Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

RESEARCH ARTICLE

www.advhealthmat.de

Remote Manipulation of TRPV1 Signaling by Near-Infrared


Light-Triggered Nitric Oxide Nanogenerators for Specific
Cancer Therapy
Shuangling Wang, Yalin Wang, Jie Lv, Chunzhe Xu, Yuxin Wei, Guiying Wang,*
and Meng Li*

cancer is still a critical public health prob-


Specific activation of transient receptor potential vanilloid member 1 (TRPV1) lem worldwide.[1] The off-target effects and
channels provides a new avenue for cancer treatment by inducing excessive low pharmacologic selectivity of the thera-
Ca2+ influx. However, controllable manipulation of TRPV1 signaling for clinical peutic agents result in unsatisfactory clin-
application has remained elusive due to the challenge in finding a mild and ical outcomes.[2,3] These issues highlight
the vital importance of developing thera-
effective method of exerting external stimulus without adverse side effects in
peutic models with high antitumor activ-
living systems. Herein, a TRPV1-targeting near-infrared (NIR) triggered nitric ity via spatiotemporal control of cell behav-
oxide (NO)-releasing nanoplatform (HCuS@PDA-TRPV1/BNN6) based on ior to achieve desired therapeutic response
polydopamine (PDA) coated hollow copper sulfide nanoparticles (HCuS NPs) only to tumor cells.
is developed for specific cancer therapy. Upon NIR irradiation, the NO donor Protein ion channels have been re-
ported to be key regulators of cellular
BNN6 encapsulated in NIR-responsive nanovehicles can locally generate NO
functions.[4] Specifically, transient recep-
to activate TRPV1 channels and induce Ca2+ influx. This NIR controlled mode tor potential vanilloid member 1 (TRPV1)
enables the nanoplatform to exert its therapeutic effects below the apoptotic which is highly expressed in several types of
threshold temperature (43°C), minimizing the photothermal damage to malignant tumors,[4,5] especially in glioblas-
normal tissue. Integrating this special NO-mediated therapy with HCuS NPs toma, has emerged as a potential target for
mediated chemodynamic therapy, the designed nanoplatform exhibits a cancer therapy. As a non-selective cation
channel, TRPV1 can be activated by nox-
boosted anticancer activity with negligible systematic toxicity. Together, this
ious heat (a threshold of ≈43°C), vanil-
study provides a promising strategy for site-specific cancer therapy by loids, acidosis and various other stimuli.[4]
spatiotemporally controlled activation of surface ion channels, thus offering a Activation of TRPV1 can induce an exces-
solution to an unmet clinical need in cancer treatment. sive Ca2+ influx, thereby affecting multi-
ple cancer-related biological processes, in-
cluding cell proliferation, migration and
1. Introduction apoptosis.[6–8] Therefore, accurate modulation of TRPV1 signal-
ing would be a promising approach for precision cancer therapy.
Despite chemotherapy as the preferred option for cancer ther- To this end, on the basis of the inherent thermosensitivity of
apy has made tremendous progress over the last few decades, TRPV1, many efforts have been devoted to designing nanoma-
terials with photothermal behavior to regulate TRPV1 signaling
S. Wang, J. Lv, C. Xu, Y. Wei, M. Li in a remote-controlled manner.[9,10] Although promising, a suf-
College of Pharmacy ficient heat dose (>43°C) to achieve therapeutic temperatures is
Key Laboratory of Innovative Drug Development and Evaluation required for activation of TRPV1, which would inevitably result
Hebei Medical University
Shijiazhuang 050017, China
in severe damage to surrounding normal tissue. Manipulation of
E-mail: limeng@hebmu.edu.cn TRPV1 signaling without adverse side effects in living systems
S. Wang remains challenging.
Department of Environmental and Chemical Engineering Nitric oxide (NO) as a biologically active signaling molecule
Hebei College of Industry and Technology plays critical roles in multiple physiological processes.[11–13] Early
Shijiazhuang 050091, China studies have shown that NO can enable the activation of TRPV1
Y. Wang, G. Wang signaling and then induce an increase of intracellular Ca2+
The Second Department of General Surgery
The Fourth Hospital of Hebei Medical University concentration,[14,15] which prompts to utilize NO as a novel stim-
Shijiazhuang 050011, China ulator to controllably activate TRPV1 signaling within the phys-
E-mail: wangguiying@hebmu.edu.cn iological temperature range for cancer specific therapy. To real-
ize this function, designing NO-releasing materials to achieve
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/adhm.202303579 controlled and localized delivery of NO with very little systemic
toxicity is highly desirable. Moreover, the appropriate releasing
DOI: 10.1002/adhm.202303579

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (1 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

Figure 1. Schematic illustration of the NIR-responsive NO nanogenerator integrating CDT and photoactivation of TRPV1 channels for specific cancer
treatment.

doses of NO in a spatiotemporal manner is also greatly im- In the present study, by integrating NO-mediated therapy and
portant for clinical applications. On the basis of this concept, CDT, a TRPV1 targeted NIR-triggered intelligent nanoplatform
various internal or external stimuli have been explored to real- (HCuS@PDA-TRPV1/BNN6) based on polydopamine (PDA)
ize on-demand NO release from NO-releasing materials.[16–19] coated hollow copper sulfide nanoparticles (HCuS NPs) has
Among them, near-infrared (NIR) light irradiation with high been developed for specific cancer therapy (Figure 1). HCuS
tissue-penetration capability and minimal damage to nearby nor- NPs with the potential to convert endogenous H2 O2 into •OH
mal tissues has shown an exciting possibility to trigger controlled through Fenton-like reaction can exert cancer CDT. In con-
NO release.[19–21] Consequently, an opportunity has risen to de- trast to Fe- and Mn-based catalysts, Cu-containing materials still
sign TRPV1 targeted NIR-triggered NO-releasing materials as show relatively high catalytic activities in slightly acidic envi-
an efficient strategy for remote-controlled activation of TRPV1 ronments, leading to an enhanced CDT efficiency.[29–31] Subse-
signaling. quently, PDA possessing a loose structure and high hydrophilic-
Intracellular Ca2+ overloading induced by TRPV1 activation ity was coated on the surface of HCuS NPs to not only con-
can cause mitochondrial dysfunction to disrupt the intracellu- trol the exposure of HCuS NPs and their derived chemody-
lar redox homeostasis.[22–24] Interestingly, amplifying intracel- namic reaction but also serve as a nanocarrier to encapsulate
lular oxidative stress can in turn efficiently destroy the mito- the NIR-responsive NO donors (BNN6, N,N′-di-sec-butyl-N,N′-
chondrial Ca2+ buffering capacity to induce an enhanced Ca2+ - dinitroso-1,4-phenylenediamine). Finally, the monoclonal anti-
overloading, which would further aggravate cell apoptosis.[24,25] body TRPV1 peptide was covalently conjugated onto the PDA
Benefiting from these features, combination of reactive oxy- layer (HCuS@PDA-TRPV1/BNN6) to enable specific binding to
gen species (ROS)-mediated therapy with modulation of TRPV1 the plasma membrane TRPV1 channels of cancer cells. The PDA
channel function would produce synergistic effects on cancer layer can absorb the NIR light and transform the photons to ac-
treatment. As a noninvasive emerging ROS-based anticancer tive electrons, inducing NO release from BNN6 at temperatures
modality, chemodynamic therapy (CDT) which principally cat- below the apoptotic threshold (43°C), which can then specifically
alyzes the decomposition of H2 O2 to produce hydroxyl radical activate TRPV1 channels and simultaneously minimize the pho-
(•OH) via Fenton/Fenton-like reactions can result in escalated tothermal damage to normal tissue. In addition, upon being in-
oxidase stress inside cells.[26–28] Therefore, combination TRPV1 ternalized into cancer cells after long-term incubation, the ex-
targeted therapy with CDT can be a feasible solution to selectively posed HCuS NPs could deplete cellular glutathione (GSH) to en-
amplify intracellular oxidative stress. hance the efficiency of CuS-mediated CDT, which could further

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (2 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

Figure 2. Characterization of HCuS@PDA-TRPV1. A) The TEM image of HCuS NPs. B) The XRD patterns of HCuS NPs and HCuS@PDA. C) FTIR
spectra of HCuS NPs, HCuS@PDA, and HCuS@PDA-TRPV1. D) TEM image of HCuS@PDA.

accelerate the oxidative damage. As expected, the NIR-triggered the surface area of HCuS NPs, which was calculated to be 16.19
multifunctional nanoplatform exhibited an amplified anticancer m2 g−1 (Figure S1B, Supporting Information). Through self-
activity with negligible systematic toxicity both in vitro and in polymerization of dopamine in a slightly alkaline environment,
vivo. To the best of our knowledge, this is the first report of de- PDA layer was coated onto the surface of HCuS NPs.[32,34] The
veloping spatiotemporally controllable NO-releasing materials as broad absorbance band occurred at 3500– 3000 cm−1 in the FTIR
activators of TRPV1 for site-specific cancer synergetic therapy. spectrum of HCuS@PDA was attributed to the stretching vibra-
tion of N─H and O─H groups of PDA, which authenticated the
2. Results and Discussion successful fabrication of HCuS@PDA. The TEM image revealed
that at the 1:1 molar ration of HCuS NPs to PDA, PDA coating
2.1. Synthesis and Characterization of formed a smooth layer with thickness of 15 nm on the surface
HCuS@PDA-TRPV1/BNN6 of HCuS NPs (Figure 2D) and the mean size of HCuS@PDA
was ≈ 130 nm (Figure S1C, Supporting Information). Further-
HCuS NPs were first prepared by solvothermal method using more, the surface of the nanocomposites became more negatively
sacrificial Cu2 O nanospheres as hard templates.[32] The trans- charged after PDA coating, changing from −8.8 to −28.1 mV
mission electron microscopy (TEM) image indicated that the (Figure S1D, Supporting Information). Dynamic light scatter-
monodispersed HCuS NPs exhibited an average size of ≈100 nm ing (DLS) measurements suggested that the average sizes of
with hollow interiors (Figure 2A; Figure S1A, Supporting Infor- the synthesized HCuS NPs and HCuS@PDA were 199.2 ± 14.4
mation). The X-ray powder diffraction (XRD) pattern of HCuS and 285.54 ± 7.0 nm, respectively (Figure S1E, Supporting In-
NPs (Figure 2B) was well-indexed to the hexagonal phase of formation). Besides, the structure of HCuS NPs was not dis-
CuS (JCPDS No. 06–0464).[33] Fourier translation infrared (FTIR) turbed by modification with PDA, which was validated by the
spectroscopy was used to determine the chemical composition results of XRD analysis and UV–vis–NIR spectra (Figure 2B;
of HCuS NPs. As displayed in Figure 2C, the peaks at 1123 Figure S1F, Supporting Information). Subsequently, TRPV1 anti-
cm−1 and 618 cm−1 were the characteristic stretching vibra- body was covalently conjugated onto the surface of the nanocom-
tions of the C─O and Cu─S in HCuS NPs, respectively. The posites to realize the TRPV1 targeting capability. As indicated
N2 adsorption–desorption measurement was used to determine in the FTIR spectrum of HCuS@PDA-TRPV1 (Figure 2C), af-

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (3 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

ter conjugation of TRPV1 antibody, the characteristic absorption effect even after heating/cooling for five cycles, suggesting their
band of C─O in the carboxyl groups at ≈ 1100 cm−1 occurred. excellent photothermal stability (Figure 3D). To ensure complete
The average size obtained from DLS analysis for HCuS@PDA- release of NO and minimize the thermal damage to healthy
TRPV1 was 294.91 ± 8.4 nm, which was comparable to that tissues, a relatively low laser power density of 0.5 W cm−2 that
of HCuS@PDA (Figure S1E, Supporting Information). All only caused a mild temperature elevation upon irradiation for
these characteristics suggested the successful synthesis of the 5 min was chosen for the following study (Figure S6, Supporting
nanocomposites. Information).

2.2. The CDT Efficiency 2.4. NO Release in HCuS@PDA/BNN6

The efficiency of the nanocomposites induced CDT was in- Encapsulation of the NO donor, BNN6, into the PDA layer
vestigated using colorimetric assay. To verify the ability of the of the nanocomposites was achieved via an in situ polymer-
nanocomposites to generate ROS in acidic tumor microenviron- ization method for responsive release of NO. BNN6 was syn-
ment (TME), 3,3′,5,5′-tetramethylbenzidine (TMB) which can be thesized according to the previous report.[38] And its success-
oxidized by •OH to produce oxidant TMB (oxTMB) with a max- ful synthesis was confirmed by nuclear magnetic resonance
imum absorption at 652 nm was selected as the probe.[35,36] As (NMR) (Figure S7, Supporting Information). The PDA layer in
shown in Figure S2A (Supporting Information), after incubat- HCuS@PDA/BNN6 is capable of absorbing NIR light and con-
ing HCuS NPs with H2 O2 and TMB, the reaction system dis- verting photons into active electrons,[39,40] triggering the release
played a strong absorption peak at 652 nm, indicating their po- of NO from BNN6 (Figure S8, Supporting Information). In or-
tential as Fenton-like catalysts. Critically, compared with the con- der to further study the overall release of NO by NIR laser ir-
ventional solid CuS nanoparticles which were synthesized ac- radiation, the NO release behavior of HCuS@PDA/BNN6 was
cording to previously reported method (Figure S2B,C, Support- monitored under continuous laser irradiation by a commonly
ing Information),[37] the HCuS NPs possessed a higher catalytic used Griess assay.[41] As shown in Figure 3E, negligible NO
activity (Figure S2A,D, Supporting Information). And the bind- was released from the solutions of HCuS@PDA/BNN6 with-
ing affinity between HCuS NPs and H2 O2 was stronger (Figure out laser exposure, revealing the good stability of BNN6. In con-
S2E,F and Table S1, Supporting Information). All these phenom- trast, laser irradiation remarkably induced the NO release from
ena verified the superior catalytic performance of HCuS NPs with HCuS@PDA/BNN6. Moreover, the amount of NO molecules re-
hollow structures, which was of great significance for achieving leased by HCuS@PDA/BNN6 increased significantly with in-
great anticancer efficiency. More interestingly, after coating with creasing irradiation time. Critically, the release of NO can be
PDA, HCuS@PDA still exhibited a relatively high catalytic activ- well switched off/on by controlling the switching of an ex vivo
ity in acidic or even weakly acidic conditions (Figure 3A), thereby NIR laser to manipulate the active drug at the appropriate dose
enabling them to sever as promising CDT agents to transform in vivo within the required time to produce a continuous thera-
H2 O2 to •OH. peutic effect and minimize the risk of NO poisoning. To address
whether HCuS@PDA/BNN6 possessed a NIR controllability for
NO release, the NO released from the samples that heated di-
2.3. The Photothermal Effect rectly to 60°C without NIR irradiation was also monitored. As ex-
hibited in Figure 3F, there was no significant release of NO from
To demonstrate the photothermal performance of the nanocom- HCuS@PDA/BNN6 during 10 min of direct heating at 60°C,
posites, the photothermal effects of the HCuS NPs, PDA while NIR irradiation (0.5 W cm−2 , 10 min) did appreciably cause
nanoparticles (Figure S3, Supporting Information) and NO release. All these results demonstrated that the release of NO
HCuS@PDA nanocomposites were investigated. As indi- from HCuS@PDA/BNN6 can be efficiently controlled by NIR
cated in Figure S4 (Supporting Information), a significant laser, revealing a unique and intelligent release mode.
increase of temperature was observed in all these samples upon
NIR irradiation. And the photothermal conversion efficiency
(ƞ) was also calculated. According to the results (Figure S4, 2.5. Optimization of the Nanocomposites
Supporting Information), HCuS@PDA (ƞ = 23.86%) showed
an enhanced photothermal conversion capability compared to The thickness of PDA shell was critical for the therapeutic ef-
HCuS NPs (ƞ = 20.92%), which was probably attributed to the ficacy of the nanoplatform. To achieve satisfactory CDT effi-
high NIR absorption and excellent photothermal conversion ciency together with high sensitivity of NIR-triggered NO re-
efficiency of PDA (ƞ = 30.40%). Therefore, the excellent pho- lease, the PDA shell thickness was optimized. In this case,
tothermal performance of HCuS@PDA was originated from three types of HCuS@PDA were synthesized by varying the
both the PDA layer and HCuS NPs. Importantly, conjugation mass ratios of PDA/HCuS NPs (Figure 4A–C). As displayed in
of TRPV1 antibody onto the surface of the nanocomposites Figure 4D and Figure S9 (Supporting Information), the change
showed no influence on their photothermal effect (Figure S5, in the PDA shell thickness largely affected the efficiency of
Supporting Information). Moreover, the photothermal effect of HCuS@PDA-mediated Fenton-like reaction in acidic condition.
the HCuS@PDA-TRPV1 showed a radiant energy- (Figure 3B) Under the conditions with an equal amount of HCuS@PDA,
and materials concentration-dependent fashion (Figure 3C). the catalytic activity of the nanocomposites gradually decreased
Additionally, HCuS@PDA-TRPV1 achieved the same heating with decreasing the mass ratio of HCuS NPs to PDA from

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (4 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

Figure 3. A) UV–vis spectra of TMB to probe the HCuS@PDA-mediated Fenton-like reaction under different pH values. Temperature curves of
HCuS@PDA-TRPV1 at B) power densities and C) different concentrations. D) Photothermal stability curves of HCuS@PDA-TRPV1 for five on/off ir-
radiation cycles. E) The in vitro NO release profiles of HCuS@PDA/BNN6 under different conditions. Arrows indicate the initiation of 808 nm laser
irradiation. F) The NO release profiles of nanocomposites under NIR laser irradiation (0.5 W cm−2 ) or direct heating at different temperature.

2:1 to 1:2. The absorbance of produced oxTMB in 2:1 system the amount of NO released from HCuS@PDA/BNN6 with the
showed a 1.6-fold increase compared to that in 1:2 system. Be- same concentration was increased with the increased mass ratio
sides, HCuS@PDA/BNN6 with different mass ratios of HCuS of PDA in HCuS@PDA. Considering the efficiency of NO re-
NPs to PDA were also synthesized. The characteristic peak lease and CDT effect, HCuS@PDA with a mass ratio of 1:1 was
of BNN6 at 252 nm was observed in the UV–vis spectra of selected for the subsequent in vitro and in vivo studies.
HCuS@PDA/BNN6, confirming the successful encapsulation
of BNN6 (Figure S10A, Supporting Information). Furthermore,
with the increase of PDA content, the absorbance intensity of 2.6. The Activation of TRPV1 Signaling
the nanocomposites at 252 nm was increased. The BNN6 load-
ing capability of HCuS@PDA/BNN6 with different mass ratios The targeting ability of the nanocomposites toward TRPV1 chan-
of HCuS NPs to PDA was determined by the UV absorption of nels is of great significance for specific activation of TRPV1 sig-
BNN6 at 252 nm (Figure S10, Supporting Information), which naling by NO released from the nanosystem. As revealed in TEM
was calculated to be 96.2 μg BNN6/mg HCuS@PDA (HCuS images (Figure 5A), after the first 2 h of incubation, different
NPs:PDA = 2:1), 115.2 μg BNN6/mg HCuS@PDA (HCuS from HCuS@PDA, a higher density of HCuS@PDA-TRPV1 was
NPs:PDA = 1:1) and 123.1 μg BNN6/mg HCuS@PDA (HCuS firmly attached on the plasma membrane of U373 cells, the well
NPs:PDA = 1:2), respectively. While, for NO release (Figure 4E), characterized TRPV1 positive cells. To further analyze the tar-

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (5 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

Figure 4. TEM images of HCuS@PDA with different mass ratios of HCuS NPs to PDA: A) HCuS NPs:PDA = 1:2, B) HCuS NPs:PDA = 1:1, and C) HCuS
NPs:PDA = 2:1. The effects of the mass ratio of HCuS NPs to PDA on D) the catalytic activity of HCuS@PDA nanocomposites (pH 5.0) and E) their in
vitro NO release efficacy upon laser irradiation (0.5 W cm−2 ).

get ability of HCuS@PDA-TRPV1 to U373 cells, a quantitative HCuS@PDA-TRPV1/BNN6 pretreated U373 cells under laser
inductively coupled plasma-atomic emission spectrometer (ICP- irradiation. To further eliminate the photothermal effect trig-
AES) analysis was also employed to determine the Cu content in gered by NIR irradiation on TRPV1 signaling activation and min-
the cell lysates. As depicted in Figure S11A (Supporting Infor- imize hyperthermia-induced side effects, the NIR laser irradia-
mation), after incubating with the nanocomposites for 2 h, the tion (0.5 W cm−2 ) was accurately controlled to keep the cell tem-
amount of Cu in the cell lysates was increased with increasing perature < 43°C (Figure S6, Supporting Information). Under this
concentrations of the nanocomposites. It was worth noting that condition, without BNN6, HCuS@PDA-TRPV1 with laser irra-
the Cu content in HCuS@PDA-TRPV1 treated cells was signif- diation did not induce an altered Ca2+ response in U373 cells
icantly higher than that in the HCuS@PDA group. TRPV1 an- (Figure 5B). Moreover, the amounts of NO released into culture
tibody functionalization led to a 3.6-fold increase of Cu content media were also measured to verify that ability of HCuS@PDA-
in cell lysates when the cells were treated with the nanocompos- TRPV1/BNN6 to produce NO in the cells in the presence of
ites at the concentration of 10 μg mL−1 . Meanwhile, the lower laser irradiation (Figure S12, Supporting Information). All the
Cu amount in the cell culture supernatant of the HCuS@PDA- above results revealed that the NIR-triggered release of NO from
TRPV1 group (Figure S11B, Supporting Information) also sug- HCuS@PDA-TRPV1/BNN6 was sufficient to evoke TRPV1 acti-
gested that the TRPV1 antibody modified on the surface could vation.
endow the nanosystem with a specific and strong binding ability
with TRPV1 channels.
To ensure that the released NO was sufficient to induce the 2.7. In Vitro Therapeutic Effect
activation of TRPV1 signaling, the intracellular Ca2+ levels were
measured using a Ca2+ -sensitive fluorescent dye, Fluo-3 AM.[23] Encouraged by the excellent properties of HCuS@PDA-
As depicted in Figure 5B, compared with the untreated cells, NIR TRPV1/BNN6, the in vitro therapeutic effect of this nanocom-
irradiation only induced a remarkable increased fluorescence sig- posites was evaluated. As indicated in Figure 5E, after treated
nal in U373 cells pretreated with HCuS@PDA-TRPV1/BNN6. U373 cells for 24 h, HCuS@PDA-TRPV1 and HCuS@PDA-
By contrast, no apparent fluorescence change was detected in TRPV1/BNN6 showed comparable cytotoxicity in which the
HeLa cells, the well-known TRPV1-negative cells[4,42] (Figure 5C). cell viabilities reduced gradually with the increasing concen-
Moreover, when TRPV1 channels in U373 cells were blocked by tration of the nanocomposites. The anticancer activity can be
capsazepine (Cpz), a selective TRPV1 antagonist, the cellular flu- predominantly attributed to the CDT effect of HCuS NPs.
orescence intensity did not increase obviously as well (Figure 5D). Fluorescence images indicated that with a prolonged incuba-
Concurrently, HCuS@PDA-TRPV1/BNN6 itself without laser ir- tion time, cellular internalization of the nanocomposites was
radiation could not induce noticeable intracellular Ca2+ influx observed in U373 cells but not in TRPV1-negative Hela cells
(Figure 5B). Hence, obvious Ca2+ influx was only initiated in (Figure S13, Supporting Information). After internalization, the

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (6 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

Figure 5. A) Representative TEM images of U373 cells incubated with HCuS@PDA or HCuS@PDA-TRPV1. Fluorescence images of B) U373 cells, C)
HeLa cells, or D) Cpz pretreated U373 cells treated with different nanocomposites before and after laser irradiation. E) The relative viability of U373
cells treated with HCuS@PDA-TRPV1 or HCuS@PDA-TRPV1/BNN6 at various concentrations with or without laser irradiation (n = 3, *** p < 0.001). F)
Fluorescence images of U373 cells after various treatments stained with DCFH-DA. G) Cell viability of U373 cells or HeLa cells after being treated with
HCuS@PDA-TRPV1 and HCuS@PDA-TRPV1/BNN6. H) MMP of U373 cells analysis using JC-1 mitochondrial membrane dye. Scale bar = 50 μm.

nanocomposites largely accumulated in lysosomes. The highly with the ROS sensitive probe 2′,7′-dichlorodihydrofluorescein
acidic environment of lysosome could induce the peeling of PDA diacetate (DCFH-DA). As expected, the intracellular fluorescence
coating and facilitate HCuS NPs mediated Fenton-like reaction, images demonstrated that the fluorescence signals observed in
thus remarkably enhancing the CDT efficiency. To confirm this, HCuS@PDA-TRPV1 or HCuS@PDA-TRPV1/BNN6 treated
direct visualization of intracellular ROS level was conducted U373 cells were stronger than that in untreated cells (Figure 5F),

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (7 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

Figure 6. The biocompatibility of nanomaterials. A) Hemolysis assays for HCuS NPs and HCuS@PDA. The concentration of these nanocomposites
varied from 1 to 100 μg mL−1 . B) The body weights from tumor-free mice after intravenous administration of saline, HCuS NPs or HCuS@PDA (5 mg
kg−1 B.W.). (n = 6). C) The H&E-stained images of main organ slices collected from mice of different groups. Scale bars = 50 μm.

indicating the generation of ROS through HCuS NPs medi- Since both the Ca2+ influx and ROS production could in-
ated Fenton reaction. Critically, under the mild photothermal duce mitochondrial damage, we moved on to investigate the
condition, the HCuS@PDA-TRPV1/BNN6 treatment showed synergistic outcomes of our nanoplatform on the change of
greatly increased cytotoxicity than HCuS@PDA-TRPV1, which mitochondrial membrane potential (MMP) to further explore
was attributed to the amplified therapeutic effect of combined their toxic mechanism toward TRPV1-positive cancer cells. As
TRPV1 targeted therapy and CDT (Figure 5E). The NIR laser illustrated in Figure 5H and Figure S15A (Supporting Infor-
irradiation induced enhanced cytotoxicity of HCuS@PDA- mation), unlike the red fluorescence observed in the control
TRPV1/BNN6 was also supported by the elevated ROS level group, a red-to-green fluorescence conversion of JC-1 was ob-
in U373 cells (Figure 5F), since the intracellular Ca2+ overload served in HCuS@PDA-TRPV1 and HCuS@PDA-TRPV1/BNN6
could further amplify ROS burst. It has been reported that pretreated U373 cells, manifesting the contribution of HCuS NPs
CuS-based nanomaterials possess GSH depletion property to mediated CDT to the decrease of MMP and the damage of mi-
increase the sensitivity of tumors to ROS-associated therapy.[43] tochondria. More importantly, with laser irradiation treatment,
Based on this, the intracellular GSH content after treated with HCuS@PDA-TRPV1/BNN6-incubated U373 cells showed obvi-
nanocomposites was detected using the GSH assay kits. As ous green fluorescence, which indicated the strong synergistic
shown in Figure S14 (Supporting Information), HCuS@PDA- effect of Ca2+ influx and CDT on the alteration of MMP. Interest-
TRPV1 dramatically reduced the intracellular GSH level in a ingly, the alteration of MMP could be significantly inhibited by
concentration dependent manner. The depletion of cellular GSH Cpz, highlighting the critical role of TRPV1 activation in mito-
could efficiently elevate the intercellular ROS level and enhance chondrial dysfunction (Figure S15B, Supporting Information).
the efficiency of HCuS NPs-mediated CDT.
To demonstrate the possibility of the nanoplatform to in-
duce killing activities specifically in TRPV1-positive cancer 2.8. In Vivo Biocompatibility
cells, their toxicity on HeLa cells was also assessed. As
shown in Figure 5G, with the less localization on HeLa The in vivo biocompatibility of the nanoplatform was also de-
cells membranes, both HCuS@PDA-TRPV1 and HCuS@PDA- tected to evaluate the safety of our nanoplatform in cancer treat-
TRPV1/BNN6 were non-toxic to the TRPV1-negative cells even ment. Hemolysis assay was employed for quantitative determi-
at the concentration up to 40 μg mL−1 . Besides, NIR irradia- nation of hemoglobin to estimate the biocompatibility of HCuS
tion apparently had no effect on the cytotoxity of HCuS@PDA- NPs and HCuS@PDA via their interactions with the red blood
TRPV1 and HCuS@PDA-TRPV1/BNN6 to HeLa cells. Taken to- cells (RBCs). Figure 6A illustrated that both HCuS NPs and
gether, these findings suggested that the specific binding of the HCuS@PDA over the concentration range of 1–100 μg mL−1
nanocomposites to TRPV1 channels was of a crucial importance only exhibited 1% of RBCs hemolysis, which was clinically ac-
to trigger CDT and the photoactivation of TRPV1. ceptable according to ISO/TR 7406.[44] To further evaluate the in

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (8 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

vivo biocompatibility of HCuS NPs and HCuS@PDA, the weight sity in the in vivo therapy experiments. The average tumor vol-
fluctuation in the mice was recorded for 21 days following in- umes of mice in all groups were recorded as a function of time af-
travenous injection. As displayed in Figure 6B, treatment with ter treatment. As represented in Figure 7A, the tumor size of the
the nanocomposites even at the dose of 5 mg kg−1 body weight control group increased rapidly throughout the study. However,
(B.W.) showed no effects on the body weights gain in mice within due to the CDT activity induced by HCuS NPs, administration
21 days. On day 21, all mice pre-treated with saline and differ- of HCuS@PDA-TRPV1 and HCuS@PDA-TRPV1/BNN6 could
ent nanomaterials were sacrificed with major organs collected partially reduce tumor growth, with tumor growth inhibition
for Hematoxylin-eosin (H&E) staining (Figure 6C). Similar to (TGI) values of 40.62% and 43.03%, respectively. Laser irradiation
those from the control group, no obvious pathological changes itself showed almost no effect on the control and HCuS@PDA-
were observed in H&E-stained pathological sections of the main TRPV1 treated group. By contrast, the tumor size was suppressed
organs (hearts, livers, spleens, lungs, kidneys and brains) of obviously in mice treated with HCuS@PDA-TRPV1/BNN6 plus
the nanomaterials-treated mice. All these data revealed the good laser irradiation, with a TGI value of 82.15%, which highlighted
biocompatibility and safety of the synthesized HCuS NPs and the synergistic effect of the CDT and photo-activation of TRPV1
HCuS@PDA for in vivo therapeutic applications. channels. These findings were further supported by the images
of the nude mice bearing U373 tumors (Figure 7B).
To further evaluate the synergistic results, the H&E-stained
2.9. In Vivo Anticancer Effect tumor sections were collected for measuring cell necrosis of
each group (Figure 7C). Among all the groups, HCuS@PDA-
Inspired by the excellent in vitro therapeutic effect and good bio- TRPV1/BNN6 with laser irradiation induced the most noticeable
compatibility, we next investigated the in vivo antitumor efficacy cell apoptosis in tumor tissues, which was consistent with their
on tumor mouse models. The in vivo behavior of the nanocom- highest TGI value. Importantly, no significant difference was ob-
posites after intravenous injection were first systematically as- served in the body weights among all the experimental mice
sessed to ensure the effectiveness of the nanoplatform for in vivo (Figure 7D), indicating that our nanoplatform showed no acute
application. First, the in vivo pharmacokinetics of HCuS@PDA- toxicity in vivo. These results further highlighted the promis-
TRPV1 was investigated through detecting the Cu concentrations ing potential of our nanoplatform for spatiotemporally controlled
in blood samples after intravenous injection of HCuS@PDA- synergetic cancer therapy.
TRPV1 into BALB/c mice. As shown in Figure S16 (Support-
ing Information), the HCuS@PDA-TRPV1 nanocomposites per-
formed a prolonged blood circulation half-life of 2.69 h. In ad-
3. Conclusion
dition, the in vivo circulation behavior and biodistribution of
the nanocomposites were also measured (Figure S17, Support- In summary, a TRPV1 targeted NO-releasing nanoplatform has
ing Information). Taking advantage of the excellent photother- been fabricated and successfully applied for specific cancer ther-
mal performance of HCuS@PDA-TRPV1, IR thermal imaging apy. In this design, the biocompatible HCuS@PDA acted as not
was conducted after intravenously injected with HCuS@PDA- only Fenton-like catalysts to evoke CDT but also nanocarriers for
TRPV1. As displayed in Figure S17A (Supporting Information), encapsulation of NIR-responsive NO donors BNN6. Upon NIR
the signals of tumor areas gradually strengthened post injec- irradiation, the PDA layer can absorb the NIR light and trans-
tion and reached peak values at 4 h following injection. More- form the photons to active electrons, inducing NO release from
over, ICP-AES was also employed to analyze the distribution of BNN6 and then specific activation of TRPV1 channels. This NIR
HCuS@PDA-TRPV1 in the main tissues of U373 tumor-bearing controlled mode enabled the nanoplatform to exert its therapeu-
mice at 4 h post intravenous injection. The accumulation of tic effects below the threshold temperature (43°C) to induce pho-
HCuS@PDA-TRPV1 in tumor tissues was 10.00 ± 1.52% ID/g tothermal cell damage, minimizing the damage to normal tissue.
(Figure S17B, Supporting Information). All these results demon- Together with its CDT action, the designed nanoplatform exhib-
strated the passive accumulation of HCuS@PDA-TRPV1 in tu- ited an amplified anticancer activity with negligible systematic
mor areas. toxicity both in vitro and in vivo. As an example of NO-releasing
To evaluate the in vivo synergistic therapeutic activities of material based TRPV1 agonist, this work provides a promising
HCuS@PDA-TRPV1/BNN6, U373 tumor-bearing mice were di- strategy for site-specific cancer synergetic therapy in a spatiotem-
vided into 6 groups randomly: 1) saline (control group), 2) saline porally controlled manner.
+ laser irradiation, 3) HCuS@PDA-TRPV1, 4) HCuS@PDA-
TRPV1 + laser irradiation, 5) HCuS@PDA-TRPV1/BNN6,
and 6) HCuS@PDA-TRPV1/BNN6 + laser irradiation. The
tumor-bearing mice were intravenously injected with different
4. Experimental Section
nanocomposites (2.8 mg kg−1 B.W.) on day 0. For the photother- Synthesis of HCuS NPs: HCuS NPs were prepared using a reported
apeutic groups, the laser treatment process was conducted under method with slight modifications.[32] Briefly, copper chloride solution
strict control to keep the temperature at tumor sites lower than (0.5 m, 100 μL) was mixed with polyvinylpyrrolidone (PVP-K30) solution
43°C (Figure S18, Supporting Information). In this condition, (0.24 g, 25 mL) under continuous stirring. Next, the reaction system was
diluted with an aqueous NaOH solution (pH 9.0, 25 mL), followed by the
to avoid the burn injury by hyperthermia and simultaneously addition of hydrazine anhydrous solution (50%, 6.4 μL) within 5 min. After
achieve satisfactory therapeutic effect, 0.4 W cm−2 , the human mixed with the sodium sulphide aqueous solution (320 mg mL−1 , 200 μL),
skin-sustainable maximum value permitted by the American Na- the reaction was allowed to proceed at 60°C for 2 h. Then, the precipitate
tional Standards Institute,[45] was chosen as the laser power den- was collected by centrifugation at 10,000 rpm for 10 min and washed with

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (9 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

Figure 7. A) The mean tumor volumes of U373-tumor-bearing nude mice after different treatments within 16 days (n = 6, ** p ˂ 0.01, *** p ˂ 0.001). For the
phototherapeutic groups, mice at 4 h after intravenously injecting with different nanocomposites (2.8 mg kg−1 B.W.) were exposed to the 808 nm laser
(0.4 W cm−2 , 5 min). B) Representative digital photos of mice in day 0 and 16 after different treatments. C) The H&E-staining images of the dissected
tumor tissues after 16 days of treatment. Scale bar = 50 μm. D) The body weights of mice from different groups within 16 days (n = 6). Note that
1–6 represents different groups: 1) saline, 2) saline + 808 nm laser, 3) HCuS@PDA-TRPV1, 4) HCuS@PDA-TRPV1 + 808 nm laser, 5) HCuS@PDA-
TRPV1/BNN6, and 6) HCuS@PDA-TRPV1/BNN6 + 808 nm laser.

D.I. water and ethanol for three times, respectively. The product was finally TRPV1/BNN6 at different concentrations. For phototherapeutic groups,
dried under vacuum. the cells were exposed to 808 nm laser irradiation (0.5 W cm−2 , 5 min) at
Preparation of HCuS@PDA/BNN6: The HCuS@PDA/BNN6 2 h after pretreated with nanocomposites. After further incubated for 24 h,
nanocomposites were synthesized through in situ polymerization standard MTT assay was employed to evaluate the cell viability.
method. HCuS NPs (1 mg) were first diluted to the tris(hydroxymethyl) Intracellular Calcium Analysis: U373 cells or HeLa cells (1 × 105 cells
aminomethane solution (10 mm, pH 8.5, 10 mL) under magnetic stirring. per well) were cultured in 24-well plates for 24 h. Then, fresh medium con-
Then, a DMSO solution of BNN6 (0.48 mg, 1 mL) and different weight of taining CaCl2 (150 mg L−1 ) was added to replace the culture medium.
dopamine hydrochloride (2 mg, 1 mg and 0.5 mg) were added into the In the case of Cpz treatment groups, cells were pre-incubated with Cpz
suspension. Afterward, the reaction mixture was further stirred overnight (1 mm) for 30 min, followed by the addition of HCuS@PDA-TRPV1 or
at 25°C. Finally, the HCuS@PDA/BNN6 nanocomposites were obtained HCuS@PDA-TRPV1/BNN6 (40 μg mL−1 ). After 2 h of treatment, the cells
via centrifugation and washing several times with D.I. water. The loading were loaded with Fluo-3 AM for 1 h in the dark at 37°C following the stan-
capacity of BNN6 was measured by assessing its UV–vis absorption at dard protocol. Finally, the cells were irradiated by an 808 nm laser (0.5 W
252 nm. The BNN6 loading efficiency of nanocomposites was calculated cm−2 ) for 5 min, and the intracellular calcium images were collected by
as follows: fluorescence microscope.
Intracellular Mitochondrial Membrane Potential Test: Briefly, U373 cells
Loading efficiency (%) = (m0 − m1 ) ∕m × 100% (1) were cultured in 24-well plates (1 × 105 cells per well) for 24 h. After that,
the culture medium was changed by the fresh medium containing CaCl2
where m0 and m1 were the initial and equilibrium mass of BNN6, respec- (150 mg L−1 ). Subsequently, the cells were treated with HCuS@PDA-
tively, and m was the mass of HCuS@PDA. TRPV1 or HCuS@PDA-TRPV1/BNN6 (40 μg mL−1 ) for 2 h. Then, the cells
Preparation of HCuS@PDA-TRPV1/BNN6: The HCuS@PDA/BNN6 were irradiated by an 808 nm laser (0.5 W cm−2 ) for 5 min, followed by a
(10 mg) and 50% glutaraldehyde solution (1 mL) were added into phos- further incubation of 12 h. Finally, the membrane potential dye JC-1 was
phate buffer (pH 7.4, 20 mL), followed by stirring at 25°C for 2 h. After used to stain with the cells following the standard protocol. The images
addition of anti-TRPV1 antibody (60 μg), the reaction was continued for were recorded by fluorescence microscope.
24 h under 8°C with constant stirring. The product was centrifuged, and Animals Experiment: All animal experimental and handling procedures
washed with D.I. water for several times. were in accordance with the guidelines of the Hebei committee for care
Detection of NO Release In Vitro: The release of NO from and use of laboratory animals, and approved by the Ethics Committee
HCuS@PDA/BNN6 was triggered by NIR laser irradiation. Specifi- for Animal Experimentation of Hebei Medical University (IACUC-Hebmu-
cally, the HCuS@PDA/BNN6 suspension (0.5 mg mL−1 ) was exposed to 2022040). All the animals were housed in a controlled environment with
an 808 nm laser (0.5 W cm−2 ). After irradiation for different times, the free access to food and water.
supernatant was obtained via centrifugation at 12 000 rpm for 2 min. NO In Vivo Toxicity Test: Six-week-old female BALB/c mice were provided
assay kit was applied to monitor the amount of released NO. Moreover, by the Experimental Animal Center of the Chinese Academy of Medical Sci-
the NO release behavior of HCuS@PDA/BNN6 at different temperatures ences. To test the toxicity of the nanocomposites in vivo, the mice were ran-
(25, 37, and 60°C) was also detected with the same protocols. domized into three groups (n = 6), and then were intravenously injected
Cell Toxicity Assays: U373 cells (5 × 103 cells per well) were cultured in with saline, saline containing HCuS NPs or HCuS@PDA (5 mg kg−1 B.W.),
96-well plates for 24 h and treated by HCuS@PDA-TRPV1 or HCuS@PDA- respectively. The body weights of the mice were recorded every day. 21 days

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (10 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

after treatment, the hearts, livers, spleens, lungs, kidneys and brains of the Keywords
mice were harvested and fixed with 4% paraformaldehyde for 24 h. After-
ward, the organs were embedded in paraffin and cut into 4 μm sections to chemodynamic therapy, NO nanogenerators, remote manipulation, spe-
carry out the Hematoxylin-eosin (H&E) staining. Images were taken using cific cancer therapy, TRPV1 channels
an optical microscope.
In Vivo Biodistribution: The nude mice bearing subcutaneous U373 tu- Received: October 17, 2023
mors were treated with HCuS@PDA-TRPV1 (2.8 mg kg−1 B.W.) via intra- Revised: December 19, 2023
venous injection. After administration for 0, 2, 4, 8, and 24 h, the tumor Published online: January 2, 2024
sites were irradiated by an 808 nm laser (0.5 W cm−2 ) for 5 min. And the
photothermal images were collected using FLIR infrared camera.
The U373 tumor-bearing mice were treated with saline or HCuS@PDA-
TRPV1 (25 mg kg−1 B.W.) via intravenous injection. After 4 h, mice were
sacrificed, hearts, livers, spleens, lungs, kidneys and tumors of the mice
[1] R. L. Siegel, K. D. Miller, N. S. Wagle, A. Jemal, CA Cancer J. Clin. 2023,
were collected to determine the Cu content in the samples by ICP-AES.
73, 17.
In Vivo Tumor Therapy: The tumor-bearing models were established
[2] M. I. Khan, M. I. Hossain, M. K. Hossain, M. H. K. Rubel, K. M.
by subcutaneous injection of 1 × 108 U373 cells suspended in PBS
(100 μL) into six weeks-old female BALB/c nude mice. Then, the mice Hossain, A. M. U. B. Mahfuz, M. I. Anik, ACS Appl. Bio. Mater. 2022,
were randomized into six groups (n = 6) for different treatment when 5, 971.
the tumor volume reached ≈ 110–130 mm3 : 1) saline, 2) saline + [3] M. T. Manzari, Y. Shamay, H. Kiguchi, N. Rosen, M. Scaltriti, D. A.
808 nm laser, 3) HCuS@PDA-TRPV1, 4) HCuS@PDA-TRPV1 + 808 nm Heller, Nat. Rev. Mater. 2021, 6, 351.
laser, 5) HCuS@PDA-TRPV1/BNN6, and 6) HCuS@PDA-TRPV1/BNN6 + [4] X. Zhen, C. Xie, Y. Jiang, X. Ai, B. Xing, K. Pu, Nano Lett. 2018, 18,
808 nm laser. On day 0, the mice were intravenously injected with saline, 1498.
HCuS@PDA-TRPV1 (2.8 mg kg−1 B.W.) or HCuS@PDA-TRPV1/BNN6 [5] L. Li, C. Chen, C. Chiang, T. Xiao, Y. Chen, Y. Zhao, D. Zheng, Int. J.
(2.8 mg kg−1 B.W.). For the phototherapeutic groups, the tumor sites of Biol. Sci. 2021, 17, 2034.
the mice were exposed to 808 nm laser irradiation (0.4 W cm−2 , 5 min) at [6] X. Ni, W. Shi, Y. Liu, L. Yin, Z. Guo, W. Zhou, Q. Fan, Small 2022, 18,
4 h after administration. The tumor size and body weight of all mice were 2200152.
recorded every other day. The equation V = a × b2 /2 was used to calculate [7] T. T. L. Wu, A. A. Peters, P. T. Tan, S. J. Roberts-Thomson, G. R.
the tumor volume, where a and b represent the longest and shortest tu- Monteith, Cell Calcium 2014, 56, 59.
mor diameters, respectively. In addition, the tumor growth inhibition rate [8] S. Xu, X. Cheng, L. Wu, J. Zheng, X. Wang, J. Wu, H. Yu, J. Bao, L.
(TGI%) was determined via using the following equation: Zhang, Cellular Signal 2020, 75, 109733.
[9] Z. Ma, J. Zhang, W. Zhang, M. F. Foda, Y. Zhang, L. Ge, H. Han,
TGI (%) = (Vc − Vt ) ∕ (Vc − V0 ) × 100% (2) iScience 2020, 23, 101049.
[10] J. Song, J.-B. Pan, W. Zhao, H.-Y. Chen, J.-J. Xu, Chem. Commun. 2020,
where Vc and Vt refer to the mean tumor volumes of mice in the control
56, 6118.
and treated groups on day 16, respectively, and V0 refers to the mean tu-
[11] J. O. Lundberg, E. Weitzberg, M. T. Gladwin, Nat. Rev. Drug Discov.
mor volume of control group on day 0.
2008, 7, 156.
Statistical Analysis: All of the data in this study were presented as mean
± standard deviation. One-way analysis of variance (ANOVA) was used to [12] X. Chen, F. Jia, Y. Li, Y. Deng, Y. Huang, W. Liu, Q. Jin, J. Ji, Biomaterials
evaluate the statistical significance between different groups. Significant 2020, 246, 119999.
differences were denoted by * p < 0.05, ** p < 0.01 and *** p < 0.001, re- [13] R. C. Maurya, J. M. Mir, Chem. Biol. Potent. Nat. Prod. Syn. Comp.
spectively. 2021, 20, 343.
[14] T. Yoshida, R. Inoue, T. Morii, N. Takahashi, S. Yamamoto, Y. Hara,
M. Tominaga, S. Shimizu, Y. Sato, Y. Mori, Nat. Chem. Biol. 2006, 2,
Supporting Information 596.
[15] N. Ito, U. T. Ruegg, A. Kudo, Y. Miyagoe-Suzuki, S. Takeda, Nat. Med.
Supporting Information is available from the Wiley Online Library or from
2013, 19, 101.
the author.
[16] H.-J. Xiang, J.-G. Liu, Y. Zhao, Acta Phys. Chim. Sin. 2017, 33, 903.
[17] Y. Zhao, X. Ouyang, Y. Peng, S. Peng, Pharmaceutics 2021, 13, 1917.
Acknowledgements [18] J. Kim, D. M. Francis, L. F. Sestito, P. A. Archer, M. P. Manspeaker, M.
J. O’melia, S. N. Thomas, Nat. Commun. 2022, 13, 1479.
S.W. and Y.W. contributed equally to this work. This work was supported by [19] Y. Yang, Z. Huang, L.-L. Li, Nanoscale 2021, 13, 444.
the Central Guidance on Local Science and Technology Development Fund [20] T. Feng, J. Wan, P. Li, H. Ran, H. Chen, Z. Wang, L. Zhang, Biomaterials
of Hebei Province (226Z2601G), the Youth Top-notch Talents Support- 2019, 214, 119213.
ing Plan of Hebei Province (QNBJ19004), the Scientific Research Foun- [21] Z. Liu, Y. Zhong, X. Zhou, X. Huang, J. Zhou, D. Huang, Y. Li, Z. Wang,
dation of Hebei Province for the Returned Overseas Chinese Scholars
B. Dong, H. Qiao, W. Chen, Biomaterials 2021, 277, 121118.
(C20220508), the Science and Technology Project of Hebei Education De-
[22] A. De La Harpe, N. Beukes, C. L. Frost, Biotechnol. Appl. Biochem.
partment (no. ZD2021072), and the Postdoctoral Fund of Hebei Medical
2022, 69, 420.
University.
[23] S. Wang, J. Lv, Y. Pang, S. Hu, Y. Lin, M. Li, J. Mater. Chem. B 2022,
10, 748.
Conflict of Interest [24] D. Wang, X. Zhu, X. Wang, Q. Wang, K. Yan, G. Zeng, G. Qiu, R. Jiao,
X. Lin, J. Chen, Q. Yang, W. Qin, J. Liu, K. Zhang, Y. Liu, Adv. Funct.
The authors declare no conflict of interest. Mater. 2023, 33, 2303869.
[25] J. Zhu, A. Jiao, Q. Li, X. Lv, X. Wang, X. Song, B. Li, Y. Zhang, X. Dong,
Data Availability Statement Acta Biomater. 2021, 137, 252.
[26] D. Jana, Y. Zhao, Exploration 2022, 2, 20210238.
The data that support the findings of this study are available from the cor- [27] Y. Huang, S. Wu, L. Zhang, Q. Deng, J. Ren, X. Qu, ACS Nano 2022,
responding author upon reasonable request. 16, 4228.

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (11 of 12) © 2024 Wiley-VCH GmbH
21922659, 2024, 10, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/adhm.202303579 by Universidad De Salamanca, Wiley Online Library on [30/04/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advhealthmat.de

[28] J. Xin, C. Deng, O. Aras, M. Zhou, C. Wu, F. An, J. Nanobiotechnol. [37] K. Yang, L. Zhu, L. Nie, X. Sun, L. Cheng, C. Wu, G. Niu, X. Chen, Z.
2021, 19, 192. Liu, Theranostics 2014, 4, 134.
[29] Y.-N. Hao, W.-X. Zhang, Y.-R. Gao, Y.-N. Wei, Y. Shu, J.-H. Wang, J. [38] J. Fan, N. He, Q. He, Y. Liu, Y. Ma, X. Fu, Y. Liu, P. Huang, X. Chen,
Mater. Chem. B 2021, 9, 250. Nanoscale 2015, 7, 20055.
[30] L.-H. Fu, Y. Wan, C. Qi, J. He, C. Li, C. Yang, H. Xu, J. Lin, P. Huang, [39] Q. Deng, L. Zhang, W. Lv, X. Liu, J. Ren, X. Qu, ACS Nano 2021, 15,
Adv. Mater. 2021, 33, 2006892. 6604.
[31] Z. Wang, Y. Shi, Y. Shi, J. Zhang, R. Hao, G. Zhang, L. Zeng, ACS Appl. [40] G. Wei, G. Yang, B. Wei, Y. Wang, S. Zhou, Acta Biomater. 2019, 100,
Mater. Interfaces 2022, 14, 54478. 365.
[32] J. Lv, S. Wang, D. Qiao, Y. Lin, S. Hu, M. Li, J. Nanobiotechnol. 2022, [41] J. Gehring, B. Trepka, N. Klinkenberg, H. Bronner, D. Schleheck, S.
20, 42. Polarz, J. Am. Chem. Soc. 2016, 138, 3076.
[33] X. Deng, K. Li, X. Cai, B. Liu, Y. Wei, K. Deng, Z. Xie, Z. Wu, P. ’. Ma, [42] Y. Lyu, C. Xie, S. A. Chechetka, E. Miyako, K. Pu, J. Am. Chem. Soc.
Z. Hou, Z. Cheng, J. Lin, Adv. Mater. 2017, 29, 1701266. 2016, 138, 9049.
[34] S. Bhogal, I. Mohiuddin, S. Kumar, A. K. Malik, K.-H. Kim, K. Kaur, [43] M. Luo, H. Yukawa, K. Sato, M. Tozawa, M. Tokunaga, T. Kameyama,
Sci. Total Environ. 2022, 847, 157356. T. Torimoto, Y. Baba, ACS Appl. Mater. Interfaces 2022, 14, 34365.
[35] F. Cao, Y. Zhang, Y. Sun, Z. Wang, L. Zhang, Y. Huang, C. Liu, Z. Liu, [44] I. Carreira-Barral, C. Rumbo, M. Mielczarek, D. Alonso-Carrillo, E.
J. Ren, X. Qu, Chem. Mater. 2018, 30, 7831. Herran, M. Pastor, A. Del Pozo, M. García-Valverde, R. Quesada,
[36] F. Xu, Y. Tang, H. Wang, H. Deng, Y. Huang, C. Fan, J. Zhao, C. Lin, Y. Chem. Commun. 2019, 55, 10080.
Lin, Small 2022, 18, 2201205. [45] H. Yuan, A. M. Fales, T. Vo-Dinh, J. Am. Chem. Soc. 2012, 134, 11358.

Adv. Healthcare Mater. 2024, 13, 2303579 2303579 (12 of 12) © 2024 Wiley-VCH GmbH

You might also like