nanomaterials Article

Near-Infrared-Triggered Photodynamic Therapy toward Breast Cancer Cells Using Dendrimer-Functionalized Upconversion Nanoparticles Bing-Yen Wang 1,2,3,4 , Ming-Liang Liao 5 , Guan-Ci Hong 6 , Wen-Wei Chang 6, *,† and Chih-Chien Chu 5,7, *,† ID 1 2 3 4 5 6 7

* †

ID

Division of Thoracic Surgery, Department of Surgery, Changhua Christian Hospital, Changhua County 50006, Taiwan; [email protected] School of Medicine, Chung Shan Medical University, Taichung City 40201, Taiwan Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung City 40227, Taiwan School of Medicine, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan Department of Medical Applied Chemistry, Chung Shan Medical University, Taichung City 40201, Taiwan; [email protected] Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 40201, Taiwan; [email protected] Department of Medical Education, Chung Shan Medical University Hospital, Taichung City 40201, Taiwan Correspondence: [email protected] (W.-W.C.); [email protected] (C.-C.C.); Tel.: +886-4-2473-0022 (ext. 12317) (W.-W.C.); +886-4-2473-0022 (ext. 12227) (C.-C.C.) These authors contributed equally to this work.

Received: 3 August 2017; Accepted: 7 September 2017; Published: 11 September 2017

Abstract: Water-soluble upconversion nanoparticles (UCNPs) that exhibit significant ultraviolet, blue, and red emissions under 980-nm laser excitation were successfully synthesized for performing near infrared (NIR)-triggered photodynamic therapy (PDT). The lanthanide-doped UCNPs bearing oleate ligands were first exchanged by citrates to generate polyanionic surfaces and then sequentially encapsulated with NH2 -terminated poly(amido amine) (PAMAM) dendrimers (G4) and chlorine6 (Ce6) using a layer-by-layer (LBL) absorption strategy. Transmission electron microscopy and X-ray diffraction analysis confirm that the hybrid UCNPs possess a polygonal morphology with an average dimension of 16.0 ± 2.1 nm and α-phase crystallinity. A simple calculation derived through thermogravimetric analysis revealed that one polycationic G4 dendrimer could be firmly accommodated by approximately 150 polyanionic citrates through multivalent interactions. Moreover, zeta potential measurements indicated that the LBL fabrication results in the hybrid nanoparticles with positively charged surfaces originated from these dendrimers, which assist the cellular uptake in biological specimens. The cytotoxic singlet oxygen based on the photosensitization of the adsorbed Ce6 through the upconversion emissions can be readily accumulated by increasing the irradiation time of the incident lasers. Compared with that of 660-nm lasers, NIR-laser excitation exhibits optimized in vitro PDT effects toward human breast cancer MCF-7 cells cultured in the tumorspheres, and less than 40% of cells survived under a low Ce6 dosage of 2.5 × 10−7 M. Fluorescence microscopy analysis indicated that the NIR-driven PDT causes more effective destruction of the cells located inside spheres that exhibit significant cancer stem cell or progenitor cell properties. Moreover, an in vivo assessment based on immunohistochemical analysis for a 4T1 tumor-bearing mouse model confirmed the effective inhibition of cancer cell proliferation through cellular DNA damage by the expression of Ki67 and γH2AXser139 protein markers. Thus, the hybrid UCNPs are a promising NIR-triggered PDT module for cancer treatment.

Nanomaterials 2017, 7, 269; doi:10.3390/nano7090269

www.mdpi.com/journal/nanomaterials

Nanomaterials 2017, 7, 269

2 of 18

Keywords: upconversion; dendrimers; photodynamic therapy; layer-by-layer; cancer stem cells; near-infrared; tumorspheres

1. Introduction Recently, the biomedical applications of lanthanide-doped upconversion nanoparticles (UCNPs) have attracted much attention [1–6]. Because UCNP-based materials can sequentially absorb two or more photons, they exhibit a unique anti-Stokes shift of fluorescence emission in ultraviolet (UV)-visible wavelengths (300–700 nm) under NIR light excitation (750–1400 nm). The unique “photon upconversion” process has several merits, including reduced fluorescence background, improved tissue penetration, and lower phototoxicity in biological fields [7]. Moreover, UCNPs usually exhibit multiple fluorescence emission bands of UV, blue-green, and red light simultaneously under NIR excitation. Additionally, finely manipulating the doped-metal composition or the host crystal structure can lead to specific upconverted emissive patterns. For example, the nanoparticles doped with erbium (Er3+ ) instead of thulium (Tm3+ ) can switch emission wavelengths from blue to green light as well as enhance red-light intensity; a single band, intense red-light emission is achieved by incorporating manganese (Mn2+ ), ytterbium (Yb3+ ), and Er3+ into the nanostructure [8–10]. Combined with the robust, well-developed synthetic protocols, UCNPs bearing specific upconverted emission profiles are promising for photo-assisted cancer treatments, particularly photodynamic therapy (PDT) [11–14]. PDT based on the photochemical reactions of photosensitizers (PSs) is a treatment strategy involving light irradiation at appropriate wavelengths, by which PS molecules can produce cytotoxic reactive oxygen species (ROS) and singlet oxygen (1 O2 ) to destroy nearby cancer cells [15]. The greatest advantage of PDT is the ability to selectively treat tumor cells by using light under spatiotemporal control. However, most PS molecules are excited by visible or UV light, which have poor tissue penetration, thus limiting the treatment of large or internal tumors. Loading the PS molecules onto the UCNPs surface avoids this drawback, as it facilitates NIR-triggered PDT, which is based on upconverted fluorescence emission and resonance energy transfer. Several strategies have been developed to immobilize the PS molecules on the surface including mesoporous silica coating and polymer encapsulation [9,16–19]. Intense upconverted emission at suitable wavelengths and higher loading capacity of the active molecules was proposed for guaranteeing more effective therapeutic outcomes. Liu and coworkers demonstrated a layer-by-layer (LBL) method for loading multiple layers of PS-conjugated polymers onto the surface of UCNPs surface through electrostatic interaction [13]. Both in vitro and in vivo PDT effects were substantially improved through their multiloading strategy. As clinical PDT develops, fabricating PS-conjugated-UCNP systems with commercialization potential by using more facile and robust synthetic protocols remains challenging. This paper introduces the application of dendrimers in the encapsulation layers of UCNPs for trapping PS molecules according to the multivalency of the dendritic structures [20]. Dendrimers, well-defined hyperbranched polymers consisting of a central core, radiating branches ending in multivalent terminal groups, and cavities within the branched structure have aroused much interest in the past two decades [21–24]. For example, full-generation poly(amido amine) (PAMAM) dendrimers bear multiple primary amines on the surface and internal tertiary amines as branching sites. These tertiary amines can be protonated to generate polycations at physiological pH, thus allowing for electrostatic interaction and subsequent formation of complexes with molecules bearing opposite charges [25–28]. Apart from electrostatic binding, the internal cavities within the dendrimers can accommodate small molecules through host-guest affinity. Accordingly, typical fourth-generation PAMAM dendrimers (G4), bearing 64 peripheral amines, were immobilized on the surface by using the facile LBL strategy to create positively charged UCNPs. The material design enhanced the cellular uptake of the nanoparticles as well as the absorption of negatively charged PS molecules by those

Nanomaterials 2017, 7, 269

3 of 18

dendrimers (Scheme 1). A 980-nm NIR laser diode (LD) was used as the light excitation source to analyze upconversion and PDT effect. To elucidate the NIR-triggered PDT efficacy, we explore the 1 O2 production combined with the apoptosis of breast cancer cells cultured in two-dimensional (2D) arrays Nanomaterials 2017, 7, 269 3 of 18 and three-dimensional (3D) spheres and with the cellular DNA damage of tumor tissues [29,30].

Scheme 1. Near-infrared (NIR)-light-triggered therapy based a lanthanide Scheme 1. Near-infrared (NIR)-light-triggered photodynamic photodynamic therapy based on aon lanthanide (Ln)- (Ln)doped NaYF upconversion nanoparticle (UCNP) sequentially encapsulated with citrates, PAMAM doped NaYF4 upconversion nanoparticle (UCNP) sequentially encapsulated with citrates, PAMAM 4 dendrimers, and chlorin e6 (Ce6) viaa alayer-by-layer layer-by-layer (LBL) absorption method. dendrimers, and chlorin e6 (Ce6) via (LBL) absorption method. 2. Results and Discussion 2. Results and Discussion 2.1. Preparation of Lanthanide-Doped UCNPs

2.1. Preparation of Lanthanide-Doped UCNPs 3+ 3+

Yb - and Tm -doped NaYF4 nanocrystals were used as core materials in the NIR-induced because their high efficacy in NIR-to-visible Herein, chlorin e6 (Ce6), PDT Yb - and Tm3+of-doped NaYF 4 nanocrystals were light usedupconversions. as core materials in the NIR-induced a chlorophyll analog with significant absorption, maximum at approximately 400 nm (Soret band) because of their high efficacy in NIR-to-visible light upconversions. Herein, chlorin e6 (Ce6), a and 660 nm (Q-band), was used as the PS in the PDT investigation at the cellular level [31,32]. chlorophyll analog with significant absorption, maximum at approximately 400 nm (Soret band) and The UCNPs were synthesized using a solvothermal procedure at 300 ◦ C with oleic acid (OA) as 660 nm (Q-band), was used as the PS in the PDT investigation at the cellular level [31,32]. The UCNPs the stabilizer [33]. To enhance the energy transfer between the doped ions and host materials, were synthesized using a solvothermal procedure at 300 °C with oleic acid (OA) as the stabilizer [33]. the core of the UCNPs was further covered by a layer of undoped NaYF4 to form a core-shell [34]. To enhance the energy between the doped ions and host materials, core of(XRD) the UCNPs The crystallinity andtransfer composition for the UCNPs were characterized using X-raythe diffraction was further covered by adispersive layer of X-ray undoped NaYF4 (EDXS), to formsuggesting a core-shell [34].(cubic-like) The crystallinity and equipped with energy spectroscopy α-phase NaYF4 3+ 3+ with successful Yb andwere Tm characterized doping (Supplementary Materials, Figures S1 and S2). As shown composition for the UCNPs using X-ray diffraction (XRD) equipped with energy in Figure 1a, the images of the core-shell nanocrystals, taken using TEM, display a polygonal dispersive X-ray spectroscopy (EDXS), suggesting α-phase (cubic-like) NaYF4 with successful Yb3+ morphology an average dimension of 16.0 ± 2.1 S1 nm.and Moreover, 3+ doping with and Tm (Supplementary Materials, Figures S2). Asa cubic-like shown innanocrystal Figure 1a,larger the images than 50 nm was clearly observed in certain grid areas, which is consistent with the XRD data (Figure 1b). of the core-shell nanocrystals, taken using TEM, display a polygonal morphology with an average As shown in Figure 2c, the prepared UCNPs emitted upconverted fluorescence at multiple bands of UV dimension 16.0nm), ± 2.1blue nm.(λMoreover, a cubic-like nanocrystal larger than 50 nm was clearly observed (λmax of = 360 max = 480 nm), red (λmax = 650 nm), and NIR (λmax = 795 nm) wavelengths in certain grid is consistent with the XRDintensity data (Figure 1b). As shown in Figure under 980areas, nm LDwhich excitation. Notably, the fluorescence was considerably increased by the 2c, the prepared UCNPs emitted upconverted fluorescence multiple (λmaxthe = Ce6 360 that nm), blue core-shell strategy. Among the bands, the UV and red at emissions arebands capableofofUV exciting absorption, NIR emissions allow tracing the (λmax =possesses 480 nm),corresponding red (λmax = 650 nm), andand NIRmore (λmaxintense = 795 blue nm)and wavelengths under 980fornm LD excitation. biodistribution of the UCNPs through fluorescence microscopy. Notably, the fluorescence intensity was considerably increased by the core-shell strategy. Among the 3+ PDT

bands, the UV and red emissions are capable of exciting the Ce6 that possesses corresponding absorption, and more intense blue and NIR emissions allow for tracing the biodistribution of the UCNPs through fluorescence microscopy.

Nanomaterials 2017, 7, 269

4 of 18

Nanomaterials 2017, 7, 269

4 of 18

(a)

(b)

(c)

(d)

Figure 1. Transmittance electron microscopic (TEM) images of: (a) polygonal (small); and (b) cubic-

Figure 1. Transmittance electron microscopic (TEM) images of: (a) polygonal (small); and (b) cubic-like like (large) NaYF4 nanocrystals. The average dimension for (a) was calculated based on 100 (large) NaYF Thea average dimension for (a) was calculated based nanoparticles 4 nanocrystals. nanoparticles selected from TEM micrograph. Upconversion fluorescence images (c)on and100 spectra (d) selectedoffrom a TEM micrograph. Upconversion fluorescence images (c) and (d) of the: the: (i) core; and (ii) core-shell nanocrystals, under 980-nm laser excitation. Thespectra spectra recorded by (i) core; a CCD imaging spectrometer exhibits UV, blue, red, and NIR wavelengths and (ii) core-shell nanocrystals, under 980-nm laser excitation. The spectrasimultaneously. recorded by a CCD imaging spectrometer exhibits UV, blue, red, and NIR wavelengths simultaneously. 2.2. Fabrication of UCNPs through Ligand Exchange and the LBL Strategy

Synthetic strategies involving encapsulation of mesoporous 2.2. Fabrication of UCNPs through Ligand the Exchange and the LBL Strategy silica and water-soluble

polymers have been developed for preparing the hydrophilic UCNPs used in biomedicinal

Synthetic strategies involving encapsulation mesoporous silica water-soluble polymers applications. To fabricate the the nanoparticle surfaceofwith polycationic G4 and in this hybrid system, negatively chargedfor UCNPs are produced and, the dendrimers areused electrostatically adsorbed applications. using have been developed preparing the hydrophilic UCNPs in biomedicinal the LBL [13]. Thesurface first attempt using an ozonolysis to cleave the double bonds ofcharged To fabricate themethod nanoparticle withofpolycationic G4 inreaction this hybrid system, negatively the oleate ligands for generating anionic carboxylate groups was unsuccessful, because the oxidative UCNPs are produced and, the dendrimers are electrostatically adsorbed using the LBL method [13]. treatment under acidic condition led to the unexpected desorption of the surface ligands [35]. The The firstremoval attempt using an ozonolysis reaction to cleave the double the oleate ligands of ofthe ligands was confirmed by thermogravimetric analysisbonds (TGA)ofmeasurement for generating anionicMaterials, carboxylate groups was unsuccessful, because thewere oxidative treatment under (Supplementary Figure S3) [12]. Alternatively, the oleate ligands readily exchanged by a trivalent sodium citrate in diethylene glycol under ligands 200 °C, thus for the of the acidic condition led ligand to theofunexpected desorption of the surface [35].allowing The removal of theby UCNPs with multiple analysis carboxylate groups as the first layer [36]. The NHMaterials, 2ligands fabrication was confirmed thermogravimetric (TGA) measurement (Supplementary determinated G4 dendrimers on the UCNPs were then immobilized through electrostatic interaction Figure S3) [12]. Alternatively, the oleate ligands were readily exchanged by a trivalent ligand of at ambient temperature, thus generating water-dispersible nanoparticles with excess positive charges sodium citrate in diethylene glycol under 200 ◦ C, thus allowing for the fabrication of the UCNPs on the surface and forming the second layer. After the exchange with the citrate ligands, the zeta with multiple carboxylate groups asrevealed the first layer [36]. Thesurface NH2 -determinated G4 dendrimers potentials of the modified UCNPs a negatively charged (ζ = −8.54 mV), however, a on the UCNPs were then immobilized through interaction at ambient temperature, positive charge (ζ = +31.7 mV) presented afterelectrostatic the dendrimer encapsulation (Supplementary Materials, Figure S4). Moreover, FT-IR analysis confirmed the exchanged ligands the thus generating water-dispersible nanoparticles with excess positivecitrate charges onthrough the surface and −1and 1400 cm−1, and the featured amide stretching of characteristic carboxylate stretching at 1586 cm forming the second layer. After the exchange with the citrate ligands, the zeta potentials of the the PAMAM dendrimers adsorbed on the surface was well characterized at 1640 cm−1and 1560 cm−1 modified UCNPs revealed a negatively charged surface (ζ = −8.54 mV), however, a positive charge (Supplementary Materials, Figure S5). As illustrated by the TEM images shown in Figure 2a,b, the (ζ = +31.7 mV) presented after theofdendrimer (Supplementary Figure S4). morphology and dimensions the UCNPs encapsulation were mostly unaltered after citrate Materials, and dendrimer Moreover, FT-IR analysis confirmed the exchanged citrate ligands through the characteristic carboxylate stretching at 1586 cm−1 and 1400 cm−1 , and the featured amide stretching of the PAMAM dendrimers adsorbed on the surface was well characterized at 1640 cm−1 and 1560 cm−1 (Supplementary Materials, Figure S5). As illustrated by the TEM images shown in Figure 2a,b, the morphology and dimensions of the UCNPs were mostly unaltered after citrate and dendrimer modification, but the boundary of each dendrimer-encapsulated nanoparticle was less defined. This observation was presumably because

Nanomaterials 2017, 7, 269

5 of 18

Nanomaterials 2017, 7, 269

5 of 18

modification, but the boundary of each dendrimer-encapsulated nanoparticle was less defined. This observation was presumably because of the strengthened interactions between the nanoparticles of the strengthened interactions includingby thethe hydrogen bonding and van including the hydrogen bondingbetween and vanthe dernanoparticles Waals force produced G4 PAMAM dendrimers. der Waals force produced by the G4 PAMAM dendrimers. Moreover, the hydrodynamic dimension Moreover, the hydrodynamic dimension of the G4-functionalized UCNPs determined by dynamic of thescattering G4-functionalized UCNPs180 determined dynamic light scattering is approximately 180 nm light is approximately nm with aby narrow distribution, suggesting that these dendrimers with narrow distribution, suggesting that these were successfullyMaterials, immobilized on S6). the wereasuccessfully immobilized on the surfaces of dendrimers the UCNPs (Supplementary Figure surfaces of the UCNPs (Supplementary Materials, Figure S6). Although the dendrimer interaction may Although dendrimer interaction may have led to substantial aggregation in these waterhave led to substantial in these water-dispersible upconversion emission still dispersible UCNPs, theaggregation upconversion emission still exhibitedUCNPs, distinctthe fluorescence bands in the UVexhibited distinct fluorescence bands in the UV-, blue-, and red-light regions (Figure 2c). , blue-, and red-light regions (Figure 2c).

(a)

(b)

(c) Figure 2.2.Transmittance Transmittance electron microscopic (TEM) of: (a) and citrate; and (b)dendrimerPAMAM Figure electron microscopic (TEM) imagesimages of: (a) citrate; (b) PAMAM dendrimer-encapsulated nanoparticles (c) Upconversion in encapsulated nanoparticles (G4-UCNP).(G4-UCNP). (c) Upconversion fluorescencefluorescence of G4-UCNPof in G4-UCNP water under water under 980-nm laser excitation. The fluorescence intensity was extinguished after encapsulation 980-nm laser excitation. The fluorescence intensity was extinguished after encapsulation of Ce6 of Ce6 (Ce6-G4-UCNP). (Ce6-G4-UCNP).

The organic organiccontents contents modified UCNPs were calculated using the weight-loss data The of of thethe modified UCNPs were calculated using the weight-loss data obtained obtained TGA, through and approximately 15%ofand the citrate oleate ligands, and citrate ligands, through and TGA, approximately 15% and 25% the 25% oleateof and respectively, respectively, were on immobilized the (Supplementary UCNP surface (Supplementary Figure that: S3). were immobilized the UCNP on surface Materials, FigureMaterials, S3). Assuming Assuming that: (1) every UCNP exhibited spherical shape uniform size upon TEM analysis; (1) every UCNP exhibited a spherical shapea and uniform sizeand upon TEM analysis; and (2) the mass 3), the ligand 3 and (2) the mass of a nanoparticle is calculated from the bulk density of NaYF 4 (4.21 g/cm of a nanoparticle is calculated from the bulk density of NaYF4 (4.21 g/cm ), the ligand density per density can per be particle can beusing determined using the simple calculation proposed Winnik [37]. and particle determined the simple calculation proposed by Winnik and by coworkers 2, coworkers [37].densities The coverage densities of oleate- and citrate-modified UCNPs, were 4.8 and 11 The coverage of oleateand citrate-modified UCNPs, were 4.8 and 11 ligands/nm ligands/nm2, thus, respectively; thus, approximately 4000 oleate andmolecules 9000 citrate molecules each respectively; approximately 4000 oleate and 9000 citrate covered each covered nanoparticle. nanoparticle. by finely adjusting the feeding of concentration of UCNPs, dendrimers to UCNPs, Moreover, by Moreover, finely adjusting the feeding concentration dendrimers to approximately approximately dendrimers delivered to one citrate-modified Basedand on Winnik and 60 dendrimers 60 were deliveredwere to one citrate-modified UCNP. BasedUCNP. on Winnik coworkers’ coworkers’ calculation, one polycationic G4 dendrimer was theoretically accommodated by 150 calculation, one polycationic G4 dendrimer was theoretically accommodated by 150 polyanioinic

Nanomaterials 2017, 7, 269

6 of 18

citrate molecules. Accordingly, the dendrimers could be firmly immobilized on the UCNP surface Nanomaterials 2017, 7, interactions. 269 6 of 18 through multivalent The negatively charged Ce6 molecules were then directly loaded onto the dendrimer-encapsulated polyanioinic citrate molecules. Accordingly, the dendrimers could be firmly immobilized on the UCNPs to form the third layer through the electrostatic binding of the COOH and NH2 groups. UCNP surface through multivalent interactions. The zeta The potential of the hybrid UCNPs decreased from +31.7 mV to +24.0 mV, suggesting that negatively charged Ce6 molecules were then directly loaded onto the dendrimernegatively charged Ce6tomolecules covered the surface of the nanoparticles and thus partially encapsulated UCNPs form the third layer through the electrostatic binding of the COOH and NH 2 neutralized the cationic density produced by the dendrimers (Supplementary Materials, Figure groups. The zeta potential of the hybrid UCNPs decreased from +31.7 mV to +24.0 mV, suggesting S4). Moreover, as showncharged in Figure the UV-Vis spectrum alsoofconfirmed the Ce6and adsorption through that negatively Ce63a, molecules covered the surface the nanoparticles thus partially neutralized the cationic produced by the dendrimers Materials, Figure Notably, S4). the characteristic Soret anddensity Q-band absorption bands at 407 (Supplementary nm and 643 nm, respectively. Moreover, as shown Figure exhibited 3a, the UV-Vis spectrum also confirmed the Ce6 adsorption the Q-band of the hybridinUCNPs substantial blue-shifted wavelength comparedthrough with that of the characteristic Soretthat andthe Q-band absorptionwere bands at 407 nmby andthe 643surface nm, respectively. Notably, free Ce6, which suggests PS molecules entrapped dendrimers. Generally, the Q-band of the hybrid UCNPs exhibited substantial blue-shifted wavelength compared with that the interior cavities of PAMAM dendrimers possess a hydrophobic domain. Once chromophores or of free Ce6, which suggests that the PS molecules were entrapped by the surface dendrimers. fluorophores are trapped inside the dendritic cavities, their absorption or emission wavelength shifts Generally, the interior cavities of PAMAM dendrimers possess a hydrophobic domain. Once hypsochromically in contrast to the free probe molecule in aqueous solution [38]. Therefore, the red chromophores or fluorophores are trapped inside the dendritic cavities, their absorption or emission fluorescence emission of the water-immersed hybrid under the excitation the Soret wavelength shifts hypsochromically in contrast to theUCNPs free probe molecule in aqueousof solution [38].band was also blue shifted by approximately 3b). In addition, the loading of the Therefore, the red fluorescence emission20ofnm the (Figure water-immersed hybrid UCNPs under thecapacity excitation Ce6 molecules be readily determined based on the 20 fluorescence intensity of thethe non-adsorbed of the Soretcould band was also blue shifted by approximately nm (Figure 3b). In addition, loading capacity of theoff Ce6the molecules be readily based onincreased the fluorescence intensity of feeding the Ce6 that washed hybridcould UCNPs. Thedetermined loading amount with the initial non-adsorbed Ce6 that washed off the hybrid UCNPs. The loading amount increased with the initial concentration of the Ce6. This was due to the multivalent character of the dendrimers, assisting the feedingofconcentration of the Ce6. This due and to the multivalent character of the dendrimers, and adsorption the PS molecules through the was exterior interior interactions caused by electrostatic assisting the adsorption of the PS molecules through the exterior and interior interactions caused by host-guest affinity, respectively. Figure 3b also shows a blue-to-green fluorescence band (430–580 nm), electrostatic and host-guest affinity, respectively. Figure 3b also shows a blue-to-green fluorescence which is presumably attributable to the intrinsic emission of the G4 dendrimer [28,39,40]. Notably, band (430–580 nm), which is presumably attributable to the intrinsic emission of the G4 dendrimer the fluorescence intensity of the G4 decreased with increasing Ce6 content. This result implies a strong [28,39,40]. Notably, the fluorescence intensity of the G4 decreased with increasing Ce6 content. This interaction dendrimer andbetween the entrapped Ce6, because theentrapped emission of thebecause dendrimer result between implies athe strong interaction the dendrimer and the Ce6, the and the absorption Ce6 occupied regions, thus facilitating fluorescence emission ofof thethe dendrimer and the proximal absorption wavelength of the Ce6 occupied proximal wavelength the regions, thus energy transfer. Moreover, this result clearly indicates that the PSresult molecules were firmly that immobilized facilitating the fluorescence energy transfer. Moreover, this clearly indicates the PS by molecules were firmlyon immobilized byof thethe PAMAM dendrimers the surfaces of the UCNPs.fluorescence Figure the PAMAM dendrimers the surfaces UCNPs. Figure 2con depicts the upconversion 2c of depicts the upconversion fluorescence of the aqueous The hybrid UCNPswavelengths under NIR-laser spectra the aqueous hybrid UCNPs under spectra NIR-laser excitation. emission centered excitation. The emission wavelengths centered at the UV-, blue-, and red-light regions were unaltered at the UV-, blue-, and red-light regions were unaltered after the surface modification. However, after the surface modification. However, the visible-light emissions of the UCNPs were extinguished the visible-light emissions of the UCNPs were extinguished by the peripheral Ce6, principally through by the peripheral Ce6, principally through the Soret and Q-band absorptions, a manifestation of the Soret and Q-band absorptions, a manifestation of effective energy transfer from the UCNPs to the effective energy transfer from the UCNPs to the surrounding PS molecules [41–43]. Notably, even the surrounding PS molecules Notably, the weak absorption band of the Ce6 (Figure 3a) weak absorption band of[41–43]. the Ce6 (Figure 3a) even at approximately 480–520 nm demonstrated substantial at approximately 480–520 nm demonstrated substantial absorption of the blue fluorescence emitted absorption of the blue fluorescence emitted from the UCNPs. Finally, the PS molecules were from successfully the UCNPs.photoexcited Finally, theby PSthe molecules were successfully photoexcited by the 980-nm NIR 980-nm NIR laser, using the dendrimer-encapsulated UCNPs as thelaser, usingnanoplatform. the dendrimer-encapsulated UCNPs as the nanoplatform.

(a) Figure 3. Cont.

Nanomaterials 2017, 7, 269

7 of 18

Nanomaterials 2017, 7, 269

7 of 18

Nanomaterials 2017, 7, 269

7 of 18

(b) Figure 3. (a) UV-Vis absorption; and (b) fluorescence spectra of the hybrid nanoparticles sequentially

Figure 3. (a) UV-Vis absorption; and (b) fluorescence spectra of the hybrid nanoparticles sequentially encapsulated with dendrimers (G4-UCNP) and Ce6 (Ce6-G4-UCNP) in comparison with that of encapsulated with dendrimers (G4-UCNP) and Ce6 (Ce6-G4-UCNP) in comparison with that of pristine pristine Ce6; (I) and (II) denote different loading capacity of Ce6. Ce6; (I) and (II) denote different loading capacity (b) of Ce6. 1O2) Formation 2.3. The Figure Evaluation of Photoactivity and Oxygen (spectra 3. (a) UV-Vis absorption; andSinglet (b) fluorescence of the hybrid nanoparticles sequentially

1 O ) Formation 2.3. The Evaluation of Photoactivity and(G4-UCNP) Singlet Oxygen 2 encapsulated with dendrimers and Ce6( (Ce6-G4-UCNP) in 1

comparisonCypridina with that of To assess the capability of the hybrid UCNPs to generate O2, a fluoresceinyl luciferin pristine Ce6; (I) and (II) denote different loading capacity of Ce6. 1 1O2 analog (FCLA) was employed a chemiluminescence probe [14]. FCLA was oxidized by luciferin To assess the capability of theashybrid UCNPs to generate O2 , The a fluoresceinyl Cypridina and thus markedly enhanced fluorescence by 524 nm. Figure 4a,b illustrates the correlation of analog (FCLA) was employed as a chemiluminescence [14]. The FCLA was oxidizedthe by 1 O2 2.3. The Evaluation of Photoactivity and Singlet Oxygen (1O2) probe Formation laser irradiation time and fluorescence intensities of FCLA in the presence of PSs. The Ce6 and hybrid and thus markedly enhanced fluorescence by 524 nm. Figure 4a,b illustrates the correlation of the assess the fluorescence capability of the hybrid UCNPs to generate 1O2, 2.3-fold a fluoresceinyl Cypridinaincrements, luciferin UCNPsToexhibited intensities of approximately and 3.5-fold laser irradiation timewas and fluorescence intensities of FCLA in the of PSs. The and analog (FCLA) employed as660-nm a chemiluminescence probe Thepresence FCLA wasthe oxidized by 1OCe6 2 respectively, and plateaued upon laser irradiation for [14]. 16 min, by which Ce6 molecules hybrid UCNPs exhibited fluorescence intensities ofnm. approximately 2.3-foldthe and 3.5-foldofincrements, and thus markedly enhanced fluorescence by 524 Figure 4a,b illustrates correlation the were directly photoexcited through the Q-band absorption. The FCLA assay could detect the laser irradiation time and fluorescence intensities of FCLAfor in the of PSs. The Ce6 and hybrid respectively, and plateaued upon laser irradiation 16presence min, Ce6 molecules accumulation of the 1O2 in water660-nm by increasing the exposure time of theby redwhich light. the Notably, the hybrid were UCNPs exhibited fluorescence intensities of approximately 2.3-fold and 3.5-fold increments, directly photoexcited throughtothe absorption. The FCLA assay could detect the accumulation UCNPs were also sensitive theQ-band irradiation using a 980-nm NIR laser, displaying a two-fold increase respectively, and plateaued upon 660-nm laser irradiation for 16 min, by which the Ce6 molecules of thein1 O in water by increasing the exposure time of the red light. Notably, the hybrid UCNPs fluorescence; whereas, the Ce6 was completely unreactive to the NIR light. This result clearly were 2 were directly photoexcited through the Q-band absorption. The FCLA assay could detect the also sensitive to the irradiation using a 980-nm laser, displaying a two-foldby increase in fluorescence; confirmed that the Ce6 immobilized by theNIR dendrimers was photoexcited the upconverted accumulation of the 1O2 in water by increasing the exposure time of the red light. Notably, the hybrid fluorescence emitted from the nanoplatform. Moreover, as shown Figure 4c, the UCNPs whereas, the Ce6 was unreactive to thea 980-nm NIR light. This resultinclearly confirmed that the UCNPs were alsocompletely sensitive to UCNPs the irradiation using NIR laser, displaying a two-fold increase 1O2 under 980-nm laser irradiation, attributed to with higher by loading of Ce6the generated substantial Ce6 immobilized the dendrimers by the upconverted fluorescence emitted from in afluorescence; whereas, Ce6was wasphotoexcited completely unreactive to the NIR light. This result clearly theconfirmed merits of that usingthetheCe6 polyvalent dendrimers as adsorption layer to accommodate more PS immobilized the dendrimers was photoexcited by thea upconverted the UCNPs nanoplatform. Moreover, asby shown in the Figure 4c, the UCNPs with higher loading of molecules through both exterior and nanoplatform. interior interactions. Considering all the 4c, assay data, the 1 fluorescence emitted from the UCNPs Moreover, as shown in Figure the UCNPs Ce6 generated substantial O2 under 980-nm laser irradiation, attributed to the merits of using the 1O2 under photoactivation permitted energy transfer between the excited surrounding oxygen with a higher loading of Ce6 generated substantial 980-nmCe6 laserand irradiation, attributed to polyvalent dendrimers as the adsorption layer to accommodate more PS molecules through both 1 molecules, generating O2 as the majoras ROS NIR-light-triggered PDT for solid tumors. the merits of using significant the polyvalent dendrimers theinadsorption layer to accommodate more PS

exteriormolecules and interior interactions. Considering all the assay data, the photoactivation through both exterior and interior interactions. Considering all the assaypermitted data, the energy 1 O as the transferphotoactivation between the excited Ce6 and surrounding oxygen molecules, generating significant 2 permitted energy transfer between the excited Ce6 and surrounding oxygen 1O2 for molecules, generating significant as the major ROS in NIR-light-triggered PDT for solid tumors. major ROS in NIR-light-triggered PDT solid tumors.

(a) (a)

Figure 4. Cont.

Nanomaterials 2017, 7, 269

8 of 18

Nanomaterials 2017, 7, 269

8 of 18

(b)

(c) Figure 4. The FCLA assay for determining the accumulation of singlet oxygen using: (a) pristine Ce6;

Figure 4. The FCLA assay for determining the accumulation of singlet oxygen using: (a) pristine Ce6; and (b) hybrid nanoparticles (Ce6-G4-UCNP) as the photosensitizer: (i) dark condition; (ii) 980; and and (b) (iii) hybrid nanoparticles (Ce6-G4-UCNP) as the photosensitizer: (i) dark condition; (ii) 980; and 660-nm laser excitation; and (c) the Ce6-G4-UCNP with increasing loading capacity of Ce6 (0.43, (iii) 660-nm laser excitation; (c) the Ce6-G4-UCNP with increasing loading capacity to ofthe Ce6 (0.43, 2.50, and 13.6 μM) underand 980-nm laser excitation. The fluorescence intensity was normalized 2.50, and 13.6 µM) under 980-nm laser excitation. Theoffluorescence intensity was normalized to the intrinsic fluorescence of FCLA solution in the absence additives and light exposure. intrinsic fluorescence of FCLA solution in the absence of additives and light exposure. 2.4. In Vitro PDT Analysis for Human-Breast Cancer Cells was performed Cancer on the MCF-7 2.4. In Vitro Laser-triggered PDT Analysis PDT for Human-Breast Cells cell lines that were cultured in the 2D and 3D models [44,45]. Generally, 2D cell-culture systems are a convenient way to study cancer cells in vitro;

Laser-triggered PDT wasarchitecture performed on cell–cell the MCF-7 lines that were cultured in the however, tissue-specific with and cell cell-extracellular matrix interactions are2D and reduced when Generally, cells grow on and adherent substrates. tumors growing in study a 3D spatial 3D models [44,45]. 2Dflat cell-culture systems are Solid a convenient way to cancer cells are currently being developed with as more advanced in cell-extracellular vitro models. A keymatrix advantage of in vitro;conformation however, tissue-specific architecture cell–cell and interactions 3D cultures in preclinical research is their capacity to modulate the molecular gradients that exist in spatial are reduced when cells grow on flat and adherent substrates. Solid tumors growing in a 3D living tissues, such as oxygen, nutrients, metabolites, and signaling molecules. Hence, 3D models conformation are currently being developed as more advanced in vitro models. A key advantage of 3D mimic the complexity of tissues more precisely than conventional 2D monolayers, thus potentially culturesbridging in preclinical is their capacity to modulate the molecular gradients exist in living the gapresearch between 2D cultures and animal models. Moreover, cells cultured in athat 3D scaffold tissues, generate such as aoxygen, nutrients, metabolites, and signaling molecules. Hence, 3D models mimic the cell population by enhancing the properties of cancer stem cells (CSCs), which have been demonstrated to retain cancer-initiating potential and self-renewal capability and lead to complexity of tissues more precisely than conventional 2D monolayers, thus potentially bridging the tumorigenesis and drug resistance in malignant tumors in vivo. In addition, 3D cell cultures usually gap between 2D cultures and animal models. Moreover, cells cultured in a 3D scaffold generate a cell upregulate the expression of hypoxia-responsive genes and are thus more resistant to PDT than 2D population by enhancing the properties of cancer stem cells (CSCs), which have been demonstrated monolayers, because of limited oxygen supply in the 3D spheroids. Accordingly, performing NIRto retaintriggered cancer-initiating potential and self-renewal capability and lead to tumorigenesis and drug PDT in a 3D model of the MCF-7 CSCs using the hybrid UCNPs as the PS is a therapeutic resistance in malignant tumors in vivo. In addition, 3D cell cultures usually upregulate the expression challenge. of hypoxia-responsive genes and are thus more resistant to PDT than 2D monolayers, because of limited oxygen supply in the 3D spheroids. Accordingly, performing NIR-triggered PDT in a 3D model of the MCF-7 CSCs using the hybrid UCNPs as the PS is a therapeutic challenge. Prior to PDT analysis, the control experiment shows that 660-nm or 980-nm laser irradiation has no influence on the survival of MCF-7 cell lines. Under darkness, Ce6 was nontoxic for the cells cultured in a conventional 2D system at a high dose (10 µM), but it immediately became a chemotherapy drug

Prior to PDT analysis, the control experiment shows that 660-nm or 980-nm laser irradiation has no influence on the survival of MCF-7 cell lines. Under darkness, Ce6 was nontoxic for the cells cultured in a conventional 2D system at a high dose (10 μM), but it immediately became a chemotherapy drug and completely destroyed target cells under 660-nm laser excitation for 10 min, Nanomaterials 2017, 7, 269 9 of 18 which potentially favored photodynamic medication under spatiotemporal control. However, the PDT outcome was ineffective when the 980-nm NIR laser was used as the excitation source, because andwas completely target cellswavelength under 660-nm excitation for1O10 min, which potentially the Ce6 nearly destroyed transparent at this andlaser photoinduced 2 production was inhibited. favored photodynamic medication under spatiotemporal control. However, the PDTwas outcome was The median lethal dose of Ce6 for the MCF-7 cells under 660-nm laser irradiation below 1 μM, ineffective when the 980-nm NIR laser was used as the excitation source, because the Ce6 was nearly suggesting that Ce6 is an effective PS responsive to1 the red light. transparent at this wavelength and photoinduced O2 production was inhibited. The median lethal Figure 5 presents the survival test results for the 2D cell cultures, using the hybrid UCNPs as the dose of Ce6 for the MCF-7 cells under 660-nm laser irradiation was below 1 µM, suggesting that Ce6 is PS under laser excitation. Herein, the loading concentration of the Ce6 on the UCNP determined an effective PS responsive to the red light. through Figure fluorescence was approximately μM. The experiments 5 presentsspectroscopy the survival test results for the 2D cell 0.25 cultures, using the control hybrid UCNPs as demonstrated that the hybrid UCNPs were also nontoxic in the dark and that 86% and 91% of the the PS under laser excitation. Herein, the loading concentration of the Ce6 on the UCNP determined MCF-7 cells fluorescence survived inspectroscopy the presence pristine Ce60.25 under 660-nm and 980-nm demonstrated laser irradiation, through wasof approximately µM. The control experiments that the hybrid UCNPs were also the dark was and quite that 86% 91%Ce6 of the MCF-7 cellslaser respectively. This result suggests thatnontoxic the PDTin efficiency lowand at this dosage under survived the presence of pristine Ce6Ce6 under 660-nm and onto 980-nm irradiation, respectively. excitation. Ininsharp contrast, when the was loaded thelaser dendrimer-modified UCNPs, This result suggests that the PDT efficiency was quite low at this Ce6 dosage under laser excitation. approximately 50% of the cells died under 660-nm light. Compared with the Ce6, the hybrid UCNPs In sharp contrast, the Ce6 was loaded onto the dendrimer-modified UCNPs, efficiency approximately exhibited higher PDT when performance, presumably because of higher cellular-uptake through 50% of the cells died under 660-nm light. Compared with the Ce6, the hybrid UCNPs exhibited endocytosis. Generally, the accumulation of Ce6 in target cells is inefficient because the negatively higher PDT performance, presumably because of higher cellular-uptake efficiency through endocytosis. charged –COOH groups may be electrostatically repulsed by the cell membranes; based on the Generally, the accumulation of Ce6 in target cells is inefficient because the negatively charged –COOH surface PAMAM dendrimers, the hybrid UCNPs possess multiple positive charges, thus favoring the groups may be electrostatically repulsed by the cell membranes; based on the surface PAMAM cellular uptake of nanoparticles. Therefore, accumulation of PS in thethe cells, usinguptake the UCNPs dendrimers, thethe hybrid UCNPs possess multiplethe positive charges, thus favoring cellular of as thethe drug carrier, would be considerably enhanced under the same incubation time. Most crucially, nanoparticles. Therefore, the accumulation of PS in the cells, using the UCNPs as the drug carrier, approximately 70% of the cells were under 980-nm laser irradiation, indicating the would be considerably enhanced underkilled the same incubation time. Most crucially, approximately 70%PDT efficiency was increased under NIR light. FCLA analysis revealed that the hybrid UCNPs exposed of the cells were killed under 980-nm laser irradiation, indicating the PDT efficiency was increased to under NIR light. FCLAhigher analysis1O revealed that the hybrid UCNPs laser exposed tobecause the 660-nm induced the 660-nm laser induced 2 formation than the 980-nm did, thelaser photoexcitation 1 O formation than the 980-nm laser did, because the photoexcitation of the Ce6 was less higher of the Ce6 was2 less effective through the upconversion fluorescence emitted from the UCNPs (Figure effective through upconversion fluorescence emitted from the UCNPs (Figure 4b). However, 4b). However, the in the vitro PDT experiments demonstrated a contrary result: NIR-laser irradiation the in vitro PDT experiments demonstrated a contrary result: NIR-laser irradiation provides more provides more positive therapeutic outcomes in MCF-7 cell lines. Because the polycationic UCNPs positive therapeutic outcomes in MCF-7 cell lines. Because the polycationic UCNPs improved the improved the cellular uptake, the marked NIR-triggered PDT effect was due to longer laser cellular uptake, the marked NIR-triggered PDT effect was due to longer laser wavelength and thus wavelength and thus deeper tissue penetration.1 Although higher 1O2 was produced under red-light deeper tissue penetration. Although higher O2 was produced under red-light excitation in the excitation in the tube NIR lightpass could easilythe pass theand cellsome membrane andand some organelles tube test, NIR lighttest, could easily through cellthrough membrane organelles reach the and reach hybridTherefore, UCNPs. surface Therefore, Ce6 was effectively under photoexcited under 980-nm laser hybridthe UCNPs. Ce6 surface was effectively photoexcited 980-nm laser irradiation, irradiation, thus dramatically thus dramatically enhancingenhancing the overall the PDToverall effect. PDT effect.

Figure 5. Photo-inducedcytotoxicity cytotoxicity for cellscells cultured in a 2Dinsystem the presence pristine of Figure 5. Photo-induced forMCF-7 MCF-7 cultured a 2D insystem in theof presence Ce6 (red) and hybrid nanoparticles (blue) as the photosensitizer under laser irradiation. [Ce6] = 0.25 µM.[Ce6] pristine Ce6 (red) and hybrid nanoparticles (blue) as the photosensitizer under laser irradiation. = 0.25 μM. The MCF-7 cells were also cultured on ultralow attachment surfaces in serum-free mediums with limited growth factors to form the 3D scaffolds. These tumorspheres, which have been proven to

The MCF-7 cells were also cultured on ultralow attachment surfaces in serum-free mediums with limited growth factors to form the 3D scaffolds. These tumorspheres, which have been proven

Nanomaterials 2017, 7, 269 Nanomaterials 2017, 7, 269

10 of 18 10 of 18

to possess CSC-like properties, served as an effective in vitro platform for screening anti-CSCs drugs. The 3D CSC-like cells were also exposed to as thean660-nm 980-nm lasersfor to screening compare anti-CSCs the wavelengthpossess properties, served effectiveand in vitro platform drugs. dependent efficiencies. after980-nm exposure, staining was performed to The 3D cells PDT were also exposed Immediately to the 660-nm and laserslive-dead to compare the wavelength-dependent evaluate viabilityImmediately distribution after of theexposure, cells in tumorspheres [46].was Figure 6a shows the pretreatment PDT efficiencies. live-dead staining performed to evaluate viability distribution of the living and dead cells in the 3D spheres specifically labeled with green and red distribution of the cells in tumorspheres [46]. Figure 6a shows the pretreatment distribution of the living fluorophores. Figure 6b demonstrates thatgreen after and treating the cells withAdditionally, the hybrid and dead cellsAdditionally, in the 3D spheres specifically labeled with red fluorophores. UCNPs under darkness, the dead cells marked in red were mainly located at the peripheral 3D Figure 6b demonstrates that after treating the cells with the hybrid UCNPs under darkness, of thethe dead spheres. The natural apoptosis oflocated the surrounding cells during of the tumorspheres cells marked in red were mainly at the peripheral of thethe 3Dcultivation spheres. The natural apoptosis was the possible cause of this observation. When the treated 3D cells were exposed to the laser of the surrounding cells during the cultivation of the tumorspheres was the possible cause oflight, this the dead portions clearly3D located inside the spheres, indicating thatdead the portions UCNPs successfully observation. Whenwere the treated cells were exposed to the laser light, the were clearly absorbed by thethe tumorspheres produced thethe PDT effect,successfully not only at the surface by butthe also at the center located inside spheres, indicating that UCNPs absorbed tumorspheres of spheres. Moreover, comparing Figure 6c,d, we noticed that the 980-nm laser induced cell produced the PDT effect, not only at the surface but also at the center of spheres. Moreover, higher comparing death inside the spheres. This result indicates that the NIR-light penetrated the 3D scaffolds more Figure 6c,d, we noticed that the 980-nm laser induced higher cell death inside the spheres. This result deeply, the PDT outcome to the accumulated hybrid UCNPs. Furthermore, indicatesthus thattriggering the NIR-light penetrated theaccording 3D scaffolds more deeply, thus triggering the PDT outcome as shown in Figure 6e, the quantitative analysis was conducted by manually counting the live and according to the accumulated hybrid UCNPs. Furthermore, as shown in Figure 6e, the quantitative dead cells, based on the spheres selected from the fluorescence images. result wasspheres consistent with analysis was conducted by manually counting the live and dead cells,The based on the selected the survival test for 2D cells, confirming the highest PDT performance, the lowest percentage of cell from the fluorescence images. The result was consistent with the survival test for 2D cells, confirming survival under 980-nm laser irradiation. To our knowledge, this is the first example of NIR-lightthe highest PDT performance, the lowest percentage of cell survival under 980-nm laser irradiation. driven PDT on a this 3D is tumorspherical platform using UCNPs ason the PS,tumorspherical carrier, and activator, To our knowledge, the first example of NIR-light-driven PDT a 3D platform simultaneously. using UCNPs as the PS, carrier, and activator, simultaneously.

(a)

(b)

(c)

(d)

Figure 6. Cont.

Nanomaterials 2017, 7, 269 Nanomaterials 2017, 7, 269

11 of 18 11 of 18

Nanomaterials 2017, 7, 269

11 of 18

(e)

(e)

Figure 6. 6. Fluorescence MCF-7cells cellscultured cultured a 3D tumorsphere system: (a) Figure Fluorescencestaining stainingimages images for MCF-7 in in a 3D tumorsphere system: (a) control: control: live (calcein AM, green) and (EthD-1, dead (EthD-1, red)incells in the absence of hybrid nanoparticles live (calcein AM, green) and dead red) cells the absence of hybrid nanoparticles and light and light exposure; andin (b–d) in the presence of hybrid nanoparticles incondition, dark condition, and under exposure; and (b–d) the presence of hybrid nanoparticles in dark and under 660-nm 660-nm and 980-nm laser irradiation, respectively. forsystem: the average Figure Fluorescence staining images for Quantitative MCF-7 (e) cells Quantitative cultured in a 3D tumorsphere (a) and 980-nm laser 6.irradiation, respectively. (e) analysis foranalysis the average distribution of live AM, in green) and a dead (EthD-1, red) inbased the absence of spheres hybrid nanoparticles distribution ofcontrol: live and cells a with tumorsphere with a cells standard deviation based on 10 spheres live and dead cells in dead a(calcein tumorsphere standard deviation on 10 selected from the and light exposure; and (b–d) in the presence of hybrid nanoparticles in dark condition, and under selected from the fluorescence images. fluorescence images. The scale bar isThe 100 scale µm. bar is 100 μm. 660-nm and 980-nm laser irradiation, respectively. (e) Quantitative analysis for the average distribution of live and dead cells in a tumorsphere with a standard deviation based on 10 spheres Tumorspheres driven cancer cells have been display characteristics of Tumorspheres driven cancer cells have been display characteristics selected from from thefrom fluorescence images. The scale bar is proven 100proven μm. to to

CSCs, of CSCs, which areare considered thethe main cause of cancer recurrence. Because thethe 3D3D spheres grow divergently, which considered main cause of cancer recurrence. Because spheres grow divergently, Tumorspheres driven from cancer cells have been proven to display characteristics of CSCs, from core peripheral, the inside cells inside the may spheres may more CSC/progenitor characteristic fromthe the core totoperipheral, the cells the spheres exhibit moreexhibit characteristic which are considered the main cause of cancer recurrence. Because the 3D spheres grow divergently, CSC/progenitor cell properties than the cells located on the surface. For the MCF-7 cells cultured in cell properties than cells on the For the the MCF-7 in the 3D tumorspheres, from thethe core to located peripheral, the surface. cells inside spheres cells may cultured exhibit more characteristic CSC/progenitor cell properties than laser the cells located onremarkable the surface. For the MCF-7 cells cultured thethe 3Dhybrid tumorspheres, the hybrid UCNPs combined with a NIR laser exhibited remarkable deepUCNPs combined with a NIR exhibited deep-tissue PDT effects,inallowing the 3Dallowing tumorspheres, the hybrid UCNPs combined NIR CSCs laserMoreover, exhibited deeptissue PDT effective effects, forof more effective destruction ofaspheres. the locatedremarkable inside the spheres.can for more destruction the CSCs located insidewith the the PDT outcome tissue PDT effects, allowing for more effective destruction of the CSCs located inside the spheres. Moreover, PDT outcome can bethe readily enhanced increasing dosagetime, of hybrid be readilythe enhanced by increasing dosage of hybridbyUCNPs, lightthe exposure and theUCNPs, power of Moreover, the PDT outcome can be readily enhanced by increasing the dosage of hybrid UCNPs, light time, and the power the of incident laser. Figure illustrates that the NIR-lighttheexposure incidentlight laser. Figure 7 illustrates that the NIR-light-triggered killedthat most the cells in the exposure time, and the of power the incident laser. Figure 77PDT illustrates the of NIR-lightPDT most of the cells in the tumorspheres when the exposure exposure time waswas increased to triggered PDTtriggered killed of the cells in was the tumorspheres the time increased to tumorspheres whenmost the killed exposure time increased to 20when min under the treatment of the UCNPs with 20 min under the treatment of the UCNPs with higher Ce6 loading (1.36 μM). Considering all 20 higher min under the treatment of Considering the UCNPs all with higher Ce6 loading μM). Considering all Ce6 loading (1.36 µM). aforementioned factors, (1.36 hybrid UCNPs are promising aforementioned factors, hybrid UCNPs are promising NIR-triggered PDT modules for application in aforementioned factors, hybrid UCNPs are promising NIR-triggered PDT modules for application in NIR-triggered PDT modules for application in the treatment of spheroidal CSCs. the treatment of spheroidal CSCs. the treatment of spheroidal CSCs.

(a)

(b)

Figure 7. (a) Fluorescence staining images for live and dead MCF-7 cells in a tumorsphere in the

Figure 7. (a)presence Fluorescence staining images for loading live and deadofMCF-7 in a tumorsphere of hybrid nanoparticles with higher capacity Ce6 (1.36cells μM). Control: live (calcein in the presence of hybrid nanoparticles withred) higher loading capacity of Ce6 (1.36 µM). live (calcein AM, green) and dead (EthD-1, cells in the absence of hybrid nanoparticles and Control: light exposure. AM, green) and dead (EthD-1, red) cells in the absence of hybrid nanoparticles and light exposure. (a) (b) The scale bar is 50 µm. (b) The quantitative analysis based on 10 spheres selected from the images Figure 7. that (a) Fluorescence images for live980-nm and dead MCF-7 cells in in the shows less than 20%staining of cells survived under laser irradiation fora20tumorsphere min. presence of hybrid nanoparticles with higher loading capacity of Ce6 (1.36 μM). Control: live (calcein AM, green) and dead (EthD-1, red) cells in the absence of hybrid nanoparticles and light exposure.

Nanomaterials 2017, 7, 269

12 of 18

2.5. In Vivo Assessment for DNA Damage in Tumor Tissues We further performed in vivo PDT analysis based on a 4T1 tumor-bearing mouse model treated with the hybrid UCNPs in combination with 980-nm laser exposure. The hybrid UCNPs with different dosages (7 µg and 21 µg) was loaded by intratumoral injection followed by exposure to laser for 10 min. From the immunohistochemical (IHC) analysis for the tissue slices, the expression of Ki67, a protein marker for cell proliferation [47], was obviously decreased in the tumor under higher dose combined with laser exposure (Figure 8a–c). Because of the observation of in vitro 1 O2 production, which is a strong inducer for DNA damage including DNA strand breaks [48], we further examined whether DNA damage in tumors could be induced by the photosensitizing effect of the UCNPs with increasing dosage. The expression of γH2AXser139 , a protein marker for DNA double strand breaks [49], was massively induced under higher dose combined with laser exposure (Figure 8d–f). Moreover, based on the IHC images taken in four different fields, statistical analysis conducted by manually counting the cells indicates that the ratio of positively stained cells for the Ki67 marker decreased from 24.0% to 4.88% and for the γH2AXser139 marker increased from 2.16% to 15.6% after laser exposure (Supplementary Materials, Figures S7 and S8). Accordingly, the preliminary data suggest that the tumor growth was effectively inhibited through the cellular DNA damage induced by the NIR-triggered PDT effect.

Figure 8. In vivo analysis for a murine 4T1 breast cancer model based on 980-nm laser light-trigged photodynamic therapy (PDT) using the hybrid nanoparticles as the photosensitizer. The tumors were treated with lower (7 µg) and higher (21 µg) dosage of the nanoparticles combined with (+) and without (−) light exposure. Immunohistochemical analysis of the tissue slices for: (a–c) Ki67; and (d–f) γH2AXser139 , corresponding to the protein markers for cell proliferation and DNA damage, respectively. The brown colors indicated positive staining for the markers. The cell nuclei were counterstained with hematoxylin (blue colors).

Nanomaterials 2017, 7, 269

13 of 18

3. Conclusions We successfully synthesized a dendrimer-functionalized hybrid UCNPs for performing NIR-light-driven PDT on human breast cancer cell lines and a murine breast cancer model. The water-soluble nanoparticles composed of lanthanide-doped NaYF4 nanocrystals encapsulated by the PAMAM dendrimers exhibited significant upconversion fluorescence under 980-nm laser irradiation. Thereby, the Ce6 molecules immobilized on the surface through an LBL strategy were effectively photoexcited to generate cytotoxic ROS. The chemiluminescence assay confirmed that 1 O2 as the main ROS was readily accumulated during the photoexcitation and that the 660-nm LD was a more effective light source for photoexciting the hybrid UCNPs through the Q-band absorption of the Ce6; however, in vitro experiments revealed that the 980-nm-laser light had higher cytotoxicity in the MCF-7 cells cultured according to a conventional 2D model. The reverse PDT outcome was most likely due to the NIR light demonstrating higher penetration of a living system than visible light did. Moreover, compared with pristine Ce6 under visible-light excitation, the hybrid UCNPs displayed optimal in vitro PDT efficacy under NIR-light excitation. This finding is attributed to the enhancement of cellular uptake by the polycationic dendrimers through electrostatic interactions with polyanionic membrane lipids. Most notably, NIR light also induced an efficient PDT effect in the 3D cell model of MCF-7 tumorspheres, suggesting that hybrid UCNPs can be effectively applied in more complicated cellular textures. In vivo assessment based on immunohistochemical analysis for tumor tissues on a 4T1 tumor-bearing female mouse eventually shows significant inhibition of tumor cell proliferation through cellular DNA damage. Thus, the 1 O2 produced from the hybrid UCNPs is the main cause for the successful in vitro and in vivo NIR-triggered PDT. 4. Materials and Methods 4.1. Materials and Instruments The chemical reagents and organic solvents for materials synthesis were obtained as high-purity reagent-grade from commercial suppliers and used without further purification. The fourth generation PAMAM dendrimer (G4-NH2 ) was purchased from Dendritech, Inc. (Midland, MI, USA). For the materials analysis, FT-IR was performed on a Bruker Alpha FT spectrometer (Bruker Corp., Billerica, MA, USA). UV-Vis absorption spectra were recorded on a Thermo Genesys 10S UV-vis spectrometer (ThermoFisher Scientific, Waltham, MA, USA) equipped with a thermostatic cuvette holder. Fluorescence emission spectra for 1 O2 analysis were recorded on a Hitachi F-2500 spectrometer (Hitachi High-Tech., Tokyo, Japan). TEM, EDXS, and XRD were performed on a Jeol JEM-2100 instrument, a Jeol JSM-6330F instrument (Jeol Ltd., Toyko, Japan), and a Bruker D2 PHASER X-ray Diffractometer (Bruker Corp., Billerica, MA, USA), respectively. Dynamic light scattering analysis involving particle size and zeta potential measurements was carried out by a Malvern Zetasizer Nano ZS (Malvern Instrument Ltd., Malvern, UK). TGA was recorded on a Thermo Cahn VersaTherm HS TG analyzer (ThermoFisher Scientific, Waltham, MA, USA). The analysis for upconversion fluorescence and photodynamic reaction were carried out on a homemade experimental apparatus using either 660-nm or 980-nm laser diode as the excitation source (output power: 2 W). The upconversion emission from the samples in a cuvette folder was collected and induced by a fiber bundle into a CCD imaging spectrometer (USB-4000, Ocean Optics, Largo, FL, USA) for spectra recording. 4.2. Synthesis of Hybrid UCNPs NaYF4 :Yb, Tm/NaYF4 UCNPs with a core-shell structure was synthesized by a solvothermal procedure [33]. In brief, for the synthesis of the core nanoparticles, anhydrous Y(CH3 CO2 )3 (372 mg), Yb(CH3 CO2 )3 (210 mg), and Tm(CH3 CO2 )3 (3.5 mg) were added to a 100 mL two-neck round-bottom flask containing oleic acid (35 mL). The solution was first purged with N2 and then heated slowly to 110 ◦ C under reduced pressure until complete removal of residual water and oxygen. The temperature of the reaction flask was then lowered to 50 ◦ C under a gentle N2 flow. During this time, an anhydrous

Nanomaterials 2017, 7, 269

14 of 18

methanol solution of ammonium fluoride (0.3 g) and sodium hydroxide (0.4 g) was prepared via sonication, and the mixture solution was then transferred to the two-neck flask via cannulation. The reaction flask was heated to 65 ◦ C under reduced pressure until complete removal of methanol. Subsequently, the reaction temperature was increased to 300 ◦ C as quick as possible and maintained at this temperature for 30 min under N2 . After reaction, the nanoparticles were precipitated by adding excess ethanol (400 mL) and collected by centrifugation at 4500 rpm. The resulting pellet was redispersed in hexane and precipitated with excess ethanol. The dispersion-precipitation process was repeated until the removal of free oleic acids. The core-shell UCNPs with an outer layer of NaYF4 was synthesized following the same solvolthermal procedure except that the reaction time was increased to 90 min. For the synthesis of citrate-modified UCNPs, 100 mg of core-shell UCNPs was dispersed in 60 mL of diethylene glycol (DEG) followed by adding 200 mg of trisodium citrate. The mixture was then heated to 200 ◦ C with gentle stirring for 17 h. After cooling to room temperature, the modified UCNPs were collected by centrifugation under 12,000 rpm. The precipitate was rinsed with 20 mL of DEG for 3 times to remove nonadsorbed citrate ligands followed by rinsed with 20 mL of CH2 Cl2 to remove DEG. The final product was obtained by drying under room temperature. For the synthesis of dendrimer-encapsulated UCNPs via LBL method, 20 mg of citrate-modified UCNPs was dispersed in 15 mL of H2 O followed by adding 40 µL of NH2 -terminated PAMAM dendrimer (10 wt % in methanol). The mixture solution was then carefully adjusted to pH = 4 by adding HCl solution and allowed to stirring at room temperature for 24 h. After reaction, water was first removed by vacuum distillation, and the modified UCNPs was precipitated by adding 200 mL of cold acetone. The final product collected by centrifugation under 12,000 rpm was drying under vacuum under the temperature less than 35 ◦ C. For the synthesis of hybrid UCNPs, 10 mg of the dendrimer-encapsulated UCNPs was dispersed in 5 mL of methanol with gentle stirring followed by adding Ce6 stock solution (1.5 × 10−3 M in methanol). The feeding amount of Ce6 was fine adjusted by controlling the injection volume of the stock solution. The mixture solution was stirred under room temperature for 2 h, and the solvent was removed under reduced pressure. The crude UCNPs was rinsed with CH2 Cl2 to wash off the nonadsorbed Ce6 until the CH2 Cl2 layer was colorless. The final hybrid nanoparticles was drying under room temperature, and the loading capacity of the Ce6 was determined by fluorescence spectroscopy analysis. Prior to biological experiments, the hybrid UCNPs was prepared in stock aqueous solutions with corresponding Ce6 concentrations of 0.43, 2.50 and 13.6 µM. 4.3. The Singlet Oxygen Assay In a typical FCLA experiment, 20 µL of a FCLA/ethanol solution (0.7 mg/mL) was added to 2 mL of a UCNPs solution and transferred into a 1-cm quartz cuvette. The solution was irradiated with an either 660-nm or 980-nm laser diode, and the emission intensity of the exposed solutions at 519-nm was recorded every 2 min under 496-nm excitation by fluorescence spectroscopy. For the control experiment, FCLA emission was also recorded at the same conditions in the absence of laser irradiation. 4.4. Cell Culture and Tumorsphere Cultivation For a conventional 2D cell model, MCF-7 cells were obtained from American Type Culture Collection (Manassas, VA, USA) and cultured in DMEM medium (Invitrogen Corporation, Grand Island, NY, USA) containing 10% fetal bovine serum (Invitrogen) and 5 µg/mL insulin (Sigma-Aldrich, St. Louis, MO, USA). For a 3D tumorsphere cultivation, MCF-7 cells were prepared as a density of 1 × 104 cells/mL in DMEM/F12 medium (Invitrogen Corporation) containing 0.5% methylcellulose (Sigma-Aldrich), 0.4% bovine serum albumin (Sigma-Aldrich), 10 ng/mL of EGF (PeproTech, Rocky Hill, NJ, USA), 10 ng/mL bFGF (PeproTech, Rocky Hill, NJ, USA), 5 µg/mL insulin, 1 µM hydrocortisone (Sigma-Aldrich) and 4 µg/mL heparin (Sigma-Aldrich). Cell suspension (2 mL)

Nanomaterials 2017, 7, 269

15 of 18

was seeded into wells of suspension culture 6-well-plate (Greiner Bio One International GmbH, Frickenhausen, Germany) and incubated for 7 days. 4.5. Cell Viability Assay To determine the PDT efficiency, cell proliferation for the cells cultured in a 2D model after light exposure were determined by CCK8 cell viability assay reagent (Sigma-Aldrich) using a microplate reader (Molecular Devices, Sunnyvale, CA, USA) according to the manufacturer’s recommendations. For the in vitro photoexicitation experiment, MCF-7 cells were firstly suspended as 1 × 105 per mL for 2.5 mL and transferred into a 1-cm quartz cuvette and directly exposed to a laser beam with either 660-nm or 980-nm wavelength at fixed time intervals under gentle agitation. The exposed cells were then seeded into 96-well-plate at 1 × 104 per well and cultured for another 48 h. The absorbance values at 440 nm of non-treatment control were set as 100% of cell proliferation. Cell proliferation for the cells cultivated in a 3D model was determined by Live/Dead cell viability assay (Thermo Fisher Scientific, Waltham, MA, USA). The viable or dead cells were specifically stained by green and red fluorophores derived from calcein AM and ethidium homodimer-1, respectively. Following the same protocol of the in vitro photoexcitation, the exposed tumorspheres were collected and washed in phosphate buffered saline (PBS) and then suspended in 250 µL PBS containing 4 µM of calcien AM/2 µM of ethidium homodimer-1. After incubation at 37 ◦ C for 30 min, fluorescence images with respect to the distributions of viable and dead cells in tumorspheres were captured by inverted fluorescence microscopy (AE30, Motic Electric Group Co., Ltd., Xiamen, China) and counted with ImageJ software (NIH, Bethesda, MD, USA). 4.6. In Vivo Assessment for Tumor Tissues Murine 4T1 breast cancer cells were obtained from American Type Culture Collection (ATCC, Manassas, VA, USA) and maintained by DMEM containing 10% fetal bovine serum. The orthotopic breast cancer model was conducted by injection of 4T1 cells into mammary fat pads of BALB/c female mice (purchased from the National Laboratory Animal Center of Taiwan, Taipei, Taiwan) according to the previous report [50]. All the animal studies were operated following a protocol approved by Institutional Animal Care & Utilization Committee of Chung Shan Medical University (IACUC Approval No. 1379). Briefly, 2 × 105 4T1 cells were suspended in 50 µL matrigel (BD Biosciences, San Jose, CA, USA) at a concentration of 2 mg/mL and injected the 4th pair of mammary fat pad. The treatment was begun when tumor volume reached to 100 mm3 (around 21 days after injection). The tumors were exposed with 980-nm laser after intratumoral injection of the hybrid UCNPs for 10 min per time and the treatment was performed every 3 days for total 3 times. Three mice were sacrificed at day 3 after the last treatment and tumors were taken out for performing immunohistochemistrial analysis. The tumors were fixed with 3.7% formaldehyde and embedded in paraffin. Four micrograms of paraffin sections were sliced followed by dewaxed/rehydration steps and used for detection of Ki-67 (a marker for cell proliferation) and γH2AXser139 (a marker for DNA double strand breaks) expression. Briefly, the sections were incubated with monoclonal rabbit anti-Ki67 (Cat. No. GTX16667, GeneTex International Corporation, Hsinchu City, Taiwan) or monoclonal rabbit anti-γH2AXser139 (Cat. No. NB100-79967, Novus Biologicals, Littleton, CO, USA) using a standard avidin-biotin-peroxidase complex method. 3,30 -Diaminobenzidine (DAKO, Carpinteria, CA, USA) was then used to detect the positive staining. Supplementary Materials: The following are available online at http://www.mdpi.com/2079-4991/7/9/269/s1, Figure S1: XRD analysis of: (a) core-shell; and (b) core NaYF4 nanocrystals with α-phase (cubic) pattern; Figure S2: EDXS analysis for the NaYF4 nanocrustals with successful Yb and Tm-doping; Figure S3: TGA analysis for: (a) ligand-free; (b) oleate; and (c) citrate-modified upconversion nanoparticles (UCNPs); Figure S4: zeta potential measurements for UCNPs sequentially encapsulated with: (a) citrate; (b) PAMAM dendrimer; and (c) chlorin e6 (Ce6); Figure S5: FT-IR analysis for: (a) citrate; and (b) PAMAM dendrimer-modified UCNPs. The arrows indicate the characteristic absorption bands of the surface functional groups; Figure S6: particle size distribution for PAMAM dendrimer-modified UCNPs; Figure S7: the Immunohistochemical (IHC) analysis of the tissue slices

Nanomaterials 2017, 7, 269

16 of 18

for Ki67 protein marker in four different fields. The tumors were treated with lower (7 µg) and higher (21 µg) dosage of the UCNPs combined with (+) and without (−) 980-m laser exposure: (a) 21 µg/−; (b) 7 µg/+; and (c) 21 µg/+; and (d) the ratios of the positively stained cells for (a–c) analyzed by manually counting the cells in the IHC images; Figure S8: the Immunohistochemical (IHC) analysis of the tissue slices for γH2AXser139 protein marker in four different fields. The tumors were treated with lower (7 µg) and higher (21 µg) dosage of the UCNPs combined with (+) and without (−) 980-m laser exposure: (a) 21 µg/−; (b) 7 µg/+; and (c) 21 µg/+; and (d) the ratios of the positively stained cells for (a–c) analyzed by manually counting the cells in the IHC images. Acknowledgments: The authors would like to thank Chung Shan Medical University and Ministry of Science and Technology (MOST) of Taiwan, for financially supporting this research (MOST104-2119-M-040-001) and for instrumental analysis. The authors are also grateful to Jing-Yun Wu from National Chi-Nan University for support with TGA analysis and to Hsien-Ming Lee from Academic Sinica for support with dynamic light scattering analysis. Author Contributions: Chih-Chien Chu conceived the idea and designed the experiments; Wen-Wei Chang contributed the biological evaluation; Bing-Yen Wang, Ming-Liang Liao, and Guan-Ci Hong performed the experiments and analyzed the data; and Wen-Wei Chang and Chih-Chien Chu wrote the paper. Conflicts of Interest: The authors declare no conflict of interest.

References 1. 2. 3.

4. 5.

6.

7.

8.

9.

10.

11.

12.

13.

Chen, G.; Qiu, H.; Prasad, P.N.; Chen, X. Upconversion Nanoparticles: Design, Nanochemistry, and Applications in Theranostics. Chem. Rev. 2014, 114, 5161–5214. [CrossRef] [PubMed] Shanmugam, V.; Selvakumar, S.; Yeh, C.-S. Near-infrared light-responsive nanomaterials in cancer therapeutics. Chem. Soc. Rev. 2014, 43, 6254–6287. [CrossRef] [PubMed] Li, J.; Lee, W.Y.-W.; Wu, T.; Xu, J.; Zhang, K.; Wong, D.S.H.; Li, R.; Li, G.; Bian, L. Near-infrared light-triggered release of small molecules for controlled differentiation and long-term tracking of stem cells in vivo using upconversion nanoparticles. Biomaterials 2016, 110, 1–10. [CrossRef] [PubMed] Wang, C.; Li, X.; Zhang, F. Bioapplications and biotechnologies of upconversion nanoparticle-based nanosensors. Analyst 2016, 141, 3601–3620. [CrossRef] [PubMed] Xu, Y.; Xiang, J.; Zhao, H.; Liang, H.; Huang, J.; Li, Y.; Pan, J.; Zhou, H.; Zhang, X.; Wang, J.H.; et al. Human amniotic fluid stem cells labeled with up-conversion nanoparticles for imaging-monitored repairing of acute lung injury. Biomaterials 2016, 100, 91–100. [CrossRef] [PubMed] Tian, G.; Zheng, X.; Zhang, X.; Yin, W.; Yu, J.; Wang, D.; Zhang, Z.; Yang, X.; Gu, Z.; Zhao, Y. TPGS-stabilized NaYbF4:Er upconversion nanoparticles for dual-modal fluorescent/CT imaging and anticancer drug delivery to overcome multi-drug resistance. Biomaterials 2015, 40, 107–116. [CrossRef] [PubMed] Punjabi, A.; Wu, X.; Tokatli-Apollon, A.; El-Rifai, M.; Lee, H.; Zhang, Y.; Wang, C.; Liu, Z.; Chan, E.M.; Duan, C.; et al. Amplifying the Red-Emission of Upconverting Nanoparticles for Biocompatible Clinically Used Prodrug-Induced Photodynamic Therapy. ACS Nano 2014, 8, 10621–10630. [CrossRef] [PubMed] Tian, G.; Gu, Z.; Zhou, L.; Yin, W.; Liu, X.; Yan, L.; Jin, S.; Ren, W.; Xing, G.; Li, S.; et al. Mn2+ Dopant-Controlled Synthesis of NaYF4:Yb/Er Upconversion Nanoparticles for In Vivo Imaging and Drug Delivery. Adv. Mater. 2012, 24, 1226–1231. [CrossRef] [PubMed] Yang, S.; Li, N.; Liu, Z.; Sha, W.; Chen, D.; Xu, Q.; Lu, J. Amphiphilic copolymer coated upconversion nanoparticles for near-infrared light-triggered dual anticancer treatment. Nanoscale 2014, 6, 14903–14910. [CrossRef] [PubMed] Qiu, H.; Yang, C.; Shao, W.; Damasco, J.; Wang, X.; Ågren, H.; Prasad, P.; Chen, G. Enhanced Upconversion Luminescence in Yb3+ /Tm3+ -Codoped Fluoride Active Core/Active Shell/Inert Shell Nanoparticles through Directed Energy Migration. Nanomaterials 2014, 4, 55–68. [CrossRef] [PubMed] Wang, X.; Yang, C.-X.; Chen, J.-T.; Yan, X.-P. A Dual-Targeting Upconversion Nanoplatform for Two-Color Fluorescence Imaging-Guided Photodynamic Therapy. Anal. Chem. 2014, 86, 3263–3267. [CrossRef] [PubMed] Wang, M.; Chen, Z.; Zheng, W.; Zhu, H.; Lu, S.; Ma, E.; Tu, D.; Zhou, S.; Huang, M.; Chen, X. Lanthanide-doped upconversion nanoparticles electrostatically coupled with photosensitizers for near-infrared-triggered photodynamic therapy. Nanoscale 2014, 6, 8274–8282. [CrossRef] [PubMed] Wang, C.; Cheng, L.; Liu, Y.; Wang, X.; Ma, X.; Deng, Z.; Li, Y.; Liu, Z. Imaging-Guided pH-Sensitive Photodynamic Therapy Using Charge Reversible Upconversion Nanoparticles under Near-Infrared Light. Adv. Funct. Mater. 2013, 23, 3077–3086. [CrossRef]

Nanomaterials 2017, 7, 269

14.

15. 16.

17. 18. 19.

20. 21.

22.

23. 24. 25. 26. 27.

28.

29. 30.

31.

32.

33.

17 of 18

Liu, X.; Zheng, M.; Kong, X.; Zhang, Y.; Zeng, Q.; Sun, Z.; Buma, W.J.; Zhang, H. Separately doped upconversion-C60 nanoplatform for NIR imaging-guided photodynamic therapy of cancer cells. Chem. Commun. 2013, 49, 3224–3226. [CrossRef] [PubMed] Rapozzi, V.; Jori, G. Resistance to Photodynamic Therapy in Cancer; Springer International Publishing: Cham, Switzerland, 2015; p. 248. Zhao, L.; Peng, J.; Huang, Q.; Li, C.; Chen, M.; Sun, Y.; Lin, Q.; Zhu, L.; Li, F. Near-Infrared Photoregulated Drug Release in Living Tumor Tissue via Yolk-Shell Upconversion Nanocages. Adv. Funct. Mater. 2014, 24, 363–371. [CrossRef] Qian, H.S.; Guo, H.C.; Ho, P.C.-L.; Mahendran, R.; Zhang, Y. Mesoporous-Silica-Coated Up-Conversion Fluorescent Nanoparticles for Photodynamic Therapy. Small 2009, 5, 2285–2290. [CrossRef] [PubMed] Wu, X.-J.; Xu, D. Formation of Yolk/SiO2 Shell Structures Using Surfactant Mixtures as Template. J. Am. Chem. Soc. 2009, 131, 2774–2775. [CrossRef] [PubMed] Budijono, S.J.; Shan, J.; Yao, N.; Miura, Y.; Hoye, T.; Austin, R.H.; Ju, Y.; Prud’homme, R.K. Synthesis of Stable Block-Copolymer-Protected NaYF4:Yb3+ , Er3+ Up-Converting Phosphor Nanoparticles. Chem. Mater. 2010, 22, 311–318. [CrossRef] Bogdan, N.; Vetrone, F.; Roy, R.; Capobianco, J.A. Carbohydrate-coated lanthanide-doped upconverting nanoparticles for lectin recognition. J. Mater. Chem. 2010, 20, 7543–7550. [CrossRef] Mohammadifar, E.; Nemati Kharat, A.; Adeli, M. Polyamidoamine and polyglycerol; their linear, dendritic and linear-dendritic architectures as anticancer drug delivery systems. J. Mater. Chem. B 2015, 3, 3896–3921. [CrossRef] Liu, H.; Shen, M.; Zhao, J.; Zhu, J.; Xiao, T.; Cao, X.; Zhang, G.; Shi, X. Facile formation of folic acid-modified dendrimer-stabilized gold-silver alloy nanoparticles for potential cellular computed tomography imaging applications. Analyst 2013, 138, 1979–1987. [CrossRef] [PubMed] Matai, I.; Gopinath, P. Chemically Cross-Linked Hybrid Nanogels of Alginate and PAMAM Dendrimers as Efficient Anticancer Drug Delivery Vehicles. ACS Biomater. Sci. Eng. 2016, 2, 213–223. [CrossRef] Siriviriyanun, A.; Imae, T.; Calderó, G.; Solans, C. Phototherapeutic functionality of biocompatible graphene oxide/dendrimer hybrids. Colloids Surf. B Biointerfaces 2014, 121, 469–473. [CrossRef] [PubMed] Figueroa, E.R.; Lin, A.Y.; Yan, J.; Luo, L.; Foster, A.E.; Drezek, R.A. Optimization of PAMAM-gold nanoparticle conjugation for gene therapy. Biomaterials 2014, 35, 1725–1734. [CrossRef] [PubMed] Tomalia, D.A.; Baker, H.; Dewald, J.; Hall, M.; Kallos, G.; Martin, S.; Roeck, J.; Ryder, J.; Smith, P. A New Class of Polymers: Starburst-Dendritic Macromolecules. Polym. J. 1985, 17, 117–132. [CrossRef] Worden, J.G.; Dai, Q.; Huo, Q. A nanoparticle-dendrimer conjugate prepared from a one-step chemical coupling of monofunctional nanoparticles with a dendrimer. Chem. Commun. 2006, 14, 1536–1538. [CrossRef] [PubMed] Tsai, Y.-J.; Hu, C.-C.; Chu, C.-C.; Imae, T. Intrinsically Fluorescent PAMAM Dendrimer as Gene Carrier and Nanoprobe for Nucleic Acids Delivery: Bioimaging and Transfection Study. Biomacromolecules 2011, 12, 4283–4290. [CrossRef] [PubMed] Weiswald, L.-B.; Bellet, D.; Dangles-Marie, V. Spherical Cancer Models in Tumor Biology. Neoplasia 2015, 17, 1–15. [CrossRef] [PubMed] Alemany-Ribes, M.; García-Díaz, M.; Busom, M.; Nonell, S.; Semino, C.E. Toward a 3D Cellular Model for Studying In Vitro the Outcome of Photodynamic Treatments: Accounting for the Effects of Tissue Complexity. Tissue Eng. Part A 2013, 19, 1665–1674. [CrossRef] [PubMed] Huang, X.; Tian, X.-J.; Yang, W.-L.; Ehrenberg, B.; Chen, J.-Y. The conjugates of gold nanorods and chlorin e6 for enhancing the fluorescence detection and photodynamic therapy of cancers. Phys. Chem. Chem. Phys. 2013, 15, 15727–15733. [CrossRef] [PubMed] Gong, H.; Dong, Z.; Liu, Y.; Yin, S.; Cheng, L.; Xi, W.; Xiang, J.; Liu, K.; Li, Y.; Liu, Z. Engineering of Multifunctional Nano-Micelles for Combined Photothermal and Photodynamic Therapy Under the Guidance of Multimodal Imaging. Adv. Funct. Mater. 2014, 24, 6492–6502. [CrossRef] Boyer, J.-C.; Carling, C.-J.; Gates, B.D.; Branda, N.R. Two-Way Photoswitching Using One Type of Near-Infrared Light, Upconverting Nanoparticles, and Changing Only the Light Intensity. J. Am. Chem. Soc. 2010, 132, 15766–15772. [CrossRef] [PubMed]

Nanomaterials 2017, 7, 269

34.

35.

36.

37. 38. 39.

40. 41.

42. 43. 44. 45.

46.

47. 48. 49. 50.

18 of 18

Vetrone, F.; Naccache, R.; Mahalingam, V.; Morgan, C.G.; Capobianco, J.A. The Active-Core/Active-Shell Approach: A Strategy to Enhance the Upconversion Luminescence in Lanthanide-Doped Nanoparticles. Adv. Funct. Mater. 2009, 19, 2924–2929. [CrossRef] Zhou, H.-P.; Xu, C.-H.; Sun, W.; Yan, C.-H. Clean and Flexible Modification Strategy for Carboxyl/Aldehyde-Functionalized Upconversion Nanoparticles and Their Optical Applications. Adv. Funct. Mater. 2009, 19, 3892–3900. [CrossRef] Wang, D.; Chen, C.; Ke, X.; Kang, N.; Shen, Y.; Liu, Y.; Zhou, X.; Wang, H.; Chen, C.; Ren, L. Bioinspired Near-Infrared-Excited Sensing Platform for In Vitro Antioxidant Capacity Assay Based on Upconversion Nanoparticles and a Dopamine-Melanin Hybrid System. ACS Appl. Mater. Interfaces 2015, 7, 3030–3040. [CrossRef] [PubMed] Tong, L.; Lu, E.; Pichaandi, J.; Cao, P.; Nitz, M.; Winnik, M.A. Quantification of Surface Ligands on NaYF4 Nanoparticles by Three Independent Analytical Techniques. Chem. Mater. 2015, 27, 4899–4910. [CrossRef] Chu, C.-C.; Imae, T. Fluorescence Investigations of Oxygen-Doped Simple Amine Compared with Fluorescent PAMAM Dendrimer. Macromol. Rapid Commun. 2009, 30, 89–93. [CrossRef] [PubMed] Lee, W.I.; Bae, Y.; Bard, A.J. Strong Blue Photoluminescence and ECL from OH-Terminated PAMAM Dendrimers in the Absence of Gold Nanoparticles. J. Am. Chem. Soc. 2004, 126, 8358–8359. [CrossRef] [PubMed] Wang, D.; Imae, T. Fluorescence Emission from Dendrimers and Its pH Dependence. J. Am. Chem. Soc. 2004, 126, 13204–13205. [CrossRef] [PubMed] Chen, X.; Zhao, Z.; Jiang, M.; Que, D.; Shi, S.; Zheng, N. Preparation and photodynamic therapy application of NaYF4:Yb, Tm-NaYF4:Yb, Er multifunctional upconverting nanoparticles. New J. Chem. 2013, 37, 1782–1788. [CrossRef] Wang, C.; Tao, H.; Cheng, L.; Liu, Z. Near-infrared light induced in vivo photodynamic therapy of cancer based on upconversion nanoparticles. Biomaterials 2011, 32, 6145–6154. [CrossRef] [PubMed] Dou, Q.Q.; Teng, C.P.; Ye, E.; Loh, X.J. Effective near-infrared photodynamic therapy assisted by upconversion nanoparticles conjugated with photosensitizers. Int. J. Nanomed. 2015, 10, 419–432. Lee, C.-H.; Yu, C.-C.; Wang, B.-Y.; Chang, W.-W. Tumorsphere as an effective in vitro platform for screening anti-cancer stem cell drugs. Oncotarget 2015, 7, 1215–1226. [CrossRef] [PubMed] Peng, C.-Y.; Fong, P.-C.; Yu, C.-C.; Tsai, W.-C.; Tzeng, Y.-M.; Chang, W.-W. Methyl Antcinate A Suppresses the Population of Cancer Stem-Like Cells in MCF7 Human Breast Cancer Cell Line. Molecules 2013, 18, 2539–2548. [CrossRef] [PubMed] Chen, Y.-C.; Lou, X.; Zhang, Z.; Ingram, P.; Yoon, E. High-Throughput Cancer Cell Sphere Formation for Characterizing the Efficacy of Photo Dynamic Therapy in 3D Cell Cultures. Sci. Rep. 2015, 5, 12175. [CrossRef] [PubMed] Scholzen, T.; Gerdes, J. The Ki-67 protein: From the known and the unknown. J. Cell. Physiol. 2000, 182, 311–322. [CrossRef] Cadet, J.; Wagner, J.R. DNA Base Damage by Reactive Oxygen Species, Oxidizing Agents, and UV Radiation. Cold Spring Harb. Perspect. Biol. 2013, 5, a012559. [CrossRef] [PubMed] Siddiqui, M.S.; François, M.; Fenech, M.F.; Leifert, W.R. Persistent γH2AX: A promising molecular marker of DNA damage and aging. Mutat. Res. Rev. Mutat. Res. 2015, 766, 1–19. [CrossRef] [PubMed] Chang, W.-W.; Kuan, Y.-D.; Chen, M.-C.; Lin, S.-T.; Lee, C.-H. Tracking of mouse breast cancer stem-like cells with Salmonella. Exp. Biol. Med. 2012, 237, 1189–1196. [CrossRef] [PubMed] © 2017 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).

Near-Infrared-Triggered Photodynamic Therapy toward Breast Cancer Cells Using Dendrimer-Functionalized Upconversion Nanoparticles.

Water-soluble upconversion nanoparticles (UCNPs) that exhibit significant ultraviolet, blue, and red emissions under 980-nm laser excitation were succ...
6MB Sizes 0 Downloads 11 Views