Therapy


Cyanine-Containing Polymeric Nanoparticles with Imaging/Therapy...

3 downloads 144 Views 3MB Size

Subscriber access provided by UNIV OF CAMBRIDGE

Article

Cyanine-Containing Polymeric Nanoparticles with Imaging/TherapySwitchable Capability for Mitochondria-Targeted Cancer Theranostics Guang-Yu Pan, Hao-Ran Jia, Ya-Xuan Zhu, Wei Sun, Xiao-Tong Cheng, and Fu-Gen Wu ACS Appl. Nano Mater., Just Accepted Manuscript • DOI: 10.1021/acsanm.8b00527 • Publication Date (Web): 25 May 2018 Downloaded from http://pubs.acs.org on May 25, 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 42 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 Nano Materials

Cyanine-Containing Polymeric Nanoparticles with Imaging/Therapy-Switchable Capability for Mitochondria-Targeted Cancer Theranostics

Guang-Yu Pan, Hao-Ran Jia, Ya-Xuan Zhu, Wei Sun, Xiao-Tong Cheng, and Fu-Gen Wu*

State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China.

Corresponding Author *(F.G.W.) E-mail: [email protected]

KEYWORDS: near-infrared heptamethine cyanine dye, mitochondrial imaging, cancer/normal cell differentiation, early diagnosis of cancer, photothermal therapy

1

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

Theranostic nanoparticles (NPs) capable of mitochondrial targeting/imaging, cancer/normal cell differentiation, early-stage cancer diagnosis, and mitochondria-based photothermal therapy (PTT) were developed. The NPs were fabricated by physical encapsulation of near-infrared (NIR) heptamethine cyanine dye me-IR825 into the inner core of the micelle-forming copolymer Pluronic F127 (PF127). The PF127/me-IR825 NPs demonstrated two fluorescence emissions at ~610 nm (excited by 550 nm) and 845 nm (excited by 780 nm). The former was used for in vitro mitochondrial fluorescence imaging, cancer/normal cell differentiation, and early-stage cancer detection with high fluorescence contrast. The latter was used for in vivo NIR fluorescence imaging. Besides, the NPs could also be used for in vivo photoacoustic imaging under 808 nm excitation. After irradiation by an 808 nm laser at an elevated power density, the NPs achieved excellent photothermal tumor ablation both in vitro and in vivo. Furthermore, me-IR825 inside the inner core of PF127/me-IR825 NPs could be degraded into biocompatible products after PTT treatment, which guaranteed the post-treatment biosafety of the NPs. Benefiting from their simple preparation, good colloidal dispersibility/stability, excellent cancer/normal cell differentiation ability, and superb in vivo dual-modal imaging-guided therapeutic outcome, the PF127/me-IR825 NPs may be used as a theranostic nanoplatform for applications in the biomedical field.

2

ACS Paragon Plus Environment

Page 2 of 42

Page 3 of 42 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 Nano Materials

1. INTRODUCTION

Early and accurate diagnosis of cancer before metastasis can dramatically increase the chance of successful treatment, thus alleviating patients’ suffering and raising overall survival rate.1 Existing traditional clinical cancer diagnostic methods include physical examination, biopsy, endoscopy, computed tomography imaging, magnetic resonance imaging, ultrasound imaging, and photoacoustic imaging (PAI), etc.2 However, these techniques are still suboptimal due to many limitations, e.g., low signal-to-noise ratio, health risk caused by radiation overdose, relatively high cost, and unsatisfactory sensitivity.3 As a noninvasive, low-cost, and real-time technique with high spatial resolution, fluorescence imaging has received considerable attention in cancer diagnosis. In particular, near infrared (NIR) fluorescence-emitting probes are highly desirable due to the low absorption of tissue and blood in this transparent window (700–900 nm).4-8 However, the detection sensitivity and signal-to-noise ratio of most fluorescent probes are often insufficient for accurate cancer diagnosis, possibly due to their poor capacity to differentiate cancer cells from normal ones. To overcome these drawbacks, tumor-targeting ligands are usually conjugated to fluorescent probes to endow these probes with better tumor specificity. In addition, some imaging probes have been developed that can respond to the specific characteristics of tumor microenvironment (e.g., acidic pH and hypoxia) for amplified detection sensitivity.9 However, despite these achievements, the limited number of targeted biomarkers overexpressed on cancer cells and the insufficient difference between the microenvironment of early-stage tumors and that of normal tissues severely impede the development of highly sensitive and accurate cancer detection methods.

3

ACS Paragon Plus Environment

ACS Applied Nano Materials 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 4 of 42

Phototherapy, a light-activated local treatment modality that generally includes photothermal therapy (PTT) and photodynamic therapy (PDT),10-17 has been extensively studied due to its high anticancer efficiency. Specifically, PTT uses heat generated from optical energy to kill cancer cells and possesses huge advantages such as easy operation, short treatment time, and minimal invasiveness.18,19 In recent years, various photothermal agents (PTAs) such as metal-based nanoparticles (NPs),20-27 carbon-based nanoparticles,28,29 black phosphorus-based nanomaterials,30-33 and conjugated polymer nanoparticles have been developed.34-40 However, many developed PTAs are lack of intrinsic fluorescence property for cancer diagnosis. In addition, they usually suffer from poor biodegradability/biocompatibility (especially for heavy or toxic metal-containing PTAs), non-photodegradability, and long body retention time with potential long-term toxicity. Therefore, small organic molecule-based PTAs such as pyropheophorbide-based porphysomes41,42 and NIR cyanine dye-based PTAs43-61 have been developed for PTT. Moreover, to improve therapeutic efficiency and simultaneously minimize adverse side effects, organelle-specific precision targeted therapy is highly desired. It is known that mitochondrion is a key organelle for cell survival because of its key role in energy production and cellular apoptosis.62,63 It has been known that mitochondrion is susceptible to heat shock,64 and therefore mitochondria-targeted PTT may possibly achieve better cancer treatment performance. In view of the above facts, to develop a theranostic

PTA

with

biocompatible/biodegradable,

self-traceable,

and

mitochondria-targeting properties is becoming a focus.

In

this

work,

Pluronic

F127

(poly(ethylene 4

ACS Paragon Plus Environment

oxide)106-block-poly(propylene

Page 5 of 42 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 Nano Materials

oxide)70-block-poly(ethylene oxide)106, PEO106PPO70PEO106, abbreviated as PF127), an amphiphilic triblock copolymer which has been widely used as a building block of drug carriers in phase II/III clinical studies,65,66 was used to encapsulate a hydrophobic NIR heptamethine cyanine dye, me-IR825. The resultant PF127/me-IR825 micellar NPs can be used for mitochondrial imaging, cancer/normal cell differentiation, and mitochondria-targeted NIR PTT. (Scheme 1). Interestingly, it was found that the PF127/me-IR825 NPs have dual channel-activatable fluorescence emissions at two different excitation wavelengths: 550 nm for in vitro red-emitting fluorescence imaging (for mitochondrial imaging and normal/cancer cell differentiation), and 780 nm for in vivo NIR fluorescence imaging. Additionally, the PF127/me-IR825 NPs with strong absorption in the NIR region can be used for mitochondria-targeted PTT. On the other hand, the in vivo experiments showed that the PF127/me-IR825 NPs can target tumor tissue after intravenous injection via the enhanced permeability and retention (EPR) effect and be retained in the tumor site for a long time period. Moreover, the PF127/me-IR825 NPs exhibit excellent in vivo NIR fluorescence and photoacoustic (PA) imaging-guided PTT, resulting in effective tumor ablation. After PTT treatment, me-IR825 molecules inside the inner core of PF127/me-IR825 NPs can be photodegraded into non-toxic products, which further ensures the good biosafety of the nanoagents.

Scheme 1. Schematic Illustrating (a) the Fabrication of PF127/me-IR825 NPs and 5

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

(b)

Their

Applications

in

Mitochondrial

Imaging,

Page 6 of 42

Cancer/Normal

Cell

Differentiation, and Mitochondria-Targeted PTT.

2. EXPERIMENTAL SECTION

2.1. Preparation of PF127/me-IR825 NPs. Pluronic F127 (PF127) and me-IR825 were first dissolved in CH2Cl2 at a concentration of 40 mg mL−1 and 10 mg mL−1, respectively. Then 1800 µL of PF127 solution and 400 µL of me-IR825 solution were mixed in a 10 mL centrifuge tube, and dried under a stream of nitrogen. After thoroughly drying under vacuum at 25 oC for 24 h, 3800 µL DI water was added and the resulting suspension was sonicated at 25 oC for 30 min. The me-IR825 molecules that were not encapsulated in the nanoparticles were precipitated and 6

ACS Paragon Plus Environment

Page 7 of 42 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 Nano Materials

discarded after centrifugation at 20000 rpm for 10 min, and the transparent dark green supernatant was collected and stored at 4 oC in the dark as the stock solution for further use. The drug loading content (DLC) and encapsulation efficiency (EE) of PF127/me-IR825 NPs were quantified according to the standard curve of the concentration-dependent absorbance of me-IR825 solutions by UV–vis spectroscopy. The DLC and EE were calculated using the following formulae: DLC% = (weight of me-IR825 in nanoparticles)/(weight of nanoparticles) × 100%; EE% = (weight of me-IR825 in nanoparticles)/(weight of me-IR825 fed initially) × 100%.67 The DLC and EE of PF127/me-IR825 NPs were determined to be 5.0% and 95.0%, respectively.

2.2. Evaluation of the Photothermal Property of PF127/me-IR825. The photothermal property of PF127/me-IR825 NPs was determined using our previous method.19,20 Briefly, PF127/me-IR825 NP solutions at different concentrations (0, 100, 200, 300, 500, 800, and 1000 µg mL−1) were placed in 500 µL centrifuge tubes, followed by irradiation with an 808 nm NIR laser (beam spot 0.8 × 0.8 cm2, 1.0 W cm−2, 5 min) at room temperature. The temperature of the solutions during laser irradiation was recorded in real time using an Ai50 NIR thermal imaging camera to acquire the thermal performance curves.

2.3.

Cell

Culture.

HeLa

(human

cervical

cancer

cell

line),

A549

(adenocarcinomic human alveolar basal epithelial cell line, a type of lung cancer cell line), MCF-7 (human breast adenocarcinoma cell line), Hep G2 (human liver cancer cell line), L02 (human normal liver cell line), COS-7 (monkey normal kidney cell line), AT II (human normal lung cell line), RGC-5 (rat normal retinal ganglion cell line), and U14 (murine 7

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

cervical cancer cell line) cells were propagated in Dulbecco's modified Eagle's medium (DMEM, KeyGen

Biotech

Inc., Nanjing, China) supplemented with 100 IU mL−1

penicillin−streptomycin and 10% fetal bovine serum (FBS). All the cells were cultured at 37 °C in a humidified 5% CO2 incubator.

2.4. Other Experimental Details. Materials and reagents, characterization, other detailed descriptions on the synthesis and characterization of me-IR825, calculation of photothermal conversion efficiency (η), preparation of sulfo-cyanine5 (Cy5)-labeled PF127/me-IR825 NPs, photodegradation of PF127/me-IR825 NPs and me-IR825, and the in vitro and in vivo experiments can be found in Supporting Information.

3. RESULTS AND DISCUSSION

3.1. Preparation and Characterization of PF127/me-IR825 NPs. IR825, with a characteristic absorption peak at ~825 nm, is a type of ideal organic PTA with strong absorption but low quantum yields (QYs) in the NIR region. Generally, a high η of organic dyes is contradictory to a high QY. This is because part or a majority of the absorbed optical energy is dissipated through fluorescence emission rather than heat.68 Therefore, to prepare PTAs with high photothermal conversion efficiencies, organic dyes are encapsulated in an aggregated state in the inner core of NPs to quench the fluorescence of the organic molecules, so that the absorbed optical energy was converted into heat in its greatest extent.18 IR825, containing two carboxyl groups, possesses a certain degree of hydrophilicity. To increase the 8

ACS Paragon Plus Environment

Page 8 of 42

Page 9 of 42 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 Nano Materials

hydrophobicity of IR825 but not sacrifice its photothermal property, we herein turned the two carboxyl groups of an IR825 molecule into two methyl ester moieties and the thus-obtained methyl-esterified IR825 was abbreviated as me-IR825.

The hydrophobic heptamethine cyanine dye me-IR825 with strong adsorption at 828 nm (in ethanol) was successfully synthesized following the procedures described in the Supporting Information (see Scheme S1 and the corresponding text). The chemical structures of the intermediates and me-IR825 were confirmed by 1H nuclear magnetic resonance (NMR), 13

C NMR, and electrospray ionization mass spectrometry (ESI-MS) (Figures S1‒S6).

Me-IR825 possesses a high molar absorption coefficient of 3.13 × 105 M−1 cm−1 at the absorption peak of 828 nm (Figure S7a) but low NIR fluorescence QY of < 1% (using indocyanine green as a standard reference58,69) in ethanol, revealing that me-IR825 is an excellent PTA candidate when it is in a less polar environment. Therefore, PF127, a triblock polymeric amphiphile, was used to solubilize me-IR825 (which has poor water-solubility) and create an ethanol-like less polar environment.

PF127 molecules can self-assemble into polymeric micelles in aqueous solution with PPO segments as the hydrophobic inner core and PEO chains as the hydrophilic outer shell. The hydrophobic PPO inner core serves as a hydrophobic cavity for the encapsulation of hydrophobic drugs, and the hydrophilic PEO outer shell provides aqueous stability and sometimes endows the formed NPs with the “stealth” property, which prevents the NPs from aggregation, protein adsorption, and recognition by the mononuclear phagocyte system (MPS), thus prolonging the circulation time after systemic administration.16 Because of its 9

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

good biocompatibility and capability to encapsulate a high content of drugs and cross some biological barriers, PF127 triblock copolymer has been widely used to prepare drug carriers for phase II/III clinical studies.65,66 Therefore, we herein used PF127 to incorporate the poorly water-soluble me-IR825 molecules.

We first used dynamic light scattering (DLS) to characterize the hydrodynamic diameter of the PF127/me-IR825 complexes with different mass ratios of the two components. It was found that when the mass ratio of PF127 to me-IR825 changed from 10 : 1 to 12 : 1 and to 15 : 1, the hydrodynamic diameter of the complexes decreased from 550 ± 23 nm to 300 ± 16 nm and to 150 ± 9 nm, respectively. When the mass ratio increased to 18 : 1, the hydrodynamic diameter of the PF127/me-IR825 NPs became 20.5 ± 2.5 nm, which was larger than that of pure PF127 NPs (11.2 ± 2.0 nm) in PBS (Figure S8a and b). These results indicated the successful fabrication of PF127/me-IR825 NPs. It has been well known that nanoparticles with sizes of 10−100 nm can achieve an optimum tumor accumulation effect, and therefore, 18 : 1 was selected as the optimized mass ratio since the formed NPs have a suitable hydrodynamic diameter and a proper DLC (~5.0 wt% according to Figure S7). The successful formation of PF127/me-IR825 NPs was further confirmed by TEM. The result shown in Figure 1a demonstrates that these NPs were spherical in shape and had a homogeneous dispersion with an average diameter of 19.5 ± 10.2 nm, which was consistent to that measured by DLS (Figure S8b). Moreover, the zeta potential of PF127/me-IR825 NPs was determined to be + 37.4 ± 2.2 mV (in pure water) and + 0.8 ± 0.1 mV (in 10 mM phosphate-buffered saline, PBS). We then investigated the stability of PF127/me-IR825 NPs in physiological 10

ACS Paragon Plus Environment

Page 10 of 42

Page 11 of 42 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 Nano Materials

solutions. According to Figure S9a, the PF127/me-IR825 NPs exhibited good stability and dispersibility in these biological media, and no floccule or precipitation could be observed in all the samples even after centrifugation at 5000 rpm for 15 min. Besides, there were no significant changes in the hydrodynamic diameter of the NPs dispersed in PBS and complete culture medium during a week (Figure S9, b and c). Also, no discernible hydrodynamic diameter or zeta potential changes of the NPs were observed in the samples after storage at 4 o

C in the dark for 60 days (Figure S9, d and e), suggesting the excellent colloidal stability of

PF127/me-IR825 NPs, which was very important for their further applications in biological systems.

We next investigated the UV−vis absorption and fluorescence properties of PF127/me-IR825 NPs using UV−vis absorption spectroscopy and fluorescence spectroscopy, respectively. The UV−vis absorption spectra of me-IR825 and PF127/me-IR825 NPs in ethanol solutions exhibited the same UV−vis absorption pattern (Figure S7), while the strong peak at 828 nm became significantly weakened and broadened in aqueous solutions (Figure 1b, Figure S7b, and Figure S10), with a 7 nm red-shift as compared with the absorption peaks in the spectra of me-IR825 in ethanol and PF127/me-IR825 NPs in ethanol. The UV−vis absorption spectrum of PF127/me-IR825 NPs in water was similar to that of me-IR825 in aqueous solution, which may be due to the fact that some me-IR825 molecules inside the inner core of the NPs form aggregates via π-π stacking. The similar absorption pattern of me-IR825 and PF127/me-IR825 NPs in ethanol indicates that when dispersed in ethanol, the PF127/me-IR825 NPs would dis-assemble into free PF127 and me-IR825 molecules, forming 11

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

similar microenvironment (the ethanol solvent) of the me-IR825 molecules in both samples and thus the same absorption property. This further confirms that the two components are self-assembled into NPs via hydrophobic interaction because ethanol can disassociate the hydrophobic interaction in self-assembled complexes. Besides, as revealed in Figure 1c, when excited separately at 550 and 780 nm, PF127/me-IR825 NPs showed fluorescence emission peaks at 610 and 845 nm, respectively. Herein, we selected 550 nm for in vitro visible-light fluorescence imaging (with a relatively high fluorescence QY), and 780 nm for in vivo NIR fluorescence imaging (with minimum background interference and deep tissue penetration). It should be noted that the fluorescence emission intensities of PF127/me-IR825 NPs in water at both 610 and 845 nm were stronger than that of me-IR825 in water, but weaker than that of me-IR825 in ethanol at the same concentration (Figure S11). These results can be explained by the fact that me-IR825 molecules mainly existed as aggregates in water, so that the fluorescence of me-IR825 at both wavelengths was quenched; whereas me-IR825 mainly existed as monomers in ethanol, thus emitting strong fluorescence. For PF127/me-IR825 NPs, there were both aggregated forms and monomers of me-IR825 molecules inside the hydrophobic PPO inner core, leading to a moderate fluorescence emission intensity at the two wavelengths.

We finally investigated the photothermal properties of the PF127/me-IR825 NPs. PF127/me-IR825 NP solutions at different concentrations were irradiated by an 808 nm laser (1.0 W cm−2) for 5 min. Remarkable concentration-dependent temperature increase of PF127/me-IR825 NP solutions was observed under laser irradiation; by contrast, PBS solution 12

ACS Paragon Plus Environment

Page 12 of 42

Page 13 of 42 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 Nano Materials

showed negligible change in temperature under the same irradiation condition (Figure 1d). Additionally, the η of PF127/me-IR825 NPs was calculated to be 17.2% (Figure S12). These results demonstrate high η of PF127/me-IR825 NPs upon NIR laser irradiation, and suggest that PF127/me-IR825 NPs can be used as an efficient PTA for inducing tumor hyperthermia (above 42 oC53,54).

Figure 1. (a) TEM image of PF127/me-IR825 NPs and the corresponding size distribution histogram (inset). (b) UV−vis absorption spectrum of PF127/me-IR825 NPs dispersed in PBS (500 µg mL−1). Inset shows the photograph of the corresponding solution. (c) Fluorescence spectra of PF127/me-IR825 NPs dispersed in PBS (100 µg mL−1). (d) Photothermal heating curves of PF127/me-IR825 NP solutions at different concentrations under the continuous 808

13

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

nm laser irradiation (1.0 W cm−2) for 5 min.

3.2. Cellular Uptake and Localization of PF127/me-IR825 NPs. We first investigated the cellular internalization and localization of PF127/me-IR825 NPs using confocal laser scanning microscopy and flow cytometry. HeLa cells treated with the PF127/me-IR825 NPs (100 µg mL−1) showed a time-dependent cellular internalization within 24 h, as demonstrated by the confocal imaging and flow cytometric results (Figure 2a and c). Specifically, the fluorescence intensity of the treated HeLa cells reached a relatively stable plateau after 4 h, and therefore, 4 h was selected as the appropriate incubation time to achieve a high drug internalization. Importantly, PF127/me-IR825 NPs did not disassemble after being internalized by the cells, which was confirmed by the Cy5-labeled PF127/me-IR825 NPs (Figures S13–15). Cy5-labeled PF127 molecules alone showed negligible uptake by A549 cells (Figure S14). However, the fluorescence signal of Cy5 overlaps well with that of me-IR825 after the cellular uptake of Cy5-labeled PF127/me-IR825 NPs with Pearson’s correlation coefficient (PCC) of 0.89 (Figure S15), suggesting the integrity of Cy5-labeled PF127/me-IR825 after cellular internalization. Then the cellular uptake mechanism of PF127/me-IR825 NPs was also investigated, and a significantly decreased uptake quantity of the NPs was observed after low temperature treatment (Figure 2b and d), indicating that the internalization of PF127/me-IR825 NPs is a temperature- and energy-dependent process, since the low temperature treatment can inhibit ATP production. Further, the possible internalization pathways that would associate with the energy-dependent cellular uptake of 14

ACS Paragon Plus Environment

Page 14 of 42

Page 15 of 42 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 Nano Materials

PF127/me-IR825 NPs were explored. We first pretreated HeLa cells with specific endocytosis inhibitors: 5-(N,N-dimethyl)-amiloride hydrochloride (amiloride, a macropinocytosis inhibitor), chlorpromazine hydrochloride (CPZ, a clathrin-mediated endocytosis inhibitor), genistein (a caveolae-mediated endocytosis inhibitor), and methyl-β-cyclodextrin (MβCD, a lipid raft-mediated endocytosis inhibitor), respectively, followed by incubation with PF127/me-IR825 NPs. Flow cytometry was used to quantify the cellular uptake amounts of the NPs. As depicted in Figure 2b and 2d, the uptake quantity of PF127/me-IR825 NPs was markedly inhibited (by ~30%) only in the genistein-pretreated group, indicating that the cellular internalization of PF127/me-IR825 NPs was caveolae-mediated energy-dependent endocytosis. Because caveolae is a mitochondrial function-related domain,70 and therefore, this endocytosis pathway of PF127/me-IR825 NPs may be related to their mitochondrial

accumulation.

15

ACS Paragon Plus Environment

ACS Applied Nano Materials 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) Confocal fluorescence images of HeLa cells after treatment with PF127/me-IR825 NPs (100 µg mL−1) for different time periods as indicated. Scale bar = 20 µm. (b) Confocal fluorescence images of HeLa cells treated with PF127/me-IR825 NPs (100 µg mL−1) after pretreatment without (control) and with 4 °C or different endocytosis inhibitors as indicated. Scale bar = 20 µm. (c) Flow cytometry analysis of the fluorescence intensity of HeLa cells after incubation with PF127/me-IR825 NPs (100 µg mL−1) for various time periods. (d) Flow cytometry analysis of the relative uptake efficiency of PF127/me-IR825 NPs for cells in (b).

16

ACS Paragon Plus Environment

Page 16 of 42

Page 17 of 42 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 Nano Materials

Next, we investigated the intracellular location of PF127/me-IR825 NPs by co-staining the live HeLa cells with PF127/me-IR825 NPs and one of the following commercially available organelle-selective trackers: rhodamine 123 (Rhod 123, for staining mitochondria), ER-Tracker Green (ER-Tracker, for staining endoplasmic reticulum, ER), LysoTracker Green (LysoTracker, for staining lysosomes), and Golgi-Tracker Green (Golgi-Tracker, for staining Golgi apparatus), respectively. As shown in Figure 3, the fluorescence of PF127/me-IR825 NPs overlapped nicely with that of Rhod 123. The corresponding PCC and overlap coefficient (OLC) were 0.95 and 0.96, respectively. In contrast, PF127/me-IR825 NPs did not stain ER well, and the corresponding PCC and OLC were 0.64 and 0.68, respectively. Further, a poor overlap was seen in the case of lysosomes and Golgi apparatus, with PCC of 0.09 and 0.04, respectively. These results suggest that PF127/me-IR825 NPs have a high binding specificity towards mitochondria. To further verify this conclusion, cells were treated with 1 µM staurosporine (STS), which can disrupt the mitochondrial membrane potential (MMP). It is well known that the MMP of mitochondria is up to −180 mV, and such a large MMP may be responsible for the selective accumulation of cationic species in mitochondria instead of other organelles.3 As shown in Figure S16, after treatment with STS for 6 h, the fluorescence of PF127/me-IR825 NPs and Rhod 123 in HeLa cells were largely reduced. This result not only points out that the binding of the cationic PF127/me-IR825 NPs to mitochondria was MMP-dependent,

but

also

confirms

the

specific

mitochondrial

accumulation

of

PF127/me-IR825 NPs. We further investigated the time-dependent mitochondrial localization of PF127/me-IR825 NPs, and the results shown in Figure S17 demonstrate that the NPs could

17

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

realize long-term (at least 24 h) mitochondrial localization with a high PCC of 0.96 after 24 h of incubation. Furthermore, the excellent mitochondrial targeting of PF127/me-IR825 NPs could also be realized in various other cell types. As shown in Figure S18, the fluorescence emission of PF127/me-IR825 NPs coincided well with that of Rhod 123 in A549 (PCC = 0.91), MCF-7 (PCC = 0.92), and Hep G2 (PCC = 0.92) cells. The above results indicate that PF127/me-IR825 NPs are excellent mitochondria-targeted nanodrugs.

Figure 3. Confocal microscopy images of HeLa cells costained with PF127/me-IR825 NPs (100 µg mL−1) and one of the various commercially available organelle-selective trackers as

18

ACS Paragon Plus Environment

Page 18 of 42

Page 19 of 42 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 Nano Materials

indicated. Scale bar = 20 µm. Plots shown in the right column represent the colocalization correlation intensity plots of PF127/me-IR825 NPs and the commercial probes.

3.3. Cancer/Normal Cell Differentiation Using PF127/me-IR825 NPs. It has been reported that cancer cells have a more negative MMP than normal cells, with a difference of larger than 60 mV,71 and thus some positively charged molecules or nanoparticles may preferably target the mitochondria of cancer cells but not those in normal cells. Besides, because tumor tissue has a much faster (~9 times) metabolic rate than normal tissue,3 cancer cells will absorb more substances than normal cells. These above-mentioned reasons make cancer cells strive for more PF127/me-IR825 NPs than normal cells. To verify if PF127/me-IR825 NPs are suitable for differentiating cancer cells from normal ones, four kinds of normal cells (L02, RGC-5, COS-7, and AT II) and four types of cancer cells (HeLa, Hep G2, MCF-7, and A549) were separately incubated with 100 µg mL−1 PF127/me-IR825 NPs for 30 min, followed by observation using a confocal microscope. Interestingly, the mitochondria with strong red fluorescence signals were clearly observed in all the cancerous cells; however, much weaker fluorescence was seen in all the normal cells (Figure 4a). These results were further confirmed by the three-dimensional confocal imaging of A549/AT II cells (Figure S19). Additionally, the quantitative results measured by flow cytometry indicated that the uptake amount of the NPs by cancer cells was much higher than that by normal cells (Figure 4b and c). The above results demonstrate that PF127/me-IR825 NPs are capable of cancer/normal cell differentiation with high efficiency.

19

ACS Paragon Plus Environment

ACS Applied Nano Materials 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) Confocal fluorescence images of four kinds of normal cells (L02, RGC-5, COS-7, and AT II) and four types of cancerous cells (HeLa, Hep G2, MCF-7, and A549) after treatment with PF127/me-IR825 NPs (100 µg mL−1). Scale bar = 20 µm. (b) Flow cytometric results of different cells treated with PF127/me-IR825 NPs (100 µg mL−1) for 30 min. (c) Relative mean fluorescence intensities of different cells obtained from the data in (b).

Next, to mimic the real situation of the tumor site, the A549 lung cancer cells and AT II 20

ACS Paragon Plus Environment

Page 20 of 42

Page 21 of 42 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 Nano Materials

normal lung cells were co-cultured in a 96-well confocal dish separately, with the A549/AT II cell number ratio of 1 : 1, 1 : 2, 1 : 5, and 1 : 10, respectively. After 24 h of incubation, all the cells in the same dish were incubated with PF127/me-IR825 NPs (100 µg mL−1) for 30 min, followed by imaging using a confocal microscope. Interestingly, only the cancerous A549 cells displayed bright red fluorescence signals, while the normal AT II cells emitted negligible fluorescence in all the co-cultured cell samples (Figure 5). Furthermore, the markedly different fluorescence signals of two cells did not change within 24 h (Figure S20). The results revealed that even under the cancerous/normal cell co-culture conditions, the PF127/me-IR825 NPs could still selectively interact with the mitochondria of cancerous cells, indicating the feasibility of using the PF127/me-IR825 NPs as mitochondria-targeted nanotheranostics for cancer.

21

ACS Paragon Plus Environment

ACS Applied Nano Materials 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) Confocal microscopy images of the co-cultured cancerous A549 and normal AT II cells with the A549 to AT II cell number ratios of (a−c) 1 : 10, (d−f) 1 : 5, (g−i) 1 : 2, and (j−l) 1 : 1. The A549 cells are highlighted by white dotted circles in the fluorescence channel. Scale bar = 25 µm.

22

ACS Paragon Plus Environment

Page 22 of 42

Page 23 of 42 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 Nano Materials

Finally, to further mimic the real early-stage situation of cancer disease (in which a very small quantity of cancerous cells are usually surrounded by numerous normal cells71), the A549 and AT II cells were co-cultured in the same well of a confocal dish with the A549 to AT II cell number ratio of 1 : 100. After 24 h, all the cells were treated with 100 µg mL−1 PF127/me-IR825 NPs for 30 min, followed by imaging via a confocal microscope. As displayed in Figure S21, even when the number ratio of the two cells reached 1 : 100, we could still observe clearly the red fluorescent spots that indicate the presence of the PF127/me-IR825 NP-stained cancerous A549 cells. Besides, the marked fluorescence difference of the two types of cells did not change within 24 h (Figure S21). Collectively, the above-mentioned results indicate that PF127/me-IR825 NPs can be used as an effective fluorescent nanoprobe for early-stage cancer diagnosis.

3.4. Cytotoxicity Evaluation of PF127/me-IR825 NPs. As an ideal theranostic PTA, its biocompatibility is a prerequisite for its practical applications. The potential dark cytotoxicity of

PF127/me-IR825

NPs

to

cells

was

first

investigated

by

MTT

(3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide) assay. Our results show that after incubation of cells with PF127/me-IR825 NPs (0–500 µg mL−1) for 24 h, the cell viabilities of four types of cells (HeLa, A549, MCF-7, and U14) were larger than 85% even at a concentration of up to 500 µg mL−1 (Figure 6a and S22), suggesting the negligible adverse effect of PF127/me-IR825 NPs to cells in the dark.

23

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

We then assessed the cytotoxicity of PF127/me-IR825 NPs to HeLa cells after treatment with NIR irradiation. The MTT assay results revealed a concentration-dependent cytotoxicity of the NPs. Specifically, after 10 min of NIR light irradiation (808 nm, 1.0 W cm−2), > 90% of the cells were killed at the concentration of 500 µg mL−1 (Figure 6a). Meanwhile, confocal fluorescence imaging of calcein acetoxymethyl ester (calcein-AM, a fluorescent dye that stains live cells green)- and propidium iodide (PI, a fluorescent dye that stains dead cells red)-costained HeLa cells after PTT treatment also verified the high PTT efficiency mediated by PF127/me-IR825 NPs (Figure 6b). Furthermore, the phototoxicity of PF127/me-IR825 NPs toward other tumor cell lines such as A549, MCF-7, and U14 was examined. The killing efficiency of the NPs towards all the tested cancer cells was > 90% under the same irradiation condition, whereas the NP-treated cells without NIR laser irradiation exhibited negligible cytotoxicity over the entire concentration range (Figure S22), indicating the great potential of the PF127/me-IR825 NPs to be used as a biocompatible and effective PTA for cancer therapy. It has been reported that, mitochondria-targeted PTT is a very effective cancer treatment modality, and a slight temperature elevation in mitochondria can result in cell apoptosis due to the high sensitivity of mitochondria to hyperthermia.64 Thus, the mitochondrial localization of the PF127/me-IR825 NPs would be highly beneficial for enhancing their PTT efficiency. Additionally, the MTT assay results showed that upon NIR laser irradiation, the viability of the PF127/me-IR825 NP-treated co-cultured normal AT II/cancer A549 cells (with a cell number ratio of 1 : 1) became ~55% (Figure S23a). Similarly, in the calcein-AM and PI costaining assay, around half of the cells were dead (stained red by PI) after laser irradiation

24

ACS Paragon Plus Environment

Page 24 of 42

Page 25 of 42 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 Nano Materials

(Figure S23b). These results indicated that the cancerous A549 cells in the co-culture system might be killed by the combined PF127/me-IR825 NP and laser treatment.

Figure 6. (a) Relative viabilities of PF127/me-IR825 NP-treated HeLa cells without or with NIR laser irradiation. (b) Confocal fluorescence images of PF127/me-IR825 NP (500 µg mL−1)-treated HeLa cells after 808 nm laser irradiation at different power densities for 10 min. The cells were costained by calcein-AM and PI before imaging. Scale bar: 200 µm. (c−h) Flow cytometric analysis of HeLa cell death induced by various treatments as indicated.

25

ACS Paragon Plus Environment

ACS Applied Nano Materials 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 42

Finally, we used flow cytometry to investigate the effect of NIR and/or PF127/me-IR825 NPs on the apoptosis/necrosis of HeLa cells using the annexin V-fluorescein isothiocyanate (annexin V-FITC) and PI double staining assay. NIR laser irradiation or PF127/me-IR825 NPs alone did not elicit discernible cytotoxicity (Figure 6c−e). In sharp contrast, the cell viability of “PF127/me-IR825 NPs + laser”-treated groups decreased with increasing NP concentration (Figure 6f−h). Specifically, in the “500 µg mL−1 PF127/me-IR825 NPs + laser”-treated group, most of the cells exhibited features of late apoptosis and/or necrosis, and only 5.44% cells remained survival. These results indicate that PTT mainly induced the death of HeLa cells via late apoptosis and/or necrosis.

3.5. Cytotoxicity Evaluation of PF127/me-IR825 NPs after Photodegradation. A key feature of cyanine dyes is that they can easily be photodegraded into several small molecules upon NIR laser irradiation due to the regioselective oxidation of their polyene chains by the generated singlet oxygen.72-74 As shown in Figure S24, the absorption peak of me-IR825 in PF127/me-IR825 NPs (500 µg mL−1) significantly decreased after 6 cycles of irradiation, indicating the photodegradation of me-IR825 molecules in PF127/me-IR825 NPs. Besides, the

matrix-assisted

laser

desorption/ionization

time-of-flight

(MALDI-TOF)

mass

spectrometry result showed that the me-IR825 molecules were photodegraded into several carbonyl compounds (Figure S25). Moreover, the MTT results revealed that the products of PF127/me-IR825 NPs after laser irradiation elicited negligible cytotoxicity towards normal AT II cells over the entire concentration range of 0−2000 µg mL−1 (Figure S26), indicating the 26

ACS Paragon Plus Environment

Page 27 of 42 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 Nano Materials

good biocompatibility of the PF127/me-IR825 NPs even after the PTT treatment.

3.6. In Vivo NIR Fluorescence/PA Dual Modal Imaging Guided PTT. Motivated by the excellent cancer cell-targeting performance and satisfactory in vitro PTT efficacy of PF127/me-IR825 NPs, we then evaluated the in vivo tumor targeting specificity, biodistribution, and therapeutic potential of PF127/me-IR825 NPs in tumor-bearing nude mice. Figure 7 (a and c) shows that, after intravenous injection of the NPs, the average NIR fluorescence signals at the tumor sites first increased with time during the 0‒24 h period, reached the maximum at 24 h postinjection, and then gradually decreased over time after 24 h. These results confirm the effective accumulation of PF127/me-IR825 NPs in the tumor tissues due to the EPR effect. By contrast, no NIR fluorescence signals were observed in the tumor sites for the me-IR825 group (Figure 7, a and c), suggesting the poor tumor accumulation of me-IR825. We then investigated the biodistribution of PF127/me-IR825 NPs via ex vivo NIR fluorescence imaging of major organs and tumors. As displayed in Figure 7 (b and d), the results further confirm the excellent tumor targeting of the PF127/me-IR825 NPs. Apart from NIR fluorescence imaging, PF127/me-IR825 NPs also hold great potential in PAI in vivo. We first investigated the PAI performance of PF127/me-IR825 NP solutions. Upon excitation at 808 nm, the PA signal intensities of PF127/me-IR825 NP solutions increased with concentration (Figure 7e), suggesting the excellent PAI performance of the nanoagents. Next, the in vivo PAI capability of PF127/me-IR825 NPs was further investigated. As shown in Figure 7f and g, the PA signal of tumor-bearing mice intravenously injected with PF127/me-IR825 NP solution was clearly observed. The PAI result further confirmed the 27

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

preferential accumulation of PF127/me-IR825 NPs in the tumor tissue. Furthermore, it should be noted that the PF127/me-IR825 NPs could be gradually cleared from the mice body. As shown in Figure S27, a relatively complete body clearance of the NPs was achieved in mice after 5 and 7 days postinjection, ensuring the good biosafety of the PF127/me-IR825 NPs.

Figure 7. (a) In vivo NIR fluorescence images of tumor-bearing mice taken at different time

28

ACS Paragon Plus Environment

Page 28 of 42

Page 29 of 42 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 Nano Materials

points after intravenous injection of PF127/me-IR825 NP (dose: 300 mg kg−1) solutions or me-IR825 (dose: 5 mg kg−1) solutions. The green dotted circles indicate the tumor sites. (b) Ex vivo NIR fluorescence images of various main organs and tumors excised from mice after treatment with PF127/me-IR825 NPs. (c) Corresponding average fluorescence intensities of tumors in PF127/me-IR825 NP- or me-IR825-treated mice in (a). (d) Corresponding average fluorescence intensities of various main organs and tumors in (b). (e) PA images of PF127/me-IR825 NPs in water at various concentrations. (f) PA images of mice taken at 12 h after intravenous injection of PF127/me-IR825 NP (dose: 300 mg kg−1) solution. Tumor sites are highlighted with white dotted circles. Scale bar: 4 mm. (g) Corresponding PA intensites in the tumor sites in (f).

Based on the high in vitro PTT efficacy and excellent tumor accumulation performance of the PF127/me-IR825 NPs, we next investigated the hyperthermia effect and photothermal tumor ablation capability of the NPs in vivo. As exhibited in Figure S28, the average temperature of the tumor region in the mouse treated with PF127/me-IR825 NPs and laser irradiation reached 47.5 °C, which is sufficient to cause tumor damage. In sharp contrast, the PBS-treated group only had a slight temperature elevation (~4 °C) in the tumor region after the same laser irradiation. The above results demonstrate that the PF127/me-IR825 NPs can induce satisfactory in vivo hyperthermia effect. To evaluate the in vivo photothermal tumor ablation ability and biosafety of the NPs, four groups of tumor-bearing mice were treated separately as follows: PBS, PBS + laser irradiation, PF127/me-IR825 NPs, PF127/me-IR825 29

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

NPs + laser irradiation. After that, we monitored the tumor size and body weight every other day during the subsequent two weeks. As shown in Figure 8 (a and b), the combined treatment of PF127/me-IR825 NPs and laser irradiation completely ablated the tumors in mice, leaving black scars at the original sites. By contrast, the tumors in the other groups all grew rapidly with time. These encouraging in vivo PTT results were further confirmed by the histological examination of the tumor slices. Severe damages could only be observed in the tumor tissues from mice receiving both PF127/me-IR825 NPs administration and laser irradiation (Figure 8a). Besides, the body weights of mice in each group maintained in a normal range throughout the experimental period (Figure 8d), revealing that all treatments were safe. To further assess the biocompatibility of the PF127/me-IR825 NPs, histological examination was performed for the major organs including hearts, livers, spleens, lungs, and kidneys of mice on the 14th day postinjection. It can be seen that the hematoxylin and eosin (H&E)-stained histological sections of these major organs did not show noticeable differences as compared with those in the control group (Figure 8c), proving the good biosafety of the PF127/me-IR825 NPs for tumor ablation.

30

ACS Paragon Plus Environment

Page 30 of 42

Page 31 of 42 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 Nano Materials

Figure 8. (a) Representative photographs of tumor-bearing mice taken on the 14th day after different treatments and the corresponding H&E-stained tumor slices. Scale bar = 100 µm. (b) The tumor growth curves of mice in different groups. ***p < 0.001 (one way analysis of variance, ANOVA). (c) H&E-stained slices of major organs from mice sacrificed on the 14th day after treatment with PBS (control) or PF127/me-IR825 NP (dose: 300 mg kg−1) solution. Scale bar = 100 µm. (d) Time-dependent changes of the body weights of mice in different groups.

4. CONCLUSIONS

In summary, we propose a simple method of fabricating water-dispersible and multifunctional PF127/me-IR825

NPs

for

applications

including

mitochondrial

targeting/imaging,

cancer/normal cell differentiation, and mitochondria-targeted PTT. The PF127/me-IR825 NPs

31

ACS Paragon Plus Environment

ACS Applied Nano Materials 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 32 of 42

have dual-channel activatable fluorescence emissions with 610 nm for in vitro red-emitting fluorescence imaging and 845 nm for in vivo NIR fluorescence imaging. With their remarkable

NIR

fluorescence

emission,

strong

NIR

absorption,

and

excellent

tumor-targeting/retention properties, the PF127/me-IR825 NPs can successfully realize in vivo NIR fluorescence and PA dual-modal imaging-guided PTT. More interestingly, the NPs achieve high fluorescence contrast in cancerous and normal cells, allowing for the accurate early-stage cancer diagnosis. Furthermore, the good biocompatibility of the two components and the photodegradability of me-IR825 ensure the excellent biosafety of the PF127/me-IR825 NPs. Overall, the PF127/me-IR825 NPs may hold great promise for mitochondrial imaging, early-stage cancer diagnosis, and mitochondria-targeted photothermal cancer therapy.

ASSOCIATED CONTENT Supporting Information The Supplementary Information is available free of charge on the ACS Publications website at DOI:

Materials and reagents, characterization, and other experimental details; NMR and ESI-MS spectra, UV−vis absorption spectra, DLS data, colloidal stability assay, fluorescence spectra, calculation of photothermal conversion efficiency, confocal images, cell viabilities, MALDI-TOF mass spectrum, ex vivo fluorescence images, and tumor-site temperature (PDF) 32

ACS Paragon Plus Environment

Page 33 of 42 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 Nano Materials

AUTHOR INFORMATION Corresponding Author E-mail: [email protected] Notes

The authors declare no competing financial interest.

ACKNOWLEDGMENTS This work was supported by grants from the Natural Science Foundation of Jiangsu Province (BK20170078) and National Natural Science Foundation of China (21673037).

REFERENCES (1) Huang, X. L.; Liu, Y. J.; Yung, B.; Xiong, Y. H.; Chen, X. Y. Nanotechnology-Enhanced No-Wash Biosensors for in Vitro Diagnostics of Cancer. ACS Nano 2017, 11, 5238‒5292. (2) Fan, W. P.; Yung, B.; Huang, P.; Chen, X. Y. Nanotechnology for Multimodal Synergistic Cancer Therapy. Chem. Rev. 2017, 117, 13566‒13638. (3) Gao, G.; Jiang, Y. W.; Yang, J. J.; Wu, F. G. Mitochondria-Targetable Carbon Quantum Dots for Differentiating Cancerous Cells from Normal Cells. Nanoscale 2017, 9, 18368‒ 18378. (4) Gao, M.; Yu, F. B.; Lv, C. J.; Choo, J.; Chen, L. X. Fluorescent Chemical Probes for Accurate Tumor Diagnosis and Targeting Therapy. Chem. Soc. Rev. 2017, 46, 2237‒ 2271. 33

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

(5) Zheng, X. C.; Wang, X.; Mao, H.; Wu, W.; Liu, B. R.; Jiang, X. Q. Hypoxia-Specific Ultrasensitive Detection of Tumours and Cancer Cells in Vivo. Nat. Commun. 2015, 6, 5834. (6) Liu, Z. G.; Rong, P. F.; Yu, L.; Zhang, X. T.; Yang, C. J.; Guo, F.; Zhao, Y. Z.; Zhou, K. C. Wang, W.; Zeng, W. B. Dual-Modality Noninvasive Mapping of Sentinel Lymph Node by Photoacoustic and Near-Infrared Fluorescent Imaging Using Dye-Loaded Mesoporous Silica Nanoparticles. Mol. Pharmaceutics, 2015, 12, 3119−3128. (7) Gao, T.; Cao, X. Z.; Dong, J.; Liu, Y.; Lv, W. W.; Li, C. M.; Feng, X. P.; Zeng, W. B. A Novel Water Soluble Multifunctional Fluorescent Probe for Highly Sensitive and Ultrafast Detection of Anionic Surfactants and Wash Free Imaging of Gram-Positive Bacteria Strains. Dyes Pigm. 2017, 143, 436−443. (8) Wang, X. B.; Wang, S. L.; Huang, K. Z.; Liu, Z. G.; Gao, Y. J.; Zeng, W. B. A Ratiometric Upconversion Nanosensor for Visualized Point-of-Care Assay of Organophosphonate Nerve Agent. Sens. Actuator B–Chem. 2017, 241, 1188−1193. (9) Zheng, X. C.; Mao, H.; Huo, D.; Wu, W.; Liu, B. R.; Jiang, X. Q. Successively Activatable Ultrasensitive Probe for Imaging Tumour Acidity and Hypoxia. Nat. Biomed. Eng. 2017, 1, 0057. (10) Chakrabortty, S.; Agrawalla, B. K.; Stumper, A.; Vegi, N. M.; Fischer, S.; Reichardt, C.; Kögler, M.; Dietzek, B.; Feuring-Buske, M.; Buske, C.; Rau, S.; Weil, T. Mitochondria Targeted Protein-Ruthenium Photosensitizer for Efficient Photodynamic Applications. J. Am. Chem. Soc. 2017, 139, 2512−2519. (11) Zhang, Y.; Wang, F. M.; Liu, C. Q.; Wang, Z. Z.; Kang, L. H.; Huang, Y. Y.; Dong, K.; Ren, J. S.; Qu, X. G. Nanozyme Decorated Metal–Organic Frameworks for Enhanced Photodynamic Therapy. ACS Nano 2018, 12, 651–661. (12) Li, X. S.; Yu, S.; Lee, D.; Kim, G.; Lee, B.; Cho, Y.; Zheng, B. Y.; Ke, M. R.; Huang, J. D.; Nam, K. T.; Chen, X. Y.; Yoon, J. Facile Supramolecular Approach to Nucleic-Acid-Driven Activatable Nanotheranostics That Overcome Drawbacks of Photodynamic Therapy. ACS Nano 2018, 12, 681–688. (13) Cheng, Y. H.; Cheng, H.; Jiang, C. X.; Qiu, X. F.; Wang, K. K.; Huan, W.; Yuan, A. H.; Wu, J. H.; Hu, Y. Q. Perfluorocarbon Nanoparticles Enhance Reactive Oxygen Levels and Tumour Growth Inhibition in Photodynamic Therapy. Nat. Commun. 2015, 6, 8785. 34

ACS Paragon Plus Environment

Page 34 of 42

Page 35 of 42 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 Nano Materials

(14) Jia, H. R.; Jiang, Y. W.; Zhu, Y. X.; Li, Y. H.; Wang, H. Y.; Han, X. F.; Yu, Z. W.; Gu, N.; Liu, P. D.; Chen, Z.; Wu, F. G. Plasma Membrane Activatable Polymeric Nanotheranostics with Self-Enhanced Light-Triggered Photosensitizer Cellular Influx for Photodynamic Cancer Therapy. J. Control. Release 2017, 255, 231–241. (15) Xia, L. Y.; Zhang, X. D.; Cao, M.; Chen, Z.; Wu, F. G. Enhanced Fluorescence Emission and Singlet Oxygen Generation of Photosensitizers Embedded in Injectable Hydrogels for Imaging-Guided Photodynamic Cancer Therapy. Biomacromolecules 2017, 18, 3073– 3081. (16) Zhu, Y. X.; Jia, H. R.; Chen, Z.; Wu, F. G. Photosensitizer (PS)/Polyhedral Oligomeric Silsesquioxane (POSS)-Crosslinked Nanohybrids for Enhanced Imaging-Guided Photodynamic Cancer Therapy. Nanoscale 2017, 9, 12874−12884. (17) Zhang, X. D.; Xia, L. Y.; Chen, X. K.; Chen, Z.; Wu, F. G. Hydrogel-Based Phototherapy for Fighting Cancer and Bacterial Infection. Sci. China Mater. 2017, 60, 487−503. (18) Zou, Q. L.; Abbas, M.; Zhao, L. Y.; Li, S. K.; Shen, G. Z.; Yan, X. H. Biological Photothermal Nanodots Based on Self-Assembly of Peptide–Porphyrin Conjugates for Antitumor Therapy. J. Am. Chem. Soc. 2017, 139, 1921‒1927. (19) Pan, G. Y.; Jia, H. R.; Zhu, Y. X.; Wu, F. G. Turning Double Hydrophilic into Amphiphilic: IR825-Conjugated Polymeric Nanomicelles for Near-Infrared Fluorescence Imaging-Guided Photothermal Cancer Therapy. Nanoscale 2018, 10, 2115‒2127. (20) Ma, N. N.; Jiang, Y. W.; Zhang, X. D.; Wu, H.; Myers, J. N.; Liu, P. D.; Jin, H. Z.; Gu, N.; He, N. Y.; Wu, F. G.; Chen, Z. Enhanced Radiosensitization of Gold Nanospikes via Hyperthermia in Combined Cancer Radiation and Photothermal Therapy. ACS Appl. Mater. Interfaces 2016, 8, 28480‒28494. (21) Lin, H.; Gao, S. S.; Dai, C.; Chen, Y.; Shi, J. L. A Two-Dimensional Biodegradable Niobium Carbide (MXene) for Photothermal Tumor Eradication in NIR-I and NIR-II Biowindows. J. Am. Chem. Soc. 2017, 139, 16235−16247. (22) Manikandan, M.; Hasan, N.; Wu, H. F. Platinum Nanoparticles for the Photothermal Treatment of Neuro 2A Cancer Cells. Biomaterials 2013, 34, 5833–5842. (23) Zhao, R. F.; Han, X. X.; Li, Y. Y.; Wang, H.; Ji, T. J.; Zhao, Y. L.; Nie, G. J. Photothermal Effect Enhanced Cascade Targeting Strategy for Improved Pancreatic Cancer Therapy by Gold Nanoshell@Mesoporous Silica Nanorod. ACS Nano 2017, 11, 8103−8113. 35

ACS Paragon Plus Environment

ACS Applied Nano Materials 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 36 of 42

(24) Shi, S. G.; Huang, Y. Z.; Chen, X. L.; Weng, J.; Zheng, N. F. Optimization of Surface Coating on Small Pd Nanosheets for in Vivo Near-Infrared Photothermal Therapy of Tumor. ACS Appl. Mater. Interfaces 2015, 7, 14369−14375. (25) Dai, C.; Chen, Y.; Jing, X. X.; Xiang, L. H.; Yang, D. Y.; Lin, H.; Liu, Z.; Han, X. X.; Wu, R. Two-Dimensional Tantalum Carbide (MXenes) Composite Nanosheets for Multiple Imaging Guided Photothermal Tumor Ablation. ACS Nano 2017, 11, 12696−12712. (26) Cai, W.; Gao, H. Y.; Chu, C. C.; Wang, X. Y.; Wang, J. Q.; Zhang, P. F.; Lin, G.; Li, W. G.; Liu, G.; Chen, X. Y. Engineering Phototheranostic Nanoscale Metal−Organic Frameworks for Multimodal Imaging-Guided Cancer Therapy. ACS Appl. Mater. Interfaces 2017, 9, 2040–2051. (27) Kong, L. D.; Xing, L. X.; Zhou, B. Q.; Du, L. F.; Shi, X. Y. Dendrimer-Modified MoS2 Nanoflakes as a Platform for Combinational Gene Silencing and Photothermal Therapy of Tumors. ACS Appl. Mater. Interfaces 2017, 9, 15995–16005. (28) Ge, J. C.; Jia, Q. Y.; Liu, W. M.; Guo, L.; Liu, Q. Y.; Lan, M. H.; Zhang, H. Y.; Meng, X. M.; Wang, P. F. Red-Emissive Carbon Dots for Fluorescent, Photoacoustic, and Thermal Theranostics in Living Mice. Adv. Mater. 2015, 27, 4169–4177. (29) Li, S. L.; Chen, Y. H.; Liu, H. B.; Wang, Y. X.; Liu, L. B.; Lv, F. T.; Li, Y. L.; Wang, S. Graphdiyne Materials as Nanotransducer for in Vivo Photoacoustic Imaging and Photothermal Therapy of Tumor. Chem. Mater. 2017, 29, 6087–6094. (30) Zhao, Y. T.; Tong, L. P.; Li, Z. B.; Yang, N.; Fu, H. D.; Wu, L.; Cui, H. D.; Zhou, W. H.; Wang, J. H.; Wang, H. Y.; Chu, P. K.; Yu, X. F. Stable and Multifunctional Dye-Modified Black Phosphorus

Nanosheets

for

Near-Infrared

Imaging-Guided

Photothermal Therapy. Chem. Mater. 2017, 29, 7131–7139. (31) Shao, J. D.; Ruan, C. S.; Xie, H. H.; Li, Z. B.; Wang, H. Y.; Chu, P. K.; Yu, X. F. Black-Phosphorus-Incorporated

Hydrogel

as

a

Sprayable

and

Biodegradable

Photothermal Platform for Postsurgical Treatment of Cancer. Adv. Sci. 2018, 1700848. (32) Chen, W. S.; Ouyang, J.; Liu, H.; Chen, M.; Zeng, K.; Sheng, J. P.; Liu, Z. J.; Han, J. Y.; Wang, L. Q.; Li, J.; Deng, L.; Liu, Y. N.; Guo, S. J. Black Phosphorus Nanosheet-Based Drug Delivery System for Synergistic Photodynamic/Photothermal/Chemotherapy of Cancer. Adv. Mater. 2017, 29, 1603864. (33) Li, Y.; Liu, Z. M.; Hou, Y. Q.; Yang, G. C.; Fei, X. X.; Zhao, H.; Guo, Y. X.; Su, C. K.; Wang, Z.; Zhong, H. Q.; Zhuang, Z. F.; Guo, Z. Y. Multifunctional Nanoplatform Based 36

ACS Paragon Plus Environment

Page 37 of 42 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 Nano Materials

on Black Phosphorus Quantum Dots for Bioimaging and Photodynamic/Photothermal Synergistic Cancer Therapy. ACS Appl. Mater. Interfaces 2017, 9, 25098–25106. (34) Lyu, Y.; Zeng, J. F.; Jiang, Y. Y.; Zhen, X.; Wang, T.; Qiu, S. S.; Lou, X.; Gao, M. Y.; Pu, K. Y. Enhancing Both Biodegradability and Efficacy of Semiconducting Polymer Nanoparticles for Photoacoustic Imaging and Photothermal Therapy. ACS Nano 2018, 12, 1801–1810. (35) Jiang, Y. Y.; Li, J. C.; Zhen, X.; Xie, C.; Pu, K. Y. Dual-Peak Absorbing Semiconducting Copolymer Nanoparticles for First and Second Near-Infrared Window Photothermal Therapy: A Comparative Study. Adv. Mater. 2018, 1705980. (36) Zhang, S. B.; Guo, W. S.; Wei, J.; Li, C.; Liang, X. J.; Yin, M. Z. Terrylenediimide-Based Intrinsic Theranostic Nanomedicines with High Photothermal Conversion Efficiency for Photoacoustic Imaging-Guided Cancer Therapy. ACS Nano 2017, 11, 3797–3805. (37) Guo, B.; Sheng, Z. H.; Hu, D. H.; Li, A. R.; Xu, S. D.; Manghnani, P. N.; Liu, C. B.; Guo, L.; Zheng, H. R.; Liu, B. Molecular Engineering of Conjugated Polymers for Biocompatible Organic Nanoparticles with Highly Efficient Photoacoustic and Photothermal Performance in Cancer Theranostics. ACS Nano 2017, 11, 10124–10134. (38) Zhu, C. L.; Liu, L. B.; Yang, Q.; Lv, F. T.; Wang, S. Water-Soluble Conjugated Polymers for Imaging, Diagnosis, and Therapy. Chem. Rev. 2012, 112, 4687–4735. (39) Hu, P.; Han, L.; Dong, S. J. A Facile One-Pot Method to Synthesize a Polypyrrole/Hemin Nanocomposite and Its Application in Biosensor, Dye Removal, and Photothermal Therapy. ACS Appl. Mater. Interfaces 2014, 6, 500–506. (40) Cai, Y.; Liang, P. P.; Tang, Q. Y.; Yang, X. Y.; Si, W. L.; Huang, W.; Zhang, Q.; Dong, X. C. Diketopyrrolopyrrole–Triphenylamine Organic Nanoparticles as Multifunctional Reagents for Photoacoustic Imaging-Guided Photodynamic/Photothermal Synergistic Tumor Therapy. ACS Nano 2017, 11, 1054–1063. (41) MacDonald, T. D.; Liu, T. W.; Zheng, G. An MRI-Sensitive, Non-Photobleachable Porphysome Photothermal Agent. Angew. Chem., Int. Ed. 2014, 53, 6956–6959. (42) Lovell, J. F.; Jin, C. S.; Huynh, E.; Jin, H. L.; Kim, C.; Rubinstein, J. L.; Chan, W. C. W.; Cao, W. G.; Wang, L. V.; Zheng, G. Porphysome Nanovesicles Generated by Porphyrin Bilayers for Use as Multimodal Biophotonic Contrast Agents. Nat. Mater. 2011, 10, 324−332. 37

ACS Paragon Plus Environment

ACS Applied Nano Materials 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 38 of 42

(43) Lin, W. H.; Li, Y.; Zhang, W.; Liu, S.; Xie, Z. G.; Jing, X. B. Near-Infrared Polymeric Nanoparticles with High Content of Cyanine for Bimodal Imaging and Photothermal Therapy. ACS Appl. Mater. Interfaces 2016, 8, 24426–24432. (44) Wang, W. Q.; Wang, L.; Liu, S.; Xie, Z. G. Metal–Organic Frameworks@Polymer Composites Containing Cyanines for Near-Infrared Fluorescence Imaging and Photothermal Tumor Therapy. Bioconjugate Chem. 2017, 28, 2784−2793. (45) Tan, X.; Luo, S. L.; Long, L.; Wang, Y.; Wang, D. C.; Fang, S. T.; Ouyang, Q.; Su, Y. P.; Cheng, T. M.; Shi, C. M. Structure-Guided Design and Synthesis of a Mitochondria-Targeting Near-Infrared Fluorophore with Multimodal Therapeutic Activities. Adv. Mater. 2017, 29, 1704196. (46) Li, L.; Pang, X.; Liu, G. Near-Infrared Light-Triggered Polymeric Nanomicelles for Cancer

Therapy

and

Imaging.

ACS

Biomater.

Sci.

Eng.

2018,

DOI:

10.1021/acsbiomaterials.7b00648. (47) Luo, S. L.; Tan, X.; Qi, Q. R.; Guo, Q. Y.; Ran, X. Z.; Zhang, L. L.; Zhang, E. L.; Liang,Y. F.; Weng, L. L.; Zheng, H.; Cheng, T. M.; Su, Y. P.; Shi, C. M. A Multifunctional

Heptamethine

Near-Infrared

Dye

for

Cancer

Theranosis.

Biomaterials 2013, 34, 2244−2251. (48) Meng, X. Q.; Li, W. J.; Sun, Z. H.; Zhang, J. L.; Zhou, L. H.; Deng, G. J.; Gong, P.; Cai, L. T. Tumor-Targeted Small Molecule for Dual-Modal Imaging-Guided Phototherapy Upon Near-Infrared Excitation. J. Mater. Chem. B, 2017, 5, 9405−9411. (49) Zhao, P. F.; Zheng, M. B.; Yue, C. X.; Luo, Z. Y.; Gong, P.; Gao, G. H.; Sheng, Z. H.; Zheng, C. F.; Cai, L. T. Improving Drug Accumulation and Photothermal Efficacy in Tumor

Depending

on

Size

of

ICG

Loaded

Lipid-Polymer

Nanoparticles.

Biomaterials 2014, 35, 6037−6046. (50) Zhang, J. X.; Liu, S.; Hu, X. L.; Xie, Z. G.; Jing, X. B. Cyanine-Curcumin Assembling Nanoparticles for Near-Infrared Imaging and Photothermal Therapy. ACS Biomater. Sci. Eng. 2016, 2, 1942−1950. (51) Zhang, Y. Y.; Qu, Q. Y.; Cao, X.; Zhao, Y. L. NIR-Absorbing Dye Functionalized Hollow Mesoporous Silica Nanoparticles for Combined Photothermal–Chemotherapy. Chem. Commun. 2017, 53, 12032−12035. (52) Li, M. H.; Teh, C.; Ang, C. Y.; Tan, S. Y.; Luo, Z.; Qu, Q. Y.; Zhang, Y. Y.; Korzh, V.; Zhao, Y. L. Near-Infrared Light-Absorptive Stealth Liposomes for Localized Photothermal Ablation of Tumors Combined with Chemotherapy. Adv. Funct. Mater. 2015, 25, 5602–5610. 38

ACS Paragon Plus Environment

Page 39 of 42 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 Nano Materials

(53) Zhu, A. J.; Miao, K.; Deng, Y. B.; Ke, H. T.; He, H.; Yang, T.; Guo, M.; Li, Y. L.; Guo, Z. Q.; Wang, Y. Y.; Yang, X. L.; Zhao, Y. L.; Chen, H. B. Dually pH/Reduction-Responsive Vesicles

for

Ultrahigh-Contrast

Fluorescence

Imaging

and

Thermo-Chemotherapy-Synergized Tumor Ablation. ACS Nano 2015, 9, 7874–7885. (54) Wang, Y.; Yang, T.; Ke, H. T.; Zhu, A. J.; Wang, Y. Y.; Wang, J. X.; Shen, J. K.; Liu, G.; Chen, C. Y.; Zhao, Y. L.; Chen, H. B. Smart Albumin-Biomineralized Nanocomposites for Multimodal Imaging and Photothermal Tumor Ablation. Adv. Mater. 2015, 27, 3874– 3882. (55) Gao, C.; Liang, X. L.; Mo, S. Y.; Zhang, N. S.; Sun, D. S.; Dai, Z. F. Near-Infrared Cyanine-Loaded Liposome-like Nanocapsules of Camptothecin–Floxuridine Conjugate for Enhanced Chemophotothermal Combination Cancer Therapy. ACS Appl. Mater. Interfaces 2018, 10, 3219–3228. (56) Xu, Y. X.; Liang, X. L.; Bhattarai, P.; Sun, Y.; Zhou, Y. M.; Wang, S. M.; Chen, W.; Ge, H. Y.; Wang, J. R.; Cui, L. G.; Dai, Z. F. Enhancing Therapeutic Efficacy of Combined Cancer Phototherapy by Ultrasound-Mediated In Situ Conversion of Near-Infrared Cyanine/Porphyrin Microbubbles into Nanoparticles. Adv. Funct. Mater. 2017, 27, 1704096. (57) Pan, G. Y.; Jia, H. R.; Zhu, Y. X.; Wang, R. H.; Wu, F. G.; Chen, Z. Dual Channel Activatable Cyanine Dye for Mitochondrial Imaging and Mitochondria-Targeted Cancer Theranostics. ACS Biomater. Sci. Eng. 2017, 3, 3596−3606. (58) Chen, Q.; Wang, C.; Zhan, Z. X.; He, W. W.; Cheng, Z. P.; Li, Y. Y.; Liu, Z. Near-Infrared Dye Bound Albumin with Separated Imaging and Therapy Wavelength Channels for Imaging-Guided Photothermal Therapy. Biomaterials 2014, 35, 8206‒8214. (59) Zhang, Y. Y.; Teh, C.; Li, M. H.; Ang, C. Y.; Tan, S. Y.; Qu, Q. Y.; Korzh, V.; Zhao, Y. L. Acid-Responsive Polymeric Doxorubicin Prodrug Nanoparticles Encapsulating a Near-Infrared Dye for Combined Photothermal-Chemotherapy. Chem. Mater. 2016, 28, 7039−7050. (60) Zhang, Y. Y.; Ang, C. Y.; Li, M. H.; Tan, S. Y.; Qu, Q. Y.; Zhao, Y. L. Polymeric Prodrug Grafted Hollow Mesoporous Silica Nanoparticles Encapsulating Near-Infrared Absorbing Dye for Potent Combined Photothermal-Chemotherapy. ACS Appl. Mater. Interfaces 2016, 8, 6869−6879. (61) Huang, P.; Rong, P. F.; Jin, A.; Yan, X. F.; Zhang, M. G.; Lin, J.; Hu, H.; Wang, Z.; Yue, X. Y.; Li, W. W.; Niu, G. Zeng, W. B. Wang, W.; Zhou, K. C.; Chen, X. Y. Dye-Loaded Ferritin Nanocages for Multimodal Imaging and Photothermal Therapy. Adv. Mater. 2014, 26, 6401–6408. 39

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

(62) Ju, E. G.; Li, Z. H.; Liu, Z. Ren, J. S.; Qu, X. G. Near-Infrared Light-Triggered Drug-Delivery Vehicle for Mitochondria-Targeted Chemo-Photothermal Therapy. ACS Appl. Mater. Interfaces 2014, 6, 4364−4370. (63) Kumar, R.; Han, J. Y.; Lim, H. J.; Ren, W. X.; Lim, J. Y.; Kim, J. H.; Kim, J. S. Mitochondrial Induced and Self-Monitored Intrinsic Apoptosis by Antitumor Theranostic Prodrug: In Vivo Imaging and Precise Cancer Treatment. J. Am. Chem. Soc. 2014, 136, 17836−17843. (64) Jung, H. S.; Han, J. Y.; Lee, J. H.; Lee, J. H.; Choi, J. M.; Kweon, H. S.; Han, J. H.; Kim, J. H.; Byun, K. M.; Jung, J. H.; Kang, C.; Kim, J. S. Enhanced NIR Radiation-Triggered Hyperthermia by Mitochondrial Targeting. J. Am. Chem. Soc. 2015, 137, 3017‒3023. (65) Liu, L. M.; Yong, K. T.; Roy, I.; Law, W. C.; Ye, L.; Liu, J. W.; Liu, J.; Kumar, R.; Zhang, X. H.; Prasad, P. N. Bioconjugated Pluronic Triblock-Copolymer Micelle-Encapsulated Quantum Dots for Targeted Imaging of Cancer: In Vitro and In Vivo Studies. Theranostics 2012, 2, 705‒713. (66) Zhang, W.; Shi, Y.; Chen, Y. Z.; Ye, J.; Sha, X. Y.; Fang, X. L. Multifunctional Pluronic P123/F127 Mixed Polymeric Micelles Loaded with Paclitaxel for the Treatment of Multidrug Resistant Tumors. Biomaterials 2011, 32, 2894‒2906. (67) Chen, Y. J.; Li, Z. H.; Wang, H. B.; Wang, Y.; Han, H. J.; Jin, Q.; Ji, J. IR-780 Loaded Phospholipid Mimicking Homopolymeric Micelles for Near-IR Imaging and Photothermal Therapy of Pancreatic Cancer. ACS Appl. Mater. Interfaces 2016, 8, 6852‒ 6858. (68) Cheng, L.; He, W. W.; Gong, H.; Wang, C.; Chen, Q.; Cheng, Z. P.; Liu, Z. PEGylated Micelle Nanoparticles Encapsulating a Non-Fluorescent Near-Infrared Organic Dye as a Safe and Highly-Effective Photothermal Agent for in Vivo Cancer Therapy. Adv. Funct. Mater. 2013, 23, 5893‒5902. (69) Resch-Genger, U.; Grabolle, M.; Cavaliere-Jaricot, S.; Nitschke, R.; Nann, T. Quantum Dots versus Organic Dyes as Fluorescent Labels. Nat. Methods 2008, 5, 763−775. (70) Xia, T.; Kovochich, M.; Liong, M.; Zink, J. I.; Nel, A. E. Cationic Polystyrene Nanosphere Toxicity Depends on Cell-Specific Endocytic and Mitochondrial Injury Pathways. ACS Nano 2008, 2, 85‒96. (71) Gui, C.; Zhao, E. G.; Kwok, R. T. K.; Leung, A. C. S.; Lam, J. W. Y.; Jiang, M. J.; Deng, H. Q.; Cai, Y. J.; Zhang, W. J.; Su, H. F.; Tang, B. Z. AIE-Active Theranostic System: Selective Staining and Killing of Cancer Cells. Chem. Sci. 2017, 8, 1822‒1830. (72) Nani, R. R.; Kelley, J. A.; Ivanic, J.; Schnermann, M. J. Reactive Species Involved in the Regioselective Photooxidation of Heptamethine Cyanines. Chem. Sci. 2015, 6, 6556‒ 40

ACS Paragon Plus Environment

Page 40 of 42

Page 41 of 42 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 Nano Materials

6563. (73) Gorka, A. P.; Nani, R. R.; Zhu, J. J.; Mackem, S.; Schnermann, M. J. A Near-IR Uncaging Strategy Based on Cyanine Photochemistry. J. Am. Chem. Soc. 2014, 136, 14153‒14159. (74) Yuan, A. H.; Qiu, X. F.; Tang, X. L.; Liu, W.; Wu, J. H.; Hu, Y. Q. Self-Assembled PEG-IR-780-C13 Micelle as a Targeting, Safe and Highly-Effective Photothermal Agent for in Vivo Imaging and Cancer Therapy. Biomaterials 2015, 51, 184‒193.

41

ACS Paragon Plus Environment

ACS Applied Nano Materials 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

TOC Graphic

42

ACS Paragon Plus Environment

Page 42 of 42