ArticlePDF Available

Fe3O4/Ag/Bi2MoO6 Photoactivatable Nanozyme for Self-Replenishing and Sustainable Cascaded Nanocatalytic Cancer Therapy

Wiley
Advanced Materials
Authors:

Abstract and Figures

Catalytic cancer therapy based on nanozymes has recently attracted much interest. But the types of the current nanozymes are limited and their efficiency is usually compromised and not sustainable in the tumor microenvironment (TME). Therefore, combination therapy involving additional therapeutics is often necessary and the resulting complication may jeopardize the practical feasibility. Herein, an unprecedented “all‐in‐one” Fe3O4/Ag/Bi2MoO6 nanoparticle (FAB NP) is rationally devised to achieve synergistic chemodynamic, photodynamic, photothermal therapy with guidance by magnetic resonance, photoacoustic, and photothermal imaging. Based on its manifold nanozyme activities (mimicking peroxidase, catalase, superoxide dismutase, glutathione oxidase) and photodynamic property, cascaded nanocatalytic reactions are enabled and sustained in TME for outstanding therapeutic outcomes. The working mechanisms underlying the intraparticulate interactions, sustainability, and self‐replenishment arising from the coupling between the nanocatalytic reactions and nanozyme activities are carefully revealed, providing new insights into the design of novel nanozymes for nanocatalytic therapy with high efficiency, good specificity, and low side effects. This article is protected by copyright. All rights reserved
This content is subject to copyright. Terms and conditions apply.
2106996 (1 of 10) ©  Wiley-VCH GmbH
www.advmat.de
ReseaRch aRticle
Fe3O4/Ag/Bi2MoO6 Photoactivatable Nanozyme for
Self-Replenishing and Sustainable Cascaded Nanocatalytic
Cancer Therapy
Changyu Cao, Hai Zou, Nan Yang, Hui Li, Yu Cai, Xuejiao Song,* Jinjun Shao,
Peng Chen,* Xiaozhou Mou,* Wenjun Wang, and Xiaochen Dong*
C. Cao, N. Yang, H. Li, X. Song, J. Shao, X. Dong
Key Laboratory of Flexible Electronics and Institute of Advanced Materials
School of Physical and Mathematical Sciences
Nanjing Tech University
Nanjing , China
E-mail: xjsong@njtech.edu.cn; iamxcdong@njtech.edu.cn
W. Wang
School of Physical Science and Information Technology
Liaocheng University
Liaocheng , China
H. Zou
Department of Oncology
Shanghai Medical College
Fudan University
Shanghai , China
DOI: 10.1002/adma.202106996
good specificity, and low side-eect.[–]
The recent advance in nanochemistry
and nanocatalysis has stimulated rapid
development of nanomaterials with
enzyme-mimicking activities.[–] These
nanozymes generally outperform natural
enzymes because of excellent stability,
high catalytic activity, multifunctionality,
and low cost. Moreover, nanozymes may
be designed to be photoactivatable for
localized treatment.[–]
But most of current nanozymes mimic
peroxidase (POD) and a few simulate
catalase (CAT) to convert HO into ·OH
for chemodynamic therapy (CDT) or into
oxygen to enhance O-dependent photo-
dynamic therapy (PDT).[–] The eec-
tiveness of these strategies is, however,
not sustainable owing to limited supply
of HO and suppression of the gener-
ated reactive oxygen species (ROS) by
the elevated antioxidants in TME.[–]
In addition, most nanozymes are not
able to catalyze multielectron reactions, such as simulating
superoxide dismutase (SOD) to reduce O to HO. Therefore,
combination therapy involving additional therapeutics is nec-
essary.[–] This however complicates fabrication, encapsula-
tion, delivery, and release processes, making it not practical
Catalytic cancer therapy based on nanozymes has recently attracted much
interest. However, the types of the current nanozymes are limited and their
eciency is usually compromised and not sustainable in the tumor micro-
environment (TME). Therefore, combination therapy involving additional ther-
apeutics is often necessary and the resulting complication may jeopardize the
practical feasibility. Herein, an unprecedented “all-in-one” Fe3O4/Ag/Bi2MoO6
nanoparticle (FAB NP) is rationally devised to achieve synergistic chemo-
dynamic, photodynamic, photothermal therapy with guidance by magnetic
resonance, photoacoustic, and photothermal imaging. Based on its manifold
nanozyme activities (mimicking peroxidase, catalase, superoxide dismutase,
glutathione oxidase) and photodynamic property, cascaded nanocatalytic
reactions are enabled and sustained in TME for outstanding therapeutic out-
comes. The working mechanisms underlying the intraparticulate interactions,
sustainability, and self-replenishment arising from the coupling between
the nanocatalytic reactions and nanozyme activities are carefully revealed,
providing new insights into the design of novel nanozymes for nanocatalytic
therapy with high eciency, good specificity, and low side eects.
1. Introduction
Catalytic cancer therapy, enabled by enzymatic reactions in
the tumor microenvironment (TME), has attracted increasing
interest owing to its potential to achieve high eectiveness,
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/./adma..
Y. Cai, X. Mou
Clinical Research Institute
Zhejiang Provincial People’s Hospital
Aliated People’s Hospital
Hangzhou Medical College
Hangzhou , China
E-mail: mouxz@zju.edu.cn
P. Chen
School of Chemical and Biomedical Engineering
Lee Kong Chian School of Medicine
Nanyang Technological University
 Nanyang Drive, Singapore , Singapore
E-mail: chenpeng@ntu.edu.sg
Adv. Mater. 2021, 33, 
©  Wiley-VCH GmbH
2106996 (2 of 10)
www.advmat.dewww.advancedsciencenews.com
due to poor reproducibility, reduced eectiveness, high side-
eects, and high cost.
BiMoO (BMO) NP has been widely used as photocatalysts
to decompose organic pollutants.[] Its potential for photody-
namic therapy was suggested based on in vitro experiments.[]
However, the UV absorption property of BMO NP makes its
clinical use impractical because short-wavelength lights cannot
penetrate through the skin and cause phototoxicity. To tackle
the aforementioned issues, we rationally designed a FeO/Ag/
BiMoO nanoparticle (FAB NP) which simultaneously pos-
sesses high POD, CAT, SOD, glutathione oxidase (GSHOD),
and photodynamic activities. Doping of FeO and Ag NPs
endows UV-absorbing BMO NP, which with strong NIR-II
absorption, greatly enhanced photocatalytic activities, ferro-
magnetic and photothermal eects which enable magnetic
resonance (MR), photoacoustic (PA), and photothermal (PT)
imaging to guide the nanocatalytic therapy. Based on both in
vitro and in vivo experiments, we demonstrated that such an
unprecedented photoactivatable “all-in-one” nanoparticulate
system enables synergistic chemodynamic, photodynamic,
and photothermal therapy as illustrated in Figure 1. Further,
the working mechanisms underlying the intraparticulate
interactions, sustainability, and self-replenishment arising from
the coupling between the cascaded nanocatalytic reactions and
manifold nanozyme activities are carefully revealed. This study
provides new insights to design novel nanozymes for catalytic
cancer therapy with high eciency, good specificity, and low
side eects, as well as for other theranostic applications.
2. Results and Discussion
FAB NPs were fabricated by a three-step synthesis (Figure ),
involving hydrothermal synthesis of BMO NPs, photoreduc-
tion of Ag NPs, and solvothermal doping of FeO together with
surface capping with hydrophilic polyvinylpyrrolidone (PVP)
(Figure2a; and Figure S, Supporting information). As shown
by transmission electron microscopy (TEM), FAB NPs have an
aspect ratio of . with an average length of . ± . nm
(n= ) (Figure a, b). Such size is favorable for nanopar-
ticle retention in tumor tissues via enhanced permeability and
retention (EPR) eect. FAB NPs can well and stably disperse
in various media because of the small size and PVP coating
(Figure S, Supporting Information). The high-resolution TEM
Adv. Mater. 2021, 33, 
Figure 1. Schematic illustration for the fabrication of FAB nanozyme and mechanisms of synergistic CDT/PDT/PTT therapy.
©  Wiley-VCH GmbH
2106996 (3 of 10)
www.advmat.dewww.advancedsciencenews.com
(HRTEM) reveals three crystalline lattice spacings of ., .,
and .nm, corresponding to (), (), and () planes of
Ag, FeO, and BMO NPs, respectively. These high-index facets
were also identified by the selected area electron diraction
(SAED) pattern (Figurea).
The elemental mapping of FAB NPs demonstrates the
homogenous distribution of Bi, Mo, O, Fe, and Ag (Figurec).
The atomic composition is also confirmed by X-ray photo-
electron spectroscopy (XPS) (Figure d; and Figure S, Sup-
porting Information). The high-resolution XPS Mo spectrum in
Figuree exhibits four major peaks derived from Mo d/ and
Mo d/, which can be deconvolved into the double peaks at
. and . eV from Mo+ and double peaks at . and
.eV from Mo+. The XPS spectrum of Fe can be fitted by
eight peaks (Figuref). Among them, the peaks at ., .,
., and . eV are assigned to Fe+, while the other four
peaks at ., ., ., and . eV are ascribed to Fe+.
Besides, as shown in Figure S (Supporting Information), in
the presence of oxidant (HO), the binding energy of FAB NP
upshifts, indicating the transition of Fe+/Mo+ to the higher
valence state (Fe+/Mo+); while in the presence of reductant
(GSH), the binding energy shifts downward, indicating the
reduction of Fe+/Mo+ to Fe+/Mo+. Taken together, the exist-
ence of Fe+/Fe+ and Mo+/Mo+ redox couples endow the nan-
oparticle with enzymatic properties.
As schematically proposed in Figure 3a, electrons are greatly
enriched in the conduction band of BMO NP because of pho-
toexcitation, migration of excited electrons from FeO NP,
Adv. Mater. 2021, 33, 
Figure 2. Characterization of FAB NPs. a) TEM, HRTEM, SAED pattern. b) Size distribution. c) Elemental mapping. d) XPS survey spectrum. e,f) High-
resolution XPS spectra of Mo d and Fe p.
©  Wiley-VCH GmbH
2106996 (4 of 10)
www.advmat.dewww.advancedsciencenews.com
and electron injection from Ag NP with lower work function,
whereby eciently catalyzing O reduction. On one hand, O
is reduced to ·O via one electron transfer and generated
·O is then oxidized toO by down-transition of Fe+/Mo+
to Fe+/Mo+ in FAB NP for photodynamic therapy. On the
other hand, O is reduced to HO via a two-electron pathway
mimicking superoxide dismutase (SOD). Coupling with up-
transition of Fe+/Mo+ to Fe+/Mo+, HO is then converted
to ·OH (mimicking peroxidase – POD) for chemodynamic
therapy or O (mimicking catalase – CAT) to sustain photo-
dynamic therapy in hypoxic TME. The eciency of PDT and
CDT are maintained by catalytic depletion of antioxidant GSH
via down-transition of Fe+/Fe+ and Mo+/Mo+ redox couples,
mimicking glutathione oxidase (GSHOD). Finally, the nonradi-
ative relaxation of photoexcited electrons enables photothermal
therapy. In sum, a cascade of nanocatalytic reactions (Figure S,
Adv. Mater. 2021, 33, 
Figure 3. Photo-enhanced catalytic activities of FAB nanozymes. FAB NP concentration is  µg mL–. (a) Schematic illustration of the working
mechanisms (GSH: glutathione, GSSG: glutathione disulfide). Orange arrows represent oxidation reactions. Green arrows represent reduction reac-
tions. Arrows with frames indicate the valence transition reactions. (b) O generation curve at pH . with or without HO addition or nm
laser irradiation (L). (c) HO production curves by FAB NPs and FeO/BMO NPs at pH = . or .. (d) Degradation of MB by FAB NPs. (e) Light
adsorption of DPBF decreases due to FAB-NP inducedO generation under laser irradiation with dierent durations (-s). (f) GSH depletion by FAB
NPs with dierent concentrations (-µg mL–).
©  Wiley-VCH GmbH
2106996 (5 of 10)
www.advmat.dewww.advancedsciencenews.com
Supporting Information) is triggered in TME to continuously
produce cytotoxic ·OH andO, which are sustained by photo-
stimulation, coupling between reactions and nanozyme activi-
ties, and cycling between Fe+/Mo+ and Fe+/Mo+.
The CAT-like activity of FAB nanozyme was evidenced by
quick increase of dissolved O upon adding them into HO at
pH . (analogous to acidic TME) (Figureb). As expected, O
generation was enhanced by exposure to NIR-II laser irradiation
( nm), testifying the light enhancement of the nanozyme
activity. To assess the SOD-like activity of FAB nanozyme under
dierent conditions, KI was used as the detection probe which
reacts with HO to generate I. As presented in Figure c,
with nm laser irradiation, much HO ( µmol L) was
produced at pH .. In contrast, no obvious generation of
HO was observed without laser irradiation or in the absence
of Ag, implying their critical role to enrich free electrons for
catalyzing the two-electron reduction of O. Further, the POD-
likely activity of FAB nanozyme to reduce HO into ·OH for
chemodynamic therapy (CDT) was probed by methylene blue
(MB) whose light adsorption decreases upon reaction with
·OH. As shown in Figured, FAB NP is more POD-active in
acidic environment and with laser stimulation. Production of
·OH via Fenton-like reaction was further confirmed by its char-
acteristic signals in electron spin resonance (ESR) (Figure Sa,
Supporting Information).
As evidenced by the decrease of light absorption of
,-diphenylisobenzofuran (DPBF) and the characteristic sig-
nals in ESR, FAB NPs were also able to produce abundantO
upon NIR-II irradiation (Figuree; and Figures Sb and S, Sup-
porting Information). Thus, FAB NP can serve as an eective
photosensitizer for photodynamic therapy (PDT). Noteworthy,
the eciency of the current chemodynamic and photodynamic
therapies are largely compromised by the antioxidant defense
system in tumor tissues, mainly because highly elevated GSH
molecules neutralize the generated ·OH andO. Interestingly,
FAB nanozyme can also mimic GSH oxidase (GSHOD) to
deplete GSH, thereby self-enhancing its CDT and PDT. This is
supported by the observation that the adsorption of ,-dithiobis-
(-nitrobenzoic acid) decreases upon reaction with the oxidized
GSH in a FAB concentration-dependent manner (Figuref).
As shown in Figure S (Supporting Information), the band-
gaps of BMO, Ag/BMO, and FeO/Ag/BMO are ., .,
and . eV, respectively. Doping of Ag NP, which introduces
impurity energy level, and FeO NP, which has a higher
valence band level than that of BMO, greatly narrows the eec-
tive bandgap, and thus transforms UV-absorbing BMO NP to
a hybrid NP with strong adsorption in both NIR-I and NIR-II
regions (Figure S, Supporting Information). Ag@FeO NPs
have been previously synthesized for photothermal and chemo-
dynamic cancer therapy.[,] But, because of the wider bandgap
than FAB NP, it is not capable of NIR-II adsorption. Compared
with the commonly used nm laser in NIR-I, nm laser
in NIR-II used in this study is more desirable due to its higher
maximum permissible exposure (vs . W cm) and deeper
tissue penetration depth. Being suitable for photothermal
therapy (PTT), FAB NP exhibits concentration-, laser power
density-, and irradiation time-dependent photothermal eect
under  nm laser irradiation, with a photothermal conver-
sion eciency of .% and outstanding stability (Figure S,
Supporting Information). Taken together, utilizing the multiple
nanozyme activities (mimicking CAT, SOD, POD, GSHOD),
photothermal eect, and ferromagnetic property from FeO
NP, FAB NP can serve as an unprecedented “all-in-one” thera-
peutic agent for synergistic CDT, PDT, and PTT guided by mul-
timodal (magnetic resonance, photoacoustic, photothermal)
imaging.
Using T murine breast cancer cells, it was found that
green fluorescence FITC-labeled FAB NPs, but not free
FITC molecules, were eectively endocytosed to enter the
nucleus (Figure S, Supporting Information) and caused a
quick increase of intracellular HO under laser irradiation
(Figure 4a) and depletion of GSH (Figureb). The intracellular
O level was reported by tris (,-diphenyl-,-phenanthroline)
ruthenium(II) dichloride (RDPP), whose fluorescence can be
quenched by O. As depicted in Figure c, addition of HO
(mimicking TME) plus laser greatly stimulated intracellular
O production by FAB nanozyme. Altogether, these in vitro
observations suggest that FAB nanozyme can eectively modu-
late TME to boost ROS generation by suppressing antioxidant
defense, producing HO, and relieving hypoxia.
FAB NP exhibited no apparent toxicity to human immor-
talized keratinocytes (HaCaTs) whereas it was cytotoxic to T
tumor cells in a dose-dependent manner (Figure d), pre-
sumably because tumor cells have higher endogenous HO
than normal cells. And the tumor-killing eect was greatly
enhanced by laser irradiation or adding HO (Figure e).
With µg mL nanozyme, nm irradiation ( W cm,
min), and  mM extracellular HO, essentially all T cells
were killed. Live/dead cell staining experiment (Figuref) and
flow cytometry (Figure g) further confirmed the CDT and
PDT eects of FAB NP toward tumor cells and the enhance-
ments by NIR-II irradiation and HO. Moreover, western blot-
ting analysis showed that Cas- expression was significantly
induced by FAB NP under laser irradiation, suggesting the
occurrence of Cas- dependent apoptosis. (Figure S, Sup-
porting Information). Intracellular ROS level was reported by
fluorescence increase of a cell-permeable probe (,-dichloro-
fluorescein diacetate). Consistently, with laser and external
HO, FAB NP became very potent to induce intracellular
ROS (Figureh,i).
The hemocompatibility of FAB NP was firstly confirmed
(Figure S, Supporting Information) before using T tumor-
bearing Balb/c mice to investigate the in vivo therapeutic per-
formance of FAB nanozyme. After intravenous (i.v.) injection,
the pharmacokinetic profiles of NPs were investigated and
the blood circulation half-time was determined to be . h
(Figure 5a). Endowed by FeO, FAB NP has a typical ferromag-
netic behavior with saturation magnetization of  emu g
(Figureb) and r relaxivity of . ×   s (Figure c),
therefore can act as a good contrast agent for MRI. After i.v.
injection of FAB NPs, the MR signal of tumor site increased
over time and reached the maximum at  h with a time con-
stant of  h (Figure d,e), indicating the dynamic accumu-
lation of NPs through the EPR eect. This is similar to the
dynamics revealed by PAI based on the photothermal eect of
FAB NP (Figuref). FAB NP accumulation in the tumor was
further confirmed by iron ion-enabled Prussian blue staining
(Figureg).
Adv. Mater. 2021, 33, 
©  Wiley-VCH GmbH
2106996 (6 of 10)
www.advmat.dewww.advancedsciencenews.com
Adv. Mater. 2021, 33, 
The accumulation of FAB NPs in the heart, liver, spleen,
lung, kidney, and tumor site at dierent time points (– h)
was quantified by inductively coupled plasma-optical emis-
sion spectrometry (ICP-OES). As shown in Figureh, FAB
NPs were also retained in these major organs except heart
and reached maximum accumulation in the tumor at  h
before being metabolized. Therefore, in the following experi-
ments, laser stimulation was applied at this optimal time
point.
The tumor temperature of FAB NP injected mouse rose
to . °C in  min upon laser irradiation, indicating the
realization of photothermal therapy (Figure 6a,b). Without
lasering, the nonsustainable CDT enabled by FAB NPs sup-
pressed tumor growth to an extent. In comparison, min laser
irradiation once a day significantly inhibited tumor growth
(% reduction comparing to control) and irradiation twice a
day completely eliminated the tumor in  days, testifying the
outstanding potency of FAB NPs due to sustained and syner-
gistic combination of CDT, PDT, and PTT (Figurec–e).
Tumor tissues were harvested and stained with dihydro-
ethidium to report ROS levels. As expected, FAB NP alone
increased ROS in tumors due to chemodynamic generation
Figure 4. Intracellular catalyzes and cytotoxicity by FAB NPs. Unless otherwise stated, cells are T cells and FAB NP concentration is µg mL–.
(a) Intracellular HO generation under nm laser irradiation. (b) GSH depletion with dierent concentrations of FAB NPs ( to µg mL–).
(c) Cellular O level reflected by RDPP staining. Scar bar: µm. (d) Relative cell viability of HaCaTs and T cells after incubation with FAB NPs of
dierent concentrations. (e) Relative cell viability of T cells treated with FAB NPs of dierent concentrations, without or with HO addition and laser
irradiation. (f) Fluorescence images of T cells treated by FAB NPs and then stained by PI (red, dead cells) and Ca-AM (green, live cells). Scar bar:
µm. (g) Flow cytometry using Annexin-V-FTIC/PI assay. (h) Fluorescence images of T cells stained by DCFH-DA to indicate nanoparticle-induced
ROS generation. Scar bar: µm. Laser irradiation was applied only to the right sides of the dashed lines. (i) Corresponding quantitative analysis of
ROS generation.
©  Wiley-VCH GmbH
2106996 (7 of 10)
www.advmat.dewww.advancedsciencenews.com
Adv. Mater. 2021, 33, 
of ·OH, while additional laser irradiation caused much more
ROS generation because of additional photodynamic pro-
duction ofO and photodriven self-replenishment of the
coupled cascaded nanocatalytic reactions (Figure f). Apop-
tosis of tumor cells caused by the photoactivatable nano-
catalytic therapy was confirmed by Hematoxylin and eosin
(H&E) staining (Figure g). Moreover, inhibition of tumor
cell proliferation was evidenced by the Ki- immunohisto-
chemistry assay (Figureh; and Figure S in the Supporting
Information). Noteworthy, the mouse body weight was not sig-
nificantly aected by laser or FAB NP treatment (Figure S,
Supporting Information). Based on H&E staining, no notice-
able damage was observed in heart, liver, spleen, lung, or
kidney (Figure S, Supporting Information). Blood bio-
chemistry assay and complete blood panel analysis after the
treatments also did not show any abnormality (Figure S,
Supporting Information). Taken together, the catalytic therapy
does not exert apparent o-target toxicity.
Figure 5. Pharmacokinetic, imaging, and biodistribution studies. (a) Blood circulation curve after i.v. injection of FAB NPs ( µg mL–). (b) Field-
dependent magnetization curves at  K for Ag/BMO and FAB NPs. (c) MR images of aqueous solutions containing dierent amounts of FAB NPs
and the corresponding relative T relaxation rates. (d) MR images of T tumor on mice, at dierent time points after injection of FAB NPs. (e) Quan-
tification of MR signals. (f) PA images of the tumor at dierent time points. (g) Prussian blue staining of the tumor tissues after injection of FAB NPs.
Scar bar: µm. (h) Biodistribution of FAB NPs at , , ,  and  h.
©  Wiley-VCH GmbH
2106996 (8 of 10)
www.advmat.dewww.advancedsciencenews.com
Adv. Mater. 2021, 33, 
3. Conclusion
In summary, to tackle the current problems of catalytic cancer
therapy, including compromised and nonsustainable e-
ciency, incapability to catalyze multielectron reactions, and
complexity due to involvement of multiple therapeutic compo-
nents, we herein developed a simple unprecedented “all-in-one”
nanozyme – FeO/Ag/BiMoO nanoparticle (FAB NP).
Through essential intraparticulate coupling, the NP exhibits a
narrowed bandgap for ecient NIR-II adsorption, enhanced
photocatalytic property, replenishable catalytic activity due
to existence of Mo+/Mo+ and Fe+/Fe+ redox sites, and fer-
romagnetic property. It enables eective chemodynamic,
photo dynamic, and photothermal therapy guided by magnetic
Figure 6. In vivo nanocatalytic therapy. (a) Photothermal images of T tumor-bearing mice injected with PBS (control) or FAB NPs (µg mL–,
µL), under nm laser irradiation ( W cm–) for dierent durations. (b) Corresponding temperature change. (c-e) Changes in tumor volume,
relative tumor weight and tumor size after dierent treatments. P-values were calculated by Student’s two-sided t-test. **P<., ***P<. (n = ).
(f) DHE staining of tumors with dierent treatments to indicate ROS level. Scar bar: µm. (g, h) H&E (chromatins stained by purple-blue) and Ki
(cell proliferation indicated by brown staining) staining of tumors with dierent treatments. Scar bar: µm.
©  Wiley-VCH GmbH
2106996 (9 of 10)
www.advmat.dewww.advancedsciencenews.com
Adv. Mater. 2021, 33, 
resonance, photoacoustic, and photothermal imaging, as
evidenced by both in vitro and in vivo experiments. The out-
standing performance is attributed to synergy between the
multi-mode therapy, guidance by multimode imaging, sustain-
ability of the coupled catalytic reactions, and the simplicity of
the system. This study provides new insights to design novel
nanozymes for nanocatalytic therapy with high eciency, good
specificity, and low side-eects, as well as for other theranostic
applications.
4. Experimental Section
Preparation of BMO, Ag/BMO, and FAB NPs: First, . g
NaMoO·HO and . g Bi(NO)·HO were separately dissolved in
mL glycol under sonication, followed by mixing and addition of mL
ethanol. After stirring for . h, the mixture was hydrothermally treated
in a mL Teflon-lined autoclave at  °C for  h. The centrifugated
precipitate was washed thrice with deionized (DI) water and ethanol,
then dried in oven at  °C for  h. The resulting powder was calcined
at  °C for  h ( °C min heating rate) to obtain BMO NPs. Ag/
BMO NPs were synthesized by the photoreduction method. Specifically,
g of BMO NPs was dissolved in  mL ethanol under sonication, and
subsequently, mL AgNO solution (mg mL) was added. The mixture
was then irradiated with a Xe lamp ( W,  h) for photoreduction of Ag
NPs onto BMO NPs. The centrifugated precipitate was washed thrice
with ethanol to remove the unreacted AgNO. Next, the sample was
dried at  °C for  h to obtain Ag/BMO NPs. To produce FAB NPs,
.g Ag/BMO NPs,  g FeCl·HO, g PVP, and  g natrium acetate
were dissolved into mL propanediol with ultrasonication for  min,
followed by titrating pH to . using NaOH ( ). After stirring for  h
at  °C, the above solution was hydrothermally treated in a mL of
Teflon-lined autoclave at  °C for  h. The centrifugated precipitate
was dried at  °C for  h and washed thrice with DI water and ethanol
to obtain FAB NPs.
Cell Culture, Cytotoxicity Assay, and Apoptosis Assay: Human
keratinocytes cells (HaCaTs) and mouse breast carcinoma cells (T
cells) were cultured with dulbecco’s modified eagle medium (DMEM)
medium supplemented with % fetal bovine serum (FBS), streptomycin
( µg mL), and penicillin ( units mL) in % CO atmosphere at
 °C. Cytotoxicity of FAB NPs was evaluated by MTT (-(,-Dimethylthiazol-
-yl)-,-diphenyltetrazolium bromide) assay. Specifically, cells ( µL,
×  cells mL) were seeded into -well plates and incubated
with NPs with dierent concentrations (, ., ., , , , or
 µg mL) for  h. Then, µL MTT solution (mg mL) was added
for another  h incubation. After rinsing with phosphate buer solution
(PBS) to remove NPs,  µL dimethyl sulfoxide (DMSO) was used to
dissolve the formazan crystals sediment in each well. The absorbance at
nm was determined using a microplate spectrophotometer (Epoch ,
Biotek, USA). To test apoptosis induced by FAB NPs, T cells (. × )
were incubated with NPs (µg mL) for  h, then rinsed with cold PBS
and immediately dyed with Annexin V-FITC/PI kit (KeyGEN, BioTECH),
followed by detection using flow cytometry (AccuriC, Biosciences).
Animal Experiments: Female Balb/c mice (– weeks old) were
ordered from the Comparative Medicine Centre of Yangzhou University.
The procedure was approved by Nanjing Tech University. PBS containing
T cells ( µL,  ×  cells) was subcutaneously injected into the
back of Balb/c mice to induce tumor. The tumor-bearing mice were
randomly divided into five groups: ) control (i.v. injection with µL
PBS); ) NIR irradiation only (nm, . W cm, min, once a day);
) treatment with FAB NPs (i.v. injection with µL NP solution with
a concentration of  µg mL); ) treatment of FAB NPs and NIR
irradiation at  h after injection once a day; ) treatment of FAB NPs
with NIR irradiation twice a day. Magnetic resonance imaging (MRI)
was conducted with a Bruker BioSpec / USR MR scanner (Bruker,
Germany). Photoacoustic imaging was performed through Vevo LAZR-X
Multimodal Imaging (FUJIFILM VisualSonics). Tumor tissues and blood
samples were digested with HNO and hydrochloric acid for ICP-OES
analysis.
Supporting Information
Supporting Information is available from the Wiley Online Library or
from the author.
Acknowledgements
C,C., H.Z., and N.Y. contributed equally to this work. The work was
supported by NNSF of China (Nos. , , and
), Jiangsu Province Policy Guidance Plan (No. BZ),
Natural Science Foundation of Shandong Province (No. ZRKB),
“Taishan scholars” construction special fund of Shandong Province, and
AcRF Tier- Grant (No. MOE-T--) from Ministry of Education
of Singapore.
Conflict of Interest
The authors declare no conflict of interest.
Data Availability Statement
Research data are not shared.
Keywords
chemodynamic therapy, nanocatalytic therapy, nanozymes,
photodynamic therapy, photothermal therapy
Received: September , 
Revised: October , 
Published online: October , 
[] J.Chen, Y.Zhu, C.Wu, J.Shi, Chem. Soc. Rev. 2020, 49, .
[] L.-H.Fu, C.Qi, J.Lin, P.Huang, Chem. Soc. Rev. 2018, 47, .
[] H.Lin, Y.Chen, J.Shi, Chem. Soc. Rev. 2018, 47, .
[] X. Hu, F. Li, F. Xia, X. Guo, N. Wang, L.Liang, B. Yang, K. Fan,
X.Yan, D.Ling, J. Am. Chem. Soc. 2020, 142, .
[] L. Gao, J. Zhuang, L. Nie, J. Zhang, Y. Zhang, N. Gu,
T. Wang, J. Feng, D. Yang, S. Perrett, X. Yan, Nat. Nanotechnol.
2007, 2, .
[] H.Li, Y.Li, J.Jiao, H.-M.Hu, Nat. Nanotechnol. 2011, 6, .
[] B. Jiang, D. Duan, L. Gao, M. Zhou, K. Fan, Y. Tang, J.Xi, Y. Bi,
Z.Tong, G. F.Gao, N. Xie, A. Tang, G.Nie, M. Liang, X.Yan, Nat.
Protoc. 2018, 13, .
[] Y.Zhang, X.Han, G.Nie, Nat. Protoc. 2020, 16, .
[] S.Ji, B. Jiang, H.Hao, Y.Chen, J.Dong, Y.Mao, Z.Zhang, R.Gao,
W. Chen, R. Zhang, Q. Liang, H. Li, S. Liu, Y. Wang, Q. Zhang,
L.Gu, D. Duan, M. Liang, D. Wang, X.Yan, Y.Li, Nat. Catal. 2021,
4, .
[] L.Shi, Y.Wang, C.Zhang, Y.Zhao, C. Lu, B. Yin, Y.Yang, X.Gong,
L.Teng, Y. Liu, X.Zhang, G.Song, Angew. Chem., Int. Ed. 2021, 60,
.
[] B.Yang, Y.Chen, J.Shi, Chem. Rev. 2019, 119, .
©  Wiley-VCH GmbH
2106996 (10 of 10)
www.advmat.dewww.advancedsciencenews.com
Adv. Mater. 2021, 33, 
[] S. Li, L. Shang, B. Xu, S.Wang, K.Gu, Q. Wu, Y. Sun, Q. Zhang,
H.Yang, F.Zhang, L.Gu, T.Zhang, H. Liu, Angew. Chem., Int. Ed.
2019, 58, .
[] G.Liu, J. Zhu, H. Guo, A. Sun, P.Chen, L. Xi, W.Huang, X. Song,
X.Dong, Angew. Chem., Int. Ed. 2019, 58, .
[] S.Sun, Q.Chen, Z.Tang, C.Liu, Z.Li, A.Wu, H.Lin, Angew. Chem.,
Int. Ed. 2020, 59, .
[] Y.Xie, K. A.Krug, K. S.Cay, M.Kalaj, N. C.McCallum, Z. E.Siwicka,
Z. Wang, N. C. Gianneschi, M. D. Burkart, J. D. Rinehart, CCS
Chem. 2021, 3, .
[] P.-C. Lo, M. S. Rodríguez-Morgade, R. K. Pandey, D. K. P. Ng,
T.Torres, F.Dumoulin, Chem. Soc. Rev. 2020, 49, .
[] X.Li, J. F.Lovell, J.Yoon, X.Chen, Nat. Rev. Clin. Oncol. 2020, 17, .
[] H.Yang, B.Xu, S.Li, Q.Wu, M.Lu, A.Han, H.Liu, Small 2021, 17,
.
[] M. Chang, M. Wang, M.Wang, M. Shu, B. Ding, C. Li, M. Pang,
S.Cui, Z.Hou, J.Lin, Adv. Mater. 2019, 31, .
[] Y. Cheng, X. Kong, Y. Chang, Y. Feng, R. Zheng, X. Wu, K. Xu,
X.Gao, H.Zhang, Adv. Mater. 2020, 32, .
[] L.Fu, Y. Wan, C.Qi, J. He, C. Li, C. Yang, H.Xu, J.Lin, P.Huang,
Adv. Mater. 2021, 33, .
[] C.Wang, F. Cao, Y.Ruan, X. Jia, W.Zhen, X. Jiang, Angew. Chem.,
Int. Ed. 2019, 58, .
[] X.Cai, L.Jiao, H. Yan, Y. Wu, W.Gu, D.Du, Y.Lin, C. Zhu, Mater.
Today 2021, 44, .
[] Z.Wang, R.Zhang, X.Yan, K.Fan, Mater. Today 2020, 41, .
[] C. Fang, Z.Deng, G. Cao, Q.Chu, Y.Wu, X.Li, X. Peng, G. Han,
Adv. Funct. Mater. 2020, 30, .
[] F. Gong, M. Chen, N. Yang, Z. Dong, L. Tian, Y. Hao,
M. Zhuo, Z. Liu, Q. Chen, L. Cheng, Adv. Funct. Mater. 2020, 30,
.
[] S.Wu, P.Wang, J.Qin, Y.Pei, Y.Wang, Adv. Funct. Mater. 2021, 31,
.
[] X.Zhong, X.Wang, L.Cheng, Y.Tang, G.Zhan, F.Gong, R.Zhang,
J.Hu, Z.Liu, X.Yang, Adv. Funct. Mater. 2020, 30, .
[] H.Li, T.Zhang, C.Pan, C.Pu, Y.Hu, X.Hu, E.Liu, J.Fan, Appl. Surf.
Sci. 2017, 391, .
[] Y.Xing, X.Gao, G.Ji, Z.Liu, C.Du, Appl. Surf. Sci. 2019, 465, .
[] M.Wang, Y. Liang, Z. Zhang, G. Ren, Y.Liu, S. Wu, J. Shen, Anal.
Chim. Acta 2019, 1086, .
[] J. C. Pieretti, W. R. Rolim, F. F. Ferreira, C. B. Lombello, M. H.
M.Nascimento, A. B.Seabra, J. Cluster Sci. 2020, 31, .
... Reproduced with permission. [187] Copyright 2021, Wiley-VCH. comprehensively explained by the H 2 O 2 decomposition regulation and electron transfer promotion due to the UiO-66 supporting. ...
... [185] In addition, combining nanomaterials with photothermal activity is also effective for enhancing the POD-like activity of nanozymes at weakly acidic or neutral pH, the generated heat could significantly promote the overall catalytic process. [186][187][188] OXD-Like and CAT-Like Nanozymes: In case of photoactivated OXD-like nanozymes, cofactors were widely applied to break through their pH-restricted catalytic behavior. As shown in Figure 8a(i), Ce 3+ was introduced to form a hybrid catalytic system with CDs. ...
... Thus, adding photothermal component was again beneficial to improve the CAT-like activity of composite nanozymes under weakly acid pH. [187,201,202] As concluded in Table 3, composite methods that combine different active components together to enhance the catalytic activity of the original nanozymes via the promotion of kinetic catalytic process are demonstrated applicable to break through the pH limitation of nanozymes. ...
Article
Full-text available
Although nanozymes have drawn great attention over the past decade, the activities of peroxidase‐like, oxidase‐like, and catalase‐like nanozymes are often pH dependent with elusive mechanism, which largely restricts their application. Therefore, a systematical discussion on the pH‐related catalytic mechanisms of nanozymes together with the methods to overcome this limitation is in need. In this review, various nanozymes exhibiting pH‐dependent catalytic activities are collected and the root causes for their pH dependence are comprehensively analyzed. Subsequently, regulatory concepts including catalytic environment reconstruction and direct catalytic activity improvement to break this pH restriction are summarized. Moreover, applications of pH‐independent nanozymes in sensing, disease therapy, and pollutant degradation are overviewed. Finally, current challenges and future opportunities on the development of pH‐independent nanozymes are suggested. It is anticipated that this review will promote the further design of pH‐independent nanozymes and broaden their application range with higher efficiency.
... To further quantitatively evaluate the catalytic activity of the CAT samples, the consumption of H 2 O 2 was recorded at determined time points. [36] As shown in Figure 3i, over 50% of H 2 O 2 was rapidly catalyzed and decomposed by CeVO 4 , CeVO 4 -Ti, and CeVO 4 -Ti@PPy, especially the decomposition of 50% H 2 O 2 induced by CeVO 4 -Ti@PPy needed only 15 min. Therefore, CeVO 4 -Ti@PPy has the potential to effectively alleviate tumor hypoxia. ...
Article
Full-text available
Compromises between enhanced on‐targeting reactivity and precise real‐time monitoring in the tumor microenvironment (TME) are the main roadblocks for catalytic cancer therapy. The hallmark of a high level of hydrogen peroxide (H2O2) and acidic extracellular environment of the hypoxia solid tumor can underpin therapeutic and tracking performance. Herein, this work provides an activatable wintersweet‐like nanohybrid consisting of titanium (Ti) doped cerium vanadate nanorods with the modification of polypyrrole (PPy) nanoparticles (CeVO4‐Ti@PPy) for combinatorial therapies of breast carcinoma. The Ti dopants in the size‐controllable CeVO4 nanorods lower the energy barrier (0.5 eV) of the rate‐determining steps and elaborate peroxidase‐like (POD‐like) activities to improve the generation of toxic hydroxyl radical (·OH) according to the density functional theory (DFT) calculation. The multiple enzyme‐like activities, including the intrinsic glutathione peroxidase (GPx) and catalase (CAT), achieve a record‐high therapeutic efficiency. Coupling this oxidative stress with the photothermal effects of PPy enables enhanced catalytic tumor necrosis. The exterior PPy heterogeneous structure can be further doped with protons in the local acidic environment to intensify photoacoustic signals, allowing the non‐invasive accurate tracking of tumors. The theranostic performance displayed negligible attenuated signals in near‐infrared (NIR) windows. This organic‐inorganic nanohybrid with a heterogeneous structure provides the potential to improve the overall outcomes of catalytic therapy.
Article
Nanozyme‐mediated chemodynamic therapy has emerged as a promising strategy due to its tumor specificity and controlled catalytic activity. However, the poor efficacy caused by low hydrogen peroxide (H 2 O 2 ) levels in the tumor microenvironment (TME) poses challenges. Herein, an H 2 O 2 self‐supplying nanozyme is constructed through loading peroxide‐like active platinum nanoparticles (Pt NPs) on zinc peroxide (ZnO 2 ) (denoted as ZnO 2 @Pt). ZnO 2 releases H 2 O 2 in response to the acidic TME. Pt NPs catalyze the hydroxyl radical generation from H 2 O 2 while reducing the mitigation of oxidative stress by glutathione, serving as a reactive oxygen (ROS) amplifier through self‐cascade catalysis. In addition, Zn ²⁺ released from ZnO 2 interferes with tumor cell energy supply and metabolism, enabling ion interference therapy to synergize with chemodynamic therapy. In vitro studies demonstrate that ZnO 2 @Pt induces cellular oxidative stress injury through enhanced ROS generation and Zn ²⁺ release, downregulating ATP and NAD ⁺ levels. In vivo assessment of anticancer effects showed that ZnO 2 @Pt could generate ROS at tumor sites to induce apoptosis and downregulate energy supply pathways associated with glycolysis, resulting in an 89.7% reduction in tumor cell growth. This study presents a TME‐responsive nanozyme capable of H 2 O 2 self‐supply and ion interference therapy, providing a paradigm for tumor‐specific nanozyme design.
Article
Full-text available
Due to the limitations of single-model tumor therapeutic strategies, multimodal combination therapy have become a more favorable option to enhance efficacy by compensating for its deficiencies. However, in nanomaterial-based multimodal therapeutics for tumors, exploiting synergistic interactions and cascade relationships of materials to achieve more effective treatments is still a great challenge. Based on this, we constructed a nanoplatform with a “triple-linkage” effect by cleverly integrating polydopamine (PDA), silver nanoparticles (AgNPs), and glucose oxidase (GOx) to realize enhanced photothermal therapy (PTT) and activatable metal ion therapy (MIT) for hepatocellular carcinoma (HCC) treatment. First, the non-radiative conversion of PDA under light conditions was enhanced by AgNPs, which directly enhanced the photothermal conversion efficiency of PDA. In addition, GOx reduced the synthesis of cellular heat shock proteins by interfering with cellular energy metabolism, thereby enhancing cellular sensitivity to PTT. On the other hand, H2O2, a by-product of GOx-catalyzed glucose, could be used as an activation source to activate non-toxic AgNPs to release cytotoxic Ag⁺, achieving activatable Ag⁺-mediated MIT. In conclusion, this nanosystem achieved efficient PTT and MIT for HCC by exploiting the cascade effect among PDA, AgNPs, and GOx, providing a novel idea for the design of multimodal tumor therapeutic systems with cascade regulation. Graphical abstract
Article
Full-text available
Asymmetric transformations and synthesis have garnered considerable interest in recent decades due to the extensive need for chiral organic compounds in biomedical, agrochemical, chemical, and food industries. The field of chiral inorganic catalysts, garnering considerable interest for its contributions to asymmetric organic transformations, has witnessed remarkable advancements and emerged as a highly innovative research area. Here, we review the latest developments in this dynamic and emerging field to comprehensively understand the advances in chiral inorganic nanocatalysts and stimulate further progress in asymmetric catalysis.
Article
Full-text available
Regeneration of bone defects in diabetic patients has always been a significant challenge in clinical treatment. The pathologic diabetic micromilieu, characterized by mitochondrial dysfunction, excessive reactive oxygen species (ROS) accumulation, cellular senescence, and chronic inflammation, compromises innate bone healing capacity. 3D cryo‐printing technology is utilized in bone tissue engineering to fabricate hierarchical porous scaffolds that promote a conducive microenvironment for cellular adhesion, migration, proliferation, and nutrient exchange. Nanozymes are used as synthetic mimics of natural enzymes to scavenge ROS, addressing the limitations of natural antioxidative enzymes. To remodel the diabetic bone regeneration micromilieu, a 3D cryo‐printed polyaryletherketone with carboxyl groups (PAEK‐COOH) and 45S5 bioactive glass (BG) hierarchical porous scaffold (PBG scaffold), harmonized with hybrid nanozymes comprising SS31‐enhanced manganese dioxide (MnO2)‐ferritin biomimetic nanozyme (MF@S nanozyme), is developed for combinatorial mitochondrial therapy. The MF@S nanozyme specifically targets mitochondria to enhance mitochondrial function, scavenge ROS accumulated in mitochondria, and suppress mitochondrial ROS (mtROS) production, and thus rejuvenate aging cells, regulate macrophage polarization, and modulate differentiation of osteoblasts and osteoclasts. This 3D cryo‐printed PBG‐MF@S hierarchical porous scaffold combines with a combinatorial mitochondrial therapy system to remodel the diabetic micromilieu and presents a promising therapeutic approach for the regeneration of bone defects in diabetes.
Article
Full-text available
Although inspiring progress has been achieved in tumor nanocatalytic therapies based on tailor‐made nanozymes for converting hydrogen peroxide into reactive oxygen species (ROS) efficiently, most cytotoxic hydroxyl radicals do not spread far enough within a cell to damage the primary organelles for effective tumor therapy due to their short half‐life time (≈1 µs). Developing a novel nanocatalyst platform involving longer half‐life time ROS is desired. To this end, Fe3O4‐Schwertmannite nanocomposites (Fe3O4‐Sch) with triple‐effect tumor therapy are constructed through a facile method. The Schwertmannite shell converts the •OH produced by Fe3O4 via the Fenton reaction into sulfate radicals with a longer half‐life time (30 µs). Combination of dual radicals exhibits overwhelming tumor inhibition efficacy. The nanocomposites also show the multifunctionality of good photothermal efficiency (33.2%) and synergistic oxidative stress amplification upon glutathione biosynthesis (GSH) depletion by the l‐buthionine sulfoximine (BSO) molecules loaded in the hollow Fe3O4 cores. The comprehensive properties of the nanoplatform including the dual‐radical production, Fe3O4 nanocrystal mediated PTT, and the BSO mediated GSH depletion result in remarkable tumor inhibition both in vitro and in vivo, which may pave a way to constructing a synergic catalytic nanoplatform for efficient tumor therapy. A multifunctional catalytic nanoplatform with cascade dual‐radical enzyme activities based on Schwertmannite coated Fe3O4 hollow nanoparticles produces both hydroxyl radicals and sulfate radicals, conducts photothermal therapy, and induces synergistic oxidative stress amplification by the loaded l‐buthionine sulfoximine molecules mediated glutathione biosynthesis depletion, enabling an effectively synergetic breast cancer therapy.
Article
Full-text available
Developing artificial enzymes with the excellent catalytic performance of natural enzymes has been a long-standing goal for chemists. Single-atom catalysts with well-defined atomic structure and electronic coordination environments can effectively mimic natural enzymes. Here, we report an engineered FeN3P-centred single-atom nanozyme (FeN3P-SAzyme) that exhibits comparable peroxidase-like catalytic activity and kinetics to natural enzymes, by controlling the electronic structure of the single-atom iron active centre through the precise coordination of phosphorus and nitrogen. In particular, the engineered FeN3P-SAzyme, with well-defined geometric and electronic structures, displays catalytic performance that is consistent with Michaelis–Menten kinetics. We rationalize the origin of the high enzyme-like activity using density functional theory calculations. Finally, we demonstrate that the developed FeN3P-SAzyme with superior peroxidase-like activity can be used as an effective therapeutic strategy for inhibiting tumour cell growth in vitro and in vivo. Therefore, SAzymes show promising potential for developing artificial enzymes that have the catalytic kinetics of natural enzymes.
Article
Full-text available
Chemodynamic therapy is an emerging tumor therapeutic strategy. However, the anticancer effects are greatly limited by the strong acidity requirements for effective Fenton‐like reaction, and the inevitably “off‐target” toxicity. Herein, we develop an acidity‐unlocked nanoplatform (FePt@FeOx@TAM‐PEG) that can accurately perform the high‐efficient and tumor‐specific catalysis for anticancer treatment, through dual pathway of cyclic amplification strategy. Notably, the pH‐responsive peculiarity of tamoxifen (TAM) drug allows for the catalytic activity of FePt@FeOx to be “turn‐on” in acidic tumor microenvironments, while keeping silence in neutral condition. Importantly, the released TAM within cancer cells is able to inhibit mitochondrial complex I, leading to the upregulated lactate content and thereby the accumulated intracellular H⁺, which can overcome the intrinsically insufficient acidity of tumor. Through the positive feedback loop, large amount of active FePt@FeOx nanocatalyzers are released and able to access to the endogenous H2O2, exerting the improved Fenton‐like reaction within the more acidic condition. Finally, such smart nanoplatform enables self‐boosting generation of reactive oxygen species (ROS) and induces strong intracellular oxidative stress, leading to the substantial anticancer outcomes in vivo, which may provide a new insight for tumor‐specific cascade catalytic therapy and reducing the “off‐target” toxicity to surrounding normal tissues.
Article
Full-text available
Dual phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), has shown a great prospect in cancer treatment. However, its therapeutic effect is restricted by the depth of light penetration in tissue and tumor hypoxia environment. Herein, inspired by the specific response of nanozymes to the tumor microenvironment (TME), a simple and versatile nanozyme‐mediated synergistic dual phototherapy nanoplatform (denoted as FePc/HNCSs) is constructed using hollow nitrogen‐doped carbon nanospheres (HNCSs) and iron phthalocyanine (FePc). FePc/HNCSs simultaneously exhibit peroxidase (POD)‐ and catalase (CAT)‐like activities, which not only can convert endogenous hydrogen peroxide (H2O2) into highly toxic hydroxyl radicals (•OH) for catalytic therapy, but also decompose H2O2 to oxygen (O2) to enhance O2‐dependent PDT. In addition, their enzyme‐like activities are significantly enhanced under light irradiation. Combining with the excellent photothermal effect, FePc/HNCSs realize a high tumor inhibition rate of 96.3%. This strategy opens a new horizon for exploring a more powerful tumor treatment nanoplatform.
Article
Full-text available
Chemodynamic therapy (CDT) is an emerging therapy method that kills cancer cells by converting intracellular hydrogen peroxide (H2O2) into highly toxic hydroxyl radicals (•OH). To overcome the current limitations of the insufficient endogenous H2O2 and the high concentration of glutathione (GSH) in tumor cells, an intelligent nanocatalytic theranostics (denoted as PGC‐DOX) that possesses both H2O2 self‐supply and GSH‐elimination properties for efficient cancer therapy is presented. This nanoplatform is constructed by a facile one‐step biomineralization method using poly(ethylene glycol)‐modified glucose oxidase (GOx) as a template to form biodegradable copper‐doped calcium phosphate nanoparticles, followed by the loading of doxorubicin (DOX). As an enzyme catalyst, GOx can effectively catalyze intracellular glucose to generate H2O2, which not only starves the tumor cells, but also supplies H2O2 for subsequent Fenton‐like reaction. Meanwhile, the redox reaction between the released Cu²⁺ ions and intracellular GSH will induce GSH depletion and reduce Cu²⁺ to Fenton agent Cu⁺ ions, and then trigger the H2O2 to generate •OH by a Cu⁺‐mediated Fenton‐like reaction, resulting in enhanced CDT efficacy. The integration of GOx‐mediated starvation therapy, H2O2 self‐supply and GSH‐elimination enhanced CDT, and DOX‐induced chemotherapy, endow the PGC‐DOX with effective tumor growth inhibition with minimal side effects in vivo.
Article
Full-text available
Modulating the hostile tumor microenvironment (TME) rather than directly killing cancer cells may be an effective strategy to improve the therapeutic benefits in cancer treatment. Herein, FeWOX nanosheets are constructed as cascade bioreactors to modulate the TME and enhance radiotherapy and immunotherapy of tumors. Synthesized by the thermal‐decomposition method and modified by poly(ethylene glycol) (PEG), the obtained FeWOX‐PEG with multivalent metal elements (Fe2+/3+, W5+/6+) exhibit efficient catalytic decomposition of hydrogen peroxide (H2O2) to generate hydroxyl radicals (•OH) for chemo‐dynamic therapy (CDT). The generated high valence of metal ions (Fe3+/W6+) in FeWOX‐PEG are reduced by endogenous glutathione (GSH), both leading to depletion of GSH and further amplified oxidative stress, and resulting in the reduced metal valence statuses (Fe2+/W5+) enabling cascade bioreactions. Such FeWOX‐PEG bioreactors enhance the oxidative stress in the tumor and interact with X‐rays, significantly improving cancer radiotherapy (RT). Furthermore, the reactive oxygen species (ROS)‐induced inflammation caused by FeWOX‐PEG in TME activates the immune system and promotes the tumor‐infiltration of various types of immune cells, which working together with cytotoxic T‐lymphocyte antigen‐4 (CTLA‐4) checkpoint blockade could elicits a robust immune response to defeat tumors. This work presents an intelligent cascade bioreactor based on multivalent bimetallic oxide FeWOX nanosheets, which exhibit efficient catalytic decomposition of hydrogen peroxide (H2O2) to generate hydroxyl radicals (•OH), as well as glutathione (GSH) depletion capability to further amplify oxidative stress. This good tumor microenvironment (TME)‐modulation performance can further enhance both radiotherapy and immunotherapy of tumors.
Article
Enzyme cascade reactions in biological systems can effectively improve the catalytic performances owing to their high local concentration, efficient mass transfer and reduced intermediate decomposition. However, the disadvantages of high cost, low stability, and easy inactivation limit their practical applications in harsh environments. Nanozymes as the substitutes for enzymes not only have enzyme-like activities but also possess high stability, tunable catalytic property, and high pH/temperature tolerance, which have been intensively investigated for the establishment of biomimetic catalysis systems. In this review, we first summarized the recent development of the construction and advantage of the nanozyme-involved cascade reaction system. Then, the biomedical applications of the nanozyme-enhanced biomimetic cascade catalytic systems, including biosensing, therapies and antioxidation, were highlighted in detail. Finally, current challenges and future perspectives for nanozyme-activated cascade reactions are discussed and outlooked.
Article
Here we describe two protocols for the construction of responsive and activable nanomedicines that regulate the tumor microenvironment (TME). The TME is composed of all non-cellular and cellular components surrounding a tumor, including the surrounding blood vessels, immune cells, fibroblasts, signaling molecules, and extracellular matrix and has a crucial role in tumor initiation, growth, and metastasis. Owing to the relatively stable properties of the TME compared to tumor cells, which exhibit frequent genetic mutations and epigenetic changes, therapeutic strategies targeting the TME using multifunctional nanomedicines hold great potential for anti-tumor therapy. By regulating tumor-associated platelets and pancreatic stellate cells (PSCs), the two major players in the TME, we can effectively manipulate the physiological barriers for enhanced drug delivery and significantly improve the tumor penetration and therapeutic efficacy of chemotherapeutics. The preparation and characterization of the multifunctional nanoparticles takes ~10 h for tumor-associated platelet regulation and 16 h for PSC regulation. These nanoformulations can be readily applied to regulate other components in the TME to realize synergistic or additive anti-tumor activity.
Article
Various therapeutic techniques have been studied for treating cancer precisely and effectively, such as targeted drug delivery, phototherapy, tumor-specific catalytic therapy, and synergistic therapy, which, however, evoke numerous challenges due to the inherent limitations of these therapeutic modalities and intricate biological circumstances as well. With the remarkable advances of nanotechnology, nanoplatform-based cascade engineering, as an efficient and booming strategy, has been tactfully introduced to optimize these cancer therapies. Based on the designed nanoplatforms, pre-supposed cascade processes could be triggered under specific conditions to generate/deliver more therapeutic species or produce stronger tumoricidal effects inside tumors, aiming to achieve cancer therapy with increased anti-tumor efficacy and diminished side effects. In this review, the recent advances in nanoplatform-based cascade engineering for cancer therapy are summarized and discussed, with an emphasis on the design of smart nanoplatforms with unique structures, compositions and properties, and the implementation of specific cascade processes by means of endogenous tumor microenvironment (TME) resources and/or exogenous energy inputs. This fascinating strategy presents unprecedented potential in the enhancement of cancer therapies, and offers better controllability, specificity and effectiveness of therapeutic functions compared to the corresponding single components/functions. In the end, challenges and prospects of such a burgeoning strategy in the field of cancer therapy will be discussed, hopefully to facilitate its further development to meet the personalized treatment demands.
Article
Nanozymes, nanomaterials with enzyme-like activities, are becoming powerful competitors and potential substitutes for natural enzymes because of their excellent performance, including design from scratch, controllable activity, and environmental resistance. In recent years, various nanozymes have been discovered or designed, and gradually applied to molecular detection, biomedical treatment and environmental management. Nevertheless, nanozymes are often regarded as fascinating and confusing black boxes as their catalytic mechanisms remain largely indistinct. Interestingly, recent researches have shed light into these black boxes. It appears that the enzymatic activities of nanozymes are closely related to their size, surface lattice, surface modification and composition, etc. Some regular structure–activity relationships have been elucidated in recent reports. In this review, we systematically summarized the studies on the structure–activity relationship of nanozymes in recent years, aiming to illustrate the catalytic mechanism of nanozymes and clarify the key factors regulating their behavior, so as to provide ideas and inspiration for the de novo design of nanozymes.