You are on page 1of 6

Chinese Chemical Letters 29 (2018) 1725–1730

Contents lists available at ScienceDirect

Chinese Chemical Letters


journal homepage: www.elsevier.com/locate/cclet

[29_TD$IF]Review

Au-Fe3O4 heterostructures for catalytic, analytical, and biomedical


applications
Baoling Liua , Hongchen Zhangb , Ya Dinga,*
a
Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China
b
Beijing Key Laboratory of Magnetoelectric Materials and Devices (BKL-MEMD), Beijing Innovation Center for Engineering Science and Advanced Technology
(BIC-ESAT), Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing 100871, China

A R T I C L E I N F O [31_TD$IF]A B S T R A C T

Article history: Heterostructures are a series of nanomaterials combining different components into a single
Received 1 October 2018 nanostructure. Au-Fe3O4 heterostructures have received considerable attentions because of their
Received in revised form 2 December 2018 superior properties coming from both individual and combinational features of gold and iron oxide
Accepted 3 December 2018
nanoparticles. Their intrinsically peculiar magnetic, optical properties, and structure designability
Available online 5 December 2018
greatly enhance and broaden their potential applications in catalysis, assay, multimodal imaging, and
synergistic treatment for tumor. In this review, we systematically introduce the preparation methods of
Keywords:
Au-Fe3O4 heterostructures and their potential applications in the biomedical field, focusing on the unique
Au-Fe3O4 heterostructures
Janus nanoparticles
synergistic effect caused by the combination of gold and iron oxide structures. This review will provide
Catalyst insights into the structure control in adjusting the function of heterogeneous or hybrid material, such as
Multimodal imaging Au-Fe3O4 heterostructures, to implement their biomedical applications.
Synergistic treatment © 2018 Chinese Chemical Society and Institute of Materia Medica, Chinese Academy of Medical Sciences.
Published by Elsevier B.V. All rights reserved.

1. Introduction nanoparticles, PB nanocubes coated with AuNPs, were developed


to carry out [3_TD$IF]magnetic resonance imaging (MRI) and computed
Inorganic nanoparticles (NPs) and their oxides with various tomography (CT) imaging and synergistic photothermal and
compositions, structures, and functions have been synthesized radiosensitive therapy [7]. Similarly, dumbbell-like MnFe2O4–
and widely used as contrast agents and drug vehicles [1,2]. NaYF4 Janus nanoparticles was synthesized via a two-step
Heterostructures composed of more than one kind of NPs, thermolysis approach. This magnetic-luminescent nanocomposite
present unique physicochemical properties from different combined functions of magnetic and luminescent materials that
nanomaterials and provide “all-in-one” excellent performance endow them bimodal imaging, magnetic field directed drug
[3]. Compared with single nanomaterial, it is facile to expand delivery, and imaging-guided cancer therapies [8]. Among these
the potential functions and applications of nanomaterials with heterostructures, Au-Fe3O4 is one of the most important bifunc-
multi-compositions. tional heterostructures.
Due to the complexity and heterogeneity of tumor tissue, Gold nanoparticles (AuNPs) are a widely used biomedical
multimodal diagnosis and multifunctionally therapeutic system probes [9] and drug carriers [10,11] due to their controllable shape
attracts more attention than ever for precise detection and and size, flexible surface chemistry, adjustable optical properties,
effective cocktail therapy [4]. Two-photon active silica-coated and biological inert [12,13]. Owing to the desirable photothermal
Au@MnO Janus particles were prepared by a seed-mediated performance of gold nanorods ([34_TD$IF]AuNRs), nanoshell, and nano-
nucleation for simultaneous magnetic and optical detection of spheres, AuNPs are also applied to achieve laser-induced PTT of
tumor cells [5]. Au-bis-pyrene@Si-PEG (Au-BP@SP), composed by cancers [14]. CT scan makes use of computer-processed combi-
inorganic gold stars and organic bis-pyrene nanoaggregates, was nations of X-ray measurements taken from different angles to
prepared to exert photothermal therapy [32_TD$IF](PTT) upon irradiation by produce cross-sectional images of specific areas of a scanned
the laser beam (808 nm) and emit strong fluorescence occurred object [15,16]. Due to the high atomic number and X-ray
from BP nanoaggregates [6]. Prussian blue (PB)@Au core-satellite absorption coefficient of gold atom, AuNPs are considered as
desirable contrast agent for CT [17]. In addition, because of their
sufficiently large optical absorption to enhance the agent’s
* Corresponding author.
photoacoustic signal, [35_TD$IF]AuNPs are also used in photoacoustic
E-mail address: dingya@cpu.edu.cn (Y. Ding). imaging (PAI) [18].

https://doi.org/10.1016/j.cclet.2018.12.006
1001-8417/ © 2018 Chinese Chemical Society and Institute of Materia Medica, Chinese Academy of Medical Sciences. Published by Elsevier B.V. All rights reserved.
1726 B. Liu et al. / Chinese Chemical Letters 29 (2018) 1725–1730

Superparamagnetic iron oxide nanoparticles (IONPs, commonly On the basis of above preparation method, the regulation on the
Fe3O4 NPs) are often used as the contrast agent of [36_TD$IF]MRI because of structure of either Au or Fe3O4 [40_TD$IF]NPs offers possible property
its high relaxivity and contrast effect [19]. Directly guided by the modulation of Au-Fe3O4 nanostructure [37,38]. Increasing the
magnetic field, Fe3O4 NPs can also be applied to chemodynamic molar ratio of iron precursor, such as Fe(CO)5, to gold, dumbbell-
therapy (CDT) via Fenton reaction under high H2O2 concentrations like morphologies of the dimer NPs can be changed into flowers
in tumor [20]. Under alternating current (AC) magnetic field, Fe3O4 having two to six Fe3O4 leaves around the gold core [39]. Increasing
NPs can generate heat for the ablation of cancers [21,22]. in the reaction time enlarged individual iron oxide domains and
The intrinsic appeal of Au-Fe3O4 heterostructures are inherited generated particles with smaller gold and larger Fe3O4. Further
not only from the unique properties of AuNPs or Fe3O4 NPs, but also modulation of the molar ratio between iron and gold precursors
from their complementary function and synergistic effect. Firstly, produced the Au-Fe3O4 and Fe3O4-Au core-shell morphologies
these two different nanoparticle domains supplied different [40,41].
surfaces to be modified for multi-anchoring functionalization. For obtaining superior in vitro and in vivo T2 relaxivity for
Next, the catalytic activity [23–28] and heat performance [29] of magnetic resonance imaging, 25 nm octahedral-shaped Fe3O4
Au-Fe3O4 heterostructures have been improved by increasing the magnetite nanocrystals are epitaxially grown on 9 nm Au seed
interface between gold and iron oxide surface. Moreover, nanoparticles using a modified wet-chemical synthesis [42].
combining these two NPs into one system, multimodal imaging Oleylamine-stabilized Au NPs as prepared firstly [43]. After that
of CT, PAI, and MRI can be achieved simultaneously [30,31] for Fe3O4 is grown on the Au seed particles referred to a previous
making up their individual technical shortcomings. Finally, the PTT method [35,39] with some modifications. 1-Octadecene was
and chemotherapy of Au-Fe3O4 heterostructures and their con- replaced by phenyl ether and the reaction time was increased
jugates would be more precise under the magnetic field guided from 45 min to 3 h allowing for a full crystallization process. The
delivery [32,33]. Thus, the Au-Fe3O4 heterostructures show strongly improved crystallinity and a highly facetted growth mode
excellent performance in [37_TD$IF]catalytic, analytical, and biomedical characterized by X-ray diffraction (XRD) patterns led to a better T2
applications, including, multimodal imaging and combination contrast in MRI as compared to other hybrids or commercial
therapy. materials. Ultraviolet-visible spectroscopy declared the existence
of [35_TD$IF]AuNPs and curves of hysteresis loops revealed the magnetism of
2. Preparation of Au-Fe3O4 heterostructures Au-Fe3O4 heterostructures. The simultaneous functionalization of
fluorescence dyes and drugs on two different surfaces offered an
The methods for the preparation of Au-Fe3O4 heterostructures all-in-one platform for theranostics.
can be divided into two categories, oil phase synthesis and aqueous In the case of aqueous phase synthesis, Fe3O4 NPs were
phase synthesis. generally synthesized at first by thermal decomposition under
Using oil phase synthesis [34], [38_TD$IF]AuNPs were synthesized firstly relatively high temperature and pressure using ferric chloride as
and then iron oxide epitaxially grew on the Au seeds. Dumbbell- raw material and trisodium citrate as stabilizer [44]. And then,
like bifunctional composite nanoparticles of Au-Fe3O4 with the chloroauric acid was subsequently reduced on the surface of Fe3O4
size tuned from 2 nm to 8 nm for Au and 4 nm to 20 nm for Fe3O4 NPs by sodium borohydride to form Au-Fe3O4 heterostructures
were synthesized as the first proof of this double functional (Fig. 1B).
nanostructure [35]. The shape and size were characterized by
transmission electron microscope (TEM, Fig. 1A). The strength of 3. Au-Fe3O4 heterostructures in catalytic applications
metal-support bonding between gold nanoparticles and “nano-
engineered” Fe3O4 substrates was significantly related to the size AuNPs show high catalysis selectivity towards CO oxidation
and morphology control of a metal oxide support [36]. The shape [45], oxidation of hydrocarbons [46], and water-gas shift reactions
and size were mainly affected by the temperature for the [47], while Fe3O4 NPs display peroxidase-like activity that activates

nucleation and growth of nanoparticles, the ratio between Au H2O2 to produce [41_TD$IF]OH in the presence of Fe2+ species [48]. Due to
and Fe3O4, and the polarity of the solvent. The dumb-bell structure different surface energy of Au and Fe3O4, the electrons on the
offers an ideal model nanoparticle with two distinct surfaces and contact interface of Au-Fe3O4 heterostructures are more active.
functionalities for potential [39_TD$IF]applications. Associated with the magnetic property of Fe3O4, Au-Fe3O4
[(Fig._1)TD$IG]

Fig. 1. (A) Oil phase synthesis of dumbbell-like Au-Fe3O4 dimers and their TEM images. Reprinted with permission [35]. Copyright 2005, American Chemical Society. (B)
Water phase synthesis of Fe3O4-supported Au NPs with satellite structure and their TEM images. Reprinted with permission [44]. Copyright 2014, Elsevier.
B. Liu et al. / Chinese Chemical Letters 29 (2018) 1725–1730 1727
[(Fig._2)TD$IG]

Fig. 2. (A) The mechanism of catalytic degradation of 4-chlorophenol by Au-Fe3O4 heterostructures and (B) the catalytic degradation of 4-chlorophenol by all test systems.
Reprinted with permission [50]. Copyright 2016, Royal Society of Chemistry.

heterostructures not only exhibit synergistic catalytic ability with photodegradation of Rhodamine B (RhB) under visible light
higher activity [49,50], but also are able to be separated irradiation and the catalytic reduction of 4-nitrophenol to 4-
magnetically for recycling (Fig. 2). aminophenol by sodium borohydride at room temperature [52].
The catalytic activity of Au-Fe3O4 heterostructures with various Because most of photocatalytic processes is highly oxidative, lethal
structures has been widely studied. The bifunctional Fe3O4/Au to most microorganisms [53–55], photocatalysis is used to kill not
heterostructures with core-satellites structures exhibited high only pathogens, but also cancer cells [56–58].
performance in catalytic reduction of 4-nitrophenol [44]. The The catalytic efficiency of Au-Fe3O4 heterostructures is related
designed Au-Fe3O4@metalorganic framework (MOF) catalysts to many factors. The increase of interfaces between Au and FexOy
showed excellent functions in rapid catalytic reduction of nitro- caused by the redox pretreatments improved the catalytic
phenol to 4-aminophenol [51]. The Au/Fe3O4@hTiO2 nanospheres performance of Au-FexOy dumbbells nanoparticles in CO oxidation
exhibited favorable catalytic performance in both the [59]. After the pretreatment, FexOy shells suffered great

[(Fig._3)TD$IG]

Fig. 3. (A) Schematic representation of detected As(III) by Au-Fe3O4 heterostructures and (B) HRTEM images of Au-Fe3O4. (C) Comparison of sensitivities for SWASV detection
of As(III) at 7 nm Au, 10 nm Fe3O4, and Au-Fe3O4 SPCE. The inset compares limit of detection of test samples (LODs). (D) High-resolution XPS spectra of Au-Fe3O4 NPs after
adsorption of 10 ppm As(III). Reprinted with permission [66]. Copyright 2018, American Chemical Society.
1728 B. Liu et al. / Chinese Chemical Letters 29 (2018) 1725–1730

morphological and structural changes, becoming irregular and 5. Au-Fe3O4 heterostructures in biomedical applications
presenting lower crystallinity, which enhanced the interface in Au-
FexOy and subsequently was of benefit for the catalytic activity. The 5.1. Multimodal imaging
amount ratio of Au:[42_TD$IF]Fe3O4 is another impact factor in controlling
the catalytic capability of Au-Fe3O4 heterostructures. In the Multimodality imaging of tumor tissues is important in the
photocatalytic reaction of methylene blue, Au-Fe3O4 heterostruc- diagnosis and precise surgical navigation for tumor treatment. MRI
tures showed higher photocatalytic efficiency than pure Fe3O4 and is a medical imaging technique used in radiology to form pictures
the higher concentration of AuNPs in the range from 1 wt% to 5 wt% of the anatomy and the physiological processes of the body in both
[60]. Moreover, after iron oxides were coated on the surface of health and disease. Fe3O4 NPs are the commonly used contract
cetyltrimethylammonium bromide (CTAB)-capped [43_TD$IF]AuNRs, the agent for MRI to reflect the chemical structure information in
photocatalytic efficiency of Fe3O4 was about 1.7-fold higher than human tissues. It has great superiority for early diagnosis of tissue
Fe2O3 as more surface defects were present on the Fe3O4 shell. This necrosis, malignant disease and degenerative diseases, which
result demonstrated the impact of oxidation state of FexOy in makes the contrast of soft tissue more accurate [68,69]. AuNPs are
promoting the adsorption and activation of reagents on the surface considered as desirable contrast agent for CT and PAI [70,71].
during the catalytic reactions [41]. In addition, the catalytic Embracing both AuNPs and Fe3O4 NPs, Au-Fe3O4 heterostructures
efficiency showed close relationship with the particle size of the Au featuring a magneto-plasmonic structure show their multimodal
core and the substituent groups of the substrate. For the reduction imaging ability [72]. It has also reported that folic acid (FA)-
of nitroarenes by Au@Fe3O4 yolk-shell nanocatalyst, small-sized Au modified Au-Fe3O4 heterostructures increased the specific accu-
core NPs (e.g., 2.5 nm) in the yolk-shell nanostructures exhibited mulation in FA receptor-overexpressed cancer cells and were
superior catalytic performance for nitroarene reduction. The further used as an efficient nanoprobe for dual mode CT/MR
reduction rates of nitroarenes with electron-withdrawing groups imaging of a xenografted FAR-overexpressing tumor model [25].
were found to be 2.3–2.6 times higher than those with electron- In addition, adding other imaging agent in the system of Au-
donating groups [61]. Fe3O4 heterostructures would offer multimodal imaging functions.
When adding fluorescent reagent into the Au-Fe3O4 heterostruc-
4. Au-Fe3O4 heterostructures in analytic applications tures, it could not only simultaneously enhance the contrast effect
for both MR and CT imaging, but also be effective for both dark-
Taking advantages of heterogeneous surface of Au-Fe3O4 field light scattering and fluorescence imaging [73]. Attaching
heterostructures, more different specific moieties can be modified [99mTc(CO)3]+ fragment on the surface of either Fe3O4–Au core–
on their surface. The S Au bond stabilized on [35_TD$IF]AuNPs surface [62] shell or Fe3O4–Au dumbbell-like NPs, single photon emission
and diphosphate, hydroxamate, or catechol decorated on Fe3O4 computed tomography (SPECT) imaging could be also achieved
surface [63] facile the connection of multi-modifiers, and the [74].
magnetic properties of Fe3O4 NPs offer easy separation by Besides adding imaging probes, adjusting the structure of
magnetic fields. Thus, Au-Fe3O4 heterostructures were widely nanocomposite can also affect its imaging function [49]. Compared
applied to detect the related substances. with Au-Fe3O4 shell-core NPs, the Janus dimer of Au- Fe3O4 offered
Based on the combined the optical, electrochemical, and high availability of the iron oxide surface, which consequently gave
magnetic properties of Au-Fe3O4 heterostructures, some toxic rise to high r2 relaxivity values and the exposed iron oxide part
substances in foods or water can be detected. A primary could facilitate the reaction of potassium ferrocyanide with iron
monoclonal anti-aflatoxin antibody (anti AFB1) covalently immo- atoms, giving rise to blue staining of the sample where the
bilized on a monolayer modified gold coated quartz crystal nanoparticles were located. In addition, the Janus NPs composed of
electrode (anti AFB1/4-ATP/Au) by 4-amino thiophenol. The a branched gold nanostar could extend their use in photoacoustic
secondary rabbit-immunoglobulin antibodies (r-IgGs) tagged with and [4_TD$IF]surface enhanced Raman spectroscopy (SERS) imaging in the
core-shell structure of gold coated iron oxide nanoparticles (r-IgG- near infrared (NIR) biological window [75].
Au-Fe3O4) were used to achieve the sandwiched system and the
regeneration of the bioelectrode. This immuno sensor could 5.2. Combination therapy
regenerate about 15–16 times with 2%–3% loss of activity. Using the
electrochemical quartz crystal microbalance-cyclic voltammetry Taking advantages of the leaky tumor blood vessels, NPs tend to
technology, the prepared immuno electrode was highly promising accumulate in tumor tissues via the enhanced permeability and
for detection of AFB1 in food, such as corn flakes samples [64]. In retention (EPR) effect. In recent years, various NPs have been
addition, l-(2-mercaptoethyl)-1,2,3,4,5,6-hexanhydro-s-triazine- employed as promising carriers for the delivery of small molecule
2,4,6-trione (MTT) modified Au-Fe3O4 heterostructures was and gene drugs [76–78]. Different from the homogeneous [40_TD$IF]NPs, Au-
explored for the bimodal detection of melamine. The bimodal Fe3O4 heterostructures have two kinds of different surfaces, gold
detection was based on the aggregation of Au-Fe3O4@MTT NPs and iron oxide, and show possibility to anchor drug molecules via
from the dispersed state upon the formation of special triple more types of interactions. The aptamer–siRNA chimera con-
hydrogen bonds between melamine and MTT. This phenomenon structed by VEGF RNA aptamer and Notch3 siRNA was bonded with
caused the red shift of the absorption peak and the increase of the cationic Au-Fe3O4 NPs through electrostatic interaction [79].
spin–spin relaxation time (T2) of the water protons upon addition Herceptin and oxaliplatin can be conjugated to the surface of
of melamine, which enhanced the detection selectivity toward PEG- and dopamine-modified Au- Fe3O4 NPs through a covalent
melamine in foods [65]. Furthermore, heavy metals in water could bond [80].
be also detected by Au-Fe3O4 NP-fabricated systems. For As(III) In order to improve the selective targeting capacity to tumor,
detection, Au-Fe3O4 NPs was modified the screen-printed carbon targeting and therapeutic moieties can be modified on the surface
electrode to serve as an efficient sensing interface. The signals were of Au-Fe3O4 NPs simultaneously. Folate-conjugated gold shell
attributed to the participation of Fe(II)/Fe(III) cycle on Fe3O4 NPs coated magnetite nanoparticles were synthesized to target cancer
surface in the electrochemical reaction of As(III) redox. The cells with folate receptor that is overexpressed on the cell surface
formation of dumbbell-like Au-Fe3O4 NPs by adding Au NPs can [81]. When DNA aptamers targeting vascular endothelial growth
accelerate the redox electrocatalysis of As(III), and subsequently factor (VEGF) assembled onto the surface of Au-Fe3O4 by
enhanced the electrochemical response (Fig. 3) [66,67]. electrostatic absorption, Apt-Au-Fe3O4 could be found to bind
B. Liu et al. / Chinese Chemical Letters 29 (2018) 1725–1730 1729
[(Fig._4)TD$IG]
achieved through near-infrared absorption by the gold shell, and
simultaneously facilitated the release of the anticancer drug
doxorubicin (Dox) loaded by Fe3O4@Au nanoroses [89]. For
synergistic PTT and gene therapy, polycationic Au nanorod (NR)-
coated Fe3O4 nanosphere (Au@pDM/Fe3O4, pDM representing poly
(2-dimethyl amino)ethyl methacrylate) was used to compress
plasmid DNA into pDNA/Au@pDM/Fe3O4. The combined near-
infrared absorbance properties of Fe3O4-PDA and AuNR-pDM were
applied to photoacoustic imaging and photothermal therapy,
which achieved a trimodal imaging and combined photothermal
and gene therapy in a xenografted rat glioma nude mouse model
(Fig. 4) [90].
Due to the limited ability of near-infrared light in the
penetration of tissues, the treatment effect of PTT for tumors
located deeply in tissues would be weaken [91]. However, [46_TD$IF]MH
demonstrated the advantages of hyperthermia for deep tumors in
tissues [92]. Under the AC, the magnetic particles injected into the
tumor site can generate heat and raised the local temperature to
above 42  C, thus killing tumor cells [93]. With the high magnetic
thermal conversion efficiency of Fe3O4 [21,22], Au-Fe3O4 hetero-
structures can exert hyperthermia at the edge and in the deep of
tumors by combining PTT with MH [33]. It has been reported
recently that Au-Fe3O4 heterostructures with the size of 25 nm
showed the best thermal efficiency under AC [94].
In addition, for the dumbbell-like Au-Fe3O4 dimer, combining a
plasmonic nanosphere with a highly reactive Fe3O4 surface, may
significantly enhance the impact of X-rays on tumor tissue. So,
nitrosyl tetrafluoroborate was modified on the surface of dimer by
the ligand exchange to enabled the X-ray-induced NO release. The
Fig. 4. (A) Images of mice at various axial positions before and 5 min after production of NO radical at the Fe3O4 surface and the superoxide
intratumoral injection of pDNA/Au@PDM/Fe3O4: overlay PAI images (left), T2- radical at the Au surface accelerated the generation of peroxyni-
weighted MRI images (middle), CT images (right), where the tumor regions are
trite, and then caused DNA strand breaks and the nitration of
indicated by the white circles, (B) infrared thermal images (at different time points),
(C) the tumor growth curves, and (D) photographs of dissected tumors after various various proteins. It was the example of Au-Fe3O4 dimer in the
treatments of two weeks. Reprinted with permission [90]. Copyright 2016, Wiley application of X-ray-enhanced radiation therapy [95].
Online Library.
6. Conclusions
with SKOV-3 ovarian cancer cells with high selectivity [82]. For
delivering platin to Her2-positive tumor cells specifically, the Heterostructures integrate the properties and functions of
dumbbell-like Au-Fe3O4 were functionalized with thiol-modified various components in one platform, which greatly enhance and
platin complexes on the Au domain, and with herceptin on the expand the potential applications of these heterogeneous or hybrid
Fe3O4 domain [83]. In this case, Herceptin can treat metastatic materials. This review introduces the preparation methods and
breast cancer as an antibody drug. In addition, it also helps the biomedical applications of Au-Fe3O4 heterostructures in recent
preferred targeting of platin complexes to Sk-Br3 cells that are years. Au-Fe3O4 heterostructures enhance catalytic activity of
Her2-positive breast cancer cells as opposed to Her2-negative AuNPs when the interface between Au and Fe3O4 increased. The
MCF-7 breast cancer cells. multimodal imaging of MRI, CT, and PAI by Au-Fe3O4 hetero-
Inorganic heterogeneous nanoparticles are capable of combin- structures is expected to improve the accuracy of surgical
ing the advantages of inorganic nanoparticles (e.g.,[45_TD$IF] imaging, navigation. Utilizing the magnetic targeting function of Fe3O4
magnetic guided delivery, magnetic hyperthermia (MH) for NPs, drug loaded on Au-Fe3O4 NPs could be delivered precisely, and
magnetic nanoparticles or photoablation therapy for plasmonic then released effectively with the help of photothermal conversion
nanoparticles) with drug delivery/therapy, which is highly of Au or magnetic thermal conversion of Fe3O4. Based on the
attractive to theranostics [28,40,84]. The structure of Au-Fe3O4 individual functions coming from [35_TD$IF]AuNPs and Fe3O4 NPs, the
heterostructures can be adjusted to proper optical properties to structure combination of these two heterostructures provides an
obtain high photothermal conversion efficiency of AuNPs [85,86]. excellent “all-in-one” system encompassing enhanced catalytic
Irradiated by the laser, the Au-Fe3O4 heterostructures can generate activity, multimodal imaging, and combination therapy.
heat and raise the local temperature. This is beneficial for the
release of drugs embedded in thermosensitive materials, such as Acknowledgments
the release of Ampicillin from AuNPs decorated Fe3O4@poly(N-
isopropylacrylamide-co-acrylamide) [87]. This work was supported by grants from the National Natural
Although chemotherapy is one of the most important approach Science Foundation of China (Nos. 31870946, 31470916). We thank
in cancer treatment, it is limited by drug resistance and systematic Prof. Yanglong Hou at Perking University for his guidance on
toxicity. Based on the attractive photothermal effect of plasmonic writing this article.
nanomaterials, the combination of photothermal agents and
chemotherapy is a promising approach to achieve enhanced References
cancer killing efficiency through the synergistic effect [88]. As
such, Fe3O4@Au nanorose was fabricated. The inner Fe3O4 core [1] T. Cui, J.J. Liang, H. Chen, et al., ACS Appl. Mater. Interface 9 (2017) 8569–8580.
[2] R. Hao, J. Yu, Z.G. Ge, et al., Nanoscale 5 (2013) 11954–11963.
functioned as the MR imaging agent. The photothermal effect was
1730 B. Liu et al. / Chinese Chemical Letters 29 (2018) 1725–1730

[3] F. Oyarzun-Ampuero, A. Vidal, M. Concha, et al., Curr. Pharm. Des. 21 (2015) [49] C.J. Jin, J. Han, F.Y. Chu, X.X. Wang, R. Guo, Langmuir 33 (2017) 4520–4527.
4329–4341. [50] J. Liu, Z.W. Zhao, Z.X. Ding, Z.D. Fang, F.Y. Cui, RSC Adv. 6 (2016) 53080–53088.
[4] S. Wang, J. Lin, Z.T. Wang, et al., Adv. Mater. 29 (2017) 1701013. [51] F. Ke, L.H. Wang, J.F. Zhu, Nanoscale 7 (2015) 1201–1208.
[5] I. Schick, S. Lorenz, D. Gehrig, et al., J. Am. Chem. Soc. 136 (2014) 2473–2483. [52] J.C. Cheng, S.L. Zhao, W.B. Gao, P.B. Jiang, R. Li, React. Kinet. Mech. Catal. 121
[6] P.P. Yang, Y.G. Zhai, G.B. Qi, et al., Small 12 (2016) 5423–5430. (2017) 797–810.
[7] Y. Dou, X. Li, W.T. Yang, et al., ACS Appl. Mater. Interface 9 (2017) 1263–1272. [53] A. Fakhri, M. Naji, J. Photochem. Photobiol. B 167 (2017) 58–63.
[8] Q. Wu, Y.N. Lin, F.J. Wo, et al., Small 13 (2017)1701129. [54] Y.Y. Song, H.J. Jiang, H.K. Bi, et al., ACS Omega 3 (2018) 973–981.
[9] B. Ankudze, A. Philip, T.T. Pakkanen, Sensor. Actuat. B-Chem. 265 (2018) 668– [55] Y. Wang, H.B. Fang, Y.Z. Zheng, et al., Nanoscale 7 (2018) 19118–19128.
674. [56] T.Y. Lai, W.C. Lee, J. Photochem. Photobiol. A 204 (2009) 148–153.
[10] Y. Ding, Z. Jiang, K. Saha, et al., Mol. Ther. 22 (2014) 1075–1083. [57] Z. Li, X.B. Pan, T.L. Wang, et al., Nanoscale Res. Lett. 8 (2013) 96.
[11] J.J. Liang, Y.Y. Zhou, J. Wu, Y. Ding, Curr. Drug Met. 15 (2014) 620–631. [58] X.H. Feng, S.K. Zhang, X. Lou, Colloid Surf. B 107 (2013) 220–226.
[12] M.C. Daniel, D. Astruc, Chem. Rev. 104 (2004) 293–346. [59] L. Souza da Costa, D. Zanchet, Catal. Today 282 (2017) 151–158.
[13] C.M. Cobley, J. Chen, E.C. Cho, L.V. Wang, Y. Xia, Chem. Soc. Rev. 40 (2011) 44– [60] A. Mahmood, S.M. Ramay, Y.S. Al-Zaghayer, et al., Desalin. Water Treat. 57
56. (2015) 20069–20075.
[14] S.S. Xing, X.W. Zhang, L.Y. Luo, et al., Nanotechnology 29 (2018) 405101. [61] F.H. Lin, R.A. Doong, J. Phys. Chem. C 121 (2017) 7844–7853.
[15] J. Kirchner, L.M. Sawicki, F. Nensa, et al., Eur. J. Nucl. Med. Mol. Imaging 46 [62] Y.F. Chen, J. Hong, D.Y. Wu, et al., RSC Adv. 6 (2016) 8336–8345.
(2019) 437–445. [63] J.F. Zeng, L.H. Jing, Y. Hou, et al., Adv. Mater. 26 (2014) 2694–2698.
[16] B.W. Lan, X.J. Li, Acad. Radiol. (2018), doi:http://dx.doi.org/10.1016/j. [64] R. Chauhan, J. Singh, P.R. Solanki, et al., Biochem. Eng. J. 103 (2015) 103–113.
acra.2018.05.002. [65] J.C. Shen, Y. Yang, Y. Zhang, et al., Sensor. Actuat. B-Chem. 226 (2016) 512–517.
[17] K.T. Butterworth, S.J. McMahon, F.J. Currell, K.M. Prise, Nanoscale 4 (2012) [66] S.S. Li, W.Y. Zhou, M. Jiang, et al., Anal. Chem. 90 (2018) 4569–4577.
4830–4838. [67] J. Wei, S.S. Li, Z. Guo, et al., Anal. Chem. 88 (2016) 1154–1161.
[18] J.V. Jokerst, A.J. Cole, D. Van de Sompel, S.S. Gambhir, ACS Nano 6 (2012) [68] H. Zhang, X.T. Tian, Y. Shang, Y.H. Li, X.B. Yin, ACS Appl. Mater. Interface 10
10366–10377. (2018) 28390–28398.
[19] T. Wang, Y. Hou, B. Bu, et al., Small 14 (2018) 1800573. [69] R. Zhou, P. Bagga, K. Nath, et al., Cancer Res. 78 (2018) 5521–5526.
[20] C. Zhang, W.B. Bu, D.L. Ni, et al., Angew. Chem. Int. Ed. 55 (2016) 2101–2106. [70] X.H. Liu, C.H. Gao, J.H. Gu, et al., ACS Appl. Mater. Interface 8 (2016) 27622–
[21] K.S. Sharma, R.S. Ningthoujam, A.K. Dubey, et al., Sci. Rep. 8 (2018) 14766. 27631.
[22] G.K. Thirunavukkarasu, K. Cherukula, H. Lee, et al., Biomaterials 180 (2018) [71] J.Q. Xi, W.J. Wang, L.Y. Da, et al., ACS Biomater. Sci. Eng. 4 (2018) 1083–1091.
240–252. [72] J. Zhu, Y.J. Lu, Y.G. Li, et al., Nanoscale 6 (2014) 199–202.
[23] H. Kang, H.J. Jung, D.S.H. Wong, et al., J. Am. Chem. Soc. 140 (2018) 5909–5913. [73] W.J. Dong, Y.S. Li, D.C. Niu, et al., Small 9 (2013) 2500–2508.
[24] W. Shi, X.Y. Liu, C. Wei, et al., Nanoscale 7 (2015) 17249–17253. [74] M. Felber, R. Alberto, Nanoscale 7 (2015) 6653–6660.
[25] H.D. Cai, K.A. Li, J.C. Li, et al., Small 11 (2015) 4584–4593. [75] J. Reguera, D.J. de Aberasturi, M. Henriksen-Lacey, et al., Nanoscale 9 (2017)
[26] Q. Gao, Y. Xing, M.L. Peng, et al., Chin. J. Chem. 35 (2017)1700032. 9467–9480.
[27] F. Pang, R.F. Zhang, D.P. Lan, J.P. Ge, ACS Appl. Mater. Interface 10 (2018) 4929– [76] R.H. Jin, Z.N. Liu, Y.K. Bai, Y.S. Zhou, X. Chen, ACS Omega 3 (2018) 4306–4315.
4936. [77] S. Lee, A. Stubelius, N. Hamelmann, V. Tran, A. Almutairi, ACS Appl. Mater.
[28] L. Shang, Y.H. Liang, M.Z. Li, et al., Adv. Funct. Mater. 27 (2017) 1606215. Interface 10 (2018) 40378–40387.
[29] P. Guardia, S. Nitti, M.E. Materia, et al., J. Mater. Chem. B 5 (2017) 4587–4594. [78] S.H. Jeong, J.H. Jang, H.Y. Cho, Y.B. Lee, Arch. Pharm. Res. 41 (2018) 797–814.
[30] Y. Hu, J. Yang, P. Wei, et al., J. Mater. Chem. B 3 (2015) 9098–9108. [79] Y. Chen, M.J. Xu, Y. Guo, et al., Nanotechnology 28 (2017) 025101.
[31] J.C. Li, L.F. Zheng, H.D. Cai, et al., ACS Appl. Mater. Interface 5 (2013) 10357– [80] D.R. Liu, X.W. Li, C.L. Chen, et al., Oncol. Lett. 15 (2018) 8079–8087.
10366. [81] S. Karamipour, M.S. Sadjadi, N. Farhadyar, Spectrochim. Acta A 148 (2015) 146–
[32] J.R. Peng, T.T. Qi, J.F. Liao, et al., Theranostics 4 (2014) 678–692. 155.
[33] D. Yan, X. Liu, G. Deng, et al., J. Colloid Interface Sci. 530 (2018) 547–555. [82] Z. Jian, K.Y. Tu, Y.L. Liu, et al., Mater. Sci. Eng. C 80 (2017) 88–92.
[34] C. Wang, C.J. Xu, H. Zeng, S.H. Sun, Adv. Mater. 21 (2009) 3045–3052. [83] C. Xu, B. Wang, S. Sun, J. Am. Chem. Soc. 131 (2009) 4216–4217.
[35] H. Yu, M. Chen, P.M. Rice, et al., Nano Lett. 5 (2005) 379–382. [84] B.H. Wu, S.H. Tang, M. Chen, N.F. Zheng, Chem. Commun. 50 (2014) 174–176.
[36] C.W. Han, T. Choksi, C. Milligan, et al., Nano Lett. 17 (2017) 4576–4582. [85] J.F. Ren, S. Shen, Z.Q. Pang, et al., Chem. Commun. 47 (2011) 11692–11694.
[37] W.L. Shi, H. Zeng, Y. Sahoo, et al., Nano Lett. 6 (2006) 875–881. [86] X.J. Chen, G.L. Li, Q.H. Han, et al., Chemistry 23 (2017) 17204–17208.
[38] F.H. Lin, R.A. Doong, J. Colloid Interface Sci. 417 (2014) 325–332. [87] C.N. Wang, Y.Y. Wang, Y.L. Jin, et al., J. Nanosci. Nanotechnol. 15 (2015) 6784–
[39] Y.H. Wei, R. Klajn, A.O. Pinchuk, B.A. Grzybowski, Small 4 (2008) 1635–1639. 6789.
[40] J. Canet-Ferrer, P. Albella, A. Ribera, J.V. Usagre, S.A. Maier, Nanoscale Horiz. 2 [88] X.Y. Nan, X.J. Zhang, Y.Q. Liu, et al., ACS Appl. Mater. Interface 9 (2017) 9986–
(2017) 205–216. 9995.
[41] Y. Li, J.W. Zhao, W.L. You, D.H. Cheng, W.H. Ni, Nanoscale 9 (2017) 3925–3933. [89] C.M. Li, T. Chen, I. Ocsoy, et al., Adv. Funct. Mater. 24 (2014) 1772–1780.
[42] M.V. Efremova, V.A. Naumenko, M. Spasova, Sci. Rep. 8 (2018) 11295. [90] Y. Hu, Y.Q. Zhou, N.N. Zhao, F.S. Liu, F.J. Xu, Small 12 (2016) 2459–2468.
[43] X.O. Liu, M. Atwater, J.H. Wang, Q. Huo, Colloid. Surf. B 58 (2007) 3–7. [91] T. Suto, M. Ito, T. Uehara, et al., Int. Congress Ser. 1232 (2002) 383–388.
[44] F. Yan, R.Y. Sun, Mater. Res. Bull. 57 (2014) 293–299. [92] Z. Hedayatnasab, F. Abnisa, W.M.A.W. Daud, Mater. Des. 123 (2017) 174–196.
[45] Y. He, J.C. Liu, L.L. Luo, et al., Proc. Natl. Acad. Sci. U. S. A. 115 (2018) 7700–7705. [93] O.L. Gobbo, K. Sjaastad, M.W. Radomski, Y. Volkov, A. Prina-Mello, Theranostics
[46] R. Ye, A.V. Zhukhovitskiy, R.V. Kazantsev, et al., J. Am. Chem. Soc. 140 (2018) 5 (2015) 1249–1263.
4144–4149. [94] M.V. Efremova, Y.A. Nalench, E. Myrovali, et al., Beilstein J. Nanotechnol. 9
[47] M. Yang, L.F. Allard, M. Flytzani-Stephanopoulos, J. Am. Chem. Soc. 135 (2013) (2018) 2684–2699.
3768–3771. [95] S. Klein, C. Harreiss, C. Menter, et al., ACS Appl. Mater. Interface 10 (2018)
[48] Y.L. Dai, Z. Yang, S.Y. Cheng, et al., Adv. Mater. 30 (2018) 1704877. 17071–17080.

You might also like