Enhanced Antitumor Activity of EGFP-EGF1-Conjugated Nanoparticles

Feb 18, 2016 - Tumor stromal cells have been increasingly recognized to interact with tumor parenchyma cells and promote tumor growth. Therefore, we ...
1 downloads 0 Views 2MB Size
Subscriber access provided by The University of British Columbia Library

Article

Enhanced anti-tumor activity of EGFP-EGF1conjugated nanoparticles by a multi-targeting strategy Bo Zhang, Ting Jiang, Li Ling, Zhonglian Cao, Jingjing Zhao, Yanyan Tuo, Xiaojian She, Shun Shen, Xinguo Jiang, Yu Hu, and Zhiqing Pang ACS Appl. Mater. Interfaces, Just Accepted Manuscript • DOI: 10.1021/acsami.6b00036 • Publication Date (Web): 18 Feb 2016 Downloaded from http://pubs.acs.org on February 25, 2016

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

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

Page 1 of 35

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

ACS Applied Materials & Interfaces

Enhanced

Anti-Tumor

Activity

of

EGFP-EGF1-Conjugated

Nanoparticles

by

a

Multi-Targeting Strategy Bo Zhang a,1, Ting Jiang a,1, Li Lingb, Zhonglian Caoc, Jingjing Zhaob, Yanyan Tuob, Xiaojian Sheb, Shun Shenb, Xinguo Jiangb, Yu Hua*, Zhiqing Pangb* a

Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of

Science & Technology, Wuhan, Hubei, 430022, China; b

School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of

Education, 826 Zhangheng Road, Shanghai, 201203, China; c

Instrumental Analysis Center of School of Pharmacy, Fudan University, 826 Zhangheng

Road, Shanghai, 201203, China 1: equal contribution to the work * Corresponding author: Yu Hu, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China. Tel.: +86-27-85726335; fax: +86-27-85776343. E-mail address: [email protected] (Y. Hu). Zhiqing Pang, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, China Tel.: +86-21-51980069; fax: +86-21-51980069. E-mail address: [email protected]

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

Abstract Tumor stromal cells have been increasingly recognized to interact with tumor parenchyma cells and promote tumor growth. Therefore, we speculated that therapeutics delivery to both parenchyma cells and stromal cells simultaneously might treat tumor more effectively. Tissue factor (TF) was shown to extensively locate in tumor and abundantly site in both tumor parenchyma cells and stromal cells including neo-vascular cells, tumor-associated fibroblasts and tumor-associated macrophages, indicating it might function as a favorable target for drug delivery to multiple cell types simultaneously. EGFP-EGF1 is a fusion protein derived from factor VII, the natural ligand of TF. It retains the specific TF binding capability but does not cause coagulation. In the present study, nanoparticle modified with EGFP-EGF1 (ENP) was constructed as a multi-targeting drug delivery system. The protein binding experiment showed EGFP-EGF1 could bind well to A549 tumor cells and other stromal cells including neo-vascular cells, tumor-associated fibroblasts, and tumor-associated macrophages. Compared with unmodified nanoparticles (NP), ENP uptake by A549 cells and those stromal cells was significantly enhanced but inhibited by excessive free EGFP-EGF1. In addition, ENP induced more A549 tumor cell apoptosis than Taxol and NP when paclitaxel (PTX) was loaded. In vivo, ENP accumulated more specially in TF-overexpressed A549 tumors by in vivo imaging, mainly regions unoccupied by factor VII and targeted tumor parenchyma cells as well as different types of stromal cells by immunofluorescence staining. Treatment with PTX-loaded ENP (ENP-PTX) significantly reduced the A549 tumor growth in nude mice while NP-PTX- and Taxol-treated mice had lower response to the therapy. Furthermore, H&E and TUNEL staining revealed that ENP-PTX induced more severe tumor necrosis and more extensive cell apoptosis. Altogether, the present study

ACS Paragon Plus Environment

Page 2 of 35

Page 3 of 35

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

ACS Applied Materials & Interfaces

demonstrated that ENP could target multiple key cell types in tumors through TF, which could be utilized to improve the therapeutic effect of anticancer drugs. Keywords:EGFP-EGF1,TF,tumor,multi-targeting,nanoparticle Introduction Effective tumor targeted drug delivery is of vital importance for the treatment of malignant tumors as conventional chemotherapeutics lack selectivity and are always associated with severe adverse effects.1, 2 The key issue in tumor targeting therapy is to understand the proliferative foundation of tumor and utilize it as the therapeutic target to destroy tumors.3 For a long time, it has been believed that tumor growth mainly depends on tumor parenchyma cells and neo-vascular cells, and accordingly for tumor treatment, most researches are focused on targeting either of these two types of cells or both of them simultaneously via the enhanced permeability and retention (EPR) effect as well as active targeting strategy.2, 4 However, the benefits of these strategies are only modest and clinical success is limited mainly due to the heterogeneous expression of targeted receptors. Therefore, targetable receptors more widely and homogeneously distributed in tumors may enable improved drug efficacy,5, 6 which is still in continued and urgent need. It is now increasingly recognized that tumor stroma contains distinct cell types including tumor-associated fibroblasts, tumor-associated macrophages and neo-vascular cells which interact with each other, exert variable roles in enabling tumor progression and function as the “soil” of tumor growth.3, 7, 8 Furthermore, the tumor parenchymal cell-centric paradigm has been gradually shifted to a stroma-centric paradigm9 and recently some approaches targeting to distinct types of tumor stromal cells have also been designed. For example, fibroblast activation protein (FAP)-responsive pro-drug or docetaxel conjugate nanoparticle has been established to selectively

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

kill tumor-associated fibroblasts to produce a therapeutic response.10 Moreover, survival benefits are also obtained by altering macrophages11 or delivering proapoptotic peptides selectively to tumor-associated macrophages.7 However, these approaches targeting therapeutics only to a limited number of cell types in a tumor, which could not eradicate the tumor “soil” totally and there is still large room for improvement. Therefore, we propose that the multi-therapeutic strategy targeting both tumor parenchyma cell and different types of tumor stromal cells and destroying tumor “seeds” and “soil” simultaneously might be a more promising approach for tumor treatment.1 To the best of our knowledge, few studies in this filed has been reported, partly because both targeting moieties capable of targeting multiple cell types in tumors simultaneously and the targetable receptors widely and homogeneously distributed in tumors are scarce. Tissue factor (TF) is a 47 KD trans-membrane protein widely presented in normal tissues12 and its expression gets dramatically up-regulated in pathological conditions, such as thrombosis,13 atherosclerosis,14 and tumor.15 Cancer cell-expressed TF contributes to cancer progression, facilitation of metastasis and induction of neo-vascular formation in a wide spectrum of tumors,16 including lung cancer,17 glioma,18 breast cancer19 and so on. TF not only locates on tumor parenchyma cells but also sites on tumor stromal cells such as tumor-associated fibroblasts, tumor-associated macrophages and neo-vascular cells to varying degrees.20-22 The wide distribution of TF in tumor would enable it function as a favorable target for drug delivery to multiple cell types in tumor simultaneously. EGFP-EGF1 is a fusion protein derived from rat factor VII, the natural ligand of TF. EGFP-EGF1 retains the specific affinity for TF without induction of coagulation.23, 24 It is well established in our previous studies that EFGP-EGF1 could mediate drug delivery system specially to TF highly

ACS Paragon Plus Environment

Page 4 of 35

Page 5 of 35

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

ACS Applied Materials & Interfaces

expressed sites, such as rat brain thrombosis25,

26

or C6 glioma,2 showing potential clinical

application in targeted drug delivery. As TF is regarded as an evolutionarily conserved protein,27 EGFP-EGF1 might also efficiently mediate drug delivery system to human tumor where human TF is highly expressed by the multi-targeting strategy as mentioned above, which will open a new avenue for tumor targeting strategy design and according widely extend the disease coverage of EGFP-EGF1. In the present study, EGFP-EGF1-conjugated nanoparticles (ENP) were constructed as the multi-targeting drug delivery system. The multi-targeting ability of ENP in vitro was investigated by protein binding experiment and nanoparticles uptake experiment. In addition, the related endocytosis mechanisms of ENP were investigated in vitro by cellular uptake inhibition and intracellular trafficking experiments. Furthermore, the targeting ability and related mechanisms in vivo were investigated by in vivo imaging and immuno-fluorescence staining experiments. Finally, the pharmacodynamics of paclitaxel(PTX)-loaded ENP (ENP-PTX) was also assessed in vitro and in vivo, compared with that of Taxol and conventional NP-PTX. Methods and materials The EGFP-EGF1 fusion protein was expressed from E. coli BL21 cells as previously reported.2 D, L-lactide (purity: 99.5%) was obtained from PURAC (Arkelsedijk, Holland). Methoxy-poly (ethylene glycol) (MPEG, MW 3000 Da) was from NOF (Tokyo, Japan) and Maleimide-poly (ethylene glycol) (Mal-PEG, MW 3400 Da) was custom-synthesized by Nektar (Huntsville, AL, USA). Methoxy-poly (ethylene glycol)-poly (lactic acid) (MPEG–PLA, Mw 33000 Da) and Maleimide-poly (ethylene glycol)-poly (lactic acid) (Mal–PEG–PLA, Mw 35000 Da) block copolymers were both synthesized by ring-opening polymerization of lactide with MPEG and

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

Mal-PEG as the initiator as described elsewhere.28

Page 6 of 35

Coumarin-6, lipopolysaccharide (LPS),

transforming growth factor-β (TGF-β) and interleukin-4 (IL-4) were from sigma (Saint Louis, MO, USA). PTX was purchased from Xi’ an San jiang Bio-Engineering Co. Ltd. (Xi’an, China). Sodium cholate was purchased from Shanghai Chemical Reagent Company (Shanghai, China). 1,1'-Dioctadecyl-3,3,3',3'-tetramethylindo-tricarbocyanineiodide (DiR), a near-infrared dye, was obtained from Biotium (Invitrogen, CA, USA). The AnnexinV-FITC apoptosis detection kit and Hoechst 33342 were purchased from Beyotime® Biotechnology Co., Ltd. (Nantong, China). TF goat polyclonal primary antibody and F4/80 rabbit polyclonal primary antibodywere from Santa Cruz biotechnology (Santa Cruz, CA, USA), factor VII and fibroblasts activation protein (FAP) rabbit polyclonal primary antibody was bought from BIOSS Biotechnology Co., Ltd. (Beijing, China), CD31 rabbit polyclonal primary antibody was obtained from Abcam (Hong Kong) Ltd. (Hong Kong, China). Cy™ 3-conjugated affinipure donkey anti-goat secondary antibody was from

Jackson

ImmunoResearch

Laboratories,

Inc

(West

Grove,

PA,

USA).

Alexa

fluor®647-conjugated affinipure donkey anti-rabbit was from Invitrogen (Carlsbad, USA). Plastic cell culture dishes were obtained from Corning Incorporation (Corning, NY, USA). Foetal bovine serum (FBS), F-12K cell culture medium, Dulbecco’s Modified Eagle’s Medium (high glucose) (DMEM), RPMI Medium 1640, trypsin-EDTA (0.25%) and penicillin-streptomycin were obtained from Gibco (CA, USA). Puromycin was from Aladdin ® (Shanghai, China). Deionised water from the Millipore Simplicity System (Millipore, Bedford, MA) was used throughout the entire study. All other reagents and chemicals were analytical reagent grade and were obtained from Sinopharm Chemical Reagent (Shanghai, China). Human lung cancer cells A549, primary human umbilical vein endothelial cells (HUVEC), mouse NIH3T3 fibroblasts and THP1 were ordered from the

ACS Paragon Plus Environment

Page 7 of 35

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

ACS Applied Materials & Interfaces

Chinese Academy of Sciences Cell Bank (Shanghai, China). A549-mCherry-puro cell lines were bought from the Shanghai SBO Medical biotechnology (Shanghai, China). Cells were cultured at 37℃, 5% CO2 with A549 cultured in F-12K, HUVEC, and NIH3T3 cultured in DMEM, THP1 cultured in RPMI Medium 1640 and A549-mCherry-puro cell lines in F-12K supplemented with 2µg/ml puromycin. Besides, all the cell culture mediums were supplemented with 10% FBS, 100 IU/ml penicillin and 100 µg/ml streptomycin. Male Balb/c nude mice (20 ± 2 g) were from the Shanghai Slac Lab Animal Ltd. (Shanghai, China) and housed under specific-pathogen-free (SPF) conditions. All procedures were performed in accordance with guidelines evaluated and approved by the ethics committee of Fudan University. In vitro binding of EGFP-EGF1 A549 cells were seeded at a density of 2×105 cells/well in 12-well plates, 24 h later A549 were incubated in F-12K with 3, 6 and 9 µM of EGFP-EGF1 protein for 6 h. For qualitative analysis, cells were rinsed with PBS (0.01 M, pH=7.4) for three times, fixed with 4% paraformaldehyde and then analyzed using fluorescence microscope (Leica, DMI4000B, Germany). For quantitative analysis, cells were collected by trypsinization and analyzed by flow cytometry (BD, USA). In addition, HUVEC, NIH3T3 and THP1were activated with LPS (1µg/ml for 4 h),23 TGF-β (100 ng/ml for 24 h)29 and IL-4 (20 ng/ml for 24 h),30 respectively for TF induction, and these types without activation served as control to incubate with 9 µM of EGFP-EGF1 protein for 6 h before collected by trypsinization and analyzed by flow cytometry (BD, USA). The characterization of nanoparticles Blank NP and ENP as well as coumarin-6-, DiR- or PTX-loaded NP and ENP were developed as previously described.2 Particle size and zeta potential of both NP and ENP were measured using a

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

Malvern Nano ZS (Malvern Instruments, UK). After negative stained with 2% phosphatotungstic acid, the morphology of both nanoparticles was observed under a transmission electron microscope (TEM) (H-600, Hitachi, Japan). The amount of coumarin-6 and PTX in NP or ENP was measured by the high performance liquid chromatography (HPLC) (Agilent, 1200, USA).41 The mobile phase was a mixture of methyl alcohol and water (CH3OH:H2O=96:4) for coumarin-6 and a mixture of acetonitrile and water (CH3CN:H2O =55:45, v/v) for PTX, respectively. The flow rate was 1.0 ml/min and the sample injection volume was 20 µl. The detection wavelength was 465 nm and 227 nm for coumarin-6 and PTX, respectively. The amount of DiR in NP or ENP was determined by a universal fluorescence spectrophotometer (Excitation: 748 nm, Emission: 780 nm) using a Tecan Infinite M200 Pro Multiplate Reader (TECAN Safire2, Switzerland). Cellular uptake experiment A549 cells, HUVEC, NIH3T3 and THP1were seeded into 24-well plates at a density of 1×105 cells /well, 24 h later, coumarin-6-labeled NP and ENP were added into the well and incubated for 1 h. For EGFP-EGF1 inhibition experiment, excessive free EGFP-EGF1 (10 µg/ml) was added 2 h before ENP incubation. For qualitative analysis, cells were rinsed with PBS (0.01 M, pH=7.4) for three times, fixed with 4%paraformaldehyde and then analyzed under fluorescence microscopy (Leica, Germany). For quantitative analysis, cells were harvested by trypsinization and then subject to flow cytometry (BD, USA). Cellular uptake mechanisms A549 cells were seeded into 12-well plates at a density of 2×105 cells/well and incubated for 24 h. After a 30 min pre-incubation in F-12K, the cells were treated with coumarin-6-labeled NP or ENP (coumarin-6 concentration adjusted to 20 ng/ml) and various endocytic inhibitors including:

ACS Paragon Plus Environment

Page 8 of 35

Page 9 of 35

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

ACS Applied Materials & Interfaces

F-12K (control), 0.1% w/v sodium azide, 2 µmol/L phenylarsine oxide, 20 µg/ml chlorpromazine, 450 mmol/L sucrose, 10 µg/ml filipin, 50 µmol/L nacodazole, 40 µmol/L cytochalasin B, 20 µg/ml Brefeldin A, 100 nmol/L monensin. After incubation for 1 h, cells were washed with ice-cold PBS (0.01 M, pH=7.4) and subsequently with acid buffer (consisting of 120 mmol/L NaCl, 20 mmol/L sodium barbital, and 20 mmol/L sodium acetate, pH=3) at 4 ℃ for 5 min. Then the cells were collected by trypsinization, suspended in 0.5 ml PBS (0.01 M, pH=7.4) and the mean fluorescence intensity was analyzed by flow cytometry (BD, USA). Subcellular localization of the nanoparticles In order to determine which organelles were involved in the intracellular distribution of the nanoparticles, lysosome and mitochondria staining was performed on A549 cells. A549 cells were seeded at a density of 2×104 cells per dish into a 35-mm glass-bottom culture dish (NEST, China), and cultured at 37 ℃ in the presence of 5% CO2 for 24 h. After incubation with coumarin-6-labeled NP or ENP with coumarin-6 concentration adjusted to 20 ng/ml, cells were further stained with Lyso-tracker Red (50 nmol/L, 30 min) or Mito-tracker Red (200 nmol/L, 30 min) and Hoechst 33342 (1 µg/ml, 10 min) to visualize endosome/lysosome (endolysosomes) or mitochondria and cell nucleus, respectively. After being fixed with 4% paraformaldehyde at room temperature for 10 min, the cells were rinsed with PBS (0.01 M, pH=7.4) and observed under confocal microscopy (ZEISS, 710, LSM, Germany). Cytotoxicity assay A549 cells were seeded into 12-well plates at a density of 1 × 105 cells /well. After 24 h, Taxol, NP-PTX and ENP-PTX were applied with the final PTX concentration in each well 100 ng/ml. Cells without any drug treatment served as control. After incubation for 24 h, cells from each

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

Page 10 of 35

group were collected by trypsinization and stained according to procedures of AnnexinV-FITC apoptosis detection kit for quantitative analysis using flow cytometry. (BD, USA). For qualitative analysis, cells were staining with Hoechst 33342 (1 µg/ml) for 10 min at room temperature and the morphology of cell nuclei were observed under fluorescence microscopy (Leica, Germany). In vivo imaging The A549 xenograft-bearing nude mice models were established by subcutaneous injection of 5×106 A549 cells in 100 µl of PBS (0.01 M, pH=7.4). After administration with DiR-labeled NP or ENP with the dose of DiR 0.5 mg/Kg, the mice models were imaged in vivo at various time points (1 h, 2 h, 4 h, 6 h, 8 h, 12 h and 24 h) by the In Vivo IVIS spectrum imaging system (PerkinElmer, USA). 24 h post administration, the mice were sacrificed and subjected to perfusion with 4% paraformaldehyde. Then, tumor tissues and other major organs including liver, spleen, kidney, heart, lung and brain were collected and the corresponding fluorescence signals ex vivo were also analyzed by the in vivo IVIS spectrum imaging system. In vivo distribution The A549 xenograft-bearing nude mice models were injected with coumarin-6-labeled NP and ENP at the dose of coumarin-6 0.05 mg/Kg. 12 h later, the mice were sacrificed, perfused with 4% paraformaldehyde, and prepared for frozen slices

with the

thickness of 20 µm.

Immunofluorescence staining was performed as elsewhere described.31, 32 TF expression in the tumor slices was stained with TF goat polyclone antibody (1:100) at 4 ℃ overnight and then exposed to Cy™3-conjugated donkey anti-goat secondary antibody (1:100) for 1 h at room temperature. A549 tumor parenchymal cells were labeled with red fluorescence protein (RFP) expressed by the A549-mCherry-puro cell lines. Neo-vascular cells, tumor-associated

ACS Paragon Plus Environment

Page 11 of 35

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

ACS Applied Materials & Interfaces

macrophages, tumor-associated fibroblasts, and factor VII were stained with primary rabbit polyclone CD 31, F4/80, FAP, or factor VII antibody (1:100) at 4 ℃ overnight, respectively, and then further stained with Alexa fluor®647-conjugated donkey anti-rabbit IgG secondary antibody (1:100) for 1 h at room temperature. Cell nuclei were counterstained by Hoechst 33342 (1 µg/ml) at room temperature for 10 min. Finally, the slices were mounted in Dako fluorescent mounting medium and observed under confocal microscopy (ZEISS, 710, LSM, Germany). In vivo anti-tumor effects 5.0 ×106 A549 cells in 100 µl of PBS (0.01 M, pH=7.4) were subcutaneously injected into nude mice to establish nude mice xenograft tumor models. When the diameters of tumors reached around 5 mm, the mice were randomly divided into four groups (n= 6) to minimize the weight and tumor size differences among the groups: control group treated with saline, Taxol group, NP-PTX group, and ENP-PTX group (PTX dose 5 mg/kg). Therapy was continued every four days through tail vein administration for four times. The tumor size and body weight were measured every other day. The tumor volumes were calculated using the formula: V=1/2×a×b2, where a and b represented the maximum and minimum diameters of tumors, respectively. After four cycles of treatments, the tumor size and body weight were recorded for another three times. When the whole experiment ended, tumors from all mice models were collected and the tumor weight was measured. The tumors were fixed with 4% paraformaldehyde, imbedded in paraffin and sectioned at 5 µm for H&E staining and terminal dUTP-mediated nick-end-labeling (TUNEL) staining as previously described.2 The tumor slices of H&E staining and TUNEL staining were observed under the fluorescence microscope (Leica, Germany) at 200× and 400× magnification, respectively, and the semi-quantitative data of cell apoptosis in vivo were obtained by Image J

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

software. Statistical Analysis All data were presented as the mean ± standard deviation. Statistical differences in tumor volumes and tumor weight were determined by one-way analysis of variance (ANOVA), followed by post hoc analysis of Bonferroni for multiple groups comparison. A value of p< 0.05 was considered as significant. Results and discussion In vitro binding of EGFP-EGF1 with A549 cells To assess multi-targeting ability of ENP in vitro, TF-overexpressed human lung cancer cells line A549 was chosen as the parenchyma cell model.33 HUVEC, NIH3T3 and THP1 activated with special stimulating factor were selected as the cell model of tumor-associated neo-vascular cells,2 tumor-associated fibroblasts1 and tumor-associated macrophage,34 respectively. The results in Fig. S 1 showed EGFP-EGF1 protein bound well to TF-overexpressed A549 cells with a concentration-depend manner, which consisted well to previous work.2 In addition, the average fluorescence intensity of activated HUVEC, NIH3T3 and THP1 was 3.34, 2.23 and 2.70 fold higher than that of unstimulated HUVEC, NIH3T3 and THP1, respectively (Fig. S 2). These results indicated that the fusion protein EGFP-EGF1 could also bind with human TF and be applied to human derived tumor cells and other stromal cells. This might be due to the evolutionary conservation of TF.27 The encouraging outcome indicated the protein EGFP-EGF1 could function well as a multi-targeting moiety in the present study. Characterization of nanoparticles As shown in Fig. 1, the diameters for both NP and ENP were around 110 nm with a narrow size

ACS Paragon Plus Environment

Page 12 of 35

Page 13 of 35

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

ACS Applied Materials & Interfaces

distribution (Fig. 1 C). Both NP and ENP in the TEM photographs were of regular size and smooth surface (Fig. 1 A & B). The size observed under TEM was slightly smaller than that measured by the DLS method. The targeting moiety EGFP-EGF1 conjugation to nanoparticles slightly increased the diameter and lowered the zeta potential of nanoparticles (Fig. 1 C) as previously described.2 Encapsulation of coumarin-6, DiR or PTX did not obviously influence the size or zeta potential of nanoparticles as previously reported.2, 35 As few changes happened to the physical and chemical parameters of NPs when coumarin-6 or DiR was loaded, furthermore, the amount of coumarin-6 or DiR released from NPs was less than 1% in 24 h,35 which confirmed that the fluorescence tracker including coumarin-6 or DiR could represent NPs. Nanoparticles uptake and related mechanism As shown in Fig. 2, EGFP-EGF1 modification significantly enhanced nanoparticles uptake by A549 cells and stromal cells including activated HUVEC, NHI3T3 and THP-1 as compared with traditional NP, which could be inhibited by free EGFP-EGF1 co-incubation. These results demonstrated the targeting moiety EGFP-EGF1 protein could facilitate nanoparticles uptake by tumor parenchyma cell and different types of stromal cells, indicating EGFP-EGF1-mediated drug delivery system might be used for the treatment of human tumors by multi-targeting mechanism. This was in good agreement with previous study in which EGF1-EGFP conjugation significantly increased the nanoparticles uptake by C6 cells and perturbed HUVEC.2 Results of related uptake mechanism was shown in Fig. 3, sodium azide, an energy depletory, decreased cellular uptake of both NP and ENP to 58.4% and 53.4% of that of control, respectively, indicating both NP and ENP uptake was energy-dependent process. Phenylarsine oxide, the endocytosis inhibitor, also significantly reduced the cellular uptake of NP and ENP to 36.1% and 26.6%, respectively,

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

indicating that endocytosis was involved in both NP and ENP uptake. Chlorpromazine and sucrose which prevented clathrin-coated pit formation and functioned as the inhibitors of clathrin-associated endocytosis pathway,36 reduced NP uptake to 64.0% and 69.6% as compared with control group, respectively. Furthermore, chlorpromazine and sucrose also decreased ENP uptake to 78.3% and 66.2%, respectively, indicating clathrin-associated endocytosis pathway was involved in both NP and ENP uptake. Filipin, a special sterol-binding agent that served as an inhibitor of caveolae-dependent endocytosis,37 significantly decreased the ENP uptake to 76.5%, while it had no effect on the NP uptake, suggesting that caveolae-dependent endocytosis was involved in the ENP uptake rather than NP uptake. Cytochalasin B and nocodazole, both as typical inhibitors of the macropinocytosis pathway,38 significantly reduced NP uptake to 89.0% and 75.1% , respectively, but almost had no effect on ENP uptake, suggesting that macropinocytosis was involved in NP uptake but not in ENP uptake. Lysosomes and Golgi apparatus had been shown to play important roles in both intracellular cargo transport and disposition.39, 40 In the present study, the lysosome inhibitor monensin considerably decreased both NP and ENP uptake to 87.0% and 88.4%, respectively. Brefeldin A, which disrupted the Golgi apparatus and intracellular trafficking, significantly decreased the uptake of NP and ENP to 83.8% and 81.2%, respectively, suggesting the intracellular transport of both NP and ENP were related to lysosome and Golgi apparatus. Altogether, the results demonstrated clathrin-and caveolae-associated endocytosis pathway participated in ENP uptake, a different manner as compared with NP uptake, in which clathrin-associated endocytosis pathway and micropinocytosis was involved. The active targeting nanoparticles were always associated with clathrin- and caveolae-mediated endocytosis, such as peptide-22-modified nanoparticles for low-density lipoprotein receptor (LDLR) targeting41

ACS Paragon Plus Environment

Page 14 of 35

Page 15 of 35

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

ACS Applied Materials & Interfaces

and angiopep-2-modified nanoparticles for low-density lipoprotein receptor-related protein (LRP) targeting.42 However, the uptake mechanism was also closely associated with the type of targeting moiety, the targeting receptor, the concentration of inhibitors, and the cell line model used.43, 44 To more precisely determine the main uptake pathway of ENP, further studies were still needed by using different concentration of inhibitors, siRNA inhibition of clathrin or caveolae,45 and TEM observation as elsewhere reported.38 Intercellular distribution of nanoparticles Confocal microscopy was utilized to analyze nanoparticles distribution in A549 cells. As shown in Fig. 4, after incubation for 1 h, both NP (B & J) and ENP (F & N) were internalized by A549 cells. Nanoparticles co-localization with endosome/lysosome (endolysosomes) (C & K) or mitochondria (G & O) indicated by yellow color (D, H, L, P) suggested that both NP and ENP uptake by the cells were delivered to lysosome and mitochondria. In addition, the fluorescence intensity of A549 cells incubated with coumarin-6-labeled ENP was stronger than that of coumarin-6-labeled NP, which was consistent with the results of cell uptake experiment analyzed by fluorescence microscopy and flow cytometry (Fig. 2 A & E). Cytotoxicity assay Results from in vitro cytotoxicity assay determined by flow cytometry using Annexin V/propidiumiodide double staining was shown in Table S 1, ENP-PTX induced significantly more cell apoptosis than any other group. The cytotoxicity of NP-PTX was comparable to that of Taxol, presumably because of the added cell-killing effect of the solvent Cremophor EL-ethanol of Taxol. 46

The qualitative analysis displayed similar outcomes (Fig. S 3). The morphology of cell nuclei in

control group was integral with homogenous fluorescence. However, the cell nuclei displayed

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

fragmentation and dense staining with varying degrees in drug-treatment groups, among which the most severe impairment was observed in ENP-PTX treatment group. These results indicated ENP-PTX presented the strongest cytotoxicity against A549 cells, which consisted well to the nanoparticles uptake experiment. In vivo imaging In vivo fluorescence imaging was utilized to investigate the targeting potential of ENP in ectopic subcutaneous A549 tumor models, based on the fluorescence signal of DiR encapsulated in nanoparticles. The in vivo three-dimensional reconstituted imaging (Fig. 5 A-C) at 24 h showed the fluorescence signal intensity in the tumor site of ENP-treated mice was much stronger than that of NP. The in vivo imaging at different time points after drug administration also showed consistent results (Fig. S 4). At 24 h, the mice models were sacrificed, tumors and major organs were harvested and the findings were validated ex vivo. In line with the results obtained by in vivo imaging, the semi-quantitative results of ex vivo tumors revealed the tumor fluorescence intensity for ENP-treated mice was 2.3 times stronger than that for NP-treated group (Fig. 5 D). Moreover, the nanoparticles distribution in normal organs of ENP was similar to that of NP (Fig. S 5). These results altogether showed that the targeting ability of EGFP-EGF1 did work in human tumor models after conjugation to nanoparticles surface, leading to more efficient retention of nanoparticles within tumors. The interaction between the targeting moiety EGFP-EGF1 and TF might be the main contributor to the targeting effect. In vivo distribution As the natural ligand for TF, factor VII could reach tumor site by the EPR effect or be produced by tumor cells themselves,19, 21 so how to explain the possible competition between EGFP-EGF1

ACS Paragon Plus Environment

Page 16 of 35

Page 17 of 35

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

ACS Applied Materials & Interfaces

protein and factor VII which have already located in tumor tissues prior to EGFP-EGF1 arriving? Immuno-fluorescence staining was conducted to preliminary analyze the issue. As shown in Fig. 6, TF was highly and extensively expressed in A549 tumor tissues (Fig. 6 C & H). As the natural ligand of TF, factor VII also located in tumor tissues in some regions (Fig. 6 D & I).21 For ENP group, nanoparticles mainly distributed in regions unoccupied by factor VII and much less in the factor VII-occupied region (Fig. 6 G). For NP group, there was no significant difference in nanoparticles distribution whether TF was occupied or not (Fig. 6 B). However, NP distribution in a local region occupied by factor VII was relative higher than other regions (Fig. 6 B & D), which might be due to the presence of tumor vascular indicated by the lumen structure nearby (Fig. 6 C). In addition, ENP distribution in the tumor slices was much more prominent than traditional NP. The outcome could be reasonably explained as follows: TF occupied by factor VII in advance would no longer be open to TF-targeting ENP and would accordingly decrease ENP accumulation in these regions. In contrast, the regions unoccupied by factor VII were still open to ENP and thus TF could bind with ENP and augment ENP accumulation in the factor VII-unoccupied regions. As for traditional NP, TF occupied by factor VII or not had no effect on the nanoparticles distribution. Factor VII would certainly exert negative influence on drug delivery system modified by EGFP-EGF1 or those similar targeting moieties including factor VII itself17, 47 or the light chain of factor VII.48 However, even though TF was occupied by factor VII, there were still some other domains of TF open for targeted therapy. To screen targeting moieties with specific affinity for these domains may open another avenue to utilize all forms of TF in tumor and thus might obtain a more satisfactory outcome. Multi-targeting mechanism of ENP in vivo

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

It was now believed that there were different types of cells in tumor tissues including tumor parenchymal cells, tumor neo-vascular cells, tumor-associated macrophages and tumor-associated fibroblasts, which were closely associated and compensated for each other to drive tumor progression.3, 6-8 TF was reported to be highly expressed in these types of cells with varying degrees.20-22 And here we tried to observe nanoparticles distribution in these cells to analyze the possible multi-targeting potential of ENP to A549 tumor xenograft in vivo. As shown in Fig. 7, ENP not only resided in neo-endothelial but also reached other types of cells in tumor microenvironment far away from tumor vessels (Fig. 7 H). In contrast, NP mainly sited near tumor vessels and few was observed to reach neo-endothelial (Fig. 7 G). To further reveal the possible multi-targeting mechanism of ENP, other tumor stromal cells were also stained by immune-fluorescence staining and tumor parenchymal cells were tracked by the fluorescence of RFP as A549-mCherry-puro cell lines stably expressed RFP. ENP distribution in these types of cells in tumor xenograft (Fig. 7 B, D & F) was much more prominent than that of NP (Fig. 7 A, C & E), indicating TF sited in all these cells could function as the targeting receptor for ENP to varying degrees. As the Figures showed, ENP distribution in different cell types varied greatly from each other and was not so homogeneously. This might be due to the varying TF levels on different cells and also the possible competition from factor VII distribution (Fig. 6). As tumor parenchymal cells proliferation and neo-vessels formation were the two basic characterizations of many tumors, strategies targeting tumor parenchymal cells35 or destroying tumor-associated vessels to block nutrition supplement for tumor therapy2, 49 have been extensively reported. In contrast, ENP in the present study could target multiple types of cells in tumor tissues. This strategy shifted from the tumor parenchymal cell-centric paradigm9 to a more global paradigm that

ACS Paragon Plus Environment

Page 18 of 35

Page 19 of 35

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

ACS Applied Materials & Interfaces

covered almost all important cell types in tumor, which established a new strategy for drug delivery system design and suggested an encouraging anti-tumor outcome when loaded with chemotherapeutics.3 In vivo anti-tumor effect Next, the antitumor effect of ENP-PTX in vivo was evaluated. The final size and weight of tumors from drug-treated mice were significantly reduced, especially for mice treated with ENP-PTX (Fig. 8 A-D), and the tumor growth curve showed that tumor growth was delayed most prominently in the group treated with ENP-PTX (Fig. 8 A). The inhibitive rate calculated based on tumor volume and weight were 60.53% and 55.60% in the ENP-PTX group, respectively, about 2.1 times higher than those in the NP-PTX group and 2.6 times higher than those in the commercial Taxol group. During the whole study, there were no significant differences in the average body weights between the four groups (Fig. 8 B), indicating no obvious side effects arising from the treatments. To more intuitively detect pharmacodynamics of different PTX formulations in tumor tissue after four cycles

of

drug

administration,

excised

tumors

were

subsequently

assessed

by

immunohistochemistry (IHC). The H&E staining and TUNEL staining of the tumor slices were displayed in Fig. 9 & S6. There were negligible necrosis and sporadic cell apoptosis in saline treated group. Tumor tissue cell necrosis including pyknosis, karyorrhexisor and karyolysis and apoptosis in the three PTX treated groups were much more obvious than control group, among which ENP-PTX group showed the most extensive necrosis and most significant cell apoptosis. The hemi-quantitative data of cell apoptosis (Fig. 9 I) consisted well to the qualitative representative image (Fig. 9 E-H). In addition, the necrosis scope consisted well with the cell apoptosis region in all groups (Fig. 9 & S6). These data altogether indicated that multi-targeting

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

parenchymal cells and stromal cells simultaneously could contribute to the significant therapeutic benefit. Conclusion The present study successfully established ENP as a multi-targeting drug delivery system to human tumor. In vitro experiments demonstrated that EGFP-EGF1 could bind well to A549 tumor parenchyma cells and other different types of stromal cells including tumor-associated neo-vascular cells, tumor-associated fibroblasts and tumor-associated macrophages and enhanced nanoparticles uptake by those types of cells. In vivo experiments displayed ENP targeted multiple types of key cells in tumor tissues, accumulated more specially in TF-expressing region unoccupied by factor VII and thus achieved the most promising therapeutic benefits. Acknowledgement The work was supported by the National Natural Science Foundation of China (81472757, 81301974, 81302043, 81302714), the State Scholarship Fund, the Doctoral fund of Ministry of Education of China (20100071120050), and “Zhuoxue” program of Fudan University. Supporting Information Available: EGFP-EGF1 protein binding experiment (Fig. S1-2), the pharmacodynamics experiment in vitro (Table S 1 and Fig. S 3), in vivo imaging of A549 tumor xenograft treated with DIR-labeled NP or ENP and ex vivo imaging of major normal organs (Fig. S 4-5). HE staining and TUNEL staining of tumor slice in different treatment group at 200× (Fig. S 6). This information is available free of charge via the Internet at http://pubs.acs.org/. Reference (1) Prakash, J.; de Jong, E.; Post, E.; Gouw, A. S.; Beljaars, L.; Poelstra, K., A Novel Approach to Deliver Anticancer Drugs to Key Cell Types in Tumors Using a PDGF Receptor-Binding Cyclic

ACS Paragon Plus Environment

Page 20 of 35

Page 21 of 35

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

ACS Applied Materials & Interfaces

Peptide Containing Carrier. J. Controlled Release 2010, 145 (2), 91-101. (2) Zhang, B.; Wang, H.; Liao, Z.; Wang, Y.; Shi, W.; Pang, Z.; Jiang, X., EGFP-EGF1-conjugated Nanoparticles for Targeting Both Neovascular and Glioma Cells in Therapy of Brain Glioma. Biomaterials 2014, 35 (13), 4133-4145. (3) Hanahan, D.; Weinberg, R. A., Hallmarks of Cancer: the Next Generation. Cell 2011, 144 (5), 646-674. (4) Gao, H.; Yang, Z.; Cao, S.; Xiong, Y.; Zhang, S.; Pang, Z.; Jiang, X., Tumor Cells and Neovasculature Dual Targeting Delivery for Glioblastoma Treatment. Biomaterials 2014, 35 (7), 2374-2382. (5) Chauhan, V. P.; Jain, R. K., Strategies for Advancing Cancer Nanomedicine. Nat. Mater. 2013, 12 (11), 958-962. (6) Luo, Y.; Zhou, H.; Krueger, J.; Kaplan, C.; Lee, S.-H.; Dolman, C.; Markowitz, D.; Wu, W.; Liu, C.; Reisfeld, R. A., Targeting Tumor-Associated Macrophages as a Novel Strategy Against Breast Cancer. J. Clin. Invest. 2006, 116 (8), 2132-2141. (7) Maryelise Cieslewicz, S. H. P., Targeted Delivery of Proapoptotic Peptides to Tumor-Associated Macrophages Improves Survival. Proc. Natl. Acad. Sci. U.S.A. 2013, 110 (40), 15919-15924. (8) Dorit Granot, Y. A., Vyacheslav Kalchenko, In Vivo Imaging of the Systemic Recruitment of Fibroblasts to the Angiogenic Rim of Ovarian Carcinoma Tumors. Cancer Res. 2007, 67, 9180-9189. . (9) Tchou, J.; Conejo-Garcia, J., Targeting the Tumor Stroma as a Novel Treatment Strategy for Breast Cancer: Shifting from the Neoplastic Cell-Centric to a Stroma-Centric Paradigm. Adv.

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

Pharmacol. 2012, 65, 45-61. (10) Brennen, W. N.; Rosen, D. M.; Wang, H.; Isaacs, J. T.; Denmeade, S. R., Targeting Carcinoma-Associated Fibroblasts within the Tumor Stroma with a Fibroblast Activation Protein-Activated Prodrug. J. Natl. Cancer Inst. 2012, 104 (17), 1320-1334. (11) Pyonteck, S. M.; Akkari, L.; Schuhmacher, A. J.; Bowman, R. L.; Sevenich, L.; Quail, D. F.; Olson, O. C.; Quick, M. L.; Huse, J. T.; Teijeiro, V., CSF-1R Inhibition Alters Macrophage Polarization and Blocks Glioma Progression. Nat. Med. 2013, 19 (10), 1264-1272. (12) Luther, T.; Flossel, C.; Mackman, N.; Bierhaus, A.; Kasper, M.; Albrecht, S.; Sage, E. H.; Iruela-Arispe, L.; Grossmann, H.; Strohlein, A.; Zhang, Y.; Nawroth, P. P.; Carmeliet, P.; Loskutoff, D. J.; Muller, M., Tissue Factor Expression During Human and Mouse Development. Am. J. Pathol. 1996, 149 (1), 101-113. (13) Mei, H.; Shi, W.; Pang, Z.; Wang, H.; Lu, W.; Jiang, X.; Deng, J.; Guo, T.; Hu, Y., EGFP -EGF1 Protein-Conjugated PEG-PLA Nanoparticles for Tissue Factor Targeted Drug Delivery. Biomaterials 2010, 31 (21), 5619-5626. (14) Kocaturk, B.; Versteeg, H. H., Tissue Factor Isoforms in Cancer and Coagulation: May the Best Isoform Win. Thromb. Res. 2012, 129 Suppl 1, S69-75. (15) Bluff, J. E.; Brown, N. J.; Reed, M. W.; Staton, C. A., Tissue Factor, Angiogenesis and Tumour Progression. Breast Cancer Res. 2008, 10 (2), 204. (16) van den Berg, Y. W.; Osanto, S.; Reitsma, P. H.; Versteeg, H. H., The Relationship Between Tissue Factor and Cancer Progression: Insights from Bench and Bedside. Blood 2011, 119 (4), 924-932. (17) Hu, Z., Effective Treatment of Human Lung Cancer by Targeting Tissue Factor with a Factor

ACS Paragon Plus Environment

Page 22 of 35

Page 23 of 35

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

ACS Applied Materials & Interfaces

VII-Targeted Photodynamic Therapy Curr. Cancer Drug Targets. 2011, 11, 1069-1081. (18) Carneiro-Lobo, T. C.; Konig, S.; Machado, D. E.; Nasciutti, L. E.; Forni, M. F.; Francischetti, I. M.; Sogayar, M. C.; Monteiro, R. Q., Ixolaris, a Tissue Factor Inhibitor, Blocks Primary Tumor Growth and Angiogenesis in a Glioblastoma Model. J. Thromb. Haemostasis 2009, 7 (11), 1855-1864. (19) Cole, M.; Bromberg, M., Tissue Factor as a Novel Target for Treatment of Breast Cancer. Oncologist 2013, 18 (1), 14-18. (20) Guan, M.; Jin, J.; Su, B.; Liu, W. W.; Lu, Y., Tissue Factor Expression and Angiogenesis in Human Glioma. Clin. Biochem. 2002, 35 (4), 321-325. (21) Liu, Y.; Jiang, P.; Capkova, K.; Xue, D.; Ye, L.; Sinha, S. C.; Mackman, N.; Janda, K. D.; Liu, C., Tissue Factor-Activated Coagulation Cascade in the Tumor Microenvironment Is Critical for Tumor Progression and an Effective Target for Therapy. Cancer Res. 2011, 71 (20), 6492-6502. (22) Shoji, M.; Hancock, W. W.; Abe, K.; Micko, C.; Casper, K. A.; Baine, R. M.; Wilcox, J. N.; Danave, I.; Dillehay, D. L.; Matthews, E., Activation of Coagulation and Angiogenesis in Cancer: Immunohistochemical Localization in Situ of Clotting Proteins and Vascular Endothelial Growth Factor in Human Cancer. Am. J. Pathol. 1998, 152 (2), 399-411. (23) Mei, H.; Hu, Y.; Wang, H.; Shi, W.; Deng, J.; Guo, T., Binding of EGF1 Domain Peptide in Coagulation Factor VII with Tissue Factor and Its Implications for the Triggering of Coagulation. J. Huazhong Univ. Sci. Technol., Med. Sci. 2010, 30 (1), 42-47. (24) Huile, G.; Shuaiqi, P.; Zhi, Y.; Shijie, C.; Chen, C.; Xinguo, J.; Shun, S.; Zhiqing, P.; Yu, H., A Cascade Targeting Strategy for Brain Neuroglial Cells Employing Nanoparticles Modified with Angiopep-2 Peptide and EGFP-EGF1 Protein. Biomaterials 2011, 32 (33), 8669-8675.

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

(25) Shi, W.; Mei, H.; Deng, J.; Chen, C.; Wang, H.; Guo, T.; Zhang, B.; Li, L.; Pang, Z.; Jiang, X.; Shen, S.; Hu, Y., A Tissue Factor Targeted Nanomedical System for Thrombi-Specific Drug Delivery. Biomaterials 2012, 33 (30), 7643-7654. (26) Shi, W.; Mei, H.; Deng, J.; Chen, C.; Wang, H.; Guo, T.; Zhang, B.; Pang, Z.; Jiang, X.; Wang, X.; Lei, H.; Hu, Y., The Delivery of Thrombi-Specific Nanoparticles Incorporating Oligonucleotides into Injured Cerebrovascular Endothelium. Biomaterials 2013, 34 (16), 4128-4136. (27) Versteeg, H. H., Tissue Factor as an Evolutionary Conserved Cytokine Receptor: Implications for Inflammation and Signal Transduction. Semin. Hematol. 2004, 41 (1 Suppl 1), 168-172. (28) Pang, Z.; Lu, W.; Gao, H.; Hu, K.; Chen, J.; Zhang, C.; Gao, X.; Jiang, X.; Zhu, C., Preparation and Brain Delivery Property of Biodegradable Polymersomes Conjugated with OX26. J. Controlled Release 2008, 128 (2), 120-127. (29) Lohr, M.; Schmidt, C.; Ringel, J.; Kluth, M.; Muller, P.; Nizze, H.; Jesnowski, R., Transforming Growth Factor-beta1 Induces Desmoplasia in an Experimental Model of Human Pancreatic Carcinoma. Cancer Res. 2001, 61 (2), 550-555. (30) Martinez, F. O.; Gordon, S.; Locati, M.; Mantovani, A., Transcriptional Profiling of the Human Monocyte-to-Macrophage Differentiation and Polarization: New Molecules and Patterns of Gene Expression. J. Immunol. 2006, 177 (10), 7303-7311. (31) Gao, H.; Yang, Z.; Zhang, S.; Cao, S.; Pang, Z.; Yang, X.; Jiang, X., Glioma-Homing Peptide with a Cell-Penetrating Effect for Targeting Delivery with Enhanced Glioma Localization, Penetration and Suppression of Glioma Growth. J. Controlled Release 2013, 172 (3), 921-928.

ACS Paragon Plus Environment

Page 24 of 35

Page 25 of 35

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

ACS Applied Materials & Interfaces

(32) Gao, H.; Yang, Z.; Zhang, S.; Pang, Z.; Liu, Q.; Jiang, X., Study and Evaluation of Mechanisms of Dual Targeting Drug Delivery System with Tumor Microenvironment Assays Compared with Normal Assays. Acta Biomater 2014, 10 (2), 858-687. (33) Ishibashi, H.; Nakagawa, K.; Onimaru, M.; Castellanous, E. J.; Kaneda, Y.; Nakashima, Y.; Shirasuna, K.; Sueishi, K., Sp1 Decoy Transfected to Carcinoma Cells Suppresses the Expression of Vascular Endothelial Growth Factor, Transforming Growth Factor-beta1, and Tissue Factor and Also Cell Growth and Invasion Activities. Cancer Res. 2000, 60 (22), 6531-6536. (34) Genin, M.; Clement, F.; Fattaccioli, A.; Raes, M.; Michiels, C., M1 and M2 Macrophages Derived from THP-1 Cells Differentially Modulate the Response of Cancer Cells to Etoposide. BMC Cancer 2015, 15, 577. (35) Gao, H.; Qian, J.; Yang, Z.; Pang, Z.; Xi, Z.; Cao, S.; Wang, Y.; Pan, S.; Zhang, S.; Wang, W.; Jiang,

X.;

Zhang,

Q.,

Whole-cell

Poly(ethyleneglycol)-Poly(epsilon-caprolactone)

SELEX

Nanoparticles

Aptamer-Functionalised for

Enhanced

Targeted

Glioblastoma Therapy. Biomaterials 2012, 33 (26), 6264-6272. (36) Pang, Z.; Gao, H.; Yu, Y.; Chen, J.; Guo, L.; Ren, J.; Wen, Z.; Su, J.; Jiang, X., Brain Delivery and Cellular Internalization Mechanisms for Transferrin Conjugated Biodegradable Polymersomes. Int. J. Pharm. 2011, 415 (1-2), 284-292. (37) Chang, J., Characterization of Endocytosis of Transferrin-Coated PLGA Nanoparticles by the Blood–Brain Barrier. Int. J. Pharm. 2009, 379, 285-292. (38) Gao, H.; Yang, Z.; Zhang, S.; Cao, S.; Shen, S.; Pang, Z.; Jiang, X., Ligand Modified Nanoparticles Increases Cell Uptake, Alters Endocytosis and Elevates Glioma Distribution and Internalization. Sci. Rep. 2013, 3, 2534.

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

Page 26 of 35

(39) Gabor, F.; Schwarzbauer, A.; Wirth, M., Lectin-Mediated Drug Delivery: Binding and Uptake of BSA-WGA Conjugates Using the Caco-2 Model. Int. J. Pharm. 2002, 237 (1-2), 227-239. (40) Jiang, S.; Rhee, S. W.; Gleeson, P. A.; Storrie, B., Capacity of the Golgi Apparatus for Cargo Transport Prior to Complete Assembly. Mol. Biol. Cell 2006, 17 (9), 4105-4117. (41) Zhang, B.; Sun, X.; Mei, H.; Hu, Y.; Pang, Z.; Jiang, X., LDLR-mediated peptide-22-conjugated nanoparticles for dual-targeting therapy of brain glioma. Biomaterials 2013, 34 (36), 9171-9182. (42) Xin, H.; Jiang, X.; Gu, J.; Sha, X.; Chen, L.; Law, K.; Chen, Y.; Wang, X.; Jiang, Y.; Fang, X., Angiopep-conjugated poly(ethylene glycol)-co-poly(epsilon-caprolactone)

nanoparticles

as

dual-targeting drug delivery system for brain glioma. Biomaterials 2011, 32 (18), 4293-4305. (43) Hu, Q.; Gu, G.; Liu, Z.; Jiang, M.; Kang, T.; Miao, D.; Tu, Y.; Pang, Z.; Song, Q.; Yao, L.; Xia, H.; Chen, H.; Jiang, X.; Gao, X.; Chen, J., F3 peptide-functionalized PEG-PLA nanoparticles co-administrated with tLyp-1 peptide for anti-glioma drug delivery. Biomaterials 2012, 34 (4), 1135-1145. (44) Gu, G.; Gao, X.; Hu, Q.; Kang, T.; Liu, Z.; Jiang, M.; Miao, D.; Song, Q.; Yao, L.; Tu, Y.; Pang, Z.; Chen, H.; Jiang, X.; Chen, J., The influence of the penetrating peptide iRGD on the effect of paclitaxel-loaded MT1-AF7p-conjugated nanoparticles on glioma cells. Biomaterials 2013, 34 (21), 5138-5148. (45) Azizi, P. M.; Zyla, R. E.; Guan, S.; Wang, C.; Liu, J.; Bolz, S. S.; Heit, B.; Klip, A.; Lee, W. L., Clathrin-dependent entry and vesicle-mediated exocytosis define insulin transcytosis across microvascular endothelial cells. Mol. Biol. Cell 2015, 26 (4), 740-750. (46) Brannon-Peppas, L.; Blanchette, J. O., Nanoparticle and Targeted Systems for Cancer

ACS Paragon Plus Environment

Page 27 of 35

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

ACS Applied Materials & Interfaces

Therapy. Adv. Drug Delivery Rev. 2004, 56 (11), 1649-1659. (47) Hu, Z.; Garen, A., Intratumoral Injection of Adenoviral Vectors Encoding Tumor-Targeted Immunoconjugates for Cancer Immunotherapy. Proc. Natl. Acad. Sci. U.S.A. 2000, 97 (16), 9221-9225. (48) Zhang, Q.; Liu, X. J.; Hu, L.; Liao, D. S.; Zheng, Y. B.; Zhen, Y. S.; Song, X., Factor VII Light Chain-Targeted Lidamycin Targets Tissue Factor-Overexpressing Tumor Cells for Cancer Therapy. Int. J. Mol. Med. 201129 (3), 409-415. (49) Bieker, R.; Kessler, T.; Schwoppe, C.; Padro, T.; Persigehl, T.; Bremer, C.; Dreischaluck, J.; Kolkmeyer,

A.;

Heindel,

W.;

Mesters,

R.

M.,

Infarction

of

Tumor

Vessels

by

NGR-Peptide-Directed Targeting of Tissue Factor: Experimental Results and First-in-Man Experience. Blood 2009, 113 (20), 5019-5027.

Fig. 1 The characterizations of NP and ENP. TEM images of NP (A) and ENP (B) negatively stained by 2% phosphotungstic acid. Particle size distribution and zeta potential of NP and ENP (C). The bar represented 200 nm.

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

Fig. 2 Cellular uptake of coumarin-6-labeled NP (the first row), ENP (the middle row) and ENP with EGFP-EGF1 protein incubation in advance (the third row) by A549 cells (A & E), activated HUVEC (B & F), activated NIH3T3 (C & G) and activated THP-1 (D & H) analyzed by fluorescence microscopy (A, B, C, D) and flow cytometry (E, F, G, H). Blue: Hoechst 33342 counterstained nucleus. Green: coumarin-6-labeled nanoparticles. The bar was 200 µm. *p< 0.01 vs ENP.

ACS Paragon Plus Environment

Page 28 of 35

Page 29 of 35

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

ACS Applied Materials & Interfaces

Fig. 3 Cellular uptake of coumarin-6-labeled NP and ENP by A549 cells in the presence of different endocytic inhibitors including NaN3 (0.1%), phenylarsine oxide (2 µmol/L), chlorpromazine (20 µg/ml), sucrose (450 mmol/L), filipin (10 µg/ml), nacodazole (50 µmol/L), cytochalasinB (40 µmol/L),brefeldin A (20 µg/ml), monensin (100 nmol/L), respectively (n = 3). The concentration of coumarin-6 were 20 ng/ml in each well. *p< 0.05 vs control.

ACS Paragon Plus Environment

ACS Applied Materials & Interfaces

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

Fig. 4 Intracellular distribution of coumarin-6-labeled NP (A, B, C, D, I, J, K, L) and ENP (E, F, G, H, M, N, O, P) in A549 cells observed by confocal microscope after incubation for 1 h. Image D was merged by image A, B and C. Image H was merged by image E, F and G. Image L was merged by image I, J and K. Image P was merged by image M, N and O. Blue: cell nuclei. Green: nanoparticles. Red: Lysotracker Red (C & K) or Mitotracker Red (G & O). Yellow: Red co-localized with green. The bar was 200 µm.

Fig. 5 In vivo three-dimensional reconstruction reconstructed imaging (with individual bars) of A549 xenograft-bearing nude mice 24 h after injection with DiR-labeled NP (A) or ENP (B).

ACS Paragon Plus Environment

Page 30 of 35

Page 31 of 35

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

ACS Applied Materials & Interfaces

Whole body fluorescence imaging (C), ex vivo fluorescence imaging of tumors at 24 h and the corresponding semi-quantitative results of tumors (D) **p