ArticlePDF Available

A novel pH‐responsive nanoniosomal emulsion for sustained release of curcumin from a chitosan‐based nanocarrier: Emphasis on the concurrent improvement of loading, sustained release, and apoptosis induction

Wiley
Biotechnology Progress
Authors:

Abstract and Figures

Curcumin application as an anti‐cancer drug is faced with several impediments. This study has developed a platform that facilitates the sustained release of curcumin, improves loading efficiency, and anti‐cancer activity. Montmorillonite (MMT) nanoparticles were added to chitosan (CS)‐agarose (Aga) hydrogel and then loaded with curcumin (Cur) to prepare a curcumin‐loaded nanocomposite hydrogel. The loading capacity increased from 63% to 76% by adding MMT nanoparticles to a chitosan‐agarose hydrogel. Loading the fabricated nanocomposite in the nanoniosomal emulsion resulted in sustained release of curcumin under acidic conditions. Release kinetics analysis showed diffusion and erosion are the dominant release mechanisms, indicating non‐fickian (or anomalous) transport based on the Korsmeyer‐Peppas model. FTIR spectra confirmed that all nanocomposite components were present in the fabricated nanocomposite. Besides, XRD results corroborated the amorphous structure of the prepared nanocomposite. Zeta potential results corroborated the stability of the fabricated nanocarrier. Cytotoxicity of the prepared CS‐Aga‐MMT‐Cur on MCF‐7 cells was comparable with that of curcumin‐treated cells (p < 0.001). Moreover, the percentage of apoptotic cells increased due to the enhanced release profile resulting from the addition of MMT to the hydrogel and the incorporation of the fabricated nanocomposite into the nanoniosomal emulsion. To recapitulate, the current delivery platform improved loading, sustained release, and curcumin anti‐cancer effect. Hence, this platform could be a potential candidate to mitigate cancer therapy restrictions with curcumin.
This content is subject to copyright. Terms and conditions apply.
RESEARCH ARTICLE
Formulation and Engineering of Biomaterials
A novel pH-responsive nanoniosomal emulsion for sustained
release of curcumin from a chitosan-based nanocarrier:
Emphasis on the concurrent improvement of loading, sustained
release, and apoptosis induction
Shabnam Haseli
1
| Mehrab Pourmadadi
1
| Amirmasoud Samadi
1
|
Fatemeh Yazdian
2
| Majid Abdouss
3
| Hamid Rashedi
1
| Mona Navaei-Nigjeh
4,5
1
Department of Biotechnology, School of
Chemical Engineering, College of Engineering,
University of Tehran, Tehran, Iran
2
Department of Life Science Engineering,
Faculty of New Science and Technologies,
University of Tehran, Tehran, Iran
3
Department of Chemistry, Amirkabir
University of Technology, Tehran, Iran
4
Pharmaceutical Sciences Research Center,
The Institute of Pharmaceutical Sciences
(TIPS), Tehran University of Medical Sciences,
Tehran, Iran
5
Department of Pharmaceutical Biomaterials
and Medical Biomaterials Research Center,
Faculty of Pharmacy, Tehran University of
Medical Sciences (TUMS), Tehran, Iran
Correspondence
Fatemeh Yazdian, Department of Life Science
Engineering, Faculty of New Science and
Technologies, University of Tehran, Tehran,
Iran.
Email: yazdian@ut.ac.ir
Hamid Rashedi, Department of Biotechnology,
School of Chemical Engineering, College of
Engineering, University of Tehran, Tehran,
Iran.
Email: hrashedi@ut.ac.ir
Abstract
Curcumin application as an anti-cancer drug is faced with several impediments. This
study has developed a platform that facilitates the sustained release of curcumin,
improves loading efficiency, and anti-cancer activity. Montmorillonite (MMT) nano-
particles were added to chitosan (CS)-agarose (Aga) hydrogel and then loaded with
curcumin (Cur) to prepare a curcumin-loaded nanocomposite hydrogel. The loading
capacity increased from 63% to 76% by adding MMT nanoparticles to a chitosan-
agarose hydrogel. Loading the fabricated nanocomposite in the nanoniosomal emul-
sion resulted in sustained release of curcumin under acidic conditions. Release kinet-
ics analysis showed diffusion and erosion are the dominant release mechanisms,
indicating non-fickian (or anomalous) transport based on the Korsmeyer-Peppas
model. FTIR spectra confirmed that all nanocomposite components were present in
the fabricated nanocomposite. Besides, XRD results corroborated the amorphous
structure of the prepared nanocomposite. Zeta potential results corroborated the
stability of the fabricated nanocarrier. Cytotoxicity of the prepared CS-Aga-MMT-
Cur on MCF-7 cells was comparable with that of curcumin-treated cells (p< 0.001).
Moreover, the percentage of apoptotic cells increased due to the enhanced release
profile resulting from the addition of MMT to the hydrogel and the incorporation of
the fabricated nanocomposite into the nanoniosomal emulsion. To recapitulate, the
current delivery platform improved loading, sustained release, and curcumin anti-
cancer effect. Hence, this platform could be a potential candidate to mitigate cancer
therapy restrictions with curcumin.
KEYWORDS
cancer therapy, chitosan, curcumin, montmorillonite, niosome, pH-sensitive nanocarrier
1|INTRODUCTION
During the last decade, cancer has skyrocketed alarmingly fast as the
world's second cause of fatality across the globe.
1
It is estimated that
there are 1.8 million new diagnosed cases of cancer, with over
600 thousand deaths in the United States alone.
2,3
It is approximated
that breast cancer makes up 30% of all cancers diagnosed in the
United States; the most prevalent cancer among women after skin
Received: 11 April 2022 Revised: 30 April 2022 Accepted: 11 May 2022
DOI: 10.1002/btpr.3280
Biotechnol. Prog. 2022;e3280. wileyonlinelibrary.com/journal/btpr © 2022 American Institute of Chemical Engineers. 1of18
https://doi.org/10.1002/btpr.3280
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
cancer.
4
Multiple cancer treatments, including chemotherapy, have
been tried; however, several impediments, including toxicity to
healthy cells and cardiac tissues, multiple drug resistance (MDR),
non-specific distribution, and destructive damages on normal tissues
hamper the effective treatment of cancer with chemotherapy.
5,6
Therefore, safer anti-cancer drugs, and efficient, targeted delivery
methods are needed. In this direction, taking advantage of the anti-
cancer properties of naturally derived polyphenols such as curcumin
and delivering it via stimuli-responsive delivery systems has opened
up a new avenue for cancer therapy.
7,8
Curcumin (Cur), a plant-derived flavonoid polyphenol, is one of the
main constituents of food and beverages.
9
Curcumin has several fea-
tures, involving anti-inflammatory,
9
antiviral,
10
anti-Leishmanial
activity,
11
and anti-cancer properties through controlling tumor progres-
sion by increasing reactive oxygen species (ROS) after targeting ROS
metabolic pathway enzymes.
12
Several physiological activities have been
ascribed to curcumin for controlling tumor progression, including inhibi-
tion of the WNT/β-catenin pathway and pyruvate kinase M2
(PKM2).
13,14
Curcumin is also utilized as a chemosensitizer to reduce
cancer cells' multidrug resistance by blocking the nuclear factor NF-κB.
15
The mentioned attributes render curcumin a promising compound
for cancer treatment; however, low solubility(1μg.ml
1
), instability,
short biological half-life, and poor permeability are among the striking
negative aspects that impede employing anti-cancer properties of curcu-
min.
16,17
The aforementioned deficiencies of curcumin for cancer therapy
can be mitigated with the fabrication of hydrogels using natural polymers
as a promising approach for employing hydrogels in drug delivery applica-
tions.
18,19
In this study, the hydrogel is prepared by chitosan and agarose.
Chitosan (CS) is a biocompatible biopolymer comprised of
β-linked D-glucosamine and N-acetyl-D-glucosamine. Enhancing pro-
longed release and drug penetration, low toxicity, and structural vari-
ability are some of the properties that render chitosan a desirable
natural polymer for hydrogel fabrication in biomedical applications,
including drug delivery.
2022
As well, chitosan amine groups proton-
ation at the low pH of cancer cells leads to the pH-sensitive release of
loaded drugs in hydrogel; on the other hand, chitosan retains its gel-
like structure at pH 7.4. Indeed, this pH-sensitive property of chitosan
renders the fabricated hydrogel pH-responsive and prevents the
release of drugs under neutral conditions before reaching tumor
cells.
23
For instance, Díaz-Zepeda et al. coated microemulsions con-
taining curcumin in the oil phase with chitosan to obtain a pH-
sensitive platform for curcumin delivery.
24
Also, due to chitosan's
semi-crystalline structure and high hydrophilicity, it forms hydrogen
bonds with other compounds within the hydrogel network, includ-
ing agarose. Agarose (Aga) is a porous polysaccharide with d-galac-
tose-3,6-anhydro-l-galactopyranose as building blocks.
25
Contrary
to PEG and alginate, which are prone to reactive chemistry and lack
of effectuation capability, agarose physical crosslinking presents an
attractive property for hydrogel preparation.
26
Agarose-based bio-
materials, e.g., hydrogels, can be a matrix for the incorporation of
pH-sensitive compounds like diblock copolymers and chitosan to
produce pH-sensitive delivery platforms.
21,27
Indeed, agarose has
been employed as a matrix to produce pH-sensitive carriers.
28,29
Consequently, agarose is suitable for fabricating controlled stimuli-
responsive drug delivery systems.
The lack of multiresponsiveness and low loading efficiency of cur-
cumin due to hydrogels' hydrophilic nature are among the major factors
limiting their use in curcumin delivery as a hydrophobic drug. To
address these shortcomings, nanoparticles are incorporated into hydro-
gels to prepare nanocomposite hydrogels.
30,31
Nanoparticles' unique
properties, including high surface area, controlled shape, reduced parti-
cle size, and the potential to be loaded with hydrophobic molecules,
render their incorporation into hydrogels an apt solution to extend the
capacity of hydrogels for the delivery of a broader range of therapeu-
tics, including hydrophobic anti-cancer medicines.
32,33
In this vein,
montmorillonite (MMT), a common clay mineral, which exhibits biocom-
patibility, biodegradability, and good mechanical properties,
34
was
incorporated into the prepared CS-Aga hydrogel. As stated for nano-
particles, MMT incorporation as a biocompatible compound in the
CS-Aga hydrogel can enhance the loading capacity of hydrogel, homo-
geneity of the hydrogel, and burst release control.
35,36
This potential of
MMT is mainly ascribed to the presence of siloxane (SiOSi), which
plays a pivotal role in interaction with polymers in the hydrogel struc-
ture and drugs loaded in hydrogels.
37
Hydroxyl groups in agarose and
curcumin can create hydrogen bonds with the oxygen of the SiOSi
functional group of MMT, causing a high affinity for curcumin.
38,39
These interactions between components of the nanocomposite hydro-
gel maintain nanocomposite integrity under neutral conditions.
Anti-cancer drug delivery platforms that respond to pH are
among the most popular stimuli-responsive drug delivery options.
40,41
The change in the pH of cancer cells during their proliferation is one
of the main reasons for this popularity.
42,43
This reduction in pH,
known as the Warburg effect, is a hallmark in cancer and has led to
applying pH-sensitive drug delivery platforms like hydrogels for the
controlled delivery of anti-cancer therapeutics.
44,45
As mentioned before, a hydrogel of CS-Aga was prepared in this
study. MMT nanoparticles were embedded in this hydrogel to pro-
duce a CS-Aga-MMT nanocomposite hydrogel. Hydroxyl groups in
agarose provided gelling property in the hydrogel, known as physical
crosslinking property. Besides, the H-atoms of the CH group of chit-
osan and its amine groups, agarose and curcumin hydroxyl groups,
and the oxygen atoms of the siloxane group of MMT can form hydro-
gen bonds.
37
Indeed, MMT nanoparticles were non-covalently incor-
porated into the hydrogel before the addition of a crosslinking agent.
Then, curcumin was added to the nanocomposite hydrogel, which led
to hydrogen bonding between MMT and curcumin.
38
Afterwards,
glyoxal was used as a crosslinker to further entrap the drug inside the
CS-Aga-MMT nanocomposite hydrogel by forming covalent bonds in
the hydrogel 3D network. The prepared hybrid hydrogel includes
chitosan-chitosan, chitosan-agarose, and agarose-agarose crosslinks
using glyoxal. Due to these interactions and the high surface area pro-
vided by MMT nanoparticles, curcumin loading is improved as a
hydrophobic compound in the fabricated nanocomposite hydrogel.
Furthermore, the fabricated nanocomposite has pH-responsive prop-
erty due to the alterations in the structure of nanocomposite com-
pounds with the change in pH. Indeed, Al and Si of MMT dissolve
2of18 HASELI ET AL.
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
faster under acidic conditions as the pH decreases.
46
Also, amine
groups in chitosan, hydroxyl groups in agarose and curcumin are pro-
tonated in acidic media. The release of Al and Si ions along with
protonation in the mentioned groups provides repulsive forces
between nanocomposite components. These repulsive forces lead to
the disintegration of the nanocomposite and faster release in an acidic
environment; however, the interactions between the drug and
components preserve the nanocomposite structure under neutral con-
ditions. The nanocomposite can release the drug at the tumor location
with a reduced pH by retaining the drug under neutral conditions.
Since the controlled release of curcumin and prolonging the release
time to enhance the apoptosis induction of curcumin was a major
objective of this study, the curcumin-loaded nanocomposite hydrogel
was then encapsulated in a nanoniosomal emulsion. The nanoniosomal
emulsion is a bilayer vesicle produced by a non-ionic surfactant and an
oil phase as per the steps in the literature.
47,48
The prolonged release
by encapsulating the nanocomposite in the niosome is due to separat-
ing the aqueous phase with an oil phase. Indeed, the prolonged release
is achieved since the oil layer separates the internal and external aque-
ous phases. The oil layer functions like a liquid membrane, reducing cur-
cumin release from the internal aqueous phase.
49
Comparing the
current study with our previous research reveals that loading the drug-
loaded nanocomposite in the nanoniosome prolongs the release.
50
Sev-
eral attributes make niosomes suitable platforms for curcumin delivery,
including high stability and cheaper cost of production.
51
The nanoniosomal emulsion is based on a water in oil emulsion
that contains the drug-loaded nanocomposite in its aqueous phase.
Span 80 is used as a non-ionic surfactant to form the first layer of the
vesicle. Then, the formed vesicle is added to the water phase to form
niosomes as double-layer vesicles. The water phase also washes the
free unbound drug from niosomes.
Hence, by incorporating MMT nanoparticles in the pH-responsive
CS-Aga hydrogel, curcumin loading was improved. Then, CS-Aga-MMT
nanocomposite hydrogel containing curcumin was encapsulated in a
niosomal emulsion to enhance the prolonged release of curcumin from
the nanocomposite. We inspected the in vitro antitumor activity of the
fabricated drug delivery system on the MCF-7 cells as a way to examine
the potential of this platform for cancer treatment. As far as we know,
this is an original method to ameliorate predominant restrictions regard-
ing cancer therapy with curcumin. This approach has the potential to
concurrently improve the loading capacity and stimuli-responsive
release at the target site within a prolonged period. Moreover, the study
of curcumin delivery by this platform demonstrated enhanced apoptosis
induction of curcumin. These results represent that the common draw-
backs of using curcumin as an anti-cancer drug, including poor loading
and low solubility, have been addressed to a good extent; thus, the use
of this platform may expand to treat other cancer cells as well.
2|MATERIALS
MCF-7 breast cancer cells were taken from the American Type Cul-
ture Collection (ATCC) (Manassas, VA). Culture flasks and dishes were
gained from Corning (Corning, NY, USA). Fetal bovine serum (FBS)
was purchased from Gibco-Life Technologies. Dulbecco's Modified
Eagle's Medium (DMEM) High Glucose was obtained from Thermo
Fisher Scientific (US). Penicillin/streptomycin and 0.25% (w/v)
trypsin0.1% (w/v) ethylenediaminetetraacetic acid (EDTA) were pro-
cured from Thermo Fisher Scientific (US). 3-(4, 5-Dimethyl-thiazol-
2-yl)-2, 5-diphenyl-tetrazolium bromide (MTT), and dimethylsulfoxide
(DMSO) were bought from Merck. Phosphate buffer saline (PBS) was
purchased from Sigma-Aldrich. Chitosan (CS), agarose (Aga), montmo-
rillonite (MMT), and curcumin (Cur) ((1E,6E)-1,7-bis(4-hydroxy-
3-methoxyphenyl)-1,6-heptadiene-3,5-dione) were obtained from
Sigma. Span 80 and olive oil were purchased from Sigma-Aldrich.
Glyoxal was bought from Merck.
3|METHODS
3.1 |Synthesis of CS-Aga-MMT hydrogel
At first, 30 ml of acetic acid 2% v/v was prepared to slowly add chito-
san (CS) to it at 25C with magnetic stirring (500 rpm) and prepare a
2% w/v solution. Then, agarose (1% w/v) was mixed with the former
2% w/v solution and intensely stirred. After the preparation of the
CS-Aga hydrogel, montmorillonite (MMT) was added under stirring
with 0.1% w/v. The presence of MMT in the CS-Aga hydrogel matrix
leads to physical crosslinking and the establishment of hydrogen
bonds in the nanocomposite hydrogel. These interactions result in the
enhancement of curcumin loading in the nanocomposite.
32,38
3.2 |Curcumin loading in the fabricated hydrogel
Firstly, curcumin was added to ethanol 96% v/v to dissolve it since it is
a hydrophobic compound. Then, the dissolved curcumin was incorpo-
rated into the CS-Aga-MMT nanocomposite. The model drug, curcu-
min, was added to the hydrogel with 10 μg.ml
1
concentration. The
addition of curcumin was done gradually while intensely stirred. The
nanocomposite was crosslinked once curcumin was added using glyoxal
(0.1% v/v). Crosslinking ensues from chitosan amine groups binding
together in addition to interaction with agarose hydroxyl groups. Also,
hydroxyl functional groups in agarose cause crosslinking between Aga
and Aga using glyoxal. The schematics of crosslinks between polymers
are depicted in Figure 1. A portion of the prepared nanocomposite
hydrogels with/without curcumin was lyophilized for characterization
study and loading/entrapment efficiency determination.
3.3 |Preparing niosomal emulsion
The primary emulsion is a water in oil (W/O) emulsion produced by
emulsifying the water phase containing the drug-loaded hydrogel.
Briefly, 10 ml of the prepared hydrogel was added little by little to
olive oil and span 80 as an emulsifier to prepare the first layer of
HASELI ET AL.3of18
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
FIGURE 1 Schematics of crosslinking between (a) chitosan and chitosan, (b) chitosan and agarose, and (c) agarose and agarose using glyoxal
as a crosslinker
4of18 HASELI ET AL.
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
niosomes. Then, the water phase was used to produce the second
layer and form the bi-layer vesicles, called niosomes. The hydrophobic
phase was removed from the top of the suspension by a laboratory
sampler due to the affinity of the external aqueous phase of the fabri-
cated niosomal emulsion to the hydrophilic (water) phase. The remain-
ing hydrophilic phase, which includes the fabricated niosomes, was
centrifuged at 6000 rpm for 8 min. The nanocomposite fabrication
and loading in niosomes are depicted in the graphical abstract
(Created with BioRender.com).
3.4 |Measurement of curcumin loading and
encapsulation efficiency
Curcumin loading and encapsulation (entrapment) capacity was speci-
fied to assess the effect of MMT on curcumin loading efficiency in
CS-Aga hydrogel. Lyophilized nanocarriers of CS-Aga (1 mg) and CS-
Aga-MMT loaded with curcumin (1 mg) were dispersed in 1 ml PBS,
then added 1 ml ethyl acetate. Separation of ethyl acetate was done
after stirring the mixtures. The free content of curcumin (Cur) in the
ethyl acetate phase was measured by a UVVis spectrophotometer at
419 nm.
52
The tests were performed in triplicate. Curcumin encapsu-
lation and loading efficiency were determined using Equations 1 and
2,
52,53
respectively.
3.5 |Characterization of the prepared delivery
system
A Nanotrac wave analyzer (Microtrac, Osaka, Japan) was used to
determine nanocarriers' average size and surface charge by dynamic
light scattering (DLS) and zeta potential measurement, respectively.
The surface morphology of the nanocomposites was studied by Field
Emission Scanning Electron Microscope (FESEM) (TESCAN, MIRA III
model, Czech Republic) at an accelerated voltage of 30 kV. The exper-
iments were done at a scattering angle of 90after probe sonication
of the solution. FTIR was used to verify the presence of chitosan, aga-
rose, montmorillonite, and curcumin in the fabricated platform by
observing the change in FTIR spectra after adding each component.
The spectroscopy was done at 25C by a KBr pellet. A pellet of thick-
ness 0.1 cm was made by grinding 1 mg of each sample in a mortar
pestle with KBr. KBr pellet was used as blank. The range of scanning
was 600 to 4000 cm
1
. The spectra of CS, CS-Aga, CS-Aga-MMT,
and CS-Aga-MMT-Cur were recorded on freeze-dried samples using
a PerkinElmer Spectrum Version 10.03.06. The changes in the crys-
tallinity of samples were examined to verify the interactions
between chitosan, agarose, nanoparticles and curcumin, which lead
to covering the crystalline peaks of curcumin and forming an amor-
phous phase. The analysis was done using an X-ray diffractometer
(XRD-6100, Shimadzu, Japan) in the range of 102θ80using a
Cu Kαsource with λ=1.54056. The step size was 0.05 degrees with
a 1-s interval between each step. XRD patterns were analyzed by
HighScore plus 3.
3.6 |Drug release analysis
Curcumin release from nanoniosomal emulsions was inspected by the
dialysis method at pH 7.4 and 5.4 to evaluate the pH-sensitive feature
of the fabricated nanocarrier and curcumin controlled-release.
54
One milliliter of the nanoniosomal solution was added into a dialysis
bag (Mw cutoff 12,000 g mol
1
) and immersed in 60 ml of PBS con-
taining ethanol 20% v/v.
At particular time points of 0, 12, 24, 48, 72, 96, and 120 hours,
300 μl of samples were elicited and substituted with the same volume
of fresh PBS. The released curcumin was measured by UVVis spec-
trophotometric analysis of the elicited buffer (Thermo Biomate
5, Thermo Scientific, US) at 419 nm. The above procedure was done
three times. Curcumin release percentage was calculated using
Equation 3:
Curcumin released %ðÞ¼Curcumin½rel
Curcumin½load 100 ð3Þ
In the above relation, [Curcumin]
load
is curcumin loaded in the
hydrogel, and [Curcumin]
rel
is curcumin released from the nanonio-
somes. The statistical analysis (ANOVA) was done based on curcumin
release percentage from CS-Aga-MMT-Cur at pH 5.4 and curcumin
release from all other groups to corroborate the pH sensitivity of the
nanocarrier.
Encapsulation Efficiency %
ðÞ
¼ðCurcumin initial amountÞðFree Curcumin in ethyl acetateÞ
ðInitial CurcuminÞð1Þ
Loading Efficiency %
ðÞ
¼ðCurcumin initial amountÞðFree Curcumin in ethyl acetateÞ
ðNanocomposite total amountÞð2Þ
HASELI ET AL.5of18
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
3.7 |Mathematical models of curcumin release
Curcumin release results were fitted to drug release models to deter-
mine the release mechanism in neutral and acidic conditions. The
zero-order, first-order, Higuchi, HixsonCrowell, and Korsmeyer
Peppas models were used for mathematic modeling. The fitting was
performed with GraphPad 7. The R squares of the above models were
calculated, and according to the best-fitting regression parameters,
the release mechanism was specified at pH 7.4 and 5.4. After fitting,
factors without statistical meaning based on a p-value less than 0.05
were expunged from models. Also, the drug release data below 60%
of the cumulative drug release were used for the KorsmeyerPeppas
model based on the instructions for this model.
55
3.8 |Cell Culture
DMEM media containing 10% fetal bovine serum (FBS), penicillin
(100 U.ml
1
) and streptomycin (100 μg.ml
1
) were used to culture
cells at 37C in a humidified incubator with 5% CO
2
.
3.9 |In vitro cytotoxicity assay
The cytotoxicity of curcumin, CS-Aga-MMT nanocomposite loaded
with curcumin (CS-AGA-MMT-Cur), and nanoniosomes containing
CS-Aga-MMT without curcumin on MCF-7 cell line were studied
using MTT assay. A growth medium of 2 10
4
MCF-7 cells was
added to wells of a 96-well plate and incubated for one night to
achieve cell adherence. After 24 h, with 70% confluency of cells
attached to the plate, the samples at 6.4 μg.ml
1
, with regard to the
loading of curcumin (64%) in the prepared hydrogel, were incubated
with the cells to evaluate each sample's cytotoxicity. Selecting this
concentration leads to comparing equal concentrations of the samples
to study the impact of the prepared platform on cytotoxicity via
changing the release profile. The MTT assay was conducted as in the
former study.
29
The optical density of samples was read using an
ELISA reader at 570 nm to determine the viability of samples com-
pared with the control group's viability put on 100%. The mean and
the standard error of the mean (SEM) of cell viability for each group
were specified. GraphPad InStat 3 was used for the statistical analysis
to compare the means, standard errors, and the number of replicates
(N=3) between the groups.
3.10 |Flow cytometry
The induction of apoptosis and necrosis was analyzed after Annexin
V-FITC and Propidium Iodide (PI) double staining (BioLegend, UK). Ini-
tially, MCF-7 cells were treated with curcumin, CS-Aga-MMT-Cur
nanoniosome, and CS-Aga-MMT without curcumin for 3 days. The
concentration of all these samples was 6.4 μg.ml
1
to compare sam-
ples at an equal concentration according to the same reason
mentioned above. The fluorescence intensity of cells was determined
in FL-1 (FITC) and FL-3 (PI) channels with the same procedure con-
ducted in former studies.
29,40
Quadrant statistics were used to deter-
mine the percentage of cells in different stages of cell death in each
quadrant. The values in the upper left, upper right, lower right, and
lower left quadrants represent necrotic (Q1), late apoptotic (Q2), early
apoptotic (Q3), and viable (Q4) cells, respectively. The described assay
was conducted in triplicate to examine the enhanced apoptosis induc-
tion of nanoniosomes loaded with CS-Aga-MMT-Cur, owing to curcu-
min's sustained-release from the fabricated niosomes.
4|RESULTS AND DISCUSSION
4.1 |FTIR
The addition of all components to the fabricated nanocomposite was
ascertained via FTIR based on the observation of peaks signifying
chitosan (CS), agarose (Aga), montmorillonite (MMT), and curcumin
(Cur) interactions. In the chitosan spectrum, the 890 cm
1
peak is
attributed to the CH bending out of the plane of the monosaccha-
ride's ring. Two bands at 1020 cm
1
and 1060 cm
1
were detected
due to CO stretching. The band at 1149 cm
1
could be ascribed to
the asymmetric extension of the COC bridge. The small peak
observed at 1272 cm
1
is due to the chitosan hydroxyl groups bend-
ing vibrations. A band at 1317 cm
1
is characteristic of the amide III
CN stretching. The absorption band detected at 1375 cm
1
is in
agreement with former detections in the literature and is ascribed to
the CH
3
symmetrical deformation.
56
At 1413 cm
1
, the peak is
referred to as CH
2
bending. The band located at 1583 cm
1
corre-
sponds to C=O stretching of amide I, which confirms N-acetyl groups
presence in the acetylglucosamine unit of chitosan.
57
The observed
band at 2856 cm
1
could be credited to CH stretching. OH stretch-
ing band can be observed at 3355 cm
1
, in agreement with previous
studies.
56
The FTIR spectrum of CS-Aga was compared with the chit-
osan spectrum to investigate the interaction between chitosan and
agarose and confirm the addition of agarose by new observed peaks.
The FTIR spectrum of CS-Aga shows two additional peaks at 613 and
655 cm
1
that represent the CH bending characteristic of aga-
rose.
58,59
The observed band at 890 cm
1
, which was ascribed to the
CH bending of chitosan moved to 917 cm
1
, probably because of
alterations in molecular interactions between chitosan and agarose. A
new band at 954 cm
1
denotes the anhydrogalactose units present in
the anhydrogalactose residues of agarose and is in accordance with
the spectrum of pure chitosan reported before.
6062
The peak
observed at 1060 cm
1
in the pure chitosan spectrum shows an
increased intensity ratio, which is ascribed to the presence of CO
stretch in both agarose, in line with the former results in the
literature,
58,63
and in chitosan. Besides, the intensity ratio of the band
at 1150 cm
1
increased in the CS-Aga likewise. The band at
1270 cm
1
representing OH in the chitosan spectrum was observed
again in the CS-Aga spectrum. The intensity ratio of the observed
CH
2
bending peak at 1413 cm
1
and the C=O stretching peak at
6of18 HASELI ET AL.
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1560 cm
1
increased in the CS-Aga as compared with the CH
because of the presence of CC stretch in agarose with CH
2
bending
of chitosan at 1413 cm
1
and the OH bending of agarose with C=O
stretching of chitosan at 1583 cm
1
. Indeed, the presence of these
peaks at the same wavenumber leads to the intensity ratio increase.
The band at 1630 cm
1
signifies the OH bending and is in accord
with reported spectrums of agarose in the literature.
64
The intensity
ratio of this band also increased because of the NH bending vibra-
tion of NH
2
in chitosan and OH bending of agarose at this wave-
number.
65
Additional peaks like the band at 2160 cm
1
are among
the characteristic peaks of agarose, affirming agarose addition to chit-
osan.
58
The band at 2856 cm
1
observed in the CS spectrum was
detected in the CS-Aga. The intensity ratio of the band at 3374 cm
1
increased in comparison with CS. This increase is ascribed to the OH
stretching vibration of agarose and OH stretching in chitosan.
64,66
In
the spectrum of CS-Aga-MMT, a new band at 776 cm
1
is observed
that refers to the CH
2
in montmorillonite.
67
The band denoting CH
at 890 cm
1
detected in the FTIR of pure chitosan was detected again
in CS-Aga-MMT. Also, the band attributed to anhydrogalactose resi-
dues of agarose in CS-Aga was observed in the CS-Aga-MMT spec-
trum at 930 cm
1
.
With regard to the CS-Aga spectrum, the intensity ratio of the
band at 1150 cm
1
increased since montmorillonite has SiO stretch-
ing and agarose has CO stretch at the same wavenumber.
67
The
peaks at 1413 cm
1
and 1583 cm
1
were observed again in the CS-
Aga-MMT spectrum. Due to the addition of MMT nanoparticles con-
taining HOH bending to CS-Aga hydrogel, the band at 1629 cm
1
shows an increased intensity ratio.
68
The band at 2856 cm
1
was
detected again in the CS-Aga-MMT spectrum.
In the spectrum of CS-Aga-MMT-Cur, the band at 930 cm
1
shifted to 925 cm
1
with a reduction in intensity ratio that indicates
probable electrostatic interactions between curcumin and hydrogel
polymers. These interactions lead to the enhanced complexation of
components in the nanocomposite. The band at 1150 cm
1
decreased
in the intensity ratio due to COC interaction in curcumin with other
components.
69
The bands at 1413 cm
1
and 1560 cm
1
were
detected again with a reduced intensity ratio compared with former
spectrums, mainly due to the complexation with curcumin. The bands
at 2918 cm
1
and 3371 cm
1
ascribed to CH stretch and OH
stretch, respectively, are characteristic peaks of curcumin detected
with reduced intensity ratio due to the complexation with other com-
ponents and are in accord with the spectrum of curcumin.
70,71
The
existence of the characteristic peaks verifies the addition of chitosan,
agarose, and montmorillonite to the fabricated nanocomposite.
Figure 2represents the FTIR spectra of samples.
4.2 |XRD analysis
XRD patterns of samples were used to determine crystalline structure
alterations by the addition of components to the nanocomposite.
XRD spectra for CS, CS-Aga, CS-Aga-MMT, and CS-Aga-MMT-Cur
were taken, as displayed in Figure 3. The diffractogram of CS shows
two characteristic peaks at 2θequal to 14.5 and 20.78 θ, which are
representative of crystalline chitosan, in agreement with the litera-
ture's results.
72
The XRD pattern of the CS-Aga sample indicates that
the crystalline peak in the spectrum of chitosan (2θ=20.78θ) has
become broader, with a reduced intensity ratio, and shifted to the
right, at 22.57θ. This reduction can be ascribed to the addition of
agarose to chitosan. Agarose has a broad peak around 1829 θ.
73
The addition of agarose to chitosan results in a semi-crystalline struc-
ture, which is displayed in the spectrum with a reduction in intensity
ratio and the formation of a broad hump (2θ=22.57θ) compared
with pure chitosan. The reduction in the intensity ratio of crystalline
chitosan was also observed after crosslinking in a study by Julkapli
et al
72
In the XRD spectrum of CS-Aga-MMT, broad peaks were
observed at 12.01θand 20.78θ. The peak observed at 12.1 θis a
characteristic of montmorillonite (JCPDS card no. 13-0135), showing
FIGURE 2 FTIR spectra of
the samples
HASELI ET AL.7of18
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
its crystalline structure, and complies with the reported results by
Samudrala et al
74
The reduction in the intensity ratio of this peak and
its broadening demonstrates the successful complexation of crystal-
line montmorillonite with other components and the formation of an
amorphous phase. Moreover, the peak at 28.51θin CS-Aga-MMT is
another characteristic peak of montmorillonite, which verifies the
incorporation of MMT in CS-Aga hydrogel.
74,75
In the CS-Aga-MMT-
Cur pattern, the intensity ratio of the peak at 20.78θincreased in
comparison with CS-Aga-MMT. This increase is due to the crystalline
peaks of curcumin between 5and 30and confirms the addition of
curcumin with regard to characteristic peaks of curcumin.
76
The suc-
cessful incorporation of curcumin in the nanocomposite with an amor-
phous structure was verified as the XRD pattern of CS-Aga-MMT-Cur
displays no crystalline peak attributed to curcumin between 20and
30.
76
Besides, the FTIR results confirmed the loading of Cur in the
nanocomposite according to the interactions observed in FTIR after
the addition of curcumin.
4.3 |Morphology observation
Lyophilized nanocomposite hydrogels that incorporated curcumin
were observed by FESEM (Figure 4). According to the images, the
spherical shape of nanocomposites is evident. The spherical shape is
an apt shape of nanocarriers for drug delivery applications.
77
From
the homogeneous surface on the nanocomposites, good compatibility
between its components was patent. According to measurements
conducted by ImageJ software, the average size of the nanocompo-
sites was 61.5 nm.
4.4 |DLS and Zeta potential
Polydispersity and nanoniosomes size were measured by DLS. The
niosome size distribution was around 400 nm with a PDI of 0.3
(Figure 5), which was sonicated to reduce their size to 241 nm. This
size distribution is satisfactory for applying niosomes in drug delivery
and is in the range of niosomal platforms fabricated before.
7880
Addi-
tionally, zeta potential can verify nanocarriers' stability. The zeta
potential was +47 mV for the nanocarriers demonstrating good stabil-
ity (Figure 6).
81
Also, the positive charge of niosomes improves their
interaction with cell membranes.
82
4.5 |Effect of montmorillonite nanoparticles on
loading and encapsulation efficiency
Poor solubility of anti-cancer agents is a prevalent challenge.
29,30,40,83
The bioavailability of curcumin has been widely reported to be very
FIGURE 3 XRD patterns of
the samples
FIGURE 4 FESEM of lyophilized nanocomposite hydrogels of CS-
Aga-MMT loaded with curcumin
8of18 HASELI ET AL.
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
poor as a consequence of its low solubility (1 μg.ml
1
).
16,17
In this
regard, improving curcumin's loading efficiency is a challenge. The
encapsulation and loading efficiencies of curcumin in CS-Aga-Cur and
CS-Aga-MMT-Cur were determined by Equations 1and 2, respec-
tively, to study MMT nanoparticles' effect on loading and entrapment
efficiency. The loading efficiency for CS-Aga-Cur was 63%, which
increased to 76% in CS-Aga-MMT-Cur. This enhanced loading is
ascribed to the role of MMT nanoparticles. In fact, the siloxane group
in montmorillonite interacts with chitosan amine groups and hydroxyl
groups in agarose and curcumin to form hydrogen bonds and create a
high interpenetrating network. The interlocked structure of the nano-
composite leads to the model drug entrapment within the structure.
84
The FTIR spectrum indicated these interactions between the curcumin
and MMT nanoparticles when the drug was loaded into the hydrogel
containing MMT nanoparticles. Besides, the introduction of MMT into
CS-Aga hydrogel leads to the provision of a higher surface for
interactions between hydrogel polymers (chitosan and agarose),
montmorillonite, and the model drug (curcumin). The loading of cur-
cuminachievedinthisstudywashigherthanthepH-responsive
platforms developed before. Ahmadi Nasab et al. developed pH-
responsive chitosan mesoporous silica nanoparticles to treat glio-
blastoma cancer. The loading of curcumin in this drug delivery sys-
tem was 8.81%.
85
In another study, solid lipid nanoparticles (SLN)
were synthesized to deliver curcumin. The loading efficiency of cur-
cumin was 71%.
86
The loading efficiency of saponin-coated curcu-
min nanoparticles was 15%.
87
The average curcumin loading
efficiency in diblock copolymer micelles developed for the treatment
of breast cancer was 5.97%.
88
Also, Li et al. fabricated thiolated
chitosan-coated liposomal hydrogels as curcumin carriers to prevent
breast cancer recurrence. The loading efficiency of curcumin was
reported 3.96%.
89
Also, the entrapment efficiency of curcumin in the chitosan-
agarose hydrogel without/ with MMT was 77% and 92%, respectively.
Similarly, the entrapment percentage in this study was higher than
curcumin encapsulation efficiency in saponin-coated curcumin
nanoparticles,
87
thiolated chitosan-coated liposomal hydrogels,
89
and
poly (lactic-co-glycolic) acid nanoparticles (PLGA NPs) loaded with
curcumin.
90
Table. 1recapitulates the efficiencies before and after the
addition of montmorillonite. Also, Table 2summarizes the loading or
encapsulation efficiencies of other curcumin delivery platforms.
0
10
20
30
40
50
60
70
80
90
100
0 100 200 300 400 500 600 700 800 900 1000
Frequency (%)
Diameter (nm)
FIGURE 5 Size distribution of
CS-Aga-MMT-Cur nanocarrier
0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1
0 102030405060
Intensity (a.u)
Zeta Potential (mV)
FIGURE 6 Zeta potential for
the fabricated CS-Aga-MMT-Cur
nanocarrier
TABLE 1 Alteration of loading and entrapment after the addition
of montmorillonite (MMT)
CS-Aga CS-Aga-MMT Effect of MMT (%)
Loading (%) 63 76 +13
Entrapment (%) 77 92 +15
HASELI ET AL.9of18
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4.6 |Curcumin release from the nanocarriers
The release profile was studied to confirm the sustained and pH-
sensitive release of curcumin from the prepared platform. As men-
tioned above, the release of curcumin was assessed by dialysis
method at pH 7.4 and 5.4 at 37C for a period of 120 h (Figure 7). As
illustrated in Figure 7, the cumulative release of curcumin from CS-
Aga-MMT-Cur and CS-Aga-Cur at pH 7.4 during 24 h was 24% and
30%, respectively. In contrast, the cumulative release during 24 h at
pH 5.4 (acidic conditions) was 67% for CS-Aga-MMT-Cur and 48%
for CS-Aga-Cur, respectively. At pH 7.4, the cumulative release from
CS-Aga-MMT-Cur is less than CS-Aga-Cur because of the impact of
MMT nanoparticles on the release behavior. The interactions of func-
tional groups in polymers like amine groups in chitosan and hydroxyl
groups in agarose and curcumin with the oxygen of siloxane group in
montmorillonite form hydrogen bonds between components and keep
the structure of the nanocomposite intact, which results in reduced
cumulative release at pH 7.4 in comparison with CS-Aga-Cur. At
pH 5.4, 98% of curcumin was released after 120 h, while at pH 7.4,
only 73% was released after 120 h. Moreover, a gradual release of
curcumin was observed prior to the sustained-release at pH 7.4,
which can be observed in Figure 7.
The observed protracted-release under neutral conditions
enables the nanocarrier to retain the drug and release more drug at
the tumor site over a long period. The release behavior of the groups
containing montmorillonite at pH 5.4 and 7.4 express the pH-
sensitive release from the prepared nanocarriers. This feature is
attributed to the dissolution of MMT nanoparticles and the release
of Al and Si ions, along with the pH-responsive property of chito-
san.
21
Indeed, the protonation of the chitosan amine group and
hydroxyl groups in agarose and curcumin, along with the release of
ions, provides repulsive forces between nanocomposite components,
which lead to the increased release under acidic conditions.
46
There-
fore, the increase in the drug release rate for CS-Aga-MMT-Cur at
pH 5.4 than at pH 7.4 can be attributed to the MMT nanoparticles'
pH-responsiveness and protonation of polymers in low pH.
46,92
There was a significant difference between pH 5.4 with montmoril-
lonite (MMT) as the control and all other groups with p< 0.001, as
presented in Figure 8.
Besides, the enhanced sustained release by niosomes can be
attributed to the oil phase in the niosomes, which functions as a
membrane to protract the release period after the nanocomposite
dissolution. A similar outcome was achieved in the fabrication of a
Chitosan-based platform.
23
Hence, the niosomal emulsion stabilizes
the nanocarrier at pH 7.4 and protracts the release from the prepared
pH-responsive hydrogel at pH 5.4.
29
The impact of niosomal emul-
sions on protracting the release period is manifest compared with the
release behavior of curcumin-loaded CS-Aga-MMT nanocomposite
fabricated in our former study and liposomal hydrogels fabricated by
Li et al
50,89
At pH 7.4, 31.57% of curcumin was released from liposo-
mal hydrogels after 12 h, while in the current study, only 15% of
curcumin was released at pH 7.4 after 12. Ju et al. prepared pH-
responsive liposomes for curcumin delivery, which released 35.25% of
curcumin at pH 7.4. after 24 h. Curcumin release from CS-Aga-MMT
nanocarrier is 24% after 24 h at pH 7.4. Also, the cumulative release
from the fabricated nanocarriers after 24 h at pH 5.4 is less than
reported by Ju et al
93
Also, in the current study, 67% of curcumin was
released at pH 5.4 after 24 h, whereas 60% of curcumin was released
from iron oxide nanoparticles loaded with curcumin after 8 h at
pH 5.
94
The simultaneous improvement of the loading and entrapment
efficiency of curcumin in niosomes and attaining sustained drug
TABLE 2 Encapsulation and loading efficiency of curcumin in other platforms
Curcumin delivery platforms Loading % Encapsulation % Reference
pH-responsive chitosan mesoporous silica
nanoparticles
8.81 85
Solid lipid nanoparticles (SLN) 71 86
Saponin-coated curcumin nanoparticles 15 91 87
Diblock copolymer micelles 5.97 64 88
Thiolated chitosan-coated liposomal hydrogels 3.96 88.75 89
Curcumin incorporated in montmorillonite (MMT) 67 91
Curcumin delivery by PLGA nanoparticles 75 90
024487296120
0
25
50
75
100
time(h)
Cumulative Release % of Curcumin
pH = 5.4 without MMT
pH = 5.4 with MMT
pH = 7.4 without MMT
pH = 7.4 with MMT
FIGURE 7 Cumulative release of curcumin from nanocarriers
encapsulating chitosan-agarose hydrogels with/without
montmorillonite (MMT) at acidic and neutral conditions
10 of 18 HASELI ET AL.
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
release is evident when the release profile of the current platform is
compared with former studies. In a study conducted by Xu et al., the
entrapment efficiency of curcumin was 92%, whereas 80% of curcu-
min was released after 24 h.
95
In contrast, this study improved the
entrapment efficiency and prolonged the release period. In another
study, optimizing the formulation of niosomes for curcumin delivery
resulted in the entrapment efficiency similar to this study but compar-
ing the release of curcumin shows the current platform has better sus-
tained release.
96
4.7 |Mathematical models of drug release
By fitting the drug release data to mathematical models of drug
release, the mechanism of drug release was determined.
54
In neutral
and acidic pH, the drug release data were fitted to zero-order, first-
order, Higuchi, Hixson-Crowell, and the Korsmeyer-Peppas models to
determine the probable alterations in the release mechanism by pH
change. Analyzing the above models exhibited the KorsmeyerPeppas
model best fits the release data at pH 7.4.
The KorsmeyerPeppas model is used to examine drug release
when the release mechanisms are complicated and involve more than
one mechanism. In the KorsmeyerPeppas model, n is the diffusional
exponent demonstrating the release mechanism. The value of
0.43 < n< 0.85 in this model designates non-fickian (or anomalous
transport) release at pH 7.4. In this case, the release is controlled by
diffusion and erosion of the polymeric matrix according to the model
and previous studies.
97,98
The same kinetics was observed in the
release of curcumin from poly (lactic-co-glycolic) acid nanofibers fabri-
cated by Sampath et al
99
The time-dependent anomalous transport
occurs from the rearrangement of polymeric chains (dissolution) and
diffusion simultaneously.
100
Also, the Higuchi model describes
the release mechanism, including both diffusion and dissolution.
101
The better fitting of the release data to the Higuchi model after the
KorsmeyerPeppas at pH 7.4 is because the Higuchi model represents
simultaneous dissolution and diffusion, which is the same as the
mechanism represented by the KorsmeyerPeppas model based on
the n value in this study.
Additionally, the same analysis was performed for pH 5.4. The
R
2
values showed that the KorsmeyerPeppas model best fits
the release data (Figure 10) at pH 5.4. According to this model, the
release mechanism is Super Case II for n> 0.85. In this mechanism,
the dissolution of the polymeric matrix occurs much faster than dif-
fusion. The reason for this faster dissolution is the protonation of
components under acidic conditions, which produces repulsive
forces between components. Also, the alteration of the mechanism
governing the model drug's release represents the prepared nanocar-
rier's pH-responsive feature. The mathematical analysis at pH 7.4
andpH5.4isillustratedinFigure9. The modeling results are recapit-
ulated in Table. 3.
4.8 |In vitro cytotoxicity study
MTT assay was used to determine the fabricated curcumin-loaded
nanocarriers' cytotoxicity and to find out if the fabricated platform for
curcumin delivery can be applied with substantial cytotoxicity in com-
parison with free curcumin. The MTT assay measured the cytotoxic
effects of CS-Aga-MMT-Cur nanocarriers and free curcumin on
MCF-7 cells after 72 hours of incubation (Figure 10). Cytotoxicity of
cells treated with CS-Aga-MMT not loaded with curcumin was also
assessed to evaluate the unloaded nanocarriers' effect on the viability
of cells. According to results represented in Figure 10, cell viability in
the CS-Aga-MMT group that was not loaded with curcumin was not
significantly reduced compared with the control group. This result
demonstrates that the fabricated nanocarrier does not have cytotoxic
effects on cancer cells by itself and only improves the release behav-
ior, as discussed before. A significant decrease in viability was
0
12
24
48
72
96
120
0
50
100
time(h)
CurcuminC
umulativeRelease%
pH=5.4 without MMT
pH=5.4 with MMT
pH=7.4 without MMT
pH=7.4 with MMT
***
***
*** *** ***
*p<0.05
** p<0.01
*** p<0.001
Com
p
arison with
p
H=5.4 with MMT
***
FIGURE 8 Cumulative release
percent of curcumin at each time point for
each group. Difference between pH 5.4
with MMT and all other groups is
significant at p< 0.001(***)
HASELI ET AL.11 of 18
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
observed in the group treated with free curcumin compared with the
control group (p< 0.001). The same result was observed for the CS-
Aga-MMT-Cur compared with the control group. In addition, the via-
bility of the group treated with CS-Aga-MMT-Cur decreased with
p< 0.001 compared with free curcumin. As mentioned before, curcu-
min is a highly hydrophobic drug with poor solubility.
102
Thus, the
effective delivery of free curcumin is hindered due to its poor solubil-
ity and instability in the cell medium. By incorporating curcumin in the
CS-Aga-MMT nanocarrier, the prolonged-release behavior leads to
the longer presence of soluble curcumin in the tumor cells' environ-
ment, causing higher cytotoxicity.
4.9 |Flow cytometry study
Apoptosis induction in breast cancer cell lines, including MCF-7, is
based on triggering a set of reactions that lead to cell membrane per-
meability, followed by swelling and potential loss in the membrane.
103
Several pathways have been identified for promoting apoptosis in can-
cer cells with curcumin. Regulation of the NF-κB signaling pathway,
104
targeting enzymes involved in the production of reactive oxygen spe-
cies (ROS),
8,12
down-regulation of bcl2 as an antiapoptotic gene,
8
and
up-regulation of bax and p53 genes are among the pathways proposed
for apoptosis induction on MCF-7 cells by curcumin.
105
pH 5.4
pH 7.4
Zero-Order
First-Order
Higuchi
Hixon-Crowell
re
y
em
sro
K -Peppas
y = 0.6535x
R = 0.9826
0
20
40
60
80
100
0 20 40 60 80 100 120
Cumulative Drug
Release %
Time (h)
y = 1.0724x
R = 0.8813
0
20
40
60
80
100
0 20 40 60 80 100 120
Cumulative Drug Release
%
Time (h)
y = -0.0333x + 4.3861
R = 0.9777
0
1
2
3
4
5
6
0 20 40 60 80 100 120
Ln ( Non-released
percentage of drug)
Time (h)
y = -0.0128x + 4.6514
R = 0.9727
0
1
2
3
4
5
6
0 20 40 60 80 100 120
Ln ( Non-released
percentage of drug)
Time (h)
y = 10.46x
R = 0.9801
0
20
40
60
80
100
0 5 10 15
cumulative release
percentage
sqrt Time (h)
y = -0.0272x + 4.1186
R = 0.8974
0
1
2
3
4
5
6
0 20 40 60 80 100 120
Cubic root of non-
released percentage
Time (h)
y = -0.016x + 4.577
R = 0.9088
0
1
2
3
4
5
6
0 20 40 60 80 100 120
Cubic root of non-
released percentage
Time (h)
y = 1.0087x
R = 0.9988
0
1
2
3
4
0246
Log (Cumulative
release percentage)
Log (Time(h))
y = 0.7778x
R = 0.9998
0
1
2
3
4
5
0246
Log( Cumulative
release percentage)
Log (Time(h))
FIGURE 9 Drug release
modeling
12 of 18 HASELI ET AL.
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
The mitochondrial-dependent pathway is another pathway
for apoptosis induction in cancer cells by curcumin. In this path-
way, cytochrome C is released from cells, which leads to a
caspase-9-caspase-3 cascade. Subsequently, poly (ADP-ribose)
polymerase (PARP) by the effectorcaspases-3 acts as a cleav-
ing agent to fragment DNA leading to cell disintegration and
apoptosis.
106,107
Externalization of phosphatidylserine (PS) from the cell's inner
membrane to its outer layer is a prominent feature of apoptosis induc-
tion. The annexin-V protein binds to PS specifically, and fluorescent
labeling of annexin-V allows flow cytometric detection of externalized
PS and, as a result, apoptotic cells. Since the loss of membrane perme-
ability in which PS externalization occurs is regarded as the onset of
apoptosis, early apoptotic cells are known as Annexin V-positive.
108
In
the early apoptosis stage, cell membrane integrity is not totally com-
promised; thus, PI cannot enter the cells making early apoptotic
cells PI-negative (Annexin V-FITC
+
/PI
). However, in the late apo-
ptosis stage, cell membrane degradation allows PI to enter the cells
making late apoptotic cells Annexin V-positive/ PI-double-positive
(Annexin V-FITC
+
/PI
+
).
109
Dual staining with Annexin V and PI
enables discriminating apoptotic and necrotic cell death since
necrotic cells are Annexin V-negative/PI-positive (Annexin V-
FITC
/PI
+
).
110
The apoptosis assay was conducted to discover the effect of the
sustained-release of curcumin achieved by the application of MMT
nanoparticles in hydrogel and niosomal emulsions on curcumin's anti-
cancer activity. As can be seen in Figure 11, the apoptotic and
necrotic induction impact of CS-Aga-MMT-Cur, free curcumin, and
blank CS-Aga-MMT (without drug) was studied by using Annexin
V-FITC and PI double staining.
The percentage of viable cells (Q4) in blank CS-Aga-MMT did not
decrease significantly compared with the viable cells in control, com-
plying with the cytotoxicity analysis mentioned earlier. Also, viable
cells treated with free curcumin (26.9%) were more than viable cells
observed in CS-Aga-MMT-Cur (9.36%), which agrees with the cyto-
toxicity results. The percentage of necrotic cells (Q1) for free curcu-
min was 62.1%, while necrotic cells in CS-Aga-MMT-Cur was 20.7%.
This difference is due to the direct exposure of cells to free curcumin.
Besides, the apoptotic cells (early and late) increased in CS-Aga-
MMT-Cur compared with free curcumin. This outcome reflects the
enhanced cytotoxicity of curcumin via increased apoptosis achieved
by curcumin sustained-release. Therefore, the prolonged-release in
the tumor microenvironment (pH 5.4), as illustrated before, is due to
the copolymer and crosslinked structure of the CS-Aga matrix and the
interaction between drug and MMT nanoparticles and niosomal emul-
sion, which lead to the protracted release of the drug and increased
apoptosis.
***$$$
***
0
10
20
30
40
50
60
70
80
90
100
Control CS-Aga-MMT CS-Aga-MMT-Cur Cur
Viability %
FIGURE 10 Cytotoxicity of curcumin, CS-Aga-MMT nanocarrier
loaded with curcumin, and CS-Aga-MMT on MCF-7 cells after 72 h
incubation. Each experiment was conducted three times in duplicate
to obtain mean ± SEM. The viability of free curcumin and CS-Aga-
MMT-Cur is significantly different from the control group (p< 0.001
(***)). Also, the viability of CS-Aga-MMT-Cur is considerably less than
free curcumin with p< 0.001 ($$$)
TABLE 3 Equations used to fit models to release data
Model Equation R
2
pH 7.4 Zero-order C
t
=1.0724 t 0.8813
First-order ln 1Mt
M

¼4:6514 0:0128t0.9727
Higuchi Q¼6:0712t1
20.9914
Hixon-Crowell 1Mt
M

1
3¼4:577 0:016t0.9088
KorsmeyerPeppas ln Mt
M

¼0:7778ln t 0.9998
pH 5.4 Zero-order C
t
=0.6535 t 0.9828
First-order ln 1Mt
M

¼4:3861 0:0333t0.9777
Higuchi Q¼10:46t1
20.9801
Hixon-Crowell 1Mt
M

1
3¼4:1186 0:0272t0.8962
KorsmeyerPeppas ln Mt
M

¼1:0087ln t 0.9988
HASELI ET AL.13 of 18
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
5|CONCLUSION
Although curcumin shows promising anti-cancer properties, its
insoluble and unstable nature is a challenge for using it as a free
drug for cancer therapy during a long period of time. To this end,
fabricating a delivery system with stimuli-sensitive properties for
targeted delivery, improved loading capacity, and prolonging its
release period is indispensable. In this study, pH-responsive nano-
compositesweredevelopedtoattainbothenhancedloadingand
sustained-release of curcumin. Incorporation of MMT nanoparticles
into nanocomposites improves loading efficiency up to 76% and
provides pH-responsive release due to the interaction with hydro-
gel polymers. The addition of nanoparticles to the hydrogel matrix
provides interactions between curcumin and the polymers to pre-
servecurcuminatpH7.4andreleaseitintheacidicconditionof
cancer cells. Besides, the encapsulation of nanocomposites into
niosomes extended the release interval by the oil phase of the nio-
somal emulsion to further prolong the release period. In addition,
nanocarriers prepared by this method improved the apoptosis
activity of curcumin and reduced cancer cells viability in compari-
son with curcumin-treated cells. Accordingly, the current fabricated
platformcanbeemployedasaprospectivepH-responsive
nanocarrier which can be employed for curcumin delivery and ame-
liorate the mentioned shortcomings assigned to curcumin for can-
cer therapy.
AUTHOR CONTRIBUTIONS
Shabnam Haseli: Formal analysis (equal); investigation (equal);
methodology (equal). mehrab pourmadadi: Formal analysis (equal);
funding acquisition (equal); methodology (equal). Amirmasoud
Samadi: Conceptualization (equal); methodology (equal); writing
original draft (equal). Fatemeh Yazdian: Project administration
(equal); supervision (equal); validation (equal). Majid Abdouss: For-
mal analysis (equal); investigation (equal). Hamid Rashedi: Concep-
tualization (equal); data curation (equal); supervision (equal). Mona
Navaei-Nigjeh: Formal analysis (equal); investigation (equal); super-
vision (equal).
CONFLICT OF INTEREST
The authors declare no conflicts of interest.
PEER REVIEW
The peer review history for this article is available at https://publons.
com/publon/10.1002/btpr.3280.
FIGURE 11 Apoptosis and
necrosis induction in MCF-7 cells
by free curcumin, CS-Aga-MMT-
Cur, and blank CS-Aga-MMT. The
values in the upper left, upper
right, lower right, and lower left
quadrants represent necrotic
(Q1), late apoptotic (Q2), early
apoptotic (Q3), and viable
(Q4) cells, respectively
14 of 18 HASELI ET AL.
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
DATA AVAILABILITY STATEMENT
The data that support the findings of this study are available from the
corresponding author upon reasonable request.
ORCID
Fatemeh Yazdian https://orcid.org/0000-0002-0550-9821
Hamid Rashedi https://orcid.org/0000-0002-8460-0841
REFERENCES
1. Pourmadadi M, Nouralishahi A, Shalbaf M, Shabani Shayeh J,
Nouralishahi A. An electrochemical aptasensor for detection of
prostate-specific antigen-based on carbon quantum dots-gold nano-
particles. Biotechnol Appl Biochem. 2022;1,2:30-40. doi:10.1002/bab.
2340
2. Cancer facts & figures 2020; 2020. https://www.cancer.org/
research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-
figures-2020.html#:~:text=EstimatedNnumbersofnewcancer,factors
%2Cearlydetection%2Candtreatment
3. Pourmadadi M, Ahmadi M, Abdouss M, Yazdian F, Rashedi H,
Hesari Y. The synthesis and characterization of double nanoemulsion
for targeted co-delivery of 5-fluorouracil and curcumin using pH-
sensitive agarose/chitosan nanocarrier. J Drug Deliv Sci Technol.
2021;70;102849. doi:10.1016/j.jddst.2021.102849
4. NIH National Cancer Institute. Cancer facts & figures 2020. 2020;
https://www.cancer.org/content/dam/cancer-org/research/cancer-
facts-and-statistics/annual-cancer-facts-and-figures/2020/cancer-
facts-and-figures-2020.pdf
5. Kaminskas LM, McLeod VM, Porter CJH, Boyd BJ. Association
of chemotherapeutic drugs with dendrimer nanocarriers: an assess-
ment of the merits of covalent conjugation compared to noncovalent
encapsulation. Mol Pharm. 2012;9(3):355-373. doi:10.1021/
mp2005966
6. Ying L, Yazdani M, Koya R, Zhao R. Engineering tumor stromal
mechanics for improved T cell therapy. Biochim Biophys Acta - Gen
Subj. 2022;1866(4):130095. doi:10.1016/J.BBAGEN.2022.130095
7. Tuli HS, Mittal S, Aggarwal D, et al. Path of Silibinin from diet to
medicine: a dietary polyphenolic flavonoid having potential anti-
cancer therapeutic significance. Semin Cancer Biol. 2020;73:196-
218. doi:10.1016/j.semcancer.2020.09.014
8. Syng-Ai C, Kumari AL, Khar A. Effect of curcumin on normal and
tumor cells: role of glutathione and bcl-2. Mol Cancer Ther. 2004;
3(9):1101-1108.
9. Hewlings S, Kalman D. Curcumin: a review of Its' effects on human
health. Foods. 2017;6(10):92. doi:10.3390/foods6100092
10. Zorofchian Moghadamtousi S, Abdul Kadir H, Hassandarvish P,
Tajik H, Abubakar S, Zandi K. A review on antibacterial, antiviral, and
antifungal activity of curcumin. Biomed Res Int. 2014;2014:1-12. doi:
10.1155/2014/186864
11. Fattahi Bafghi A, Haghirosadat BF, Yazdian F, et al. A novel delivery
of curcumin by the efficient nanoliposomal approach against Leish-
mania major. Prep Biochem Biotechnol. 2021;51(10):990-997. doi:10.
1080/10826068.2021.1885045
12. Larasati YA, Yoneda-Kato N, Nakamae I, Yokoyama T, Meiyanto E,
Kato JY. Curcumin targets multiple enzymes involved in the ROS
metabolic pathway to suppress tumor cell growth. Sci Rep. 2018;
8(1):1-13. doi:10.1038/s41598-018-20179-6
13. Vallée A, Lecarpentier Y, Vallée JN. Curcumin: a therapeutic strategy in
cancers by inhibiting the canonical WNT/β-catenin pathway. JExpClin
Cancer Res. 2019;38(1):1-16. doi:10.1186/s13046-019-1320-y
14. Siddiqui FA, Prakasam G, Chattopadhyay S, et al. Curcumin
decreases Warburg effect in cancer cells by down-regulating pyru-
vate kinase M2 via mTOR-HIF1αinhibition. Sci Rep. 2018;8(1):2-10.
doi:10.1038/s41598-018-25524-3
15. Limtrakul P. Curcumin as chemosensitizer. Adv Exp Med Biol. 2007;
595:269-300. doi:10.1007/978-0-387-46401-5_12
16. Her C, Venier-Julienne MC, Roger E. Improvement of curcumin bio-
availability for medical applications. Med Aromat Plants. 2018;07(06):
30-35. doi:10.4172/2167-0412.1000326
17. Kharat M, Du Z, Zhang G, McClements DJ. Physical and chemical
stability of curcumin in aqueous solutions and emulsions: impact of
pH, temperature, and molecular environment. J Agric Food Chem.
2017;65(8):1525-1532. doi:10.1021/acs.jafc.6b04815
18. Maghsoudi A, Yazdian F, Shahmoradi S, Ghaderi L, Hemati M,
Amoabediny G. Curcumin-loaded polysaccharide nanoparticles: opti-
mization and anticariogenic activity against Streptococcus mutans.
Mater Sci Eng C. 2017;75:1259-1267. doi:10.1016/j.msec.2017.
03.032
19. Lynch CR, Kondiah PPD, Choonara YE, du Toit LC, Ally N, Pillay V.
Hydrogel biomaterials for application in ocular drug delivery. Front
Bioeng Biotechnol. 2020;8:1-18. doi:10.3389/fbioe.2020.00228
20. Li L, Zhang Y, Han S, et al. Penetration enhancement of lidocaine
hydrochlorid by a novel chitosan coated elastic liposome for trans-
dermal drug delivery. J Biomed Nanotechnol. 2011;7(5):704-713. doi:
10.1166/jbn.2011.1333
21. Wang R, Shou D, Lv O, Kong Y, Deng L, Shen J. pH-controlled drug
delivery with hybrid aerogel of chitosan, carboxymethyl cellulose
and graphene oxide as the carrier. Int J Biol Macromol. 2017;103:
248-253. doi:10.1016/j.ijbiomac.2017.05.064
22. Azadmanesh F, Pourmadadi M, Zavar Reza J, Yazdian F, Omidi M,
Haghirosadat BF. Synthesis of a novel nanocomposite containing
chitosan as a three-dimensional printed wound dressing technique:
emphasis on gene expression. Biotechnol Prog. 2021;37(4):e3132.
doi:10.1002/btpr.3132
23. Zavareh HS, Pourmadadi M, Moradi A, Yazdian F, Omidi M. Chito-
san/carbon quantum dot/aptamer complex as a potential anticancer
drug delivery system towards the release of 5-fluorouracil. Int J Biol
Macromol. 2020;165:1422-1430. doi:10.1016/j.ijbiomac.2020.09.166
24. Díaz-Zepeda D, Peralta-Rodríguez RD, Puente-Urbina B, Cortez-
Mazatan G, Meléndez-Ortiz HI. pH responsive chitosan-coated
microemulsions as drug delivery systems. Int J Polym Mater Polym
Biomater. 2020;11:1-12. doi:10.1080/00914037.2020.1857761
25. Qi X, Su T, Tong X, et al. Facile formation of salecan/agarose hydro-
gels with tunable structural properties for cell culture. Carbohydr
Polym. 2019;224:115208. doi:10.1016/j.carbpol.2019.115208
26. Forget A, Christensen J, Lüdeke S, et al. Polysaccharide hydrogels
with tunable stiffness and provasculogenic properties via α-helix to
β-sheet switch in secondary structure. Proc Natl Acad Sci USA. 2013;
110(32):12887-12892. doi:10.1073/pnas.1222880110
27. Jin N, Morin EA, Henn DM, et al. Agarose hydrogels embedded with
pH-responsive diblock copolymer micelles for triggered release of
substances. Biomacromolecules. 2013;14(8):2713-2723. doi:10.
1021/bm4005639
28. Khodadadi Yazdi M, Taghizadeh A, Taghizadeh M, et al. Agarose-
based biomaterials for advanced drug delivery. J Control Release.
2020;326:523-543. doi:10.1016/j.jconrel.2020.07.028
29. Samadi A, Pourmadadi M, Yazdian F, Rashedi H, Navaei-Nigjeh M,
Eufrasio-da-silva T. Ameliorating quercetin constraints in cancer therapy
with pH-responsive agarose-polyvinylpyrrolidone -hydroxyapatite nano-
composite encapsulated in double nanoemulsion. Int J Biol Macromol.
2021;26:11-25. doi:10.1016/j.ijbiomac.2021.03.146
30. Ahmadi M, Pourmadadi M, Ghorbanian SA, Yazdian F, Rashedi H.
Ultra pH-sensitive nanocarrier based on Fe
2
O
3
/chitosan/montmoril-
lonite for quercetin delivery. Int J Biol Macromol. 2021;191:738-745.
doi:10.1016/J.IJBIOMAC.2021.09.023
31. Kazemi S, Pourmadadi M, Yazdian F, Ghadami A. The synthesis and
characterization of targeted delivery curcumin using chitosan-mag-
netite-reduced graphene oxide as nano-carrier. Int J Biol Macromol.
2021;186:554-562. doi:10.1016/j.ijbiomac.2021.06.184
HASELI ET AL.15 of 18
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
32. Argenta DF, dos Santos TC, Campos AM, Caon T. Hydrogel nano-
composite systems. Nanocarriers for Drug Delivery; Elsevier 2019:81-
131. doi:10.1016/b978-0-12-814033-8.00003-5
33. Samadi A, Yazdian F, Navaei-Nigjeh M, Rashedi H. Nanocomposite
hydrogels: a promising approach for developing stimuli-responsive
platforms and their application in targeted drug delivery. J Shahid
Sadoughi Univ Med Sci. 2021;29(7):3877-3897. doi:10.18502/ssu.
v29i7.7263
34. Katti DR, Sharma A, Katti KS. Predictive methodologies for design of
bone tissue engineering scaffolds. MaterialsandDevicesforBoneDisor-
ders. Elsevier Inc.; 2017:453-492. doi:10.1016/B978-0-12-802792-9.
00010-0
35. Sharifzadeh G, Hezaveh H, Muhamad II, Hashim S, Khairuddin N.
Montmorillonite-based polyacrylamide hydrogel rings for controlled
vaginal drug delivery. Mater Sci Eng C. 2020;110:110609. doi:10.
1016/j.msec.2019.110609
36. Park JH, Shin HJ, Kim MH, et al. Application of montmorillonite in ben-
tonite as a pharmaceutical excipient in drug delivery systems. JPharm
Investig. 2016;46(4):363-375. doi:10.1007/s40005-016-0258-8
37. Jayrajsinh S, Shankar G, Agrawal YK, Bakre L. Montmorillonite nano-
clay as a multifaceted drug-delivery carrier: a review. J Drug Deliv Sci
Technol. 2017;39:200-209. doi:10.1016/j.jddst.2017.03.023
38. Karatas¸ D, Tekin A, Bahadori F, Çelik MS. Interaction of curcumin in
a drug delivery system including a composite with poly(lactic-: co
-glycolic acid) and montmorillonite: a density functional theory and
molecular dynamics study. J Mater Chem B. 2017;5(40):8070-8082.
doi:10.1039/c7tb01964e
39. Silva GS, Oliveira PC, Giordani DS, De Castro HF. Chitosan/siloxane
hybrid polymer: synthesis, characterization and performance as a
support for immobilizing enzyme. J Braz Chem Soc. 2011;22(8):
1407-1417. doi:10.1590/S0103-50532011000800003
40. Nematollahi E, Pourmadadi M, Yazdian F, Fatoorehchi H, Rashedi H,
Nigjeh MN. Synthesis and characterization of chitosan/
polyvinylpyrrolidone coated nanoporous γ-alumina as a pH-sensitive
carrier for controlled release of quercetin. Int J Biol Macromol. 2021;183:
600-613. doi:10.1016/j.ijbiomac.2021.04.160
41. Gerami SE, Pourmadadi M, Fatoorehchi H, Yazdian F, Rashedi H,
Nigjeh MN. Preparation of pH-sensitive chitosan/
polyvinylpyrrolidone/α-Fe2O3 nanocomposite for drug delivery
application: emphasis on ameliorating restrictions. Int J Biol Macro-
mol. 2021;173:409-420. doi:10.1016/j.ijbiomac.2021.01.067
42. Aayanifard Z, Alebrahim T, Pourmadadi M, et al. Ultra pH-sensitive
detection of total and free prostate-specific antigen using electro-
chemical aptasensor based on reduced graphene oxide/gold nano-
particles emphasis on TiO
2
/carbon quantum dots as a redox probe.
Eng Life Sci. 2021;21(11):739-752. doi:10.1002/elsc.202000118
43. Mehdipour G, Shabani Shayeh J, Omidi M, Pour Madadi M,
Yazdian F, Tayebi L. An electrochemical aptasensor for detection of
prostate-specific antigen using reduced graphene gold nanocompo-
site and cu/carbon quantum dots. Biotechnol Appl Biochem. 2021;1-
16. doi:10.1002/bab.2271
44. Ninan N, Forget A, Shastri VP, Voelcker NH, Blencowe A. Antibac-
terial and anti-inflammatory pH-responsive tannic acid-carboxylated
agarose composite hydrogels for wound healing. ACS Appl Mater
Interfaces. 2016;8(42):28511-28521. doi:10.1021/acsami.6b10491
45. Che Y, Li D, Liu Y, et al. Physically cross-linked pH-responsive
chitosan-based hydrogels with enhanced mechanical performance
for controlled drug delivery. RSC Adv. 2016;6(107):106035-106045.
doi:10.1039/c6ra16746b
46. Ni X, Li Q, Chen W. Dissolution kinetics of Si and Al from montmoril-
lonite in carbonic acid solution. Int J Coal Sci Technol. 2014;1(1):31-
38. doi:10.1007/s40789-014-0005-6
47. Pando D, Gutiérrez G, Coca J, Pazos C. Preparation and characteri-
zation of niosomes containing resveratrol. J Food Eng. 2013;117(2):
227-234. doi:10.1016/j.jfoodeng.2013.02.020
48. Chen S, Hanning S, Falconer J, Locke M, Wen J. Recent advances in
non-ionic surfactant vesicles (niosomes): fabrication, characteriza-
tion, pharmaceutical and cosmetic applications. Eur J Pharm Bio-
pharm. 2019;144:18-39. doi:10.1016/j.ejpb.2019.08.015
49. Chouaibi M, Mejri J, Rezig L, Abdelli K, Hamdi S. Experimental Study
of Quercetin Microencapsulation Using Water-in-Oil-in-Water
(W1/O/W2) Double Emulsion. Vol 273. Elsevier B.V; 2019. doi:10.
1016/j.molliq.2018.10.030
50. Samadi A, Haseli S, Pourmadadi M, Rashedi H, Yazdian F, Navaei-
Nigjeh M. Curcumin-loaded chitosan-agarose- montmorillonite
hydrogel nanocomposite for the treatment of breast cancer. 2020
27th National and 5th International Iranian Conference on Biomedical
Engineering (ICBME); Amirkabir University 2020:148-153. Accessed
January 24, 2021. https://ieeexplore.ieee.org/document/9319425
51. Bartelds R, Nematollahi MH, Pols T, et al. Niosomes, an alternative
for liposomal delivery. PLoS One. 2018;13(4):1-18. doi:10.1371/
journal.pone.0194179
52. Jahanizadeh S, Yazdian F, Marjani A, Omidi M, Rashedi H. Curcu-
min-loaded chitosan/carboxymethyl starch/montmorillonite bio-
nanocomposite for reduction of dental bacterial biofilm formation.
Int J Biol Macromol. 2017;105:757-763. doi:10.1016/j.ijbiomac.
2017.07.101
53. Rajesh Kumar S, Priyatharshni S, Babu VN, et al. Quercetin
conjugated superparamagnetic magnetite nanoparticles for in-vitro
analysis of breast cancer cell lines for chemotherapy applications.
J Colloid Interface Sci. 2014;436:234-242. doi:10.1016/j.jcis.2014.
08.064
54. Shah BR, Li Y, Jin W, et al. Preparation and optimization of Pickering
emulsion stabilized by chitosan-tripolyphosphate nanoparticles for
curcumin encapsulation. Food Hydrocoll. 2016;52:369-377. doi:10.
1016/j.foodhyd.2015.07.015
55. Hervault A, Dunn AE, Lim M, et al. Doxorubicin loaded dual pH- and
thermo-responsive magnetic nanocarrier for combined magnetic
hyperthermia and targeted controlled drug delivery applications.
Nanoscale. 2016;8(24):12152-12161. doi:10.1039/c5nr07773g
56. Queiroz MF, Melo KRT, Sabry DA, Sassaki GL, Rocha HAO. Does
the use of chitosan contribute to oxalate kidney stone formation?
Mar Drugs. 2015;13(1):141-158. doi:10.3390/md13010141
57. Jiménez-Ocampo R, Valencia-Salazar S, Pinz
on-Díaz CE, et al. The
role of chitosan as a possible agent for enteric methane mitigation in
ruminants. Animals. 2019;9(11):1-12. doi:10.3390/ani9110942
58. Singh R, Bhattacharya B, Tomar SK, Singh V, Singh PK. Electrical,
optical and electrophotochemical studies on agarose based biopoly-
mer electrolyte towards dye sensitized solar cell application. Meas J
Int Meas Confed. 2017;102:214-219. doi:10.1016/j.measurement.
2017.02.014
59. Shamsuri AA, Daik R. Utilization of an ionic liquid/urea mixture as a
physical coupling agent for agarose/talc composite films. Materials
(Basel). 2013;6(2):682-698. doi:10.3390/ma6020682
60. Zarei Z, Akhlaghinia B. Cu(II) immobilized on Fe
3
O
4
@agarose nano-
magnetic catalyst functionalized with ethanolamine phosphate
salicylaldehyde schiff base: a magnetically reusable nanocatalyst for
preparation of 2-substituted imidazolines, oxazolines, and thiazo-
lines. Turkish J Chem. 2018;42(1):170-191. doi:10.3906/kim-
1704-53
61. Garcia RB, Vidal RRL, Rinaudo M. Preparation and structural characteri-
zation of O-acetyl agarose with low degree of substitution. Polímeros.
2000;10(3):155-161. doi:10.1590/s0104-14282000000300012
62. MacGregor EA. Biopolymers. Encyclopedia of Physical Science and
Technology. Elsevier; 2003:207-245. doi:10.1016/B0-12-227410-5/
00064-8
63. Wang F, Xiao W, Elbahnasawy MA, et al. Optimization of the linker
length of mannose-cholesterol conjugates for enhanced mRNA
delivery to dendritic cells by liposomes. Front Pharmacol. 2018;9:1-
14. doi:10.3389/fphar.2018.00980
16 of 18 HASELI ET AL.
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
64. Hu Z, Hong P, Liao M, et al. Preparation and characterization of
chitosan-agarose composite films. Materials (Basel). 2016;9(10):1-9.
doi:10.3390/ma9100816
65. Oh JW, Chun SC, Chandrasekaran M. Preparation and in vitro char-
acterization of chitosan nanoparticles and their broad-spectrum anti-
fungal action compared to antibacterial activities against
phytopathogens of tomato. Agronomy. 2019;9(1):86-90. doi:10.
3390/agronomy9010021
66. El-Hefian EA, Nasef MM, Yahaya AH. Preparation and characteriza-
tion of chitosan/agar blended films: part 1. Chemical structure and
morphology. E-Journal Chem. 2012;9(3):1431-1439. doi:10.1155/
2012/781206
67. Karaca S, Gürses A, Ejder KM. Investigation of the orientation of
CTA +ions in the interlayer of CTAB pillared montmorillonite.
J Chem. 2013;50:1-10. doi:10.1155/2013/274838
68. Bishop JL, Pieters CM, Edwards JO. Infrared spectroscopic analyses
on the nature of water in montmorillonite. Clays Clay Miner. 1994;
42(6):702-716. doi:10.1346/CCMN.1994.0420606
69. Chen X, Zou LQ, Niu J, Liu W, Peng SF, Liu CM. The stability, sustained
release and cellular antioxidant activity of curcumin nanoliposomes. Mol-
ecules. 2015;20(8):14293-14311. doi:10.3390/molecules200814293
70. Chopra M, Jain R, Dewangan AK, Varkey S, Mazumder S. Design of
curcumin loaded polymeric nanoparticles-optimization, formulation
and characterization. J Nanosci Nanotechnol. 2016;16(9):9432-9442.
doi:10.1166/jnn.2016.12363
71. Yallapu MM, Jaggi M, Chauhan SC. β-Cyclodextrin-curcumin self-
assembly enhances curcumin delivery in prostate cancer cells. Col-
loids Surf B Biointerfaces. 2010;79(1):113-125. doi:10.1016/j.
colsurfb.2010.03.039
72. Julkapli NM, Ahmad Z, Akil HM. X-ray diffraction studies of cross
linked chitosan with different cross linking agents for waste water
treatment application. AIP Conference Proceedings. Elsevier Vol
1202; 2009:106-111. doi:10.1063/1.3295578
73. Singh R, Jadhav NA, Majumder S, Bhattacharya B, Singh PK. Novel
biopolymer gel electrolyte for dye-sensitized solar cell application.
Carbohydr Polym. 2013;91(2):682-685. doi:10.1016/j.carbpol.2012.
08.055
74. Samudrala SP, Kandasamy S, Bhattacharya S. Turning biodiesel
waste glycerol into 1,3-Propanediol: catalytic performance of Sul-
phuric acid-activated montmorillonite supported platinum catalysts
in glycerol Hydrogenolysis. Sci Rep. 2018;8(1):1-13. doi:10.1038/
s41598-018-25787-w
75. Praus P, Turicová M, Karlíková M, Kvítek L, Dvorský R. Nanocompo-
site of montmorillonite and silver nanoparticles: characterization and
application in catalytic reduction of 4-nitrophenol. Mater Chem Phys.
2013;140(23):493-498. doi:10.1016/j.matchemphys.2013.03.059
76. Liu C, Cheng F, Yang X. Fabrication of a soybean Bowman-Birk
inhibitor (BBI) Nanodelivery carrier to improve bioavailability of cur-
cumin. J Agric Food Chem. 2017;65(11):2426-2434. doi:10.1021/acs.
jafc.7b00097
77. Prabha G, Raj V. Preparation and characterization of chitosan -
polyethylene glycol-polyvinylpyrrolidone-coated superparamagnetic
iron oxide nanoparticles as carrier system: drug loading and in vitro
drug release study. J Biomed Mater Res - Part B Appl Biomater. 2016;
104(4):808-816. doi:10.1002/jbm.b.33637
78. Sedaghat Doost A, Kassozi V, Grootaert C, et al. Self-assembly, func-
tionality, and in-vitro properties of quercetin loaded nanoparticles
based on shellac-almond gum biological macromolecules. Int J Biol
Macromol. 2019;129:1024-1033. doi:10.1016/j.ijbiomac.2019.
02.071
79. Sadeghi Ghadi Z, Dinarvand R, Asemi N, Talebpour Amiri F,
Ebrahimnejad P. Preparation, characterization and in vivo evaluation
of novel hyaluronan containing niosomes tailored by box-Behnken
design to co-encapsulate curcumin and quercetin. Eur J Pharm Sci.
2019;130:234-246. doi:10.1016/j.ejps.2019.01.035
80. Akbari J, Saeedi M, Enayatifard R, et al. Curcumin Niosomes (curcu-
somes) as an alternative to conventional vehicles: a potential for effi-
cient dermal delivery. J Drug Deliv Sci Technol. 2020;60:102035. doi:
10.1016/j.jddst.2020.102035
81. Joseph E, Singhvi G. Multifunctional nanocrystals for cancer therapy:
a potential nanocarrier. Nanomaterials for Drug Delivery and Therapy.
Elsevier Inc.; 2019:91-116. doi:10.1016/B978-0-12-816505-8.
00007-2
82. Salem HF, Kharshoum RM, Abou-Taleb HA, Farouk HO, Zaki RM.
Fabrication and appraisal of simvastatin via tailored niosomal
nanovesicles for transdermal delivery enhancement: in vitro and
in vivo assessment. Pharmaceutics. 2021;13(2):1-25. doi:10.3390/
pharmaceutics13020138
83. Yazdani M, Tavakoli O, Khoobi M, et al. Beta-carotene/cyclodextrin-
based inclusion complex: improved loading, solubility, stability, and
cytotoxicity. J Incl Phenom Macrocycl Chem. 2022;102(12):55-64.
doi:10.1007/s10847-021-01100-7
84. Kumarasamy D, Giri TK. Biopolysaccharide-based hydrogel materials
for drug delivery. Polysaccharide Carriers for Drug Delivery. Elsevier
Ltd.; 2019:585-613. doi:10.1016/B978-0-08-102553-6.00020-9
85. Nasab NA, Kumleh HH, Beygzadeh M, Teimourian S, Kazemzad M.
Delivery of curcumin by a pH-responsive chitosan mesoporous silica
nanoparticles for cancer treatment. Artif Cells, Nanomedicine Biotech-
nol 2018;46(1):44-48. doi:10.1080/21691401.2017.1290648,81
86. Jourghanian P, Ghaffari S, Ardjmand M, Haghighat S,
Mohammadnejad M. Sustained release curcumin loaded solid lipid
nanoparticles. Adv Pharm Bull. 2016;6(1):17-21. doi:10.15171/apb.
2016.04
87. Peng S, Li Z, Zou L, Liu W, Liu C, McClements DJ. Improving curcumin
solubility and bioavailability by encapsulation in saponin-coated cur-
cumin nanoparticles prepared using a simple pH-driven loading
method. Food Funct. 2018;9(3):1829-1839. doi:10.1039/c7fo01814b
88. Alizadeh AM, Sadeghizadeh M, Najafi F, et al. Encapsulation of cur-
cumin in diblock copolymer micelles for cancer therapy. Biomed Res
Int. 2015;2015:1-14. doi:10.1155/2015/824746
89. Li R, Lin Z, Zhang Q, et al. Injectable and in situ-formable Thiolated
chitosan-coated liposomal hydrogels as curcumin carriers for pre-
vention of in vivo breast cancer recurrence. ACS Appl Mater Inter-
faces. 2020;12(15):17936-17948. doi:10.1021/acsami.9b21528
90. Sampath M, Pichaimani A, Kumpati P, Sengottuvelan B. The remark-
able role of emulsifier and chitosan, dextran and PEG as capping
agents in the enhanced delivery of curcumin by nanoparticles in
breast cancer cells. Int J Biol Macromol. 2020;162:748-761. doi:10.
1016/j.ijbiomac.2020.06.188
91. Kar S, Kundu B, Reis RL, Sarkar R, Nandy P, Basu R, Das S. Curcumin
ameliorates the targeted delivery of methotrexate intercalated
montmorillonite clay to cancer cells. Eur J Pharm Sci 2019;135(May):
91102. doi:10.1016/j.ejps.2019.05.006
92. Hua S, Yang H, Wang A. A pH-sensitive nanocomposite microsphere
based on chitosan and montmorillonite with in vitro reduction of the
burst release effect. Drug Dev Ind Pharm. 2010;36(9):1106-1114.
doi:10.3109/03639041003677798
93. Ju L, Cailin F, Wenlan W, Pinghua Y, Jiayu G, Junbo L. Preparation
and properties evaluation of a novel pH-sensitive liposomes based
on imidazole-modified cholesterol derivatives. Int J Pharm. 2017;
518(12):213-219. doi:10.1016/j.ijpharm.2016.11.044
94. Saikia C, Das MK, Ramteke A, Maji TK. Controlled release of curcu-
min from Thiolated starch coated iron oxide magnetic nanoparticles:
an in vitro evaluation. Int J Polym Mater Polym Biomater. 2017;66(7):
349-358.
95. Xu YQ, Chen WR, Tsosie JK, et al. Niosome encapsulation of curcu-
min: characterization and cytotoxic effect on ovarian cancer cells.
J Nanomater. 2016;2016:1-9. doi:10.1155/2016/6365295
96. Akbarzadeh I, Shayan M, Bourbour M, et al. Preparation, optimiza-
tion and in-vitro evaluation of curcumin-loaded Niosome@calcium
HASELI ET AL.17 of 18
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
alginate Nanocarrier as a new approach for breast cancer treatment.
Biology (Basel). 2021;10(3):173. doi:10.3390/biology10030173
97. Korsmeyer RW, Gurny R, Doelker E, Buri P, Peppas NA. Mechanisms
of solute release from porous hydrophilic polymers. Int J Pharm.
1983;15(1):25-35. doi:10.1016/0378-5173(83)90064-9
98. Jeon S, Yoo CY, Park SN. Improved stability and skin permeability of
sodium hyaluronate-chitosan multilayered liposomes by layer-by-
layer electrostatic deposition for quercetin delivery. Colloids Surf B
Biointerfaces. 2015;129:7-14. doi:10.1016/j.colsurfb.2015.03.018
99. Sampath M, Lakra R, Korrapati PS, Sengottuvelan B. Curcumin
loaded poly (lactic-co-glycolic) acid nanofiber for the treatment of
carcinoma. Colloids Surf B Biointerfaces. 2014;117:128-134. doi:10.
1016/j.colsurfb.2014.02.020
100. Bruschi ML. Mathematical models of drug release. Strateg to Modify
Drug Release from Pharm Syst. 2015;11:63-86. doi:10.1016/b978-0-
08-100092-2.00005-9
101. Baishya H. Application of mathematical models in drug release
kinetics of carbidopa and levodopa ER tablets. J Dev Drugs. 2017;
06(02):1-8. doi:10.4172/2329-6631.1000171
102. Aditya NP, Aditya S, Yang H, Kim HW, Park SO, Ko S. Co-delivery of
hydrophobic curcumin and hydrophilic catechin by a water-in-oil-in-
water double emulsion. Food Chem. 2015;173:7-13. doi:10.1016/j.
foodchem.2014.09.131
103. Karunagaran D, Rashmi R, Kumar T. Induction of apoptosis by curcu-
min and its implications for cancer therapy. Curr Cancer Drug Targets.
2005;5(2):117-129. doi:10.2174/1568009053202081
104. Liu JL, Pan YY, Chen O, et al. Curcumin inhibits MCF-7 cells by mod-
ulating the NF-κB signaling pathway. Oncol Lett. 2017;14(5):5581-
5584. doi:10.3892/OL.2017.6860
105. Fatemizadeh M, Tafvizi F, Shamsi F, Amiri S. Apoptosis induction,
cell cycle arrest and anti-cancer potential of tamoxifen-curcumin
loaded niosomes against MCF-7 cancer cells. Iran J Pathol. 2022;
17(2):183-190. doi:10.30699/IJP.2022.124340.2356
106. Montazeri M, Sadeghizadeh M, Pilehvar-Soltanahmadi Y, et al. Den-
drosomal curcumin nanoformulation modulate apoptosis-related genes
and protein expression in hepatocarcinoma cell lines. Int J Pharm.
2016;509(12):244-254. doi:10.1016/J.IJPHARM.2016.05.039
107. Mortezaee K, Salehi E, Mirtavoos-mahyari H, et al. Mechanisms of
apoptosis modulation by curcumin: implications for cancer therapy.
J Cell Physiol. 2019;234(8):12537-12550. doi:10.1002/JCP.28122
108. Brauchle E, Thude S, Brucker SY, Schenke-Layland K. Cell death
stages in single apoptotic and necrotic cells monitored by Raman
microspectroscopy. Sci Rep. 2014;4(1):1-9. doi:10.1038/srep04698
109. Wlodkowic D, Telford W, Skommer J, Darzynkiewicz Z. Apoptosis and
beyond: cytometry in studies of programmed cell death. Methods Cell
Biol. 2011;103:55-98. doi:10.1016/B978-0-12-385493-3.00004-8
110. Luschnig P, Kienzl M, Roula D, et al. The JAK1/2 inhibitor baricitinib
suppresses eosinophil effector function and restricts allergen-
induced airway eosinophilia. Biochem Pharmacol. 2021;192:114690.
doi:10.1016/J.BCP.2021.114690
How to cite this article: Haseli S, Pourmadadi M, Samadi A,
et al. A novel pH-responsive nanoniosomal emulsion for
sustained release of curcumin from a chitosan-based
nanocarrier: Emphasis on the concurrent improvement of
loading, sustained release, and apoptosis induction. Biotechnol.
Prog. 2022;e3280. doi:10.1002/btpr.3280
18 of 18 HASELI ET AL.
15206033, 0, Downloaded from https://aiche.onlinelibrary.wiley.com/doi/10.1002/btpr.3280 by University Of California - Irvine, Wiley Online Library on [12/10/2022]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
... In addition, the niosomal formulation of drug molecules and the level of toxicity against the same order of cancerous cell lines were shown to be significantly correlated. Finally, since niosomes have not been found toxic to cells in our prior research and some other studies, in which they served as vehicles to deliver drugs, we were able to choose them for this study as well [3,[38][39][40]. ...
Article
Full-text available
In the present study, we present a pyranopyrazole-TiO2 which is encapsulated with a niosome as nanocarrier for delivery of curcumin into breast cancer cells. Nanocarrier porous TiO2 is biocompatible and with a high specific surface area and a large pore volume and was used to carry pyranopyrazole, which has been reported as an anti-cancer. Niosome in the outer layer, helpful for loading curcumin into the niosomal layer, demonstrates a pH-dependent release and can be effective for cancer treatment. Entrapment efficiency of curcumin was found at 81.02% in carriers. The results of MTT and flow cytometry revealed that apoptosis is notably enhanced by loading curcumin on pyranopyrazole-TiO2@niosome. Also, there was high biocompatibility with MCF-10A, while exhibiting significant anti-cancer and anti-metastatic effects on MCF-7, whose cell viability was 38.79% in the loaded curcumin on carrier and was more than other samples even, than free curcumin (42.82%). Furthermore, the regulation of gene expression in cancer cells decreased the regulation of MMP-2 and MMP-9 genes and increased the expression of caspase-3 and caspase-9 genes. Finally, fluorescence activity in MCF-7 significantly increased after treatment with samples. Graphical Abstract
... Also, previous studies have reported that NCs containing CUR can induce apoptosis in breast cancer cells, which is compatible with our findings. Overall, our results illustrate that the developed hydrogel NCs are more efficient than free CUR in inducing cell apoptosis [16,47,75]. ...
Article
Full-text available
Nanoparticles have promisingly served as therapeutic agents in drug delivery systems to supress side effects while improving the effectiveness of treatment to deadly cancers. The present study aims to fabricate chitosan/agarose/graphene oxide/montmorillonite nanocomposites to serve as drug carriers for the prolonged release of curcumin to suppress MCF7 breast cancer cells. Offering a range of advantages, including biocompatibility and pH-sensitivity, the nanocomposites were synthesized via a water-in-oil-in-water (W/O/W) emulsification technique. Characterization studies were conducted with FTIR and XRD analyses. Dynamic light scattering (DLS) analysis was conducted to measure the mean size of the produced nanoscale emulsions, with zeta potential assessments further performed to characterize the nanocomposites in terms of surface charge. FE-SEM imagery indicated the uniform and flat surface of the synthesized nanocomposites. MTT assay results showed that the nanocomposites were most toxic to MCF7 breast cancer cells. The occurrence of apoptosis to MCF7 breast cancer cells was evident from the flow cytometry findings. Implementing a dialysis procedure, the curcumin was found to be released at a higher rate in acidic media. Altogether, our findings proved that the designed CS/AG/GO/MMT@CUR nanocomposites could be a drug carrier for treating breast cancer cells. Graphical Abstract
... The percentage of living cells for nanocarrier and free drug is 28% and 36%, respectively. Also, the nanocarrier without the presence of drug did not show many changes in cytotoxicity in the breast cancer cell line, which means that this nanocarrier has little cytotoxic effect [19][20][21][22][23][24][25][26]. ...
Conference Paper
Full-text available
Nanotechnology-based drug delivery systems can reduce side effects, increase drug release time, and control drug release. Polymeric nanoparticles used as drug nanocarriers increase drug solubility and stability. Growing concerns about the safety of chemicals and additives have necessitated the use of bioactive substances. Drug concentration is an important factor in cytotoxicity studies as it can play a role in cell death and growth inhibition. Thus, beneficial substances released by nanocarriers remove negative and unstable substances from cells. Nanocarriers are designed for prolonged release of drugs at the cancer site, resulting in potent cytotoxicity. In the most recent study, nanocarriers are prepared by double emulsion methods that stabilize and fix the shape of the nanoparticles and also prevent the nanoparticles from accelerating during drug release in vivo. In this study, Cytotoxicity investigated the effect of a newly developed nanocarrier on anti-inflammatory drug delivery to breast cancer cells(MCF-7).
... In another study, a nanocomposite hydrogel platform for montmorillonite nanoparticles added to chitosan-agarose was constructed, increasing the loading capacity of curcumin from 63 to 76% and increasing the apoptotic rate. The delivery platform in the present study enhanced the curcumin load, sustained its release and increased its anticancer effects (100). In another study, magnetic nanoparticles containing curcumin with carboxymethyl chitosan (MNP-CMC-CUR) were designed and used to treat breast cancer MCF-7 and MDA-MB-231 cell lines and human fibroblasts, and their effect was compared to that of curcumin alone in MTT assays (101). ...
Article
Full-text available
Curcumin is a plant polyphenol in turmeric root and a potent antioxidant. It binds to antioxidant response elements for gene regulation by nuclear factor erythroid 2-related factor 2, thereby suppressing reactive oxygen species (ROS) and exerting anti-inflammatory, anti-infective and other pharmacological effects. Of note, curcumin induces oxidative stress in tumors. It binds to several enzymes in tumors, such as carbonyl reductases, glutathione S-transferase P1 and nicotinamide adenine dinucleotide phosphate to induce mitochondrial damage, increase ROS production and ultimately induce tumor cell death. However, the instability and poor pharmacokinetic profile of curcumin in vivo limit its clinical application. Therefore, the effects of curcumin in vivo may be enhanced through its combination with drugs, derivative development and nanocarriers. In the present review, the mechanisms of curcumin that induce tumor cell death through oxidative stress are discussed. In addition, the methods used to enhance the antitumor activity of curcumin are described. Finally, the existing knowledge on the functions of curcumin in tumors, particularly in terms of oxidative stress, are summarized to facilitate future curcumin research.
... The pH-responsive emulsion has emerged as a significant subject of interest in biomedical and pharmaceutical research due to its potential for targeted drug release in specific physiological environments (Jeshvaghani et al., 2023). These emulsions are typically stabilized by surfactants that respond to changes in environmental pH, triggering alterations in their surface activity and hence the stability of the emulsion (Haseli et al., 2022). This phenomenon can be exploited for precision drug delivery, with the drug being released in specific physiological locations where the pH triggers destabilization of the emulsion (Li et al., 2021). ...
Article
Cancer disease is one of the most frequent life-threatening, with a high fatality rate worldwide. However, recent immunotherapy studies in various tumours have yielded unsatisfactory outcomes, with just a few individuals experiencing long-term responses. To overcome these issues, nowadays internal stimuli-responsive nanocarriers have been widely exploited to transport a wide range of active substances, including peptides, genes and medicines. These nanosystems could be chemically adjusted to produce target-based drug release at the target location, minimizing pathological and physiological difficulties while increasing therapeutic efficiency. This review highlights the various types of internal stimuli-responsive nanocarriers and applications in cancer diagnosis. This study can provide inspiration and impetus for exploiting more promising internal stimuli-responsive nanosystems for drug delivery.
Article
The advent of nanotechnology has led to an increased interest in nanocarriers as a drug delivery system that is efficient and safe. There have been many studies addressing nano-scale vesicular systems such as liposomes and niosome is a newer generation of vesicular nanocarriers. The niosomes provide a multilamellar carrier for lipophilic and hydrophilic bioactive substances in the self-assembled vesicle, which are composed of non-ionic surfactants in conjunction with cholesterol or other amphiphilic molecules. These non-ionic surfactant vesicles, simply known as niosomes, can be utilized in a wide variety of technological applications. As an alternative to liposomes, niosomes are considered more chemically and physically stable. The methods for preparing niosomes are more economic. Many reports have discussed niosomes in terms of their physicochemical properties and applications as drug delivery systems. As drug carriers, nano-sized niosomes expand the horizons of pharmacokinetics, decreasing toxicity, enhancing drug solvability and bioavailability. In this review, we review the components and fabrication methods of niosomes, as well as their functionalization, characterization, administration routes, and applications in cancer gene delivery, and natural product delivery. We also discuss the limitations and challenges in the development of niosomes, and provide the future perspective of niosomes.
Article
Full-text available
Curcumin, a well-known dietary pigment derived from Curcuma longa, inhibited growth of several types of malignant cells both in vivo and in vitro. However, its mechanism of action still remains unclear. In this study, we have focused primarily on the cytotoxic effects of curcumin on three human tumor cell lines and rat primary hepatocytes. Curcumin induced apoptosis in MCF-7, MDAMB, and HepG2 cells in a dose-dependent and time-dependent manner. Apoptosis was mediated through the generation of reactive oxygen species. Attempts were made to establish the role played by endogenous glutathione on the apoptotic activity of curcumin. Depletion of glutathione by buthionine sulfoximine resulted in the increased generation of reactive oxygen species, thereby further sensitizing the cells to curcumin. Interestingly, curcumin had no effect on normal rat hepatocytes, which showed no superoxide generation and therefore no cell death. These observations suggest that curcumin, a molecule with varied actions, could be developed into an effective chemopreventive and chemotherapeutic agent.
Article
Full-text available
Background & objective: Breast cancer is the most common cancer among women. One of the most effective treatments for breast cancer is chemotherapy, in which specific drugs destroy the mass and its proliferation is inhibited. Chemotherapy is the most effective adjunctive therapy when multiple medications are used concurrently. Also, combining the drugs with nanocarrier has become an important strategy in targeted therapy. This study is designed to assess the apoptosis induction, cell cycle arrest, and anti-cancer potential of Tamoxifen-Curcumin-loaded niosomes against MCF-7 Cancer Cells. Methods: A novel niosomal formulation of tamoxifen-curcumin with Span 80 and lipid to drug ratio of 20 was employed. The MCF-7 cells were cultured and then treated with IC50 value of tamoxifen-curcumin-loaded niosomes, the combination of tamoxifen and curcumin, tamoxifen, and curcumin alone. Flow cytometry, Real-Time PCR, and cell cycle analysis tests were conducted to evaluate the induction of apoptosis. Results: Drug-loaded niosomes caused up-regulation of bax and p53 genes and down-regulation of bcl2 gene. Flow cytometry studies showed that niosomes containing tamoxifen-curcumin increased apoptosis rate in MCF-7 cells compared to the combination of tamoxifen and curcumin owing to the synergistic effect between the two drugs along with higher cell uptake by formulation niosomal. These results were also confirmed by cell cycle analysis. Conclusion: Co-delivery of curcumin and tamoxifen using optimized niosomal formulation revealed that at acidic pH of MCF-7 cancer cells, released drugs from niosomal carriers would be more effective than physiological pH. This feature of niosomal nanoparticles can reduce the side effects of drugs in normal cells. Niosomal nanoparticles might be used as a biological anti-cancer factor in treatment of breast cancer.
Article
Full-text available
In this work, an electrochemical aptasensor was described for determination of Prostate specific antigen (PSA). Aptamer chains were decorated on the surface of a glassy carbon electrode (GCE) via carbon quantum dots/Au nanoparticles (Au/CQD). Structural analysis that used to characterize the prepared materials shows that Au/CQD nanoparticles synthesized spherically with an average size of 70 nm. Furthermore, the combination of Au nanoparticles with CQD caused the formation of crystallinity in the structure of the Au/CQD composite. For study the electrochemical performance of prepared aptasensor, Cyclic voltammetry (CV), square wave voltammetry (SWV), and electrochemical impedance spectroscopy (EIS) were used. The results show that the aptasensor has a good selectivity to PSA over other biomaterials with the time optimized about 30 min. K4 [Fe (CN)6] was used as an electrochemical probe with the limit of detection about 2 fg.ml–1. To avoid the hazardous nature of K4 [Fe (CN)6], label based aptasensor prepared using methylene blue as an electrochemical signal producer. They provide the capability of electrochemical detection in buffer phosphate solution with high sensitivity. This article is protected by copyright. All rights reserved
Article
Full-text available
We report a label‐free electrochemical aptamer‐based biosensor for the detection of human prostate‐specific antigen (PSA). The thiolate DNA aptamer against PSA was conjugated to the reduced graphene oxide/Au (RGO‐Au) nanocomposite through the self‐assembly of Au‐S groups. Owing to the large volume to surface ratio, the RGO‐Au nanocomposite provides a large surface for aptamer loading. The RGO‐Au/aptamer was combined with Nafion polymer and immobilized on a glassy carbon electrode. The interaction of aptamer with PSA was studied by cyclic voltammetry (CV), square wave voltammetry (SW), and electrochemical impedance spectroscopy (EIS). The detection of limit for prepared electrode was obtained about 50 Pg/ml at the potential of 0.4V in Potassium hexacyanoferrate [K4 Fe (CN)6] medium. To decrease the LOD and applied potential of the prepared nanoprobe CuCQD is introduced as a new redox probe. The results show that this new electrochemical medium provides better conditions for the detection of PSA. LOD of nanoprobe in CuCQD media was obtained as 40 pg/ml at the potential of ‐0.2V. Under optimal conditions, the aptasensor exhibits a linear response to PSA with a LOD as small as 3 Pg/ml. The present aptasensor is highly selective and sensitive and shows satisfactory stability and repeatability. This article is protected by copyright. All rights reserved
Article
Full-text available
Introduction: With the development of hydrogels from half a century ago, their application in various medical fields, including drug delivery has been widely expanded. Hydrogels used in this field are produced with synthetic polymers such as polyvinylpyrrolidone, polyvinyl alcohol, or natural polymers like chitosan, agarose, and hyaluronic acid to develop biocompatible, biodegradable, and non-immunogenic drug carriers. However, limitations such as inadequate response to stimulation, low homogeneity, and poor loading capacity for hydrophobic drugs have limited the use of hydrogels for drug delivery due to the hydrophilic nature of the hydrogel. The use of nanoparticles in the structure of hydrogels to produce hydrogel nanocomposites leads to more diverse interactions such as hydrogen and electrostatic bonds in addition to covalent interactions between hydrogel polymers. In addition to enhancing the mechanical properties of the hydrogel and further homogeneity, these interactions lead to the formation of platforms responsive to various stimuli, attaining sustained release, and ameliorating the poor loading of hydrophobic drugs used in cancer treatment and wound dressing. Conclusion: A review of a research conducted in the last 20 years represents that the application of nanocomposite hydrogels in drug delivery includes a wide range of production methods, nanoparticles to create various stimulation mechanisms, and therapeutic applications. Indeed, research has been focused on developing smart systems for controlled release with stimuli to reduce side effects of conventional cancer treatment methods, such as chemotherapy, by targeting drug delivery and reducing drug administration frequency and mitigating chronic wound complications by the release of growth factors.
Article
Full-text available
Beta-carotene (BC) is a vitamin A precursor and has potential anticancer benefits, but the delivery of BC is hindered by its low solubility and storage instability. To overcome these challenges, this study investigated the use of fabricated cyclodextrin-based nanosponges (CDNS) using different ratios of two cross-linkers, epiclon (EPI) and hexamethylene diisocyanate (HMDI) to form inclusion complex with BC. The ratios of crosslinkers to βCD for two most optimaly encapsulated CDNSs-BC were determined to be 2:1 for EPI and 4:1 for HMDI with loading efficiency of 61.46% and 59.61%, respectively. The charachterization tests were carefully done for two optimal CDNSs. Encapsulation significantly improved the solubility by ~ 10 folds, 30-day storage stability by 40% compared to BCs. The in vitro release of the two encapsulated products showed no burst release. The MTT assay revealed a variable increase in cytotoxic effect in both normal and cancer cells compared to free BC. Overall, the CDNSs appear to be promising carriers for the delivery of BCs.
Article
Background & Objective: Breast cancer is the most common cancer among women. One of the most effective treatments for breast cancer is chemotherapy, in which specific drugs destroy the mass and its proliferation is inhibited. Chemotherapy is the most effective adjunctive therapy when multiple medications are used concurrently. Also, combining the drugs with nanocarrier has become an important strategy in targeted therapy. This study is designed to assess the apoptosis induction, cell cycle arrest, and anti-cancer potential of Tamoxifen-Curcumin-loaded niosomes against MCF-7 Cancer Cells. Methods: A novel niosomal formulation of tamoxifen-curcumin with Span 80 and lipid to drug ratio of 20 was employed. The MCF-7 cells were cultured and then treated with IC50 value of tamoxifen-curcumin-loaded niosomes, the combination of tamoxifen and curcumin, tamoxifen, and curcumin alone. Flow cytometry, Real-Time PCR, and cell cycle analysis tests were conducted to evaluate the induction of apoptosis. Results: Drug-loaded niosomes caused up-regulation of bax and p53 genes and down-regulation of bcl2 gene. Flow cytometry studies showed that niosomes containing tamoxifen-curcumin increased apoptosis rate in MCF-7 cells compared to the combination of tamoxifen and curcumin owing to the synergistic effect between the two drugs along with higher cell uptake by formulation niosomal. These results were also confirmed by cell cycle analysis. Conclusion: Co-delivery of curcumin and tamoxifen using optimized niosomal formulation revealed that at acidic pH of MCF-7 cancer cells, released drugs from niosomal carriers would be more effective than physiological pH. This feature of niosomal nanoparticles can reduce the side effects of drugs in normal cells. Niosomal nanoparticles might be used as a biological anti-cancer factor in treatment of breast cancer.
Article
Adoptive cellular therapies (ACT), including the engineered T cell receptor (TCR) therapy and chimeric antigen receptor (CAR) T Cell Therapy, are currently at the forefront of cancer immunotherapy. However, their efficacy for the treatment of solid tumors has not been confirmed. The fibrotic stroma surrounding the solid tumor has been suggested as a main barrier in the disarmament and suppression of the engineered T cells. In this review, we will discuss the recent findings on the mechanism of T cell suppression by the tumor stroma with a special emphasis on the effect of stromal mechanics. We will also discuss the engineering approaches used to dissect the mechanism of the T cell suppression by the stromal mechanical factors. Finally, we will provide a future outlook on the strategies to improve the efficacy of T cell therapy through altering the tumor stromal fibrosis.
Article
Due to the dangerous side effects of chemotherapy, advanced methods for curing cancer, such as drug delivery, have attracted much attention because of their high efficacies. Using a green synthesizing method, we prepared a double nano-emulsion agarose/chitosan nanocomposite as the carrier of this work to adjust stability and hydrogel releasing. Curcumin, 5-fluorouracil, and curcumin/5-fluorouracil were loaded into the nano-carrier, and the synthesized systems were characterized by XRD, FESEM, FTIR, DLS, and zeta potential analysis. Loading efficacy and entrapment efficacy of curcumin in the agarose/chitosan nanocomposite were 85.6% and 79.2%, respectively. Drug release profiles proved the pH sensitivity of the nanocomposite by providing a 24–44% difference in drug cumulative release rate between the tumor-simulated and the normal cell-simulated buffers. Also, MTT and the flow cytometry analysis showed the biocompatibility of the carrier and the effect of using both drugs, as a co-delivery system, on cytotoxicity against the MCF-7 cells. These results demonstrate the potential of the chitosan/[email protected]/5-fluorouracil to be a candidate for the targeted treatment of cancer cells.
Article
Harmful side effects of the chemotherapeutic agent have been investigated in many recent studies. Since Fe2O3 nanoparticles have proper porosity, they are capable for loading noticeable amount of drugs and controlled release. We developed Fe2O3/chitosan/montmorillonite nanocomposite. Quercetin (QC) nanoparticles, which have fewer side effects than chemical anti-tumor drugs, were encapsulated in the synthesized nanocarrier and were characterized by X-ray diffraction (XRD), Fourier transforms infrared spectroscopy (FTIR), field emission scanning electron microscopy (FE-SEM), vibrating sample magnetometer (VSM), dynamic light scattering (DLS), and zeta potential. For quercetin, the encapsulation efficiency and the loading efficiency of the drug in Fe2O3[email protected] were found to be about 94% and 57%, respectively. The release profile of QC in different mediums indicated pH-dependency and controlled release of the nanocomposite, adhering to The Weibull kinetic model. Biocompatibility of the Fe2O3/CS/MMT nanoparticles against the MCF-7 cells was shown by MTT assay and confirmed by flow cytometry. These data demonstrate that the designed Fe2O3[email protected] would have potential drug delivery to treat cancer cells.