ArticlePDF Available

L-Arginine Self-Delivery Supramolecular Nanodrug for NO Gas Therapy

Authors:

Abstract and Figures

NO gas therapy is a supplementary approach for tumor treatment due to the advantages of minimal invasion, little drug resistance, low side effect and amplified efficacy. l-Arginine (L-Arg), a natural NO source with good biocompatibility, can release NO under the stimulation of H2O2 in tumor microenvironment. However, the conventional l-Arg-delivery systems via noncovalent loading usually lead to inevitable premature leakage of nano-cargos during blood circulation. In this work, an efficient l-Arg-self-delivery supramolecular nanodrug (SDSND) for tumor treatment is demonstrated by combining Mannich reaction and π-π stacking. l-Arg-links to (-)-epigallocatechin gallate (EGCG) with the assistance of formaldehyde through Mannich reaction, and then assembles into nanometer-sized particles via π-π stacking. The guanidine group of l-Arg-and the phenolic hydroxyl groups of EGCG are preserved in the SDSNDs, which allows for accomplishing gas therapy by provoking tumor cell apoptosis and combining with EGCG to amplify apoptosis, respectively. In addition, the SDSNDs exhibit high biocompatibility and avoid the premature leakage of l-Arg-in blood circulation, providing an alternative l-Arg-delivery system for NO gas therapy. STATEMENT OF SIGNIFICANCE: NO gas therapy has attracted emerging interest in tumor treatment. However, the controlled NO release and the avoidance of premature leakage of NO donors remain challenging. In this work, l-Arg (L-Arginine) self-delivery supramolecular nanodrug for efficient tumor therapy is demonstrated through the Mannich reaction of l-Arg, (-)-epigallocatechin gallate (EGCG) and formaldehyde. Stimulated by tumor microenvironment, the guanidine groups of l-Arg-allow for accomplishing NO release and thus provoking tumor cell apoptosis. The nanodrug also avoids the premature leakage of l-Arg-in blood circulation. Moreover, the preserved phenolic hydroxyl groups of EGCG combine with l-Arg-to amplify apoptosis. The nanodrug exhibits high biocompatibility and good therapeutic effect, providing an alternative l-Arg-delivery system for NO gas therapy.
Content may be subject to copyright.
Acta Biomaterialia 169 (2023) 517–529
Contents lists available at ScienceDirect
Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actbio
Full length article
L-Arginine self-delivery supramolecular nanodrug for NO gas therapy
Mengsi Zhang
a
, Hao Jin
a
, Yi Liu
a , b
, Lanlan Wan
c , , Shuwei Liu
b , , Hao Zhang
a , b , d , ∗∗
a
State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012 , PR China
b
Joint Laboratory of Optical Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 13 00 21, PR China
c
Department of Anesthesia, The Second Hospital of Jilin University, Changchun 13 00 41, PR China
d
Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou 450 0 01, PR China
a r t i c l e i n f o
Article history:
Received 19 April 2023
Revised 3 July 2023
Accepted 27 July 2023
Available online 1 August 2023
Keywo rds:
Gas therapy
L-Arginine
Supramolecular nanodrugs
Polyphenol
Antitumor
a b s t r a c t
NO gas therapy is a supplementary approach for tumor treatment due to the advantages of minimal
invasion, little drug resistance, low side effect and amplified efficacy. l -Arginine (L-Arg), a natural NO
source with good biocompatibility, can release NO under the stimulation of H
2
O
2
in tumor microenviron-
ment. However, the conventional l -Arg delivery systems via noncovalent loading usually lead to inevitable
premature leakage of nano-cargos during blood circulation. In this work, an efficient l -Arg self-delivery
supramolecular nanodrug (SDSND) for tumor treatment is demonstrated by combining Mannich reac-
tion and π- πstacking. l -Arg links to (-)-epigallocatechin gallate (EGCG) with the assistance of formalde-
hyde through Mannich reaction, and then assembles into nanometer-sized particles via π- πstacking. The
guanidine group of l -Arg and the phenolic hydroxyl groups of EGCG are preserved in the SDSNDs, which
allows for accomplishing gas therapy by provoking tumor cell apoptosis and combining with EGCG to
amplify apoptosis, respectively. In addition, the SDSNDs exhibit high biocompatibility and avoid the pre-
mature leakage of l -Arg in blood circulation, providing an alternative l -Arg delivery system for NO gas
therapy.
Statement of significance
NO gas therapy has attracted emerging interest in tumor treatment. However, the controlled NO release
and the avoidance of premature leakage of NO donors remain challenging. In this work, L-Arginine (L-Arg)
self-delivery supramolecular nanodrug for efficient tumor therapy is demonstrated through the Mannich
reaction of L-Arg, (-)-epigallocatechin gallate (EGCG) and formaldehyde. Stimulated by tumor microenvi-
ronment, the guanidine groups of L-Arg allow for accomplishing NO release and thus provoking tumor
cell apoptosis. The nanodrug also avoids the premature leakage of L-Arg in blood circulation. Moreover,
the preserved phenolic hydroxyl groups of EGCG combine with L-Arg to amplify apoptosis. The nanodrug
exhibits high biocompatibility and good therapeutic effect, providing an alternative L-Arg delivery system
for NO gas therapy.
©2023 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
1. Introduction
Gas therapy, which utilizes gaseous molecules accumulation to
accomplish diseases therapy, has attracted emerging interest in the
past decade [1–4] . As the most competitive candidate among gas
signaling molecules, such as carbon monoxide, hydrogen sulfide
and oxygen, nitric oxide (NO) is prominent in neural communi-
Corresponding authors.
∗∗ Corresponding author at: State Key Laboratory of Supramolecular Structure and
Materials, College of Chemistry, Jilin University, Changchun 13 00 12, PR China.
E-mail addresses: wanll@jlu.edu.cn (L. Wan) , liushuwei@jlu.edu.cn (S. Liu),
hao_zhang@jlu.edu.cn (H. Zhang) .
cation, vascular regulation, wound healing and other physiological
activities, making it the preferred model in innovative tumor ther-
apeutics featured with combination and cascade [5–9] . High con-
centrations of NO ( > 1 μM) can kill tumor cells through a vari-
ety of pathways, including nitrite stress, mitochondrial/DNA dam-
age, apoptosis and et al. [ 10 , 11 ]. Although a variety of NO sources,
for instance, S-nitrosoglutathione, S-nitroso-N-acetyl penicillamine
and nitrate functionalized d - α-tocopheryl polyethylene glycol suc-
cinate, are demonstrated to support NO-mediated tumor treatment
[12–14] , the complicated preparation procedures, high costs and
potential toxicity cause the doubt in clinical utilization.
L-Arginine (L-Arg), an endogenously existing and exogenously
supplemental amino acid with high biocompatibility, can release
https://doi.org/10.1016/j.actbio.2023.07.055
1742-7061/© 2023 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
NO in organisms as its guanidine group is catalyzed by nitric oxide
synthase [15–17] . In addition, l -Arg can react with the excessive
H
2
O
2
in tumor microenvironment (TME) to produce NO via non-
enzymatic pathway, achieving targeting NO release for tumor ther-
apy with high biosafety [18–23] . Since the direct supply of exoge-
nous l -Arg molecules suffers from the shortcomings of short half-
life and low bioavailability, efficient l -Arg delivery to tumor sites
is required [24–27] . However, the conventional l -Arg delivery sys-
tems constructed through noncovalent linkages [28–32] inevitably
lead to premature leakage of the cargos during blood circulation.
Alternative delivery systems with stable l -Arg loading and control-
lable NO release are greatly welcome [ 33 , 34 ].
In this context, besides guanidine group, l -Arg possesses amino
and carboxyl groups, which is covalently linkable with other
segments to avoid premature leakage [ 35 , 36 ]. Mannich reac-
tion, a phenol-aldehyde type reaction, is expandable to multi-
component condensation in the presence of amine, aldehyde and
the molecules with active hydrogens [37] . With respect to l -Arg, its
amino group is available for Mannich reaction to link with a vari-
ety of functional molecules for designing novel therapeutic systems
[38] . (-)-Epigallocatechin gallate (EGCG) is a widely employed nat-
ural polyphenol in therapeutic applications, which exhibits plenty
of pharmacological functions caused by the abundant phenolic hy-
droxyl groups, such as inhibiting tumor cell proliferation and in-
ducing apoptosis [ 39 , 40 ]. Meanwhile, the active hydrogens adja-
cent to the phenolic hydroxyl groups of EGCG can be conducive
to modifying functional molecules [41] . And the conjugated frame-
work of EGCG favors the π- πstacking of neighboring molecules
[42] . According to the structural characteristics of EGCG and l -Arg,
it is reasonable to design self-delivery supramolecular nanodrugs
(SDSNDs) for tumor treatment with the combination of NO gas
therapy and EGCG-contributed apoptosis amplification.
In this work, Mannich reaction is employed to link l -Arg and
EGCG with the assistance of formaldehyde. The condensation prod-
ucts further assemble into SDSNDs through the π- πstacking of
EGCG framework. The covalent linkage between l -Arg and EGCG
avoids premature leakage of l -Arg, significantly improving the half-
life and bioavailability in drug delivery. Because the guanidine
group of l -Arg and the phenolic hydroxyl groups of EGCG are pre-
served in the as-produced SDSNDs, NO gas therapy by provoking
tumor cell apoptosis and the apoptosis amplification from EGCG
are realized. Further investigations reveal that the release of NO
is stimulated by the excessive H
2
O
2
in TME, and the efficacy in
HepG2 tumor model therapy is co-contributed by NO-resulted tu-
mor cell mitochondrial/DNA damage and apoptosis, EGCG-induced
tumor cell proliferation inhibition and apoptosis.
2. Materials and methods
2.1. Chemicals and reagents
(-)-Epigallocatechin gallate (EGCG) was purchased from Al-
addin Industrial Corporation (Shanghai, China). l -Arginine (L-
Arg), acetonitrile and paraf ormaldehyde were bought from Mack-
lin Biochemical Co., Ltd. (Shanghai, China). Formaldehyde (HCHO)
was purchased from Yongsheng Fine Chemical Co., Ltd (Tianjin,
China). 2,4-Dinitrophenylhydrazine (DNPH) was purchased from
Xiya reagent (Shandong, China). Folin-Ciocalteu’s phenol reagent,
cell counting kit-8 (CCK-8), Annexin V-FITC/PI apoptosis detection
kit, saline and Dulbecco’s modified eagle medium (DMEM) were
obtained from Beijing Solarbio Science & Technology Co., Ltd. (Bei-
jing, China). Trypsin-EDTA solution and phosphate buffer saline
(PBS) were supplied by Biosharp. Fetal bovine serum (FBS) was
provided by Zhejiang Tianhang Biotechnology Co., Ltd. (Hangzhou,
China). Indocyanine green (ICG) was provided from Shanghai
yuanye Bio-Technology Co., Ltd. Griess reagent, hydrogen peroxide
assay kit, hoechst 33,342 staining solution for live cells, 3-amino,4-
aminomethyl-2
,7
-difluorescein, diacetate (DAF-FM DA), Calcein/PI
cell viability/cytotoxicity assay kit, enhanced mitochondrial mem-
brane potential assay kit with JC-1, immunostaining permeabiliza-
tion buffer with Triton X-100 and crystal violet staining solution
were purchased from Beyotime Biotechnology Co., Ltd. (Shanghai,
China). γ-H2AX (phospho S139, Alexa Fluor 568) antibody was
purchased from Abcam (Cambridge, UK). All reagents are used
without further purification.
2.2. Characterization
The transmission electron microscopy (TEM) images and scan-
ning transmission electron microscopy with energy-dispersive X-
ray spectroscopy (STEM-EDS) elemental mapping images were cap-
tured by using a transmission electron microscopy (JEOL JEM-
2100F). The scanning electron microscopy (SEM) images were ob-
tained by field emission scanning electron microscopy (HITACHI
SU8020). The hydrated diameter and zeta potential were recorded
by using a zetasizer (Malvern NanoZS). The ultraviolet-visible-
near infrared (UV–vis-NIR) absorption spectra were performed on
a spectrophotometer (Shimadzu 2600). The Fourier-transform in-
frared (FTIR) spectra were measured by a Fourier transform in-
frared spectrometer (Brucker VRTEX 80 V). The content of N ele-
ment was detected on a CHNS/O elemental analyser (Agilent). The
HCHO content was measured by using a high performance liquid
chromatograph mass spectrometer (HPLC-MS, Agilent1290 - Bruker
micrOTOF QII). The absorbance tests in vitro were performed on
a microplate reader (BioTek Synergy LX). The quantity and mor-
phology of cells were observed by an inverted microscope. The cell
apoptosis was measured by a flow cytometer (BD FACSCalibur). The
fluorescent images were recorded by using the confocal laser scan-
ning microscope (CLSM, Nikon AXR). The fluorescent images were
performed with a near-infrared fluorescence imager (Raptor pho-
tonics Ninox 640 II).
2.3. Preparation of L-Arg SDSNDs
EGCG (1 mM) and HCHO (2 mM) were first stirred for seconds
in an aqueous solution containing 5% ethanol, then mixed with l -
Arg (2 mM) and stirred for 1 h at room temperature. The l -Arg
SDSNDs were obtained by centrifugation at 15, 0 0 0 rpm for 10 min,
and washed 3 times with diluted water.
2.4. Preparation of ICG-L-Arg SDSNDs
The pre-prepared l -Arg SDSNDs (1 mg/mL) were stirred with
ICG (0.5 mg/mL) at room temperature for 24 h. The ICG- l -Arg SD-
SNDs were obtained by centrifuging at 15, 0 0 0 rpm for 10 min.
2.5. Determination of EGCG content in L -Arg SDSNDs
The Folin-Ciocalteu method was adopted to determine the
EGCG content. Folin-Ciocalteu’s phenol reagent A and B were
freshly prepared. Briefly, l -Arg SDSNDs solution was mixed with
reagent A for 10 min, then mixed with reagent B followed by
30 min incubation at room temperature. And the absorbance at
750 nm was measured by a spectrophotometer. The calibration
curve was also measured by using EGCG at different concentrations
(0 to 150 μg/mL).
2.6. Measurement of HCHO content
The HCHO residue was measured by using HPLC-MS. The l -Arg
SDSNDs solution (4 mg/mL) was incubated with DNPH at 40 °C
for 60 min. Then the supernatant was collected for HPLC-MS test.
518
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
Standard HCHO solutions (0, 1 and 2 μg/mL) were also detected in
the same way.
2.7. Verification of interaction
The l -Arg SDSNDs were incubated with DMSO, urea, NaCl and
Triton X-100 solution, respectively. After 12 h incubation, the pre-
cipitates were centrifugated for TEM tests.
2.8. Exploration of composition release
A dialysis bag containing l -Arg SDSNDs is placed and stirred in
PBS aqueous solution at 37 °C. At the designated time points (0,
0.5, 1, 2, 4, 8, 12 and 24 h), the PBS aqueous solution was tested
using UV–vis absorption spectra.
2.9. NO detection in vitro
The generation of NO was detected by using Griess reagent
method and DAF-F M DA method. Different solutions (H
2
O
2
, l -Arg,
l -Arg SDSNDs, l -Arg + H
2
O
2
and l -Arg SDSNDs + H
2
O
2
) were in-
cubated at 37 °C for 1 h, respectively. For NO quantitative detec-
tion, the l -Arg SDSNDs at different concentrations (0, 250, 500 and
10 0 0 μg/mL) were incubated in the aqueous solution containing
H
2
O
2
(500 μM) at 37 °C for 10 min, 30 min, 1 h, 6 h and 24 h, re-
spectively. The l -Arg SDSNDs (200 μg/mL) were also incubated in
the aqueous solution containing H
2
O
2
(0, 10, 50, 100 and 200 μM)
at 37 °C for 2 and 4 h. Then the Griess reagent was added and
the supernatant was collected for UV–vis absorption detection. The
calibration curve was also obtained in the same procedure. For an-
other method, DAF -FM DA was added and incubated at 37 °C for
30 min in dark. The supernatant was collected for fluorescent de-
tection (Ex 495 nm, Em 515 nm).
2.10. Cell toxicity assay
The l -Arg SDSNDs with different concentrations were incubated
with mouse fibroblast cell lines (L929) and human hepatoma cell
lines (HepG2), respectively. After 24 h incubation, the cell viability
was measured by using the CCK-8 method.
2.11. Cell apoptosis assay
The l -Arg SDSNDs with different concentrations (0 to
200 μg/mL) were incubated with HepG2 cells. After 24 h in-
cubation, the cell apoptosis was measured by using Annexin
V-FITC/PI apoptosis detection kit on a flow cytometer.
2.12. Cell live/dead assay
The l -Arg SDSNDs with different concentrations (0 to
200 μg/mL) were incubated with HepG2 cells. After 24 h
incubation, the cells were stained with Calcein/PI cell viabil-
ity/cytotoxicity assay kit. The fluorescent images were recorded
using CLSM.
2.13. Cellular NO detection
The cellular NO generation was revealed with DAF-FM DA by
CLSM. The HepG2 cells were incubated with l -Arg SDSNDs (0, 50,
10 0 and 20 0 μg/mL) for 6 h, then washed with PBS. The DAF-FM
DA was added and incubated at 37 °C for 30 min in dark. After-
ward, the hoechst solution was employed to stain cell nucleus for
10 min. Then the fluorescent images were captured (Ex = 405 and
488 nm).
2.14. Mitochondrial membrane potential detection
The HepG2 cells were incubated with l -Arg SDSNDs (0, 50, 10 0
and 200 μg/mL) for 24 h. The JC-1 staining working solution was
added and incubated at 37 °C for 20 min in dark. Then the cells
were washed with JC-1 staining buffer solution 2 times. Afterward,
the mitochondrial membrane potential was observed using CLSM.
2.15. DNA damage assay
The HepG2 cells were incubated with l -Arg SDSNDs (0, 50, 10 0
and 200 μg/mL) for 24 h. Then the cells were fixed, permeabilized,
blocked and stained with γ-H2AX antibody. And the hoechst solu-
tion was employed to stain the cell nucleus for 10 min. Then the
fluorescent images were captured.
2.16. Colony forming cell (CFC) assay
The HepG2 cells were incubated with l -Arg SDSNDs (0, 25,
50, 100, 150 and 200 μg/mL) for 24 h. Then 500 cells of each
group were reseeded, respectively. After a 15-day incubation, the
cells were stained with crystal violet staining solution and pho-
tographed.
2.17. Hemolysis assay
The mouse red blood cells (2%) were incubated with Triton X-
100 (positive group), PBS (negative group) and PBS solution con-
taining l -Arg SDSNDs (0, 25, 50, 100, 150 , 200 and 500 μg/mL, ex-
perimental groups) at 37 °C for 1 h. Each group was centrifugated
at 30 0 0 rpm for 10 min and photographed. The supernatant was
collected for 541 nm absorbance (A) measurement. The hemolysis
ratio was calculated according to the following formula: hemolysis
ratio (%) = (A
experimental
- A
negative
) / (A
positive
- A
negative
)
100%.
2.18. Fluorescent images in vivo
The mouse was injected with 10 0 μL ICG- l -Arg SDSNDs
(5 mg/mL) through vein tails. The fluorescent images of the mouse
were captured by using a near-infrared fluorescence imager before
injection (control) and after injection.
2.19. Animal experiment
The animal experiments were carried out with the approval
(NO. 0693) of the Animal Laboratory Ethics Committee of the First
Hospital of Jilin University. The Balb/c nude mice (5 weeks, female)
were bought from Beijing Vital River Laboratory Animal Technol-
ogy Co., Ltd. The mice were subcutaneously injected with HepG2
cells on the right buttocks about 12 days in advance ( 12 d). Af-
ter building the tumor models, the mice were randomly divided
into four groups: control, l -Arg, EGCG and l -Arg SDSNDs. From 0
d to 16 d, the mice were injected without or with 127.5 μg l -Arg,
359.5 μg EGCG and 500 μg l -Arg SDSNDs solutions, respectively,
every two days. All solutions were at the equivalent concentration
of 5 mg/mL l -Arg SDSNDs. At the same time, the body weight and
tumor volume also need to be recorded. On 16 d, the blood was
taken for the liver and kidney function test. And all tumors, hearts,
livers, lungs, kidneys and spleens were fetched for H&E, TUNEL and
Ki67 staining.
2.20. Statistical analysis
The data were provided as the mean ±standard deviation. Stu-
dent’s t -test was used to calculate the difference for statistical sig-
nificance (
p < 0.05,
∗∗ p < 0.01,
∗∗∗ p < 0.001).
519
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
Fig. 1. The TEM image (a), SEM image (b), and Line STEM-EDS elemental mapping images (c) of l -Arg SDSNDs. The scale bar in (a), (b) and (c) is 50 0, 50 0 and 200 nm,
respectively.
3. Results and discussion
3.1. Preparation and characterization of L -Arg SDSNDs
The l -Arg SDSNDs are prepared through room-temperature
Mannich reaction by stirring an aqueous solution of l -Arg, EGCG
and HCHO for 1 h. As revealed by transmission electron mi-
croscopy (TEM) and scanning electron microscopy (SEM), the as-
prepared l -Arg SDSNDs appear as monodispersed spherical parti-
cles ( Fig. 1a and b ). The scanning transmission electron microscopy
with energy-dispersive X-ray spectroscopy (STEM-EDS) elemental
mapping images of l -Arg SDSNDs indicate the existence of C, O
and N elements (Fig. 1c and S1). In the Fourier-transform infrared
(FTIR) spectra, the broad peak from 30 0 0 to 3600 cm
1
is assigned
to the overlap of O-H and N-H stretching vibration, and the peaks
at 167 5 and 1213 cm
1 are attributed to C = N and C-N-C stretch-
ing vibration, which imply the guanidine group of l -Arg and the
phenol hydroxyls of EGCG are preserved in SDSNDs ( Fig. 2a ) [43] .
The appearance of C-O-C vibration peak at 1117 cm
1 proposes
the generation of ether bond, indicating the occurrence of phenol-
formaldehyde type reaction. Moreover, the ultraviolet-visible (UV–
vis) absorption spectra exhibit a slight red-shift of EGCG charac-
teristic peak to 280 nm, which may be attributed to the molecular
π- πstacking and the modification of EGCG in SDSNDs ( Fig. 2b ).
The disassembly of l -Arg SDSNDs is investigated to reveal the non-
covalent bonds in the formation of SDSNDs using dimethyl sul-
foxide (DMSO), urea, NaCl and Triton X-100 to disrupt π- πin-
teraction, hydrogen bond, electrostatic interaction and hydropho-
bic interaction, respectively [44] . As indicated in Fig. S2, the l -
Arg SDSNDs disassemble only after incubating in DMSO solution,
proving that π- πstacking is dominant in the formation of SD-
SNDs. By combining the aforementioned results and the previous
report [45] , a proposed illustration of SDSND formation is shown
in Scheme 1 .
In order to regulate the size of l -Arg SDSNDs, the molar feeding
ratio of l -Arg/HCHO/EGCG is adjusted and the hydrated diameters
of the products are measured by dynamic light scattering (DLS).
As shown in Table S1, if the amount of l -Arg is higher than HCHO,
SDSNDs are failed to prepare. Whereas the hydrated diameters of
SDSNDs increase with the increment of EGCG proportion, reflecting
the increased SDSNDs size. In this context, the π- πstacking and
the formation of SDSNDs are mainly dependent on the conjugated
framework of EGCG. The π- πstacking is favored as more EGCG
participates in the formation of SDSNDs. To achieve better cellu-
lar uptake efficiency, the SDSNDs with different l -Arg/HCHO/EGCG
molar feeding ratio are prepared (Table S1). The SDSNDs are fur-
ther labeled with FITC for flow cytometry analysis. As shown in
Fig. S3, the SDSNDs with high l -Arg proportion are favored for cel-
lular uptake, which is attributed to the homing of Arg-modified
SDSNDs for Arg-starved HepG2 cells [46] . Ethanol is also intro-
duced to regulate the size of l -Arg SDSNDs, because it can adjust
solvent polarity and therewith interfere the π- πstacking (Table
S2). And the solution containing 5% ethanol is most favored for
preparing small SDSNDs. With the consideration of the size and ef-
ficient uptake for therapeutic applications [47] , the l -Arg SDSNDs
prepared with l -Arg/HCHO/EGCG molar feeding ratio of 2/2/1 and
5 v/v% ethanol are selected for further investigations. Under this
condition, the as-prepared SDSNDs present the average diameter
of 114 . 7 ±28.5 nm, average hydrated diameter of 152.4 nm and
zeta potential of 43.9 ±5.7 mV (Figs. S4b, 2c and d).
L-Arg SDSNDs are tried to prepare in the absence of HCHO,
but no monodispersed particles are produced (Fig. S5), showing
that HCHO is indispensable in the preparation of l -Arg SDSNDs.
To explore the biosafety, the HCHO residue is detected using 2,4-
520
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
Fig. 2. The FTIR spectra (a) and UV–vis absorption spectra (b) of EGCG, l -Arg and l -Arg SDSNDs. The hydrated diameter (c) and zeta potential (d) of l -Arg SDSNDs.
dinitrophenylhydrazine (DNPH), because DNPH can be transformed
to DNPH derivative by HCHO [48] . In comparison with standard
HCHO solutions, the high performance liquid chromatograph mass
spectrometer (HPLC-MS) of 4 mg/mL l -Arg SDSNDs is far lower
than that of 1 μg/mL HCHO solution (Fig. S6). It means that the
l -Arg SDSNDs are completely safe by avoiding the potential health
risk of HCHO for further in vivo utilization [46] .
The quantitative analysis of EGCG and l -Arg contents is also in-
vestigated. EGCG content is measured according to Folin-Ciocalteu
method [46] . According to the absorbance at 750 nm, the calibra-
tion curve of EGCG is fitted ( Fig. 3a and b ). The absorbance of
200 μg/mL l -Arg SDSNDs is 0.1189. Thus, the EGCG content is cal-
culated as 143.8 μg/mL, approximately 71.9 wt% in the l -Arg SD-
SNDs. The content of N element in l -Arg SDSNDs is determined by
a CHNS/O elemental analyser, which is measured as 8.2 wt%, cor-
responding to the l -Arg content of 25.5 wt%.
3.2. Exploration of L -Arg and NO release
The l -Arg SDSNDs are incubated in phosphate buffer saline
solution (PBS, pH 7.4 ) at 37 °C to explore the possible release
of l -Arg and EGCG. All the spectra are basically same with PBS,
and no characteristic peaks of l -Arg and EGCG are observed
( Fig. 3c ). This result illustrates that l -Arg SDSNDs avoid the un-
necessary cargo release in normal physiological environment. Nev-
ertheless, l -Arg SDSNDs can produce NO under simulated TME,
which is proved both by Griess reagent method and 3-amino,4-
aminomethyl-2
,7
-difluorescein, diacetate (DAF-FM DA) method
[49] . In Griess reagent method, the absorption peak at 540 nm rep-
resents the generation of NO as l -Arg and l -Arg SDSNDs are trig-
gered with H
2
O
2
( Fig. 3d ). For DAF-FM DA method, DAF-FM DA can
react with NO and generate strong fluorescence at 515 nm. The en-
hanced fluorescence intensity with the increased l -Arg SDSND con-
centration demonstrates the effective generation of NO ( Fig. 3e ).
The quantitative analysis of NO generation is further studied by
Griess reagent method. According to the calibration curve ( Fig. 3f ),
the dependence of NO content on the concentration of l -Arg SD-
SNDs and the duration of H
2
O
2
trigger is summarized in Fig. 3 g .
The NO content indicates a positive correlation with the concen-
tration of l -Arg SDSNDs, which consists with the results in Fig. 3e .
In addition, the maximum absorbance appears after 1 h H
2
O
2
trig-
ger, probably attributing to the NO effuse along with the duration.
Besides, more NO is generated with more H
2
O
2
stimulation (Fig.
S7). These results demonstrate that the l -Arg SDSNDs can generate
NO with H
2
O
2
trigger ( Fig. 3h ) [18] . The no leakage of l -Arg and
H
2
O
2
-stimulated NO release are capable to facilitate antitumor gas
therapy.
3.3. Cellular NO generation
The NO gas antitumor of l -Arg SDSNDs is studied in vitro . First,
the physical stability of l -Arg SDSNDs is monitored. After incubat-
ing at room temperature for 1 month, the morphology, size and
hydrated diameter of SDSNDs are basically unchanged in compar-
521
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
Scheme 1. Schematic illustration of the preparation of l -Arg SDSNDs and their application in NO gas tumor therapy.
ison with the newly prepared ones, reflecting the good physical
stability (Figs. S8 and S9). Then the physiological stability of l -Arg
SDSNDs is conducted by incubating in H
2
O, PBS, Dulbecco’s mod-
ified eagle medium (DMEM) and DMEM with fetal bovine serum
(FBS) at 37 °C. After incubating for 16 days, l -Arg SDSNDs dis-
perse well in all solutions (Fig. S10), exhibiting the good stabil-
ity in physiological environment. To explore the dispersion stabil-
ity in biological media, the TEM image of SDSNDs is measured af-
ter incubation in culture medium for 16 days. The morphology and
size of SDSNDs are basically unchanged, indicating the good dis-
persion stability in biological media (Fig. S11). The aforementioned
good stability guarantees the further utilization in the biological
applications. The toxicity of l -Arg SDSNDs to mouse fibroblast cell
lines (L929) and human hepatoma cell lines (HepG2) is evaluated
through the CCK-8 method. With the increment of SDSND concen-
tration, the viability of L929 cells still keeps at a high level (Fig.
S12). While the viability of HepG2 cells reduces to approximately
50% at 200 μg/mL (Fig. S13). The selective toxicity of l -Arg SDSNDs
to cancerous HepG2 cells is attributed to two reasons. On the one
aspect, the great demand of nutrition in cancerous cell survival and
proliferation facilitates the uptake of SDSNDs via micropinocytosis
[50] . On the other aspect, the excessive H
2
O
2
in tumor cells accel-
erates the release of NO from l -Arg SDSNDs (Figs. S14 and S15).
Both of the two effects favor the selective NO damage towards tu-
mor cells.
To certify this consideration, the NO content of l -Arg SDSNDs-
treated HepG2 cells is conducted by virtue of DAF-FM DA-based
fluorescent detection. As presented in Fig. 4a , the intensity of flu-
orescent images is positively related to the concentrations of l -
Arg SDSNDs, illustrating more NO is produced. Accordingly, the
three-dimensional (3D) surface plots and relative intensity intu-
itively manifest the effective generation of NO from l -Arg SDSNDs
in HepG2 cells (Figs. 4b and S16). For further quantitative analysis,
the intracellular NO concentration of HepG2 cells after incubation
with different concentration of l -Arg SDSNDs is studied by using
Griess reagent. There is far beyond 1 μM NO generated with the in-
creasing SDSNDs concentration, which is enough to induce apopto-
sis [10] , indicating the good capability of l -Arg SDSNDs for NO de-
livery (Fig. S17). The mitochondrial membrane potential is further
assessed to evaluate NO-induced mitochondrial dysfunction using
JC-1 fluorescent probe. When mitochondrial membrane potential is
high, JC-1 aggregates in the mitochondrial matrix and produces red
fluorescence. When mitochondrial membrane potential is low, JC-1
exists as monomer and exhibits green fluorescence. In the experi-
ment, stronger green fluorescence is observed with the increment
of l -Arg SDSNDs concentration, representing the lower mitochon-
drial membrane potential (Figs. 4c and S18). This means that the
generated NO from l -Arg SDSNDs reduces the mitochondrial mem-
brane potential of HepG2 cells. The increase of intracellular NO
levels also leads to DNA damage of tumor cells. In the γ-H2AX im-
munofluorescence study, the l -Arg SDSNDs-treated HepG2 cells ex-
hibit stronger fluorescence, revealing the occurrence of DNA dam-
age ( Fig. 4d ). The relative intensity of the γ-H2AX fluorescent im-
ages is also measured by imageJ, showing the enhanced intensity
with SDSND concentrations (Fig. S19). Both the DNA damage and
the decrease of mitochondrial membrane potential are the hall-
marks of apoptosis. The control experiments are also conducted in
L929 cells. There is barely NO generation (Figs. S20 and S21), be-
cause of the low H
2
O
2
concentration in L929 cells (Fig. S15). After-
ward, there is basically no mitochondrial membrane potential dys-
function (Figs. S22 and S23) and DNA damage (Figs. S24 and S25)
after SDSNDs treatment, because of few NO generation.
522
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
Fig. 3. (a) The vis-NIR absorption spectra of EGCG at different concentrations (0 to 15 0 μg/mL) mixed with Folin-Ciocalteu’s phenol reagent. (b) The fitting calibration curve
according to the absorbance at 750 nm in (a). (c) The UV–vis absorption spectra of the supernatants of l -Arg SDSNDs incubated in PBS for different duration. (d) The UV–vis
absorption spectra of the supernatants of different solutions (H
2
O
2
, l -Arg, l -Arg SDSNDs, l -Arg + H
2
O
2 and l -Arg SDSNDs + H
2
O
2
) mixed with Griess reagent. (e) The
fluorescent spectra of the supernatants of l -Arg SDSNDs incubated with DAF-F M DA (Ex 495 nm, Em 515 nm). (f) The calibration curve of NO detection by Griess reagent.
(g) The generate d NO quantitative detection of the l -Arg SDSNDs at different concentrations
(0, 250, 500 and 1000 μg/mL) by Griess reagent. (h) Schematic illustration of
NO ge neration from l -Arg SDSNDs.
3.4. Apoptosis amplification of EGCG in L-Arg SDSNDs
To evaluate the apoptosis amplification of EGCG in l -Arg SD-
SNDs, Annexin V-FITC/PI apoptosis detection kit is employed and
detected by flow cytometer. As mentioned in the previous reports
[ 39 , 40 ], EGCG can induce apoptosis and inhibit tumor cell pro-
liferation. To clarify the function of EGCG in l -Arg SDSNDs, the
apoptosis of HepG2 cells is estimated after treating by l -Arg, EGCG
and l -Arg SDSNDs with the equivalent concentration of 0, 25, 50,
100, 150 and 200 μg/mL l -Arg SDSNDs. Compared with the control
group, there is obvious cell apoptosis in l -Arg, EGCG and l -Arg SD-
SNDs groups (Figs. S26, S27 and 5 a). Moreover, the cell apoptosis
ratio of 30.0 ±1. 0% in l -Arg SDSNDs group is much higher than
that treated with l -Arg (10.9 ±3.9%) at the equivalent concentra-
tion of 200 μg/mL l -Arg SDSNDs, which results from the apoptosis
amplification of EGCG in l -Arg SDSNDs. On the one hand, it means
that l -Arg SDSNDs remain the activity of EGCG for inducing can-
cerous cell apoptosis and the combination of NO-induced HepG2
cell apoptosis with EGCG-induced one leads to an apoptosis am-
plification. On the other hand, the l -Arg SDSNDs avoid the high
toxicity of EGCG to normal cells (Figs. S12, S27 and S28). In addi-
tion, the proliferation of HepG2 cells is estimated by treating with
l -Arg SDSNDs at different concentrations (
Fig. 5b ). The live/dead
assay demonstrates that more dead cells are observed at higher
SDSND concentrations, though many cells are still alive. To figure
out the proliferation of these alive cells, colony forming cell assay
is conducted. After reseeded and incubated for 15 days, the cell
proliferation exhibits a declining trend ( Fig. 5c ). The relative clone
formation rate in the group of 200 μg/mL is only 26.0%, which re-
veals that the proliferation of l -Arg SDSNDs-treated HepG2 cells is
greatly inhibited ( Fig. 5d ).
3.5. In vivo tumor therapy of L -Arg SDSNDs
Because of the efficacy of l -Arg SDSNDs on HepG2 cells in vitro,
in vivo investigation is further performed on the basis of HepG2
tumor model. The hemolysis assay is first carried out to evalu-
ate the biocompatibility of l -Arg SDSNDs. As shown in Fig. S29,
l -Arg SDSNDs do not destroy red blood cells even the concentra-
tion is up to 500 μg/mL, exhibiting the potential for in vivo utiliza-
tion. To analyze the pharmacokinetics, indocyanine green (ICG) is
labeled on l -Arg SDSNDs via the π- πstacking with EGCG frame-
work to produce ICG- l -Arg SDSNDs according to the red-shift of
the ICG absorption peak at 217 nm in Fig. S30 and the previous re-
523
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
Fig. 4. The CLSM images (a) and 3D surface plots (b) of l -Arg SDSNDs (0, 50, 100 and 200 μg/mL) treated HepG2 cells after incubating with DAF-FM DA . The CLSM images
of l -Arg SDSNDs (0, 50, 10 0 and 200 μg/mL) treated HepG2 cells after incubating with JC-1 (c) and γ-H2AX antibody (d). The scale bar in (a), (c) and (d) is 100 , 50 and
100 μm, respectively.
port
[51] . The appearance of the typical absorption at 795 nm (Fig.
S30) and the photoluminescent emission at 805 nm (Fig. S31) con-
firms the successful labeling of ICG on l -Arg SDSNDs. As indicated
in Figs. S32 and 6 a, the ICG- l -Arg SDSNDs exhibit good fluores-
cence stability, which guarantees the pharmacokinetics studies in
vivo . The half-life of SDSNDs in blood circulation is calculated to
be 8.3 ±1.0 h ( Fig. 6b ) according to the blood fluorescence inten-
sity of the mice after injecting with ICG- l -Arg SDSNDs (Fig. S33),
which is much higher than that of l -Arg (80 ±9 min) and EGCG
(62 ±11 min), showing the improved bioavailability [ 52 , 53 ]. The
distribution and metabolism of ICG- l -Arg SDSNDs in various or-
gans are also evaluated through the quantitative analysis of fluo-
rescence intensity of various organs over time. In the first 12 h,
the SDSNDs mainly exist in liver, lungs, kidneys, spleen and tu-
mor. Subsequently, the SDSNDs content in major organs decreases
over time, while the retention in the tumors can keep for 5 days,
which implies the good metabolism and potential long-time treat-
ment of the tumors ( Fig. 6c -e). To explore the excretion, after be-
ing administrated with ICG- l -Arg SDSNDs, the feces and urine of
mice are collected for fluorescence imaging. Both of them show
strong fluorescent signals at 4, 8 and 12 h ( Fig. 6f ), which indi-
cates that the SDSNDs can excrete via renal excretion and biliary
excretion.
The good metabolization and biodistribution of l -Arg SDSNDs in
tumor tissues intimate the viability for in vivo antitumor. The ani-
mal experiment is conducted by pre-building subcutaneous HepG2
tumor models in advance of 12 days ( Fig. 7a ). The mice bearing
tumor are randomly divided into four groups, namely Control, l -
524
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
Fig. 5. (a) The flow cytometry of l -Arg SDSNDs treated HepG2 cells by using Annexin V-FITC/PI apoptosis detection kit. (b) The CLSM images of l -Arg SDSNDs (0, 50, 100
and 20 0 μg/mL) treated HepG2 cells after incubating with Calcein/PI cell viability/cytotoxicity assay kit. The scale bar is 200 μm. (c) The images of CFC assay. (d) The relative
clone formation rate according to (c).
525
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
Fig. 6. (a) The quantitative fluorescent intensity analysis of ICG- l -Arg SDSNDs incubated in culture medium for 5 days in Fig. S32. (b) The half-life of l -Arg SDSNDs. (c) The
fluorescent images of mice after treated with ICG- l -Arg SDSNDs before (Control) and after 4, 8, 12, 24, 48, 72 and 120 h. (d) The fluorescent images of major organs (heart,
liver, spleen, lungs and kidneys) and tumor of the mice after treated with ICG- l -Arg SDSNDs. (e) The biodistribution of l -Arg SDSNDs in major organs and tumors according
to Fig. 6d. (f) The fluorescent images
of the feces and urine of the mice after treated with ICG- l -Arg SDSNDs. The scale bar is used for 6c, 6d and 6f.
Arg, EGCG and l - Arg SDSNDs. From 0 to 16 days, the mice in each
group are administrated with corresponding drug solutions, re-
spectively, every two days. At the same time, the body weight and
tumor volume of mice in all groups are measured and recorded. On
the 16t h day, the mice are captured and euthanatized. As shown
in Fig. 7b , the relative tumor volume of l -Arg SDSNDs group is
far smaller than other groups, directly manifesting the good an-
titumor effect of l -Arg SDSNDs. The average tumor weight shows
the same trend, according to which the tumor growth inhibition
(TGI) is calculated ( Fig. 7c and d ). The TGI of l -Arg SDSNDs is up
to 91.4 ±6.5%, much higher than those of other groups, which ex-
hibits the superiority of SDSNDs by combining the antitumor ef-
fect of l -Arg and EGCG. The images of mice bearing tumors and
dissected tumors are shown in Fig. 7e . Furthermore, the slices of
the tumors are stained by H&E, TUNEL and Ki67 to estimate the
state of the tumor cells ( Figs. 7 f, S34 and S35). The l -Arg SDSNDs
therapy group induces highest apoptosis and lowest proliferation
in tumor tissues, suggesting l -Arg SDSNDs possess good antitumor
effect. Note that although l -Arg is the source of NO and EGCG can
induce apoptosis, the therapeutic effect of l -Arg group and EGCG
group is very limited in animal experiments owing to two reasons.
First, the half-lives of free l -Arg and EGCG in blood circulation are
much lower than that of SDSNDs ( Fig. 6b ), and the quick renal
clearance results in the lower bioavailability. Second, the combi-
nation of NO-induced cell apoptosis with EGCG-induced one by l -
Arg SDSNDs leads to the apoptosis amplification ( Fig. 5a ). With re-
spect to biosafety, the relative body weight of mice in all groups is
not significantly different and shows steadily increase from 0 to 16
days (Fig. S36). On the 16th day, the blood of mice is obtained for
major liver and renal function tests. All indexes of each group are
normal (Fig. S37). The state of major organs, including heart, liver,
lungs, kidneys and spleen, are also investigated by H&E staining,
showing no obvious side effect in all groups (Fig. S38). All these
results confirm the good biosafety of l -Arg SDSNDs.
526
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
Fig. 7. (a) The Schematic illustration of animal experiment. The records of relative tumor volume (b) and average tumor weig ht (c) of the mice in each group (Control, l -Arg,
EGCG and l -Arg SDSNDs). (d) The TGI rate according to (c). (e) The images of mice bearing tumor and tumors dissected in each group on 16 d (f) The images of H&E stained
tumor tissue slices of the mice in each group. The scale bar in (f) is 20 μm.
4. Conclusion
In summary, carrier- and surfactant-free SDSNDs are prepared
by combining the Mannich reaction of l -Arg, EGCG and HCHO and
the assembly of the products via molecular π- πstacking. The for-
mation of l -Arg SDSNDs not only improves the half-life of l -Arg,
but also avoids the leakage of l -Arg in physiological environment.
With the stimulation of excessive H
2
O
2
in TME, the retained guani-
dine group of l -Arg can release NO for gas antitumor by provok-
ing tumor cell mitochondrial/DNA damage and apoptosis. Mean-
while, the combination of the preserved phenolic hydroxyl groups
of EGCG with NO leads to tumor cell apoptosis amplification and
proliferation inhibition, exhibiting good efficacy in HepG2 tumor
models. This work provides “no loss and NO release” SDSNDs,
which is an alternative for further designing innovative tumor ther-
apeutic systems with NO-associated synergism and cascade.
Declaration of Competing Interest
The authors declare no conflict of interest.
Acknowledgements
This work was supported by the Science and Technology Devel-
opment Program of Jilin Province (YDZJ202201ZYTS055), the Sci-
527
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
ence and Technology Project of Jilin Provincial Department of Ed-
ucation (JJKH20221079KJ), the Project funded by China Postdoc-
toral Science Foundation (2021TQ0125, 2022M711303), the Inter-
disciplinary Integration and Innovation Project of Jilin University
(JLUXKJC2021QZ10), and Special Project from MOST of China.
Supplementary materials
Supplementary material associated with this article can be
found, in the online version, at doi:10.1016/j.actbio.2023.07.055 .
References
[1] W. Fan , B.C. Yung , X. Chen, Stimuli-responsive NO release for on-demand
gas-sensitized synergistic cancer therapy, Angew. Chem. Int. Ed. 57 (2018)
8383–8394
.
[2] W. Zhang, Q. Xuan, Q. Zhang, T. Wa ng, C. Wang, H. Li, C. Chen, P. Wan g,
Near-infrared light switching nitric oxide nanogenerator with “linkage mecha-
nism” for tumor targeting multimodal synergistic therapy, Sci. China Chem. 66
(2022) 586–60 0
.
[3] S. Wan, J. Zeng, H. Cheng, X. Zhang, ROS-induced NO generation for gas ther-
apy and sensitizing photodynamic therapy of tumor, Biomaterials 18 5 (2018)
51–62
.
[4] F. Rong, T. Wang, Q. Zhou, H. Peng, J. Yan g, Q. Fan, P. Li, Intelligent polymeric
hydrogen sulfide delivery systems for therapeutic applications, Bioact. Mater.
19 (2023) 198–216
.
[5] Y. Li, N. Lu, Q. Lin, H. Wang, Z. Liang, Y. Lu, P. Zhang, Sono-ReCORMs for syner-
getic sonodynamic-gas therapy of hyp oxic tumor, Chin. Chem. Lett. 34 (2023)
107653
.
[6] X. Ya ng , N. Zhang, G. Li, M. Zhang, C. Pang, S. Re n, H. An, Polyphenol-mediated
biomimetic MOFs hybrid nanoplatform for catalytic cascades-enhanced cancer
targeted combination therapy, Mater. Des. 223 (2022) 111 217
.
[7] C.R. Powell, K.M. Dillon, J.B. Matson, A review of hydrogen sulfide (H
2
S)
donors: chemi stry and potential therapeutic applications, Biochem. Pharmacol.
149 (2018) 110 12 3
.
[8] C. Farah, L.Y.M. Michel, J.L. Balligand, Nitric oxide signalling in cardiovascular
health and disease, Nat. Rev. Cardiol. 15 (2018) 292–316
.
[9] Y. Lin, W. Zhong, M. Wang , Z. Chen, C. Lu, H. Ya ng , Multifunctional car-
bon monoxide prodrug-loaded nanoplatforms for effective photoacoustic imag-
ing-guided photothermal/gas synergistic therapy, ACS Appl. Bio Mater 4 (2021)
4557–4564
.
[10] Z. Zhou, Z. Gao, W. Chen, X. Wang, Z. Chen, Z. Zheng, Q. Chen, M. Tan, D. Liu,
Y. Zhang, Z. Hou, Nitric oxide-mediated regulation of mitochondrial protective
autophagy for enhanced chemodynamic therapy based on mesoporous Mo–
doped Cu
9
S
5
nanozymes, Ac ta Biomater. 151 (2022) 600–612
.
[11] W. Yu, T. Liu, M. Zhang, Z. Wang, J. Ye, C.X. Li, W. Liu, R. Li, J. Feng, X.Z. Zhang,
O
2 economizer for inhibiting cell respiration to combat the hypoxi a obstacle
in tumor treatments, ACS Nano 13 (2019) 1784–1794
.
[12] J. An, Y.G. Hu, C. Li, X.L. Hou, K. Cheng, B. Zhang, R.Y. Zhang, D.Y. Li, S.J. Liu,
B. Liu, D. Zhu, Y. D. Zhao, A pH/Ultrasound dual-response biomimetic nanoplat-
form for nitric oxide gas-sonodynamic combined therapy and repeated ultra-
sound for rel ieving hypoxia, Biomaterials 230 (2020) 119636
.
[13] S.Y. Chen, J. Wang, F. Jia, Z.D. Shen, W.B . Zhang, Y. X. Wa ng, K.F. Ren, G.S. Fu, J. Ji,
Bioinspired NO release coating enhances endothelial cells and inhibits smooth
muscle cells, J. Mater. Chem. B 10 (2022) 2454–2462
.
[14] Y. Huang, J. Huang, M. Jiang, S. Zeng, NIR-triggered theranostic Bi
2
S
3 light
transducer for on-demand NO release and synergistic gas/photothermal com-
bination therapy of tumors, ACS Appl. Bio Mater. 2 (2019) 4769–4776
.
[15] M. Sun, Y. Sang, Q. Deng, Z. Liu, J. Ren, X. Qu, Specific generation of nitric oxide
in mitochondria of cancer cell for selective oncotherapy, Na no Res. 15 (2022)
5273–5278
.
[16] C. Han, Q. Yu, J. Jiang, X. Zhang, F. Wang, M. Jiang, R. Yu, T. Deng, C. Yu, Bioen-
zyme-responsive l -arginine-based carbon dots: the replenishment of nitric ox-
ide for nonpharmaceutical therapy, Biomater. Sci. 9 (2021) 7432–7443
.
[17] Z. Yuan, C. Lin, Y. He, B. Tao, M. Chen, J. Zhang, P. Liu, K. Cai, Near-infrared
light-triggered nitric-oxide-enhanced photodynamic therapy and low-temper-
ature photothermal therapy for biofilm elimination, ACS Nano 14 (2020)
3546–3562
.
[18] W. Fan, N. Lu, P. Huang, Y. Liu, Z. Yang, S. Wang, G. Yu, Y. Liu, J. Hu, Q. He, J. Qu,
T. Wang , X. Chen, Glucose-responsive sequential generation of hydrogen per-
oxide and nitric oxide for synergistic cancer starving-like/gas therapy, Angew.
Chem. Int. Ed. 56 (2017) 1229–1233
.
[19] F. Gong, N. Yan g, X. Wa ng, Q. Zhao, Q. Chen, Z. Liu, L. Cheng, Tumor microen-
vironment-responsive intelligent nanoplatforms for cancer theranostics, Nano
Tod ay 32 (2020) 1008 51
.
[20] R. Jin, Z. Liu, T. Liu, P. Yuan, Y. Bai, X. Chen, Redox-responsive micelles inte-
grating catalytic nanomedicine and selective chemotherapy for effective tumor
treatment, Chin. Chem. Lett. 32 (2021) 3076–3082
.
[21] T. Li, Y. Zhang, J. Zhu, F. Zhang, A. Xu, T. Zhou, Y. Li, M. Liu, H. Ke, T. Yang ,
Y. Tan g, J. Tao, L. Miao, Y. Deng, H. Chen, pH-activatable copper-biomineral-
ized proenzyme for synergistic chemodynamic/chemo-immunotherapy against
aggressive cancers, Adv. Mater. (2022) 2210201
.
[22] H. Lin, Y. Chen, J. Shi, Nanoparticle-triggered in situ catalytic chemical reactions
for tumour-specific therapy, Chem. Soc. Rev. 47 (2018) 1938–1958
.
[23] Q. Zhang, G. Kuang, W. Li, J. Wang, H. Ren, Y. Zhao, Stimuli-responsive gene
delivery nanocarriers for cancer therapy, Nano-Micro Lett 15 (2023) 44
.
[24] H. Chen, Z. Gu, H. An, C. Chen, J. Chen, R. Cui, S. Chen, W. Chen, X. Chen,
X. Chen, Z. Chen, B. Ding, Q. Dong, Q. Fan, T. Fu, D. Hou, Q. Jiang, H. Ke , X. Jiang,
G. Liu, S. Li, T. Li, Z. Liu, G. Nie, M. Ovais, D. Pang, N. Qiu, Y. Shen, H. Tian,
C. Wa ng , H. Wang , Z. Wan g, H. Xu, J.F. Xu, X. Yang, S. Zhu, X. Zheng, X. Zhang,
Y. Zhao, W. Ta n, X. Zhang, Y. Zhao, Precise nanomedicine for intelligent therapy
of cancer, Sci. China Chem. 61 (2018) 1503–1552
.
[25] J. Ding, J. Chen, L. Gao, Z. Jiang, Y. Zhang, M. Li, Q. Xiao, S.S. Lee, X. Chen,
Engineered nanomedicines with enhanced tumor penetration, Nano Today 29
(2019) 10 080 0
.
[26] X. Guo, X. Wei, Z. Chen, X. Zhang, G. Yang, S. Zhou, Multifunctional nanoplat-
forms for subcellular delivery of drugs in cancer therapy, Prog. Mater Sci. 107
(2020) 100599
.
[27] H. Li, J. Yan, D. Meng, R. Cai, X. Gao, Y. Ji, L. Wa ng , C. Chen, X. Wu, Gold
nanorod-based nanoplatform catalyzes constant NO generation and protects
from cardiovascular injury, ACS Nano 14 (2020) 12854–12865
.
[28] T. Gu, T. Chen, L. Cheng, X. Li, G. Han, Z. Liu, Mesoporous silica decorated
with platinum nanoparticles for drug delivery and synergistic electrodynam-
ic-chemotherapy, Nano Res. 13 (2020) 2209–2215
.
[29] W. Chen, S. Zhou, L. Ge, W. Wu, X. Jiang, Translatable high drug loading drug
delivery systems based on biocompatible polymer nanocarriers, Biomacro-
molecules 19 (2018) 1732–1745
.
[30] Y. Feng, H. Zhang, X. Xie, Y. Chen, G. Ya ng, X. We i, N. Li, M. Li, T. Li, X. Qin,
S. Li, F. You, C. Wu, H. Yan g, Y. Liu, Cascade-activatable NO release based on
GSH-detonated "nanobomb" for multi-pathways cancer therapy, Mater. To da y
Bio 14 (2022) 100288 .
[31] K. Wa ng, L. Jiang, L. Qiu, Near infrared light triggered ternary synergistic can-
cer therapy via l -arginine-loaded nanovesicles with modification of PEGylated
indocyanine green, Acta Biomater. 140 (2022) 506–517
.
[32] M. Wang , Z. Hou, S. Liu, S. Liang, B. Ding, Y. Zhao, M. Chang, G. Han,
A .A .A . Kheraif, J. Lin, A multifunctional nanovaccine based on l -arginine-loaded
black mesoporous titania: ultrasound-triggered synergistic cancer sonody-
namic therapy/gas therapy/immunotherapy with remarkably enhanced effi-
cacy, Small 17 (2021) 2005728
.
[33] F. Gao, B. Yu, H. Cong, Y. Shen, Delivery process and effective design of vectors
for cancer therapy, J. Mater. Chem. B 10 (2022) 6896–6921
.
[34] B. Lu, E. Hu, R. Xie, K. Yu, F. Lu, R. Bao, C. Wang, G. Lan, F. Dai, Magnetically
guided nanoworms for precise delivery to enhance in situ production of nitric
oxide to combat focal bacterial infection in vivo , ACS Appl. Bio Mater. 13 (2021)
22225–22239
.
[35] Z. Yi, G. Chen, X. Chen, Z. Sun, X. Ma, W. Su, Z. Deng, L. Ma, Y. Ran, Q. Tong,
X. Li, Modular assembly of versatile nanoparticles with epigallocatechin gal-
late, AC S Sustain. Chem. Eng 8 (2020) 9833–9845
.
[36] Y. Ya ng , W. Zhu, L. Cheng, R. Cai, X. Yi, J. He, X. Pan, L. Yan g, K. Yan g,
Z. Liu, W. Tan, M. Chen, Tumor microenvironment (TME)-activatable circular
aptamer-PEG as an effective hierarchical-targeting molecular medicine for pho-
todynamic therapy, Biomaterials 246 (2020) 1199 71 .
[37] L. Tang , X. Chen, Q. Ton g, Y. Ran, L. Ma, Y. Tan, Z. Yi, X. Li, Biocompatible,
bacteria-targeting resveratrol nanoparticles fabricated by Mannich molecular
condensation for accelerating infected wound healing, J. Mater. Chem. B 10
(2022) 9280–9294
.
[38] Y. Hou, Y. Kuang, Q. Jiang, S. Zhou, J. Yu, Z. He, J. Sun, Arginine-peptide com-
plex-based assemblies to combat tumor hypoxi a for enhanced photodynamic
therapeutic effect, Nano Re s. 15 (2022) 5183–5192
.
[39] K. Li, G. Xiao, J.J. Richardson, B.L. Ta rd y, H. Ejima, W. Huang, J. Guo, X. Liao,
B. Shi, Targ ete d therapy against metastatic melanoma based on self-assem-
bled metal-phenolic nanocomplexes comprised of green tea catechin, Adv. Sci.
6 (2019) 18016 8 8
.
[40] L. Shan, G. Gao, W. Wang, W. Tang, Z. Wan g, Z. Yang, W. Fan, G. Zhu, K. Zhai,
O. Jacobson, Y. Dai, X. Chen, Self-assembled green tea polyphenol-based co-
ordination nanomaterials to improve chemotherapy efficacy by inhibition of
carbonyl reductase 1, Biomaterials 210 (2019) 62–69
.
[41] C. Wang , H. Sang, Y. Wang, F. Zhu, X. Hu, X. Wan g, X. Wang, Y. Li, Y. Cheng,
Foe to friend: supramolecular nanomedicines consisting of natural polyphenols
and bortezomib, Nano Lett. 18 (2018) 7045–7051
.
[42] B. Zhang, Y. Qin, L. Ya ng , Y. Wu, N. Chen, M. Li, Y. Li, H. Wan, D. Fu,
R. Luo, L. Yuan, Y. Wang, A polyphenol-network-mediated coating modulates
inflammation and vascular healing on vascular stents, ACS Nano 16 (2022)
6585–6597
.
[43] X. Liu, Y. Liu, A.S. Thakor, B.D. Kevadiya, J. Cheng, M. Chen, Y. Li, Q. Xu, Q. Wu,
Y. Wu, G. Zhang, Endogenous NO-releasing carbon nanodots for tumor-specific
gas therapy, Acta Biomater. 136 (2021) 4 85–4 94
.
[44] Z. Lin, J. Zhou, Y. Qu, S. Pan, Y. Han, R.P.M. Lafleur, J. Chen, C. Cortez-Jugo,
J.J. Richardson, F. Caruso, Luminescent metal-phenolic networks for multicolor
particle labeling, Angew. Chem. Int. Ed. 60 (2021) 24 96 8–24 975
.
[45] Z. Yi, G. Chen, X. Chen, X. Ma, X. Cui, Z. Sun, W. Su, X. Li, Preparation of
strong antioxidative, therapeutic nanoparticles based on amino acid-induced
ultrafast assembly of te a polyphenols, ACS Appl. Mater. Interfaces 12 (2020)
33550–33563
.
[46] S. Wang, F. Li, R. Qiao, X. Hu, H. Liao, L. Chen, J. Wu, H. Wu, M. Zhao, J. Liu,
R. Chen, X. Ma, D. Kim, J. Sun, T.P. Davis, C. Chen, J. Tian, T. Hyeon, D. Ling,
Arginine-rich manganese silicate nanobubbles as a ferroptosis-inducing agent
for tumor-targeted theranostics, ACS Nan o 12 (2018) 12380–12392 .
528
M. Zhang, H. Jin, Y. Liu et al. Acta Biomaterialia 169 (2023) 517–529
[47] S. Li, X. Feng, J. Wang , W. Xu, M.A. Islam, T. Sun, Z. Xie, C. Wan g, J. Ding,
X. Chen, Multiantigenic nanoformulations activate anticancer immunity de-
pending on size, Adv. Funct. Mater. 29 (2019) 1903391
.
[48] I.M. Melo Cardozo, E.T. Sousa, G.O. da Rocha, J. Pereira Dos Anjos, J.B. de An-
drade, Determination of free- and bound-carbonyl compounds in airborne par-
ticles by ultra-fast liquid chromatography coupled to mass spectrometry, Ta-
lanta 217 (2020) 12103 3
.
[49] W. Liu, F. Semcheddine, Z. Guo, H. Jiang, X. Wang, Glucose-responsive ZIF-8
nanocomposites for targeted cancer therapy through combining starvation
with stimulus-responsive nitric oxide synergistic treatment, ACS Appl. Bio
Mater. 5 (2022) 2902–2912
.
[50] C. Commisso, S.M. Davidson, R.G. Soydaner-Azeloglu, S.J. Parker, J.J. Kamphorst,
S. Hackett, E. Grabocka, M. Nofal , J.A. Drebin, C.B. Thompson, J.D. Rabinowitz,
C.M. Metallo, M.G. Vander Heiden, D. Bar-Sagi, Macropinocytosis of protein
is an amino acid supply route in Ras-transformed cells, Nature 497 (2013)
633–637
.
[51] D. Hu, C. Liu, L. Song, H. Cui, G. Gao, P. Liu, Z. Sheng, L. Cai, Indocyanine
green–loaded polydopamine–iron ions coordination nanoparticles for photoa-
coustic/magnetic resonance dual-modal imaging-guided cancer photothermal
therapy, Nanoscale 8 (2016) 17 15 0
.
[52] S.M. Bode-Boger, R.H. Boger, A. Galland, D. Tsikas, J.C. Frolich,
L -arginine-induced vasodilation in healthy humans: pharmacokinetic–phar-
macodynamic relationship, Br. J. Clin. Pharmacol. 46 (1998) 4 89–4 97
.
[53] L. Lin, M. Wang , T. Tseng, J. Sung, T. Tsai, Pharmacokinetics of
( )-epigallocatechin-3-gallate in conscious and freely moving rats and its
brain regional distribution, J. Agric. Food Chem. 55 (2007) 1517–1524
.
529
... Figure 5d showed the NO release profiles of the L-Arg@Yb-DOBDC and control groups incubated in PBS solutions after 48 hours of incubation, both L-Arg and L-Arg@Yb-DOBDC exhibited absorption peaks for NO at 540 nm due to H 2 O 2 excitation, and the L-Arg@Yb-DOBDC group was slightly lower than that of L-Arg. The total NO release amount of L-Arg@Yb-DOBDC was 1.271 μM, achieving a relatively high NO concentration (>1 μM), [26] which possessed the potential to induce apoptosis in tumor cells. Adjusting the concentration of H 2 O 2 revealed www.advancedsciencenews.com www.advopticalmat.de ...
Article
Full-text available
Exogenous supplementation of L‐arginine (L‐Arg) facilitates the release of nitric oxide (NO) for gas therapy applications in tumor microenvironments rich in H2O2. However, direct administration inevitably leads to the premature release of hydrophilic L‐Arg in the bloodstream, potentially resulting in unstable plasma L‐Arg concentrations and subsequent adverse reactions. Therefore, it is crucial to identify efficient and stable high‐dose L‐Arg delivery carriers, albeit challenging. Here, the study has achieved high (≈94 mg g⁻¹) and stable L‐Arg loading within a 12‐coordinated Yb‐based lanthanide metal‐organic framework (named Yb‐DOBDC, where DOBDC is 2,5‐dihydroxyterephthalic acid). Yb‐DOBDC features size‐matched apertures and oxygen‐rich pore microenvironments, securely entrapping L‐Arg through electrostatic interactions and conjugation effects, thereby enhancing the biostability of the drug during delivery. In vitro experiments demonstrate that L‐Arg‐loaded Yb‐DOBDC (L‐Arg@Yb‐DOBDC) promotes NO release upon H2O2 stimulation, and the Yb‐DOBDC framework exhibits good biocompatibility within cells. More interestingly, the near‐infrared‐luminescence‐emitting Yb‐DOBDC exhibits a “turn‐off” behavior upon gradual loading of L‐Arg, enabling self‐quantifying luminescence detection during the encapsulation process. Consequently, the L‐Arg@Yb‐DOBDC system designed in this work, with its high/stable loading capacity and self‐quantifying luminescence detection capability, holds theoretical research value and potential practical application prospects.
Article
Full-text available
Chemotherapy is crucial in oncology for combating malignant tumors but often encounters obatacles such as severe adverse effects, drug resistance, and biocompatibility issues. The advantages of degradable silica nanoparticles in tumor diagnosis and treatment lie in their ability to target drug delivery, minimizing toxicity to normal tissues while enhancing therapeutic efficacy. Moreover, their responsiveness to both endogenous and exogenous stimuli opens up new possibilities for integrating multiple treatment modalities. This review scrutinizes the burgeoning utility of degradable silica nanoparticles in combination with chemotherapy and other treatment modalities. Commencing the elucidation of degradable silica synthesis and degradation mechanisms, emphasis is placed on the responsiveness of these materials to endogenous (e.g., pH, redox reactions, hypoxia, and enzymes) and exogenous stimuli (e.g., light and high‐intensity focused ultrasound). Moreover, this exploration delves into strategies harnessing degradable silica nanoparticles in chemotherapy alone, coupled with radiotherapy, photothermal therapy, photodynamic therapy, gas therapy, immunotherapy, starvation therapy, and chemodynamic therapy, elucidating multimodal synergies. Concluding with an assessment of advances, challenges, and constraints in oncology, despite hurdles, future investigations are anticipated to augment the role of degradable silica in cancer therapy. These insights can serve as a compass for devising more efficacious combined tumor treatment strategies.
Article
Full-text available
Oxaliplatin (Oxa) is a commonly used chemotherapy drug in the treatment of gastric cancer, but its toxic side effects and drug resistance after long-term use have seriously limited its efficacy. Loading chemotherapy drugs with nanomaterials and delivering them to the tumor site are common ways to overcome the above problems. However, nanomaterials as carriers do not have therapeutic functions on their own, and the effect of single chemotherapy is relatively limited, so there is still room for progress in related research. Herein, we construct Oxa@Mil-100(Fe) nanocomposites by loading Oxa with a metal–organic framework (MOF) Mil-100(Fe) with high biocompatibility and a large specific surface area. The pore structure of Mil-100(Fe) is conducive to a large amount of Oxa loading with a drug-loading rate of up to 27.2%. Oxa@Mil-100(Fe) is responsive to the tumor microenvironment (TME) and can release Oxa and Fe³⁺ under external stimulation. On the one hand, Oxa can inhibit the synthesis of DNA and induce the apoptosis of gastric cancer cells. On the other hand, Fe³⁺ can clear overexpressed glutathione (GSH) in TME and be reduced to Fe²⁺, inhibiting the activity of glutathione peroxidase 4 (GPX4), leading to the accumulation of intracellular lipid peroxides (LPO), and at the same time releasing a large number of reactive oxygen species (ROS) through the Fenton reaction, inducing ferroptosis in gastric cancer cells. With the combination of apoptosis and ferroptosis, Oxa@Mil-100(Fe) shows a good therapeutic effect, and the killing effect on gastric cancer cells is obvious. In a nude mouse model of subcutaneous tumor transplantation, Oxa@Mil-100(Fe) shows a significant inhibitory effect on tumor growth, with an inhibition rate of nearly 60%. In addition to its excellent antitumor activity, Oxa@Mil-100(Fe) has no obvious toxic or side effects. This study provides a new idea and method for the combined treatment of gastric cancer.
Article
Full-text available
Tumor hypoxia diminishes the effectiveness of traditional type II photodynamic therapy (PDT) due to oxygen consumption. Type I PDT, which can operate independently of oxygen, is a viable option for treating hypoxic tumors. In this study, we have designed and synthesized JSK@PEG-IR820 NPs that are responsive to the tumor microenvironment (TME) to enhance type I PDT through glutathione (GSH) depletion. Our approach aims to expand the sources of therapeutic benefits by promoting the generation of superoxide radicals (O2−.) while minimizing their consumption. The diisopropyl group within PEG-IR820 serves a dual purpose: it functions as a pH sensor for the disassembly of the NPs to release JSK and enhances intermolecular electron transfer to IR820, facilitating efficient O2−. generation. Simultaneously, the release of JSK leads to GSH depletion, resulting in the generation of nitric oxide (NO). This, in turn, contributes to the formation of highly cytotoxic peroxynitrite (ONOO−.), thereby enhancing the therapeutic efficacy of these NPs. NIR-II fluorescence imaging guided therapy has achieved successful tumor eradication with the assistance of laser therapy.
Article
Full-text available
Gene therapy provides a promising approach in treating cancers with high efficacy and selectivity and few adverse effects. Currently, the development of functional vectors with safety and effectiveness is the intense focus for improving the delivery of nucleic acid drugs for gene therapy. For this purpose, stimuli-responsive nanocarriers displayed strong potential in improving the overall efficiencies of gene therapy and reducing adverse effects via effective protection, prolonged blood circulation, specific tumor accumulation, and controlled release profile of nucleic acid drugs. Besides, synergistic therapy could be achieved when combined with other therapeutic regimens. This review summarizes recent advances in various stimuli-responsive nanocarriers for gene delivery. Particularly, the nanocarriers responding to endogenous stimuli including pH, reactive oxygen species, glutathione, and enzyme, etc., and exogenous stimuli including light, thermo, ultrasound, magnetic field, etc., are introduced. Finally, the future challenges and prospects of stimuli-responsive gene delivery nanocarriers toward potential clinical translation are well discussed. The major objective of this review is to present the biomedical potential of stimuli-responsive gene delivery nanocarriers for cancer therapy and provide guidance for developing novel nanoplatforms that are clinically applicable.
Article
Full-text available
Artificial enzymes have demonstrated therapeutic benefit against diverse malignant tumors, yet their antitumor potencies are still severely compromised by non‐selective catalysis, low atomic utilization efficiency, and undesired off‐target toxicity. Herein, we report that the peroxidase‐like biomineralized copper (II) carbonate hydroxide nanocrystals inside single albumin nanocages (CuCH‐NCs) act as a pH‐activatable proenzyme to achieve tumor‐selective and synergistic chemodynamic/chemo‐immunotherapy against aggressive triple‐negative breast cancers (TNBCs). These CuCH‐NCs show pH‐sensitive Cu2+ release, which spontaneously undergoes glutathione (GSH)‐mediated reduction into Cu+ species for catalyzing the evolution of H2O2 into hydroxyl radicals in a single‐atom‐like manner to cause chemodynamic cell injury, and simultaneously activates non‐toxic disulfiram to cytotoxic complex for yielding selective chemotherapeutic damage via blocking cell proliferation and amplifying cell apoptosis. CuCH‐NCs exhibit considerable tumor‐targeting capacity with deep penetration depth, thus affording preferable efficacy against orthotopic breast tumors through synergistic chemodynamic/chemotherapy, together with good in vivo safety. Moreover, CuCH‐NCs arouse distinct immunogenic cell death effect and upregulate PD‐L1 expression upon disulfiram combination, and thus synergize with anti‐PD‐L1 antibody to activate adaptive and innate immunities, together with relieved immunosuppression, finally yielding potent antitumor efficacy against both primary and metastatic TNBCs. These results provide insights into smart and high‐performance proenzymes for synergistic therapy against aggressive cancers. This article is protected by copyright. All rights reserved
Article
Full-text available
Therapeutic approaches of combining conventional photodynamic therapy (PDT) with other adjuvant treatments to sensitize PDT represent an appealing strategy. Herein, a novel synergetic “nanobomb” strategy based on glutathione (GSH)-responsive biodegradation was proposed to effectively destroy tumors expeditiously and accurately. This “nanobomb” was rationally constructed via the simultaneous encapsulation of methylene blue (MB) and l-arginine (L-Arg) into polyethylene glycol (PEG) modified mesoporous organosilicon nanoparticles (MON). The resulting L-Arg/[email protected] initially exhibited prolonged blood circulation, improved bioavailability, and enhanced tumor accumulation in mice after tail vein injection according to the pharmacokinetic investigations, before the nanoparticles were entirely excreted. Under laser irradiation, L-Arg/[email protected] produced remarkable reactive oxygen species (ROS) directly for PDT therapy, while a portion of ROS may oxidize L-Arg to generate nitric oxide (NO) not only for gas therapy (GT) but also serve as a biological messenger to regulate vasodilation to alleviate the tumor hypoxia. Subsequently, the rapidly released NO was further oxidized to reactive nitrogen species, which together with ROS promote immunogenic cell death by inducing G2/M cell-cycle arrest and apoptosis in cancer cells, and eventually resulting in enhanced anti-tumor immune responses. Moreover, the GSH depletion in tumor tissues induced by L-Arg/[email protected] biodegradation can cooperate with GT to amplify the therapeutic effect of PDT. These results demonstrate that this “nanobomb” provides new ideas for clinical translation to treat tumor patients in terms of synergistic PDT-GT nanotherapy in hypoxic-solid tumors.
Article
Gaseous therapy based on nitric oxide (NO), as a potential anti-tumor treatment strategy, has attracted great attention, but the targeted and controlled gas release in the tumor site still remains a challenge. In addressing these difficulties, a near-infrared (NIR) light-triggered NO release nanogenerator with a “linkage mechanism” was designed on the basis of sodium nitroprusside-doped mesoporous Prussian blue nanoparticles, in which the outer structure was modified with pH-sensitive gatekeeper chitosan and tumor-targeting agent folic acid. The “linkage mechanism” can achieve precise release of NO under the control of photothermal effect at tumor site, which can couple photothermal therapy and gas therapy to address the premature release of gas during transportation. Meanwhile, the amount of released gas can be controlled by adjusting the irradiation time and laser intensity. Furthermore, as-fabricated nanocomposites hold high photothermal conversion efficiency under NIR laser irradiation, resulting in the on-demand release of NO and chemotherapy drugs. The released NO can inhibit the expression of hypoxia-inducible factor α (HIF-1α) and alleviate the hypoxic tumor microenvironment, thereby enhancing the efficacy of chemotherapy. Moreover, in vitro and in vivo experiments exhibited remarkable antitumor efficiency, and the synergistic gas/chemo/photothermal therapy of deep tumors was achieved. These findings indicate an effective strategy to stimulate further the development of deep tumor therapy, which may provide new insights into other NO-related medical applications.
Article
Excessive reactive oxygen species (ROS) and long-term inflammation can delay wound healing and cause tissue damage, while bacterial infection aggravates the wound environment further. It is impossible to resolve all these thorny problems simultaneously with a wound dressing that has only one function. The antioxidative and anti-inflammatory properties of resveratrol (Res) have been proven. However, the effect of Res is non-selective, and high levels of Res can inhibit cell growth and promote oxidation. Res is also difficult to dissolve and possesses insufficient antibacterial properties, so its role is limited. In this study, Res was assembled via Mannich reaction into nanoparticles and functionalized by phenylboric acid, giving rise to targeting bacteria and solving the water-insoluble dilemma of Res. In comparison with Trolox, the assembled Res NPs performed better at scavenging ABTS and DPPH free radicals. Furthermore, Res NPs that targeted bacteria also showed high biocompatibility at concentrations five times higher than pure Res. The activities of Res NPs were comparable to free Res in downregulating the expression of inflammatory cytokines, and reducing intracellular excessive ROS. The gel embedded with Res NPs accelerated the formation of granulation tissue, collagen deposition, and re-epithelialization, facilitating wound healing. The present study suggests that functionalized polyphenol-based materials are preferably suited to the development of tissue engineering biomaterials.
Article
Recently, the development of multifunctional nanoplatforms for synergistic therapy is highly attractive in cancer treatment. Herein, a cancer cell membrane-coated biomimetic nanoplatform (termed as mPAGT) was designed through a simple and versatile polyphenol-mediated coating strategy for a catalytic cascades-enhanced tumor targeted combination therapy. Porphyrin MOFs (PCN) were selected as a nano-photosensitizer and then an endogenous nitric oxide (NO) donor L-arginine (L-Arg) was loaded into the framework to obtain PA nanoplatforms; Subsequently, glucose oxidase (GOx) was coated on the surface through the interactions between protein and polyphenol and the purified murine breast cancer cell membrane was further decorated to form the resulting nanoplatforms mPAGT NPs. The prepared nanoplatforms exhibited an excellent drug loading capability (9.59% for L-Arg and 33.75% for GOx) and homologous targeting ability. Meanwhile, the mPAGT NPs could initiated a cascade reaction included GOx catalyzed oxidation of glucose, nano-photosensitizer based ¹O2-producing, and ROS-mediated the release of NO, and induce an obvious apoptosis with the highest apoptotic ratio of 86.2%. Moreover, the in vivo experimental results further confirmed that our multifunctional nanoplatforms possessed negligible systemic toxicity and promising potential for the treatment of tumor through the catalytic cascades-enhanced synergistic therapy.
Article
Carbon monoxide (CO) gas therapy, a novel anti-tumor technique based on the cytotoxicity from the CO released in situ, has become one of the hot topics in cancer treatment. Since the technique is oxygen-independent, it displays promising therapeutic effect for hypoxic tumor where traditional photodynamic therapy shows limited efficacy and insufficient penetration depth. To fully address these limitations of PDT, we propose a synergetic sonodynamic-CO gas releasing strategy for the therapy of hypoxic tumor. In this work, two rhenium(I) tricarbonyl complexes with different substituted ligands are investigated for US-triggered ROS generation and CO release. Our results indicated that the electron-donating NMe2-substituted complex (Re-NMe2) exhibits stronger luminescence intensity and generates more singlet oxygen (¹O2) than the electron-withdrawing NO2-substituted complex (Re-NO2). In addition, Re-NMe2 displays release of CO triggered by US, thus showing high sono-cytotoxicity to tumor cells in-vitro and in-vivo. The strong ROS-generating capability combined with rapid CO-releasing feature from Re-NMe2 has made it a powerful tool for the efficient treatment of hypoxic tumor.
Article
In recent years, the efficacy of nano-drugs has not been significantly better than that of the drugs themselves, mainly because nano-drugs enter the tumor vasculature, stay near the blood vessels, and cannot enter the tumor tissues or tumor cells to complete the drug delivery process. Although intratumor injection can significantly decrease this risk, the side effects are strong. The advent of drug delivery carrier materials offers an opportunity to avoid the side effects of systemic drug delivery and the damage caused by tumor resection, holding great promise for the future of cancer therapy. Here, we systematically review recent research advances in the classification of drug delivery carrier materials and the delivery process in drug delivery systems. This review is divided into several main sections, first, we summarize the classification of tumor drug carrier materials, including drug delivery vectors and gene delivery vectors, etc., which are introduced in detail, respectively. Then we describe the carrier materials to deliver the drug cascade and the transition pathways for drug delivery, including stabilization transitions, charge inversions, and size changes. Finally, we discuss the current design strategies and research progress of drug vectors and provide a summary and outlook. This review aims to summarize different drug delivery vehicles and delivery processes to provide ideas for effective cancer therapy.
Article
The depletion of reactive oxygen species (ROS) by glutathione (GSH) and oxidative stress induced protective autophagy severely impaired the therapeutic effect of chemodynamic therapy (CDT). Therefore, how to construct a CDT treatment nanosystem with high yield and full utilization of ROS in tumor site is the main issue of CDT. Herein, a multifunctional cascade bioreactor based on mesoporous Mo-doped Cu9S5 (m-MCS) nanozymes loaded with L-Arginine (LA), abbreviated as [email protected], is constructed for realizing enhanced CDT promoted by ultrasound (US) triggered gas therapy. The m-MCS based on the catalytic performance of multivalent metal ions, which were served as nanozymes, exhibit enhanced Fenton-like and glutathione (GSH) peroxidase-like activities in comparison to Cu9S5 nanoparticles without Mo-doping. Once placed in tumor microenvironment (TME), the existence of redox couples (Cu⁺/Cu²⁺ and Mo⁴⁺/Mo⁶⁺) in m-MCS enabled it to react with hydrogen peroxide (H2O2) to generate ·OH for achieving CDT effect via Fenton-like reaction. Meanwhile, m-MCS could consume overexpressed GSH in tumor microenvironment (TME) to alleviate antioxidant capability for enhancing CDT effect. Moreover, m-MCS with mesoporous structure could be employed as the carrier to load natural nitric oxide (NO) donor LA. US as the excitation source with high tissue penetration can trigger [email protected] to produce NO. As the gas transmitter with physiological functions, NO could play dual roles to kill cancer cells through gas therapy directly, and enhance CDT effect by inhibiting protective autophagy simultaneously. As a result, this US-triggered and NO-mediated synergetic cancer chemodynamic/gas therapy based on [email protected] NPs can effectively eliminate primary tumor and achieved tumor-specific treatment, which provide a possible strategy for developing more effective CDT in future practical applications. Statement of Significance The depletion of reactive oxygen species (ROS) by glutathione (GSH) and oxidative stress induced protective autophagy severely impaired the therapeutic effect of chemodynamic therapy (CDT). Herein, a multifunctional cascade bioreactor based on mesoporous Mo-doped Cu9S5 (m-MCS) nanozymes loaded with L-Arginine ([email protected]) is constructed for realizing enhanced CDT promoted by ultrasound (US) triggered gas therapy. The m-MCS with double redox couples presents the enhanced enzyme-like activities to perform cascade reactions for reducing GSH and generating ROS. LA loaded by m-MCS can produce NO triggered by US to inhibit the mitochondria protective autophagy for reactivating mitochondria involved apoptosis pathway. The US-triggered and NO-mediated CDT based on [email protected] can effectively eliminate primary tumor through the high yield and full utilization of ROS.
Article
With the rapid development of nanomedicine, low side effects and high-efficiency green antitumor approaches have attracted great attention. Herein, we report a strategy for the in situ synthesis of graphene oxide@zeolitic imidazolate framework-8 (GOx@ZIF-8) composite nanoparticles with high catalytic efficiency, under mild conditions by adding GOx molecules to the precursor of ZIF-8, and use them as a carrier to achieve efficient loading of l-Arg. In addition. folic-acid-conjugated bovine serum albumin (FA-BSA) has been used to engineer the surface of GOx@ZIF-8-l-Arg composite nanoparticles to enhance their specific recognition of tumor cells. With the high glucose level and low pH in the tumor intracellular environment, FA-BSA/GOx@ZIF-8-l-Arg rapidly consumed the intracellular glucose and produced H2O2, which profusely deteriorated the intracellular environment. Subsequently, a large amount of l-Arg was continuously released from the nanoparticles, reacting with H2O2 to continuously produce a high concentration of nitric oxide (NO), which further damaged the tumor cells. The FA-BSA/GOx@ZIF-8-l-Arg composite nanoparticles were cleverly designed to kill cancer cells efficiently through a starvation-NO synergistic process. This emerging green antitumor method has a promising application prospect in targeted therapy for the efficient clearance of cancers.