You are on page 1of 12

Materials Chemistry and Physics 263 (2021) 124381

Contents lists available at ScienceDirect

Materials Chemistry and Physics


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

Novel topical and transdermal delivery of colchicine with chitosan based


biocomposite nanofiberous system; formulation, optimization,
characterization, ex vivo skin deposition/permeation, and
anti-melanoma evaluation
Hamed Morad a, c, Mohsen Jahanshahi b, *, Jafar Akbari a, Majid Saeedi a, Pooria Gill d,
Reza Enayatifard a, **
a
Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
b
Nanotechnology Research Institute, Faculty of Chemical Engineering Babol Noushirvani University of Technology, Babol, Iran
c
Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
d
Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran

H I G H L I G H T S G R A P H I C A L A B S T R A C T

• Colchicine-loaded chitosan-based opti­


mized nanofibers depict anti-melanoma
efficiency.
• Topical delivery of colchicine by nano­
fibers shows a significant skin
deposition.
• Significant transdermal permeation of
colchicine observed from composite
nanofibers.
• Transdermal release of colchicine from
nanofibers complies with the first-order
model.

A R T I C L E I N F O A B S T R A C T

Keywords: Melanoma bears the highest mortality rate in skin cancers. The unique properties of chitosan-based nanofibers
Melanoma provide a great potential to formulate an effective topical drug delivery system. With the emphasis on Colchicine
Nanofiber systemic toxicity as an obstacle against the administration in systemic chemotherapy and considering its supe­
Colchicine
riority over approved chemotherapeutic agents for melanoma management, it was chosen to be formulated in a
Topical
Transdermal
nanofiber based topical drug delivery system. The optimization assay was conducted by AFM and SEM based on
the morphology, topographic data, and mean diameter size of the nanofibers. Other characterization studies
include FTIR, XRD, STA, contact angle measurement, tensile test, ex vivo skin permeation, deposition analysis,
release kinetic and anti-melanoma efficiency against A-375 cell line. As a result, significant colchicine deposition
in the skin with remarkable cytotoxicity against melanoma cell line makes it a desirable formulation to be
administered as a topical or local reservoir system for neoadjuvant chemotherapy before other interventions and
adjuvant therapy of tumors after surgery and, also, for other skin diseases with dose adjustment. Besides, the

* Corresponding author.
** Corresponding author.
E-mail addresses: mjahan@nit.ac.ir (M. Jahanshahi), enayatifard_r@yahoo.com (R. Enayatifard).

https://doi.org/10.1016/j.matchemphys.2021.124381
Received 12 November 2020; Received in revised form 20 January 2021; Accepted 8 February 2021
Available online 10 February 2021
0254-0584/© 2021 Elsevier B.V. All rights reserved.
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

observed first order release kinetic behavior through the skin could suggest it as a transdermal colchicine de­
livery system which improves its efficiency in systemic indications.

1. Introduction transdermal patches. The encapsulation of drugs within the NFs in


amorphous manner would prevent this problem [17,19,20]. Besides,
Melanoma is the deadliest skin cancer. It bears a high recurrence NFs provide more stability for drugs and protects sensitive and potent
rate, rapid progression, and a very high metastatic potential compared active pharmaceutical ingredients (API). Interestingly, Both hydrophilic
with other malignancies, whose fatality time is less than a year [1]. The and lipophilic drugs could be loaded in various biocompatible and
first-line and second-line FDA-approved drugs like Dacarbazine and biodegradable polymers with such inexpensive technology [18].
Paclitaxel-Carboplatin combination have not significantly altered fa­ NF-based carriers for localized delivering of chemotherapeutic agents
tality period [1,2]. BRAF inhibitors like Vemurafenib or Dabrafenib and have some additional advantages, like stability boosting of loaded
MEK inhibitors like Trametinib showed very limited responses [3]. chemotherapeutic agents with maintenance of its cytotoxic property,
Evaluation of microtubule-mediated chemotherapeutic agents demon­ controlled and targeted tumor delivery, low systemic side effects and
strates that the clinical efficiency of drugs interacting with vinca or toxicity, single-dose therapy, and reducing administered dose in the
taxane binding cites of microtubules has been decreased. The observed formulation [21].
chemoresistance is related to over expression of P-glycoprotein (P-gp), As found by latest studies, achieving a distinct and optimized
multidrug resistance-associated protein (MRP) and/or breast cancer nanofibers and reducing their diameter size is critical for fabricating
resistance protein (BCRP) [4]. Attractively, Colchicine (CL), with its applicable nanofibrous mats with perfect efficiency [22,23]. On the
specific binding site on microtubules, reported to be less sensitive to the other hand, in the recent study of Zhu.et al., the loaded Chitosan (CS) in
mentioned resistance mechanism. Moreover, it has extra anti-vascular the structure of nanofibrous delivery system of 5-FU against melanoma
activity, enhanced water solubility, and simple structure in compari­ exhibited remedying effect on normal skin cells [24]. Therefore, the aim
son with the two other groups [5]. CL, as a well-known tricyclic pseu­ of this study is to precisely optimize CL-loaded CS-based Composite-NFs
doalkaloid that could be extracted from Colchicum autumnale (autumn and fully characterize and evaluate it as an efficient topical and trans­
crocus) and Gloriosa superba L, has numerous adopted indications in dermal nano drug delivery system. As a transdermal formulation, it
systemic and topical diseases. In addition to gout, pseudogout, familial could be suggested for general diseases’ management with the dose
Mediterranean fever, and amyloidosis, as old therapeutic indications, range of 0.015 mg/kg up to less than 0.1 mg/kg like gout, osteoarthritis,
recently a multitude of novel indications have been reported for pericarditis, Behcet’s disease, Familial Mediterranean fever, hepatic
immunology, cardiology, oncology and dermatology. The dermatolog­ diseases, atherosclerosis and cardiovascular diseases or even localized
ical diseases include Actinic keratosis, Behcet’s disease, Chronic urti­ targeting inflammations managements with minimum systemic side
carial, Epidermolysis bullosa acquisita, Granuloma annulare, effects. Although, as mentioned above, CL seems to be a great choice for
Henoch–Schonlein purpura, Idiopathic plantar eccrine hidradenitis, melanoma management, the main factor that has limited its being
Linear IgA, Neutrophilic urticarial, Nodular vasculitis, Purpura annu­ applied in cancer therapies is that at high doses it could cause severe
laris telangiectoides, Pyoderma gangrenosum, Recurrent aphthous sto­ toxicity to normal cells; thus, systemic administration of CL for cancer
matitis, Relapsing polychondritis, Scleredema, Scleredema therapy has been impossible so far. Consequently, there is the need to
diabeticorum, Sweet syndrome, and Urticarial vasculitis [6–8]. jettison the obstacle against safe administration of the natural based
Applications of nanofiber-based fabrications are growing in a variety drug of CL [7,8,25]. Therefore, as one of the aims of this study, the
of fields. However, one of the most notable areas is the use of nanofibers nanofiber based formulation of CL, using CS as polymer, may be sug­
(NFs) in drug delivery, including transdermal, topical, oral, ocular and gested for localized and targeted management of not only all mentioned
even local injection systems. There are numbers of advantages attrib­ topical skin diseases, but also, especially, as a neoadjuvant and adjuvant
uted to nanofibers over conventional transdermal drug delivery systems therapy in melanoma before and after other treatments, like tumor
(TDDS), like transdermal patches containing gel, metered liquid spray or excision, radiotherapy or systemic chemotherapies particularly for
other topical formulations and also pressure sensitive adhesive (PSA) effective inhibition of tumor recurrence with sustain releasing the lower
matrix type TDDSs or even other nanostructures, like nanotubes, doses than the dose range of 0.1 mg/kg - 0.8 mg/kg in wide range of
nanocrystals, nanofilms, nanorods, nanospheres, liposomes, noisomes, cancers from melanoma to any carcinomas [8].
solid lipid nanoparticles, nanostructured lipid carriers, dendrimers, etc.
These features for NF-based scaffolds includes large surface area-to- 2. Materials
volume ratio, flexibility in achieved mat, great porosity, high entrap­
ment efficiency, the lowest polydispersity in fiber’s diameter size with CL (HPLC grade), Chitosan (CS) with medium molecular weight,
various compositions and bearing potential of being functionalized with Dulbecco’s Modified Eagle’s Medium (DMEM, high glucose), 3-(4,5-
biological agents and simulating the condition of extracellular matrix of dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), and
skin and, on the other hand, the capability of controlling drug release dimethyl sulfoxide (DMSO) were purchased from Sigma–Aldrich Co. (St.
rate. These unique properties of nanofibrous carriers yield an effective Louis, MO, U.S.A.). Poly vinyl alcohol (PVA) with 27000 molecular
and special reservoir of drug over skin with optimum absorption and weight and Acetic acid (glacial) were bought from Merck (Germany). A-
great distribution of drug molecules from nanometric fibers over the 375 cell line was obtained from Pasteur institute (Medical research
skin surface that would be dedicated to more parts of skin area per unit center, Tehran, Iran). Ultra-pure water was utilized in all the proced­
of time, which increases the drug internalization and bioavailability as ures. All the solvents and reagents applied had analytical grade in
topical or transdermal drug delivery systems, which improves the accordance with United States’ pharmacopeia standards.
therapeutic effects [9–16]. Moreover, NFs compared with other
mentioned TDDSs, not only could provide high occlusive effect, which 3. Methods
relates to its high surface area, but also brings great breathability to the
skin which refers to high porosity of the NFs [17]. The high porosity 3.1. Optimization process of pristine and CL-loaded PVA/CS composite
could also help to absorb wounds exudates in topical formulations and NFs
better cell attachment, which improves the healing period [17,18]. In
addition, drug crystallization is a major challenge of conventional The assembled electrospinning apparatus (PN 04, Pars Nano Ris Co,

2
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

Iran) consisted of a 5 ml syringe with 140 mm inner diameter and 10 ◦ C/min.


aluminum foil as the collector. The flow rate was kept constant at 0.2
ml/h and the process was performed at room temperature and 50% 3.7. Water angle measurement
relative humidity. The optimization was conducted in three steps based
on various voltages (10, 12, 15, 18 and 20 kv) and nozzle-collector The water contact angle of PVA mats alone, pristine PVA:CS Com­
distances (10, 12, 15, 20 cm) of electrospinning. The qualified NFs posite mats and CL-loaded Composite mats were carried out in a contact
were chosen by considering the AFM analysis results, including mean angle measurement apparatus (CA-500 M, Sharif solar Co, Iran) by
and distribution of diameters sizes, bead appearance, and surface depositing a drop of water (5 μl) over the surface of the mats and
roughness. recording the drop shape by digital camera. The measurements were
Initially, different concentrations of CS (2.5%, 3%, 5% and 7% w/v) repeated at least five times for each sample.
and PVA (8%, 10% and 12% w/v) were electrospuned separately to find
the best concentration of each polymer. 3.8. Tensile study (DTS)
In the second step, the selected concentrations of each polymer were
combined together in different PVA/CS ratios (60:40, 50:50, 60:40, The tensile parameters of PVA NFs, PVA/Cs Composite NFs and CL-
70:30 and 80:20 v/v) and electrospuned to reach the optimized ratio and loaded PVA/CS Composite NFs were investigated by tensile testing
related apparatus variables for pristine Composite NFs. machine (STM- 5, SANTAM, Iran) with 500 kgl capacity at ambient
Finally, the 20% w/w of CL was added to the prepared optimized temperature based on ASTM international standard test method for
Composite solution and stirred for 8 h to ensure complete solvation of Tensile Properties of Polymer Matrix Composite Materials (ASTM, D
drug. Then, each apparatus factor was respectively changed to achieve 3039/D 3039 M). Briefly, the speed of moveable head was kept constant
optimized CL-loaded NFs. at 5 mm/min with respect to stationary head, the NF mat pieces having
The solvents for CS and PVA were 70% acetic acid and ultrapure rectangular shapes (0.1 mm × 20 mm × 50 mm) were mounted in the
water, respectively. Each separate polymeric solution and the Composite grids and after fixation, and the length of the specimen in the apparatus
mixtures had an overnight stirring by magnetic stirrer. was considered as initial length. The digital micrometer (DSWQ0,
China) was used to measure the thickness of the specimens. The stress-
3.2. Optimization assay via atomic force microscopy strain response curves were studied for determination of tensile
strength as the maximum stress that was applied over the mats, elon­
Atomic force microscopy (AFM) images were obtained using a JPK gation at break as the optimum elongation at break point, and Young’s
Instrument’s NanoWizard II (Berlin, Germany) mounted on an modulus as the linear part slope of the curve. Five specimens were
OLYMPUS IX51 inverted microscope (USA). Images were performed in evaluated for each test.
contact mode (air) using ACTA silicon cantilevers (USA) with a tip
radius of 6 nm, a resonant frequency between 200 and 400 kHz, and 3.9. Skin permeation studies
spring constant of 75 N m− 1. The scan rate was set to 1 Hz and the set
point was close to 1 V. The obtained images were analyzed with the JPK The animal studies were approved by the Ethics Committee for An­
image processing software (JPK Instruments), not only evaluating the imal Experiments at Mazandaran University of medical sciences in
morphology of the NFs but also calculating the topographic character­ compliance with Ethical Guidelines for Investigations in Laboratory
istics by measuring mean and distribution of diameters’ sizes and all Animals. The ex vivo skin permeation experiments were conducted to
surface roughness values. evaluate the skin permeation parameters of CL-NFs compared with CL-
solution. The Franz glass diffusion cells were Composed of receptor
3.3. Scanning electron microscopy compartment filled by 30 ml phosphate buffered (PBS, 7.4) that was
continuously stirred at 150 rpm during the experiment, and donor part
Total and surface morphology of pristine and CL-loaded NF mats contained a piece of CL-NFs mat with about 2*2 cm2 area (25 mg) and 5
were studied by field emission scanning microscopy (FE-SEM, MIRA3 ml of CL-solution (1 mg/ml) in each related cell over the stratum cor­
TESCAN, 3.0 Kv, Czech Republic). The Mean diameter size of NFs was neum surface of the rat’s skins with 3.8 cm2 effective diffusion area and
measured by Image J software (1.51 J8, NIH, USA). sealed by sealing film. In priory, the excised clean shaved male Wistar
rat’s abdomen skin had an overnight immersion in a normal saline so­
3.4. FTIR spectroscopy lution and then mounted between the donor and receptor compart­
ments. The temperature was maintained at 32 ± 0.5 ◦ C utilizing a
The FTIR spectra of pure PVA, CS, CL, pristine PVA/CS NF mats and constant thermostatic water circulation through the cell’s surrounded
CL-loaded PVA/CS Composite NF mats were evaluated by WQF 510A jacket. Before initiation of the experiments, 30 min’ equilibration time
spectrophotometer, China with a spectral range of 400–4400 cm− 1 and was considered. 2 ml aliquots were withdrawn from the receptor sam­
resolution of 2 cm− 1. pling nozzle at 0.5, 1, 2, 4, 6, 12 and 24-h time intervals, filtered and
analyzed. An equal volume of fresh medium at the same temperature
3.5. XRD analysis was replaced into the receptor compartment. At the end of the experi­
ment, the part of the skin that was in exposure to the donor phase was
X-ray diffractometer (X’Pert-Pro MPD, PAN analytical, Netherlands) removed and the surface was washed three times with deionized water
equipped with a Cu X-ray Anode was used to investigate the crystallinity to remove excess drug. Then the skin was cut into small pieces and
of the NF Composites and CL loaded NFs. Continuous scanning was homogenized in 10 ml of PBS, 7.4. After being kept for 24 h in room
carried out over a 2θ-angle range of 2◦ –70◦ at 40 kV and 40 mA with step temperature and then 10 min centrifugation at 3000 rpm, the super­
angle size of 0.02◦ and scanning rate of 43 s/step. natant was filtered by 0.22 μm syringe filter, and the CL content was
determined by HPLC method as mentioned above.
3.6. Thermal study Please note that the animal skin model is a simulated model of
human skin and may over predict human skin CL delivery.
The simultaneous thermal analyses of pure CS, PVA, CL, PVA NF,
PVA/CS Composite NF and CL-loaded PVA/CS Composite NF were 3.10. Transdermal release kinetic
carried out by BAHR thermoanalysis GmbH STA 503 (Hullhorst, Ger­
many) from 10 ◦ C to 700 ◦ C under argon atmosphere and heating rate of Various kinetic models like zero order, first order, Higuchi,

3
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

Korsmeyer-Peppas, Hixon-Crowell, etc. were evaluated to determine the optimized 10% w/v and 12% w/v PVA was 224 nm with 29 nm and 255
most similar kinetic model related to the CL transdermal release from nm with 57 nm respectively. The diameter size distribution of 10% PVA
CL-NFs. was a little better (Fig. 1. H& J).
In the second step, since both 10% and 12% PVA demonstrated
3.11. HPLC analysis relatively similar characteristics, both concentrations were utilized for
preparing various ratios in compounding with CS. Besides, as illustrated
The CL concentration was quantified according to the described above, CS owns no electrospinnability alone. To increase the chance of
assay method in colchicine monograph of USP pharmacopeia by HPLC electrospinning, the lowest concentration of CS (2.5%) was selected to
Knauer (Germany) using Knauer XDB-C18 column (5 μm, 4.6 × 250 be mixed with PVA. All compounding ratios in all distances and voltages
mm). The mobile phase was a mixture of methanol and phosphate buffer could form Taylor cone and acceptable jet, but the results were different.
(pH 5.5 ± 0.05) with the ratio of 55:45 (v/v), and the flow rate was 1 ml/ All PVA 10%:CS ratios had formed totally bead full fibers. Additionally,
min, and the drug monitoring was performed by UV detector at 254 nm. in higher voltages and distances, spindles appeared, too (Fig. 2. A, B&C).
The CL retention time was 7.7 min, which was in accordance with the It could be concluded that 10% PVA could not bring optimum
predicted range in the monograph (5.5–9.5 min) [26]. improvement in electrospinnability of CS solution even at 80:20 ratios
[27]. The PVA 12%:CS with 40:60 and 50:50 ratios in all apparatus
variable parameters showed beads and short pieces of fibers in back­
3.12. Encapsulation yield and drug content calculation
ground of constructed fibers (Fig. 2. D). As could be seen in Fig. 2. D, the
appearing fibers in the background have very small diameters about 50
A specified piece of CL-NFs mat with about 2*2 cm2 area was
nm and also small length in micrometer scale that could be named as
weighed and dissolved in 50 ml of 1% acetic acid, and 10 μl of the so­
“micronanofibers” as a new term. The attained fibers from the ratio of
lution was injected into HPLC with the same condition explained above
60:40 in all voltages and distances also contained some beads but the
in HPLC analyses section (3.11). Encapsulation efficiency (EE) and drug
mean diameter size of the best apparatus variables at 20 cm and 20kv
content (DC) percentages were calculated by following equations.
was fine and scarcely had some beads (204 ± 23 nm) (Sa = 91.72 nm)
ACTUAL ​ DRUG ​ LOADING (Fig. 2E &F). The 70:30 ratio led to achieving beadles fibers with
EE⋅(%)⋅ = × ⋅100⋅%
PREDICTED ​ DRUG ​ LOADING continuous structure in all apparatus variables and 20 cm distance with
20kv voltage showing desirable mean diameter size (243 ± 71 nm)
DC ⋅ (%)⋅ =
ACTUAL ​ DRUG ​ LOADING
× ⋅100 ​ % (Fig. 2. G &H). In the end, the electrospun resultant of 80:20 ratio gained
MAT ​ WEIGHT bead less fibers in all distances and voltages, but the mean diameter size
even at 20 cm and 20 kv was enormous (438 ± 157 nm) and caused
3.13. Cell viability assay considerable surface roughness (Sa = 266 as a representative) in com­
parison with 70:30 ratio fibers (Sa = 44.08) (Fig. 2. I &J). Presence of
The A-375 cell line was selected due to its availability in our country. some beads in the AFM image of formulation with 60:40 ratio would
The cells were seeded overnight into 96-well plates (1 × 104 cells/well). demonstrate that the auto-calculated mean diameter for this formula­
Then the cells were treated with equal weighed pieces of pristine PVA/ tion was not real and specific for nanofibers and seems to be an average
CS composite NF-based carrier and CL-loaded PVA/CS composite NFs by of both beads and nanofibers diameters. Comparison of the reported Sa
direct exposure for 72 h. Subsequently, the cell culture medium of each value of the formulation with 60:40 ratio, which was 91.72 with the
well was drawn out and replaced with 100 μL MTT solution (0.5 mg/ml formulation of 70:30 ratio which was 44.08, would prove this hypoth­
in PBS), calculating to provide 0.05 mg per each well, and incubated for esis. Investigation of the similar reported studies determines that
4 h at 37 ◦ C in 5% CO2. Then, the culture media was removed carefully, depending on the polymer’s nature (molecular weight and degree of
and 100 μL of DMSO were added to dissolve formazan crystals. The acetylation of CS and degree of polymerization and hydrolysis of PVA)
optical density of wells was determined at 570 nm by microplate reader there would be an optimum ratio for CS to be mixed with PVA to reach
(Epoch, Biotek, U.S.A.). electrospinality that could be in the range of 11% up to 50%. Based on
our findings, the optimum ratio for the combination of utilized CS and
4. Results and discussion PVA was 70:30. It could be construed that at higher ratio of CS, more
than 30, the accumulated increase in cationic charge at the surface of the
4.1. Optimization of pristine and CL-loaded PVA/CS composite NFs Taylor cone and ejected jet which relates to CS nature would cause a
repulsive force between the polymer’s ionic groups and change the
In the first step, none of the concentrations of CS solution led to surface tension that inhibits proper formation of continuous fibers
production of fiber in all attempted voltages and distances (Fig. 1 A, B, C during electrospinning [30]. As another result, the diameter of the
&D). This phenomenon was confirmed by Koosha et al. [27]. The elec­ Composite NFs would be influenced by changing in PVA:CS ratios.
trospun results of 8% w/v PVA in all voltages and distances from 10 cm Increasing the PVA:CS ratio would increase the diameter size of NFs.
to 15 cm showed totally bead full NFs but in 20 cm at all applied volt­ This result was in agreement with previous works [27,28,30].
ages. Approximately, half of the constructed NFs contained a lot of In the final step, CL was added to the solution of optimized PVA:CS
spindles and few beads (Fig. 1 E & F). The study of Sargazi et al. also (70:30), and the achieved electrospun NFs in various distances and
confirmed that shorter distances could not provide sufficient time for voltages were evaluated. The thinnest fiber with the best diameter size
solvent evaporation. thus, the NFs did not form properly [28]. The distribution was attained at 20 cm and 20kv (Fig. 2. K& L). The mean
infeasibility of forming NFs in 20 cm distance with relatively higher diameter size was 247 ± 57 nm, and the surface roughness values were
frequency could be attributed to other related parameters. The study of Sa = 74.27, Sq = 98.56, Sy = 894.72, Sp = 646.07, Sv = − 248.6 and Sm
Elkasaby et al. determined the polymer concentration as one of the most = 297.27 nm and also for pristine Nfs were Sa = 44.08, Sq = 64.26, Sy =
significant parameters affecting NFs diameter. The 10% w/v and 12% 719.64, Sp = 598.22, Sv = − 121.43 and Sm = 297.21 nm. The loading of
w/v of the PVA were electrospuned in a variety of distances and volt­ drug into the fiber mats did not make any significant change in diameter
ages. The results showed that both concentrations in all voltages and and surface roughness (p > 0.05). This would confirm the successful
distances would form continuous NFs with great frequency, no beads or incorporation of drug within the NFs [31].
spindles. The optimized parameters for both concentrations leading to The SEM micrographs were also corroborated by AFM and illustrated
the thinnest PVA NFs were 20 cm distance and 20 kv voltage (Fig. 1 G beadles and continuous non-woven NFs with smooth surface and no
&I) [29.]. The mean diameter size with surface roughness (Sa) for phase separation. The determined mean diameter size from SEM was

4
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

Fig. 1. AFM micrographs of CS 2.5% (A), 3% (B), 5% (C) and 7% (D), PVA 8% at 10 cm, 20kv (E) and 20 cm, 18kv (F), PVA 10% at 20 cm, 20kv (G) with its size
distribution (H) and PVA 12% at 20 cm, 20kv (I) with its size distribution (J).

5
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

Fig. 2. AFM micrographs of PVA 10%:CS 60:40 (A), 70:30 (B) and 80:20 (C) and PVA 12%:CS 50:50 (D), 60:40 (E) with its size distribution (F), 70:30 (G) with its
size distribution (H), 80:20 (I) with its size distribution (J), CL-loaded PVA 12%:CS (K) and its size distribution (L) all at 20 cm and 20kv.

195 ± 0.05 nm for pristine Composite NFs and 227 ± 0.03 nm for CL- The spectra of CL exhibited two differentiated peaks at 3315 and
loaded Composite NFs. The thinner diameter size reported from SEM 3429 cm− 1 which related to O–H and N–H stretching vibrations. In the
in comparison with AFM was related to complete evaporation of residual CL-loaded Composite NFs, these two peaks were combined and orga­
solvent conducted before SEM analysis by an overnight keeping in nized an individual band with more intensity in comparison with the
vacuum oven at ambient temperature (Fig. 3 A, B, C & D). same peak in pristine Composite NFs. This phenomenon determines the
hydrogen bond formation between CL, which possesses poly oxygen
4.2. FTIR molecular analysis structure with an amide group, and the Composite’s O–H and N–H
groups. Also, the C–H stretching peak of CL at 2935 cm− 1 appeared in
According to Fig. 4 A, the FTIR spectrum of pure CS showed a broad the CL-loaded nanofibers spectra. Moreover, other distinctive peak of
band at about 3438 cm− 1 and significant peaks at 2922, 1653, and 1095 CL, including 1733 (C– – O, stretching), 1653 (C– – O of amide, stretch­
cm− 1 refers to overlapped N–H and O–H stretching, C–H bending and ing), 1559 (N–H, bending), 1253 (C–O of ether, stretching), were
stretching of alkyl groups, bending of amide type I, and C–O stretching manifested in the CL-loaded nanofibers spectra with decreased intensity,
vibration frequencies, respectively [32–34]. which confirms the participation of the drug’s N and O atoms in
Main characteristic peaks for pure PVA were observed at 3450 cm− 1 hydrogen bonding.
(O–H, stretching), 2921 (C–H of alkyls, stretching), 1652 (C– – O, No significant modification of drug’s characteristic peaks in the drug
stretching) and 1101 (C–O, stretching) [34–37]. loaded nanofibers spectrum might be due to homogeneous loading of
The PVA/CS Composite nanofibers illustrated a broad band around drug along the Composite [38].
3433 cm− 1 with higher intensity and a slight shift to higher wavelength
compared with the similar band in PVA spectrum, referring to increased 4.3. XRD pattern
formation of hydrogen bonding by adding CS [33]. Furthermore, the
exhibited peak at 1652 cm− 1 compared with the same peak in pure CS Based on Fig. 4 B, XRD diffractogram of PVA showed a high intensity
spectrum showed a very slight shift (from 1653 to 1652 cm− 1) with peak at 2θ = 19.6◦ with a shoulder at 22.7◦ and also two low intensity
decreased intensity, confirming the increase in consumption of CS’s peaks at 11◦ and 40.79◦ . These characteristic peaks are related to the
amide to interact with hydroxyl groups of PVA by hydrogen bonding presence of crystalline lattice planes in the structure of PVA polymer.
[34–36]. Other characteristic peaks related to PVA, and CS also existed The obvious crystalline peak of chitosan at 20.2◦ was also observed.
in the Composite spectra with very slight change. Evaluation of the spectrum of the pristine PVA/CS Composite

6
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

Fig. 3. SEM micrographs of PVA:CS Composite NFs (A & B) and CL-loaded PVA:CS Composite NFs (C & D).

nanofiber in comparison with the raw polymers demonstrated that only with human serum albumin, which has many available donor groups for
the main peak of pure PVA at 19.6◦ remained. Besides, this peak became hydrogen bonding, showed that CL has the potential to conduct such
weaker and had a very short shift to a lower degree (19.4◦ ). The absence spontaneous interactions [37,39].
of the polymer’s crystalline peaks in the Composite and intensity Crucially, the sharp diffraction pattern of pure CL vanished in the
reduction of the related main peak of PVA revealed that PVA/CS Com­ spectra of CL loaded NFs, which obviously demonstrates the dispersion
posite nanofibers have lower crystallinity compared to raw polymers. of drug in amorphous manner along the Composite nanofibers. The CL
The attained reduction in crystallinity could be related to hydrogen amorphization was proved by STA analysis [40].
bonding interaction occurring between the polymers molecules in the
Composite. This result was confirmed by above FT-IR analysis. More­ 4.4. STA analysis
over, the absence of chitosan diffraction peak in the PVA/CS Composite,
considering the 70:30 compounding ratios and crystallinity reduction in As Shown in Fig. 5, the DTA thermogram of pure CS exhibited two
nanofibrous form, illustrates fine compatibility of chitosan and PVA [32, characteristic peaks at 89 ◦ C (endothermic) and 309 ◦ C (exothermic)
34]. attributed to the adsorbed moisture evaporation and polymer degrada­
As shown in Fig. 4 B, loading of CL into the PVA/CS Composite tion, respectively. The TGA mass loss stages of CS were observed in the
nanofibers led to enhancement of peak’s intensity. Since the CL mole­ area of 30 ◦ C–107 ◦ C and 240◦ C–370 ◦ C, confirming the thermal be­
cules are surrounded by oxygen atoms, which could be the receptor haviors recorded by DTA related to water evaporation occurring in the
atom of hydrogen bonding from the Composite donor groups like hy­ first stage and CS degradation, including dehydration of sugar rings,
droxyl and amides, it seems that it has the potential to be entrapped by decomposition and depolymerization of the polymer in the second stage
developing all around hydrogen bonding interactions and also van der [41].
waals interactions. Thus, it may stabilize the Composite molecular The DTA curve of pure PVA showed an endothermic peak at 81 ◦ C
structure and improve the crystallinity of the Composite. As could be that refers to glass transition temperature (Tg) followed by other two
seen above, the FT-IR spectra evaluation also proved formation of these endothermic peaks at 222 ◦ C and 297 ◦ C related to melting and
hydrogen bindings. A spectroscopic analysis study on CL interaction decomposition temperatures. The remaining four peaks represent the

7
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

Fig. 4. FT-IR spectra (A) and XRD pattern (B) of pure powder of CS, PVA, CL and the attained mat of PVA/CS Composite NFs and CL-loaded PVA/CS Composite NFs.
Representative stress-strain curves (C) of PVA NFs, PVA/CS Composite NFs and CL-loaded PVA/CS Composite NFs. Ex vivo percutaneous permeation profile (D) of CL
per unit area of the rat skin from CL-loaded nanofibers and CL solution.

degradation process of PVA planes structure [34,36]. The TGA curve determining the entrapment of drug in the nanofibers as an amorphous
also illustrated weight loss phases at these three ranges. manner. This result was also confirmed by XRD and FT-IR studies con­
Comparison of the DTA curves of pure PVA and PVA NF alone ducted in this paper. The DTA curve of CL-loaded PVA/CS NFs, in
indicated that the first three peaks only remained and the four other comparison with pristine Composite NFs, depicts the three main peaks of
peaks vanished, which refers to crystallinity decrease during process of PVA with a slight shift. The movement of degradation peak at 320 ◦ C
electrospinning. However, the three phases of weight loss are still was a little more toward higher temperature that determines increase in
obvious in the TGA thermogram, confirming all thermal behaviors thermal stability. Two new peaks also appeared with a shift toward
mentioned above [35]. higher temperatures that interpreted higher Composite crystallinity and
The pristine PVA/CS Composite NFs compared with PVA NFs and increased thermal stability via addition of CL. The mass loss temperature
pure CS demonstrated the three main peaks of PVA in the DTA ther­ also increased, which confirms the enhancement of thermal stability.
mogram. However, the melting peak became broad and shifted to lower The FT-IR and XRD proved that the new hydrogen bond interactions
temperature due to reduction of PVA crystallinity with addition of CS. between CL and the Composite structure are responsible for this
Besides, the molecular hydrogen bonding between two polymers and phenomenon.
also plasticization of presenting water traces in nanofibers led to exhi­ Overall, the three phase weight loss profile is similar in both pristine
bition of a broad glass transition temperature. Due to lower proportion Composite NFs and CL-loaded NFs. Furthermore, the represented main
of CS compared with PVA in the Composite and also the crystallinity peaks in DTA curves are similar, too, demonstrating homogeneous
reduction, the two peaks of CS were not obvious in the Composite. The dispersion of amorphous CL along the Composite nanofibers [35,42].
three stages of mass loss in TGA curve also confirm the presence of PVA,
and since the first two phases are common in both PVA and CS, the
observed decrease in thermal stability of Composites indicates crystal­ 4.5. Water angle analysis
linity reduction of PVA via added CS [34,36].
The sharp endothermic peak of pure CL at 160 ◦ C, which relates to its The mean water angle of PVA mats, alone, pristine PVA:CS Com­
melting point, disappeared in CL-loaded PVA/CS NFs curve, posite mats and CL-loaded Composite mats were 43.69 ± 4.8◦ , 38.8 ±
5.5◦ and 31.35 ± 3.4 ◦ respectively (Fig. 6 A, B, C as representative).

8
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

Fig. 5. TGA, blue curves and DTA, black curves of pure powders of CS (A), PVA (B), CL (C) and attained mat of PVA NFs (D), PVA/CS Composite NFs (E) and CL-
loaded PVA/CS Composite NFs (F). (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this article.)

Fig. 6. Representative water angles images of PVA NFs (A), PVA/CS Composite NFs (B) and CL-loaded PVA/CS Composite NFs (C).

Obviously, addition of CS to PVA decreased the contact angle. As re­ 4.6. Tensile evaluation
ported above, the surface roughnesses of PVA (Sa = 57) and PVA:CS (Sa
= 44) are almost similar, so the enhancement of wettability after addi­ The mechanical features of the mats could be related to various
tion of CS does not relate to the surface roughness and could be attrib­ factors like composition, atomic interaction energies, morphology,
uted to increase in hydrophilicity of the Composite NFs in comparison consistency and average diameter of fibers [43,44]. Fig. 4C illustrates
with the PVA NFs. In fact, the amino and hydroxyl groups of CS and also the representative stress-strain response curves. The tensile strength of
newly formed hydrogen bonding with PVA are responsible for this PVA NFs, PVA/CS NFs and CL-loaded PVA/CS NFs were 8.53 ± 1.45,
development. The incorporation of CL into the Composite NFs also 15.86 ± 3.71 and 16.91 ± 1.52 respectively. Also, their calculated
decreased the contact angle. Thus, loading of CL increased the wetta­ Young’s modulus were 0.7 ± 0.39, 4.27 ± 0.94 and 3.84 ± 0.45,
bility of the mat. Based on AFM findings, no significant change in surface respectively. The difference of tensile strength and Young’s modulus
roughness was exhibited after drug loading (44 nm–66 nm). This affirms between PVA NFs group and PVA/CS NFs group were significant (p <
that CL was not present over the surface of the NFs and the reported 0.05). This shows that the addition of CS to PVA and making a fibrous
slight change in roughness refers to the diameter size enhancement nano Composite could lead to produce stronger interactions, which
developed by drug loading into the NFs. Thereby, increase in wettability based on FT-IR, XRD and STA analyses relates to formation of new
does not relate to surface roughness or presence of drug on the surface of hydrogen bonding between two polymers which provided fine unifor­
NFs. However, it relates to increase in NFs hydrophilicity due to ho­ mity composition. The study conducted by Liu et al. also stated that
mogenous dispersion of hydrophilic drug with surrounded poly oxygen consistency without phase separation or agglomeration in the Com­
groups and hydrogen bonding with Composite all along NFs with high posite nanofibers could impact on mechanical properties of the mats
surface area that provides easy penetration of water into the mat. These [45]. On the other hand, depending on previous studies, CS could
modifications in angle values are in accordance with FT-IR, XRD and improve stiffness in the mats [46,47]. Moreover, the difference between
DSC results and also previous similar studies [27,34]. PVA/CS NF group and drug-loaded PVA/CS NF group was not signifi­
cant (p > 0.05), which demonstrates that the twenty-percent drug
loading did not significantly change the mechanical properties of

9
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

PVA/CS Composite. As mentioned in FT-IR, XRD and STA studies, the CL amorphous manner to each unit of skin area [49].
could form fine hydrogen bonding within the Composite, but it seems As summarized in Table 1, significant differences in amount of CL
that more than twenty percent drug loading may be needed to enhance skin deposition from NF formulation and CL solution were observed (p
the mechanical stability of the nanofibrous Composite drug delivery < 0.001). Total drug accumulated in the skin was found to be about
system. However, this non-significant alteration in mechanical proper­ three-fold higher after applying CL-NFs compared to the CL solution.
ties after twenty-percent drug incorporation is desirable. The better accumulation of CL in skin from NF formulation could be
The elongation at break of PVA NFs was 97.93 ± 46.64, which was attributed to the higher amount of permeated CL and flux value. Pre­
significantly higher than PVA/CS NFs (29.92 ± 9.44) and CL-loaded vious study has proven that CL skin deposition is in direct relationship
PVA/CS NFs (28.37 ± 7.92) (p < 0.05). This criterion was not signifi­ with the amount and rate of CL permeation [50]. This greater affinity to
cantly different between PVA/CS NFs groups and CL loaded one (p > skin may also refers to the partition coefficient value of CL that is rela­
0.05). This result also confirms that the PVA NFs possess less stiffness tively more desirable than some other drugs [14,51]. Skin accumulation
than the fibrous nano Composites. This elasticity of PVA NFs refers to the is beneficial for localized target delivery of CL in improved management
tendency of randomly aligned nanofibers to be aligned easier through of all approved therapeutic properties of CL in dermatological diseases
the axis of tensile pulling relative to PVA/CS Composite NFs. This like skin cancers, psoriasis, and so on or even could be suggested for
behavior that occurs in Composite NFs with more resistance refers to adjuvant or neoadjuvant local chemotherapy or localized post-surgery
stronger atomic interaction which needs more stress for slipping of tumor recurrence prevention by reducing the adverse effects caused
nanofibers along each other [48]. Therefore, the PVA/CS Composite NFs by the impact on unaffected areas or systemic absorption.
and CL-loaded PVA/CS Composite NFs demonstrated more stable and
firmer mats compared with pristine PVA NFs. 4.9. Transdermal release kinetic

4.7. Assuring encapsulation yield and drug content by HPLC The permeated amounts of CL per unit area of the rat skin from CL-
NFs were analyzed by applying the related equation of each kinetic
Despite the fact that electrospun nanofibers usually provide a com­ model. Depending on reported parameters in Table 2, the transdermal
plete encapsulation of drug, considering the narrow therapeutic index of release kinetic from CL-NFs was in accordance with first order model
CL, in order to ensure full loading of CL into nanofibers mat and precise with R2 value of 0.9858 ± 0.0021. In the previous study, the release
dose adjustment for further administrations of the formulations, the EE kinetic model of another drug (gentamicin) from a chitosan based
and DC percentages were calculated based on the HPLC results. The EE nanofibrous Composite membrane was also best fitted by first order
was 98.27 ± 1.85% and DC was 19.65 ± 0.37% after repeating the model [52].
procedure several times.
4.10. Anti-melanoma activity
4.8. Ex vivo skin permeation and deposition studies
According to Fig. 7, the CL-loaded bio composite NFs exhibited a
significant anti-tumoral effect against A-375 human melanoma cell line
The cumulative permeated profiles of CL per unit area of the rat skin
at the concentrations of 500 and 1000 μg/ml of the whole mat compared
from CL-loaded nanofibers and CL solution were illustrated in Fig. 4 D.
with the control. Considering the 19.65 ± 0.37% DC concentration, that
Total permeated drug during 24h from NF formulation was 45 ± 2.29
was calculated in section 4.7, clearly shows better efficacy of the CL-
μg/cm2 and 15.29 ± 1.21 μg/cm2 from drug solution. This difference
loaded bio composite NFs compared with pure CL. Additionally, the
was significant (p < 0.001). The drug flux from NF formulation was also
pristine NFs and CL-loaded NFs in the lower concentrations shows a
significantly (p < 0.001) higher than drug solution (more than three-
relative cell proliferation compared with the control.
fold) (Fig D and Table 1).
The significant reported cytotoxicity of CL-loaded composite NFs
This increase in drug permeation and flux could refer to the high
could be ascribed to many reasons, including enhanced stability of CL
specific surface area to mass with very high aspect ratio, flexibility,
molecules that was provided in the nanofibers and large surface area
porosity structure and release sustainability of PVA/Chitosan nanofiber
that was dedicated by the nanofiberous mat which would completely
scaffold that also improves dissolution rate and drug release profile by
attach to the cells. Moreover, the sustained release of CL from the
providing a smaller amount of drug permeation through each unit of
nanometer-scale fibers, with extensive cells contact level, ensures dis­
skin surface per each unit of time and also possess occlusive property
tribution and internalization into the melanoma cells that were
with desirable breathability enhancing the drug permeation [17]. Be­
entrapped throughout the scaffold [21]. Additionally, the amorphous
sides, the amorphous status of CL in NFs would enhance the drug flux
manner of CL in the carrier and the desirable log-p of CL could be also
through the stratum corneum. In contrast, crystal status of CL in the
responsible for the reported high performance cellular entrance and
control solution would prevent drug penetrating into the rat skin [40].
wiping off function subsequently [14,40].
Moreover, water angle measurements’ results proved the high tendency
The observed proliferation of the pristine nanofibrous carrier is
of CL loaded-NFs to easily absorb even small quantity of skin moisture
related to the presence of CS in the structure of nanofibers. The remained
after being applied over the skin surface and lead to desirable penetra­
CS based scaffold, after completion of CL sustain release, could provide a
tion of water molecules into the mat, thus providing gradual swelling
desirable recovery for normal cells at the area after suffering from the
and leaching and consequently sustaining diffusion of drug in

Table 2
Table 1
Coefficients of determination (R2) and other parameters values of kinetics
Skin permeation and deposition parameters of CL from CL-loaded composite NFs
models for transdermal release.
and CL-solution.
Kors- Zero Higuchi First order Hixson
Q24h (μg/cm2) Flux (μg/cm2/h) Skin deposited24h (μg/cm2)
peppas order
CL-NF 45.00 ± 2.29 5.317 ± 0.159 51.72 ± 4.09
R2 0.907 ± 0.9756 ± 0.9355 ± 0.9858 ± 0.9828 ±
CL-solution 15.29 ± 1.21 1.638 ± 0.099 18.46 ± 1.00
0.0066 0.0030 0.0074 0.0021 0.0025
Values determined as mean ± SD. Slope 21.0743 ± 2.5289 ± 9.0526 ± − 0.0129 ± 0.0436 ±
Q24, it is the cumulative amount of permeated CL per skin area into the receptor. 1.1124 0.1463 0.4747 0.0008 0.0027
Intercept 2.2911 ± 0.4827 ± − 4.5753 ± 1.9999 ± 0.0032 ±
Flux, calculated from the curve slop of cumulative amount of permeated CL per
0.1146 0.2568 0.4716 0.0014 0.0047
skin area versus time.

10
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

Fig. 7. Cell viability of A-375 human melanoma cell line after 72h treated with various concentration of A) pristine chitosan based bio composite NFs,B) CL-loaded
chitosan based bio composite NFs and C) pure CL.

CL. The role of CS in post localized chemotherapy recovery was also Supervision, Resources, Project administration, Funding acquisition.
proved in the recent study [24]. Pooria Gill: Resources, Supervision. Reza Enayatifard: Supervision,
Considering the significant skin depot of CL from the nanofibrous Resources, Project administration, Funding acquisition.
carrier that was exhibited in section 4.8, and the observed residual wipe
out of melanoma cells, it could be suggested for safe localized target Declaration of competing interest
chemotherapy as an adjuvant or neoadjuvant intervention or even for
post-surgery tumor recurrence prevention with the lowest systemic The authors declare that they have no known competing financial
cytotoxicity. interests or personal relationships that could have appeared to influence
the work reported in this paper.
5. Conclusion
Appendix A. Supplementary data
The chitosan based bio Composite nanofiberous drug delivery system
of CL was formulated, optimized, fully characterized and exhibited such Supplementary data to this article can be found online at https://doi.
a desirable efficiency as topical and transdermal dosage forms. The org/10.1016/j.matchemphys.2021.124381.
significant skin deposition and anti-melanoma performance, could
suggest it as a neoadjuvant chemotherapy before other interventions References
and adjuvant therapy after tumor excision surgery for local delivery and
[1] T.K. Eigentler, U.M. Caroli, P. Radny, C. Garbe, Palliative therapy of disseminated
also as tumor recurrence inhibitor. Moreover, it may be suggested for
malignant melanoma: a systematic review of 41 randomised clinical trials, Lancet
management of other types of external or internal tumors. It also could Oncol. 4 (2003) 748–759, https://doi.org/10.1016/s1470-2045(03)01280-4.
be recommended for administration in other related skin diseases with [2] R.D. Rao, S.G. Holtan, J.N. Ingle, et al., Combination of paclitaxel and carboplatin
dose adjustment. as second-line therapy for patients with metastatic melanoma, Cancer 106 (2006)
375–382, https://doi.org/10.1002/cncr.21611.
Additionally, based on the reported remarkable first order release [3] Z. Eroglu, A. Ribas, Combination therapy with BRAF and MEK inhibitors for
kinetic behavior through skin it could be suggested as an efficient melanoma: latest evidence and place in therapy, Ther Adv Med Oncol 8 (2016)
external nanofibrous CL delivery patch for systemic approved in­ 48–56, https://doi.org/10.1177/1758834015616934.
[4] G.D. Leonard, T. Fojo, S.E. Bates, The role of ABC transporters in clinical practice,
dications by omission of first pass effect and providing more conve­ Oncol. 8 (2003) 411–424.
nience and compliance for patients. [5] W. Shuai, X. Li, W. Li, et al., Design, synthesis and anticancer properties of
isocombretapyridines as potent colchicine binding site inhibitors, Eur. J. Med.
Chem. 197 (2020), 112308, https://doi.org/10.1016/j.ejmech.2020.112308.
Statement of human and animal rights [6] B. Dasgeb, D. Kornreich, K. McGuinn, et al., Colchicine: an ancient drug with novel
applications, Br. J. Dermatol. 178 (2018) 350–356, https://doi.org/10.1111/
The animal studies were approved by the Ethics Committee for An­ bjd.15896.
[7] Y.Y. Leung, L.L. Yao Hui, V.B. Kraus, Colchicine–Update on mechanisms of action
imal Experiments at Mazandaran University of medical sciences in and therapeutic uses, Semin. Arthritis Rheum. 45 (2015) 341–350, https://doi.org/
compliance with Ethical Guidelines for Investigations in Laboratory 10.1016/j.semarthrit.2015.06.013.
Animals. [8] K.K. Dubey, P. Kumar, N.E. Labrou, P. Shukla, Biotherapeutic potential and
mechanisms of action of colchicine, Crit. Rev. Biotechnol. 37 (2017) 1038–1047,
https://doi.org/10.1080/07388551.2017.1303804.
Statement of biosafety considerations [9] M.R. Prausnitz, R. Langer, Transdermal drug delivery, Nat. Biotechnol. 26 (2008)
1261–1268, https://doi.org/10.1038/nbt.1504.
[10] B.D. Bozorg, A.K. Banga, Effect of different pressure-sensitive adhesives on
All cell culture experiments were performed in accordance with the performance parameters of matrix-type transdermal delivery systems,
approved biosafety level guidelines of Mazandaran University of Medi­ Pharmaceutics 12 (2020), https://doi.org/10.3390/pharmaceutics12030209.
cal Sciences. [11] X. Wang, B. Ding, B. Li, Biomimetic electrospun nanofibrous structures for tissue
engineering, Mater. Today 16 (2013) 229–241, https://doi.org/10.1016/j.
mattod.2013.06.005.
Data availability statement [12] S. Chen, R. Li, X. Li, J. Xie, Electrospinning: an enabling nanotechnology platform
for drug delivery and regenerative medicine, Adv. Drug Deliv. Rev. 132 (2018)
188–213, https://doi.org/10.1016/j.addr.2018.05.001.
Any raw/processed data required to reproduce these findings could [13] S. Thakkar, M. Misra, Electrospun polymeric nanofibers: new horizons in drug
be shared on demands. delivery, Eur. J. Pharmaceut. Sci. 107 (2017) 148–167, https://doi.org/10.1016/j.
ejps.2017.07.001.
[14] J.A. Sahu Rishabh Kumar, Ns, Development and evaluation of transdermal patches
CRediT authorship contribution statement of Colchicine, Der Pharm. Lett. 4 (2012) 330–343.
[15] R. Goyal, L.K. Macri, H.M. Kaplan, J. Kohn, Nanoparticles and nanofibers for
topical drug delivery, J. Contr. Release 240 (2016) 77–92, https://doi.org/
Hamed Morad: Conceptualization, Methodology, Software, Valida­
10.1016/j.jconrel.2015.10.049.
tion, Formal analysis, Investigation, Resources, Data curation, Writing - [16] H. Asgarirad, P. Ebrahimnejad, M.A. Mahjoub, et al., A promising technology for
original draft, Writing - review & editing, Visualization, Supervision, wound healing; In-vitro and in-vivo evaluation of chitosan nano-biocomposite
Project administration. Mohsen Jahanshahi: Supervision, Resources, films containing gentamicin, J. Microencapsul. (2020) 1–22, https://doi.org/
10.1080/02652048.2020.1851789.
Project administration, Funding acquisition. Jafar Akbari: Supervision, [17] Y. Shi, Y. Li, J. Wu, et al., A novel transdermal drug delivery system based on self-
Resources, Project administration, Funding acquisition. Majid Saeedi: adhesive Janus nanofibrous film with high breathability and monodirectional

11
H. Morad et al. Materials Chemistry and Physics 263 (2021) 124381

water-penetration, J. Biomater. Sci. Polym. Ed. 25 (2014) 713–728, https://doi. [36] Mei Yang, Runjun Sun, Yan Feng, H.D. Honghong Wang, Cl, Preparation,
org/10.1080/09205063.2014.897596. characterization and kinetics study of chitosan/PVA electrospun nanofiber
[18] K. Kataria, A. Gupta, G. Rath, et al., In vivo wound healing performance of drug membranes for the adsorption of dye from water, J. Polym. Eng. 39 (2019) 459.
loaded electrospun composite nanofibers transdermal patch, Int. J. Pharm. 469 [37] R. Ahmed, M. Tariq, I. Ali, et al., Novel electrospun chitosan/polyvinyl alcohol/
(2014) 102–110, https://doi.org/10.1016/j.ijpharm.2014.04.047. zinc oxide nanofibrous mats with antibacterial and antioxidant properties for
[19] M.A. Tahir, M.E. Ali, A. Lamprecht, Nanoparticle formulations as recrystallization diabetic wound healing, Int. J. Biol. Macromol. 120 (2018) 385–393, https://doi.
inhibitors in transdermal patches, Int. J. Pharm. 575 (2020), 118886, https://doi. org/10.1016/j.ijbiomac.2018.08.057.
org/10.1016/j.ijpharm.2019.118886. [38] S.A. Siddiqui, A. Dwivedi, A. Pandey, et al., Molecular structure, vibrational
[20] H. Morad, M. Jahanshahi, J. Akbari, et al., Formulation, optimization, and spectra and potential energy distribution of colchicine using ab initio and density
evaluation of nanofiber-based fast dissolving drug delivery system of colchicine for functional theory, J Comput Chem Japan 8 (2009) 59–72, https://doi.org/
pediatrics, Int. J. Pediatr. (2020), https://doi.org/10.22038/ijp.2020.52106.4140. 10.2477/jccj.H2028.
[21] M. Khodadadi, S. Alijani, M. Montazeri, et al., Recent advances in electrospun [39] Y.-J. Hu, Y. Liu, R.-M. Zhao, S.-S. Qu, Interaction of colchicine with human serum
nanofiber-mediated drug delivery strategies for localized cancer chemotherapy, albumin investigated by spectroscopic methods, Int. J. Biol. Macromol. 37 (2005)
J. Biomed. Mater. Res. 108 (2020) 1444–1458, https://doi.org/10.1002/jbm. 122–126, https://doi.org/10.1016/j.ijbiomac.2005.09.007.
a.36912. [40] N.-W. Kang, M.-H. Kim, S.-Y. Sohn, et al., Curcumin-loaded lipid-hybridized
[22] Y. Huang, J. Song, C. Yang, et al., Scalable manufacturing and applications of cellulose nanofiber film ameliorates imiquimod-induced psoriasis-like dermatitis in
nanofibers, Mater. Today 28 (2019) 98–113, https://doi.org/10.1016/j. mice, Biomaterials 182 (2018) 245–258, https://doi.org/10.1016/j.
mattod.2019.04.018. biomaterials.2018.08.030.
[23] F. Nasiri, S. Ajeli, D. Semnani, et al., Fuzzy VIKOR optimization for designing high [41] J. Jalvandi, M. White, Y. Gao, et al., Polyvinyl alcohol composite nanofibres
performance hydroxyapatite/polycaprolactone scaffolds for hard tissue containing conjugated levofloxacin-chitosan for controlled drug release, Mater. Sci.
engineering, J Text Polym 8 (2020) 17–36. Eng. C 73 (2017) 440–446, https://doi.org/10.1016/j.msec.2016.12.112.
[24] L.-F. Zhu, Y. Zheng, J. Fan, et al., A novel core-shell nanofiber drug delivery system [42] J.H. Park, H.W. Lee, D.K. Chae, et al., Electrospinning and characterization of poly
intended for the synergistic treatment of melanoma, Eur. J. Pharmaceut. Sci. 137 (vinyl alcohol)/chitosan oligosaccharide/clay nanocomposite nanofibers in
(2019), 105002, https://doi.org/10.1016/j.ejps.2019.105002. aqueous solutions, Colloid Polym. Sci. v. 287 (2009).
[25] A. Massarotti, A. Coluccia, R. Silvestri, et al., The tubulin colchicine domain: a [43] I.K. Kwon, S. Kidoaki, T. Matsuda, Electrospun nano- to microfiber fabrics made of
molecular modeling perspective, ChemMedChem 7 (2012) 33–42, https://doi.org/ biodegradable copolyesters: structural characteristics, mechanical properties and
10.1002/cmdc.201100361. cell adhesion potential, Biomaterials 26 (2005) 3929–3939, https://doi.org/
[26] M. Rockville, United States Pharmacopeia and National Formulary (USP 41-NF 36), 10.1016/j.biomaterials.2004.10.007.
41st-NF, 36th ed, 2018. [44] A. Baji, Y.-W. Mai, S.-C. Wong, et al., Electrospinning of polymer nanofibers: effects
[27] M. Koosha, H. Mirzadeh, Electrospinning, mechanical properties, and cell behavior on oriented morphology, structures and tensile properties, Compos. Sci. Technol.
study of chitosan/PVA nanofibers, J. Biomed. Mater. Res. 103 (2015) 3081–3093, (2010) 703–718, https://doi.org/10.1016/j.compscitech.2010.01.010.
https://doi.org/10.1002/jbm.a.35443. [45] H. Liu, W. Liu, B. Luo, et al., Electrospun composite nanofiber membrane of poly(L-
[28] G. Sargazi, D. Afzali, A. Mostafavi, S.Y. Ebrahimipour, Synthesis of CS/PVA lactide) and surface grafted chitin whiskers: fabrication, mechanical properties and
biodegradable composite nanofibers as a microporous material with well cytocompatibility, Carbohydr. Polym. 147 (2016), https://doi.org/10.1016/j.
controllable procedure through electrospinning, J. Polym. Environ. 26 (2018) carbpol.2016.03.096.
1804–1817, 101007/s10924-017-1080-8. [46] C. Mahoney, D. Conklin, J. Waterman, et al., Electrospun nanofibers of poly
[29] M. Elkasaby, H.A. Hegab, A. Mohany, G.M. Rizvi, Modeling and optimization of (epsilon-caprolactone)/depolymerized chitosan for respiratory tissue engineering
electrospinning of polyvinyl alcohol (PVA), Adv. Polym. Technol. 37 (2018) applications, J. Biomater. Sci. Polym. Ed. 27 (2016) 611–625, https://doi.org/
2114–2122, https://doi.org/10.1002/adv.21869. 10.1080/09205063.2016.1144454.
[30] M.Z. Elsabee, H.F. Naguib, R.E. Morsi, Chitosan based nanofibers, review, Mater. [47] D. Semnani, E. Naghashzargar, M. Hadjianfar, et al., Evaluation of PCL/chitosan
Sci. Eng. C 32 (2012) 1711–1726, https://doi.org/10.1016/J.MSEC.2012.05.009. electrospun nanofibers for liver tissue engineering, Int J Polym Mater Polym
[31] A.E. Torkamani, Z.A. Syahariza, M.H. Norziah, et al., Encapsulation of Biomater 66 (2017) 149–157, https://doi.org/10.1080/00914037.2016.1190931.
polyphenolic antioxidants obtained from Momordica charantia fruit within zein/ [48] J.D. Schiffman, C.L. Schauer, Cross-linking chitosan nanofibers,
gelatin shell core fibers via coaxial electrospinning, Food Biosci 21 (2018) 60–71, Biomacromolecules 8 (2007) 594–601, https://doi.org/10.1021/bm060804s.
https://doi.org/10.1016/j.fbio.2017.12.001. [49] T. Ngawhirunpat, P. Opanasopit, T. Rojanarata, et al., Development of meloxicam-
[32] M. Koosha, M. Raoufi, H. Moravvej, One-pot reactive electrospinning of chitosan/ loaded electrospun polyvinyl alcohol mats as a transdermal therapeutic agent,
PVA hydrogel nanofibers reinforced by halloysite nanotubes with enhanced Pharmaceut. Dev. Technol. 14 (2009) 70–79, https://doi.org/10.1080/
fibroblast cell attachment for skin tissue regeneration, Colloids Surf. B 10837450802409420.
Biointerfaces 179 (2019) 270–279, https://doi.org/10.1016/j. [50] H.P. Singh, P. Utreja, A.K. Tiwary, S. Jain, Elastic liposomal formulation for
colsurfb.2019.03.054. sustained delivery of colchicine: in vitro characterization and in vivo evaluation of
[33] J. Jacob, G. Peter, S. Thomas, et al., Chitosan and polyvinyl alcohol anti-gout activity, AAPS J. 11 (2009) 54–64, https://doi.org/10.1208/s12248-008-
nanocomposites with cellulose nanofibers from ginger rhizomes and its 9078-8.
antimicrobial activities, Int. J. Biol. Macromol. 129 (2019) 370–376, https://doi. [51] R. Shawahna, N. Rahman, Evaluation of the use of partition coefficients and
org/10.1016/j.ijbiomac.2019.02.052. molecular surface properties as predictors of drug absorption: a provisional
[34] S. Yang, P. Lei, Y. Shan, D. Zhang, Preparation and characterization of antibacterial biopharmaceutical classification of the list of national essential medicines of
electrospun chitosan/poly (vinyl alcohol)/graphene oxide composite nanofibrous Pakistan, Daru 19 (2011) 83–99.
membrane, Appl Surf SCINCE 435 (2018) 832–840. [52] F. Amini, D. Semnani, S. Karbasi, S.N. Banitaba, A novel bilayer drug-loaded
[35] A.C. Alavarse, F.W. de Oliveira Silva, J.T. Colque, et al., Tetracycline wound dressing of PVDF and PHB/Chitosan nanofibers applicable for post-surgical
hydrochloride-loaded electrospun nanofibers mats based on PVA and chitosan for ulcers, Int J Polym Mater Polym Biomater 68 (2019) 772–777, https://doi.org/
wound dressing, Mater Sci Eng C Mater Biol Appl 77 (2017) 271–281, https://doi. 10.1080/00914037.2018.1506982.
org/10.1016/j.msec.2017.03.199.

12

You might also like