You are on page 1of 25

REVIEW

Cancer Therapy www.advmat.de

Emerging Strategies of Nanomaterial-Mediated


Tumor Radiosensitization
Jiani Xie, Linji Gong, Shuang Zhu, Yuan Yong, Zhanjun Gu,* and Yuliang Zhao

1. Introduction
Nano-radiosensitization has been a hot concept for the past ten years, and
the nanomaterial-mediated tumor radiosensitization method is mainly Over 14 million new cases and more
than 8 million cancer-caused deaths
focused on increasing intracellular radiation deposition by high atomic
were reported worldwide in 2012, and the
number (high Z) nanomaterials, particularly gold (Au)-mediated radiation number of cancer-caused death per year
enhancement. Recently, various new nanomaterial-mediated radiosensitive was projected to raise by 45% from 2007
approaches have been successively reported, such as catalyzing reactive to 2030 according to the World Health
oxygen species (ROS) generation, consuming intracellular reduced glu- Organization (WHO).[1,2] In today’s society,
despite the massive financial and intellec-
tathione (GSH), overcoming tumor hypoxia, and various synergistic radio-
tual investments are put into the preven-
therapy ways. These strategies may open a new avenue for enhancing the tion and treatment of cancer, cancer is still
radiotherapeutic effect and avoiding its side effects. Nevertheless, reviews one of the world’s leading causes of death
systematically summarizing these newly emerging methods and their to threaten human health. Therefore, it is
radiosensitive mechanisms are still rare. Therefore, the general strategies highly desired to find better cancer thera-
of nanomaterial-mediated tumor radiosensitization are comprehensively peutic approaches to complement and
improve current cancer treatments.
summarized, particularly aiming at introducing the emerging radiosensitive
Radiotherapy (RT) is such an effec-
methods. The strategies are divided into three general parts. First, methods tive cancer therapeutic tool and has been
on account of the intrinsic radiosensitive properties of nanoradiosensitizers extensively applied in current clinical
for radiosensitization are highlighted. Then, newly developed synergistic cancer therapy. According to the reports,
strategies based on multifunctional nanomaterials for enhancing radio- over 50% cancer patients are treated with
RT either alone or cooperated with other
therapy efficacy are emphasized. Third, nanomaterial-mediated radiopro-
types of therapies.[3–5] The therapeutic
tection approaches for increasing the radiotherapeutic ratio are discussed. rationale of RT is that high-energy ion-
Importantly, the clinical translation of nanomaterial-mediated tumor radio- izing radiations (such as X-rays and γ-rays)
sensitization is also covered. Finally, further challenges and outlooks in this directly interact with cellular DNA to cause
field are discussed. DNA damage (DNA is the major target
which determines the radiobiological
effects[3]), or indirectly, react with water
molecule to produce ROS (such as superoxide O2•−, hydrogen
peroxide H2O2, hydrogen radical H•, hydroxyl radical HO•, and
H2O•+) to destroy DNA or other cellular components to induce
J. Xie, L. Gong, S. Zhu, Dr. Y. Yong, Prof. Z. Gu, Prof. Y. Zhao cell apoptosis and necrosis.[6,7] As a classical cancer therapy
CAS Key Laboratory for Biomedical Effects of Nanomaterials
and Nanosafety
method, RT has its specific superiority, such as no tissue depth
Institute of High Energy Physics restriction.[8] However, some intrinsic shortcomings of RT also
Chinese Academy of Sciences limit its long-term application, which mainly include three
Beijing 100049, China aspects as follows: 1) The ionizing radiations used for killing
E-mail: zjgu@ihep.ac.cn tumor may harm neighboring normal tissues and thus lead
Prof. Z. Gu, Prof. Y. Zhao to severe toxic side effects to body.[9] 2) The hypoxic nature
College of Materials Science and Optoelectronic Technology
University of Chinese Academy of Sciences inside the tumor microenvironment (TME) may make tumor
Beijing 100049, China cells resistant to radiation, ultimately resulting in the failure of
Prof. Y. Zhao tumor eradication.[10,11] 3) Since elevated antioxidants seems to
CAS Center for Excellence in Nanoscience be a general feature of cancer phenotype, antioxidant system
National Center for Nanoscience and Technology of China within cancer cells will prevent them against these damaging
Chinese Academy of Sciences
Beijing 100190, China
effects through quenching excessive free radicals. For example,
the intracellular glutathione (GSH) concentration in cancer
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/adma.201802244. cells is about four fold than that within normal cells,[12,13] which
has strong ability to scavenge the ROS before they arrival at the
DOI: 10.1002/adma.201802244 target site and finally reduce the radiotherapeutic efficiency to

Adv. Mater. 2018, 1802244 1802244  (1 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

a large extent. As a result, these defects discount the efficiency


of RT. Therefore, the purpose of radiosensitization aims to Jiani Xie is a graduate stu-
improve the RT therapeutic efficacy. dent of the Key Laboratory
In the last two decades, the rapid development of emerging for Biomedical Effects
advanced nanomaterials and nanobiotechnology provides a of Nanomaterials and
promising opportunity for tumor radiosensitization. A variety Nanosafety of the Institute of
of nanomaterials have been widely studied and applied in the High Energy Physics, Chinese
enhancement of radiotherapeutic efficacy attributed to their ver- Academy of Sciences, under
satile physicochemical properties, such as good biocompatibility, the supervision of Prof.
intrinsic radiosensitive activities, highly loading abilities of mul- Zhanjun Gu. She obtained
tiple types of drugs[14] as well as the enhanced permeability and her bachelor’s degree from
retention (EPR) effects in tumor tissue.[15] Previously, the nano- Sichuan Normal University,
material-mediated radiosensitization method is mainly focused China, in 2014. She is now
on enhancing the intracellular radiation energy deposition by studying the synthesis and biomedical applications of
high Z nanoradiosensitizers. In recent years, a large number of nanomaterials.
new nanoradiosensitizers and radiosensitive strategies are con-
tinuously proposed. These nanoradiosensitizers and radiosensi- Zhanjun Gu is a professor
tive strategies not only can enhance ionizing radiation energy in the Key Laboratory
deposition, but also have the ability to catalyze ROS genera- for Biomedical Effects
tion, manipulate the TME (such as reducing intracellular GSH of Nanomaterials and
concentration, converting low toxic intracellular H2O2 to high- Nanosafety of the Institute
active HO• as well as improving oxygen level in tumor) or tune of High Energy Physics,
the cell circle/signal pathway to make cancer cells more sensi- Chinese Academy of Sciences.
tive to radiation, and thus significantly enhance the RT thera- He obtained his Ph.D.
peutic efficacy and simultaneously reduce its side effects. For degree from the Institute of
example, the photocatalytic semiconductor nanoparticles can be Chemistry, Chinese Academy
activated by X-ray to facilitate ROS generation so as to enhance of Science, in 2007 under the
radiation effects.[16] Some of the nanomaterials have the ability direction of Prof. Jiannian
to deplete GSH in tumor and consequently prevent the scav- Yao. He carried out postdoctoral research at the University
enging of radiation-induced ROS from GSH, finally improving of Georgia in 2009 in the group of Prof. Zhengwei Pan.
the treatment outcomes.[17] A few iron (Fe)/copper (Cu)-based His current research interests focus on nanomaterials
nanomaterials can convert intracellular H2O2 into high reactive synthesis and biomedical applications.
HO• to damage cancer cells and thus enhance the RT thera-
peutic effects.[18,19] The nanomaterials with the ability of oxygen
level improvement can effectively overcome the tumor hypoxia Yuliang Zhao is the founder
to reduce radioresistance and eventually elevate radiation of the Key Laboratory
efficiency.[20] And some nanomaterials are able to arrest cancer for Biomedical Effects
cells at G2/M phase (the most sensitive phase to radiation) and of Nanomaterials and
hence enhance radiation sensitivity of cells.[21] Apart from these Nanosafety, and Professor
intrinsic radiosensitive properties, due to the large surface area and Deputy Director-General
and multifunction features, the nanoparticles can act as carriers of National Center for
for loading and delivering radiosensitive therapeutic agents Nanoscience and Technology
(such as some of the chemotherapeutic drugs, gene materials, of China. He was elected as a
photosensitizers (PSs), and photothermal agents) into tumors Member of Chinese Academy
to achieve synergistic RT sensitization effect. In addition, the of Sciences in 2017. His
nano­­particles with radioprotective function for healthy tissue can research interests focus on
also enhance the RT efficiency, because they can decrease side nanotoxicological chemistry and nanomedicine.
effects of RT to healthy tissues, and thus elevate the therapeutic
ratio under higher dose radiation.[5]
Currently, although plenty of new radiosensitive approaches to guide researchers to promote the development of this field.
based on nanomaterials are reported, most review articles still Therefore, in this review, we systematically sum up the gen-
focus on the high Z nanomaterials mediated radiosensitiza- eral strategies of nanomaterial-mediated radiosensitization
tion,[22–28] particularly Au-mediated RT enhancement.[3,29–43] by far, and narrate their radiosensitive principles and applica-
The systematic summarization of the newly emerging nano- tions. We divide these methods into three general parts: 1) The
material-mediated RT enhancement strategies, such as the enhancement of radiosensitivity by nanoradiosensitizers with
new nanoradiosentizers and radiosensitive methods men- intrinsic radiosensitive properties. 2) The improvement of the
tioned above, is still rare. Thus, it is necessary and important cancer therapeutic outcomes via synergy of other therapy and
to make a new summarization of the most recent advances in RT based on nanocarriers. 3) The elevation of the therapeutic
this field to review its growing tendency and more importantly, ratio through protecting healthy tissues from radiation-caused

Adv. Mater. 2018, 1802244 1802244  (2 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

injury by nanoradioprotectors. Moreover, some nanoradio- According to a series of studies, the nanomaterials with
sensitizers have come into clinical transformation stage, indi- intrinsic radiosensitive properties not only can enhance ion-
cating the great potential of current nanoradiosensitizers on izing radiation energy deposition, but also have the ability to
clinical application. Thus, current state of the art of the clinical catalyze ROS generation, regulate the TME, and manipulate
translation of nanoradiosensitizers is also introduced in this cancer cell circles/signal path ways to make cancer cells more
review. Finally, we further discuss the current challenges and sensitive to radiation-induced damage. Herein, we summarize
future research directions in this field. It is anticipated that the common intrinsic radiosensitive properties of the nanora-
such a review article can assist researchers in understanding diosensitizers as follows (Scheme 1): 1) The high Z nanomate-
the latest progresses of nanomaterial-based radiosensitization, rials can promote the radiation energy deposition in tumor to
and encourage all of us to improve the existing approaches or increase the RT efficiency. 2) The semiconductor nanomaterials
explore new ways for radiosensitization. with photocatalytic function can be activated by light to gen-
erate free radicals for enhancing radiation effects. 3) The Fe/
Cu-based nanomaterials with chemical catalytic properties can
2. General Strategies of Nanomaterial-Mediated catalyze H2O2 to produce toxic HO• for tumor radiosensitiza-
tion. 4) The nanomaterials which can deplete GSH in cancer
Tumor Radiosensitization
cells will prevent the consumption of ROS and thus improve
2.1. The Intrinsic Radiosensitive Properties of the RT effect. 5) The nanomaterials that raise the oxygen level
Nanoradiosensitizers for Tumor Radiosensitization in the TME can overcome the tumor hypoxia and thus reduce
radioresistance to realize radiosensitization. 6) The nanomate-
The multifunctional properties of nanomaterials make rials which arrest cells at G2/M phase can enhance radiation
them suitable for a wide range of biomedicine applications. sensitivity of cells. In this section, we will detailedly introduce

Scheme 1. Scheme of the general intrinsic radiosensitive properties of nanoradiosensitizers. Energy deposition: Adapted with permission.[42]
Copyright 2017, Royal Society of Chemistry. Photocatalysis: Adapted with permission.[90] Copyright 2017, Wiley-VCH. Chemcatalysis: 1) iron oxide:
Adapted with permission.[18] Copyright 2016, Elsevier. 2) Cu3BiS3: Adapted with permission.[19] Copyright 2017, Royal Society of Chemistry. Depleting
intracellular GSH: Adapted with permission.[17] Copyright 2017, American Chemical Society. Improving tumor oxygen: Adapted with permission.[117]
Copyright 2017, Elsevier. Regulating cell cycle: Adapted with permission.[119] Copyright 2014, American Chemical Society.

Adv. Mater. 2018, 1802244 1802244  (3 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

the radiosensitive mechanisms and applications of these kinds optimum delivery and effect, multiform modification of tar-
of nanoradiosensitizers on RT enhancement. geting moieties for specifically delivering to the tumor tissue
and longer systemic circulation for providing the material with
enough time to be absorbed by tumor tissue.[7] A large amount
2.1.1. Increasing the Intracellular Radiation Energy of articles about Au-induced tumor radiosensitization can be
Deposition by High Z Nanoradiosensitizers searched.[44–58] For example, Zhang et al. prepared ultrasmall
GSH-Au nanomolecules for increasing the safety and efficacy
The main mechanism for developing high Z nanoradiosensi- of RT.[48] The radiosensitization came from the direct interac-
tizers is on account of the difference in the energy absorbing tion between Au and radiation. When the GSH-Au nanomol-
abilities between high Z nanomaterials and soft tissues. There ecules received high-energy radiation, they would become a
are much more X-ray energy absorption of high Z atoms in new source of radiation and emit high energy electrons to
nanoparticles than that of low Z atoms in soft tissues (soft cause radiochemicals (ionizations and free radicals) within the
tissues are mainly consisted of organic matters with low Z), cancer cells that could damage and kill the cells. Meanwhile,
allowing physical energy dose deposition enhancement in the increased targeting specificity and tumor uptake of GSH-Au
the presence of high Z nanoparticles.[3] In a detailed process, nanomolecules in cancer cells via the improved EPR effect
when ionizing radiation hits high Z nanoparticles, the high Z could increase the accumulation of Au nanomolecules in cancer
atoms can absorb more photons to increase the local dose of cells and thus further enhance the radiosensitization effects.
radiation in cells. Then, the interaction of photons with high Yang et al. reported the enhancement of RT on cancer cells by
Z nanoparticles will induce the occurrence of multiple effects, lipid nanocapsules (NCs) mediated delivery of amphiphilic Au
such as the Compton effect and photoelectric effect. These nanoparticles (amph-NPs) into intracellular membranes.[46] As
effects can lead to various emissions, such as photoelectrons, seen in Figure  1a, the amph-NPs were embedded in the walls
Compton electrons, and Auger electrons. Subsequently, the of NCs and formed Au-NCs. And such a formation could pro-
emissive electrons either directly damage DNA or indirectly mote the transfer of amph-NPs into intracellular membranes.
interact with water to produce ROS to damage DNA to induce Clonogenic assays demonstrated that Au-NCs strengthened
cell apoptosis and necrosis (Scheme 2).[6,7,42] radiotherapeutic killing of cancer cells compared to that of
Owing to the special radiosensitive effects of high Z nano- radiation alone (Figure 1b,c). Zaki et al. used polymeric micelle-
materials, many researches have devoted great effort to apply encapsulated Au nanoparticles for RT enhancement, where the
high Z nanomaterials with good biocompatibility and tumor median survival time of tumor-bearing mice treated by both
targeting ability for improving the therapeutic outcome of of the Au and radiation showed a 1.7-fold improvement com-
RT. Here, we take the most popular high Z nanomaterials pared to that of mice receiving radiation alone.[45] Attributed to
(including Au-, gadolinium (Gd)-, bismuth (Bi)-, and tungsten the positive radiosensitive response of Au-based nanomaterials,
(W)-based nanomaterials) in the field of tumor radiosensitiza- researchers attempted to evaluate the radiosensitive efficacy of
tion as paradigms to manifest the promising potential of high Au-based nanoradiosensitizers with different physicochemical
Z nanoradiosensitizers on RT enhancement. What needs to be parameters (such as size and shape) so as to obtain the optimal
specifically mentioned is that attributing to lots of works about sensitization condition.[44,51,58,59] As a representative example,
hafnium (Hf)-based nanomaterials for cancer radiosensitiza- Zhang et al. observed that 12.1 and 27.3 nm poly(ethylene
tion have entered clinical trials, we will introduce them at the glycol) (PEG)-coated Au nanoparticles had higher radiosensi-
next section of “The clinical translation of nanomaterial-medi- tive effects than 4.8 and 46.6 nm nanoparticles, which indicated
ated tumor radiosensitization,” and thus will not repeat the that the radiosensitization of the PEG-coated Au nanoparticles
statement about them here. was size-dependent.[44]
Au-based nanomaterials are the hottest researched radiosen- Gd is a lanthanide element with powerful high energy ioni-
sitizers due to their unique advantages such as chemical inert- zation radiation absorption ability and the Gd-based nano-
ness, highly biocompatibility, varied morphology for achieving materials are widely developed as radiosensitizers in recent

Scheme 2.  The schematic illustration of the RT enhancement effect by high Z nanoradiosensitizers.

Adv. Mater. 2018, 1802244 1802244  (4 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 1.  a) Scheme of the structure of Au-NCs and amph-NPs. b) colonies assay of B16F10 cells after different treatments. c) Responding survival
fraction of B16F10 and 4T1 cells with different treatments. Adapted with permission.[46] Copyright 2014, American Chemical Society.

years.[60–68] As a paradigm, Le Duc et al. used ultrasmall Gd- to demonstrate the radiosensitive effect of Bi2S3/poly(lactic-co-
based nanoparticles (GBNs) to carry microbeam RT. The glycolic acid) (PLGA) nanocapsule against prostate cancer.[69] For
radiosensitive effect of GBNs activated by X-ray microbeams another example, Bi2Se3 has attracted wide attention in biomed-
significantly improved the survival of rats bearing brain ical applications due to the large X-ray absorption ability of Bi
tumors.[60] Miladi et al. studied the radiosensitive effect of GBNs and the effective anticancer activity of selenium (Se). Zhang et al.
for human radioresistant head and neck squamous cell carci- reported a poly(vinylpyrollidone) (PVP)-coated Bi2Se3 nano-
noma (HNSCC). Combining the GBNs with irradiation could plates for enhancing RT on cancer.[70] The net increase of
significantly decrease the HNSCC cell survival and delay tumor tumor weight was minimum after treating with Bi2Se3  + RT,
growth.[64] Dufort et al. developed the Gd-based theranostic indicating that Bi2Se3 nanoparticles possess a strong radiation
nanoparticles (USRPs) for lung tumor radiosensitization.[62] An enhancement effect. In addition, Du et al. recently synthesized
effective radiosensitive effect of USRPs could be certified by the hyaluronic acid-modified bismuth oxide nanoparticles (HA-
longer mean survival time of 112 days for irradiated H358-Luc Bi2O3 NPs) to use as adjuvants for cancer radiosensitization.[73]
lung tumor-bearing mice with USRPs treatment compared to The HA-Bi2O3 NPs with high X-ray attenuation efficiency exhib-
that of 77 days for the irradiated mice without USRPs treat- ited an ideal radiosensitivity to induce cell apoptosis and inhibit
ment. Most recently, Du et al. engineered the Gd-doped carbon tumor growth. Thus, Bi-based nanomaterials manifest the great
dots (Gd-doped CDs) for tumor RT treatment.[68] Compared to promise for RT enhancement.
treating tumor-bearing mice with radiation alone, injecting the W-based nanomaterials also hold the great attention in radio-
Gd-doped CDs to the mice during the radiation process remark- sensitization field in recent years.[76–79] For example, Yong et al.
ably inhibited the growth of the solid tumors. Consequently, reported a new ultrasmall bovine serum albumin (BSA)-coated
Gd-based nanomaterials could be envisioned as the powerful GdW10O36 nanoclusters (GdW10@BSA NCs) as radiosensi-
radiosensitizers to assist RT. tizers for RT treatment.[76] The tumors of the mice in group
Bi-based nanomaterials, such as Bi2S3, Bi2Se3 and Bi2O3, are GdW10@BSA NCs + X-ray were thoroughly eliminated at
another hot materials in radiosensitization.[69–75] For example, 4 days and showed no recurrence in the subsequent 14 days,
Yao et al. provided the first example of both in vitro and in vivo indicating the strong sensitization effect of the NCs on RT.

Adv. Mater. 2018, 1802244 1802244  (5 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

For another example, Shen et al. developed an ultrasmall coor- For example, Zhang et al. investigated the combined application
dination polymer nanodot (CPN) by the coordination of gallic of ZnO nanorods with anticancer agent daunorubicin (DNR)
acid (W-GA) and tungsten ions (WVI) (W-GA-PEG CPNs) for in PDT.[80] The ZnO nanorods could not only act as the carrier
enhancing RT.[77] Taking advantage of the radiosensitive func- to deliver DNR for cancer therapy, but also further enhance
tion of W with strong X-ray absorption, the W-GA-PEG CPNs the inhibition of cancer cells under UV irradiation by photo-
greatly inhibited the tumor growth under X-ray exposure. As a catalysis. Recently, inspired by the ROS generation ability of
result, W-based nanomaterials also have the potential for tumor semiconductors under UV irradiation, researchers attempted to
radiosensitization. design and fabricate one kind of special photocatalytic semicon-
ductor nanomaterial that could be excited by X-ray radiation to
generate ROS for improving RT treatment effect. For example,
2.1.2. Promoting ROS Generation through Photocatalysis Ghaemi et al. designed lanthanide-doped ZnO nanoparticles
by Semiconducting Nanoradiosensitizers (europium (Eu)/Gd-doped ZnO nanoparticles) to enhance the
RT of cancer.[16] The RT enhancement mechanism of this nano-
Photocatalytic semiconductor nanomaterials have also been particle is illustrated in Figure  2a. Lanthanides could interact
exploited to enhance radiation effects, because they can be with X-ray radiation, and an efficient relaxation of lanthanides
activated by light to generate the toxic free radicals. In detail, electronic excitation occurred via the transfer of excess energy
when the semiconductors are excited by light with energy levels to ZnO nanoparticles. When the transferred energy levels were
higher than their band gap, the electron-hole pairs are formed. higher than that of the ZnO band gap, electron–hole pairs were
Subsequently, the free radicals will continuously generate from formed and consequently ROS were generated and caused
these semiconductors via either oxidative pathway (involving DNA breakage in targeted cancer cells. For another example,
hole transfer to a donor (usually H2O) to form HO•) or reduc- Townley et al. synthesized earth-doped TiO2 nanoparticles for
tive pathway (involving electron transfer to an acceptor (usually ROS generation in tumors to enhance RT.[81] When tumor was
O2) to form O2•−). As a result, the DNA or proteins in cancer treated with X-ray in the existence of such nanoparticles, the
cells were damaged by the generated free radicals.[16,80,81] TiO2 tumor volume could be reduced by around half compared to
and ZnO are typical semiconductors that can produce toxic free that of X-ray alone, which exhibited a good radiosensitization
radicals by photoactivation.[82,83] Previously, researchers applied effect.
them in photodynamic therapy (PDT) due to their unique photo- In addition to lanthanides-doped semiconductor nanoparti-
toxic effect that generates ROS upon the UV irradiation.[80,84–88] cles, some of the other semiconductor nanocomposites were

Figure 2.  a,b) The diagrams of proposed “photoactivation mechanism” by Eu/Gd-doped ZnO nanoparticles and BSA-coated BiOI@Bi2S3 semiconductor
heterojunction nanoparticles under X-ray, respectively. a) Adapted with permission.[16] Copyright 2016, American Chemical Society. b) Adapted with
permission.[90] Copyright 2017, Wiley-VCH.

Adv. Mater. 2018, 1802244 1802244  (6 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

also used in photocatalysis-induced radiosensitization.[89,90] For by Fe2+/Fe3+ to produce a toxic, highly reactive HO•. These radi-
instance, BSA-coated BiOI@Bi2S3 semiconductor heterojunc- cals can lead to oxidative damage to cell components.[91]
tion nanoparticles (SHNPs) for radiosensitization have been The Haber–Weiss reaction process was presented as follows
well investigated in treatment of hepatocellular carcinoma
by Guo et al.[90] On the one hand, since SHNPs contained the Fe3+ + O2 •− → Fe 2+ + O2  (3)
high Z elements such as Bi and I, they possessed a strong
X-ray attenuation capability and thus was anticipated to be the
Fe 2+ + H2O2 → Fe3+ + HO • + OH −  (4)
radiosensitive materials to increase radiation dose deposition
in tumor. On the other hand, owing to the photocatalytic semi-
conductor property, BiOI could be excited by X-ray to generate This reaction can occur in cells and is therefore a possible
the electron–hole pair that reacted with surrounding oxygen source for oxidative stress.[92,93]
or H2O molecules to produce ROS such as HO• and O2•− to Based on the chemocatalysis of Fe-based nanomaterials,
kill tumor. Importantly, superior to the conventional semicon- Hauser et al. employed iron oxide nanoparticles to catalyze
ductor PSs, the formation of core–shell heterojunction structure H2O2 to form HO• for the enhancement of RT on A549 cells.
by coating Bi2S3 shell on the surface of BiOI could reduce the elec- Moreover, the iron oxide nanoparticles were functionalized
tron–hole recombination, and thus further increase the yield of with the TAT peptide (a cell penetrating peptide), which further
electron–hole pair, resulting in higher photocurrent and photo- elevated the efficacy of radiation via increasing intracellular
catalytic activity. As a result, the SHNPs could generate higher concentrations and promoting lysosomal escape of the nano-
level of ROS to damage DNA more effectively (Figure 2b). particles (Figure 3a).[18]

2.1.3. Promoting ROS Generation through


Chemocatalysis Processes of Fe/Cu-based
Nanoradiosensitizers

The H2O2 level within a tumor is overex-


pressed because of the fast metabolism of
cancer cells and insufficient blood supply.
Notably, it has approved that ionizing radia-
tion can increase production of the O2•−
and then these free radical are quickly con-
verted to H2O2 by superoxide dismutase (SOD).
This action further elevates H2O2 level in
cancer cells during the RT treatment. There-
fore, converting intracellular H2O2 into toxic
HO• by suitable catalytic agents under radia-
tion exposure becomes an effective way for
radiosensitization. Some Fe-based nanoma-
terials are such good catalysts. The detailed
radiosensitive process is as follow: the radia-
tion initially facilitates electron leakage
from the electron transport chain and thus
enhances the generation of the O2•−. Then,
the O2•− is converted to H2O2 by natural SOD.
Finally, the H2O2 is catalyzed by Fe-based
nanomaterials to form highly reactive HO• to
result in DNA damage.[18] The generation of
HO• by Fe-based nanoparticles is through the
Fenton’s reaction or Haber–Weiss reaction.
The catalytic process of Fenton’s reaction
was presented as follows

Fe 2+ + H2O2 → Fe3+ + HO • + OH −  (1)

Fe3+ + H2O2 → Fe 2+ + HOO • + H+  (2)


Figure 3.  a,b) Schematic illustration of the mechanism of radiosensitization by TAT-conjugated
iron oxide nanoparticles and TPGS-functionalized Cu3BiS3 nanocrystals, respectively. a) Adapted
This reaction happens in almost all Fe- with permission.[18] Copyright 2016, Elsevier. b) Adapted with permission.[19] Copyright 2017,
related vital processes. H2O2 can be catalyzed Royal Society of Chemistry.

Adv. Mater. 2018, 1802244 1802244  (7 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

In addition to Fe-based nanoparticles, some studies have combined with γ-radiation. Recently, Yong et al. designed a
demonstrated that Cu-based nanoparticles can act as Fenton- chitosan-polyoxometalates-based radiosensitization platform
like reagents for catalyzing the H2O2 substrate to produce (GdW10@CS nanosphere) for enhanced radiation response
ROS.[94,95] Its catalytic mechanism is likely to followed Fenton- by reducing GSH level to improve ROS generation.[17] As
like reaction[95] shown in Figure 4a, the GdW10@CS nanosphere was able to
consume GSH via the redox reaction between W6+ and GSH
Cu 2+ + H2O2 → Cu + + HOO • + H+  (5) to reduce the removal of the generated ROS. This mecha-
nism was verified by the Ellman’s assay, which evaluated
the oxidative stress level by the ratio of GSH and oxidized
Cu + + H2O2 → Cu 2+ + HO • + OH −  (6) glutathione (GSSG). As expected, the relative GSH level and
the GSH/GSSG ratio were significantly reduced after treat-
Based on this principle, Du et al. designed the d-α-tocopherol ment with GdW10@CS nanosphere in external analog and
polyethylene glycol 1000 succinate (TPGS)-functionalized cells (Figure 4b,c). Therefore, these results amply proved that
Cu3BiS3 nanocrystals (NCs) (TPGS-Cu3BiS3 NCs) for the GdW10@CS nanosphere could effectively reduce GSH via
enhanced RT treatment of deep-seated tumor cells.[19] Prof- redox reaction to attenuate radioresistance so as to enhance
iting from the catalytic activity, high X-ray and near-infrared radiation response.
(NIR) laser absorbable ability of TPGS-Cu3BiS3 NCs, the ther- Up to now, a lot of nanomaterials have been reported to
apeutic efficiency of RT was remarkable increased. As shown be able to decrease GSH in cancer cells for cancer therapy,
in Figure 3b, the Cu2+ located on the surface of the NCs were but they are not yet used as radiosensitizers. Herein, we
capable of catalyzing the intracellular H2O2 to generate highly also introduce some of them to inspire researchers to exploit
reactive HO• via Haber–Weiss and Fenton-like reactions to their function in tumor radiosensitization. Some oxidative
destruct cancer cells. Meanwhile, attributing to the strong X-ray compounds are such suitable candidates to exhaust GSH.
absorbability of Bi element, TPGS-Cu3BiS3 NCs could deposit For example, Platinum (Pt) (IV) pro-drugs can be reduced
more X-ray irradiation energy within tumor via generating pho- to highly cytotoxic Pt(II) by high concentrations of GSH in
toelectrons, Compton electrons and Auger electrons to induce cancer cells. Taking advantage of this feature, Bi et al. suc-
the emission of secondary electrons for radiosensitization. cessfully fabricated CuS–Pt(IV) nanoparticles for cooperated
Moreover, benefiting from the strong absorbance of NIR light Pt drug-induced chemotherapy with CuS nanoparticles-medi-
in the second NIR window, TPGS-Cu3BiS3 NCs could generate ated photothermal therapy (PTT).[100] The MnO2 nanomaterial
hyperthermia under NIR irradiation and effectively improve also can be reduced by intracellular GSH so as to decrease the
the tumor oxygenation so as to overcome the hypoxia-induced level of GSH in cancer cells. For instance, chlorin e6 (Ce6)-
radioresistance of tumors, thus further increasing the radiation loaded MnO2 nanosheets and PLGA/hematoporphyrin mono-
effect. methyl ether (HMME)@MnO2 nanoparticles were used to
In addition to Fe and Cu, some of the elements with mul- strengthen PDT efficacy by reducing the intracellular GSH
tiple redox states (such as cobalt (Co) and chromium (Cr)) level, respectively.[101,102] Moreover, the reduction of Cu2+ by
have been proved that can also convert intracellular H2O2 into GSH can also lead to GSH depletion. Ju et al. reported Cu2+–
toxic HO• via the Fenton-like reaction,[96] but have not yet been g-C3N4 nanosheets for PDT enhancement.[98] This system
widely applied into tumor radiosensitization. Thus, it is worth could facilitate light-triggered ROS generation and reduce
exploring and evaluating the tumor radiosensitization effects intracellular GSH levels. In addition to direct reduce GSH by
of the nanomaterials contained these elements to facilitate the oxidizing materials, accelerating GSH oxidation by the mate-
development of new nanoradiosensitizers. rials with corresponding catalytic activity is another effective
approach to indirectly promote the depletion of GSH. Yin
et al. reported that MoS2 was such a material to accelerate
2.1.4. Consuming Intracellular GSH by Nanoradiosensitizers GSH oxidation by catalytic reaction.[103] They used PEG-MoS2
nanoflowers (NFs) for antibacterial applications. It is known
GSH, which contains thiol, is an important antioxidant that GSH is important in the bacterial antioxidant defense
in vivo.[97] The overexpression of GSH is one of the major system, because it can prevent damage of cellular components
obstacles on improving the efficiency of RT, because it can caused by oxidative stress. Therefore, the accelerated GSH oxi-
reduce the amount of generated ROS before their arrival at dation to decrease GSH would more easily result in bacterial
target site and consequently cause the radiation resistance death. It was proved that PEG-MoS2 NFs showed a time- and
in tumor tissue.[98] Therefore, if a nanoparticle can deplete temperature-dependent catalytic oxidation behavior to GSH,
GSH in tumors, it will powerfully overcome the GSH-asso- and 808 nm irradiation induced hyperthermia could remark-
ciated radioresistance and thus improve the RT therapeutic ably promote GSH oxidation at the presence of PEG-MoS2
effect. NFs. Furthermore, the materials that can inhibit GSH syn-
Base on this property, Abdallah et al. realized the RT thesizing enzymes are also able to decrease the GSH level in
enhancement on solid Ehrlich carcinoma-bearing mice cells. For instance, Valodkar et al. studied the in vitro cytotoxic
by Withania somnifera extract/Gd(III) oxide nanocom- mechanisms of latex capped silver nanoparticles (LAgNPs) in
posite (WSGNC).[99] The experimental results showed a human lung carcinoma cells. It showed that LAgNPs resulted
remarkable decrease of GSH content in cancer tissues of in significant decrease of cellular GSH level by the inhibition
the tumor-bearing mice after treating mice with WSGNC of GSH synthesizing enzymes.[104] Therefore, the continued

Adv. Mater. 2018, 1802244 1802244  (8 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 4.  a) Schematic illustration of the mechanism of the GSH consumption. b) Relative GSH level after different treatment. c) GSH/GSSG ratios of
solution after different treatment in external analog and cells. Adapted with permission.[17] Copyright 2017, American Chemical Society.

efforts for developing and applying nanomaterials with GSH treatment of solid tumors through the oxygen-elevated syner-
down-regulated function on RT are significant for cancer getic RT. Similarly, Yi et al. constructed core-shell Au@MnO2
therapy. nanoparticles for improving RT efficacy.[20] On the one hand,
as a well-known radiosensitizer, Au nanoparticle could interact
with X-rays to generate charged particles for enhancing cancer
2.1.5. Improving Tumor Oxygen Level by Nanoradiosensitizers killing under RT. On the other hand, the MnO2 shell could act
as activator to trigger the decomposition of endogenous H2O2
Since tumors grow in an exaggerated way, the tumor interior in the tumor to produce O2 for overcoming hypoxia-induced
becomes far beyond blood supply, which causes the largely sup- radioresistance. As a result, Au@MnO2-PEG nanoparticles
pressed delivery of O2. Such an insufficient O2 supply cannot indicated remarkable enhancement of tumor therapeutic effi-
meet the ever-increasing metabolic demands of the aggres- ciency during RT both in vitro and in vivo. As another typical
sively proliferating tumor cells, bringing about the significant example, since hyperthermia can enhance tumor blood flow
hypoxia in TME and leading to the tumor resistance to ionizing and improve the overall tumor oxygenation to make tumor
irradiation.[10,11,105,106] Thus, the nanoparticles which are able more susceptible to radiation,[112] the nanoparticles that can
to reduce the tumor hypoxia can effectively elevate the RT effi- be used as photothermal agents to provide hyperthermia are
ciency. There are many nanoparticle-mediated approaches for promising for radiosensitization, such as WS2 and Bi2S3.[113,114]
increasing oxygen levels in TME to overcome tumor hypoxia for They can convert NIR into heat to increase the oxygen level in
radiosensitization applications. As a typical paradigm, MnO2 TME so as to elevate the efficacy of RT. In addition, the nano-
can act as activator to trigger the decomposition of endog- particles that load catalase for promoting the generation of O2
enous H2O2 in tumor to produce O2 for decreasing tumor or integrate with oxygen donator to release O2 in tumor have
hypoxia, and thus realize tumor radiosensitization.[20,107–111] been explored to overcome hypoxia-associated radioresist-
For instance, Fan et al. used MnO2 nanosheets anchored with ance.[72,115–118] For example, Song et al. encapsulated catalase
upconversion nanoprobes (UCSMs) for therapy of the solid within tantalum oxide (TaOx) nanoshells for enhancing RT.[115]
tumors.[107] The oxygen generation was achieved by the redox Among this bio-nanoreactor, high Z TaOx could concentrate
activity of MnO2 toward acidic H2O2, which allowed the effective radiation energy into the tumor to increase DNA damage;

Adv. Mater. 2018, 1802244 1802244  (9 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 5.  a) The schematic illustration of the oxygen-loaded TaOx@PFC-PEG nanodroplets for enhanced RT. b) Tumor growth inhibition curves of
mice with different treatments. c) Averaged tumors weights of mice with different treatments at the end of the experiment. d) TUNEL and H&E stained
images of tumor collected from different treated groups. Adapted with permission.[117] Copyright 2017, Elsevier.

meanwhile, catalase could rapidly decompose H2O2 into O2 gradually been reported. For example, Huang et al. employed
to overcome hypoxia-induced radioresistance. Soon after, they biocompatible BSA nanoparticles as carriers of organic sele-
used TaOx nanoparticles to decorate onto perfluorocarbon nocompound with targeting ligand folate to improve the
(PFC) nanodroplets (oxygen donator) for radiosensitization.[117] efficiency of RT. This nanosystem could overcome radioresist-
As illustrated in Figure  5a, the as-prepared TaOx@PFC-PEG ance by inhibiting the function of vascular endothelial growth
nanoparticles could not only increase the X-ray absorption to factor (VEGF), vascular endothelial growth factor receptor
concentrate radiation energy by TaOx, but also act as an oxygen 2 (VEGFR2) and X-ray repair cross complementing pro-
donator to gradually release oxygen and elevate intracellular O2 tein 1 (XRCC-1) and eventually triggered G2/M phase arrest
by PFC. Consequently, remarkable enhancement of RT effect in and apoptotic signaling pathways.[119] Liu et al. confirmed
vivo is achieved by TaOx@PFC-PEG nanoparticles (Figure 5b–d). that antiepidermal growth factor receptor (EGFR) mono-
Overall, it is meaningful to design antihypoxic nanoagents for clonal antibody-conjugated hollow Au nanospheres (HGNs)
radiosensitization applications. (anti-EGFR/HGNs) obviously elevated the ratio of HeLa
cells in G2/M phase.[21] When HeLa cells were treated with
naked HGNs and anti-EGFR/HGNs, the cells in the G2/M
2.1.6. Regulating Cell Cycle by Nanoradiosensitizers phase increased remarkably while cells in the G0/G1 phase
decreased compared with the control, remarkably enhancing
Cell cycle is a series of events which leads to DNA replica- radiation sensitivity of HeLa cells. As another typical example,
tion to generate two daughter cells. It consists of four distinct the glucose-capped Au nanoparticles and androgen receptor
phases: G1, S, G2, and M phase. The most sensitive phase to shRNA-loaded nanoparticle also were reported to elevate radi-
radiation of the cell cycle is G2/M phase. Thus, the nanoparti- osensitivity of cells via enhancing cycle arrest in cells at the
cles which can arrest cancer cells at G2/M phase will enhance G2/M phase under ionizing radiation exposure, which observ-
radiation sensitivity of the cells. In recent years, the studies ably enhanced the sensitivity of cells to radiation.[120,121] There-
about retention of cells in G2/M phase by nanoparticles have fore, the exploitation of the nanomaterials with the ability to

Adv. Mater. 2018, 1802244 1802244  (10 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

arrest cells at G2/M phase is promising for radiosensitization enhancing RT efficiency, these agents can not only exert their
application. own drug therapeutic function, but also render cells more sus-
ceptible to radiation-induced damage through different mech-
anisms, and thus achieve corresponding synergistic RT for
2.2. Nanomaterial-Mediated Synergetic RT for Tumor tumor radiosensitization. For example, the chemotherapeutic
Radiosensitization drugs with the potential to arrest cell cycle at the radiosensi-
tive G2/M phase can afford synergetic chemotherapy with RT.
Besides using the intrinsic radiosensitive properties of nanora- The gene materials that limit the expression of radioresistance-
diosensitizers to enhance the RT, nanomaterial-mediated syn- linked proteins can realize synergetic gene therapy with RT. PSs
ergistic RT is also a good option for tumor radiosensitization, molecule with the ability to produce more ROS can attain syn-
which employs nanoparticles as nanocarriers to delivery various ergetic PDT with RT. Moreover, the photothermal agents, which
therapeutic drugs into tumors. An ideal delivery system needs generate heat to accelerate intratumoral blood flow to enhance
to satisfy the following requirements: delivering therapeutic the tumor oxygenation so as to make the cells more sensitive to
agents through the vasculature without drug leakage, selectively RT, can accomplish synergetic PPT with RT.
accumulating and exerting therapeutic effect in tumor instead of In this section, we summarize the common nanomaterial-
normal tissue to reduce undesired side effects.[122] Nanoparticle mediated synergetic RT methods for tumor radiosensitization
is such an ideal drug delivery system due to its good biocom- (including synergy of chemotherapy with RT, synergy of gene
patibility, highly drug-loading abilities and EPR effects. When therapy with RT, synergy of PDT with RT, synergy of PPT with
using nanoparticles to delivery various radiosensitive thera- RT and trimodal synergetic RT) (Scheme 3). The radiosensi-
peutic agents (such as some of the chemotherapeutic drugs, tive mechanism and application of each kind of synergetic RT
gene materials, PSs, and photothermal agents) into tumors for strategy are concretely discussed.

Scheme 3.  The common methods of nanomaterial-mediated synergetic RT for tumor radiosensitization. Chemo&RT: Adapted with permission.[159]
Copyright 2014, Royal Society of Chemistry. PDT&RT: 1) Single excitation source: Adapted with permission.[207] Copyright 2015, Wiley-VCH. 2)
Two excitation sources: Adapted with permission.[202] Copyright 2016, Elsevier. Trimodal: Adapted with permission.[90] Copyright 2017, Wiley-VCH.
Gene&RT: Adapted with permission.[17] Copyright 2017, American Chemical Society. PTT&RT: Adapted with permission.[243] Copyright 2018, Ivyspring
International Publisher.

Adv. Mater. 2018, 1802244 1802244  (11 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

2.2.1. Synergy of Chemotherapy with RT Therefore, using nanoparticles to load and delivery radiosensitive
chemotherapeutic drugs into tumors for achieving chemo-
At present, chemotherapy is still one of the major cancer treat- therapy/RT synergetic treatment is an effective approach for
ment approaches in the clinic besides surgery and RT.[123,124] tumor radiosensitization.
Some chemotherapy drugs, such as docetaxel (Dtxl),[125–130] Up to now, various kinds of nanoparticles formulation of
Paclitaxel (Ptxl),[131–134] curcumin,[135–140] tirapazamine radiosensitive drugs for increasing the efficacy of RT have
(TPZ),[141–143] cisplatin (CDDP),[144–146] doxorubicin (DOX),[147] been designed.[125,141,150,156–178] For example, based on the
and mitomycin C (MMC),[148–150] have been verified to act as effective radiosensitization effect of chemodrug CDDP, Fan
radiosensitizers to boost the efficiency of RT through different et al. designed rattle-structured multifunctional upconversion
mechanisms. For instance, both Ptxl and curcumin can arrest core/porous silica shell nanotheranostics (UCSNs) to deliver
cells at the G2/M phase and thus enhance the RT.[138,151,152] Dtxl CDDP into tumor cells for achieving chemotherapy/RT syn-
also shows obviously radiosensitive activity, which is ascribed ergetic treatment.[158] The UCSNs-CDDP exhibited unambigu-
to the retention of cells in the G2/M phase,[130] or the pro- ously enhanced RT efficacy both in vitro and in vivo. Soon
motion of ROS generation and the consumption of GSH in afterward, they employed rattle-structured upconversion core/
cells.[153] CDDP can inhibit the repair of damaged DNA caused mesoporous silica nanoparticles (RUMSNs) to transport radi-
by radiation,[154] thus enhancing RT therapeutic effects. More- osensitive drug MMC for chemotherapy and RT synergetic
over, TPZ, a drug only active in hypoxic condition,[155] can be therapy (Figure  6a).[150] Different from the former UCSNs-
reductively activated at low oxygen partial pressure to generate CDDP system, the RUMSNs were conjugated with nuclear
toxic products and consequently damage the hypoxic cells. localization signal ligands TAT that could facilitate the efficient

Figure 6. a) The scheme of the intranuclear radiosensitization by RUMSNs-TAT-MMC nanosystem. b,c) The cell viability of MCF-7 and MCF-7/
ADR cells after different treatments, respectively. d,e) The relative tumor volume of MCF-7 and MCF-7/ADR tumor-bearing mice with time after the
corresponding treatments, respectively. Adapted with permission.[150] Copyright 2015, Royal Society of Chemistry.

Adv. Mater. 2018, 1802244 1802244  (12 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

transportation of nanoparticles into the cell nucleus. Therefore, of radioresistance-linked proteins is also an effective method
the RUMSNs-TAT system could directly transport the radiosen- for radiosensitization.
sitive drug MMC into the nucleus for the substantially elevated Yang et al. once used adenovirus-mediated transfer of siRNA
chemodrug-sensitized RT under high energy X-ray exposure to inhibit survivin expression to enhance the sensitivity of
compared to the extranuclear chemodrug-sensitized RT system. human non–small cell lung cancer (NSCLC) cells to radiation.
As seen in Figure 6b–e, both MCF-7 and MCF-7/ADR cells in The results indicated that the synergistic effect of survivin-
vitro and in vivo could be effectively inhibited by RUMSNs-TAT- targeting gene therapy and RT was significantly valid on cell-
MMC under X-ray radiation. In addition to intranuclear radio- growth inhibition than monotherapy for the NSCLC cells.[188]
sensitive technique, if the nanocarriers can absorb high energy However, the synergy of gene therapy with RT in this work
ionizing radiation, both the nanocarriers and the radiosensitive is performed without nanocarriers. It is known that deliv-
drugs will be able to serve as radiosensitizers, and thus the ering gene drug in vivo without carriers suffers from many
therapeutic effect of RT will be further improved compared to drawbacks, such as fast degradation in physiological environ-
that of single chemodrug-radiosensitized method. For example, ment, short half-life in plasma, bad targeting ability to lead
Setua et al. developed multifunctional CDDP-tethered Au nano- side effect, and inefficient translocation of substances into the
spheres for accomplishing chemotherapy/RT combined treat- cytoplasm.[189] Therefore, it is significant to develop carriers
ment of glioblastoma multiforme (GBM).[159] On the one hand, for gene drug delivery to overcome the aforementioned short-
the chemotherapeutics agent CDDP that was delivered into comings. Nanomaterial with high drug-loading ability and
cancer cells by nanospheres could achieve chemodrug-sensi- biocompatibility is such an ideal carrier. At present, the litera-
tized RT. On the other hand, both Au and Pt were high Z radio- tures about using nanomaterials as carriers for achieving gene
sensitizers. Herein, both the two sides efficiently demolished therapy/RT synergistic treatment are coming in increasing
the tumor cells. For another example, Liu et al. recently con- numbers.[17,121,181,190–197] For instance, Shen et al. used super-
structed a new type of nanoscale coordination polymers (NCPs) paramagnetic dextran iron oxide nanoparticles to carry the E1A
on account of the Hf ions and acidic pH sensitive benzoic- gene and explored the effects of this system on radiosensitiza-
imine linker (BI-linker). Chloro(triphenylphosphine)gold(I) tion of human cervical cancer in nude mice.[181] They demon-
(TPPGC, a chemotherapeutic drug) was then loaded into the strated that this system increased p53 expression but inhibited
NCPs, forming an acidic sensitive degradation and drug human epidermal growth factor receptor-2 (HER-2)/neu expres-
release nanoplatform.[170] In this system, the drug TPPGC could sion in xenograft tumor tissue and thus enhanced the tumor
cause mitochondrial damage as well as block the cell cycle of radiosensitivity. As another example, Yong et al. employed
cancer cells into radiosensitive phase, and high Z Hf ions could GdW10@CS nanosphere to deliver the hypoxia-inducible
act as radiosensitizers to deposit ionizing radiation energy into factor-1α (HIF-1α) siRNA for enhanced radiation response,
tumors. As a result, the efficacy of RT was obviously improved. which is also a typical instance of gene therapy/RT synergistic
treatment (Figure  7a).[17] On the one hand, the GdW10@CS
nanosphere could act as a radiosensitizer to promote enough
2.2.2. Synergy of Gene Therapy with RT ROS generation under X-ray radiation. On the other hand,
HIF-1α siRNA could down-regulate HIF-1α expression to atten-
Gene therapy refers to the guide of genetic materials into uate hypoxia-associated resistance and inhibit broken double-
the target cancer cells to regulate the apoptosis-linked gene’s stranded DNA restoration under RT. The two sides together
expression and corresponding biological signaling pathway to facilitated radiosensitization in hypoxic tumors. As shown in
kill the cancer cells. It is worth pointing out that since gene Figure 7b, a significantly enhanced RT efficacy could be found
therapy can fix abnormal genes (the roots of the diseases), it in the hypoxic BEL-7402 cells after treating with GdW10@
has unparalleled superiority compared to the other traditional CSsiRNA nanospheres + X-ray compared to that of single X-ray
therapeutic methods. treatment. Furthermore, in vivo experiments showed sharp
A lot of researches have shown the apparent links between tumor growth inhibition and no recrudescence in the GdW10@
some genes and/or gene-induced proteins (such as p53, early CSsiRNA+X-ray treated group (Figure 7c), further suggesting the
region 1A (E1A), survivin, VEGF and ataxia telangiectasia prominent RT enhancement effect. Recently, Mizrachi et al.
mutated (ATM)[179–185]) and radiation, because these gene encapsulated BYL719 (a phosphatidylinositol 3-kinase (PI3Kα)
materials can regulate the expression of radiation-related pro- inhibitor) into P-selectin-targeted nanoparticles and realized
tein and thus get the goal of tumor radiosensitization. For RT efficacy enhancement by specific accumulation of BYL719
example, anti-ATM oligodeoxynucleotides and anti-survivin in HNSCC tumors.[196] The nanoparticle-targeted delivery of
siRNA are the common gene materials for radiosensitiza- BYL719 in HNSCC tumors not only enhanced RT therapeutic
tion. Anti-ATM oligodeoxynucleotides can reduce expression outcome via tumor specific PI3K inhibition, but also avoided
of ATM (ATM is a key protein to prevent the DNA damage side effects caused by systemic PI3K inhibition, thus offering a
and activate DNA-repair following ionizing radiation) in novel effective strategy for tumor radiosensitization.
cells to increase sensitivity of the cells to radiation and thus
strengthen the apoptosis of cells.[112,186] For another example,
anti-survivin siRNA that limits the generation of survivin (an 2.2.3. Synergy of PDT with RT
important radioresistance-linked protein) can reverse radiation
resistance and achieve RT enhancement.[182,187] Therefore, the PDT, a potentially new approach to substitute conventional
combination of RT with gene drugs that limits the expression cancer treatment methods, has attracted plentiful attention and

Adv. Mater. 2018, 1802244 1802244  (13 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 7.  a) Schematic illustration of the synergistic gene therapy with RT for efficient radiosensitization by GdW10@CSsiRNA nanosphere. b) The
surviving fraction of BEL-7402 cells with various treatments, respectively. c) In vivo antitumor assay of BEL-7402 tumor-bearing mice after different
administrations. Adapted with permission.[17] Copyright 2017, American Chemical Society.

is widely researched in recent years. Its therapeutic mecha- other hand, SnWO4 could absorb the UV and blue light emitted
nism is as follow: when PSs are localized in cancer tissue, the by the UCNP under 980 nm lasers and then was activated to
ground-state PSs molecules can be activated by appropriate trigger the PDT process to generate ROS. Thus, the combina-
wavelength of light to induce ROS generation to destroy cancer tion of the two processes would powerfully improve the ther-
cells.[26,198–201] It is known that RT could also generate ROS apeutic efficiency. More interestingly, unlike traditional PSs
to damage DNA and cellular components. Hence, concurrent loading strategy that made plenty of molecular PSs to attach a
integration of PDT with RT will remarkably increase the yield single UCNP core, which was not guaranteed that all PSs were
of ROS than single RT, which destroy more cancer cells and activated, this UCNP-SnWO4 nanocrystal was formed by using
thus enhance the radiation effect. amount of tiny UCNPs to covalently conjugate with one needle-
The conventional nanomaterial-modulated PDT and RT like SnWO4 core. Such an especial conformation allowed that
synergetic approaches need both the X-ray and laser as exci- SnWO4 core was able to be activated by enough intensity of
tation light source.[202–204] For example, Liu et al. employed UCNP-emitted light, and generate ROS in a “full-loaded” way,
the PEGylated nanoscale metal organic framework (NMOF) which further boosted the treatment efficacy.
(NMOF-PEG) to achieve PDT/RT combined therapy under both The aforementioned approach of nanoparticle-mediated PDT/
the X-ray and 661 nm laser exposure.[202] This nanoparticle com- RT synergistic treatment requires both X-ray and laser irradiation
posed of Hf4+ and tetrakis (4-carboxyphenyl) porphyrin (TCPP) as excitation light source. Here, another effective and widely appli-
PSs. Due to the strong X-ray absorbed ability of Hf4+ and effi- cable way that only need X-ray irradiation is introduced. It uses
cient singlet oxygen generation ability of TCPP PSs under laser scintillating nanoparticles to delivery PSs into tumors for PDT/
exposure, it was easy to realize PDT/RT synergistic treatment. RT synergistic treatment. Because the scintillating nanoparticles
The study of in vivo therapy indicated that the mice treated can transduce X-rays and/or γ-rays into visible lights and subse-
with NMOF-PEG + X-ray + laser showed remarkable inhibi- quently activate adjacent PSs, this synergistic process can achieve
tion of tumor growth, demonstrating the good treatment effect under a single X-ray excitation source. This energy transfer idea
of PDT/RT synergistic therapy than other mono-therapies. For has been proposed by Chen et al. in 2006.[205] Shortly afterward,
another example, Zhang et al. designed and fabricated a SnWO4 they employed LaF3:Tb3+-meso-tetra(4-carboxyphenyl) porphine
nanocrystal-conjugated upconversion nanoparticle (UCNP- nanoparticle conjugates to generate singlet oxygen following
SnWO4). This nanoparticle could accomplish PDT/RT synergetic X-ray irradiation, which was expected to be a promising thera-
treatment under both X-ray and 980 nm NIR light exposure.[204] peutic approach for deep cancer treatment.[206] Later, Zhang et al.
On the one hand, W and tin (Sn) (high Z atoms) with strong designed a core–shell Ce(III)-doped LiYF4@SiO2@ZnO-PEG
X-ray attenuation capacity are excellent radiosensitizers. On the nanoparticles (SZNP) for combination of octahedral Ce(III)-doped

Adv. Mater. 2018, 1802244 1802244  (14 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 8.  a) The scheme of the synthetic route of SZNPs and the mechanism of ionizing radiation-induced PDT. b) The change of relative tumor
volume of mice with time after different treatments. c) H&E staining of tumor tissue from mice after treatment with: i) saline, ii) SZNPs, iii) X-ray, and
iv) SZNPs + X-ray, respectively. Adapted with permission.[207] Copyright 2015, Wiley-VCH.

LiYF4 nanoscintillator (SCNP) with ZnO nanosemiconductor to electrostatic interaction. This nanoplatform was able to produce
achieve PDT/RT synchronous treatment with reduced oxygen- singlet oxygen under X-ray irradiation.[208] This work experimen-
tension dependency.[207] As shown in Figure  8a, the SCNP was tally quantified the singlet oxygen quantum yield in scintillating
excited by ionizing radiation and thus emitted lots of low-energy CeF3-VP conjugates as a result of the X-ray. The viability of pan-
photons for exciting ZnO surface-bound to form excitons creatic cancer cells treated with the nanoparticles and radiation
(the electron–hole pairs), and the excitons subsequently inter- was clearly reduced, manifesting the good synergetic effect of
acted with water or O2 to obtain free radicals. Specially, the highly radiation-induced-PDT and RT by CeF3-VP conjugates. Most
biotoxic free radicals HO• were generated from the interaction recently, Lan et al. constructed the nanoscale metal–organic layers
between the hole and water instead of O2, which diminished the (MOLs) for X-ray-induced PDT of colon cancer. The MOLs com-
oxygen dependence for producing ROS. Meanwhile, ionizing radi- posed by [Hf6O4(OH)4(HCO2)6] secondary building units (SBUs)
ation had the intrinsic antitumor efficacy via directly interacting and [Ru(bpy)3]2+- or Ir[bpy(ppy)2]+-derived tricarboxylate ligands
with DNA or indirectly radiating water in tumors to produce (bpy = 2,2′-bipyridine, ppy = 2-phenylpyridine).[209] The Hf atoms
ROS to damage DNA. The two paths together afforded the syn- (high Z atom) could strong attenuate X-rays and transfer energy
ergy of PDT and RT for cancer treatment. As exhibited in animal to [Ru(bpy)3]2+ or Ir[bpy(ppy)2]+ ligands to induce PDT by pro-
experiment (Figure 8b), when tumors treated with SZNPs + X-ray ducing ROS, which availably afforded a superb anticancer efficacy
radiation, the relative tumor volume were almost unchanged after through PDT/RT synchronous treatment.
15 days, indicating the complete inhibition of tumor growth.
The result of hematoxylin and eosin (H&E) staining agreed well
with the tumor growth curve, which obviously indicated that the 2.2.4. Synergy of PTT with RT
tumor tissue was damaged after treating with SZNPs + X-ray
irradiation (Figure 8c). Recently, Clement et al. synthesized the PTT has received wide application in recent years due to its
CeF3 nanoparticles conjugated with the verteporfin (VP) PSs via minimal invasion or noninvasion properties.[210] It utilizes

Adv. Mater. 2018, 1802244 1802244  (15 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

heat generated from NIR optical absorbance agents to ablate dots were synthesized via a green and facile method of phys-
tumor.[113,211–213] Because of the minimal tissue absorption ical grinding and ultrasonication. For the in vitro experiment,
and scattering within the NIR or biological window, NIR light when the cells treated by WS2  + NIR + RT, it was obviously
has greater depth of penetration in tissue, which is pivotal for observed that the cell survival fraction dramatically decreased.
the utilization of PTT on biological cells and in vivo organ- Similarly, the mice treated with WS2 QDs + NIR + RT showed
isms.[214–221] More interestingly, an appropriate level of hyper- the most efficient tumor growth delay. More worthy of men-
thermia generated by PTT can accelerate intratumoral blood tion, the ultrasmall sizes WS2 QDs could be cleared out of the
flow so as to improve the overall tumor oxygenation to over- body by renal excretions, greatly minimizing the side effects of
come the tumor hypoxia.[222] It is known that RT suffers from WS2 QDs to living organisms. For another example, as depicted
resistance to hypoxic cells due to its oxygen dependence and in Figure  9a, Wang et al. reported BSA-mediated synthesis of
thus leads to anticancer failure. Therefore, when combing PTT Bi2S3 biocompatible nanoparticles for combined PTT/RT treat-
with RT, the heat generated from PTT can elevate the oxygen ments of cancers.[114] Attributing to the ultrasmall size and
level in cancer cells to make the cells more sensitive to radia- good colloidal stability, the Bi2S3 nanoparticles possessed a long
tion, and thus the RT therapeutic effect is availably strength- blood circulation time and high tumor targeting ability, which
ened. Up to date, a large number of literatures about PTT and was greatly in favor of the therapeutic applications. As shown
RT synergetic therapy can be searched.[8,54,79,113,114,211,212,222–245] in Figure 9b,c, the Bi2S3 nanoparticle-induced excellent PTT
And we roughly summarized the evolution trends of this syner- and RT synergistic effects afforded the completely eradication
getic strategy as follows. of tumors and 100% survival rate of tumor-bearing mice over
Previously, nanomaterial-modulated synergetic PTT with RT 40 day after the treatments.
was achieved by decorating the photothermal agents on nano-
carriers. For example, Xiao et al. designed a multifunctional
core/satellite nanotheranostic (CSNT) through decorating 2.2.5. Trimodal Synergetic RT
photothermal transducing agent ultrasmall CuS nanoparticles
onto the surface of silica-coated rare-earth upconversion nano­ Following the step of exploration for dual-modal synergetic cancer
particles.[222] The CuS nanoparticles absorbed NIR light and RT, trimodal synergetic RT based on nanomaterials for tumor
converted it into heat for tumor thermal-ablation. Meanwhile, radiosensitization has been gradually realized in recent years,
the heat improved tumor oxygen and thus the RT treatment such as chemotherapy/PDT/RT,[246] chemotherapy/PTT/ RT[247]
effect was enhanced. Moreover, silica-coated rare-earth upcon- and PTT/PDT/RT,[90,248,249] and so on. Fan et al. constructed the
version nanoparticles (NaYbF4: 2%Er3+/20%Gd3+@SiO2-NH2) sub-80 nm multifunctional rattle-structured Gd-UCNPs core/
containing high Z elements also could induce a highly local- mesoporous silica shell nanocomposites (UCMSNs) for the co-
ized radiation dose to strengthen radiation damage. Therefore, delivery of hematoporphyrin (HP) and Dtxl to achieve chemo-
CSNTs were the promising nanoplatform of PTT/RT syner- therapy/PDT/RT trimodal synergetic cancer therapy.[246] In this
gistic treatment for tumor radiosensitization. UCMSNs-HP-Dtxl nanosystem, HP was applied as both PS and
Lately, the construction of nanocomposites by photothermal radiosensitizer for PDT/RT synergetic therapy, and Dtxl was acted
transducing agents and radiation dose-enhancing agents as both chemotherapeutic agent and radiosensitizer for chemo-
became another effective strategy for PTT/RT synergetic treat- therapy/RT synergetic therapy. As a result, such a UCMSNs-HP-
ment. For instance, Wang et al. integrated photothermal agent Dtxl nanosystem mediated trimodal synergetic therapy afforded
MoS2 with radiation dose-enhancing agents Bi2S3 to form the complete tumor ablation. Recently, Qiu et al. employed single
MoS2/Bi2S3-PEG (MBP) composite nanosheets for cancer ultrathin PVP-decorated W18O49 nanowires to realize PTT/PDT/
therapy.[212] Among the MBP, the high X-ray attenuation effect RT trimodal synergistic cancer therapy.[249] Three properties
of Bi promoted the dose-enhancement during tumor RT, and endowed W18O49 nanowires with the above therapeutic func-
the strong NIR absorbance of MoS2 nanosheet framework tions. First, W18O49 nanowire with high photothermal conver-
enabled the highly efficient photothermal ablation of tumors. sion efficacy could serve as a photothermal agent.[250] Second,
When the mice bearing 4T1 tumors were treated with MBP the nanowires could be used as PSs to generate cytotoxic singlet
nanosheets and subsequently exposed to 808 nm laser and oxygen under the 980 nm laser excitation for PDT.[251] Third, the
X-ray, a remarkable tumor therapeutic outcome of MBP high Z atom (W, Z = 74) enabled the nanowire with the potential
nanosheets could be obtained. for local radiation dose enhancement. The In vivo experiments
Recently, the nanomaterials that play both roles of photo- showed the complete tumor clearance of mice treated with
thermal agents and radiosensitizers are widely researched due to W18O49  + NIR + RT after 4 days, and no recurrence within a 9
their multifunctional properties and simply synthetic methods, month follow-up, which demonstrated that trimodal PTT/PDT/
such as Au,[223,224,227,231,239] WS2[8,113] Bi2S3,[114,233] Bi2Se3,[228,245] RT synergistic therapeutic effect between NIR-induced PDT/PTT
ReS2,[240] Cu3BiS3,[236] and WO3−x[230] nanoparticles. The NIR and RT was more available than that of RT therapy alone.
absorption ability of these nanoparticles allows them to act as
photothermal agents for producing significant heat to elimi-
nate tumor, and the high Z atom endows these nanoparticles 2.3. Radioprotection of Healthy Tissues by Nanomaterials for
with the ability to serve as radiosensitizers for accumulating Tumor Radiosensitization
more X-ray energy in cells. For example, Yong et al. reported
that ultra-small size WS2 dots could act as multifunctional When ionizing radiation is used for tumor treatment, the sur-
nanotheranostics for PTT/RT synergistic treatment.[8] The WS2 rounding healthy tissues may inevitably receive the radiation

Adv. Mater. 2018, 1802244 1802244  (16 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 9.  a) The scheme of synthetic process of BSA-stabilized Bi2S3 nanoparticles and the BSA-stabilized Bi2S3 nanoparticles for PTT/RT synergistic
therapy. b) The changes of relative tumor volume of mice with time under various treatments. c) The survival rates of mice after various treatments.
Adapted with permission.[114] Copyright 2016, Wiley-VCH.

energy, which leads to the side effects and thus decreases the The in vivo experiments indicated that surviving fraction of
therapeutic ratio of RT. Therefore, the protection of healthy tis- mice was significantly increased under high-energy ionizing
sues from the radiation-caused injury by nanoradioprotectors radiation after the mice were treated with these quantum dots.
to increase the radiotherapeutic ratio is another useful way Moreover, the cysteine-protected MoS2 and WS2 nanodots could
for nanomaterial-mediated tumor radiosensitization. Because protect the hematological system and DNA in bone marrow
human tissues have 80% water, the major radiation damage is cells of mice from high-energy rays. Furthermore, these
caused by ROS generated from the interaction between water quantum dots could remove excessive 3,4-methylenedioxyam-
and ionizing radiation.[252] Therefore, the general radioprotec- phetamine (MDA) and recover SOD in lung and liver of mice.
tion reagents are free radical scavengers, which can scavenge These results suggested that cysteine-protected MoS2 and WS2
ROS in cells and thereby reduce the DNA damage. Up to now, nanodots have the potential to reduce radiation-caused injury
a lot of nanomaterials with free radical scavenge ability have to normal organ and thus boost the therapeutic ratio of RT. In
been reported, including graphene-based nanomaterials,[253,254] addition, it is well known that Se is beneficial to human health
fullerene-based nanomaterials,[255,256] bamboo charcoal nano­ by involving in various important bioactivities, including radio-
particles,[257] graphdiyne nanoparticles,[258] cerium oxide protection. Based on this, Du et al. constructed a new versatile
nanoparticles,[259–261] MoS2, WS2, WSe2, and Bi2Se3 nanoparti- radioprotector based on PVP- and selenocysteine (Sec)-mod-
cles,[262–265] to name a few. Most of them have been utilized as ified Bi2Se3 nanoparticles (PVP-Bi2Se3@Sec NPs).[245] Particu-
radioprotectors for radioprotection. As a paradigm example, Xie larly, this Bi2Se3 nanoplatform could simultaneously enhance
et al. found that the biocompatible bamboo charcoal nanoparti- RT efficacy in tumor and achieve radioprotection in healthy
cles have the potential applications in radioprotection.[257] They tissues (Figure  10). On the one hand, the strong X-ray attenu-
evaluated the radioprotection activity of TPGS-functionalized ation ability of Bi atom and remarkable photothermal effect
bamboo charcoal nanoparticles (TPGS-BCNPs) through the of Bi2Se3 endowed this nanoplatform with the ability of PTT/
intracellular ROS level assay and DNA damage assay (γ-H2AX RT synergistic treatment. On the other hand, it is known that
staining) in human umbilical vein endothelial cells (HUVECs). Se could be incorporated into selenoproteins by the amino
It turned out that the intracellular ROS level and DNA damage acid Sec and subsequently catalyzed a wide range of electron-
level of HUVECs in the TPGS-BCNPs + X-ray treated groups transfer reactions for antioxidant defense and redox regulation
were less than the single X-ray treated groups, revealing the to reduce side-effects of RT for normal tissues. Therefore, the
potential of TPGS-BCNPs on radiation protection of healthy traces of Se, which released from the PVP-Bi2Se3@Sec NPs
tissues. As another typical example, Zhang and co-workers and entered the blood circulation system, could increase the
reported that cysteine-protected MoS2 and WS2 nanodots could immune function (such as enhancing the SOD and GSH-Px
serve as radioprotectors due to their ROS scavenging ability in activity, improving the cytokines level, up-regulating number
vivo via the highly catalytic activity against H2O2 and O2.[262,263] of white blood cells and reducing marrow DNA suppression)

Adv. Mater. 2018, 1802244 1802244  (17 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 10.  Scheme of the therapeutic principle for simultaneously radiosensitization and radioprotection by PVP-Bi2Se3@Sec nanoparticles. Adapted
with permission.[245] Copyright 2017, Wiley-VCH.

and reduce the side-effects of ionizing radiation in body. Con- named NBTXR3, whose effective radiation sensitizing ingre-
sequently, the PVP-Bi2Se3@Sec NPs provided a promising dient is HfO2 nanoparticles.[266–270] HfO2 is relatively chem-
platform to simultaneously enhance RT in tumor and protect ical inert in biological media, which reduce its biotoxicity and
normal tissue from radiation-induced side effect, which avail- encourage its biomedical use. After being functionalized with
ably increased the radiotherapeutic ratio and thus accomplished a negative surface charge, this nanosphere with average dia­
tumor radiosensitization. meter about 50 nm can be stabilized in an aqueous solution
at pH between 6 and 8.[268,271] More importantly, Hf is a high
Z atom (Z = 72), which enables it to be used as radiosensitive
3. The Clinical Translation of Nanomaterial- agent.[266] HfO2 nanoparticles can serve as a radiosensitizer by
physical mechanism.[270,272] When HfO2 enters the tumor tis-
Mediated Tumor Radiosensitization
sues and is activated by external radiation beam, it can emit
For the sake of ultrasmall size and tailorable versatile physico- high energy electrons and increase the electron density depos-
chemical properties, nanomaterial and its related nanomedicine ited within irradiated tissues, resulting that the dose of X-ray
is the emerging field utilized in clinic biomedical applications delivered to the tumor is magnified while the dose passing
to improve human health, especially in cancer diagnosis and through healthy tissues remains unchanged. This physical
treatment. Over the last two decades, many prominent exam- interaction among high energy photons, HfO2 nanoparticles
ples of multifunctional nanotheranostics have been used to and cancer cells can promote the generation of cytotoxic free
conquer kinds of cancer. The past decade has witnessed a dra- radicals and other ROS to destroy cancer cells through double-
matic increase in interest in the use of nanoparticles as radio- stranded DNA damage,[266] and thus lead to the inhibition
sensitizers for RT. Herein, we want to analyze the state of art of tumor growth. In addition, Monte Carlo simulation also
of promising nanoradiosensitizers as well as their translation find that NBTXR3 enhances radiation doses ninefolds when
from bench to bedside. compared to water exposure alone.[268] The first human trial
Nanomedicines are undergoing clinical translation for RT. showed that preoperative NBTXR3 with external beam RT is
At present, some nanoradiosensitizers, especially the inorganic a technically feasible therapeutic approach that yields encour-
nanoradiosensitizers, have made gratifying breakthroughs and aging radiological and pathologic responses in patients with
entered clinical trials for cancer RT (Table  1). Compared with locally advanced soft tissue sarcoma of the extremity and trunk
organic nanomaterials, there is some skepticism that inorganic wall.[272] In the stage of clinical trials, NBTXR3 has shown
compounds are the underdog and hardly applied in biomedi- better antitumor performance to many types of cancers, such as
cine. However, in recent years, some inorganic nanomate- soft tissue sarcoma (ClinicalTrials.gov; NCT02379845),[272–274]
rials have been demonstrated to be promising candidates for head and neck cancer (NCT02901483),[6] prostate cancer
the enhancement of cancer radiation treatment. For example, (NCT02805894),[275] rectal cancer (NCT02465593),[272] and liver
French company Nanobiotix developed a nanoradiosensitizer cancer (NCT02721056),[272] just to name a few.

Adv. Mater. 2018, 1802244 1802244  (18 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Table 1.  Clinical translation of some nanoradiosensitizers. side effects of normal organ by nanoradiopro-
tectors, which protects healthy tissues from
Name ClinicalTrials.gov identifier Condition or disease Clinical trial phase radiation-caused injury to improve the radio-
NBTXR3 NCT02721056 Liver cancer Phase I/II therapeutic ratio. Moreover, we also discuss
NCT02805894 Prostate cancer Phase I/II
the clinical translation of nanoparticle-medi-
ated radiosensitization. It is clear that the
NCT01946867 Head and neck cancer Phase I
challenges still exist in the clinical translation
NCT01433068 Adult soft tissue sarcoma Phase I of the nanomaterials for RT enhancement
NCT02379845 Adult soft tissue sarcoma Phase II/III that we have to face and tackle:
NCT02901483 Head and neck squamous cell carcinoma Phase I/II
NCT02465593 Rectal cancer Phase I/II
1) The systematical investigations of the biosafety
and metabolism in vivo of nanomaterials. Owing
AGuIX NCT03308604 Gynecologic cancer Phase I
to the relative large size and slow biodegrad-
NCT02820454 Brain metastases Phase I able ability, nanoparticles have a relative longer
systemic circulation time and lower metabo-
lism rate than that of molecular drugs, which
In addition, Tillement and co-workers find a nanoradiosen- endows the nanoparticles with enough time to exert their ther-
sitizer AGuIX to enhance the sensitization of brain tumor cells apeutic function in vivo. However, these characters are also
to radiation.[276,277] This ultrasmall AGuIX nanoparticle displays easy to induce their long-term retention and accumulation in
hydrodynamic diameters inferior to 5 nm, indicating that it can body and thus may cause harm to health. Therefore, the sys-
be excreted through the kidney and avoiding biosafety prob- tematical investigations of their biosafety and metabolism in
lems. AGuIX is composed of Gd chelated polysiloxane, and vivo are vitally important to evaluate the prospect of their ra-
its chemical composition is anticipated to be Gd10Si40C200N50 diosensitization applications. First, for the biosafety, although
O150Hx.[277–279] Gd resulted in a strong interaction with X-rays short-term toxicity and biodistribution of nanomaterials have
and an increase of RT efficacy.[280] In addition, AGuIX can be been studied in some efforts, their long-term biological effects
accumulated in tumoral tissue due to EPR effect,[281] further and potential biosafety are largely unexplored.[285] Meanwhile,
promoting the RT efficacy. Preclinical animal experiments due to plenty of the nanomaterials studied at the present stage
show that AGuIX locally enhance the effect of RT in a variety contain heavy metal elements, which have toxicity to a certain
of tumor models and exert no significant toxicity to mice and extent, it is necessary to find more and better ways to reduce
monkeys.[277,282] Several phases I clinical trials are ongoing in their toxicity and improve their biocompatibility (such as the
patients with brain metastases (NCT02820454) to enhance the utilization of suitable surface modifiers). Then, for the metas-
sensitization of brain metastatic tumor cells to radiation.[283] bolism issue, many of the nanomaterials are faced with the
Also, AGuIX nanoparticles also have the potential to treat cer- problem of slower biodegradation compared to molecular
vical and liver cancer.[277,284] The safety, tolerability of doses drugs. Accordingly, the efforts are needed to appropriately
of AGuIX in combination with radiation and CDDP-based improve the metabolic capacity of nanomaterials and/or
chemotherapy in patients with locally advanced cervical cancer design and fabricate the new types of easily biodegradable
are being evaluated in phase 1 clinical trials (NCT03308604). nano­materials. For instance, it can prepare the nanoparticles
Besides, according to the information of NH TherAguix com- with ultrasmall size to make them clear out of the body by
pany, the potential indications of AGuIX also include lung renal excretions. Moreover, the studies on clearance pathway,
cancer, esophageal carcinoma, head and neck tumor. immune behavior and other biological effects are still insuffi-
cient. Therefore, all these issues are of significance and should
be put in the first place to investigate so as to satisfy its future
4. Conclusions and Future Perspectives radiosensitization application in the clinic.
2) The optimization of the physicochemical properties of nano­
The rapid development and application of emerging advanced materials for RT enhancement. In general, so many factors,
nanomaterials in biomedicine provides a good opportunity to such as the composition, shape, size, synthesis method, sur-
enhance the efficiency of RT treatment. In this review, we sys- face functional modification, and doses of nanoradiosensitiz-
tematically summarize the general strategies and corresponding ers, can affect the sensitization effect of RT. For instance, as
mechanisms of nanomaterial-mediated tumor radiosensitiza- mentioned above, different size of Au-based nanoparticles
tion. It mainly consists of three aspects. The first is on account can lead to the different radiosensitive effect.[44,51,59] Moreover,
of the intrinsic radiosensitive properties of nanoradiosensitizers shape-dependent radiosensitive effect of Au nanostructures
selves, which includes enhancing ionizing radiation energy dep- was also reported.[58] Therefore, it is important to compare the
osition, promoting ROS generation and modulating the TME to radiosensitive efficacy of nanomaterials with different phys-
make tumor more sensitive to radiation. The second is nanoma- icochemical parameters, and gain the optimal conditions to
terial-mediated synergistic RT, which uses the nanoparticles to obtain satisfying RT efficiency.
load and delivery the radiosensitive drugs into tumor for cancer 3) The deeper exploration of the mechanisms of radiosensitiza­
treatment. These drugs can not only exert its own drug func- tion. Human organism is a complex system that numerous
tion, but also make cells more susceptible to radiation-induced physical, chemical and biological factors will influence the RT
damage through different mechanisms. The third is to reduce efficiency when nanomaterials entered tumor region. Until

Adv. Mater. 2018, 1802244 1802244  (19 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

now, researchers have devoted numerous efforts to explain the [1] J. Ferlay, I. Soerjomataram, R. Dikshit, S. Eser, C. Mathers,
mechanisms of various kinds of radiosensitization approach- M. Rebelo, D. M. Parkin, D. Forman, F. Bray, Int. J. Cancer 2015,
es, but most of the mechanisms are still limited at cellular 136, E359.
level, and the mechanisms that deep into molecular levels are [2] L. A. Kunz-Schughart, A. Dubrovska, C. Peitzsch, A. Ewe, A. Aigner,
S. Schellenburg, M. H. Muders, S. Hampel, G. Cirillo, F. Iemma,
still rare. Therefore, it is necessary to deeper understand ra-
R. Tietze, C. Alexiou, H. Stephan, K. Zarschler, O. Vittorio,
diosensitization mechanisms at molecular or gene levels. M. Kavallaris, W. J. Parak, L. Madler, S. Pokhrel, Biomaterials 2017,
4) The exploration of the optimal modality of synergistic RT. Al- 120, 155.
though there are various combined modalities for each kind [3] S. Her, D. A. Jaffray, C. Allen, Adv. Drug Delivery Rev. 2017, 109, 84.
of synergistic RT, we cannot randomly combine these thera- [4] S. Stapleton, D. Jaffray, M. Milosevic, Adv. Drug Delivery Rev. 2017,
peutic strategies together, because the random combination 109, 119.
may not get the optimal synergistic anticancer efficiency. For [5] G. Song, L. Cheng, Y. Chao, K. Yang, Z. Liu, Adv. Mater. 2017, 29,
example, when we use PTT/RT for cancer treatment, whether 1700996.
we offer NIR laser before X-ray or offer X-ray before NIR laser [6] L. J. Gong, J. N. Xie, S. Zhu, Z. J. Gu, Y. L. Zhao, Acta Phys.-
or offer NIR laser and X-ray simultaneously, it still requires Chim. Sin. 2018, 34, 140.
[7] D. Kwatra, A. Venugopal, S. Anant, Transl. Cancer Res. 2013, 2,
us to explore thoroughly and choose the most suitable modal-
330.
ity to get the optimal therapeutic efficiency. Therefore, it still [8] Y. Yong, X. Cheng, T. Bao, M. Zu, L. Yan, W. Yin, C. Ge, D. Wang,
needs to spare no effort to find the optimal synergistic modal- Z. Gu, Y. Zhao, ACS Nano 2015, 9, 12451.
ity of synergistic RT. [9] M. Benderitter, F. Caviggioli, A. Chapel, R. P. Coppes, C. Guha,
5) The establishment of standardization of nanoradiosensitizers de­ M. Klinger, O. Malard, F. Stewart, R. Tamarat, P. V. Luijk,
sign and fabrication to satisfy the clinical translation. The bio- C. L. Limoli, Antioxid. Redox Signaling 2014, 21, 338.
logical effects and toxicological characteristics are drastically [10] J. M. Brown, W. R. Wilson, Nat. Rev. Cancer 2004, 4, 437.
different between nanodrug and molecular drugs for vary of [11] J. N. Liu, W. Bu, J. Shi, Chem. Rev. 2017, 117, 6160.
physical and chemical properties when they enter biological [12] J. Xu, W. Han, Z. Cheng, P. Yang, H. Bi, D. Yang, N. Niu, F. He,
tissue. Therefore, to make the nanoradiosensitizers suitable S. Gai, J. Lin, Chem. Sci. 2018, 9, 3233.
[13] M. H. Lee, Z. Yang, C. W. Lim, Y. H. Lee, S. Dongbang, C. Kang,
for clinic translation, rationally preclinical protocols are ex-
J. S. Kim, Chem. Rev. 2013, 113, 5071.
traordinary demanded, and the guidance as well as stand- [14] E. K. Lim, T. Kim, S. Paik, S. Haam, Y. M. Huh, K. Lee, Chem. Rev.
ardization for nanoradiosensitizers design and fabrication 2015, 115, 327.
should be established. [15] S. Acharya, S. K. Sahoo, Adv. Drug Delivery Rev. 2011, 63, 170.
6) The promotion of clinical translation of nanoradiosensitizers. [16] B. Ghaemi, O. Mashinchian, T. Mousavi, R. Karimi, S. Kharrazi,
The current status of the development of nanoradiosensitiz- A. Amani, ACS Appl. Mater. Interfaces 2016, 8, 3123.
ers is too many papers but too few clinic anticancer drugs. [17] Y. Yong, C. Zhang, Z. Gu, J. Du, Z. Guo, X. Dong, J. Xie, G. Zhang,
The clinic translation of nanoradiosensitizers remains in a X. Liu, Y. Zhao, ACS Nano 2017, 11, 7164.
dilemma. Therefore, efforts are needed to facilitate the clini- [18] A. K. Hauser, M. I. Mitov, E. F. Daley, R. C. McGarry,
cal translation of nanoradiosensitizers. In the era of nano- K. W. Anderson, J. Z. Hilt, Biomaterials 2016, 105, 127.
[19] J. Du, X. Zheng, Y. Yong, J. Yu, X. Dong, C. Zhang, R. Zhou, B. Li,
medicine, this emerging and promising interdisciplinary
L. Yan, C. Chen, Z. Gu, Y. Zhao, Nanoscale 2017, 9, 8229.
filed full of hope and will further promote the application of [20] X. Yi, L. Chen, X. Zhong, R. Gao, Y. Qian, F. Wu, G. Song, Z. Chai,
nanomedicine for RT enhancement. Z. Liu, K. Yang, Nano Res. 2016, 9, 3267.
[21] J. Liu, Y. Liang, T. Liu, D. Li, X. Yang, Nanoscale Res. Lett. 2015, 10,
218.
Acknowledgements [22] R. R. Arvizo, S. Bhattacharyya, R. A. Kudgus, K. Giri,
R. Bhattacharya, P. Mukherjee, Chem. Soc. Rev. 2012, 41, 2943.
J.X. and L.G. contributed equally to this work. This work was supported
by the National Basic Research Program of China (2016YFA0201600, [23] J. Conde, G. Doria, P. Baptista, J. Drug Delivery 2012, 2012, 751075.
2015CB932104), National Natural Science Foundation of China [24] J. A. Coulter, W. B. Hyland, J. Nicol, F. J. Currell, Clin. Oncol. 2013,
(51772292, 31571015, 11621505, 11435002, and 21320102003), Youth 25, 593.
Innovation Promotion Association CAS (2013007), and Key Research [25] X. Y. Su, P. D. Liu, H. Wu, N. Gu, Cancer Biol. Med. 2014, 11, 86.
Program of Frontier Sciences CAS (QYZDJ-SSW-SLH022). [26] P. Retif, S. Pinel, M. Toussaint, C. Frochot, R. Chouikrat,
T. Bastogne, M. Barberi-Heyob, Theranostics 2015, 5, 1030.
[27] A. Subiel, R. Ashmore, G. Schettino, Theranostics 2016, 6, 1651.
Conflict of Interest [28] Y. Liu, P. C. Zhang, F. F. Li, X. D. Jin, J. Li, W. Q. Chen, Q. Li,
Theranostics 2018, 8, 1824.
The authors declare no conflict of interest. [29] J. F. Hainfeld, F. A. Dilmanian, D. N. Slatkin, H. M. Smilowitz,
J. Pharm. Pharmacol. 2008, 60, 977.
[30] A. Mesbahi, Rep. Pract. Oncol. Radiother. 2010, 15, 176.
[31] S. Jelveh, D. B. Chithrani, Cancers 2011, 3, 1081.
Keywords [32] D. Wu, X. D. Zhang, P. X. Liu, L. A. Zhang, F. Y. Fan, M. L. Guo,
clinical translation, nanomaterial-mediated radiosensitization, Curr. Nanosci. 2011, 7, 110.
nanoradiosensitizers, radioprotection, synergistic radiotherapy [33] K. T. Butterworth, S. J. McMahon, F. J. Currell, K. M. Prise,
Nanoscale 2012, 4, 4830.
Received: April 9, 2018 [34] S. Jain, D. G. Hirst, J. M. O’Sullivan, Br. J. Radiol. 2012, 85, 101.
Revised: June 8, 2018 [35] K. T. Butterworth, S. J. McMahon, L. E. Taggart, K. M. Prise,
Published online: Transl. Cancer Res. 2013, 2, 269.

Adv. Mater. 2018, 1802244 1802244  (20 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

[36] J. F. Dorsey, L. Sun, D. Y. Joh, A. Witztum, A. Al Zaki, G. D. Kao, [63] F. Lux, L. Sancey, A. Bianchi, Y. Cremillieux, S. Roux, O. Tillement,
M. Alonso-Basanta, S. Avery, A. Tsourkas, S. M. Hahn, Nanomedicine 2015, 10, 1801.
Transl. Cancer Res. 2013, 2, 280. [64] I. Miladi, M. T. Aloy, E. Armandy, P. Mowat, D. Kryza, N. Magne,
[37] D. R. Cooper, D. Bekah, J. L. Nadeau, Front. Chem. 2014, 2, 86. O. Tillement, F. Lux, C. Billotey, M. Janier, C. Rodriguez-Lafrasse,
[38] W. Ngwa, R. Kumar, S. Sridhar, H. Korideck, P. Zygmanski, Nanomed.: Nanotechnol. Biol. Med. 2015, 11, 247.
R. A. Cormack, R. Berbeco, G. M. Makrigiorgos, Nanomedicine [65] F. Taupin, M. Flaender, R. Delorme, T. Brochard, J. F. Mayol,
2014, 9, 1063. J. Arnaud, P. Perriat, L. Sancey, F. Lux, R. F. Barth, M. Carriere,
[39] K. Haume, S. Rosa, S. Grellet, M. A. Smialek, K. T. Butterworth, J. L. Ravanat, H. Elleaume, Phys. Med. Biol. 2015, 60, 4449.
A. V. Solov’yov, K. M. Prise, J. Golding, N. J. Mason, Cancer [66] R. Delorme, F. Taupin, M. Flaender, J. L. Ravanat, C. Champion,
Nanotechnol. 2016, 7, 8. M. Agelou, H. Elleaume, Med. Phys. 2017, 44, 5949.
[40] J. Schuemann, R. Berbeco, D. B. Chithrani, S. H. Cho, R. Kumar, [67] A. Detappe, E. Thomas, M. W. Tibbitt, S. Kunjachan, O. Zavidij,
S. J. McMahon, S. Sridhar, S. Krishnan, Int. J. Radiat. Oncol., Biol., N. Parnandi, E. Reznichenko, F. Lux, O. Tillement, R. Berbeco,
Phys. 2016, 94, 189. Nano Lett. 2017, 17, 1733.
[41] L. Cui, S. Her, G. R. Borst, R. G. Bristow, D. A. Jaffray, C. Allen, [68] F. Du, L. Zhang, L. Zhang, M. Zhang, A. Gong, Y. Tan, J. Miao,
Radiother. Oncol. 2017, 124, 344. Y. Gong, M. Sun, H. Ju, C. Wu, S. Zou, Biomaterials 2017, 121,
[42] N. Goswami, Z. T. Luo, X. Yuan, D. T. Leong, J. P. Xie, Mater. 109.
Horizons 2017, 4, 817. [69] M. H. Yao, M. Ma, Y. Chen, X. Q. Jia, G. Xu, H. X. Xu, H. R. Chen,
[43] S. Rosa, C. Connolly, G. Schettino, K. T. Butterworth, K. M. Prise, R. Wu, Biomaterials 2014, 35, 8197.
Cancer Nanotechnol. 2017, 8, 2. [70] X.-D. Zhang, J. Chen, Y. Min, G. B. Park, X. Shen, S.-S. Song,
[44] X. D. Zhang, D. Wu, X. Shen, J. Chen, Y. M. Sun, P. X. Liu, Y.-M. Sun, H. Wang, W. Long, J. Xie, K. Gao, L. Zhang, S. Fan,
X. J. Liang, Biomaterials 2012, 33, 6408. F. Fan, U. Jeong, Adv. Funct. Mater. 2014, 24, 1718.
[45] A. Al Zaki, D. Joh, Z. Cheng, A. L. B. De Barros, G. Kao, J. Dorsey, [71] M. Algethami, M. Geso, T. Piva, A. Blencowe, L. Lu, K. Ai,
A. Tsourkas, ACS Nano 2014, 8, 104. P. D. Harty, E. Gan, NanoWorld J. 2015, 1, 99.
[46] Y. S. Yang, R. P. Carney, F. Stellacci, D. J. Irvine, ACS Nano 2014, [72] G. Song, C. Liang, X. Yi, Q. Zhao, L. Cheng, K. Yang, Z. Liu,
8, 8992. Adv. Mater. 2016, 28, 2716.
[47] X. D. Zhang, J. Chen, Z. Luo, D. Wu, X. Shen, S. S. Song, [73] F. Du, J. Lou, R. Jiang, Z. Fang, X. Zhao, Y. Niu, S. Zou, M. Zhang,
Y. M. Sun, P. X. Liu, J. Zhao, S. Huo, S. Fan, F. Fan, X. J. Liang, A. Gong, C. Wu, Int. J. Nanomed. 2017, 12, 5973.
J. Xie, Adv. Healthcare Mater. 2014, 3, 133. [74] Z. H. Song, Y. Z. Chang, H. H. Xie, X. F. Yu, P. K. Chu, T. F. Chen,
[48] X. D. Zhang, Z. Luo, J. Chen, X. Shen, S. Song, Y. Sun, S. Fan, NPG Asia Mater. 2017, 9, e439.
F. Fan, D. T. Leong, J. Xie, Adv. Mater. 2014, 26, 4565. [75] J. Deng, S. Xu, W. Hu, X. Xun, L. Zheng, M. Su, Biomaterials 2018,
[49] C. Wang, A. Sun, Y. Qiao, P. Zhang, L. Ma, M. Su, 154, 24.
J. Mater. Chem. B 2015, 3, 7372. [76] Y. Yong, L. J. Zhou, S. S. Zhang, L. Yan, Z. J. Gu, G. J. Zhang,
[50] X. D. Zhang, Z. Luo, J. Chen, S. Song, X. Yuan, X. Shen, H. Wang, Y. L. Zhao, NPG Asia Mater. 2016, 8, e273.
Y. Sun, K. Gao, L. Zhang, S. Fan, D. T. Leong, M. Guo, J. Xie, [77] S. Shen, D. Jiang, L. Cheng, Y. Chao, K. Nie, Z. Dong, C. J. Kutyreff,
Sci. Rep. 2015, 5, 8669. J. W. Engle, P. Huang, W. Cai, Z. Liu, ACS Nano 2017, 11,
[51] Y. Dou, Y. Guo, X. Li, X. Li, S. Wang, L. Wang, G. Lv, X. Zhang, 9103.
H. Wang, X. Gong, J. Chang, ACS Nano 2016, 10, 2536. [78] S. D. Jo, J. Lee, M. K. Joo, V. J. Pizzuti, N. J. Sherck, S. Choi,
[52] M. Li, Q. Zhao, X. Yi, X. Zhong, G. Song, Z. Chai, Z. Liu, K. Yang, B. S. Lee, S. H. Yeom, S. Y. Kim, S. H. Kim, I. C. Kwon, Y.-Y. Won,
ACS Appl. Mater. Interfaces 2016, 8, 9557. ACS Biomater. Sci. Eng. 2018, 4, 1445.
[53] Y. Yang, L. Zhang, J. Cai, X. Li, D. Cheng, H. Su, J. Zhang, S. Liu, [79] G. Yang, R. Zhang, C. Liang, H. Zhao, X. Yi, S. Shen, K. Yang,
H. Shi, Y. Zhang, C. Zhang, ACS Appl. Mater. Interfaces 2016, 8, L. Cheng, Z. Liu, Small 2018, 14, 1702664.
1718. [80] H. Zhang, B. Chen, H. Jiang, C. Wang, H. Wang, X. Wang, Bioma-
[54] Y. Dou, X. Li, W. Yang, Y. Guo, M. Wu, Y. Liu, X. Li, X. Zhang, terials 2011, 32, 1906.
J. Chang, ACS Appl. Mater. Interfaces 2017, 9, 1263. [81] H. E. Townley, J. Kim, P. J. Dobson, Nanoscale 2012, 4, 5043.
[55] G. Liang, X. Jin, S. Zhang, D. Xing, Biomaterials 2017, 144, 95. [82] D. M. Blake, P.-C. Maness, Z. Huang, E. J. Wolfrum, J. Huang,
[56] S. Liu, H. Li, L. Xia, P. Xu, Y. Ding, D. Huo, Y. Hu, Biomaterials W. A. Jacoby, Sep. Purif. Methods 1999, 28, 1.
2017, 141, 1. [83] R. J. Barnes, R. Molina, J. B. Xu, P. J. Dobson, I. P. Thompson,
[57] N. Ma, P. Liu, N. He, N. Gu, F. G. Wu, Z. Chen, ACS J. Nanopart. Res. 2013, 15, 1432.
Appl. Mater. Interfaces 2017, 9, 31526. [84] Z. Li, X. Pan, T. Wang, P. N. Wang, J. Y. Chen, L. Mi, Nanoscale
[58] N. Ma, F. G. Wu, X. Zhang, Y. W. Jiang, H. R. Jia, H. Y. Wang, Res. Lett. 2013, 8, 96.
Y. H. Li, P. Liu, N. Gu, Z. Chen, ACS Appl. Mater. Interfaces 2017, [85] E. S. Tuchina, V. V. Tuchin, Laser Phys. Lett. 2010, 7, 607.
9, 13037. [86] J. Li, D. Guo, X. Wang, H. Wang, H. Jiang, B. Chen, Nanoscale
[59] M. Misawa, J. Takahashi, Nanomed.: Nanotechnol. Biol. Med. 2011, Res. Lett. 2010, 5, 1063.
7, 604. [87] R. Hariharan, S. Senthilkumar, A. Suganthi, M. Rajarajan, J. Photo-
[60] G. Le Duc, I. Miladi, C. Alric, P. Mowat, E. Brauer-Krisch, chem. Photobiol., B 2012, 116, 56.
A. Bouchet, E. Khalil, C. Billotey, M. Janier, F. Lux, T. Epicier, [88] H. Zhang, Y. Shan, L. Dong, J. Biomed. Nanotechnol. 2014, 10,
P. Perriat, S. Roux, O. Tillement, ACS Nano 2011, 5, 9566. 1450.
[61] W. Rima, L. Sancey, M. T. Aloy, E. Armandy, G. B. Alcantara, [89] H. Zhang, N. Patel, S. Ding, J. Xiong, P. Wu, Biomater. Sci. 2016,
T. Epicier, A. Malchere, L. Joly-Pottuz, P. Mowat, F. Lux, 4, 288.
O. Tillement, B. Burdin, A. Rivoire, C. Boule, I. Anselme-Bertrand, [90] Z. Guo, S. Zhu, Y. Yong, X. Zhang, X. Dong, J. Du, J. Xie, Q. Wang,
J. Pourchez, M. Cottier, S. Roux, C. Rodriguez-Lafrasse, P. Perriat, Z. Gu, Y. Zhao, Adv. Mater. 2017, 29, 1704136.
Biomaterials 2013, 34, 181. [91] P. Ma, H. Xiao, C. Yu, J. Liu, Z. Cheng, H. Song, X. Zhang, C. Li,
[62] S. Dufort, A. Bianchi, M. Henry, F. Lux, G. Le Duc, V. Josserand, J. Wang, Z. Gu, J. Lin, Nano Lett. 2017, 17, 928.
C. Louis, P. Perriat, Y. Cremillieux, O. Tillement, J. L. Coll, Small [92] W. H. Koppenol, Redox Rep. 2001, 6, 229.
2015, 11, 215. [93] L. Yan, Z. Gu, Y. Zhao, Chem.-Asian J. 2013, 8, 2342.

Adv. Mater. 2018, 1802244 1802244  (21 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

[94] A. K. Dutta, S. Das, S. Samanta, P. K. Samanta, B. Adhikary, [125] M. E. Werner, J. A. Copp, S. Karve, N. D. Cummings, R. Sukumar,
P. Biswas, Talanta 2013, 107, 361. C. Li, M. E. Napier, R. C. Chen, A. D. Cox, A. Z. Wang, ACS Nano
[95] Y. Huang, X. Ran, Y. Lin, J. Ren, X. Qu, Chem. Commun. 2015, 51, 2011, 5, 8990.
4386. [126] L. Milas, Clin. Lung Cancer 2002, 3, S29.
[96] A. D. Bokare, W. Choi, J. Hazard. Mater. 2014, 275, 121. [127] F. B. Cui, R. T. Li, Q. Liu, P. Y. Wu, W. J. Hu, G. F. Yue, H. Ding,
[97] X. Zhang, F. G. Wu, P. Liu, N. Gu, Z. Chen, Small 2014, 10, 5170. L. X. Yu, X. P. Qian, B. R. Liu, Cancer Lett. 2014, 346, 53.
[98] E. Ju, K. Dong, Z. Chen, Z. Liu, C. Liu, Y. Huang, Z. Wang, F. Pu, [128] W. Shi, Y. Yuan, M. Chu, S. Zhao, Q. Song, X. Mu, S. Xu, Z. Zhang,
J. Ren, X. Qu, Angew. Chem. Int. Ed. 2016, 55, 11467. K. Yang, J. Biomater. Appl. 2016, 30, 1127.
[99] N. M. Abdallah, E. Noaman, N. A. Eltahawy, A. M. Badawi, [129] M. Creane, C. B. Seymour, S. Colucci, C. Mothersill,
E. Kandil, N. A. Mansour, H. E. Mohamed, Asian Pac. J. Cancer Int. J. Radiat. Biol. 1999, 75, 731.
Prev. 2016, 17, 4367. [130] A. L. Dunne, C. Mothersill, T. Robson, G. D. Wilson, D. G. Hirst,
[100] H. Bi, Y. Dai, J. Xu, R. Lv, F. He, S. Gai, D. Yang, P. Yang, Oncol. Res. 2004, 14, 447.
J. Mater. Chem. B 2016, 4, 5938. [131] P. B. Schiff, S. B. Horwitz, Proc. Natl. Acad. Sci. USA 1980, 77,
[101] H. Fan, G. Yan, Z. Zhao, X. Hu, W. Zhang, H. Liu, X. Fu, T. Fu, 1561.
X. B. Zhang, W. Tan, Angew. Chem. Int. Ed. 2016, 55, 5477. [132] C. Jin, L. Bai, H. Wu, F. Tian, G. Guo, Biomaterials 2007, 28,
[102] Y. Hao, B. Zhang, C. Zheng, M. Niu, H. Guo, H. Zhang, J. Chang, 3724.
Z. Zhang, L. Wang, Y. Zhang, Colloids Surf., B 2017, 151, 384. [133] A. L. Zhang, P. J. Russell, T. Knittel, C. Milross, Prostate 2007, 67,
[103] W. Yin, J. Yu, F. Lv, L. Yan, L. R. Zheng, Z. Gu, Y. Zhao, ACS Nano 1630.
2016, 10, 11000. [134] J. Hiro, Y. Inoue, Y. Toiyama, S. Yoshiyama, K. Tanaka, Y. Mohri,
[104] M. Valodkar, R. N. Jadeja, M. C. Thounaojam, R. V. Devkar, C. Miki, M. Kusunoki, Oncol. Rep. 2010, 24, 1029.
S. Thakore, Mater. Sci. Eng., C 2011, 31, 1723. [135] B. B. Aggarwal, Y.-J. Surh, S. Shishodia, The Molecular Targets
[105] A. L. Harris, Nat. Rev. Cancer 2002, 2, 38. and Therapeutic Uses of Curcumin in Health and Disease, Vol. 595,
[106] J. Liu, Y. Liu, W. Bu, J. Bu, Y. Sun, J. Du, J. Shi, J. Am. Chem. Soc. Springer, Boston, MA, 2007.
2014, 136, 9701. [136] D. Chendil, R. S. Ranga, D. Meigooni, S. Sathishkumar,
[107] W. Fan, W. Bu, B. Shen, Q. He, Z. Cui, Y. Liu, X. Zheng, K. Zhao, M. M. Ahmed, Oncogene 2004, 23, 1599.
J. Shi, Adv. Mater. 2015, 27, 4155. [137] M. Li, Z. Zhang, D. L. Hill, H. Wang, R. Zhang, Cancer Res. 2007,
[108] J. Chen, Q. Chen, C. Liang, Z. Yang, L. Zhang, X. Yi, Z. Dong, 67, 1988.
Y. Chao, Y. Chen, Z. Liu, Nanoscale 2017, 9, 14826. [138] Q. Qiao, Y. Jiang, G. Li, Oncol. Rep. 2013, 29, 380.
[109] J. J. Liu, Q. Chen, W. W. Zhu, X. Yi, Y. Yang, Z. L. Dong, Z. Liu, [139] L. Zhang, X. Ding, J. Huang, C. Jiang, B. Cao, Y. Qian, C. Cheng,
Adv. Funct. Mater. 2017, 27, 1605926. M. Dai, X. Guo, J. Shao, Curr. Signal Transduction Ther. 2015, 10,
[110] F. Gong, J. Chen, X. Han, J. Zhao, M. Wang, L. Feng, Y. Li, Z. Liu, 119.
L. Cheng, J. Mat. Chem. B 2018, 6, 2250. [140] C. X. Wen, Y. Zhou, C. Zhou, Y. F. Zhang, X. Hu, J. Li, H. T. Yin,
[111] Y. Tao, L. Zhu, Y. Zhao, X. Yi, L. Zhu, F. Ge, X. Mou, L. Chen, J. Nanomater. 2017, 2017, 9625909.
L. Sun, K. Yang, Nanoscale 2018, 10, 5114. [141] Y. Liu, Y. Liu, W. Bu, Q. Xiao, Y. Sun, K. Zhao, W. Fan, J. Liu, J. Shi,
[112] T. Paunesku, S. Gutiontov, K. Brown, G. E. Woloschak, Cancer Biomaterials 2015, 49, 1.
Treat. Res. 2015, 166, 151. [142] E. M. Zeman, J. Martin Brown, Int. J. Radiat. Oncol., Biol., Phys.
[113] L. Cheng, C. Yuan, S. Shen, X. Yi, H. Gong, K. Yang, Z. Liu, ACS 1989, 16, 967.
Nano 2015, 9, 11090. [143] X. Liu, Y. Liu, P. Zhang, X. Jin, X. Zheng, F. Ye, W. Chen, Q. Li,
[114] Y. Wang, Y. Y. Wu, Y. J. Liu, J. Shen, L. Lv, L. B. Li, L. C. Yang, Int. J. Nanomed. 2016, 11, 3517.
J. F. Zeng, Y. Y. Wang, L. W. Zhang, Z. Li, M. Y. Gao, Z. F. Chai, [144] C. Schaake-Koning, H. Bartelink, B. H. Adema, L. Schuster-
Adv. Funct. Mater. 2016, 26, 5335. Uitterhoeve, N. V. Zandwijk, Int. J. Radiat. Oncol., Biol., Phys. 1986,
[115] G. Song, Y. Chen, C. Liang, X. Yi, J. Liu, X. Sun, S. Shen, K. Yang, 12, 379.
Z. Liu, Adv. Mater. 2016, 28, 7143. [145] A. A. Geldof, B. J. Slotman, Cancer Lett. 1996, 101, 233.
[116] M. Gao, C. Liang, X. Song, Q. Chen, Q. Jin, C. Wang, Z. Liu, [146] C. Schaake-Koning, W. van den Bogaert, O. Dalesio, J. Festen,
Adv. Mater. 2017, 29, 1701429. J. Hoogenhout, P. van Houtte, A. Kirkpatrick, M. Koolen,
[117] G. Song, C. Ji, C. Liang, X. Song, X. Yi, Z. Dong, K. Yang, Z. Liu, B. Maat, A. Nijs, A. Renaud, P. Rodrigus, L. Schuster-Uitterhoeve,
Biomaterials 2017, 112, 257. J-P. Sculier, N. Van Zandwijk, H. Bartelink, N. Engl. J. Med. 1992,
[118] N. Lu, W. Fan, X. Yi, S. Wang, Z. Wang, R. Tian, O. Jacobson, 326, 524.
Y. Liu, B. C. Yung, G. Zhang, Z. Teng, K. Yang, M. Zhang, G. Niu, [147] R. E. Durand, Radiology 1976, 119, 217.
G. Lu, X. Chen, ACS Nano 2018, 12, 1580. [148] R. E. Durand, N. E. LePard, Radiother. Oncol. 2000, 56, 245.
[119] Y. Huang, Y. Luo, W. Zheng, T. Chen, ACS Appl. Mater. Interfaces [149] X. Tian, S. B. Warner, K. T. Wagner, J. M. Caster, T. Zhang,
2014, 6, 19217. P. Ohana, A. A. Gabizon, A. Z. Wang, Int. J. Radiat. Oncol., Biol.,
[120] W. Roa, X. Zhang, L. Guo, A. Shaw, X. Hu, Y. Xiong, S. Gulavita, Phys. 2016, 96, 547.
S. Patel, X. Sun, J. Chen, R. Moore, J. Z. Xing, Nanotechnology [150] W. Fan, B. Shen, W. Bu, X. Zheng, Q. He, Z. Cui, K. Zhao,
2009, 20, 375101. S. Zhang, J. Shi, Chem. Sci. 2015, 6, 1747.
[121] X. Zhang, N. Liu, Z. Shao, H. Qiu, H. Yao, J. Ji, J. Wang, W. Lu, [151] F. Wenz, S. Greiner, F. Germa, K. Mayer, D. Latz, K. J. Weber,
R. C. Chen, L. Zhang, Nanomed.: Nanotechnol. Biol. Med. 2017, 13, Strahlenther. Onkol. 1999, 175, 2.
1309. [152] Y. Toiyama, Y. Inoue, J. Hiro, E. Ojima, H. Watanabe, Y. Narita,
[122] Y. Li, D. Maciel, J. Rodrigues, X. Shi, H. Tomas, Chem. Rev. 2015, A. Hosono, C. Miki, M. Kusunoki, Cancer Chemother. Pharmacol.
115, 8564. 2007, 59, 733.
[123] X. Y. Zhong, K. Yang, Z. L. Dong, X. Yi, Y. Wang, C. C. Ge, [153] C. X. Geng, Z. C. Zeng, J. Y. Wang, S. Y. Xuan, C. M. Lin, World
Y. L. Zhao, Z. Liu, Adv. Funct. Mater. 2015, 25, 7327. J. Gastroenterol. 2005, 11, 2990.
[124] J. Xu, F. He, Z. Cheng, R. Lv, Y. Dai, A. Gulzar, B. Liu, H. Bi, [154] J. A. Dolling, D. R. Boreham, D. L. Brown, G. P. Raaphorst,
D. Yang, S. Gai, P. Yang, J. Lin, Chem. Mater. 2017, 29, R. E. Mitchel, Mutat. Res. 1999, 433, 127.
7615. [155] J. M. Brown, Br. J. Cancer 1993, 67, 1163.

Adv. Mater. 2018, 1802244 1802244  (22 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

[156] C. Jin, H. Wu, J. Liu, L. Bai, G. Guo, J. Clin. Pharm. Ther. 2007, Cruz-Morcillo, I. Sanchez-Perez, R. Sanchez-Prieto, Cell Cycle 2014,
32, 41. 13, 52.
[157] X. Zhang, H. Yang, K. Gu, J. Chen, M. Rui, G. L. Jiang, [181] L. F. Shen, J. Chen, S. Zeng, R. R. Zhou, H. Zhu, M. Z. Zhong,
Int. J. Nanomed. 2011, 6, 437. R. J. Yao, H. Shen, Mol. Cancer Ther. 2010, 9, 2123.
[158] W. Fan, B. Shen, W. Bu, F. Chen, K. Zhao, S. Zhang, L. Zhou, [182] X. Jin, Q. Li, Q. Wu, P. Li, Y. Matsumoto, Y. Furusawa, L. Gong,
W. Peng, Q. Xiao, H. Xing, J. Liu, D. Ni, Q. He, J. Shi, J. Hao, Z. Dai, J. Radiat. Res. 2011, 52, 335.
J. Am. Chem. Soc. 2013, 135, 6494. [183] S. Reichert, V. Reinboldt, S. Hehlgans, T. Efferth, C. Rodel,
[159] S. Setua, M. Ouberai, S. G. Piccirillo, C. Watts, M. Welland, F. Rodel, Radiother. Oncol. 2012, 103, 394.
Nanoscale 2014, 6, 10865. [184] L. Zhang, W. Ge, K. Hu, Y. Zhang, C. Li, X. Xu, D. He, Z. Zhao,
[160] K. M. Au, Y. Min, X. Tian, L. Zhang, V. Perello, J. M. Caster, J. Zhang, F. Jie, Y. Chen, Y. Zheng, Mol. Biol. Rep. 2012, 39, 89.
A. Z. Wang, ACS Nano 2015, 9, 8976. [185] J. Zou, X. Qiao, H. Ye, Y. Zhang, J. Xian, H. Zhao, S. Liu, Cancer
[161] L. He, H. Lai, T. Chen, Biomaterials 2015, 51, 30. Biother. Radiopharm. 2009, 24, 339.
[162] J. D. Rocca, M. E. Werner, S. A. Kramer, R. C. Huxford-Phillips, [186] J. Feng, J. Zou, L. Li, Y. Zhao, S. Liu, J. Exp. Clin. Cancer Res. 2011,
R. Sukumar, N. D. Cummings, J. L. Vivero-Escoto, A. Z. Wang, 30, 43.
W. Lin, Nanomed.: Nanotechnol. Biol. Med. 2015, 11, 31. [187] F. Rodel, B. Frey, W. Leitmann, G. Capalbo, C. Weiss, C. Rodel,
[163] B. Shen, K. Zhao, S. Ma, D. Yuan, Y. Bai, Chem.-Asian J. 2015, 10, Int. J. Radiat. Oncol., Biol., Phys. 2008, 71, 247.
344. [188] C. T. Yang, J. M. Li, H. H. Weng, Y. C. Li, H. C. Chen, M. F. Chen,
[164] E. C. Wang, Y. Min, R. C. Palm, J. J. Fiordalisi, K. T. Wagner, Cancer Gene Ther. 2010, 17, 120.
N. Hyder, A. D. Cox, J. M. Caster, X. Tian, A. Z. Wang, Biomaterials [189] C. Q. Mao, J. Z. Du, T. M. Sun, Y. D. Yao, P. Z. Zhang, E. W. Song,
2015, 51, 208. J. Wang, Biomaterials 2011, 32, 3124.
[165] K. Kim, K. S. Oh, D. Y. Park, J. Y. Lee, B. S. Lee, I. S. Kim, K. Kim, [190] D. Sutton, S. Kim, X. Shuai, K. Leskov, J. T. Marques,
I. C. Kwon, Y. K. Sang, S. H. Yuk, J. Controlled Release 2016, 228, B. R. Williams, D. A. Boothman, J. Gao, Int. J. Nanomed. 2006, 1,
141. 155.
[166] B. Yu, T. Liu, Y. Du, Z. Luo, W. Zheng, T. Chen, Colloids Surf., B [191] Y. Ping, Z. Jian, Z. Yi, Z. Huoyu, L. Feng, Y. Yuqiong, L. Shixi,
2016, 139, 180. Med. Oncol. 2010, 27, 715.
[167] Y. Chang, L. He, Z. Li, L. Zeng, Z. Song, P. Li, L. Chan, Y. You, [192] J. Chen, S. Zhu, L. Tong, J. Li, F. Chen, Y. Han, M. Zhao, W. Xiong,
X. F. Yu, P. K. Chu, T. Chen, ACS Nano 2017, 11, 4848. BMC Cancer 2014, 14, 114.
[168] Y. Chen, G. Song, Z. Dong, X. Yi, Y. Chao, C. Liang, K. Yang, [193] F. B. Cui, Q. Liu, R. T. Li, J. Shen, P. Y. Wu, L. X. Yu, W. J. Hu,
L. Cheng, Z. Liu, Small 2017, 13, 1602869. F. L. Wu, C. P. Jiang, G. F. Yue, X. P. Qian, X. Q. Jiang, B. R. Liu,
[169] H. Liu, Y. Xie, Y. Zhang, Y. Cai, B. Li, H. Mao, Y. Liu, J. Lu, L. Zhang, Int. J. Nanomed. 2014, 9, 2345.
R. Yu, Biomaterials 2017, 121, 130. [194] F. M. Kievit, Z. R. Stephen, K. Wang, C. J. Dayringer, J. G. Sham,
[170] J. J. Liu, H. R. Wang, X. Yi, Y. Chao, Y. H. Geng, L. G. Xu, K. Yang, R. G. Ellenbogen, J. R. Silber, M. Zhang, Mol. Oncol. 2015, 9,
Z. Liu, Adv. Funct. Mater. 2017, 27, 1703832. 1071.
[171] C. Mirjolet, J. Boudon, A. Loiseau, S. Chevrier, R. Boidot, [195] X. Zhuo, A. Chang, C. Huang, L. Yang, H. Zhao, Y. Wu, Q. Zhou,
A. Oudot, B. Collin, E. Martin, P. A. Joy, N. Millot, G. Crehange, Am. J. Cancer Res. 2015, 5, 1571.
Int. J. Nanomed. 2017, 12, 6357. [196] A. Mizrachi, Y. Shamay, J. Shah, S. Brook, J. Soong,
[172] X. Tian, M. Nguyen, H. P. Foote, J. M. Caster, K. C. Roche, V. K. Rajasekhar, J. L. Humm, J. H. Healey, S. N. Powell, J. Baselga,
C. G. Peters, P. Wu, L. Jayaraman, E. G. Garmey, J. E. Tepper, D. A. Heller, A. Haimovitz-Friedman, M. Scaltriti, Nat. Commun.
S. Eliasof, A. Z. Wang, Cancer Res. 2017, 77, 112. 2017, 8, 14292.
[173] Z. Wang, D. Shao, Z. Chang, M. Lu, Y. Wang, J. Yue, D. Yang, [197] S. Zhang, S. Gupta, T. J. Fitzgerald, A. A. Bogdanov Jr., Nanothera-
M. Li, Q. Xu, W. F. Dong, ACS Nano 2017, 11, 12732. nostics 2018, 2, 1.
[174] R. Zhang, X. Song, C. Liang, X. Yi, G. Song, Y. Chao, Y. Yang, [198] B. W. Henderson, T. J. Dougherty, Photochem. Photobiol. 1992, 55,
K. Yang, L. Feng, Z. Liu, Biomaterials 2017, 138, 13. 145.
[175] J. R. Ashton, K. D. Castle, Y. Qi, D. G. Kirsch, J. L. West, [199] T. J. Dougherty, C. J. Gomer, B. W. Henderson, G. Jori, D. Kessel,
C. T. Badea, Theranostics 2018, 8, 1782. M. Korbelik, J. Moan, Q. Peng, J. Natl. Cancer Inst. 1998, 90,
[176] X. Hu, J. Sun, F. Li, R. Li, J. Wu, J. He, N. Wang, J. Liu, S. Wang, 889.
F. Zhou, X. Sun, D. Kim, T. Hyeon, D. Ling, Nano Lett. 2018, 18, [200] D. Yang, G. Yang, S. Gai, F. He, G. An, Y. Dai, R. Lv, P. Yang,
1196. Nanoscale 2015, 7, 19568.
[177] Y. Sun, H. Miao, S. Ma, L. Zhang, C. You, F. Tang, C. Yang, X. Tian, [201] D. Yang, G. Yang, P. Yang, R. Lv, S. Gai, C. Li, F. He, J. Lin,
F. Wang, Y. Luo, X. Lin, H. Wang, C. Li, Z. Li, H. Yu, X. Liu, Y. Xiao, Adv. Funct. Mater. 2017, 27, 1700371.
Y. Gong, J. Zhang, H. Quan, C. Xie, Cancer Lett. 2018, 418, 27. [202] J. Liu, Y. Yang, W. Zhu, X. Yi, Z. Dong, X. Xu, M. Chen, K. Yang,
[178] X. Yi, L. Chen, J. Chen, D. Maiti, Z. F. Chai, Z. Liu, K. Yang, G. Lu, L. Jiang, Z. Liu, Biomaterials 2016, 97, 1.
Adv. Funct. Mater. 2018, 28, 1705161. [203] X. J. Yu, X. Tang, J. K. He, X. Yi, G. Y. Xu, L. L. Tian, R. Zhou,
[179] S. G. Swisher, J. A. Roth, R. Komaki, J. Gu, J. J. Lee, M. Hicks, C. Zhang, K. Yang, Part. Part. Syst. Charact. 2017, 34, 1600296.
J. Y. Ro, W. K. Hong, J. A. Merritt, K. Ahrar, N. E. Atkinson, [204] M. Zhang, Z. Cui, R. Song, B. Lv, Z. Tang, X. Meng, X. Chen,
A. M. Correa, M. Dolormente, L. Dreiling, A. K. El-Naggar, X. Zheng, J. Zhang, Z. Yao, W. Bu, Biomaterials 2018, 155, 135.
F. Fossella, R. Francisco, B. Glisson, S. Grammer, R. Herbst, [205] W. Chen, J. Zhang, J. Nanosci. Nanotechnol. 2006, 6, 1159.
A. Huaringa, B. Kemp, F. R. Khuri, J. M. Kurie, Z. Liao, [206] Y. F. Liu, W. Chen, S. P. Wang, A. G. Joly, Appl. Phys. Lett. 2008, 92,
T. J. McDonnell, R. Morice, F. Morello, R. Munden, V. 043901.
 Papadimitrakopoulou, K. M. Pisters, J. B. Putnam Jr., [207] C. Zhang, K. Zhao, W. Bu, D. Ni, Y. Liu, J. Feng, J. Shi, Angew.
A. J. Sarabia, T. Shelton, C. Stevens, D. M. Shin, W. R. Smythe, Chem. Int. Ed. 2015, 54, 1770.
A. A. Vaporciyan, G. L. Walsh, M. Yin, Clin. Cancer Res. 2003, 9, [208] S. Clement, W. Deng, E. Camilleri, B. C. Wilson, E. M. Goldys,
93. Sci. Rep. 2016, 6, 19954.
[180] M. L. Valero, F. J. Cimas, L. Arias, P. Melgar-Rojas, E. Garcia, [209] G. Lan, K. Ni, R. Xu, K. Lu, Z. Lin, C. Chan, W. Lin, Angew. Chem.
J. L. Callejas-Valera, J. Garcia-Cano, L. Serrano-Oviedo, M. A. de la Int. Ed. 2017, 56, 12102.

Adv. Mater. 2018, 1802244 1802244  (23 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

[210] R. Lv, C. Zhong, R. Li, P. Yang, F. He, S. Gai, Z. Hou, G. Yang, [241] Y. Yang, Y. Chao, J. J. Liu, Z. L. Dong, W. W. He, R. Zhang, K. Yang,
J. Lin, Chem. Mater. 2015, 27, 1751. M. W. Chen, Z. Liu, NPG Asia Mater. 2017, 9, e344.
[211] X. Yi, K. Yang, C. Liang, X. Y. Zhong, P. Ning, G. S. Song, [242] X. Yu, A. Li, C. Zhao, K. Yang, X. Chen, W. Li, ACS Nano 2017, 11,
D. L. Wang, C. C. Ge, C. Y. Chen, Z. F. Chai, Z. Liu, 3990.
Adv. Funct. Mater. 2015, 25, 4689. [243] J. Li, X. Zu, G. Liang, K. Zhang, Y. Liu, K. Li, Z. Luo, K. Cai, Thera-
[212] S. Wang, X. Li, Y. Chen, X. Cai, H. Yao, W. Gao, Y. Zheng, X. An, nostics 2018, 8, 1042.
J. Shi, H. Chen, Adv. Mater. 2015, 27, 2775. [244] N. Yu, Z. Wang, J. Zhang, Z. Liu, B. Zhu, J. Yu, M. Zhu, C. Peng,
[213] M. Wang, K. Deng, W. Lu, X. Deng, K. Li, Y. Shi, B. Ding, Z. Cheng, Z. Chen, Biomaterials 2018, 161, 279.
B. Xing, G. Han, Z. Hou, J. Lin, Adv. Mater. 2018, 30, 1706747. [245] J. Du, Z. Gu, L. Yan, Y. Yong, X. Yi, X. Zhang, J. Liu, R. Wu, C. Ge,
[214] J. Zhou, Z. Liu, F. Li, Chem. Soc. Rev. 2012, 41, 1323. C. Chen, Y. Zhao, Adv. Mater. 2017, 29, 1701268.
[215] L. Gong, L. Yan, R. Zhou, J. Xie, W. Wu, Z. Gu, J. Mat. Chem. B [246] W. Fan, B. Shen, W. Bu, F. Chen, Q. He, K. Zhao, S. Zhang,
2017, 5, 1873. L. Zhou, W. Peng, Q. Xiao, D. Ni, J. Liu, J. Shi, Biomaterials 2014,
[216] S. H. Yun, S. J. J. Kwok, Nat. Biomed. Eng. 2017, 1, 0008. 35, 8992.
[217] J. Xu, P. Yang, M. Sun, H. Bi, B. Liu, D. Yang, S. Gai, F. He, J. Lin, [247] J. Park, J. Park, E. J. Ju, S. S. Park, J. Choi, J. H. Lee, K. J. Lee,
ACS Nano 2017, 11, 4133. S. H. Shin, E. J. Ko, I. Park, C. Kim, J. J. Hwang, J. S. Lee,
[218] J. Xu, A. Gulzar, Y. Liu, H. Bi, S. Gai, B. Liu, D. Yang, F. He, S. Y. Song, S. Y. Jeong, E. K. Choi, J. Controlled Release 2015, 207,
P. Yang, Small 2017, 13, 1701841. 77.
[219] X. Zhang, P. Yang, Y. Dai, P. a. Ma, X. Li, Z. Cheng, Z. Hou, [248] J. Wang, X. Pang, X. Tan, Y. Song, L. Liu, Q. You, Q. Sun, F. Tan,
X. Kang, C. Li, J. Lin, Adv. Funct. Mater. 2013, 23, 4067. N. Li, Nanoscale 2017, 9, 5551.
[220] R. Lv, P. Yang, F. He, S. Gai, C. Li, Y. Dai, G. Yang, J. Lin, ACS Nano [249] J. J. Qiu, Q. F. Xiao, X. P. Zheng, L. B. Zhang, H. Y. Xing, D. L. Ni,
2015, 9, 1630. Y. Y. Liu, S. J. Zhang, Q. G. Ren, Y. Q. Hua, K. L. Zhao, W. B. Bu,
[221] S. Gai, G. Yang, P. Yang, F. He, J. Lin, D. Jin, B. Xing, Nano Today Nano Res. 2015, 8, 3580.
2018, 19, 146. [250] Z. Chen, Q. Wang, H. Wang, L. Zhang, G. Song, L. Song, J. Hu,
[222] Q. Xiao, X. Zheng, W. Bu, W. Ge, S. Zhang, F. Chen, H. Xing, H. Wang, J. Liu, M. Zhu, D. Zhao, Adv. Mater. 2013, 25, 2095.
Q. Ren, W. Fan, K. Zhao, Y. Hua, J. Shi, J. Am. Chem. Soc. 2013, [251] P. Kalluru, R. Vankayala, C. S. Chiang, K. C. Hwang, Angew. Chem.
135, 13041. Int. Ed. 2013, 52, 12332.
[223] J. F. Hainfeld, L. Lin, D. N. Slatkin, F. Avraham Dilmanian, [252] C. K. K. Nair, D. K. Parida, T. Nomura, J. Radiat. Res. 2001, 42, 21.
T. M. Vadas, H. M. Smilowitz, Nanomed.: Nanotechnol. Biol. Med. [253] Y. Qiu, Z. Wang, A. C. Owens, I. Kulaots, Y. Chen, A. B. Kane,
2014, 10, 1609. R. H. Hurt, Nanoscale 2014, 6, 11744.
[224] P. Li, Y. W. Shi, B. X. Li, W. C. Xu, Z. L. Shi, C. Zhou, S. Fu, J. Nano- [254] J. Wang, X. Cui, H. Li, J. Xiao, J. Yang, X. Mu, H. Liu, Y.-M. Sun,
biotechnol. 2015, 13, 52. X. Xue, C. Liu, X.-D. Zhang, D. Deng, X. Bao, Nano Res. 2018, 11,
[225] G. Song, C. Liang, H. Gong, M. Li, X. Zheng, L. Cheng, K. Yang, 2821.
X. Jiang, Z. Liu, Adv. Mater. 2015, 27, 6110. [255] B. Daroczi, G. Kari, M. F. McAleer, J. C. Wolf, U. Rodeck,
[226] L. Cheng, S. Shen, S. Shi, Y. Yi, X. Wang, G. Song, K. Yang, A. P. Dicker, Clin. Cancer Res. 2006, 12, 7086.
G. Liu, T. E. Barnhart, W. Cai, Z. Liu, Adv. Funct. Mater. 2016, 26, [256] Q. Zhao, Y. Li, J. Xu, R. Liu, W. Li, Int. J. Radiat. Biol. 2005, 81, 169.
2185. [257] J. Xie, Y. Yong, X. Dong, J. Du, Z. Guo, L. Gong, S. Zhu, G. Tian,
[227] N. Ma, Y. W. Jiang, X. Zhang, H. Wu, J. N. Myers, P. Liu, H. Jin, S. Yu, Z. Gu, Y. Zhao, ACS Appl. Mater. Interfaces 2017, 9, 14281.
N. Gu, N. He, F. G. Wu, Z. Chen, ACS Appl. Mater. Interfaces 2016, [258] J. Xie, N. Wang, X. Dong, C. Wang, Z. Du, L. Mei, Y. Yong,
8, 28480. C. Huang, Y. Li, Z. Gu, Y. Zhao, ACS Appl. Mater. Interfaces 2018,
[228] F. Mao, L. Wen, C. Sun, S. Zhang, G. Wang, J. Zeng, Y. Wang, https://doi.org/10.1021/acsami.8b00949.
J. Ma, M. Gao, Z. Li, ACS Nano 2016, 10, 11145. [259] H. Li, Z. Y. Yang, C. Liu, Y. P. Zeng, Y. H. Hao, Y. Gu, W. D. Wang,
[229] J. Wang, X. Tan, X. Pang, L. Liu, F. Tan, N. Li, ACS R. Li, Free Radic. Biol. Med. 2015, 87, 26.
Appl. Mater. Interfaces 2016, 8, 24331. [260] J. Colon, L. Herrera, J. Smith, S. Patil, C. Komanski, P. Kupelian,
[230] L. Wen, L. Chen, S. Zheng, J. Zeng, G. Duan, Y. Wang, G. Wang, S. Seal, D. W. Jenkins, C. H. Baker, Nanomed.: Nano-
Z. Chai, Z. Li, M. Gao, Adv. Mater. 2016, 28, 5072. technol. Biol. Med. 2009, 5, 225.
[231] A. W. Zhang, W. H. Guo, Y. F. Qi, J. Z. Wang, X. X. Ma, D. X. Yu, [261] J. Colon, N. Hsieh, A. Ferguson, P. Kupelian, S. Seal, D. W. Jenkins,
Nanoscale Res. Lett. 2016, 11, 279. C. H. Baker, Nanomed.: Nanotechnol. Biol. Med. 2010, 6, 698.
[232] J. Chen, M. F. Li, X. Yi, Q. Zhao, L. Chen, C. Yang, J. C. Wu, [262] X. D. Zhang, J. Zhang, J. Wang, J. Yang, J. Chen, X. Shen, J. Deng,
K. Yang, Part. Part. Syst. Charact. 2017, 34, 1600330. D. Deng, W. Long, Y. M. Sun, C. Liu, M. Li, ACS Nano 2016, 10,
[233] X. Cheng, Y. Yong, Y. Dai, X. Song, G. Yang, Y. Pan, C. Ge, Thera- 4511.
nostics 2017, 7, 4087. [263] X. T. Bai, J. Y. Wang, X. Y. Mu, J. Yang, H. X. Liu, F. J. Xu, Y. Q. Jing,
[234] R. Hu, M. Zheng, J. Wu, C. Li, D. Shen, D. Yang, L. Li, M. Ge, L. F. Liu, X. H. Xue, H. T. Dai, Q. Liu, Y. M. Sun, C. L. Liu,
Z. Chang, W. Dong, Nanomaterials 2017, 7, 111. X. D. Zhang, ACS Biomater. Sci. Eng. 2017, 3, 460.
[235] D. Huo, S. Liu, C. Zhang, J. He, Z. Zhou, H. Zhang, Y. Hu, ACS [264] H. X. Liu, J. Y. Wang, Y. Q. Jing, J. Yang, X. T. Bai, X. Y. Mu,
Nano 2017, 11, 10159. F. J. Xu, X. H. Xue, L. F. Liu, Y. M. Sun, Q. Liu, H. T. Dai, C. L. Liu,
[236] A. Li, X. Li, X. Yu, W. Li, R. Zhao, X. An, D. Cui, X. Chen, W. Li, Bio- X. D. Zhang, Part. Part. Syst. Charact. 2017, 34, 1700035.
materials 2017, 112, 164. [265] X. D. Zhang, Y. Jing, S. Song, J. Yang, J. Y. Wang, X. Xue,
[237] Y. Li, Y. Sun, T. Cao, Q. Su, Z. Li, M. Huang, R. Ouyang, H. Chang, Y. Min, G. Park, X. Shen, Y. M. Sun, U. Jeong, Nanomed.: Nano-
S. Zhang, Y. Miao, Nanoscale 2017, 9, 14364. technol. Biol. Med. 2017, 13, 1597.
[238] X. Liu, X. Zhang, M. Zhu, G. Lin, J. Liu, Z. Zhou, X. Tian, Y. Pan, [266] Y. Min, J. M. Caster, M. J. Eblan, A. Z. Wang, Chem. Rev. 2015, 115,
ACS Appl. Mater. Interfaces 2017, 9, 279. 11147.
[239] A. Neshastehriz, M. Tabei, S. Maleki, S. Eynali, A. Shakeri-Zadeh, [267] D. Bobo, K. J. Robinson, J. Islam, K. J. Thurecht, S. R. Corrie,
J. Photochem. Photobiol. B 2017, 172, 52. Pharm. Res. 2016, 33, 2373.
[240] S. D. Shen, Y. Chao, Z. L. Dong, G. L. Wang, X. Yi, G. S. Song, [268] L. Maggiorella, G. Barouch, C. Devaux, A. Pottier, E. Deutsch,
K. Yang, Z. Liu, L. Cheng, Adv. Funct. Mater. 2017, 27, 1700250. J. Bourhis, E. Borghi, L. Levy, Future Oncol. 2012, 8, 1167.

Adv. Mater. 2018, 1802244 1802244  (24 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

[269] A. Pottier, E. Borghi, L. Levy, Biochem. Biophys. Res. Commun. E. Porcel, S. Lacombe, G. Le Duc, E. Deutsch, J. L. Perfettini,
2015, 468, 471. A. Detappe, C. Verry, R. Berbeco, K. T. Butterworth, S. J. McMahon,
[270] A. C. Anselmo, S. Mitragotri, Bioeng. Transl. Med. 2016, 1, 10. K. M. Prise, P. Perriat, O. Tillement, Br. J. Radiol. 2014, 87,
[271] P. Sánchez-Moreno, J. Ortega-Vinuesa, J. Peula-García, J. Marchal, 20140134.
H. Boulaiz, Curr. Drug Targets 2018, 19, 339. [278] A. Carmona, S. Roudeau, B. L’Homel, F. Pouzoulet, S. Bonnet-
[272] S. Bonvalot, C. Le Pechoux, T. De Baere, G. Kantor, X. Buy, Boissinot, Y. Prezado, R. Ortega, Anal. Biochem. 2017, 523, 50.
E. Stoeckle, P. Terrier, P. Sargos, J. M. Coindre, N. Lassau, R. Ait [279] L. Sancey, S. Kotb, C. Truillet, F. Appaix, A. Marais, E. Thomas,
Sarkouh, M. Dimitriu, E. Borghi, L. Levy, E. Deutsch, J. C. Soria, B. van der Sanden, J. P. Klein, B. Laurent, M. Cottier, R. Antoine,
Clin. Cancer Res. 2017, 23, 908. P. Dugourd, G. Panczer, F. Lux, P. Perriat, V. Motto-Ros,
[273] E. Brun, C. Sicard-Roselli, Radiat. Phys. Chem. 2016, 128, O. Tillement, ACS Nano 2015, 9, 2477.
134. [280] J. Zhao, M. Zhou, C. Li, Cancer Nanotechnol. 2016, 7, 9.
[274] R. L. Haas, A. B. Miah, C. LePechoux, T. F. DeLaney, E. H. Baldini, [281] C. Verry, S. Dufort, E. L. Barbier, O. Montigon, M. Peoc’h,
K. Alektiar, B. O’Sullivan, Radiother. Oncol. 2016, 119, 14. P. Chartier, F. Lux, J. Balosso, O. Tillement, L. Sancey, G. Le Duc,
[275] N. G. Zaorsky, B. J. Davis, P. L. Nguyen, T. N. Showalter, Nanomedicine 2016, 11, 2405.
P. J. Hoskin, Y. Yoshioka, G. C. Morton, E. M. Horwitz, [282] S. Kotb, J. Piraquive, F. Lamberton, F. Lux, M. Verset, V. Di Cataldo,
Nat. Rev. Urol. 2017, 14, 415. H. Contamin, O. Tillement, E. Canet-Soulas, L. Sancey, Sci. Rep.
[276] S. Kotb, A. Detappe, F. Lux, F. Appaix, E. L. Barbier, V. L. Tran, 2016, 6, 35053.
M. Plissonneau, H. Gehan, F. Lefranc, C. Rodriguez-Lafrasse, [283] A. S. Wozny, M. T. Aloy, G. Alphonse, N. Magne, M. Janier,
C. Verry, R. Berbeco, O. Tillement, L. Sancey, Theranostics 2016, 6, O. Tillement, F. Lux, M. Beuve, C. Rodriguez-Lafrasse, Nanomed.:
418. Nanotechnol. Biol. Med. 2017, 13, 2655.
[277] L. Sancey, F. Lux, S. Kotb, S. Roux, S. Dufort, A. Bianchi, [284] P. Hu, D. Cheng, T. Huang, A. B. Banizs, J. Xiao, G. Liu, Q. Chen,
Y. Cremillieux, P. Fries, J. L. Coll, C. Rodriguez-Lafrasse, M. Janier, Y. Wang, J. He, H. Shi, Nanoscale Res. Lett. 2017, 12, 523.
M. Dutreix, M. Barberi-Heyob, F. Boschetti, F. Denat, C. Louis, [285] G. Tian, X. Zhang, Z. Gu, Y. Zhao, Adv. Mater. 2015, 27, 7692.

Adv. Mater. 2018, 1802244 1802244  (25 of 25) © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

You might also like