ArticlePDF Available

PEGylated hollow pH‐responsive polymeric nanocapsules for controlled drug delivery

Wiley
Polymer International
Authors:

Abstract and Figures

Novel pH‐responsive PEGylated hollow nanocapsules (HNCaps) were fabricated through a combination of distillation–precipitation copolymerization and surface thiol–ene ‘click’ grafting reaction. For this purpose, SiO2 nanoparticles were synthesized using the Stöber approach, and then modified using 3‐(trimethoxysilyl)propyl methacrylate (MPS). Afterward, a mixture of triethyleneglycol dimethacrylate (as crosslinker), acrylic acid (AA; as pH‐responsive monomer) and MPS‐modified SiO2 nanoparticles (as sacrificial template) was copolymerized using the distillation–precipitation approach to afford SiO2@PAA core–shell nanoparticles. The SiO2 core was etched from SiO2@PAA using HF solution, and the obtained PAA HNCaps were grafted with a thiol‐end‐capped poly(ethylene glycol) (PEG) through a thiol–ene ‘click’ reaction to produce PAA‐g‐PEG HNCaps. The fabricated HNCaps were loaded with doxorubicin hydrochloride (DOX) as a model anticancer drug, and their drug loading and encapsulation efficiencies as well as pH‐dependent drug release behavior were investigated. The anticancer activity of the drug‐loaded HNCaps was extensively evaluated using MTT assay against human breast cancer cells (MCF7). The cytotoxicity assay results as well as superior physicochemical and biological features of the fabricated HNCaps mean that the developed DOX‐loaded HNCaps have excellent potential for cancer chemotherapy. © 2020 Society of Chemical Industry
This content is subject to copyright. Terms and conditions apply.
This article has been accepted for publication and undergone full peer review but has not
been through the copyediting, typesetting, pagination and proofreading process which may
lead to differences between this version and the Version of Record. Please cite this article
as doi: 10.1002/pi.5987
PEGylated hollow pH-responsive polymeric nanocapsules for controlled
drug delivery
Bakhshali Massoumi1, Mojtaba Abbasian1, Rana Jahanban-Esfahlan2, Sanaz Motamedi1, Hadi
Samadian3, Aram Rezaei3, Hossein Derakhshankhah4, Amir FarnudiyanHabibi5,6, and Mehdi
Jaymand,3
1. Department of Chemistry, Payame Noor University, Tehran, Iran.
2. Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of
Medical Sciences, Tabriz, Iran.
3. Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical
Sciences, Kermanshah, Iran.
4. Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences,
Kermanshah, Iran.
5. Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences,
Tehran, Iran.
6. Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences,
Tehran, Iran.
Correspondence to: Mehdi Jaymand, Nano Drug Delivery Research Center, Health Technology Institute,
Kermanshah University of Medical Sciences, Kermanshah, Iran.
E-mail addresses: m_jaymand@yahoo.com; m.jaymand@gmail.com; mehdi.jaymand@kums.ac.ir
This article is protected by copyright. All rights reserved.
Abstract
Novel pH-responsive PEGylated hollow nanocapsules (HNCaps) were fabricated through
a combination of distillationprecipitation copolymerization and surface thiol-ene “click”
grafting reaction. For this purpose, silica nanoparticles (SiO2 NPs) were synthesized by
Stöber approach, and then modified using 3-(trimethoxysilyl) propyl methacrylate (MPS).
Afterward, a mixture of triethyleneglycol dimethacrylate (TEGDMA; as crosslinker),
acrylic acid (AA; as a pH-responsive monomer), and MPS-modified SiO2 NPs (as
sacrificial template) were copolymerized using distillationprecipitation approach to afford
SiO2@PAA core-shell NPs. The SiO2 core was etched from the SiO2@PAA using HF
solution, and the obtained PAA HNCaps were grafted with a thiol-end capped
poly(ethylene glycol) (PEG-SH) through thiol-ene “click” reaction to produce PAA-g-PEG
HNCaps. The fabricated HNCaps were loaded with doxorubicin hydrochloride (DOX) as
a model anticancer drug, and their drug loading and encapsulation efficiencies as well as
pH-dependent drug release behavior were investigated. The anticancer activity of the drug-
loaded HNCaps was extensively evaluated using MTT assay against human breast cancer
cells (MCF7). As the cytotoxicity assay results as well as superior physicochemical and
This article is protected by copyright. All rights reserved.
biological features of fabricated HNCaps, the developed DOX-loaded nanomedicines has
excellent potential for cancer chemotherapy.
Keywords: Hollow nanocapsules, pH-Responsive, PEGylation, Nanomedicine, Cancer
chemotherapy
1. Introduction
At current time, cancer is one of the most important health threats with an increasing
incidence worldwide [1]. The conventional strategies (e.g., radiation therapy) has some
unwanted outcomes, including treat both cancerous and surrounding normal tissues
simultaneously, which led to poor therapeutic effect on tumors and severe toxic side effects
on normal tissues. Given this fact, design and development of personalized medicine to
address traditional cancer therapy issues is pivotal for coming [2-5]. In this context, the
advent of nanotechnology open new opportunities for design and development of de novo
drug delivery systems (DDSs) in the nanometer-sized domain. Since the first use of
nanoparticles (NPs) in the late 1960s by Speiser and his colleagues for biomedicinal
applications, known as nanomedicine, this field progressed significantly [6, 7].
Polymeric hollow nanocapsules (PHNCaps) are one of the valuable advances and
innovative achievements in the field of nanomedicine [8-10]. These type of NPs consist of
This article is protected by copyright. All rights reserved.
a hollow inner core surrounded by a polymeric shell, have received a great deal of attention
mainly due to their potential biomedical applications, including drug or gene delivery
vehicles, nanoreactors, and protective shells for enzymes [11, 12]. In the case of DDSs,
hollow nanocapsules (HNCaps) exhibit excellent capability for encapsulation of large
quantities of guest molecules (e.g., drug or other bioactive agents) within their inner cavity.
In addition, HNCaps have excellent stability, relatively long circulation in blood, and better
mechanical properties than those of the other DDSs (e.g., liposome, and solid lipid
nanoparticles (SLNs)) [13, 14]. Some synthetic strategies, including layer-by-layer self-
assembly, sol–gel process, template synthesis, crosslinking of micelles, directed self-
assembly approaches, and heterophase polymerization have been applied for the
fabrication of HNCaps [15-17]. In this context, heterophase polymerization approaches
(e.g., emulsion, suspension, and dispersion) are particular of interest due to their
simplicities and efficiencies toward the fabrication of HNCap with well-defined
morphology and shell thickness [17].
On the other hand, the “smart” HNCaps consist of stimuli-responsive polymeric shell have
received increasing attention due to their high flexibility in tuning the permeability. In
DDSs based on “smart” HNCaps, the encapsulated drug can be released owing to response
to external triggers such as pH, temperature, electric field, and ionic strength [17-19].
Among the above mentioned stimuli, the design and development of pH-responsive DDSs
is particular of interest due to simplicity and efficiency of this stimuli in comparison with
This article is protected by copyright. All rights reserved.
others [20, 21]. In addition, it is worth noting that cancerous tissues have lower pH values
than those of the surrounding normal tissues, which originate from abnormal metabolism
of these tissues [22, 23]. This fact is the key point for the intense interest toward the design
and development of pH-responsive polymeric nanosystems for drug delivery purposes.
In general, pH-responsive polymers processes monomeric units containing basic (e.g.,
ammonium salt) or acidic (e.g., carboxyl) functionalities, which can either accept or release
protons in response to pH changes, accompanying reversible variation in volume, solubility
and equipoise between the extended and collapsed states [20, 21]. In this context,
poly(acrylic acid) (PAA) is the most famous pH-responsive polymer with pendent carboxyl
group in each repeat unit and received a great deal of interest due to its superior
physicochemical as well as biological features [20, 24].
In this contribution, novel pH-responsive PEGylated hollow nanocapsules (HNCaps) were
fabricated through a multistep process. In first stage, the SiO2@PAA core-shell NPs were
synthesized through distillationprecipitation copolymerization of MPS-modified SiO2
NPs, AA, and TEGDMA. Afterward, the SiO2 core was etched from the SiO2@PAA NPs
using HF solution, and the obtained PAA HNCaps were grafted with a PEG-SH through
thiol-ene “click” reaction to produce PAA-g-PEG HNCaps. The fabricated HNCaps were
loaded with DOX as a model anticancer drug, and their drug loading and encapsulation
efficiencies as well as pH-dependent drug release behavior were investigated. The
This article is protected by copyright. All rights reserved.
anticancer drug delivery performance of PAA-g-PEG HNCaps was evaluated through
MTT assay against MCF7 cell line.
2. Experimental
2.1. Materials
Acrylic acid monomer (Merck, Darmstadt, Germany) was distilled in reduced pressure,
and stored at −20 °C prior to use. The initiator 2,2′-azobisisobutyronitrile (AIBN; Fluka,
Switzerland) was recrystallized from ethanol at 50 °C before use. Tetrahydrofuran (THF)
was purchased from Merck, dried by refluxing over sodium, and distilled under argon prior
to use. Poly(ethylene glycol) monomethylether (Mn=2000 gmol-1), triethyleneglycol
dimethacrylate (TEGDMA), succinic anhydride (SA), N-(3-dimethylamino propyl-N-
ethylcarbodiimide) hydrochloride (EDC•HCl), N-hydroxysuccinimide (NHS), 3-
(trimethoxysilyl) propylmethacrylate (MPS), tetraethyl orthosilicate (TEOS), and
ammonium hydroxide (25% of ammonia) were purchased from Sigma-Aldrich (St. Louis,
MO, USA) and were used as received. Doxorubicin hydrochloride (DOX) was purchased
from Zhejiang Hisun Pharmaceutical Co., Ltd., Taizhou, China. Phosphate buffered saline
(PBS), fetal bovine serum (FBS), MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphe-
nyltetrazolium bromide), and other biological reagents were purchased from Invitrogen
(Carlsbad, CA, USA) and were used as received. All other reagents were bought from
Merck or Sigma-Aldrich and purified according to the standard methods.
2.2. Synthesis of SiO2 NPs
This article is protected by copyright. All rights reserved.
Silica NPs were synthesized by the hydrolysis of TEOS in ethanol medium in the presence
of ammonium hydroxide as the catalyst. In a typical experiment, ethanol (16 molL−1) was
taken and kept in a sonication bath. After 15 minutes, TEOS (24 mmolL−1) was added
dropwise under sonication. At the end of this stage, ammonium hydroxide (25%, 2
mmolL−1) was added as the catalyst to promote the condensation reaction. Afterward, the
content of the reactor was sonicated for another 2 hours to get a white opalescent
suspension. The synthesized SiO2 NPs were precipitated by centrifugation at 6000 rpm for
about 15 minutes. The white powder was then re-dispersed in ethanol and re-precipitated
by centrifugation several times, in order to remove unreacted reagents. The white powder
obtained was dried under vacuum at room temperature.
2.3. Synthesis of MPS-modified SiO2 NPs
In a typical experiment, SiO2 NPs (1.00 g) was suspended in ethanol (100 mL) through the
sonication for about 20 minutes. The mixture was then transferred into a 250-mL round-
bottomed flask equipped with condenser, and a magnetic stirrer. The silane coupling agent
(MPS; 1.00 mL, 4.2 mmol) was added dropwise to the reaction mixture. The content of the
flask was refluxed overnight at 65 °C. At the end of this time, the MPS-modified SiO2 NPs
were precipitated by centrifugation at 6000 rpm for 15 minutes. The modified NPs were
then re-dispersed in ethanol and re-precipitated by centrifugation several times, in order to
remove unreacted silane coupling agents and byproducts. The product obtained was dried
in a reduced pressure at room temperature.
This article is protected by copyright. All rights reserved.
2.4. Synthesis of SiO2@PAA core-shell NPs
The SiO2@PAA core-shell NPs were synthesized through distillationprecipitation
copolymerization of AA monomer with MPS-modified SiO2 NPs as template. It should be
pointed out that TEGDMA was used as crosslinker. For this purpose, a 50-mL three-necked
round-bottomed flask equipped with a condenser, gas inlet/outlet, and a magnetic stirrer
was charged with MPS-SiO2 (0.50 g), dried 1,4-dioxene (20 mL), AA monomer (0.35 mL,
5 mmol), and TEGDMA (52 µL, 0.2 mmol). The reaction mixture was stirred for about 1
hours under argon protection in order to obtain a homogeneous suspension. Afterward,
AIBN (20 mg, 0.12 mmol) was added to the flask, and the reaction mixture was refluxed
for about 12 hour at 80 °C. At the end of this time, the content of the flask was cooled using
an ice/water bath in order to stop the copolymerization. The SiO2@PAA NPs were
collected through centrifugation at 6000 rpm for 15 minutes, and washed by
tetrahydrofuran (THF) and ethanol, respectively. The obtained product was dried under
reduced pressure at room temperature.
2.5. Synthesis of PAA HNCaps
The PAA HNCaps were synthesized through HF etching of the SiO2 core from SiO2@PAA
NPs. Briefly, SiO2@PAA NPs (0.50 g) were stirred in a HF solution (50 mL; 20%) at room
temperature for about 24 hours to dissolve the silica core. The mixture was dialyzed in
double distilled water (DDW) for three days using 1000 molecular weight cut-off dialyzed
This article is protected by copyright. All rights reserved.
bag in order to remove the excess HF and SiF4 byproduct. Finally, the PAA HNCPs were
collected through freeze-drying.
2.6. Synthesis of carboxyl-end capped PEG (PEG-COOH)
The PEG-COOH was synthesized through the reaction between PEG and succinic
anhydride (SA). In a typical experiment, a 50-mL round-bottomed flask equipped with
condenser and a magnetic stirrer was charged with poly(ethylene glycol) monomethylether
(2.50 g , 1.25 mmol), SA (0.15 g, 1.5 mmol), and dried THF (30 mL). The reaction mixture
was refluxed at 60 °C overnight. At the end of this time, the solvent (THF) was evaporated
under reduced pressure, and the obtained crude product was extracted using CH2Cl2/DDW
(50:50 v/v) three times, in order to remove unreacted SA. The organic phase was dried
using Na2SO4 and evaporated under a reduced pressure to afford a PEG-COOH.
2.7. Synthesis of thiol-end capped PEG (PEG-SH)
The PEG-SH was synthesized through the esterification of PEG-COOH using 3-amino-1-
propanethiol hydrochloride in the presence of EDC and NHS. For this purpose, a 25-mL
three-necked round-bottomed flask equipped with condenser, gas inlet/outlet, and a
magnetic stirrer was charged with PEG-COOH (0.85 g , 0.40 mmol), EDC (80 mg, 0.40
mmol), NHS (48 mg, 0.40 mmol), and dried CH2Cl2 (20 mL). The reaction mixture was
stirred for about 4 hours, and then 3-amino-1-propanethiol hydrochloride (50 mg, 0.40
This article is protected by copyright. All rights reserved.
mmol) was added to the flask. The content of the flask was stirred for about 24 hours at
room temperature. At the end of this time, the crude product was extracted with
CH2Cl2/DDW (50:50 v/v) in order to remove residual reagents or by products. The organic
phase was dried using Na2SO4 and evaporated under reduced pressure to afford a PEG-SH.
2.8. Synthesis of PAA-g-PEG HNCaps
The PAA-g-PEG HNCaps were synthesized through a thiol-ene “click” reaction as follows.
A 25-mL three-necked round-bottomed flask equipped with condenser, gas inlet/outlet, and
a magnetic stirrer was charged with PAA HNCaps (0.30 g), PEG-SH (0.10 g, 0.045 mmol),
and dried 1,4-dioxane (15 mL). The content of the falsk was stirred for about one hour, and
then de-aerated through the bubbling of highly pure argon for some minutes. Afterward,
the initiator (AIBN, 20 mg, 0.12 mmol) was dissolved in 1,4-dioxane (5 mL), de-aerated
using argon gas, and then added to the reaction mixture under an argon protection. The
reaction was allowed to proceed for about 24 hours at 80 °C. At the end of this time, the
synthesized PAA-g-PEG HNCaps was obtained through centrifugation, followed by
washing using THF and ethanol, respectively in order to remove un-grafted PEG chains.
2.9. Preparation of DOX-loaded PAA-g-PEG HNCaps
The PAA-g-PEG/DOX nanomedicine was formulated through the stirring a mixture of
PAA-g-PEG HNCaps (0.30 g) and DOX (20 mg) in DDW (20 mL) for about 48 hours in
the dark at room temperature. At the end of this time, the mixture was filtered using ultra-
filtration method through centrifugal devices (Amicon® Ultra-4 12 kDa, Millipore,
This article is protected by copyright. All rights reserved.
Temecula, CA, USA) to separate unloaded DOX. The encapsulation (EE) and loading (LE)
efficiencies of the PAA-g-PEG HNCaps were determined through the analyzing of ultra-
filtrate solution for determination of unloaded drug concentration using a UV-vis
spectrophotometer at 480 nm according to following equations.
EE(%)=( )
× 100
LE(%)=( )
× 100
In these equations CT is total amount of DOX which taken for loading, CDOX is DOX
concentration in ultra-filtrate solution, and WS is the mass of sample which used for DOX
loading [25].
2.10. In vitro DOX release study
The in vitro release of DOX form the developed PAA-g-PEG/DOX nanomedicine was
evaluated under various pH values (4.0, 5.4, and 7.4) according to our previously reported
procedure [25]. For this purpose, the PAA-g-PEG/DOX nanomedicine was suspended in
PBS sealed in a dialysis membrane bag with a molecular cut-off of 2 kDa, and placed in
PBS (0.01 moll-1, 100 mL, pH=4.0, 5.4, and 7.4) as the release medium. The system was
kept at 37 °C with gentle stirring for predetermined times. Aliquots of release medium (1
mL) were collected at predetermined time intervals, for determination of released DOX
using a UV–vis spectrophotometer at 480 nm. It is worth noting that the aliquots were
This article is protected by copyright. All rights reserved.
brought back into the reactor to avoid volume change. The drug release values were
calculated according to following equation:
R=M1/M0
In this equation M1 is the cumulative mass of DOX released from the sample at a given
time, and M0 is the total loading amount of DOX in the sample that determined using UV–
vis spectroscopy at 480 nm as described above.
2.11. Cell culture
The MCF7 cells was provided from Iranian National Cell Bank (Pasteur Institute, Tehran,
Iran), and cultivated in RPMI1640 possessing 100 IU penicillin per 100 mg streptomycin,
enriched with 10% (v/v) of FBS. The cells were cultured into flasks and kept in a humidified
incubator at 37 °C and the media refreshed every two days [26].
2.12. In vitro cytotoxicity assay
The well-known MTT assay was applied to evaluate the cytotoxic effects of the developed
PAA-g-PEG/DOX nanomedicine versus free DOX as the reference. In a typical
experiment, the MCF7 cells was trypsinized, harvested and seeded in 96-well plates. After
overnight incubation, the cells were treated with various concentrations (5.0, 10.0, 20.0,
50.0, and 100.0 µgmL-1) of PAA-g-PEG/DOX nanomedicine (based on concentration of
DOX) and free DOX for 24 and 48 hours. After mentioned times, the media was removed
and MTT solution (50 µL) was added to each well followed by 150 µL cultivation medium,
then placed in humidified incubator for another 4 hours. At the end of this time, the
This article is protected by copyright. All rights reserved.
remaining MTT solution aspirated, the formed formazan crystals were dissolved in
dimethyl sulfoxide (DMSO; 200 µL) containing Sorenson buffer (25 µL) and absorbance
was measured at 570 nm using a spectrophotometric plate reader [26]. The cell viability
(%) was calculated using the following equation. In order to quantification of cytotoxic
effects.
Cell viability (%) = [A570(sample)/A570(control)] × 100
In this equation, A570(sample) and A570(control) represent the absorbances of the sample
and control wells, respectively.
2.13. Statistical analyses
One-way analysis of variance was performed to examine differences in the average values
between groups and the data were analyzed by SPSS 19.0 software (SPSS Inc., Chicago,
IL, USA). Each result is presented as mean±SD. A value of P<0.05 was considered
statistically significant.
2.14. Instrumentation
Fourier transform infrared (FTIR) spectroscopy was performed using a Shimadzu 8101M
FTIR (Kyoto, Japan) at room temperature. The samples were prepared in the pellet form
with potassium bromide (KBr) powder. Proton nuclear magnetic resonance (1H NMR)
spectra were recorded on an FT-NMR Bruker spectrometer (Bruker, Ettlingen, Germany)
This article is protected by copyright. All rights reserved.
with an operating frequency of 400 MHz at 25 °C. Transmission electron microscopy
(TEM) images were obtained using a CM10-TH microscope (Philips, Eindhoven, The
Netherlands) with a 100 kV accelerating voltage. Thermal properties of the synthesized
samples were examined using thermogravimetric analyzer (TGA-PL STA 1640 equipment
(Polymer Laboratories, Shropshire, UK)). The thermogravimetric analysis (TGA)
experiment was conducted under nitrogen atmosphere from room temperature to 700 °C
with heating rate of 10 °Cmin−1. Dynamic light scattering (DLS) experiments were carried
out using a Zetasizer Nano ZS90 (Malvern Instruments, Malvern, UK) at room
temperature. Samples were prepared as 0.5% (w/v) solutions in DDW. The powder X-ray
diffraction (XRD) experiment was performed using a Siemens D5000 diffractometer
(Aubrey, Texas, USA), X-ray generator (CuKα radiation with λ=1.5406 Å) in the scan
range from 2 to 80° (2θ) at room temperature. Ultraviolet-visible (UV-vis) spectra were
recorded with a Shimadzu 1650 PC UV-vis spectrophotometer (Kyoto, Japan).
3. Results and discussion
Among the numerous DDSs, polymeric HNCaps have received a great deal of interest due
to their superior features, including high specific surface area, low density, large pore
volume, and mechanical stability that lead to high drug loading capacity and improving
drug stability. In this context, the stimuli-responsive polymeric HNCaps are particular of
interest due to their “smart” and controlled drug release features. According to these facts,
This article is protected by copyright. All rights reserved.
the aim of this study was to the design and development of novel PEGylated pH-responsive
HNCaps for “smart” chemotherapy of solid tumors (Scheme 1).
(Scheme 1)
3.1. Characterization of SiO2@PAA NPs
The DLS diagram of SiO2 NPs, SiO2@PAA NPs, and PAA HNCaps as well as XRD
pattern of the synthesized SiO2 NPs are shown in Figure 1. According to the DLS results
(Figure 1a) the size distribution of the synthesized SiO2 NPs, SiO2@PAA NPs, and PAA
HNCaps were obtained to be 24, 51, and 40 nm, respectively. In addition, polydispersity
index (PDI) of the SiO2 NPs, SiO2@PAA NPs, and PAA HNCaps were as 0.11, 0.13, and
0.16, respectively that indicate the relatively monodisperse synthesis of all samples.
The XRD pattern of SiO2 NPs (Figure 1b) exhibited a broad diffraction peak approximately
at 23°. This broad diffraction peak related to the small size and incomplete inner structure
of the SiO2 NPs. This demonstrates that the most of the SiO2 NPs are amorphous, but a few
of them are crystalline.
(Figure 1)
The FTIR spectra of the SiO2 NPs, MPS-SiO2 NPs, and SiO2@PAA NPs are shown in
Figure 2. The most important characteristic absorption bands in the FTIR spectrum of the
SiO2 NPs are symmetric and asymmetric stretching vibrations of the Si–O–Si at 791 and
936, and 1067 cm−1, respectively as well as the bending and stretching vibrations of the
surface hydroxyl groups at 1647 and 3360 cm−1, respectively.
This article is protected by copyright. All rights reserved.
Silane coupling agents have been widely used for the surface modification of inorganic
NPs in order to prevent aggregation as well as further modification through their reactive
functional groups [27]. The appearance of new absorption band at 1722 cm−1 related to
carbonyl group as well as the stretching vibrations of aliphatic C–H at 2950–2800 cm−1
region confirms the successful surface modification of SiO2 NPs using MPS. Furthermore,
after modification of SiO2 NPs using MPS the stretching vibration of hydroxyl groups is
decreased due to coupling of MPS moiety.
The grafting and simultaneous crosslinking of AA monomer onto MPS-SiO2 NPs was
confirmed through the appearance of some characteristic bands, including the stretching
vibration of C–O group at 1196 cm-1, the bending vibration of –CH2 groups at 1467 cm-1,
the stretching vibration of carbonyl group at 1716 cm-1, the stretching vibrations of
aliphatic CH at 2950 to 2850 cm-1 region, and the hydroxyl stretching vibration (related
to both PAA and SiO2 NPs) as a broad and strong band centered at 3465 cm-1 in the FTIR
spectrum of the SiO2@PAA NPs.
(Figure 2)
3.2. Characterization of PAA-g-PEG HNCaps
The FTIR spectra of the PEG, PEG-SA, PEG-SH, and PAA-g-PEG HNCaps are shown in
Figure 3. The FTIR spectrum of the neat PEG exhibited the stretching vibration of C–O at
1108 cm-1, the bending vibrations of –CH2 groups at 1467 and 1356 cm-1, the stretching
vibrations of aliphatic CH at 2950 to 2800 cm-1 region, and the stretching vibration of
This article is protected by copyright. All rights reserved.
hydroxyl or adsorbed water as a broad band centered at 3490 cm-1. The most important
change in the FTIR spectrum of PEG-SA is the appearance of a weak peak at 1732 cm-1
attributable to the carbonyl groups of SA moiety. As seen, the FTIR spectrum of PEG-SH
showed similar absorption bands with minor differences.
The FTIR spectrum of the synthesized PAA-g-PEG HNCaps represent characteristic bands
related to both PAA as well as PEG polymeric chains. The most important absorption bands
are labeled in the FTIR spectrum of the PAA-g-PEG HNCaps.
(Figure 3)
The synthesized PEG-SA, and PEG-SH were further characterized using 1H NMR
spectroscopy as shown in Figure 4. The neat PEG exhibited the chemical resonance of
hydroxyl end group at 2.30–2.60 ppm (a), OCH3 group at 3.20 ppm (b), and O–CH2
groups of PEG backbone at 3.35–3.85 ppm (c, d). The successful synthesis of PEG-SA was
verified through the appearance of a new chemical shift at 2.85 ppm (e and f) related to
CH2 groups of SA moiety. Finally, the chemical shifts at 2.35 (j) and 3.60 (k and h) ppm
confirm the synthesis of PEG-SH compound.
(Figure 4)
3.3. TEM observation
Figure 5, shows the TEM images of SiO2@PAA NPs (a) and PAA HNCaps (b). As seen,
the SiO2@PAA NPs exhibited spherical morphology with average sizes distribution of
40±10 nm. In this image, the dark spots attributable to the SiO2 NPs and light regions
This article is protected by copyright. All rights reserved.
represent PAA matrix, which confirm the coreshell structure for the synthesized NPs. In
contrast, after HF etching of the SiO2 core from SiO2@PAA NPs the most of dark spots
disappeared at the TEM image due to removing of SiO2 core. As seen the PAA HNCaps
have spherical morphology with average size distribution of 30±10 nm.
(Figure 5)
3.4. Thermal property study
The thermal degradation of SiO2 NPs, MPS-SiO2 NPs, SiO2@PAA NPs, PAA HNCaps,
and PAA-g-PEG HNCaps in the course of heating under nitrogen flow are shown in Figure
6. As seen, the SiO2 NPs showed excellent thermal stability with only 8 wt.% weight loss
between 50 to 700 °C. This weight loss is started with evaporation of residual solvent
(ethanol) and adsorbed water (50 to 120 °C) followed by elimination of water molecules
from two unreacted silanol groups (120 to 220 °C), and after which the loss rate slows
down. By considering the TGA curve of MPS-SiO2 NPs, it was found that the weight
percentage of MPS in the modified NPs is about 5 wt.%. The MPS moiety is degrade in
the temperature range of 200 to 250 °C.
The characteristic TGA curve of SiO2@PAA NPs exhibited three-step weight loss
processes as follows. The first step involve the evaporation of residual solvent and adsorbed
water followed by elimination of water molecules from two unreacted silanol groups and
finally the degradation of modifier (MPS) (50 to 250 °C). The second step (250 to 320 °C)
is related to the de-carboxylation and anhydride formation in the PAA segments [28]. The
This article is protected by copyright. All rights reserved.
final weight loss step (400 to 480 °C) is associated with the degradation of the poly(acrylic
anhydride) backbone, and after which the loss rate slows down. The residue at 700 °C for
this sample is about 54.5 wt.%, which most portion of this residue is related to the SiO2
NPs. The characteristic TGA curve of PAA HNCaps showed similar weight loss processes
as discussed above, and the residue at 700 °C for this sample is about 5.5 wt.%. This
quantity of residue confirmed the successful etching of the SiO2 core from SiO2@PAA
NPs using HF.
The TGA trace of PAA-g-PEG HNCaps also showed three-step weight loss processes. The
decomposition of this sample was started through the evaporation of solvent and adsorbed
water followed by dehydration and degradation of small molecules (e.g., MPS) (50 to 280
°C). The second step is corresponded to de-carboxylation and anhydride formation in the
PAA segments (280 to 380 °C). The final decomposition step is related to the degradation
of the poly(acrylic anhydride) and PEG backbones (380 to 470 °C), and after which the
loss rate slows down. The residue at 700 °C for this sample is about 4 wt.%. As seen, the
thermal stability of PAA HNCaps is improved slightly after grafting of PEG segments.
(Figure 6)
3.4. Investigation of self-assembly behavior
In all pH values the micelles with PAA inner shell and a PEG outer shell is expected.
However, at lower pH values (pH˂4.2) the PAA inner shell shrinkage due to fully
protonation of carboxyl groups (Scheme 2). In detail, at pH 4 the average size of these
This article is protected by copyright. All rights reserved.
aggregates was found to be 85 nm. In contrast, when the pH value were increased to 5.5 or
7.4 the average sizes of the fabricated PAA-g-PEG HNCaps were increased to 102.2 and
121.5 nm, respectively due to deprotonation of PAA carboxyl groups. This phenomenon
may be originated from disappearance of the hydrogen bonding between the PAA segments
due to ionization of their carboxylic acid groups as well as electronic repulsion between
carboxylate groups (Figure 7).
(Scheme 2)
(Figure 7)
3.5. Stability of the micelles
The stability of a DDS in physiological condition has significantly affect its final
performance. The stability of the fabricated DDS was investigated through DLS
measurements of fresh micelles and incubated micelles for about 3 days in PBS at
physiological condition (pH=7.4 and temperature value of 37 °C). As seen in Figure 8, the
synthesized PAA-g-PEG HNCaps showed excellent stability after 3 days incubation. In
detail, the average hydrodynamic diameters of PAA-g-PEG HNCaps was increased only
23 nm due to slightly aggregation of micelles. This excellent stability of micelles may be
originated from negatively charged of core (PAA) that lead to electronic repulsion as well
as the effect of PEG chains as shell.
This article is protected by copyright. All rights reserved.
(Figure 8)
3.6. Drug loading and encapsulation efficiencies
As the hollow nature of PAA-g-PEG HNCaps as well as the strong physical interactions
between the HNCaps and DOX (e.g., firm hydrogen bonding and Van der Waals forces), it
is expected that the fabricated nanosystem has high drug loading and encapsulation
efficiencies. The DOX-encapsulation efficiency (EE) and DOX-loading efficiency (LE) of
the synthesized PAA-g-PEG HNCaps were calculated to be 83 and 71%, respectively under
mentioned experimental condition.
3.7. In vitro drug release study
The in vitro drug release behavior of the DOX-loaded PAA-g-PEG HNCaps was
investigated under various pH values (4.0, 5.4, 6.6 and 7.4) at 37 °C. It should be pointed
out that the pHs 4.0, 5.4, and 6.6 are the average pH values of cancer cell lysosomal,
endosomal, and tumor tissue environment, respectively, and the pH 7.4 is the normal
physiological pH in the human body.
As seen in release profiles (Figure 9), drug release from the DOX-loaded PAA-g-PEG
HNCaps is a pH-responsive process where the protonation of carboxyl groups of PAA in
acidic conditions resulted in a faster release of encapsulated cargo. In contrast, at neutral
pH (7.4) the fully deprotonated PAA lead to strong interactions (e.g., firm hydrogen bonds
and Van der Waals forces) between PAA-g-PEG HNCaps and DOX, which delays the
release of loaded drug. According to Figure 9, the conclusion could be drown that the drug
This article is protected by copyright. All rights reserved.
release values in pHs 4.0, 5.4, 6.6, and 7.4 after 300 hours were 42.1, 29.5, 24.8, and 22%,
respectively. It should be pointed out that in pH 4 the protonation of DOX, which increased
the dissolution of this drug is another reason for the higher drug release value.
It is worth noting that according to acidic feature of tumor microenvironment in
comparison with surrounding normal tissues and based on obtained results, the developed
PAA-g-PEG HNCaps nanomedicines can be considered as de novo drug delivery system
for “smart” chemotherapy of solid tumors.
(Figure 9)
3.8. In vitro cytotoxicity assay
The cytotoxicity effects of PAA-g-PEG HNCaps as well as DOX-loaded HNCaps were
investigated using MMT assay against MCF7 cells and the results obtained were compared
with the cytotoxicity effect of the free DOX as the reference. As seen in Figure 10, the
PAA-g-PEG HNCaps has acceptable biocompatibility during incubation time period of 24
hours, and the cells viability is more than 91% even at high concentration (200 µg mL-1).
In contrast, the free DOX as well as DOX-loaded HNCaps showed cytotoxic effects on
MCF7 cells. In detail, free DOX can kill cancer cells more efficiently than those of the
developed PAA-g-PEG/DOX nanomedicine. This may be originated from the high
availability of DOX in the case of free DOX treatments than those of the DOX-loaded
HNCaps due to slow drug release profile according to Figure 9. Despite, the use of free
DOX is not recommended due to some issues, including rapid drug clearance, poor
This article is protected by copyright. All rights reserved.
biodistribution, non-ideal physicochemical properties of drugs (e.g., poor solubility), rapid
drug degradation, and systemic toxicity. It is well established that the formulated anticancer
drugs have some advantages, including prolonged release profiles, favorable
biodistribution, higher therapeutic outcomes and minimizing the side effects of the drug
[29]. This theory is confirmed through the treatment of MCF7 cells with samples in the
time period of 48 hours as shown in Figure 11.
The statistical analysis results revealed that although the eradication of plain DOX was
slightly higher compared with PAA-g-PEG/DOX nanomedicine at 24 h (p<0.01) especially
in higher drug concentrations (100 and 200 µgmL-1). In contrast, after 48 h the PAA-g-
PEG/DOX nanomedicine exhibited slightly higher cytotoxicity against MCF7 cells
(p<0.05) especially at 20 and 50 µgmL-1 of DOX.
(Figure 10)
(Figure 11)
4. Conclusions
Novel pH-responsive PEGylated HNCaps was fabricated through a thiol-ene “click”
grafting of a thiol end-capped PEG onto hollow crosslinked PAA-based NPs for “smart”
chemotherapy of solid tumors. The fabricated HNCaps showed excellent drug loading
capacity (~ 71%) as well as “smart” drug release behavior under pH stimuli. In acidic
conditions, the protonation of carboxyl group of the PAA lead to faster release of
encapsulated DOX. In contrast, at neutral pH (7.4) the fully deprotonated PAA lead to
This article is protected by copyright. All rights reserved.
strong interactions between the PAA-g-PEG HNCaps and DOX, which delays the release
of loaded drug. The biocompatibility of the fabricated PAA-g-PEG HNCaps was
confirmed through MTT assay against MCF7 cells. In addition, the PAA-g-PEG/DOX
nanomedicine exhibited excellent cytotoxic effect against mentioned cancerous cells.
In conclusion, according to acidic feature of tumor microenvironment in comparison with
surrounding normal tissues and based on obtained results from drug loading and pH-
responsive drug release studies as well as cytotoxicity assay, the developed PAA-g-PEG
nanomedicine can be considered as de novo drug delivery nanosystem for smart
chemotherapy of solid tumors.
Acknowledgement
The authors gratefully acknowledge the financial support from Kermanshah University of
Medical Sciences, Kermanshah, Iran (grant number: 980304).
Competing interests
The authors declare that they have no competing interests.
This article is protected by copyright. All rights reserved.
References
[1] Bode AM and Dong Z, Nat Rev Cancer 9:508–516 (2009).
[2] Sharabi AB, Nirschl CJ, Kochel CM, Nirschl TR, Francica BJ, Velarde E, Deweese TL
and Drake CG, Cancer Immunol Res 3:1–11 (2015).
[3] Wicki A, Witzigmann D, Balasubramanian V and Huwyler J, J Control Release
200:138-157 (2015).
[4] Brannon-Peppas L and Blanchette JO, Adv Drug Delivery Rev 64:206-212 (2012).
[5] Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R and Langer R, Nat Nanotechnol
2:751–760 (2007).
[6] Khanna SC and Speiser P, J Pharm Sci 58: 1114–1117 (1969).
[7] Khanna SC, Jecklin T and Speiser P, J Pharm Sci 59, 614–618 (1970).
[8] Lollo G, Ullio-Gamboa G, Fuentes E, Matha K, Lautram N and Benoit JP, Mater Sci
Eng C 91:859-867 (2018).
[9] Liu G, Tsai HI, Zeng X, Cheng W, Jiang L, Chen H, Zhang X, Zhang J and Mei
L, ACS Biomater Sci Eng 4:1679-1686 (2018).
[10] Zhong S, Zhang H, Liu Y, Wang G, Shi C, Li Z, Feng Y and Cui X, Carbohyd
Polym 168:282-289 (2017).
[11] Sotelo-Boyás M, Correa-Pacheco Z, Bautista-Baños S and Gómez y Gómez Y, Int J
Biol Macromol 103:409-414 (2017).
This article is protected by copyright. All rights reserved.
[12] Yamala AK, Nadella V, Mastai Y, Prakash H and Paik P, Nanoscale 9:14006-14014
(2017).
[13] Ji F, Li J, Qin Z, Yang B, Zhang E, Dong D, Wang J, Wen Y, Tian L and Yao F,
Carbohyd Polym 177:86-96 (2017).
[14] Rivera MC, Pinheiro AC, Bourbon AI, Cerqueira MA and Vicente AA, Int J Biol
Macromol 79:95-102 (2015).
[15] Paik P and Zhang Y, Nanoscale 3:2215-2219 (2011).
[16] Liu X and Basu A, J Am Chem Soc 131:5718-5719 (2009).
[17] Chen L, Peng Z, Zeng Z, She Y, Wei J and Chen Y, J Polym Sci A 52:2202–2216
(2014).
[18] Belbekhouche S, Mansour O and Carbonnier B, J Colloid Interface Sci 522:183-190
(2018).
[19] Hu H, Zhang X, Sun J, An L, Du J, Yang H, Li F, Wu H and Yang S, RSC Adv 6:
105,103754-103762 (2016).
[20] Zhao X, Wei Z, Zhao Z, Miao Y, Qiu Y, Yang W, Jia X, Liu Z and Hou H, ACS Appl
Mater Interfaces 10: 6608-6617 (2018).
[21] Mahmoodzadeh F, Abbasian M, Jaymand M and Amirshaghaghi A, Polym Int
66:1651–1661 (2017).
[22] Poorgholy N, Massoumi B, Ghorbani M, Jaymand M and Hamishehkar H, Drug
Develop Indus Pharm 8:1254–1261 (2018).
This article is protected by copyright. All rights reserved.
[23] Mahmoodzadeh F, Abbasian M, Jaymand M, Salehi R and Bagherzadeh-
Khajehmarjan E, Mater Sci Eng C 93:880-889 (2018).
[24] Miao Y, Zhao X, Qiu Y, Liu Z, Yang W, and Jia X. ACS Appl Bio Mater 2: 895-905
(2019).
[25] Abbasian M, Roudi MM, Mahmoodzadeh F, Eskandani M and Jaymand M, Int J Biol
Macromol 118:1871-1879 (2018).
[26] Jaymand M, Lotfi M and Abbasian M, Mater Res Express 5:035406 (2018).
[27] Jaymand M, J Polym Res 18:1617–1624 (2011).
[28] Mozafari Z, Hatamzadeh M, Massoumi B and Jaymand M, J Appl Polym Sci
135:46657 (2018).
[29] Yang HK, Qi M, Mo L, Yang RM, Xu XD, Bao JF, Tang WJ, Lin JT, Zhang LM and
Jiang XQ, RSC Adv 6:114519–114531 (2016).
This article is protected by copyright. All rights reserved.
1
Schemes and Figures
Scheme 1. The overall strategy for the synthesis of PAA-g-PEG HNCaps.
Accepted Article
2
Figure 1. The DLS diagram of SiO2 NPs, SiO2@PAA NPs, and PAA HNCaps (a), and XRD pattern
of SiO2 NPs (b).
Accepted Article
3
Figure 2. The FTIR spectra of the SiO2 NPs, MPS-SiO2 NPs, and SiO2@PAA NPs.
Accepted Article
4
Figure 3. The FTIR spectra of the PEG, PEG-SA, PEG-SH, and PAA-g-PEG HNCaps.
Accepted Article
5
Figure 4. The 1H NMR spectra of PEG, PEG-SA, and PEG-SH.
Accepted Article
6
Figure 5. The TEM images of SiO2@PAA NPs (a) and PAA HNCaps (b).
Figure 6. The TGA curves of SiO2 NPs, MPS-SiO2 NPs, SiO2@PAA NPs, PAA HNCaps, and PAA-
g-PEG HNCaps.
Accepted Article
7
Scheme 2. The pH responsibility of the fabricated PAA-g-PEG HNCaps.
Figure 7. The DLS diagrams of PAA-g-PEG HNCaps at various pH values in 37 °C.
0
5
10
15
20
25
30
0 50 100 150 200 250
Intencity (%)
Size (d.nm)
pH=4
pH=5.5
pH=7.4
Accepted Article
8
Figure 8. The DLS diagrams of fresh PAA-g-PEG HNCaps micelles and micelles after 3 days
incubation in PBS (pH=7.4 and T=37 °C).
0
5
10
15
20
25
30
0 50 100 150 200 250
Intencity (%)
Size (d.nm)
Fresh
After 3 days
Accepted Article
9
Figure 9. In vitro drug release profiles of DOX-loaded PAA-g-PEG HNCaps at pH values of 7.4, 6.6,
5.4, and 4.0 in 37 °C. [SPSS results: (pH: 7.4/6.6; P<0.05); (pH: 7.4/5.4; P<0.01); (pH: 7.4/4;
P<0.01); (pH: 6.6/5.4; P<0.01); (pH: 6.6/4; P<0.01); and (pH: 5.4/4; P<0.01).
0
20
40
60
0 50 100 150 200 250 300 350
Release (%)
Time (h)
PAA-g-PEG (pH=7.4)
PAA-g-PEG (pH=6.6)
PAA-g-PEG (pH=5.4)
PAA-g-PEG (pH=4)
Accepted Article
10
Figure 10. The cytotoxicity assay results of PAA-g-PEG HNCaps, PAA-g-PEG/DOX, and free DOX
on MCF7 cells in time period of 24 hours.
0
20
40
60
80
100
120
0 10 20 50 100 200
Survival rate (%)
Concentrations (µg mL-1)
PAA-g-PEG HNCaps PAA-g-PEG/DOX Free DOX
Accepted Article
11
Figure 11. The cytotoxicity assay results of PAA-g-PEG HNCaps, PAA-g-PEG/DOX, and free DOX
on MCF7 cells in time period of 48 hours.
0
20
40
60
80
100
120
0 10 20 50 100 200
Survival rate (%)
Concentrations (µg mL-1)
PAA-g-PEG HNCaps PAA-g-PEG/DOX Free DOX
Accepted Article
... A higher weight loss of ~25% is displayed for the SiO2-P(AMPSA-co-DMA) NPs, over the temperature range 200-700 °C. This finding suggests that the polymerization of the AMPSA and DMA monomers onto the SiO2 surface was successful [43][44][45]. The fact that the decomposition of the organic NP coating takes place after 250 °C indicates that the SiO2-P(AMPSAco-DMA) NPs remain stable even at high temperatures, which are higher than those prevailing at oil reservoirs (<150 °C) [46]. ...
... A higher weight loss of~25% is displayed for the SiO 2 -P(AMPSA-co-DMA) NPs, over the temperature range 200-700 • C. This finding suggests that the polymerization of the AMPSA and DMA monomers onto the SiO 2 surface was successful [43][44][45]. The fact that the decomposition of the organic NP coating takes place after 250 • C indicates that the SiO 2 -P(AMPSA-co-DMA) NPs remain stable even at high temperatures, which are higher than those prevailing at oil reservoirs (<150 • C) [46]. ...
Article
Full-text available
Globally the overall oil recovery factors for primary and secondary recovery range from 35% to 45%, and a tertiary recovery method that can enhance the recovery factor by 10–30% could contribute to the energy supply. The use of nanoparticles in enhanced oil recovery (EOR) processes comprises an emerging and well-promising approach. Polymer-coated nanoparticles (PNPs) were synthesized through the free radical polymerization (FRP) of the monomers 2-acrylamido-2-methyl-1-propanesulfonic acid (AMPSA) and dodecyl methacrylate (DMA) on the surface of acrylic-modified spherical silica nanoparticles. The obtained PNPs were characterized using Attenuated Total Reflectance-Fourier Transform Infrared Spectroscopy (ATR-FTIR) and thermogravimetric analysis (TGA). Dispersions of PNPs were prepared in salt (NaCl, CaCl2) aqueous solutions, the static oil/water interfacial tension were measured using the Du Nouy ring method, and changes caused based on the oil/water contact angle were recorded optically. The PNP dispersions were used to stabilize and characterize shear-thinning oil-in-water Pickering emulsions. The capacity of the PNP dispersions and Pickering emulsions to mobilize the trapped ganglia of viscous paraffin oil, which remained after successive tests of drainage and primary imbibition, was tested with visualization experiments of the secondary imbibition in a transparent glass-etched pore network. The synthesized SiO2-P(AMPSA-co-DMA) nanoparticles were stable even at high temperatures (~200–250 °C) and displayed excellent stability in aqueous dispersions at high ionic strengths with the presence of divalent cations, and their dispersions generated stable oil-in-water Pickering emulsions with a shear-thinning viscosity. The oil-recovery efficiency is maximized when the most viscous Pickering emulsion is selected, but if energy cost factors are also taken into account, then the less viscous Pickering emulsion is preferable.
... This can be achieved through either active or passive targeting strategies. Medicinal agents can be incorporated into nanoparticles (NPs) that passively enter target tissue via enhanced permeation and retention (EPR) and through the charge of the nanoplatform [7][8][9]. In active targeting, therapeutic drugs are coupled to ligands specific to tissues or cells. ...
Article
Full-text available
The emerging strategy of biomimetic nanoparticles (NPs) via cellular membrane camouflage holds great promise in cancer therapy. This scholarly review explores the utilization of cellular membranes derived from diverse cellular entities; blood cells, immune cells, cancer cells, stem cells, and bacterial cells as examples of NP coatings. The camouflaging strategy endows NPs with nuanced tumor-targeting abilities such as self-recognition, homo-typic targeting, and long-lasting circulation, thus also improving tumor therapy efficacy overall. The comprehensive examination encompasses a variety of cell membrane camouflaged NPs (CMCNPs), elucidating their underlying targeted therapy mechanisms and delineating diverse strategies for anti-cancer applications. Furthermore, the review systematically presents the synthesis of source materials and methodologies employed in order to construct and characterize these CMCNPs, with a specific emphasis on their use in cancer treatment.
... Further, the combination of particulate materials with stimuli-responsive hydrogels improves the prospects of DDS such as better targeting, minimizing possible risk factors, and enhanced therapeutic outcomes [22,23] . Microgels are one of them which has advantages over bulk gels including much faster response to external stimuli, can be modified chemically to increase circulation time in the bloodstream, sustained release of loaded drugs, and quick clearance from the body via biodegradation. ...
Article
Full-text available
Stimuli-responsive, smart, or intelligent polymers are materials that significantly change their physical or chemical properties when there is a small change in the surrounding environment due to either internal or external stimuli. In the last two decades or so, there has been tremendous growth in the strategies to develop various types of stimuli-responsive polymer (SRP) materials/systems that are suitable for various fields, including biomedical, material science, nanotechnology, biotechnology, surface and colloid sciences, biochemistry, and the environmental field. The wide acceptability of SRPs is due to their availability in different architectural forms such as scaffolds, aggregates, hydrogels, pickering emulsions, core-shell particles, nanogels, micelles, membranes, capsules, and layer-by-layer films. The present review focuses on different types of SRPs, such as physical, chemical, and biological, and various important applications, including controlled drug delivery (CDD), stabilization of colloidal dispersion, diagnostics (sensors and imaging), tissue engineering, regenerative medicines, and actuators. The applications of SRPs have immense potential in various fields, and the author hopes these polymers will add a new field of applications through new concepts.
... Nanotechnology is a rapidly growing new field for producing valuable materials at the nanoscale with its applications in science and technology [1,2] It focuses on the study of nanomaterials' interaction with cells, tissues and bacteria, and their potential application in drug delivery, biomarker detection and quantification, and cancer therapy, among others [3]. The nanoscale materials generally referred to as nanoparticles (NPs) are highly desirable because of their small size, optical properties, high surface area to volume ratio and their multifunctional nature [4]. ...
Article
ARTICLE INFO Keywords: Nanoparticles Nano-drug delivery Apoptosis and necrosis Cytotoxicity Confocal microscopy ABSTRACT Nanomaterial-based delivery platforms have indeed revolutionized drug delivery systems in recent years. These platforms, which are based on various types of nanoparticles, offer several advantages over traditional drug delivery systems. Therefore, during this work, we synthesized gold nanoparticles (GNPs) and graphene oxide (GO) as representative nanomaterials and assessed their biocompatibility and cytoplasmic localization in vitro. Structural characterization of the prepared nanoparticles was performed by using different techniques. For instance, laser diffraction spectrometry using a Nano-ZS Zetasizer was used to determine the particles size, and Zeta potential. High-resolution transmission electron microscopy (HR-TEM) were utilized to evaluate Particle sizes and morphologies of the prepared nanoparticles. Additionally, the chemical structure of the prepared nanoparticles was assessed by X-ray diffraction (XRD) and Fourier transform infrared spectroscopy (FTIR). The effects of the different concentrations of GNPs and GO nanosheets on cell viability and proliferation were evaluated by WST-1 cell proliferation assays. Ultimately, in vitro studies were performed by confocal microscopy to characterize the cellular uptake of the prepared nanoparticles. The net results of this study suggest that both nanomaterials showed a cytoplasmic entry without penetrating the nucleus. They may be useful for slow drug release as well as photothermal and photodynamic therapy using a low-powered laser or IR radiation.
Article
The development and synthesis of hydrogels for chemical and biosensing are of great value. Hydrogels can be tailored to its own physical structure, chemical properties, biocompatibility and sensitivity to external stimuli when being used in a specific environment. This review mainly covers hydrogels and their applications in chemical and biosensing. In particular, the review focuses on the manner in which hydrogels serve as sensing materials to a specific analyte. Different types of responsive hydrogels are hence introduced and summarized. Researchers can modify different chemical groups on the skeleton of the hydrogels, which make them as good chemical and biosensing materials. Hydrogels have great application potential for chemical and biosensing in the biomedical field and some emerging fields, such as wearable devices. This article is protected by copyright. All rights reserved
Chapter
Hydrogels are highly hydrated three dimensional networks with the ability to mimic the extracellular matrix of bodily tissues and have thus found application in a wide range of biomedical applications. Unique physiochemical properties such as biocompatibility, water permeability, stimuli responsiveness and self-healing characteristics make them especially useful for use as scaffolds and matrices drug delivery, tissue engineering/regeneration and sensing. Their weak and brittle nature, however, often limits their widespread application where improved mechanical strength is required. To resolve this problem, there has been a significant amount of research into the improvement of their mechanical properties. Among these efforts, versatile multicomponent hydrogels have received much attention as their physiochemical properties can be structurally engineered to provide a wide range of desired properties. These multicomponent formulations also allow for the combination of natural and synthetic polymers, which offers the scope to exploit the advantages of each component, with the synergistic effects resulting from mutual interactions. This book critically discusses the fundamental chemistry, synthesis, characterisation, physiochemical and biological properties of various types of multicomponent hydrogels. It reviews the different strategies employed in designing and synthesizing cutting-edge multicomponent hydrogels and their key applications in biomedical fields. The work is suitable for researchers working in the specific area of multicomponent hydrogels, and also more generally for those working in materials science, biomedical engineering, biomaterials science and tissue engineering.
Article
Microparticles hydrogel sensitive to reduction, pH and radiation composed of starch (St) and MnO2 NPs were synthesized, and loaded with Dox for in vitro treatment of MCF-7 cells. LE and EE values were obtained as 3.12 and 93.4%, respectively. In vitro drug release study revealed pH- and reduction-responsiveness drug release behavior of the DDS. The intracellular uptake of the St/MnO2 microparticles was verified through FITC dye conjugation. The St-MnO2/Dox microparticles exhibited higher anticancer activity than free Dox after 48 h as verified by MTT and DAPI staining assays. As results, combined chemoradiotherapy approach exhibited synergistic outcomes than chemotherapy alone.
Article
Hydrogen peroxide (H2O2)/glutathione (GSH) dual-sensitive nanoplatform holds great promise to alleviate the side effects of chemo drugs and improve the therapeutic efficacy against cancer. The site-specific release of chemotherapeutic drug with low premature release still remains a challenge in the field of chemotherapy. Herein, a novel and multifunctional drug delivery system (DDS) based on polymethylacrylic acid core with cross-linked structure of disulfide bond (PMAABACy), metal-organic framework (MOF) interlayer and biologically inspired polydopamine (PDA) coating was developed, serving as a vehicle for on-demand drug release. The dual-responsive nanoplatform not only prevents the premature leakage of chemotherapeutic drug but also is sensitive to biologically relevant GSH and H2O2 for the precise delivery of chemotherapeutic drug. Considering the transmission route to DDS at the tumor site, the DDS might first respond to the extracellular H2O2 and then to the intracellular GSH, exhibiting a tunable release of chemotherapeutic drug. Through incubation using tumor cells, the growth of tumor cells could be significantly inhibited. Overall, by integrating these different building modules, this research demonstrates the advantages of MOF-assisted regulate strategy to DDS for precise site-specific release against tumor cells with much reduced side-effect on normal tissues.
Article
The chemo-photothermal therapy performance of a novel theranostic nanoparticles that fabricated through the conjugation of HS-poly(ε-caprolactone)-block-poly(N-isopropylacrylamide)-block-poly(acrylic acid) (HS-PCL-b-PNIPAAm-b-PAA) and gold nanoparticles (GNPs) was extensively investigated. The GNPs@polymer conjugate theranostic NPs was loaded with doxorubicin hydrochloride (DOX) as an anticancer drug through electrostatic interactions to afford GNPs@polymer-DOX theranostic nanomedicine. Temperature and pH-triggered in vitro drug release behavior of the developed theranostic nanomedicine were also examined. The biocompatibility of the synthesized GNPs@polymer theranostic NPs was confirmed through the assessing survival rate of breast cancer cell line (MCF7) using MTT assay. In vitro cytotoxic effects of the GNPs@polymer-DOX theranostic nanomedicine was also evaluated against MCF7 cells in both with or without laser irradiation (532 nm, 145 mJ per pulse, 5 min) conditions, and the results obtained were compared with free DOX as the reference. As the results, the developed GNPs@polymer-DOX can be considered as theranostic nanomedicine for chemo-photothermal therapy of solid tumors.
Article
The aim of this study was the design and development of a novel de novo drug delivery system for cancer chemotherapy. For this purpose, chitosan (CS) functionalized using phthalic anhydride followed by 4-cyano, 4-[(phenylcarbothioyl) sulfanyl] pentanoic acid as a chain transfer agent (CTA) to afford CS-CTA macroinitiator. The synthesized CS-CTA macroinitiator was then copolymerized with methacrylic acid (MAA) monomer using reversible addition-fragmentation chain transfer (RAFT) polymerization technique to produce chitosan-graft-poly(methacrylic acid) (CS-g-PMAA) graft copolymer. Afterward, graphene oxide (GO) nanosheets were incorporated into the synthesized copolymer through the physical interactions. The CS-g-PMAA/GO nanocomposite was loaded with doxorubicin hydrochloride (DOX) as a universal anticancer drug. The biocompatibility, DOX-loading capacity, and pH dependent drug release behavior of the developed nanocomposite were also investigated. As the experimental results, as well as superior biological and physicochemical features of CS and GO, we envision that the developed CS-g-PMAA/GO nanocomposite may be applied as de novo drug delivery nanosystem for cancer chemotherapy.
Article
In the present work, lipid nanocapsules (LNC) for curcumin (CCM) encapsulation have been developed and optimized. The objective was to increase drug cytotoxicity on 9L glioma cells and drug bioavailability following intravenous administration (IV). Using the phase inversion technique, we obtained 50 nm LNC loaded with CCM (4 and 6 mg/mL) and, due to the hydrophobic nature of the drug, the encapsulation efficiency was very high, being around 90%. Following 48 h of incubation with 9L cells, CCM-loaded LNC were able to reduce the viability of glioma cells resulting in significant twofold lower IC50 in comparison with the free drug solution. Moreover, CCM-loaded LNC induced both the apoptosis of 9L cells and a strong release of ATP. This suggests a cellular uptake of the LNC and an enhanced anti-proliferative effect. In order to evaluate any alteration in the pharmacokinetic behavior of the encapsulated drug, CCM-loaded LNC were injected IV into healthy rats, at a dose of 10 mg/kg. CCM pharmacokinetic studies were carried out quantifying the CCM concentration from the blood of rats, receiving either CCM-loaded LNC or free CCM solution as a control. The results demonstrated that loaded LNC exhibited a significantly higher AUC, Cmax and t1/2 in comparison with the control, while the clearance was strongly reduced. Globally, these results encouraged the use of CCM-loaded LNC to enhance the in vivo therapeutic activity of the drug after systemic administration.
Article
Herein, we report on the preparation of ultra-low sized (<100 nm in diameter) biodegradable polymeric capsules for potential applications as nanocontainers in antibiotic therapy. Hollow nanospheres based on the chitosan/poly(acrylic acid) pair are elaborated via (i) the layer-by-layer technique using gold nanoparticles (20 and 60 nm in size) as sacrificial templates, (ii) loading with amoxicillin, a betalactam antibiotic, and (iii) removal of the gold core via cyanide-assisted hydrolysis. Size, dispersity and concentration of the resulting nanocapsules are easily tuned by the nanoparticle templates, while wall thickness is controlled by the number of polyelectrolyte bilayers. Electrostatic interactions between the protonated amine groups of chitosan and the carboxyl groups of poly(acrylic acid) act as the driving attraction force allowing easy and fast design of robust and well-ordered multilayer films. Successful hydrolysis of the gold core is evidenced by time-dependent monitoring of the gold spectroscopic signature (absorbance at 519 nm and 539 nm for the gold nanoparticles with 20 and 60 nm, respectively). Crosslinked capsules are also prepared through crosslinking of the chitosan chains with glutaraldehyde. Chitosan-based nanocapsules are finally evidenced to be promising drug delivery vehicles of amoxicillin trihydrate with tuneable properties such as entrapment efficiency in the range of 62-75% and 3.5-5.5% concerning the drug loading.
Article
This article evaluates physicochemical, mechanical, and biological properties of a series of novel dental nanocomposites that fabricated from multifunctional methacrylate-based dental monomers, triethyleneglycol dimethacrylate (TEGDMA) monomer, and modified silica nanoparticles (SiO2 NPs). The antibacterial activities of the monomers were investigated against lactobacillus plantarum by standard agar disk diffusion method. The cytotoxicity characteristics of the monomers and fabricated nanocomposites were evaluated by MTT and trypan blue cell viability tests, respectively against NIH3T3 cell line. In addition, the mechanical properties, as well as physicochemical characteristics including water sorption, sol fraction, and double bond conversion were also investigated. According to the results, the formulated nanocomposites have potential to apply as dental nanocomposites mainly due to their acceptable physicochemical, mechanical and biological characteristics.
Article
This article evaluates the anticancer drug delivery performances of two nanohydrogels composed of poly(N-isopropylacrylamide-co-itaconic anhydride) [P(NIPAAm-co-IA)], poly(ethylene glycol) (PEG), and Fe3O4 nanoparticles. For this purpose, the magnetite nanohydrogels (MNHGs) were loaded with doxorubicin hydrochloride (DOX) as a universal anticancer drug. The morphologies and magnetic properties of the DOX-loaded MNHGs were investigated using transmission electron microscopy (TEM) and vibrating–sample magnetometer (VSM), respectively. The sizes and zeta potentials (ξ) of the MNHGs and their corresponding DOX-loaded nanosystems were also investigated. The DOX-loaded MNHGs showed the highest drug release values at condition of 41°C and pH 5.3. The drug-loaded MNHGs at physiological condition (pH 7.4 and 37°C) exhibited negligible drug release values. In vitro cytotoxic effects of the DOX-loaded MNHGs were extensively evaluated through the assessing survival rate of HeLa cells using the MTT assay, and their in vitro cellular uptake into the mentioned cell line were examined using fluorescent microscopy and fluorescence-activated cell sorting (FACS) flow cytometry analyses. As the results, the DOX-loaded MNHG1 exhibited higher anticancer drug delivery performance in the terms of cytotoxic effect and in vitro cellular uptake. Thus, the developed MNHG1 can be considered as a promising de novo drug delivery system, in part due to its pH and thermal responsive drug release behavior as well as proper magnetite character toward targeted drug delivery.
Article
A novel GON-based drug delivery system containing graphene oxide nanoparticles (GON) as carriers of anticancer drugs and chitosan/dimethylmaleic anhydride-modified chitosan (CS/CS-DMMA) as surface charge-reversible shells is fabricated via the classic self-assembly of the deprotonated carboxyl of graphene oxide nanoparticles (GON) and the protonated amine of CS backbone by electrostatic interaction, and the CS-DMMA serves as outmost layer. In this GON-based drug delivery system, the GON cores as desired carriers might adsorb doxorubicin hydrochloride (DOX) via the π−π stacking interaction between the large π conjugated structures of GO and the aromatic structure of DOX. Meanwhile, the chitosan-based polyelectrolyte shells served as smart protection screen to evade the premature release of as-loaded DOX in normal extracellular condition, and then the release of DOX was accelerated due to the detachment of chitosan coating at low pH. Furthermore, the re-exposure of amino groups after hydrolysis of CS-DMMA endowed the drug delivery system with positive surface charge by taking advantage of the pH difference between physiological condition and tumor microenvironment to enhance the cellular uptake. And then pH-dependent site-specific drug release was realized. The in vitro investigations confirmed that this promising GON/CS/CS-DMMA hybrids with charge reversible character possessed various merits including excellent encapsulation efficiency, high stability at physiological condition, enhanced cellular uptake by HepG2 cells and tunable intracellular chemotherapeutic agent release profiles, proving its capability as an intelligent anticancer agent nanocarrier with enhanced therapeutic effects. This smart GON/CS/CS-DMMA vehicle with surface charge reversible character may be used as a significant drug delivery system for cancer treatment.
Article
Multifunctional capsules have great applications in biomedical fields. In this study, novel polysaccharide-based nanocapsules were prepared via a layer-by-layer technique using silica as the templates. The shell was constructed based on the electrostatic interactions between pectin and chitosan. The pectin-chitosan nanocapsules ((Pec/Cs)3Pec) could keep good colloidal stability within 96 h in PBS solution and 48 h in BSA solution. Meanwhile, the nanocapsules exhibited a high drug loading and pH-sensitive release property for doxorubicin hydrochloride. Moreover, (Pec/Cs)3Pec nanocapsules had no cytotoxicity to both human hepatocellular carcinoma cells (HepG2 cells) and mouse fibroblast cells (L929 cells). More importantly, (Pec/Cs)3Pec nanocapsules could be more easily uptaken by HepG2 cells when compared with L929 cells. In vitro anticancer activity tests indicated the carriers could effectively kill HepG2 cells. Overall, (Pec/Cs)3Pec nanocapsules have great potential as a novel anticancer drug carrier as a result of their pH-sensitivity, good colloidal stability and anticancer activity.