All-in-One Theranostic Nanoagent with Enhanced Reactive Oxygen

May 4, 2018 - Keywords: cancer; magnetic resonance imaging; modulating tumor microenvironment; photoenhanced Fenton or Fenton-like reaction; “all in...
0 downloads 3 Views 3MB Size
Subscriber access provided by UNIVERSITY OF THE SUNSHINE COAST

Article

All-in-One Theranostic Nanoagent with Enhanced Reactive Oxygen Species Generation and Modulating Tumor Microenvironment Ability for Effective Tumor Eradication Yang Liu, Wenyao Zhen, Longhai Jin, Songtao Zhang, Guoying Sun, Tianqi Zhang, Xia Xu, Shuyan Song, Yinghui Wang, Jianhua Liu, and Hongjie Zhang ACS Nano, Just Accepted Manuscript • DOI: 10.1021/acsnano.8b01893 • Publication Date (Web): 04 May 2018 Downloaded from http://pubs.acs.org on May 5, 2018

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

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

Page 1 of 29

ACS Nano

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

All-in-One Theranostic Nanoagent with Enhanced Reactive Oxygen

Species

Generation

and

Modulating

Tumor

Microenvironment Ability for Effective Tumor Eradication Yang Liu1,2, Wenyao Zhen1,2, Longhai Jin3, Songtao Zhang1, Guoying Sun4, Tianqi Zhang3, Xia Xu1, Shuyan Song1, Yinghui Wang1*, Jianhua Liu3*, and Hongjie Zhang1,2* 1

State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied

Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China 2

University of Science and Technology of China, Hefei, Anhui, 230026, P.R. China

3

Department of Radiology, The Second Hospital of Jilin University, Changchun, 130041,

Changchun, China 4

Advanced Institute of Materials Science, Changchun University of Technology, 130012,

Changchun, China

Corresponding author:

Hongjie Zhang, Professor, PhD E-mail: [email protected] Yinghui Wang, PhD E-mail: [email protected] Jianhua Liu, PhD E-mail: [email protected]

1

ACS Paragon Plus Environment

ACS Nano

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

ABSTRACT Despite regulation of the reactive oxygen species (ROS) level is an intelligent strategy for cancer therapy, the therapeutic effects of ROS-mediated therapy (including photodynamic therapy (PDT) and chemodynamic therapy (CDT)) are limited by oxygen reliance, inherent flaws of traditional photosensitizers and strict reaction conditions of effective Fenton reaction. Herein, we reported biocompatible copper ferrite nanospheres (CFNs) with enhanced ROS production under irradiation with 650 nm laser through direct electron transfer and photoenhanced Fenton reaction and high photothermal conversion efficiency upon exposure to 808 nm laser, exhibiting a considerable improved synergistic treatment effect. Importantly, by exploiting the properties of O2 generation and glutathione (GSH) depletion of CFNs, CFNs relieve the hypoxia and antioxidant capability of tumor, achieving photo-enhanced CDT and improved PDT. The high relaxivity of 468.06 mM−1 s−1 enable CFNs to act as outstanding contrast agent for MRI in vitro and in vivo. These findings certify the potential of such “All in one” nanotheranostic agent integrated PDT, photo-enhanced CDT, photothermal therapy (PTT) and MRI imaging capabilities along with modulating tumor microenvironment function in theranostic of cancer. KEYWORDS: “All in one” nanotheranostic • cancer • photo-enhanced Fenton or Fentonlike reaction • modulating tumor microenvironment • magnetic resonance imaging

2

ACS Paragon Plus Environment

Page 2 of 29

Page 3 of 29

ACS Nano

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

Reactive oxygen species (ROS), including hydroxyl (·OH), superoxide (·O2-) and singlet oxygen (1O2), have been considered as a type of significant therapeutic agent for cancer when their level broken the threshold, inducing the cellular necrosis or apoptosis.1,2 As the widely explored ROS-mediated cancer treatment, photodynamic therapy (PDT) has a number of advantages over other conventional treatments, such as minimal invasiveness, low side effects and high selectivity. However, the further clinic application of PDT is severely impeded by the fact that the conventionally used organic photosensitizers suffer from poor stability, low solubility, low loading capacity, and early release from the carriers.3-6 Recently, some semiconductors and photocatalysts have been proposed as a potential photosensitizers owing to their high photostability, good biocompatibility, and broad light responsive range from UV to visible light.7-11 In spite of this, PDT is heavily dependent on O2 in essence, while the tumor microenvironment (TME) is hypoxia, thus significantly inhibiting PDT efficiency.12,13 Fenton or Fenton-like reaction can not only generate the most toxic ROS (·OH) by disproportionation reactions of H2O2,14-18 but also produce O2 in excess of H2O2,19 offering Fenton agents great potentials for cancer therapy (i.e. chemodynamic therapy, CDT) and modulating TME to improve the therapeutic effects of PDT. Due to the relatively high content of H2O2 in TME (concentration range from 100 µM to 1 mM),20 ·OH is produced only in tumor-specific microenvironment, avoiding the damage to normal tissues.21 Thus, integrating PDT and Fenton reaction to construct ROS mediated theranostic nanoplatform is a promising strategy for improving the antitumor effect. However, effective Fenton reaction requires a low pH environment (pH=3~4), whereas the pH value of tumor tissue is about 6.5.22 Despite the irradiation of UV light has been recognized as a feasible method to enhance the Fenton reaction,23 the unnegligible problems of UV light still restrict its application in tumor therapy, such as low penetration depth and potential tissue damage. Furthermore, the scaffold structure 3

ACS Paragon Plus Environment

ACS Nano

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

of theranostic nanoplatform often influenced their therapeutic and imaging effects. Therefore, it is highly desirable to design and synthesis of a single material responded to the light located in bio-windows (600~1000 nm) that possesses imaging, photocatalytic and photo-enhanced Fenton properties for efficient ROS-mediated cancer treatment. Herein, we report a biocompatible copper ferrite nanospheres (CFNs) as “All in One” theranostic agents with relatively narrow band gap of 1.71 eV, which are responsive to the light (650 nm) in bio-windows. CFNs have two redox pairs (Fe2+/Fe3+ and Cu+/Cu2+), and the coupling between them can produce more efficient ·OH through Fenton and Fenton-like reaction under 650 nm laser illumination,24-26 realizing an efficient photo-enhanced CDT. As an effective photosensitizer, cytotoxic ·OH and ·O2- can be generated by photogenerated electron/hole (eCB-/hVB+) pair of CFNs. Meanwhile, the CFNs can regulate the TME through catalyzing H2O2 to produce O2 via Fenton reaction and consuming glutathione (GSH) by Fe3+ and Cu2+, which relieve the hypoxia and antioxidant capability of tumor, so as to achieving photo-enhanced CDT and improved PDT. Moreover, the CFNs show a considerable photothermal performance under irradiation of 808 nm laser, realizing excellent synergistic tumor ablation of photo-enhanced CDT/PDT/photothermal therapy (PTT) in vitro and in vivo. Additionally, CFNs exhibited ultrahigh traverse relaxivity (468.06 mM-1 s-1), showing that CFNs could act as T2-MRI contrast agents. Therefore, CFNs are the promising “All in One” theranostic agents for MRI-guided photo-enhanced CDT/ PDT/ PTT of cancer with great modulating TME function. RESULTS AND DISCUSSION CFNs were synthesized by one-step hydrothermal method using bovine serum albumin (BSA) as surfactant to improve their biocompatibility (Scheme 1). The scanning electron microscopy (SEM, Figure 1A) and transmission electron microscope image (TEM, Figure 1B, 1C and S1, 4

ACS Paragon Plus Environment

Page 4 of 29

Page 5 of 29

ACS Nano

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

Supporting Information) shown the uniform morphology of CFNs with the average diameter of 70 nm. The lattice spacing of 0.254 nm and 0.298 nm obtained from high-resolution TEM (Figure 1D) corresponded well to the (220) and (311) planes of CuFe2O4, respectively. X-ray diffraction (XRD) pattern of CFNs reveals the cubic spinel structure of CuFe2O4 along with characteristic peaks of Cu, which is consistent with the result of the selected area electron diffraction pattern (Figure 1E and S2).27,28 The energy dispersive spectrometer (EDS)elemental mappings display that the homogeneous distribution of Fe, Cu, O, C, N and S in CFNs (Figure 1F-L), further confirming the chemical composition of CFNs. By X-ray photoelectron spectroscopy (XPS, Figure S3), it turned out that CFNs contained Cu2+, Fe3+, and O2-, corresponding to CuFe2O4.27 The peaks of N 1s and C 1s confirmed the presence of BSA, and FT-IR spectra further proved this result (Figure S4).29,30 CFNs can disperse well in water, PBS and DMEM with the hydrodynamic size of about 100 nm (Figure S5), falling within the optimal range (100-150 nm) for passive tumor targeting. The absorption range of CFNs crosses from UV to near-infrared (NIR) region owing to their relatively narrow band gap of 1.71 eV (Figure S6), making them respond to the lights in the bio-widows. Subsequently, 650 nm and 808 nm lasers are used to study the performances of photoenhanced CDT/PDT and PTT, respectively. To evaluate the TME-modulating capacity of CFNs, we detected the concentration change of GSH and O2 after the addition of CFNs. A sharp drop of the GSH level was observed because the Fe3+ and Cu2+ on the surface of CFNs were reduced by GSH, leading to the enhancement of GSH consumption with the increase of CFNs concentration (Figure 2A). As shown in Figure S7, the amount of Fe2+ obviously enhanced with the increase of concentration of GSH, proving Fe3+ on the surface of CFNs were reduced by GSH.31 Meanwhile, the XPS result of CFNs after reaction with GSH further indicated that some 5

ACS Paragon Plus Environment

ACS Nano

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

Fe(III) and Cu(II) turned into Fe(II) and Cu (I) (Figure S8).32 This process impaired the antioxidant capability of tumor.33,34 In addition, CFNs can generate O2 after the addition of H2O2 through Fenton reaction (Figure 2B), relieving the hypoxia of tumor. Such results manifest that TME modulated by CFNs is in favour of ROS-mediated therapy. The feasibility of CFNs as photosensitizers for PDT was assessed by ROS sensor agent 1,3diphenylisobenzofuran (DPBF, Figure 2C and S9). Comparing to the CFNs group and CFNs + H2O2 + 808 nm group, the absorbance of DPBF was diminished with the time of irradiation of 650 nm laser extending, ascribing to the production of ROS by the photogenerated electron (eCB-) of CFNs (Scheme 1). To identify the species of ROS, the redox potentials of the conduction band (CB) and valence band (VB) of CFNs were evaluated through the result of cyclic voltammetry (CV) curve. The ECB and EVB are determined to be -1.20 eV and 0.51 eV with respect to NHE, respectively (Figure S10). Because the ECB of CFNs is -1.20 eV, which is less than E0 of O2 /·O2- (-0.16 eV),35,36 the reaction of photogenerated electron in conduction band with O2 to yield ·O2- is thermodynamically favorable. Such results further demonstrated the CFNs could act as photosensitizers for type I-PDT. After adding H2O2, the ROS production ability markedly enhanced owing to the more O2 generation by Fenton reation, achiveing improved PDT. Then, the capacity of the CFNs to produce ·OH was investigated by the degradation of methylene blue (MB). As shown in Figure 2D and S11, the MB content did not change while there were only CFNs, whereas it took a slight dip after adding of H2O2 owing to a small amount of ·OH generation by Fenton and Fenton-like reaction. The significant reduce of the MB content (w/o O2) was observed under irradiation with 650 nm laser, indicating the photogenerated hole (hVB+) on the surface of CFNs could be play a major role in degradation MB (Figure S12).37 After introducing the different concentration of H2O2, the amount of ·OH generation obviously enhanced with the increase of H2O2 laser under the 6

ACS Paragon Plus Environment

Page 6 of 29

Page 7 of 29

ACS Nano

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

irradiation with 650 nm laser, whereas few of .OH is produced under irradiation with 808 nm (CFNs + H2O2 + 808 nm group) because the energy of 808 nm is not enough to make CFNs generate eCB-/hVB+. Such excellent ·OH production under the irradiation with 650 nm could be ascribe to the following facts: (i) H2O2 can capture eCB- to yield ·OH, realizing photoenhanced Fenton and Fenton-like reaction; (ii) the recombination of eCB- and hVB+ is limited owing to eCB- captured by H2O2, thus further enhanced the photocatalytic activity for PDT; and (iii) CFNs have porous structure and large surface area 80.397 m2 g-1 (Figure S13), which not only can enhance the enrichment of incident light, but also can increase the contact area with reactants such as H2O2 and O2.38 The excellent capacities of ROS-generation and TMEmodulating enable CFNs promising for effective photo-enhanced CDT and PDT of cancer. Furthermore,

CFNs

exhibited

good

photothermal

properties.

The

photothermal

performance was depended on the concentration of CFNs, irradiation time and irradiation power density (Figure 2E, 2F and S14). The temperature of CFNs increased from 23.9 to 46.5 o

C at concentration of 200 µg mL-1 exposed to 808 nm laser (1.3 W cm-2), exhibiting high

photothermal conversion efficiency (27.82%) and photostability (Figure S15 and S16). Collectively, these results validate that CFNs are potential “All in One” theranostic agents for photo-enhanced CDT/ PDT/ PTT. Encouraged by the above mentioned results, we further evaluated the in vitro anticancer effect and TME-modulating capacity of CFNs. The in vitro cytotoxicity of CFNs was assessed using a cell counting kit 8 (CCK-8) assay (Figure 3A). No obvious toxicity of CFNs to human cervix cancer cells (HeLa) was observed even at the concentration up to 200 µg mL1 39

,

which not only indicated their good biocompatibility owing to the existing of BSA in

CFNs, but also demonstrated the less effective Fenton reaction without light irradiation. The production of ROS in HeLa cellular environment further proved such result by intracellular 7

ACS Paragon Plus Environment

ACS Nano

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

ROS probe 2,7-dichlorofluorescin diacetate (DCFH-DA) that can be oxidized by ROS to display a green emission (Figure 3B). Comparing the control group and 650 nm laser alone group, the weak green emission signals were observed in the cells treated with CFNs because overexpressed H2O2 can react with Fe2+ and Cu+ to generate a little of ·OH without light exposure, which have negligible influence on the viability of HeLa cells. However, the cells treated with CFNs and 650 nm laser irradiation showed the strong green emission, which suggested more ROS were produced by photo-enhanced Fenton reaction and photodynamic effect. Then, we examined the cellular O2-evolving and GSH-depletion properties of CFNs by O2 probe [Ru(dpp)3]Cl2 (RDPP) and GSH assay kit. The green fluorescence was quenched by the generated O2 and GSH was almost completely consumed after the addition of CFNs (200 µg mL-1) for 24 h (Figure 3B and 3C), demonstrating that CFNs can effective regulate the microenvironment of cancer cells. The anticancer effect of CFNs was further evaluated by CCK-8 assay (Figure 3D). The cell viability decreased with the increase of the concentration of CFNs upon irradiation with 650 nm or 808 nm laser, manifesting the feasibility of CFNs to kill the cancer cells by ROS or photothermal effect alone. When the two treatments were combined, the cells was killed nearly 100% owing to the synergistically enhanced anticancer effect, indicating CFNs have a great potential for effective photo-enhanced CDT/PDT/PTT. This result can be further proved by live-dead cell staining technique (Figure 3E). Owing to their intrinsic paramagnetic properties (Figure 4A), CFNs are potential T2weighted MRI contrast agent. With increasing of Fe concentration from 0 to 0.6 mM, T2 signal intensity of CFNs dramatically augmented. And the transverse relaxivity (r2) value was calculated to be 468.06 mM−1 s−1, which is larger than a majority of reported T2-weight contrast agents (Figure 4B and Table S1). Inspired by the potency of CFNs in vitro, we 8

ACS Paragon Plus Environment

Page 8 of 29

Page 9 of 29

ACS Nano

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

explored the feasibility of CFNs for in vivo tumor ablation under the guidance of T2-weighted MRI using U14 tumor-bearing mice. The accumulation ability of CFNs in tumor with different times after intravenous injection was validated by T2-weighted MRI (Figure 4C and Figure S17). The whole tumor area became darker after 24 h post-injection, which uncovered that efficient passive targeting has been achieved in vivo since their dimension favored the enhanced permeability and retention (EPR) effect occurring in the vessels of the cancer.40-42 The biodistribution experiments of CFNs further confirmed this result (Figure 4D), which is a good basis for in vivo photo-enhanced CDT/PDT/PTT. To demonstrate the photothermal effect of CFNs in vivo, mice were exposed to different laser after injected with CFNs for 24 h. As shown in Figure 5A and S18, the temperature of tumor can rise to 57 oC within 10 min under irradiation with 808 nm laser, whereas the saline + 808 nm group and exposure to 650 nm laser group were not notably heated, which illustrated that the CFNs could act as effective photothermal agents to locally heat the tumor upon exposure to 808 nm laser. U14 tumor-bearing mice were randomly divided into six groups: (a) saline (control group), (b) 650+808 nm laser (laser group), (c) CFNs (CDT group), (d) CFNs + 650 nm laser (photo-enhanced CDT/PDT group), (e) CFNs + 808 nm laser (PTT group), (f) CFNs + 650 nm + 808 nm (photo-enhanced CDT/PDT/PTT group). The dimension and weight of tumors were measured every two days (Figure 5B, 5C and S19). Compared to the control group and the single CDT group, the photo-enhanced CDT/PDT and PTT groups showed more apparent inhibition of tumor growth. Particularly, the tumors of the photoenhanced CDT/PDT/PTT group were completely ablated (Figure 5B, 5C). Moreover, the histological analysis (H&E staining) of tumors slices further indicated that tumor tissues were damaged more seriously by photo-enhanced CDT/PDT/PTT treatment compared to other treatment schemes (Figure 5D). From Figure S20, The survival rate of control group and laser 9

ACS Paragon Plus Environment

ACS Nano

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

group are lower than the treatment group. Above results illustrate that the combination of multi-model therapies (photo-enhanced CDT/PDT/PTT) has enhanced therapeutic efficiency. What’s more, the hypoxia inducible factor (HIF-1α) staining assay was also conducted, and suggested that CFNs could relieve the hypoxia in tumor by reacting with H2O2 in vivo, overcoming the limitation of PDT and further improving the anticancer effect (Figure 5D). A study on long-term toxicity in vivo of CFNs was then carried out to prove their potential for further bioapplications. After different treatments for 30 days, no body weight change and obvious damage of normal organs were displayed (Figure 5E and S21), indicating that CFNs have low adverse effect for in vivo. CONCLUSIONS In summary, we successfully synthesized a single nanomaterial CFNs for MRI guided photoenhanced CDT, PDT and PTT through one-step hydrothermal method. As an effective photosensitizer, photogenerated eCB- of CFNs can convert O2 to ·O2- under the illumination of 650 nm laser for PDT. More importantly, the Fenton or Fenton-like reaction can be significantly enhanced under exposure of 650 nm laser, which dramatically enhances the CDT effect. In addition, the CFNs can regulate the TME through catalyzing H2O2 to produce O2 and consuming GSH, which relieve the tumor hypoxia and antioxidant capability of tumor and further improve the photo-enhanced CDT and PDT efficiency. Moreover, the CFNs show a considerable PTT effect. Such a single nanomaterial that integrates photo-enhanced CDT, PDT, PTT and MRI imaging functions along with TME-modulating capacity reflects the “All in One” concept and has tremendous potential in cancer theranostic. MATERIALS AND METHODS

10

ACS Paragon Plus Environment

Page 10 of 29

Page 11 of 29

ACS Nano

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

Chemicals and Reagents: Bovine serum albumin (BSA), Iron(III) chloride hexahydrate (FeCl3·6H2O) were purchased from Shanghai Aladdin Reagent Co. Sodium acetate (NaAc) and Methylene bue (MB) were purchased from Acros Organics (USA). Copper(II) chloride dihydrate (CuCl2·2H2O), Ethanol, Ethylene glycol and H2O2 (30%) were obtained from Beijing Chemical Reagents Company (Beijing, China). 5,5'-Dithiobis (2-nitrobenzoic Acid) (DTNB) was purchased from TCI (Shanghai, China). 1,3-Diphenylisobenzofuran (DPBF) was obtained from Alfa Aesar (USA). [Ru(dpp)3]Cl2 (RDPP), calcein acetoxymethyl ester (Calcein AM), propidium iodide (PI), 2,7-dichlorofluorescin diacetate (DCFH-DA) and glutathione (GSH) were obtained from Sigma-Aldrich (MO, America). The reduced glutathione (GSH) assay kit were purchased from Nanjing Jiancheng Bioengineering Institute. Dulbecco's Modified Eagle's Medium (DMEM) was purchased Thermo Scientific (Beijing, China). Cell Counting Kit 8 (CCK-8) assay was purchased from Changchun Sanbang Pharmaceutical Technology Co (Changchun China). Preparation of CFNs: BSA (0.5 g) was dissolved in 5 mL of deionized water. Then, 25 mL of ethylene glycol was added. After that, 2 mmol FeCl3·6H2O, 1 mmol CuCl2·2H2O and 0.75 g NaAc were added and vigorous stirred for 2 h. The mixture was added into autoclave and kept in an oven at 180°C for 24 h. Then, the precipitates were obtained by centrifuging at 12000 r min−1 for 20 min. Then, the products were washed three times by deionized water and stored at 4oC for further application. Extracellular Depletion of GSH: The consumption of GSH was monitored by UV-visible spectroscopy. DTNB PBS solution (2.5 mg mL-1, 240 µL), CFNs aqueous solution (200 µg mL-1, 200, 400, 800 and 1600 µL) and GSH aqueous solution (9.4 mM, 30 µL) were added into deionized water (2.53, 2.33, 1.93 and 1.13 mL), respectively. After that, the mixtures

11

ACS Paragon Plus Environment

ACS Nano

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

were maintained at 25°C for 30 minutes, then centrifuging the mixtures to remove CFNs, and the absorbance of the supernatant was measured by UV-vis spectroscopy. Extracellular Fe2+ Generation: CFNs (1.0 mg) were dispersed in different concentrations GSH aqueous solution (0~2 mM, 1 mL). Then, the mixtures was maintained at 25°C for 30 minutes, then centrifuging the mixtures to remove CFNs. After that, the phenanthroline solution (100 µL, 100 mM) was added to the supernatant and maintained 15 min. Finaly, the absorbance of the supernatant was measured by UV-vis spectroscopy. Extracellular O2 Generation from CFNs: 60 µL of H2O2 (1 M) was added to 30 mL of CFNs aqueous solution (150 µg mL-1) under vigorous stirring. Then, the generated concentration of O2 was monitored by a portable dissolved oxygen meters. Detection of Extracellular ·OH: CFNs (0.6 mg) were added in 3 mL of MB solution (8 mg L-1) to establish an adsorption/desorption equilibrium in dark for 1h. Then, H2O2 (1 M, 1.5, 3 and 6 µL) was added and exposed to 650 nm laser (0.469 W cm-2). After irradiation for different times (0, 5, 10 and 20 min), the MB aqueous solution was centrifuged to remove CFNs. The absorbance at 663 nm was measured by UV-vis spectroscopy. Extracellular ·O2- Detection: DPBF was used to measure the generation of extracellular ·O2-. In a typical process, 0.5 µL of H2O2 (1 M) and DPBF ethanol solution (10 mM, 20 µL) were dispersed in 2 mL of CFNs solution (200 µg mL-1, Vethanol : Vwater = 6:4). Then the mixtures were irradiated under 650 nm laser (0.469 W cm-2) for different times (0, 5, 10, 20 and 30 min). The generation of ·O2- was measured by UV-vis spectroscopy. Photothermal Effect and Thermal Stability of CFNs: Different concentrations (0~400 µg mL-1) of CFNs aqueous solutions were exposed to 808 nm laser (1.3 W cm-2) for 10 min and the temperature was recorded every 30 seconds by a thermocouple probe. Simultaneously, 12

ACS Paragon Plus Environment

Page 12 of 29

Page 13 of 29

ACS Nano

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

infrared thermal imaging camera (FLIR T420, Fluke, USA) was used to record thermal imaging every 2 min. Then, the thermal stability of CFNs solution was detected. Typically, CFNs solution (200 µg mL-1) was irradiated with 808 nm laser (1.3 W cm-2) in a quartz cuvette for 600 s, and then were naturally cooled to room temperature, above procedures were repeated four times. MRI Imaging Property: MRI images of CFNs solutions (Fe concentration: 0, 0.01875, 0.0325, 0.075, 0.15, 0.3 and 0.6 mM) were measured using a clinical MRI scanner (GE Discovery MR750 3.0T). The T2 relaxation time was measured using CFNs solution (VH2O:VD2O=1:1) with different Fe concentration (0.0325, 0.075, 0.15, 0.3 and 0.6 mM) by Bruker Avance III (9.4 T, 400 MHz) nuclear magnetic resonance spectrometer. Cytotoxicity Measurement of CFNs: HeLa cells were seeded into 96-well plates for 24 h (37 oC, 5% CO2). Then, CFNs (0, 50, 100 and 200 µg mL-1) were added into the HeLa cells for 24 h. Then, the HeLa cells were washed with PBS and incubated with CCK-8 solution (100 µL) for 4h. After that, plate reader was used to record the absorbance at 450 nm. Measurement of Intracellular Generation of O2: RDPP was used to detect the intracellular production of O2 by CLSM imaging. HeLa cells were seeded into culture dishes for 24 h (37 oC in 5% CO2). Then, RDPP medium solution (1 µM) was added to the cells for another 4 h. After that, the HeLa cells were washed with PBS and further incubated with CFNs (200 µg mL-1) for 24 h. Finally, the cells were washed by PBS and the strength of green fluorescence was obtained by CLSM with an excitation of 488 nm laser. Detection of Intracellular ROS Production: DCFH-DA was used as the probe to detect intracellular ROS. HeLa cells were seeded into culture dishes for 24 h (37 oC, 5% CO2). Then, 1 mL of CFNs medium solution (200 µg mL-1) was added and incubated for 24 h. After that, 13

ACS Paragon Plus Environment

ACS Nano

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

DCFH-DA solution (0.2 µM) was added to the cells for another 30 min. After that, the HeLa cells were washed with PBS and exposed to the 650 nm laser (0.469 W cm-2) for 15 min. The strength of green fluorescence was obtained by CLSM by an excitation of 488 nm. Detection of Intracellular GSH: The commercially available GSH assay kit was used to detect the depletion of GSH. HeLa cells were seeded in culture bottles for 24 h (37 oC, 5% CO2 ). Then, CFNs solution (0.3 µg mL-1 CFNs in medium) was added and incubated for 24 h. After that, each culture bottles was rinsed with PBS and centrifuged at 3000 rpm to collect HeLa cells. Then, the HeLa cells were suspended in 1 mL PBS and processed by ultrasound cell crusher. After that, 0.5 mL of above HeLa cells was added in 2 mL regent one in assay kit and centrifuged at 3500 rpm for 10 min. The depletion of intracellular GSH was measured by UV-vis spectroscopy. In Vitro Photothermal Therapy: HeLa cells were seeded in 96-well plates for 24 h (37 oC, 5% CO2). Then, the HeLa cells were incubated with CFNs solution (200 µg mL-1) for 24h. After that, the HeLa cells were washed with PBS and were irradiated with 808 nm laser (1.3 W cm-2) for 10 min. The cell viability was measured by CCK-8 assay and monitored by a CLSM after staining with Calcein AM and PI for 20 min. In Vitro Photo-enhanced CDT and PDT: HeLa cells were seeded in 96-well plates at a density 2.5 × 104 cell per well for 24 h. Then, the HeLa cells were incubated with CFNs medium solutionin (200 µg mL-1) for 24h, After that, the HeLa cells were washed with PBS. And culture medium (200 µL) was added in each well. Then, the HeLa cells were irradiated with 650 nm laser (0.469 W cm-2) for 15 min (1 min break after 3min irradiation). After that HeLa cells were incubated for another 24 h. The cell viability was measured by CCK-8 assay and monitored by a CLSM after staining with Calcein AM and PI for 20 min.

14

ACS Paragon Plus Environment

Page 14 of 29

Page 15 of 29

ACS Nano

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

In Vivo Therapy: We selected 60 female Kunming mice to establish the xenograft uterine cervical cancer cells (U14) tumor models. U14 tumor-bearing mice were randomly divided into six groups (n=10, in each group): (a) saline (control group), (b) 650 + 808 nm laser (NIR laser group), (c) CFNs (CDT group), (d) CFNs + 650 nm laser (photo-enhanced CDT/PDT group), (e) CFNs + 808 nm laser (PTT group), (f) CFNs + 650 + 808 nm (photo-enhanced CDT/PDT/PTT group). For each groups, CFNs (200 µg mL-1, 300 µL) were intravenously injected every two days. At 24 h post-injection, the tumors (group b, e, f) were exposed to 808 nm laser (1.3 W cm-2) for 10 min and the tumors (group b, d, f) were irradiated with 650 nm laser (0.469 W cm-2) for 15 min (1 min break after 3min irradiation). The body weight and tumor size were measured by electronic balance and digital calipers every two days. The tumor volume was obtained from Equation: Volume = (Tumor Length) × (Tumor Width)2 / 2. On the 15th day, the tumors were dissected and weighed to estimate the therapeutic efficacy. In Vivo Photothermal Imaging: The tumor-bearing mice were treated with (a) saline (300µL) + 808 nm, (b) CFNs (200 µg mL-1, 300 µL) + 650 nm and (c) CFNs (200 µg mL-1, 300 µL) + 650 nm. And infrared thermal imaging camera was used to detect the temperature of tumor every 2 min. In Vivo MRI: We used a clinical MRI scanner to observe MR imaging. The tumor-bearing mice were intravenously injected with CFNs (200 µg mL-1, 300 µL). T2-weighted MRI was obtained at different times (0, 2, 4, 12 and 24h) after intravenous injection. T2-weighted imaging with the following parameters: TR = 3000 ms, TE =104.6 ms, FOV = 200 × 200 mm. The Biodistribution of CFNs: The tumor-bearing mice were intravenously injected with CFNs solution (200 µg mL-1, 300 µL). The heart, liver, spleen, lungs, kidneys and tumor of mice were obtained through dissection and weighted at 24 h. Each group of organs and

15

ACS Paragon Plus Environment

ACS Nano

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

tumors were dissolved in digesting aqua regia (HNO3:HCl =1:3) for 48 h. The amounts of Cu were measured by ICP-AES. Histology and HIF-1α Staining: Tumor-bearing mice were dissected on the 9th day of treatment, the tumor tissues were fixed in 10% neutral buffered formalin and processed routinely into paraffi. And mice were dissected on the 30th day, the heart, liver, spleen, lung, kidney and muscle tissues were fixed in formalin and processed into paraffi. Then, the tumor tissues were sliced to 4 µm thickness for hematoxylin and eosin (H&E) and HIF-1α staining, normal organs were sliced to 4 µm thickness for H&E staining. Associated Content The Supporting Information is available free of charge on the ACS Publications website at DOI: XRD, XPS, FT-IR spectra, DLS results, zeta potential, UV-vis absorption spectra, N2 adsorption-desorption isotherm and pore size distribution of CFNs; depletion of DPBF, degradation of MB, T2-weighted MR images of mice in situ, body weight curve of mice, survival rate of mice and H&E-stained images of major organs. ACKNOWLEDGEMENTS This work was supported by the financial aid from the National Natural Science Foundation of China (Grant Nos. 51502284, 21521092, 21590794, 21635007 and 21210001), the Hong Kong, Macao and Taiwan Science and Technology Cooperation Special Project of Ministry of Science and Technology of China (No. 2014DFT10310), the Program of Science and Technology Development Plan of Jilin Province of China (No. 20140201007GX, 20170101186JC, and 2018010119JC), and the National Key Basic Research Program of China (No. 2014CB643802), the Strategic Priority Research Program of the Chinese Academy of Sciences (Grant No. XDB20030300). 16

ACS Paragon Plus Environment

Page 16 of 29

Page 17 of 29

ACS Nano

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

REFERENCES (1) Ramsey, M. R.; Sharpless, N. E. ROS as a Tumour Suppressor?. Nat. Cell. Biol. 2006, 8, 1213−1215. (2) Lau, A.; Wang, Y.; Chiu, J. F. Current Knowledge and Applications in Cancer Research and Therapeutic. J. Cell. Biochem. 2008, 104, 657−667. (3) Hou, Z.; Zhang, Y.; Deng, K.; Chen, Y.; Li, X.; Deng, X.; Cheng, Z.; Lian, H.; Li, C.; Lin, J. UV-Emitting Upconversion-Based TiO2 Photosensitizing Nanoplatform: Near-Infrared Light Mediated In Vivo Photodynamic Therapy Via Mitochondria-Involved Apoptosis Pathway. Adv. Funct. Mater. 2015, 9, 2584−2599. (4) Zhuang, X.; Ma, X.; Xue, X.; Jiang, Q.; Song, L.; Dai, L.; Zhang, C.; Jin, S.; Yang, K.; Ding, B.; Wang, P.; Liang, X. A Photosensitizer-Loaded DNA Origami Nanosystem for Photodynamic Therapy. ACS Nano 2016, 10, 3486−3495. (5) Hu, D.; Sheng, Z.; Gao, G.; Siu, F.; Liu, C.; Wan, Q.; Gong, P.; Zheng, H.; Ma, Y.; Cai, L. Activatable Albumin-Photosensitizer Nanoassemblies for Triple-Modal Imaging and Thermal-Modulated Photodynamic Therapy of Cancer. Biomaterials 2016, 93, 10−19. (6) Lv, G.; Guo, W.; Zhang, W.; Zhang, T.; Li, S.; Chen, S.; Eltahan, A.; Wang, D.; Wang, Y.; Zhang, J.; Wang, P.; Chang, J.; Liang, X. Near-Infrared Emission CuInS/ZnS Quantum Dots: All-in-One Theranostic Nanomedicines with Intrinsic Fluorescence/Photoacoustic Imaging for Tumor Phototherapy. ACS Nano 2016, 10, 9637−9645. (7) Lv, R.; Zhong, C.; Li, R.; Yang, P.; He, F.; Gai, S.; Hou, Z.; Yang, G.; Lin, J. Multifunctional Anticancer Platform for Multimodal Imaging and Visible Light Driven Photo-Dynamic/Photothermal Therapy. Chem. Mater. 2015, 27, 1751−1763. (8) Song, M.; Liu, T.; Shi, C.; Zhang, X.; Chen, X. Bioconjugated Manganese Dioxide Nanoparticles Enhance

Chemotherapy

Response

by Priming

17

ACS Paragon Plus Environment

Tumor-Associated

ACS Nano

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

Macrophages toward M1-Like Phenotype and Attenuating Tumor Hypoxia. ACS Nano 2016, 10, 633−647. (9) Yang, T.; Ke, H.; Wang, Q.; Tang, Y.; Deng, Y.; Yang, H.; Yang, X.; Yang, P.; Ling, D.; Chen, C.; Zhao, Y.; Wu, H.; Chen, H. Bifunctional Tellurium Nanodots for Photo-Induced Synergistic Cancer Therapy. ACS Nano 2017, 11, 10012−10024. (10)

Yu, Z.; Sun, Q.; Pan, W.; Li, N.; Tang, B. A Near-Infrared Triggered

Nanophotosensitizer Inducing Domino Effect on Mitochondrial Reactive Oxygen Species Burst for Cancer Therapy. ACS Nano 2015, 9, 11064−11074. (11)

Lv, R.; Yang, D.; Yang, P.; Xu, J.; He, F.; Gai, S.; Li, C.; Dai, Y.; Yang, G.; Lin, J.

Integration of Upconversion Nanoparticles and Ultrathin Black Phosphorus for Efficient Photodynamic Theranostics under 808 nm Near-Infrared Light Irradiation. Chem. Mater. 2016, 28, 4724−4734. (12)

Fingar, V. H.; Wieman, T. J.; Wiehle, S. A.; Cerrito, P. B. The Role of Microvascular

Damage in Photodynamic Therapy: The Effect of Treatment on Vessel Constriction, Permeability, and Leukocyte Adhesion. Cancer Res. 1992, 52, 4914−4921. (13)

Li, S.; Cheng, H.; Xie, B.; Qiu, W.; Zeng, J.; Li, C.; Wan, S.; Zhang, L.; Liu, W.;

Zhang, X. Cancer Cell Membrane Camouflaged Cascade Bioreactor for Cancer Targeted Starvation and Photodynamic Therapy. ACS Nano 2017, 11, 7006−7018. (14)

Zhang, C.; Bu, W.; Ni, D.; Zhang, S.; Li, Q.; Yao, Z.; Zhang, J.; Yao, H.; Wang, Z.;

Shi, J. Synthesis of Iron Nanometallic Glasses and Their Application in Cancer Therapy by a Localized Fenton Reaction. Angew. Chem., Int. Ed. 2016, 55, 2101−2106. (15)

Tang, Z.; Zhang, H.; Liu, Y.; Ni, D.; Zhang, H.; Zhang, J.; Yao, Z.; He, M.; Shi, J.; Bu,

W. Antiferromagnetic Pyrite as the Tumor Microenvironment-Mediated Nanoplatform for Self-Enhanced Tumor Imaging and Therapy. Adv. Mater. 2017, 29, 1701683.

18

ACS Paragon Plus Environment

Page 18 of 29

Page 19 of 29

ACS Nano

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

(16)

Zhou, Z.; Song, J.; Tian, R.; Yang, Z.; Yu, G.; Lin, L.; Zhang, G.; Fan, W.; Zhang, F.;

Niu, G.; Nie, L.; Chen, X. Activatable Singlet Oxygen Generation from Lipid Hydroperoxide Nanoparticles for Cancer Therapy. Angew. Chem., Int. Ed. 2017, 56, 6492−6496. (17)

Shen, Z.; Song, J.; Yung, B. C.; Zhou, Z.; Wu, A.; Chen, X. Emerging Strategies of

Cancer Therapy Based on Ferroptosis. Adv. Mater. 2018, 30, 1704007. (18)

Dai, Y.; Yang, Z.; Cheng, S.; Wang, Z.; Zhang, R.; Zhu, G.; Wang, Z.; Yung, B. C.;

Tian, R.; Jacobson, O.; Xu, C.; Ni, Q.; Song, J.; Sun, X.; Niu, G.; Chen, X. Toxic Reactive Oxygen Species Enhanced Synergistic Combination Therapy by Self-Assembled MetalPhenolic Network Nanoparticles. Adv. Mater. 2018, 1704877. (19)

Kim, J.; Cho, H. R.; Jeon, H.; Kim, D.; Song, C.; Lee, N.; Choi, S. H.; Hyeon, T.

Continuous

O2-Evolving

MnFe2O4

Nanoparticle-Anchored

Mesoporous

Silica

Nanoparticles for Efficient Photodynamic Therapyin Hypoxic Cancer. J. Am. Chem. Soc. 2017, 139, 10992−10995. (20)

Huo, M.; Wang, L.; Chen, Y.; Shi, J. Tumor-Selective Catalytic Nanomedicine by

Nanocatalyst Delivery. Nat. Commun. 2017, 8, 357. (21)

Buda, F.; Ensing, B.; Gribnau, M.; Baerends, E. O2 Evolution in the Fenton Reaction.

Chemistry 2003, 9, 3436−3444. (22)

Hu, P.; Wu, T.; Fan, W.; Chen, L.; Liu, Y.; Ni, D.; Bu, W.; Shi, J. Near Infrared-

Assisted Fenton Reaction for Tumor-Specific and Mitochondrial DNA-Targeted Photochemotherapy. Biomaterials 2017, 141, 86−95. (23)

Giannakis, S.; López, M.; Spuhler, D.; Pérez, J.; Ibáñez, P.; Pulgarin, C. Solar

Disinfection is an Augmentable, In Situ-Generated Photo-Fenton Reaction-Part 1: A Review of the Mechanisms and the Fundamental Aspects of the Process. Appl. Catal. BEnviron. 2016, 199, 199−223. 19

ACS Paragon Plus Environment

ACS Nano

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

(24)

Yao, Y.; Lu, F.; Zhu, Y.; Wei, F.; Liu, X.; Lian, C.; Wang, S. Magnetic Core-Shell

CuFe2O4@C3N4 Hybrids for Visible Light Photocatalysis of Orange II. J. Hazard. Mater. 2015, 297, 224−223. (25)

Guo, X.; Wang, K.; Li, D.; Qin, J. Heterogeneous Photo-Fenton Processes Using

Graphite Carbon Coating Hollow CuFe2O4 Spheres for the Degradation of Methylene Blue. Appl. Surf. Sci. 2017, 420, 792−801. (26)

Sharma, R.; Bansal, S.; Singhal, S. Tailoring the Photo-Fenton Activity of Spinel

Ferrites (MFe2O4) by Incorporating Different Cations (M = Cu, Zn, Ni and Co) in the Structure. RSC Adv. 2014, 5, 6006−6018. (27)

Zhu, M.; Meng, D.; Wang, C.; Diao, J. Facile Fabrication of Hierarchically Porous

CuFe2O4 Nanospheres with Enhanced Capacitance Property. ACS Appl. Mater. Interfaces 2013, 5, 6030−6037. (28)

Xiao, J.; Fang, X.; Yang, S.; He, H.; Sun, C. Microwave-Assisted Heterogeneous

Catalytic Oxidation of High-Concentration Reactive Yellow 3 with CuFe2O4/PAC. J. Chem. Technol. Biotechnol. 2015, 90, 1861−1868. (29)

Yong, Y.; Zhou, L.; Gu, Z.; Yan, L.; Tian, G.; Zheng, X.; Liu, X.; Zhang, X.; Shi, J.;

Cong, W.; Yin, W.; Zhao, Y. WS2 Nanosheet as a New Photosensitizer Carrier for Combined Photodynamic and Photothermal Therapy of Cancer Cells. Nanoscale 2014, 6, 10394−10403. (30)

Zhou, Z.; He, X.; Zhou, M.; Meng, F. Chemically Induced Alterations in the

Characteristics of Fouling-Causing Bio-Macromolecules − Implications for the Chemical Cleaning of Fouled Membranes. Water Res. 2017, 108, 115−123. (31)

Bai, J.; Jia, X.; Zhen, W.; Cheng, W.; Jiang, X. A Facile Ion-Doping Strategy to

Regulate Tumor Microenvironments for Enhanced Multimodal Tumor Theranostics. J. Am. Chem. Soc. 2018, 140, 106−109. 20

ACS Paragon Plus Environment

Page 20 of 29

Page 21 of 29

ACS Nano

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

(32)

Li, J.; Ren, Y.; Ji, F.; Lai, B. Heterogeneous Catalytic Oxidation for the Degradation of

P-Nitrophenol in Aqueous Solution by Persulfate Activated with CuFe2O4 Magnetic Nano-Particles. Chem. Eng. J. 2017, 324, 63−73. (33)

Ju, E.; Dong, K.; Chen, Z.; Liu, Z.; Liu, C.; Huang, Y.; Wang, Z.; Pu, F.; Ren, J.; Qu,

X. Copper(II)-Graphitic Carbon Nitride Triggered Synergy: Improved ROS Generation and Reduced Glutathione Levels for Enhanced Photodynamic Therapy. Angew. Chem., Int. Ed. 2016, 55, 11467−11471. (34)

Zhu, W.; Dong, Z.; Fu, T.; Liu, J.; Chen, Q.; Li, Y.; Zhu, R.; Xu, L.; Liu, Z.

Modulation of Hypoxia in Solid Tumor Microenvironment with MnO2 Nanoparticles to Enhance Photodynamic Therapy. Adv. Funct. Mater. 2016, 26, 5490−5498. (35)

He, W.; Kim, H.; Wamer, W.; Melka, D.; Callahan, J.; Yin, J. Photogenerated Charge

Carriers and Reactive Oxygen Species in ZnO/Au Hybrid Nanostructures with Enhanced Photocatalytic and Antibacterial Activity. J. Am. Chem. Soc. 2014, 136, 750−757. (36)

He, W.; Jia, H.; Wamer, W.; Zheng, Z.; Li, P.; Callahan, J.; Yin, J. Predicting and

Identifying Reactive Oxygen Species and Electrons for Photocatalytic Metal Sulfide Micro-Nano Structures. J. Catal. 2014, 320, 97−105. (37)

Pandiselvi, K.; Fang, H.; Huang, X.; Wang, J.; Xu, X.; Li, T. Constructing a Novel

Carbon Nnitride/Polyaniline/ZnO Ternary Heterostructure with Enhanced Photocatalytic Performance Using Exfoliated Carbon Nitride Nanosheets as Supports. J. Hazard. Mater. 2016, 314, 67. (38)

Chen, L.; Meng, D.; Wu, X.; Wang, A.; Wang, J.; Wang, Y.; Yu, M. In Situ Synthesis

of V4+ and Ce3+ Self-Doped BiVO4/CeO2 Heterostructured Nanocomposites with High Surface Areas and Enhanced Visible-Light Photocatalytic Activity. J. Phys. Chem. C 2016, 120, 18548−18559.

21

ACS Paragon Plus Environment

ACS Nano

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

(39)

Kanagesan, S.; Hashim, M.; Aziz, S.; Ismail, I.; Tamilselvan, S.; Alitheen, N.; Swamy,

M.; Rao, B. Evaluation of Antioxidant and Cytotoxicity Activities of Copper Ferrite (CuFe2O4) and Zinc Ferrite (ZnFe2O4) Nanoparticles Synthesized by Sol-Gel SelfCombustion Method. Appl. Sci. 2016, 6, 184. (40)

Bartlett, D. W.; Su, H.; Hildebrandt, I. J.; Weber, W. A.; Davis, M. E. Impact of

Tumor-Specific Targeting on the Biodistribution and Efficacy of siRNA Nanoparticles Measured by Multimodality In Vivo Imaging. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 15549−15554. (41)

Chen, F.; Hong, H.; Goel, S.; Graves, S. A.; Orbay, H.; Ehlerding, E. B.; Shi, S.;

Theuer, C. P.; Nickles, R. J.; Cai, W. In Vivo Tumor Vasculature Targeting of CuS@MSN Based Theranostic Nanomedicine. ACS Nano 2015, 9, 3926−3934. (42)

Ku, G.; Zhou, M.; Song, S.; Huang, Q.; Hazle, J.; Li, C. Copper Sulfide Nanoparticles

as a New Class of Photoacoustic Contrast Agent for Deep Tissue Imaging at 1064 nm. ACS Nano 2012, 6, 7489−7496.

22

ACS Paragon Plus Environment

Page 22 of 29

Page 23 of 29

ACS Nano

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

Scheme 1. Schematic illustration of synthetic process and therapeutic mechanism of CFNs.

23

ACS Paragon Plus Environment

ACS Nano

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

Figure 1. (A) SEM image of CFNs. (B,C) TEM image of CFNs. (D) HRTEM image of CFNs. (E) Selected area electron diffraction pattern of CFNs. (F-L) HAADF-STEM image and elemental mapping of Cu, Fe, O, S, N and C of CFNs. Scale bar: 50 nm.

24

ACS Paragon Plus Environment

Page 24 of 29

Page 25 of 29

ACS Nano

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

Figure 2. (A) GSH-depletion (94 µM) under the reduction of different concentrations of CFNs (13.3, 26.6, 53.2 and 106.4 µg mL-1). (B) O2 generation of CFNs (150 µg mL-1) with H2O2 (2 mM). (C) Depletion of DPBF due to .O2- generation: (a) 200 µg mL-1 of CFNs, (b) 200 µg mL-1 of CFNs with 650 nm laser (0.469 W cm-2), (c) 200 µg mL-1 of CFNs with 250 µM H2O2 and 650 nm laser, (d) 200 µg mL-1 of CFNs with 250 µM H2O2 and 808 nm laser. (D) Degradation of MB due to .OH generation: (a) 200 µg mL-1 of CFNs, (b) 200 µg mL-1 of CFNs with 1 mM H2O2, (c) 200 µg mL-1 of CFNs with 1 mM H2O2 and 808 nm laser, (d) 200 µg mL-1 of CFNs with 650nm laser, (e) 200 µg mL-1 of CFNs with 0.5 mM H2O2 and 650 nm laser, (f) 200 µg mL-1 of CFNs with 1 mM H2O2 and 650 nm laser, (g) 200 µg mL-1 of CFNs with 2 mM H2O2 and 650 nm laser. (E) Temperature curve of different concentrations of CFNs (0, 50, 100, 200 and 400 µg mL-1) under 808 nm laser (1.3 W cm-2). (F) Infrared thermal images of CFNs aqueous solutions irradiated with 808 nm laser (1.3 W cm-2) from 010 min at varied concentrations.

25

ACS Paragon Plus Environment

ACS Nano

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

Figure 3. (A) Cell viability of HeLa cells treated with different concentrations of CFNs (0, 50, 100 and 200 µg mL-1) determined by CCK-8 assay. (B) CLSM of ROS-production and O2 generation after HeLa cells incubated with CFNs (200 µg mL-1). (C) Intracellular GSHdepletion with different concentrations of CFNs (0, 100 and 200 µg mL-1). Assessment of photo-enhanced CDT/PDT/PTT multiple treatment of CFNs by (D) CCK-8 assay and (E) CLSM images.

26

ACS Paragon Plus Environment

Page 26 of 29

Page 27 of 29

ACS Nano

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

Figure 4. (A) Magnetic hysteresis loop of the CFNs. (B) In vitro T2-MRI and relaxivity plots of r2 versus the different concentrations of Fe. (C) In vivo MRI of U14 tumor-bearing mice before and after the intravenous injection of CFNs at 0, 2, 4, 12 and 24 h. (D) Biodistribution of tumor and major organs at 24 h time point.

27

ACS Paragon Plus Environment

ACS Nano

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

Figure 5. (A) In vivo thermal imaging of mice for 24h treated with (a) Control (saline + 808 nm), (b) CFNs + 650 nm, (c) CFNs + 808 nm. (B) Representative photos of mice and tumors. (C) Tumor volume curve of mice treated with (a) Control, (b) 650 + 808 nm laser, (c) CFNs, (d) CFNs + 650 nm, (e) CFNs + 808 nm and (f) CFNs + 650 nm + 808 nm, * p < 0.05 and ** p < 0.01 by the student’s two-tailed t-test. (D) H&E and HIF-1α staining of tumor slides from U14 tumor bearing mice on the 30th day. All of scale bars are 50 µm. (E) Body weight curve of mice treated with (a) Control, (b) 650 + 808 nm laser, (c) CFNs, (d) CFNs + 650 nm, (e) CFNs + 808 nm and (f) CFNs + 650 + 808 nm.

28

ACS Paragon Plus Environment

Page 28 of 29

Page 29 of 29

ACS Nano

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

Table of Content Graphic

29

ACS Paragon Plus Environment