You are on page 1of 6

Journal of Molecular Structure 1245 (2021) 131040

Contents lists available at ScienceDirect

Journal of Molecular Structure


journal homepage: www.elsevier.com/locate/molstr

Salicylic acid-loaded chitosan nanoparticles (SA/CTS NPs) for breast


cancer targeting: Synthesis, characterization and controlled release
kinetics
Mehdi Hassanpour a,b,¥, Hessam Jafari c,¥, Sina Sharifi d, Jafar Rezaie e,∗,
Zohreh Mehri Lighvan f, Gholam Reza Mahdavinia c, Gholamreza Gohari g, Ali Akbari e,∗
a
Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
b
Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
c
Polymer Research Laboratory, Department of Chemistry, Faculty of Science, University of Maragheh, Maragheh, 55181-83111, Iran
d
Disruptive Technology Laboratory, Massachusetts Eye and Ear and Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School,
Boston, MA, USA
e
Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
f
Department of Polymer Processing, Iran Polymer and Petrochemical Institute, P.O. Box 14965-115, Tehran, Iran
g
Department of Horticulture, Faculty of Agriculture, University of Maragheh, Maragheh, Iran

a r t i c l e i n f o a b s t r a c t

Article history: The present study aimed to develop an easy controlled release system for salicylic acid (SA) through
Received 28 April 2021 ionotropic gelation of chitosan (CTS) using tripolyphosphate (TPP) as cross-linker. The synthesized
Revised 6 June 2021
nanoparticles (SA/CTS NPs) were characterized for their physicochemical properties by various techniques
Accepted 3 July 2021
including Fourier transform infrared spectroscopy (FT-IR), transmission electron microscopy (TEM), scan-
Available online 15 July 2021
ning electron microscopy (SEM) with an energy-dispersive X-ray (EDX) spectroscopy, X-ray photoelectron
Keywords: spectroscopy (XPS) and X-ray diffraction (XRD). A maximum encapsulation efficiency (EE) of 84% was ob-
Biomaterials tained. The obtained material not only was tested as drug delivery system in neutral (pH 7.4) and acidic
Salicylic acid medium (pH 5.5) but also the kinetic release behavior of SA from the CTS NPs was investigated based
Breast cancer on two models namely Korsmeyer-Peppas and Higuchi. These data demonstrated that the drug release
Nanoparticles mechanism governed by Korsmeyer-Peppas model. Finally, the cytotoxic effects of free SA and SA/CTS NPs
Drug delivery
were evaluated in-vitro against breast cancer (MDA-MB-231) cell lines by MTT assay. Results showed that
SA/CTS NPs were more cytotoxic than free SA. This work suggested the potential of SA/CTS NPs system
as an effective anticancer system.
© 2021 Elsevier B.V. All rights reserved.

1. Introduction tumor compounds, but also target the site of tumor specifically
[7,8]. Therefore, alternative and complementary therapies are of-
Breast cancer is the most leading cancer type in females with ten appealing due to the most tumor cells are aggressive and resis-
high rate of mortality in almost all countries worldwide [1–3]. tant to therapies [9]. Recently, nanoparticle-mediated drug delivery
Antitumor chemotherapeutics compounds when administrated by systems have gained massive attentions for targeting the drug to
conventional drug delivery ways face several problems such as the cancer cells through cell signaling pathways. Till now, different
non-specificity, high toxicity, and chemoresistance [4]. In the past nanoparticles (NPs) such as solid lipid NPs, nanotubes, nanoshells,
two decades, the reduction of bulk particles to the nanoscale nanoballs, nanorods, polymeric NPs, nanopillars and many more
known as nanotechnology [5] has emerged as a promising strategy have been reported as efficient and tunable devices in drug de-
in drug delivery system to overcome aforementioned problems [6]. livery systems [10–12]. Among them, polysaccharide-based poly-
On the other hand, when these nanoscale materials act as nanocar- meric NPs exhibited some advantageous in terms of biocompati-
riers not only improve the efficiency and bioavailability of anti- bility and biodegradability and were used extensively for biomedi-
cal applications [13,14]. Chitosan (CTS) as an abundant natural lin-
ear polysaccharide not only could be extracted from the exoskele-

Corresponding author. ton of insects, fungi and arthropods but also may be chemically
E-mail address: akbari.a@umsu.ac.ir (A. Akbari).
¥
synthesized by the deacetylation of chitin [15–17]. The nontoxic,
These authors worked equally

https://doi.org/10.1016/j.molstruc.2021.131040
0022-2860/© 2021 Elsevier B.V. All rights reserved.
M. Hassanpour, H. Jafari, S. Sharifi et al. Journal of Molecular Structure 1245 (2021) 131040

biocompatibility, biodegradation properties of CTS and the exis- electron microscopy/energy dispersive X-ray instrument (SEM/EDS,
tence of a high density of positive charge in acidic media related VEGAII, XMU, Czech Republic).
to the amine groups on its backbone lead to increase massive at-
tention to the applications of CTS-based NPs in the biomedical and 2.2. Synthesis of SA-loaded CTS NPs (SA/CTS NPs)
pharmaceutical field [18–20]. Typically, CTS NPs are synthesized by
using various cross-linking agents such as polyaspartic acid, glu- Briefly, in 50 ml round bottom flask containing acetic acid
taraldehyde and tripolyphosphate (TPP) [21–23]. It should be high- (0.5%) in 10 ml distilled water, 0.5 g of CTS and 0.1 g of SA (0.05%)
lighted that the ionic gelation is the most investigated method and were dissolved and stirred at 100 rpm overnight. Finally, the pre-
extensively used for preparing CTS NPs. In this method, cationic pared stock TPP solution (0.1 g of TPP in 10 ml) was added to
CTS could interact with multivalent polyanions leading to form CTS/SA solution slowly. After stirring for 120 min, final solution
CTS NPs under simple and mild conditions [24,25]. Because of its was centrifuged at 12,0 0 0 rpm for 60 min. Resulted SA/CTS NPs
fast gelling ability and environmentally friendly property, TPP is were freeze-dried and kept in the sealed vial for the next usages.
the most studied polyanion that used in ionic gelation method Moreover, the supernatant was collected in order to calculate the
[26]. CTS-TPP NPs have been previously proved to be exceptional amount of un-loaded SA by measuring its absorbance at 297 nm
carriers throughout encapsulation of various drugs and nutrients using spectrophotometer. Finally, standard plot was utilized to cal-
such as curcumin [27], quercetin [28], tea catechins [29], essen- culate the concentration of SA and EE% was obtained using follow-
tial oil [30] and ascorbic acid [31]. Very recently, we used CTS NPs ing equation:
in the presence of hydroxypropyl methylcellulose as an efficient T otal concent rat ion o f SA − unloaded SA
pH-sensitive drug delivery system for melatonin release to MDA- EE% = × 100
T otal concent rat ion o f SA
MB 231 breast cancer cells [32]. Salicylic acid (SA), colorless crys-
talline mono-hydroxybenzoic acid, is an important phenolic com- 2.3. In vitro cytotoxicity assay and Statistical analysis
pound and typically utilized as plant hormones in agricultural ap-
plications [33]. However, its ability to reduce fevers and pains has The cells were cultured in 96 well culture plates (10 0 0 0 cells
been demonstrated previously [34,35] and the anti-cancer activity /well) and incubated in CO2 incubator. Next day, supernatants were
of SA as chemotherapeutic drug was studied by Kutlu and cowork- discarded and replaced with treatment medium. Then, at respec-
ers towards A549 human lung adenocarcinoma cell [36]. In this tive time point (48 h), 100 μl MTT solution (5 mg/ml) was added
study, the simple ionic gelation method was used to prepare CTS into each well and cells were kept in CO2 incubator in the dark
NPs containing desired amount of SA (SA/CTS NPs with EE = 84%). for 4 h. Further, supernatant was discarded and 100 μl DMSO was
Both SA release profile and release kinetics from CTS NPs (in neu- added to dissolve formazan crystals formed by the cells in each
tral and acidic medium) were studied. To our knowledge, the cyto- well. The absorbance was read at 570 nm using a microplate reader
toxic effect of SA encapsulated CTS NPs against human breast can- (BioTek). The cell viability was expressed as percentage (%) of con-
cer MDA-MB-231 cell line in vitro has not previously investigated trol.
and this study is the first report toward this aim. The viability of cells was calculated using the following equa-
tion:
V iabilit y(% ) = (ODamount o f t reatedcel l s)/(ODamount o f cont rol cel l s) × 100
2. Experimental
In vitro cytotoxic activity was assessed by the calculation of
2.1. Materials and instruments the inhibitory concentration of either SA or SA/CTS NPs required
to cause 50% of cells death recognized as the half maximal in-
All chemicals such as salicylic acid, chitosan (Molecular weight hibitory concentration (IC50). IC50 were calculated by Graph pad
of 310–375 kDa, 75–85% deacetylated) and Sodium Tripolyphos- software ver.8. Results were analyzed by SPSS 16.0 software (SPSS
phate (TPP) were obtained from Sigma-Aldrich (USA). Other chem- Inc., Chicago, IL) and expressed as mean ± SD from three indepen-
icals used in this project were all of analytical grade and no pu- dent experiments carried out in triplicate. One-way Analysis of
rification was applied. Human breast cancer cell line MDA-MB-231 Variance (ANOVA) and Tukey post hoc test were used to determine
(Pasteur Institute, Iran) were grown in Dulbecco’s modified Ea- the significance of differences between groups with significances of
gle’s medium (DMEM, Gibco) containing 10% fetal bovine serum P < 0.05.
(DMEM 10% FBS) (Gibco) and 1% penicillin/streptomycin (P/S). Cells
were kept in a humidified incubator of 5% CO2 at 37 °C. To treat 3. Results and discussion
cells with Salicylic acid (SA) and SA loaded Chitosan (SA/CTS NPs),
cells were co-cultured with different concentrations of either SA or 3.1. Preparation and characterization of SA/CTS NPs
SA/CTS NPs (10,20,40,80,160,320,640 ppm) over a period of 48 h.
Non-treated cells were considered as a control group. Furthermore, Fig. 1 illustrates schematic diagrams for preparation of SA/CTS
the half maximal inhibitory concentration (IC50) of both SA and NPs through ionic gelation method due to opposite charges of CTS
SA/CTS NPs for 48 h of post treatment were analyzed by Graph and TPP leading to successful encapsulation of SA. Moreover, the
pad software ver.8. All experiments were performed in triplicate. interaction of TPP and carboxylic acid group of SA with primary
The existence of functional groups of SA/CTS NPs was studied us- amine of CTS can be clearly seen from Fig 1.
ing Fourier transform infrared (FT-IR) spectra (Bruker 113V FT-IR The chemical composition of samples was elucidated by FTIR
spectrometer). Freeze-dried and powdered samples were mixed analysis in Fig 2A. A strong peak at the 3428 cm−1 corresponds
with dried KBr (1:99) and the data was collected from 500– to N-H and O-H stretching, as well as the intramolecular hydro-
3500 cm−1 wave numbers with 1 cm−1 resolution. For the crys- gen bonds. This peak is sharper in the CTS NPs indicating that the
tal phase investigation, Powder X-ray diffraction (XRD) pattern of hydrogen bonding is enhanced [37]. The peaks around 2921 cm−1
samples was obtained on a Siemens D-500 X-ray diffractometer and 2861 cm−1 can be attributed to C-H symmetric and asymmet-
(λ = 1.54 Å (CuKα ), voltage of 35 kV). TEM image of SA/CTS NPs ric stretching, respectively. Moreover, the presence of residual N-
was recorded with the help of transmission electron microscopy acetyl groups was confirmed by the peaks at around 1656 cm−1
(TEM; Philips CM10 operating at 60 kV tension). The morphol- (C=O stretching of amide I) and 1596 cm−1 (NH2 ) groups, re-
ogy and the elemental composition were evaluated using scanning spectively. These peaks shift hypsochromically to 1612 cm−1 and

2
M. Hassanpour, H. Jafari, S. Sharifi et al. Journal of Molecular Structure 1245 (2021) 131040

TEM image of SA/CTS NPs exhibited spherical shapes with the av-
erage size about 170–180 nm (Fig 3D and 3E).

3.2. In-vitro SA loading/ release studies and kinetic modeling

Standard calibration curve was used to calculate the SA encap-


sulation efficiency (EE %) value on CTS NPs (Fig 4A). The maximum
EE% of SA was found to be 84%. Fig 4B showed the time-dependent
release profiles of the SA/CTS NPs at two different pH (5.5 and 7.4)
at 37 °C. As it could be seen, the release of SA from CTS NPs is pro-
longed and sensitive to the pH of the medium. In both medium,
the initial fast release was obtained in the first 5 h possibly be-
cause of the SA-located at the surface of CTS NPs, and during this
period approximately 18% and 35% of the SA was released from CTS
NPs in pH of 5.5 and 7.4, respectively. After 22 h, nearly, 68% and
Fig 1. General procedure for SA/CTS NPs synthesis.
31% drug release were observed from the CTS NPs in pH of 5.5 and
7.4, respectively. Obtained results clearly demonstrated that SA/CTS
NPs have remarkably prolonged persistent and higher amount of
1537 cm−1 in the FTIR spectra of SA/CTS NPs which caused by the release at pH 5.5 than pH 7.4, which made it promising drug de-
interaction between NH3+ groups of CTS and phosphate groups livery system for cancer therapy.
of TPP [37, 38]. This interaction is also supported by the de- Korsmeyer-Peppas and Higuchi kinetic models was applied to
crease of amide I (1612 cm−1 ) peak intensity in SA/CTS NPs com- prove and explain the mechanism of SA release from CTS NPs (Eq.
pared to CTS. The CH2 bending and CH3 symmetrical deforma- 1, Eq. 2, Fig 4C and 4D) [43].
tions were confirmed by the presence of bands at around 1429 and Mt/M0 = KKP t n (Eq. 1)
1373 cm−1 , respectively [39]. In addition, FTIR spectrum of SA/CTS
NPs showed the characteristic peaks of TPP at 1215 cm−1 (stretch-
ing vibration of P=O), 1160 cm−1 (symmetric stretching vibrations F = KH × t 1/2 (Eq. 2)
in the O−P=O group), 1080 cm−1 (symmetric stretching vibrations
of the PO3 group) and 888 cm−1 (asymmetric stretching vibration Mt showed the amount of drug release at time t. F is the frac-
of the P−O −P bridge) [40, 41].. According to the Fig 2B, the XRD tion of drug release at time t. KKP and KH are the rate constants
patterns of CTS exhibited typical broad peak at 2θ of 20°. After en- of Korsmeyer-Peppas and Higuchi kinetic models, respectively. The
capsulation of SA into the CTS NPs, some intense peaks in the XRD data extracted from Korsmeyer-Peppas model such as the kinetic
pattern of SA/CTS NPs at 2θ of 11°, 17°, 23° and 28° [42] related to constant (K) and the characteristic index of the liberation (n) could
the SA could be seen in which suggested the crystalline nature of provide important information regarding the mechanism of drug
SA in the synthesized SA/CTS NPs. release from CTS NPs. Generally, when n < 0.45, the drug release
Surface morphology and elemental composition of SA/CTS NPs mechanism is related to the Fick’s diffusion. When 0.45 < n < 0.89,
were investigated using SEM and EDX analysis, respectively. From the non- Fick’s diffusion mechanism was occurred and the drug
Fig 3A, SA/CTS NPs illustrated slightly rough surface with the release mechanism is controlled by non- Fick’s diffusion. Finally,
spherical particles. Moreover, the existence of C, O, N and P ele- when n ˃ 0.89 the drug release mechanism belongs to the skele-
ments in the EDX analysis indicated successful formation of NPs by ton erosion [44].
ionic gelation in the presence of TPP and the SA encapsulation in Fig 4C and 4D showed the linear fit of Korsmeyer-Peppas and
the CTS NPs structure (Fig 3B). In addition, data obtained from XPS Higuchi kinetic models to in-vitro experimental SA drug release re-
analysis displayed CTS and TPP as C, O, N and P in NPs (Fig 3C). sults, respectively. Moreover, the calculated kinetic parameters for

Fig 2. (A) FTIR spectra of CTS, SA, SA/CTS NPs and (B) XRD patterns of CTS and SA/CTS NPs.

3
M. Hassanpour, H. Jafari, S. Sharifi et al. Journal of Molecular Structure 1245 (2021) 131040

Fig 3. (A, B and C) SEM-EDX and XPS analysis of SA/CTS NPs, (D and E) TEM image and size distribution of SA/CTS NPs.

Fig 4. (A and B) standard calibration curve and SA release profiles in two different pH of 5.5 and 7.4. (C and D) Kinetic models for SA release from CTS NPs for varying pH
conditions: (C) Korsmeyer-Peppas model and (D) Higuchi model.

4
M. Hassanpour, H. Jafari, S. Sharifi et al. Journal of Molecular Structure 1245 (2021) 131040

Fig 5. In-vitro cell viability of MDA-MB-231 cell after incubation with (A) free SA and (B) SA/CTS NPs for 48h in different concentrations.

Table 1 Peppas model. For the first time, the toxicity effects of synthe-
The calculated kinetic parameters of salicylic acid release according to Higuchi
sized sample were evaluated towards MDA-MB-231 breast cancer
and Korsmeyer-Peppas models.
cell line by MTT assay. The results of this investigation show the
Models Higuchi model Korsmeyer-Peppas model potential of SA encapsulated CTS NPs as anti-cancer drug delivery
k r2 n k r2
system towards various human breast cancer cell lines.
SA/CTS NPs pH = 5.5 0.850 0.95 0.467 1.043 0.99
pH = 7.4 1.443 0.93 0.641 0.623 0.98
Declaration of Competing Interest

studied models were summarized in Table 1. Based on the corre- The authors report no declaration of interest.
lation coefficients, r2 , the release of SA from SA/CTS NPs in pH 5.5
and 7.4 seems to follow Korsmeyer-Peppas model. Based on the Reference
data from Table 1, the value of ‘n’ is 0.467 and 0.641 at pH of 5.5
and 7.4, respectively which revealed the non-Fickian transport of [1] Z. Momenimovahed, H. Salehiniya, Epidemiological characteristics of and risk
SA from CTs NPs to in vitro solutions. factors for breast cancer in the world, Breast Cancer 11 (2019) 151.
[2] N. Jabbari, E. Akbariazar, M. Feqhhi, R. Rahbarghazi, J. Rezaie, Breast can-
cer-derived exosomes: Tumor progression and therapeutic agents, J. Cell. Phys-
3.3. Cellular cytotoxicity studies using MTT assay iol. 235 (10) (2020) 6345–6356.
[3] M. Khaksar, M. Sayyari, J. Rezaie, A. Pouyafar, S. Montazersaheb, R. Rahbarg-
After successful preparation and characterization of SA/CTS NPs, hazi, High glucose condition limited the angiogenic/cardiogenic capacity of
murine cardiac progenitor cells in in vitro and in vivo milieu, Cell Biochem.
the ability of the SA/CTS NPs to release SA and induce cell cytotox- Funct. 36 (7) (2018) 346–356.
icity was investigated in MDA-MB-231 breast cancer cell line by [4] H.L. Wong, R. Bendayan, A.M. Rauth, Y. Li, X.Y. Wu, Chemotherapy with anti-
MTT assay (Fig 5 A and 5B). The results showed that both free SA cancer drugs encapsulated in solid lipid nanoparticles, Adv. Drug. Deliv. Rev.
59 (6) (2007) 491–504.
and SA/CTS NPs had a dose-dependent toxicity toward the viabil- [5] A. Akbari, N. Arsalani, Preparation and characterization of novel hy-
ity of MDA-MB-231 breast cancer cell after 48 h incubation. Fur- brid nanocomposites by free radical copolymerization of vinyl pyrrolidone
thermore, our results demonstrated that IC50 value for free SA and with incompletely condensed polyhedral oligomeric silsesquioxane, J. Inorg.
Organomet. Polym Mater. 26 (3) (2016) 536–544.
SA/CTS NPs was 513 ppm and 445 ppm, respectively. Furthermore, [6] B.N. Ho, C.M. Pfeffer, A.T. Singh, Update on nanotechnology-based drug deliv-
cell viability for drug-loaded CTS NPs is relatively lower than free ery systems in cancer treatment, Anticancer Res. 37 (11) (2017) 5975–5981.
SA. These results show that CTS NPs can facilitate the delivery of [7] J.K. Patra, G. Das, L.F. Fraceto, E.V.R. Campos, M. del Pilar Rodriguez-Torres,
L.S. Acosta-Torres, L.A. Diaz-Torres, R. Grillo, M.K. Swamy, S. Sharma, Nano
SA into the cancer cells with good efficiency leading to high cyto-
based drug delivery systems: recent developments and future prospects, J.
toxic effects towards MDA-MB-231 cells in comparison to free SA Nanobiotechnol. 16 (1) (2018) 71.
over a period of 48 h. [8] A. Butu, S. Rodino, D. Golea, M. Butu, M. Butnariu, C. Negoescu, C.-E. Din-
u–Pirvu, Liposomal nanodelivery system for proteasome inhibitor anticancer
drug bortezomib, Farmacia 63 (2) (2015) 224–229.
4. Conclusion [9] N. Chatterjee, T.G. Bivona, Polytherapy and targeted cancer drug resistance,
Trends Cancer 5 (3) (2019) 170–182.
Today, design and fabrication of smart drug delivery systems [10] A. Akbari, M. Padervand, E. Jalilian, F. Seidi, Sodium alginate-halloysite nan-
otube gel beads as potential delivery system for sunitinib malate anticancer
are known as one of the most promising objects. Chitosan (CTS) compound, Mater. Lett. 274 (2020) 128038.
nanoparticles have been extensively investigated for the encapsu- [11] K. Ariga, Q. Ji, M.J. McShane, Y.M. Lvov, A. Vinu, J.P. Hill, Inorganic nanoarchi-
lation of numerous compounds such as drugs and nutrients. In tectonics for biological applications, Chem. Mater. 24 (5) (2012) 728–737.
[12] P. Couvreur, Nanoparticles in drug delivery: past, present and future, Adv.
this work, salicylic acid (SA) has been effectively loaded into CTS
Drug. Deliv. Rev. 65 (1) (2013) 21–23.
NPs through simple ionic gelation method in the presence of TPP [13] T. Miao, J. Wang, Y. Zeng, G. Liu, X. Chen, Polysaccharide-based controlled re-
(SA/CTS NPs) and characterized using various techniques. Optical lease systems for therapeutics delivery and tissue engineering: from bench to
bedside, Adv. Sci. 5 (4) (2018) 1700513.
microscopy observations showed that the synthesized SA/CTS NPs
[14] K. Ganguly, K. Chaturvedi, U.A. More, M.N. Nadagouda, T.M. Aminab-
had spherical shapes with the average size about 170–180 nm. havi, Polysaccharide-based micro/nanohydrogels for delivering macromolecular
The sustained release of SA from CTS NPs was investigated in pH therapeutics, J. Control. Release 193 (2014) 162–173.
5.5 and 7.4. Obtained results showed that the maximum cumula- [15] Z. Ahmadi, M. Saber, A. Akbari, G.R. Mahdavinia, Encapsulation of Satureja
hortensis L. (Lamiaceae) in chitosan/TPP nanoparticles with enhanced acaricide
tive SA release rate was 68% and 31% in pH 5.5 and pH 7.4, re- activity against Tetranychus urticae Koch (Acari: Tetranychidae), Ecotoxicol. En-
spectively. The kinetic release mechanism governed by Korsmeyer- viron. Saf. 161 (2018) 111–119.

5
M. Hassanpour, H. Jafari, S. Sharifi et al. Journal of Molecular Structure 1245 (2021) 131040

[16] W. Chen, Y. Li, S. Yang, L. Yue, Q. Jiang, W. Xia, Synthesis and antioxidant prop- [31] M. Sun, Q. Xie, X. Cai, Z. Liu, Y. Wang, X. Dong, Y. Xu, Preparation and charac-
erties of chitosan and carboxymethyl chitosan-stabilized selenium nanoparti- terization of epigallocatechin gallate, ascorbic acid, gelatin, chitosan nanopar-
cles, Carbohydr. Polym. 132 (2015) 574–581. ticles and their beneficial effect on wound healing of diabetic mice, Int. J. Biol.
[17] P. Negrea, A. Caunii, I. Sarac, M. Butnariu, The study of infrared spectrum of Macromol. 148 (2020) 777–784.
chitin and chitosan extract as potential sources of biomass, Digest J. Nano- [32] H. Jafari, M. Hassanpour, A. Akbari, J. Rezaie, G. Gohari, G.Reza Mahdavinia,
mater. Biostruct. (DJNB) 10 (4) (2015). E. Jabbari, Characterization of pH-sensitive chitosan/hydroxypropyl methylcel-
[18] S. Shah, A. Pal, V. Kaushik, S. Devi, Preparation and characterization of ven- lulose composite nanoparticles for delivery of melatonin in cancer therapy,
lafaxine hydrochloride-loaded chitosan nanoparticles and in vitro release of Mater. Lett. 282 (2021) 128818.
drug, J. Appl. Polym. Sci. 112 (5) (2009) 2876–2887. [33] M. Rivas-San Vicente, J. Plasencia, Salicylic acid beyond defence: its role in
[19] S. Naskar, S. Sharma, K. Kuotsu, Chitosan-based nanoparticles: an overview of plant growth and development, J. Exp. Bot. 62 (10) (2011) 3321–3338.
biomedical applications and its preparation, J. Drug Delivery Sci. Technol. 49 [34] R. Hosseinzadeh, K. Khorsandi, M. Jahanshiri, Combination photodynamic ther-
(2019) 66–81. apy of human breast cancer using salicylic acid and methylene blue, Spec-
[20] P. Beulah, U. Jinu, M. Ghorbanpour, P. Venkatachalam, Green engineered chi- trochim. Acta Part A 184 (2017) 198–203.
tosan nanoparticles and its biomedical applications—an overview, Adv. Phyto- [35] T. Hedner, B. Everts, The early clinical history of salicylates in rheumatology
nanotechnol. (2019) 329–341. and pain, Clin. Rheumatol. 17 (1) (1998) 17–25.
[21] N. Islam, H. Wang, F. Maqbool, V. Ferro, In vitro enzymatic digestibility of glu- [36] D. Vejselova, H.M. Kutlu, Inhibitory effects of salicylic acid on A549 human
taraldehyde-crosslinked chitosan nanoparticles in lysozyme solution and their lung adenocarcinoma cell viability, Turkish J. Biol. 39 (1) (2015) 1–5.
applicability in pulmonary drug delivery, Molecules 24 (7) (2019) 1271. [37] C. Lustriane, F.M. Dwivany, V. Suendo, M. Reza, Effect of chitosan and chitosan–
[22] J.J. Joseph, D. Sangeetha, T. Gomathi, Sunitinib loaded chitosan nanoparticles nanoparticles on post harvest quality of banana fruits, J. Plant Biotechnol. 45
formulation and its evaluation, Int. J. Biol. Macromol. 82 (2016) 952–958. (1) (2018) 36–44.
[23] C. Pan, J. Qian, C. Zhao, H. Yang, X. Zhao, H. Guo, Study on the relationship be- [38] S. Trabelsi, N. Issaoui, S.A. Brandán, F. Bardak, T. Roisnel, A. Atac, H. Marouani,
tween crosslinking degree and properties of TPP crosslinked chitosan nanopar- Synthesis and physic-chemical properties of a novel chromate compound with
ticles, Carbohydr. Polym. 241 (2020) 116349. potential biological applications, bis (2-phenylethylammonium) chromate (VI),
[24] S.K. Shukla, A.K. Mishra, O.A. Arotiba, B.B. Mamba, Chitosan-based nanomate- J. Mol. Struct. 1185 (2019) 168–182.
rials: A state-of-the-art review, Int. J. Biol. Macromol. 59 (2013) 46–58. [39] K. Karrouchi, S.A. Brandán, Y. Sert, H. El-marzouqi, S. Radi,
[25] K.G. Desai, Chitosan nanoparticles prepared by ionotropic gelation: an M. Ferbinteanu, M.E.A. Faouzi, Y. Garcia, Synthesis, X-ray struc-
overview of recent advances, Critical Rev. Ther. Drug Carrier Syst. 33 (2) (2016). ture, vibrational spectroscopy, DFT, biological evaluation and
[26] N.B.M. Agardan, Studies on the formulation optimization and controlled ionic molecular docking studies of (E)-N’-(4-(dimethylamino) benzyli-
gelation of chitosan nanoparticles using TPP-HP-[beta]-CD inclusion complex, dene)-5-methyl-1H-pyrazole-3-carbohydrazide, J. Mol. Struct. 1219 (2020)
J. Faculty Pharm. Istanbul University 50 (1) (2020) 54–60. 128541.
[27] N. Alizadeh, S. Malakzadeh, Antioxidant, antibacterial and anti-cancer activities [40] S.A. Loutfy, H.M.A. El-Din, M.H. Elberry, N.G. Allam, M. Hasanin, A.M. Abdellah,
of β -and γ -CDs/curcumin loaded in chitosan nanoparticles, Int. J. Biol. Macro- Synthesis, characterization and cytotoxic evaluation of chitosan nanoparticles:
mol. 147 (2020) 778–791. in vitro liver cancer model, Adv. Nat. Sci. 7 (3) (2016) 035008.
[28] A. Choudhary, V. Kant, B.L. Jangir, V. Joshi, Quercetin loaded chitosan [41] Y. Xu, Y. Du, Effect of molecular structure of chitosan on protein delivery prop-
tripolyphosphate nanoparticles accelerated cutaneous wound healing in Wistar erties of chitosan nanoparticles, Int. J. Pharm. 250 (1) (2003) 215–226.
rats, Eur. J. Pharmacol. 880 (2020) 173172. [42] J. Ji, S. Hao, D. Wu, R. Huang, Y. Xu, Preparation, characterization and in vitro
[29] N.P. da Silva, E.d.C.R.L. Pereira, L.M. Duarte, J.C. de Oliveira Freitas, C.G. de release of chitosan nanoparticles loaded with gentamicin and salicylic acid,
Almeida, T.P. da Silva, R.C. Melo, A.C.M. Apolônio, M.A.L. de Oliveira, Carbohydr. Polym. 85 (4) (2011) 803–808.
H. de Mello Brandão, Improved anti-Cutibacterium acnes activity of tea tree [43] S. Alinavaz, G.R. Mahdavinia, H. Jafari, M. Hazrati, A. Akbari, Hydroxyapatite
oil-loaded chitosan-poly (ε -caprolactone) core-shell nanocapsules, Colloids (HA)-based hybrid bionanocomposite hydrogels: ciprofloxacin delivery, release
Surf. B 196 (2020) 111371. kinetics and antibacterial activity, J. Mol. Struct. 1225 (2021) 129095.
[30] Z. Ahmadi, M. Saber, A. Akbari, G.R. Mahdavinia, Encapsulation of Satureja [44] A. Soumia, M. Adel, S. Amina, B. Bouhadjar, D. Amal, Z. Farouk, B. Abdelkader,
hortensis L.(Lamiaceae) in chitosan/TPP nanoparticles with enhanced acaricide S. Mohamed, Fe3O4-alginate nanocomposite hydrogel beads material: One-pot
activity against Tetranychus urticae Koch (Acari: Tetranychidae), Ecotoxicol. En- preparation, release kinetics and antibacterial activity, Int. J. Biol. Macromol.
viron. Saf. 161 (2018) 111–119. 145 (2020) 466–475.

You might also like