulations, a HUVEC/U87 co-culture was used to establish an in vitro BBTB model (HUVECs were seeded in transwell inserts and U87 cells were seeded in the lower chambers). The medium in the transwell inserts was replaced with different DTX formulations and transferred to another plate preseeded with U87 cells. After incubation for 0.5 h, the insets were removed and the cell viability of the HUVEC cells in them was assessed

evidenced by the lack of concentration dependence (Figure S4). Given that DTX’s mode of action is disruption of microtubule dynamics, we visualized the stabilization of microtubules in HUVECs (Figure 3C) and U87 cells (Figure 3D) treated with free DTX and various DTX nanoformulations. After 24 h of exposure to the drugs, the cells were stained with α-tubulin antibody for visualization. The multipolar spindles in cells of the control group were replaced by microtubule bundles in all of the different drug-treated cells. The in vitro cytotoxicity of free DTX, NC(DTX), RBCNC(DTX), and RGD-RBC-NC(DTX) against HUVECs and U87 cells was investigated by the MTT assay. The halfmaximal inhibitory concentration (IC50) was calculated after 48 h of inhibition. RGD-RBC-NC(DTX) exhibited the strongest anti-proliferative effect due to its active targeting capability and increasing cellular uptake, registering an IC50 value of 12.1 nM in HUVECs (Figure 3E) and 13.8 nM in U87 cells (Figure 3F). RBC-NC(DTX) displayed a higher IC50 value than that of free DTX and NC(DTX), which indicated 5596

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601

Article

ACS Nano

time of 62 days (p < 0.001). Brain glioma sections of each group were stained with TUNEL and CD31 for the quantification of apoptosis (Figure 5C,D) and observation of angiogenesis (Figure 5E,F), respectively. Compared to other groups, RGD-RBC-NC(DTX) induced significantly higher apoptosis and angiogenesis inhibition because of its effective and precise targeting to glioma. Tumor sections were also visualized by H&E staining (Figure S9).

to confirm the integrity of the model (Figure S6). After another 24 h of incubation, U87 cell apoptosis was significantly elevated by RGD-RBC-NC(DTX) treatment, indicating that RGD modification boosted transcytosis across the BBTB and targeted tumor cells through the interaction between RGD and integrin (Figure 3H). In Vivo Biodistribution and Antitumor Efficacy on a Subcutaneous Tumor Model. Mice were randomly divided into 4 groups and intravenously administered with PBS, TXT, RBC-NC(DTX), or RGD-RBC-NC(DTX) at a single dosage of 25 mg DTX/kg 7 days after subcutaneous U87 tumor implantation, when the tumors of each group had reached a mean volume of 150 mm3. To investigate the in vivo distribution of different DTX formulations based on this model, the DTX content of major organs and tissues were measured 2 and 24 h after administration (Figure 4A). RBCNC(DTX) showed significantly higher tumor accumulation than TXT at both time points, likely due to effective passive targeting from ready access to tumor vasculature and the EPR effect, which lessened the uptake in other organs; RGD-RBCNC(DTX) displayed an even higher accumulation than both RBC-NC(DTX) and TXT due to its active-targeting capability. An in vivo antitumor study showed excellent tumor growth inhibition achieved by RBC-NC(DTX) and RGD-RBCNC(DTX) (Figures 4B and S7). Tumor volumes of mice were measured every 2 days for 44 days after administration. RBC-NC(DTX) significantly inhibited tumor growth, mainly utilizing the EPR effect as mentioned above. RGD-RBCNC(DTX) achieved the best result because it could actively target tumor cells and their associated neovasculature; the tumors sizes were well controlled throughout the duration of the study, and mice in this group survived the longest time among the four experimental groups (Figure 4C). Tumor sections from each group were stained with TUNEL and CD31 for the quantification of apoptosis (Figure 4D,E) and the observation of angiogenesis (Figure 4F,G), respectively. Compared to the PBS and TXT groups, RBC-NC(DTX) induced apoptosis and inhibited angiogenesis, while tumors treated with RGD-RBC-NC(DTX) displayed higher apoptosis and even less angiogenesis due to the enhanced drug localization at the tumor site. Tumor sections were also visualized by H&E staining (Figure S8). In Vivo Biodistribution and Antitumor Efficacy on an Orthotropic Glioma Model. To investigate the in vivo antiglioma efficacy of RGD-RBC-NC(DTX), mice bearing orthotropic U87 glioma were randomly divided into 4 groups and treated with PBS, TXT, RBC-NC(DTX), or RGD-RBCNC(DTX) at a single dosage of 25 mg DTX/kg 10 days after implantation, at which stage the BBTB emerges as the main obstacle for nanocarriers. To investigate the in vivo distribution of different DTX formulations based on this model, the DTX content of major organs and tissues was measured 2 and 24 h after administration (Figure 5A). RBC-NC(DTX) and TXT could hardly be detected in the brains due to the existence of the BBTB, while RGD modification increased drug accumulation to a greater extent at both time points because RGDRBC-NC(DTX) could traverse the BBTB by receptormediated transcytosis. Similarly, an in vivo antiglioma study showed that treatments with TXT or RBC-NC(DTX) did little to improve mouse survival, registering respective median survivals of 32.5 days and 34.5 days versus 32 days for the PBStreated group (Figure 5B), while RGD-RBC-NC(DTX) treatment achieved a significantly prolonged median survival

CONCLUSIONS In summary, we have successfully established a cell membranecoated drug nanocrystal system with high drug loading, and these nanoparticles were able to specifically and safely deliver large amounts of hydrophobic drug into tumor sites upon intravenous administration. The results confirmed that the nanocrystal core could increase the loading of a poorly watersoluble drug. With an RBC membrane coating, the particles were stabilized, providing improved biocompatibility and decreased side effects. RGD modification bestowed the nanocarrier with active targeting ability, which increased the drug accumulation in tumor sites and further enhanced the efficacy of the loaded chemotherapeutic drug both in a subcutaneous tumor model and an orthotropic glioma model. Regarding future development of the platform, it should be noted that drug nanocrystals have long been studied and several products using the technology have been launched in the past few decades.36 The current infrastructure for blood transfusions could also be leveraged to source the membrane material, and the development of ex vivo blood cell culture techniques could help to further facilitate translation.37 The nanocarrier can be readily generalized for the delivery of various drugs with low solubility or high toxicity, especially those that need to be administered intravenously, such as paclitaxel, doxorubicin, and irinotecan.38,39 Ligands other than RGD could also be easily incorporated onto the surface through biotin functionalization and multivalent binding with streptavidin, although the long-term immunological implications of exposure to the foreign protein will then need to be elucidated. Overall, this therapeutic strategy has potential for the treatment of a wide range of cancers through tumortargeted drug delivery and represents a promising approach that could ultimately be applied in the clinic. METHODS Synthesis of Streptavidin-PEG3400-DSPE and Biotin-PEG3500c(RGDyK). Streptavidin (YeSen Biotech Co. Ltd.) was reacted with a 10-fold excess of Traut’s reagent (Aladdin) in borate buffer (pH 7.6) for 2 h under room temperature. The excess Traut’s reagent was removed by ultrafiltration at 4 °C to obtain streptavidin-SH. MalPEG3400-DSPE (Laysan Bio Inc.) was dissolved in dichloromethane to form a film through rotary evaporation. Streptavidin-SH was dissolved in phosphate (pH 7.4) and subsequently added into the resulting film for hydration. The residual mal-PEG3400-DSPE and phosphate was removed by dialysis in distilled water for 72 h. Biotin-PEG3500-c(RGDyK) was synthesized through the covalent conjugation of c(RGDyK)-SH (GL Biochem Ltd.) with malPEG3500-biotin (Jinpan Biotech Co. Ltd.) by a sulfhydryl-maleimide coupling method.40 1H NMR (400 MHz, DMSO-d6, δ): 12.19 (s, 1H), 9.16 (s, 2H,), 8.60−8.25 (m, 5H), 8.16−8.11 (m, 1H), 8.07 (s, 1H), 7.84 (t, J = 5.6 Hz, 2H), 6.96 (d, J = 8.1 Hz, 2H), 6.62 (d, J = 8.4 Hz, 2H), 6.42 (s, 2H), 6.36 (s, 1H), 4.70−4.58 (m, 1H), 4.53−4.37 (m, 3H), 4.32−4.28 (m, 1H), 4.20−4.08 (m, 1H), 3.96−3.89 (m, 1H), 3.68 (t, J = 4.9 Hz, 2H), 3.52−3.50 (m, 308H), 3.46−3.43 (m, 2H), 3.39 (t, J = 6.0 Hz, 4H), 3.27−3.24 (m, 2H), 3.19−3.09 (m, 6H), 2.82 (dd, J = 12.4, 5.1 Hz, 2H), 2.56 (s, 1H), 2.48−2.37 (m, 5597

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601

Article

ACS Nano

time and dissociation time of each sample to integrin αVβ3 were both 60 s at a flow rate of 30 μL/min. To reduce the nonspecific binding between RGD-RVs and the sensor chip, RVs were coupled to the reference flow cell of CM5 while the binding affinity of RGD-RVs was assessed. Biacore equilibrium responses recorded at the beginning and the end of the association step were used to determine the association constant (Ka) and dissociation constant (Kd). The KD values were calculated through the equation (KD = Kd/Ka). The binding of RVs and integrin αVβ3 were also assessed as a control. Data were analyzed using the Biacore software (GE Healthcare). Cell Culture. U87 cells were purchased from the ATCC and human umbilical vascular endothelial cells (HUVECs) were purchased from the Chinese Academy of Sciences Cells Bank. Both cells were maintained in Dulbecco’s modified Eagle medium (Gibco) supplemented with 10% FBS (Gibco), 100 U/mL penicillin, and 100 μg/mL streptomycin. Cells were cultured at 37 °C under a humidified atmosphere containing 5% CO2. Stabilization of Cell Microtubules. HUVECs and U87 cells were seeded into confocal dishes and treated with free DTX or DTX formulations for 24 h. Then cells were stained with α-tubulin antibody (Sigma-Aldrich) to observe microtubules under a laser scanning confocal microscope (ZEISS). Cytotoxicity Assay. In vitro cytotoxicity of free DTX, NC(DTX), RBC-NC(DTX), and RGD-RBC-NC(DTX) was measured by the MTT assay. HUVECs and U87 cells were seeded into 96-well plates at a density of 3000 cells per 200 microliters per well. After 12 h of culture, cells were treated with different concentrations of DTXloaded formulations for 48 h and the cytotoxicity was determined by the MTT assay. Cytosolic DTX Concentration Study. HUVECs and U87 cells were seeded into 6-well plates. After 12 h of culture, cells were treated with free DTX, NC(DTX), RBC-NC(DTX), and RGD-RBCNC(DTX) at a DTX concentration of 125 nM for 1 h. Cells were collected and washed with 1× PBS. After counting, cells were suspended in 1× PBS and sonicated for 15 min. Methyl alcohol was added into the samples for protein precipitation and DTX extraction. After centrifugation, the supernatant was collected and DTX concentration was analyzed by HPLC. Inhibition of In Vitro HUVEC Tube Formation. 24-well plates were coated with 100 μL of growth factor-reduced Matrigel (BD Biosciences). HUVECs were gently seeded onto the three-dimensional Matrigel overnight for tube formation. HUVECs cultured in the Matrigel were treated with free DTX, NC(DTX), RBC-NC(DTX), and RGD-RBC-NC(DTX) at a DTX concentration of 15 nM. Cells cultured with drug-free DMEM were used as a control group. After 12 h of incubation, tube structures were visualized under an invertedphase contrast microscope (DMI4000 B, Leica). The amount of tubes was calculated from three randomly selected wells. Evaluation of BBTB Transcytosis Effect in a HUVEC/U87 CoCulture Model. The HUVEC/U87 co-culture model was established as previously reported.42−44 Briefly, HUVECs were seeded into transwell inserts, and U87 cells were plated onto the lower chambers at a 1:5 ratio (HUVECs:U87 cells). After co-culturing for 3 days, the upper inserts were transferred to another 24-well plate, which was previously seeded with U87 cells at a density of 50 000 cells per well. In each apical chamber, the culture medium was replaced with free DTX and different DTX formulations containing 1.5 μM DTX. The inserts were removed after 0.5 h of incubation, and HUVEC viability was measured by the MTT assay. After another 24 h of incubation, U87 cells were stained with an Annexin V-FITC/PI apoptosis detection kit (BD Biosciences), and the percentage of apoptosis was analyzed using a FACS AriaII flow cytometer (BD Biosciences). In Vivo Safety Evaluation of DTX Formulations. All animals were purchased from the Shanghai SLAC Laboratory Animal Co. Ltd. and kept under SPF conditions. All animal experiments were carried out in accordance with guidelines evaluated and approved by the ethics committee of Fudan University. To investigate the in vivo toxicity of different DTX formulations, healthy nude mice were randomly divided into 5 groups and intravenously injected with Taxotere (TXT), NC(DTX), RBC-

4H), 2.33 (s, 2H), 2.06 (t, J = 7.4 Hz, 2H), 1.89−1.71 (m, 2H), 1.66−1.55 (m, 2H), 1.54−1.38 (m, 8H), 1.36−1.21 (m, 6H) (Figure S10). RBC Membrane Derivation. Red blood cell membranes were collected according to a previously reported method with modification.41 Briefly, fresh heparinized whole blood was collected from male ICR mice (20−22 g) and subsequently centrifuged at 1000g for 5 min at 4 °C to remove the plasma and the white buffy coat. The collected RBCs were washed with 1× PBS and suspended in 0.25× PBS for 30 min at 4 °C, and then the hemoglobin was removed by centrifugation at 15000g for 7 min. The resulting pink pellet was purified with 1× PBS, and the collected RBC membrane was suspended and stored in distilled water. The protein concentration of RBC membranes was determined using a BCA protein assay kit (Thermo Fisher Scientific Inc.). Preparation of RBC-NC(DTX) and RGD-RBC-NC(DTX). Docetaxel nanocrystals were prepared according to a previously reported method with modification.38 In brief, DTX (Dalian Meilun Bio Co. Ltd.) and Pluronic F127 (1:5, w/w) were dissolved in methanol to form a film through rotary evaporation and dried under a vacuum for 2 h. After 15 min of hydration, the resulting NC(DTX) was quickly mixed with RBC membranes and sonicated for 5 min to prepare RBCNC(DTX). For RGD-RBC-NC(DTX), RBC membranes derived from 100 μL of blood were first incubated with 200 μg of streptavidinPEG3400-DSPE to form streptavidin-inserted RBC membranes. Then, 10 μg of biotin-PEG3500-c(RGDyK) was added into the resulting sample and incubated for 15 min to ensure the complete binding between biotin and streptavidin. The membranes were subsequently mixed with NC(DTX) and sonicated for 5 min. To investigate the optimal RBC membrane ratio, RBC membranes with a protein concentration of 2 mg/mL were mixed with NC(DTX) containing 8, 6, 4, and 2 mg of DTX and sonicated. The formulations were incubated with FBS, and the aggregation of particles was detected by 560 nm absorbance measurements for 2 h. Preparation of RVs and RGD-RVs. RBC membranes were collected and sonicated through bath sonication for 5 min to form RBC membrane vesicles (RVs). RGD-RVs were prepared through the same avidin−biotin approach as stated above. Transmission Electron Microscopy Imaging. The structure of NC(DTX), RBC-NC(DTX), and RGD-RBC-NC(DTX) was observed using a transmission electron microscope. First, 20 μL of particle solution at a DTX concentration of 100 μg/mL was dropped onto a glow-discharged carbon-coated grid. After 5 min, the sample was blotted, and 20 μL of 0.75% phosphotungstic acid stain was added to the grid for 30 s. The grid was subsequently dried and observed using a Tecnai G2 20TWIN microscope (FEI). Stability Study. Particle size (diameter, in nanometers) and surface charge (ζ potential, in millivolts) were measured by dynamic light scattering using a Zen 3600 Zetasizer (Malvern). The 560 nm absorbance measurements were conducted using a Synergy 2 microplate reader (Biotek). Particles were suspended in 1× PBS or 100% FBS, and measurements were performed in triplicate at room temperature for 2 days. In Vitro DTX Release. Dialysis tubes (MWCO: 14000 Da, Millipore) with 400 μL of different DTX formulations were directly immersed into 4 mL of PBS (pH 7.4), PBS (pH 6.5), or 10% FBS with 0.5% Tween-80 (v/v) at 37 °C for 72 h under horizontal shaking at 100 rpm. The medium was withdrawn and replaced with an equal volume of fresh medium periodically. DTX in medium was analyzed by high-performance liquid chromatography (HPLC, 1100 series, Agilent Technologies). Surface Plasmon Resonance Analysis. To assess the binding affinities of RGD-RVs and c(RGDyK) with integrin αVβ3, binding analysis was conducted using a Biacore T200 system (GE Healthcare). Recombinant human integrin αVβ3 (R&D Systems) was coupled to the experiment flow cell of a Series S sensor chip CM5 (GE Healthcare) according to a standard amine coupling procedure. RGD-RVs and c(RGDyK) were dissolved in HBS-EP buffer (GE Healthcare) at defined concentrations and injected. Resonance changes were recorded to assess the binding affinity. The contact 5598

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601

Article

ACS Nano NC(DTX), and RGD-RBC-NC(DTX) at a single dosage of 25 mg DTX/kg or PBS. The weights of the mice in each group were measured every 2 days. The blood of mice from each group was collected before administration and at 1, 3, 5, 7, 9, and 11 days after administration for WBC counting. A total of 13 days after administration, all mice were euthanized, and blood was collected for biochemical parameters measurement, while the heart, liver, spleen, lung, kidney, and brain were excised and stained by H&E for histological analysis. Pharmacokinetics Study. To evaluate the circulation half-life of different DTX formulations, male ICR mice (25 g) were injected with TXT, RBC-NC(DTX), and RGD-RBC-NC(DTX) at a dosage of 25 mg DTX/kg via the tail vein (n = 3). A total of 30 μL of blood was collected at different time points (0, 5, 15, and 30 min, and 1, 2, 4, 8, 24, 48, and 72 h). The collected blood samples were centrifuged to separate the plasma. The amount of DTX, the active pharmaceutical ingredient, in plasma was analyzed by HPLC. Pharmacokinetic parameters were calculated to fit a two-compartment model and a one-way nonlinear model. Biodistribution Study. To investigate the distribution of TXT, RBC-NC(DTX), and RGD-RBC-NC(DTX) in subcutaneous tumoror orthotropic glioma-bearing mice 2 and 24 h after administration (25 mg DTX/kg), mice were euthanized and their hearts, livers, spleens, lungs, kidneys, brains, tumors, and blood were collected. The organs were weighed, homogenized in 1 mL of PBS, and analyzed by HPLC to quantify the concentration of DTX, the active pharmaceutical ingredient of each formulation. Anti-Subcutaneous U87 Tumor Study. Male nude mice (20− 22 g) were subcutaneously implanted with 5 × 106 U87 cells in the right armpit. A total of 7 days after implantation, when the solid tumor size reached approximately 150 mm3, mice were randomly divided into 4 groups and received a single intravenous injection of TXT, RBC-NC (DTX), and RGD-RBC-NC (DTX) with a dosage of 25 mg DTX/kg or PBS. Tumor diameters were measured with a Vernier caliper every 2 days and tumor volumes were calculated using the formula: tumor volume = (length) × (width)2/2. Relative tumor volume was tumor volume at each time point divided by tumor volume at day 7 (the day that mice received treatment). A total of 14 days after administration (at day 21), 3 mice in each group were randomly selected for photographs and then were euthanized for tumor immunofluorescence histochemical analysis. The rest of the mice in each group were observed until death (n = 6). Anti-Orthotropic U87 Glioma Study. The orthotropic glioma model was established according to a previously reported method.45 In brief, male nude mice (20−22 g) were anesthetized with chloral hydrate, and 6 × 105 U87 cells were implanted into the right brain (0.6 mm anterior and 1.8 mm lateral to the bregma with 3 mm depth) with a stereotactic apparatus (Stoteling). A total of 10 days after implantation, mice were randomly divided into 4 groups and received a single intravenous injection of TXT, RBC-NC(DTX), and RGDRBC-NC(DTX) with a dosage of 25 mg DTX/kg or PBS. A total of 10 days after administration (at day 20), 3 mice in each group were randomly selected and euthanized for brain immunofluorescence histochemical analysis. The rest of the mice in each group were observed until death (n = 10). Immunofluorescence Histochemical Analysis. Tumors or brains (with glioma) were fixed with 4.0% paraformaldehyde, embedded in paraffin, and sectioned. TUNEL staining was used to detect apoptotic cells according to a previously reported method.46 CD31 staining was performed to observe the microvessels.47 A total of three different TUNEL or CD31 sections for each group were analyzed by ImageJ to measure the integral optical density and the area of the slice, and the mean density of each slice was calculated. Statistical Analysis. All of the results are presented as mean with SD. Data were analyzed using the Student’s t-test or one-way analysis of variance unless otherwise indicated. The two-sided p < 0.05 was considered statistically significant. Median survival times were compared using the log-rank test. Statistics were calculated using GraphPad Prism 6.0 software.

ASSOCIATED CONTENT

S Supporting Information *

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsnano.9b00661. Figures showing TEM images, size and ζ potential of RBC-NC(DTX) formulations, the survival rate of mice administered with different DTX formulations, interactions of integrin αVβ3, cytosolic concentrations, the integrity of the in vitro BBTB model, photographs of mice bearing U87 subcutaneous tumors, H&E-stained histological sections, and HNMR spectra of biotinPEG3500-c(RGDyK); a table showing pharmacokinetic parameters of different DTX formulations after intravenous administration (PDF)

AUTHOR INFORMATION Corresponding Authors

*E-mail: [email protected]. *E-mail: [email protected]. ORCID

Liangfang Zhang: 0000-0003-0637-0654 Weiyue Lu: 0000-0001-8003-2675 Author Contributions

W.L., L.Z., and Z.C. conceived of the idea and designed the experiments; Z.C., D.R., L.L., H.R., X.H., K.J., J.L., J.Z., J.W., and Y.Z. performed the experiments. Z.C. and C.Z. analyzed the data. C.X. and C.Z. provided suggestions and technical support on the research. Z.C., C.Z., R.H.F., W.L., and L.Z. wrote the paper. All authors contributed to the general discussion. Notes

The authors declare no competing financial interest.

ACKNOWLEDGMENTS This work was supported by the National Basic Research Program of China (973 Program, no. 2013CB932500), the National Natural Science Foundation of China (nos. 81773657, 81690263, and 81473149), the Shanghai Education Commission Major Project (no. 2017-01-07-00-07-E00052), and the Shanghai International Science and Technology Cooperation Project (no. 16430723800). REFERENCES (1) Peer, D.; Karp, J. M.; Hong, S.; Farokhzad, O. C.; Margalit, R.; Langer, R. Nanocarriers as an Emerging Platform for Cancer Therapy. Nat. Nanotechnol. 2007, 2, 751−760. (2) Petros, R. A.; Desimone, J. M. Strategies in the Design of Nanoparticles for Therapeutic Applications. Nat. Rev. Drug Discovery 2010, 9, 615−627. (3) Torchilin, V. P. Recent Advances with Liposomes as Pharmaceutical Carriers. Nat. Rev. Drug Discovery 2005, 4, 145−160. (4) Wilhelm, S.; Tavares, A. J.; Dai, Q.; Ohta, S.; Chan, W. C. W.; Audet, J.; Dvorak, H. F. Analysis of Nanoparticle Delivery to Tumours. Nat. Rev. Mater. 2016, 1, 16014. (5) Hu, C. M. J.; Fang, R. H.; Copp, J.; Luk, B. T.; Zhang, L. A Biomimetic Nanosponge that Absorbs Pore-Forming Toxins. Nat. Nanotechnol. 2013, 8, 336−340. (6) Wang, F.; Gao, W.; Thamphiwatana, S.; Luk, B. T.; Angsantikul, P.; Zhang, Q.; Hu, C. M. J.; Fang, R. H.; Copp, J. A.; Pornpattananangkul, D.; Lu, W.; Zhang, L. Hydrogel Retaining Toxin-Absorbing Nanosponges for Local Treatment of MethicillinResistant Staphylococcus aureus Infection. Adv. Mater. 2015, 27, 3437− 3443. 5599

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601

Article

ACS Nano (7) Fang, R. H.; Hu, C. M. J.; Chen, K. N.; Luk, B. T.; Carpenter, C. W.; Gao, W.; Li, S.; Zhang, D. E.; Lu, W.; Zhang, L. Lipid-Insertion Enables Targeting Functionalization of Erythrocyte MembraneCloaked Nanoparticles. Nanoscale 2013, 5, 8884−8888. (8) Fang, R. H.; Hu, C. M. J.; Luk, B. T.; Gao, W.; Copp, J. A.; Tai, Y.; O’Connor, D. E.; Zhang, L. Cancer Cell Membrane-Coated Nanoparticles for Anticancer Vaccination and Drug Delivery. Nano Lett. 2014, 14, 2181−2188. (9) Kroll, A. V.; Fang, R. H.; Jiang, Y.; Zhou, J.; Wei, X.; Yu, C. L.; Gao, J.; Luk, B. T.; Dehaini, D.; Gao, W.; Zhang, L. Nanoparticulate Delivery of Cancer Cell Membrane Elicits Multiantigenic Antitumor Immunity. Adv. Mater. 2017, 29, 1703969. (10) Parodi, A.; Quattrocchi, N.; van de Ven, A. L.; Chiappini, C.; Evangelopoulos, M.; Martinez, J. O.; Brown, B. S.; Khaled, S. Z.; Yazdi, I. K.; Enzo, M. V.; Isenhart, L.; Ferrari, M.; Tasciotti, E. Synthetic Nanoparticles Functionalized with Biomimetic Leukocyte Membranes Possess Cell-Like Functions. Nat. Nanotechnol. 2013, 8, 61−68. (11) Hu, Q.; Sun, W.; Qian, C.; Wang, C.; Bomba, H. N.; Gu, Z. Anticancer Platelet-Mimicking Nanovehicles. Adv. Mater. 2015, 27, 7043−7050. (12) Pang, Z.; Hu, C. M. J.; Fang, R. H.; Luk, B. T.; Gao, W.; Wang, F.; Chuluun, E.; Angsantikul, P.; Thamphiwatana, S.; Lu, W.; Jiang, X.; Zhang, L. Detoxification of Organophosphate Poisoning Using Nanoparticle Bioscavengers. ACS Nano 2015, 9, 6450−6458. (13) Hu, C. M. J.; Fang, R. H.; Wang, K. C.; Luk, B. T.; Thamphiwatana, S.; Dehaini, D.; Nguyen, P.; Angsantikul, P.; Wen, C. H.; Kroll, A. V.; Carpenter, C.; Ramesh, M.; Qu, V.; Patel, S. H.; Zhu, J.; Shi, W.; Hofman, F. M.; Chen, T. C.; Gao, W.; Zhang, K.; et al. Nanoparticle Biointerfacing by Platelet Membrane Cloaking. Nature 2015, 526, 118−121. (14) Copp, J. A.; Fang, R. H.; Luk, B. T.; Hu, C. M. J.; Gao, W.; Zhang, K.; Zhang, L. Clearance of Pathological Antibodies Using Biomimetic Nanoparticles. Proc. Natl. Acad. Sci. U. S. A. 2014, 111, 13481−13486. (15) Zhang, Q.; Dehaini, D.; Zhang, Y.; Zhou, J.; Chen, X.; Zhang, L.; Fang, R. H.; Gao, W.; Zhang, L. Neutrophil Membrane-Coated Nanoparticles Inhibit Synovial Inflammation and Alleviate Joint Damage in Inflammatory Arthritis. Nat. Nanotechnol. 2018, 13, 1182− 1190. (16) Wang, C.; Sun, X.; Cheng, L.; Yin, S.; Yang, G.; Li, Y.; Liu, Z. Multifunctional Theranostic Red Blood Cells for Magnetic-FieldEnhanced In Vivo Combination Therapy of Cancer. Adv. Mater. 2014, 26, 4794−4802. (17) Luk, B. T.; Fang, R. H.; Hu, C. M. J.; Copp, J. A.; Thamphiwatana, S.; Dehaini, D.; Gao, W.; Zhang, K.; Li, S.; Zhang, L. Safe and Immunocompatible Nanocarriers Cloaked in RBC Membranes for Drug Delivery to Treat Solid Tumors. Theranostics 2016, 6, 1004−1011. (18) Xie, W.; Deng, W. W.; Zan, M.; Rao, L.; Yu, G. T.; Zhu, D. M.; Wu, W. T.; Chen, B.; Ji, L. W.; Chen, L.; Liu, K.; Guo, S. S.; Huang, H. M.; Zhang, W. F.; Zhao, X.; Yuan, Y.; Dong, W.; Sun, Z. J.; Liu, W. Cancer Cell Membrane Camouflaged Nanoparticles to Realize Starvation Therapy Together with Checkpoint Blockades for Enhancing Cancer Therapy. ACS Nano 2019, 13, 2849−2857. (19) Rao, L.; Xu, J. H.; Cai, B.; Liu, H.; Li, M.; Jia, Y.; Xiao, L.; Guo, S. S.; Liu, W.; Zhao, X. Z. Synthetic Nanoparticles Camouflaged with Biomimetic Erythrocyte Membranes for Reduced Reticuloendothelial System Uptake. Nanotechnology 2016, 27, 085106. (20) Aryal, S.; Hu, C. M. J.; Fang, R. H.; Dehaini, D.; Carpenter, C.; Zhang, D. E.; Zhang, L. Erythrocyte Membrane-Cloaked Polymeric Nanoparticles for Controlled Drug Loading and Release. Nanomedicine 2013, 8, 1271−1280. (21) Hu, C. M. J.; Fang, R. H.; Luk, B. T.; Chen, K. N.; Carpenter, C.; Gao, W.; Zhang, K.; Zhang, L. ’Marker-of-Self’ Functionalization of Nanoscale Particles Through a Top-Down Cellular Membrane Coating Approach. Nanoscale 2013, 5, 2664−2668. (22) Su, J.; Sun, H.; Meng, Q.; Yin, Q.; Tang, S.; Zhang, P.; Chen, Y.; Zhang, Z.; Yu, H.; Li, Y. Long Circulation Red-Blood-Cell-

Mimetic Nanoparticles with Peptide-Enhanced Tumor Penetration for Simultaneously Inhibiting Growth and Lung Metastasis of Breast Cancer. Adv. Funct. Mater. 2016, 26, 1243−1252. (23) Hu, C. M. J.; Zhang, L.; Aryal, S.; Cheung, C.; Fang, R. H.; Zhang, L. Erythrocyte Membrane-Camouflaged Polymeric Nanoparticles as a Biomimetic Delivery Platform. Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 10980−10985. (24) Gao, W.; Fang, R. H.; Thamphiwatana, S.; Luk, B. T.; Li, J.; Angsantikul, P.; Zhang, Q.; Hu, C. M. J.; Zhang, L. Modulating Antibacterial Immunity via Bacterial Membrane-Coated Nanoparticles. Nano Lett. 2015, 15, 1403−1409. (25) Zhang, J.; Gao, W.; Fang, R. H.; Dong, A.; Zhang, L. Synthesis of Nanogels via Cell Membrane-Templated Polymerization. Small 2015, 11, 4309−4313. (26) Xuan, M.; Shao, J.; Dai, L.; Li, J.; He, Q. Macrophage Cell Membrane Camouflaged Au Nanoshells for In Vivo Prolonged Circulation Life and Enhanced Cancer Photothermal Therapy. ACS Appl. Mater. Interfaces 2016, 8, 9610−9618. (27) Zhu, D. M.; Xie, W.; Xiao, Y. S.; Suo, M.; Zan, M. H.; Liao, Q. Q.; Hu, X. J.; Chen, L. B.; Chen, B.; Wu, W. T.; Ji, L. W.; Huang, H. M.; Guo, S. S.; Zhao, X. Z.; Liu, Q. Y.; Liu, W. Erythrocyte Membrane-Coated Gold Nanocages for Targeted Cancer Photothermal and Chemical Therapy. Nanotechnology 2018, 29, 084002. (28) Rabinow, B. E. Nanosuspensions in Drug Delivery. Nat. Rev. Drug Discovery 2004, 3, 785−796. (29) Maudens, P.; Seemayer, C.; Pfefferlé, F.; Jordan, O.; Allémann, E. Nanocrystals of a Potent p38 MAPK Inhibitor Embedded in Microparticles: Therapeutic Effects in Inflammatory and Mechanistic Murine Models of Osteoarthritis. J. Controlled Release 2018, 276, 102−112. (30) Mitragotri, S.; Burke, P. A.; Langer, R. Overcoming the Challenges in Administering Biopharmaceuticals: Formulation and Delivery Strategies. Nat. Rev. Drug Discovery 2014, 13, 655−672. (31) Fossella, F.; Pereira, J. R.; von Pawel, J.; Pluzanska, A.; Gorbounova, V.; Kaukel, E.; Mattson, K. V.; Ramlau, R.; Szczesna, A.; Fidias, P.; Millward, M.; Belani, C. P. Randomized, Multinational, Phase III Study of Docetaxel Plus Platinum Combinations Versus Vinorelbine Plus Cisplatin for Advanced Non−Small-Cell Lung Cancer: The TAX 326 Study Group. J. Clin. Oncol. 2003, 21, 3016− 3024. (32) Chan, S. Docetaxel vs Doxorubicin in Metastatic Breast Cancer Resistant to Alkylating Chemotherapy. Oncology 1997, 11, 19−24. (33) Baker, J.; Ajani, J.; Scotté, F.; Winther, D.; Martin, M.; Aapro, M. S.; von Minckwitz, G. Docetaxel-Related Side Effects and Their Management. Eur. J. Oncol. Nurs. 2009, 13, 49−59. (34) Popielarski, S. R.; Pun, S. H.; Davis, M. E. A NanoparticleBased Model Delivery System to Guide the Rational Design of Gene Delivery to the Liver: Synthesis and Characterization. Bioconjugate Chem. 2005, 16, 1063−1070. (35) Yu, Y. P.; Wang, Q.; Liu, Y. C.; Xie, Y. Molecular Basis for the Targeted Binding of RGD-Containing Peptide to Integrin Alpha v Beta 3. Biomaterials 2014, 35, 1667−1675. (36) Gao, L.; Liu, G.; Ma, J.; Wang, X.; Zhou, L.; Li, X. Drug Nanocrystals: In Vivo Performances. J. Controlled Release 2012, 160, 418−430. (37) Ito, Y.; Nakamura, S.; Sugimoto, N.; Shigemori, T.; Kato, Y.; Ohno, M.; Sakuma, S.; Ito, K.; Kumon, H.; Hirose, H.; Okamoto, H.; Nogawa, M.; Iwasaki, M.; Kihara, S.; Fujio, K.; Matsumoto, T.; Higashi, N.; Hashimoto, K.; Sawaguchi, A.; Harimoto, K.-i.; Nakagawa, M.; Yamamoto, T.; Handa, M.; Watanabe, N.; Nishi, E.; Arai, F.; Nishimura, S.; Eto, K. Turbulence Activates Platelet Biogenesis to Enable Clinical Scale Ex Vivo Production. Cell 2018, 174, 503−504. (38) 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. J. Controlled Release 2014, 174, 161− 170. 5600

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601

Article

ACS Nano (39) Lu, Y.; Wang, Z. H.; Li, T.; McNally, H.; Park, K.; Sturek, M. Development and Evaluation of Transferrin-Stabilized Paclitaxel Nanocrystal Formulation. J. Controlled Release 2014, 176, 76−85. (40) Wei, X.; Zhan, C.; Shen, Q.; Fu, W.; Xie, C.; Gao, J.; Peng, C.; Zheng, P.; Lu, W. A D-Peptide Ligand of Nicotine Acetylcholine Receptors for Brain-Targeted Drug Delivery. Angew. Chem., Int. Ed. 2015, 54, 3023−3027. (41) Dodge, J. T.; Mitchell, C.; Hanahan, D. J. The Preparation and Chemical Characteristics of Hemoglobin-Free Ghosts of Human Erythrocytes. Arch. Biochem. Biophys. 1963, 100, 119−130. (42) Wei, X.; Gao, J.; Zhan, C.; Xie, C.; Chai, Z.; Ran, D.; Ying, M.; Zheng, P.; Lu, W. Liposome-Based Glioma Targeted Drug Delivery Enabled by Stable Peptide Ligands. J. Controlled Release 2015, 218, 13−21. (43) Khodarev, N. N.; Yu, J.; Labay, E.; Darga, T.; Brown, C. K.; Mauceri, H. J.; Yassari, R.; Gupta, N.; Weichselbaum, R. R. TumourEndothelium Interactions in Co-Culture: Coordinated Changes of Gene Expression Profiles and Phenotypic Properties of Endothelial Cells. J. Cell Sci. 2003, 116, 1013−1022. (44) Ruan, H.; Chai, Z.; Shen, Q.; Chen, X.; Su, B.; Xie, C.; Zhan, C.; Yao, S.; Wang, H.; Zhang, M.; Ying, M.; Lu, W. A Novel Peptide Ligand RAP12 of LRP1 for Glioma Targeted Drug Delivery. J. Controlled Release 2018, 279, 306−315. (45) Zhan, C.; Li, B.; Hu, L.; Wei, X.; Feng, L.; Fu, W.; Lu, W. Micelle-Based Brain-Targeted Drug Delivery Enabled by a Nicotine Acetylcholine Receptor Ligand. Angew. Chem., Int. Ed. 2011, 50, 5482−5485. (46) Scott, A. W.; Tyler, B. M.; Masi, B. C.; Upadhyay, U. M.; Patta, Y. R.; Grossman, R.; Basaldella, L.; Langer, R. S.; Brem, H.; Cima, M. J. Intracranial Microcapsule Drug Delivery Device for the Treatment of an Experimental Gliosarcoma Model. Biomaterials 2011, 32, 2532− 2539. (47) Qu, Y.; Zhang, L.; Rong, Z.; He, T.; Zhang, S. Number of Glioma Polyploid Giant Cancer Cells (PGCCs) Associated with Vasculogenic Mimicry Formation and Tumor Grade in Human Glioma. J. Exp. Clin. Cancer Res. 2013, 32, 75.

5601

DOI: 10.1021/acsnano.9b00661 ACS Nano 2019, 13, 5591−5601