You are on page 1of 20

European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Contents lists available at ScienceDirect

European Journal of Pharmaceutical Sciences


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

Review

Electrospun polymeric nanofibers: New horizons in drug delivery MARK



Shreya Thakkar, Manju Misra
Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat 380054, India

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

Keywords: Nanofibers obtained using electrospinning technique are being used since ages especially in fields of textile
Electrospinning industry, sensors, filters, protective clothing and tissue engineering. Their use as drug delivery system is an
Nanofibers emerging platform in the field of pharmaceuticals and now-a-days formulation scientists are paying great at-
Release profile tention to the technology due to several advantages prime being easy modulation of drug release profile de-
CPPs and CMAs
pending upon the properties of polymer/polymeric blends/other materials used. Although there are several
reports citing the use of antibiotics-loaded nanofibers as wound dressing materials and as antimicrobial therapy
in periodontics; still there is a good scope of expanding the horizon for its application in newer ailments. This
article reviews various aspects related to loading and release of drug as such or in nano-particulate form to
polymeric nanofibers by taking critical process parameters (CPPs) for electrospinning and critical material at-
tributes (CMAs) into account. Commercially available products and electrospinning technologies are described
in brief along with some of the patents related to their use as drug delivery systems. The main focus of this
review is applicability of drug/drug nanoparticle loaded nanofibers in the management of diseases/disorders
related to the brain, eye, ear, cardiovascular system, lungs and oral cavity. Use in diseases with higher mortality
rates like diabetes, Acquired immunodeficiency syndrome (AIDS) and cancer is also described in brief.

1. Introduction (Mahalingam and Edirisinghe, 2013) have been explored by researchers


for nanofiber spinning. These techniques provide new advances in the
Electrospinning is a process of conversion of polymeric solution/ field of nanofiber formation and are strong competitors of electro-
melt (with or without additives/incorporable) into solid nanofibers by spinning technique offering several advantages and limitations. A
application of electrical force. Amongst numerous conventional comprehensive comparison of all these techniques is summarized in
methods reported for nanofiber formation such as drawing, template Table 1.
synthesis, self-assembly, phase separation, and electrospinning; elec- Nanofibers are solid fibers of materials having diameters below micron
trospinning is more popular due to its advantages like easy control over range with a porous structure and very high surface area. These can be
fiber diameter, morphology, surface characteristics, porosity and ease easily fabricated by electrospinning process as this process provides con-
of getting fiber diameter in nano range (Kowalewski et al., 2005; Nayak trol over properties as well as the alignment of fibers on collector used
et al., 2011). (static or rotating cylinder). Specific alignment of nanofibers can result in
Apart from electrospinning numerous other non-electro nanofiber specific structures such as nanotubes and nanowires. Nanofiber of the only
spinning techniques such as solution blowing (Polat et al., 2016), conventional polymer will result in no specific function and can be used as
centrifugal spinning (Taghavi and Larson, 2014), template synthesis scaffolds only. To form functional nanofibers polymers of specific nature,
(Ikegame et al., 2003), phase inversion freeze drying (Kaiser et al., soluble/insoluble drug particles and nanoparticles of drugs/functional
2014), spinneret based tunable engineered parameters technique material can be added to electrospinning solution (Frenot and Chronakis,
(STEP) (Stojanovska et al., 2016) and pressurized gyration process 2003; Zhang and Yu, 2014).

Abbreviations: AIDS, acquired immunodeficiency syndrome; CMAs, critical material attributes; CPPs, critical process parameters; DMA, dimethylacetamide; DMF, dimethylformamide;
DMSO, dimethyl sulfoxide; EHDA, electrohydrodynamic atomization; FA, formic acid; FGF, fibroblast growth factor; HFIP, 1,1,1,3,3,3-hexafluoro-2-propanol; HIV, human im-
munodeficiency virus; HPβCD, hydroxypropyl-β-cyclodextrin; LCST, lower critical solution temperature; MIC, minimum inhibitory concentration; MRSA, methicillin-resistant
Staphylococcus aureus; NEU, nanofibers electrospinning unit; NIR, near infrared; NMMO, N-methyl morpholine oxide; ODT, orally disintegrating tablets; PCL, polycaprolactone; PEG,
polyethylene glycol; PEO, polyethylene oxide; PLA, poly(L-Lactic acid); PLGA, poly(lactic-co-glycolic acid); PLLA, poly(L-Lactide acid); PMMA, polymethyl methacrylate; PVA, polyvinyl
chloride; PVP, polyvinyl pyrrolidone; SRP, scaling and root planing; STEP, spinneret based tunable engineered parameters; TFA, trifluoroacetic acid; THF, tetrahydrofuran; VEGF,
vascular endothelial growth factor; XRD, X-ray diffraction

Corresponding author.
E-mail address: manju@niperahm.ac.in (M. Misra).

http://dx.doi.org/10.1016/j.ejps.2017.07.001
Received 9 May 2017; Received in revised form 23 June 2017; Accepted 2 July 2017
Available online 08 July 2017
0928-0987/ © 2017 Elsevier B.V. All rights reserved.
S. Thakkar, M. Misra

Table 1
Comparison of numerous nanofiber production techniques.

Method Governing forces Working principle Parameters affecting fiber properties Productivity Nanofiber Scalability on commercial level References
diameter
obtained

Electrospinning Electromechanical and Single jet splaying due to very high Applied voltage, tip-to-collector 5 μL/min 40 nm-2 μm Possible (Stojanovska et al.,
hydrodynamic forces voltage supply, electro- distance, temperature, humidity, 2016)
hydrodynamic atomization solution viscosity, flow rate and
electrical conductivity
Solution blowing Forces from high speed compressed Partition of single jet is commenced Gas pressure, nozzle geometry, Flow 20 μL/min 40 nm-several μm Possible (Polat et al., 2016;
air by virtue of compressed air (in high rate and solution viscosity Stojanovska et al.,
speed) 2016)
Centrifugal spinning Centrifugal force Partitioning of single jet leaving Orifice diameter, rotational speed, 1 ml/min 25 nm-several μm Possible (Stojanovska et al.,
orifice due to centrifugal force guides flow rate, solution surface tension and 2016; Taghavi and
the production of nanofibers viscosity Larson, 2014)

149
Pressurized gyration Forces of pressurized gyration Technique provides a combination of Polymer concentration, rotational 0.1 kg/min 60–1000 nm Possible (Mahalingam and
process centrifugal spinning and solution speed, working pressure of processing Edirisinghe, 2013)
blowing system
a
Template synthesis Vacuum pressure or capillary action Molding process, matrices with nano- Template shape, pore size and driving 40 to some Not amenable to scale up due (Ikegame et al., 2003;
of the polymer (electrostatic forces scale pores, made up of force hundred nm to production of fibers with Stojanovska et al.,
can also be used) polycarbonate and alumina are used shorter length 2016)
as templates
a
Phase inversion Reduced pressure of freezing and Freezing of very dilute polymeric Freezing rate and polymer 50 nm-1 μm Not amenable to scale up due (Kaiser et al., 2014;
freeze drying sublimation solutions and subsequent removal of concentration to various challenges Stojanovska et al.,
solvent by sublimation associated with maintaining 2016)
proper conditions
STEP techniques Forces from rotational and Micropipette spinneret from droplet Substrate speed, properties of 15–100 μL/min 50 nm to several Not possible (Stojanovska et al.,
translational motions by rotational and translational polymer solution (concentration and μm 2016; Wang and Nain,
forces, a movable substrate extends it molecular weight) 2014)
to filament

a
Values are not reported in reference cited.
European Journal of Pharmaceutical Sciences 107 (2017) 148–167
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Fig. 1. Use of nanofibers in daily needs.

Polymeric nanofibers are attaining great interest in recent years due by very few reports, found in the literature. Nanotechnology has pro-
to its wide applications (with incorporation of some functional mate- vided the pharmaceutical industry with new means of targeted drug
rials and remodeling) in numerous fields including electronics, fabric delivery and it would be interesting to see how electrospinning com-
formation (as protective clothing), separation industries (as filter bined with nanotechnology, could be the harbinger of a new era in drug
media/aids) medical field (tissue engineering, wound dressing material, delivery. In the present review, we have discussed the various ther-
artificial organs, tissue grafts), photonics, biosensors, chemistry (as apeutic areas where electrospinning is used for drug delivery their
catalyst), enzyme immobilization, as nano-composites and many others advantages and disadvantages. Emphasis is also laid on critical pro-
(Haider et al., 2015) as shown in Fig. 1. cessing parameters (CPP) and critical manufacturing attributes (CMA)
Nanofibers are different from other fibrous structures with a dia- that may affect electrospinning process. An attempt is also made to
meter in the range of nanometers while the length in the range of elaborate on the effect of these CPP and CMA on nanofiber morphology
meters, and are thus referred to as link between nanotechnology and especially upon nanoparticulate drug loading with specific applications
micron size world (Begum et al., 2012; Bhardwaj and Kundu, 2010; in drug delivery and tissue engineering.
Demir et al., 2004; Frenot and Chronakis, 2003; Rujitanaroj et al., 2008;
Zhao et al., 2006). A number of synthetic, semisynthetic and biological
polymers have been used to form nanofibers using electrospinning 2. Drug/drug nanoparticle fabrication in nanofibers
process as summarized in Table 2. However, not all the polymers are
capable of forming nanofibers, alginates are reported to have the 2.1. Electrospinning, the basic principle
property of gelation at very low concentrations while at higher con-
centration, its solution becomes too viscous, to be injected. Often to Electrohydrodynamic atomization (EHDA) is a process well con-
improve this behavior of alginates cosolvent/surfactant are added to trolled and driven by two regulatory forces working in combination as
electrospinning solution to aid the process of nanofibre formation electromechanical and hydrodynamic forces. Since decades, this pro-
(Bhattarai and Zhang, 2007). It is reported that, polymer solutions cess has been utilized for skillful and precise production of particles or
depending upon the molecular weights and concentration used, exhibit fibers in a definite size range and morphology (Parhizkar et al., 2016b;
electrospinnability only when polymer concentration and viscosity is Xie et al., 2015). Basically EHDA process is divided into two main op-
optimum. Usually solutions with lower polymer concentration results in erative techniques as electrospinning and electrospraying depending
electrospraying instead of spinning. Electrospinnability of low mole- upon the processing conditions and properties of polymeric solution
cular weight polymers can be enhanced, utilizing supramolecular ap- (Husain et al., 2016; Pina et al., 2017). One should pay considerable
proaches which allow cross-linking via hydrogen bonding or co-ordi- attention to parameters involved in turning one process to other, as
nation interactions in solution using small molecules (Wang et al., these are critical to final product and the properties desired, more so if
2016). One should be cautious in selection of concentration and visc- the drug is potent. Polymer concentration is crucial factor affecting
osity ranges of working solutions to get desired electrospinnability of product output from particle to fiber. Recently Husain et al. have in-
polymer used. vestigated the effect of polymer concentration, flow rate, tip-to-col-
A sensible number of nanofibers based products produced by elec- lector distance and applied voltage on the transition threshold for
trospinning are already available commercially like NanoAligned™ and particle to fiber. Findings of the study on PLGA suggested polymer
NanoECM™ which are electrospun fiber multiwell plates useful for high concentration to be the key factor involved. From investigated con-
throughput cell culture, cancer research and regenerative medicine. centration range of 2–25% three different zones were differentiated as
Similarly, AVflo™ commercialized by Nicast; is a certified polyurethane 1–10%, 10–20% and 20–30% concentration zones which resulted in
vascular access graft used in hemodialysis. Fig. 2 provides a brief particles, particles with beads on strings and beaded/bead-less fibers
glimpse of several nanofibers based products obtained from electro- respectively. A complete transition from particle to fiber state was re-
spinning specifically used in drug discovery or for therapeutic appli- ported at a concentration of 20% while other investigated parameters
cations ([19-09-2016], n.d; [18-09-2016], n.d). had a minute or no impact on the transition of aforementioned opera-
Electrospinning is a well-established technique for nanofiber for- tional techniques (Husain et al., 2016). In general it could be concluded
mation, more so in the area of textile industry, separation industry in- that very low polymer concentrations results in electro spraying with
cluding biomedical applications; however the use of this technique in final product yield in particulate product. Interestingly, change in
pharmaceuticals for drug delivery is still largely unexplored, as evident concentration of polymer, was also associated with different values of
surface tension, electrical conductivity and viscosity of final product

150
Table 2
Polymers used in electrospinning process and their applications.

Polymer Solvent used Monomer Applications Reference

Biopolymers in electrospinning
S. Thakkar, M. Misra

Polysaccharides and Cellulose N-methyl morpholine oxide β(1 → 4) linked D-glucose In textile, paper, plastic industries as food (Kang et al., 2002)
derivatives (NMMO) additives, and as the propellant
Cellulose acetate Acetone β(1 → 4) linked D-glucose with two acetyl In textile, paper, plastic industries as food (Jaeger et al., 1998;
group per molecule additives, and as a propellant Konwarh et al., 2013)
Ethyl cellulose Tetrahydro furan (THF)/dimethyl β(1 → 4) linked D-glucose with some of the As a carrier for loading of functional material (Park et al., 2015; Wu et al.,
acetamide (DMA) hydroxyl groups converted to ethyl ether 2005)
Propionyl cellulose Acetone β(1 → 4) linked D-glucose with propionyl As a carrier for loading of functional material (Jaeger et al., 1998)
groups bound to some of the hydroxyl groups
Methyl cellulose Water/ethanol β(1 → 4) linked D-glucose with methyl groups As a carrier for loading of functional material (Frenot et al., 2007)
bound to some of the hydroxyl groups
Hydroxypropyl cellulose Anhydrous ethanol β(1 → 4) linked D-glucose with hydroxypropyl As a carrier for loading of functional material (Gencturk et al., 2016;
groups bound to some of the hydroxyl groups and in ceramic industry Shukla et al., 2005)
Hydroxypropyl methyl cellulose Water/ethanol β(1 → 4) linked D-glucose with hydroxypropyl As a carrier for loading of functional material (Frenot et al., 2007; Paaver
methyl groups bound to some of the hydroxyl et al., 2015)
groups
Carboxymethylcellulose Methanol/water β(1 → 4) linked D-glucose with carboxymethyl As a carrier for loading of functional material (Kessick et al., 2004)
groups bound to some of the hydroxyl groups
Chitin 1,1,1,3,3,3-hexafluoro-2-propanol N-acetyl glucosamine Tissue scaffolding and wound dressing with (Min et al., 2004; Noh et al.,
(HFIP) functional materials 2006)
Practical grade chitin HFIP N-acetyl glucosamine Tissue scaffolding and wound dressing with (Schiffman et al., 2009)
functional materials
Chitin/poly (glycolic acid) HFIP N-acetyl glucosamine Tissue scaffolding and wound dressing with (Park et al., 2006a)
functional materials
Chitin/silk fibroin HFIP N-acetyl glucosamine Clothing (Park et al., 2006b)

151
Chitosan Trifluoroacetic acid (TFA) Random distribution of β(1 → 4) linked D- Removals of metals from solutions for (Mengistu Lemma et al.,
Ethanol/water glucosamine (deacetylated) and N-acetyl- environmental use and wound dressing with 2016; Ohkawa et al., 2004)
glucosamine functional materials
Chitosan/polyvinyl alcohol (PVA) Aqueous acetic acid Random distribution of β(1 → 4) linked D- Wound dressing with functional materials and (Ohkawa et al., 2004)
glucosamine (deacetylated) and N-acetyl- as carrier material
glucosamine
Hexanoyl chitosan Chloroform Hexanoyl units connected to some of the Wound dressing with functional materials and (Neamnark et al., 2006)
hydroxyl groups of chitosan monomer as carrier material
Dextran Dimethyl sulfoxide (DMSO)/water α-1,6-linked-D-glucose For incorporation of some functional material (Jiang et al., 2004)
Proteins Collagen and gelatin/polyethylene HFIP Irreversibly hydrolyzed collagen As tissue scaffolds and wound healing (Boland et al., 2004;
oxide (PEO) Matthews et al., 2002)
Silk/PEO Formic acid (FA) Fibroin protein Fabric industry (Park et al., 2004)
Human and bovine fibrinogen fraction HFIP/Earle's medium Fibrin protein Blood clotting and other protein reactions (Wnek et al., 2003)
I
Casein/poly ethylene oxide (PEO) THF Calcium, phosphorous and serine residues Food supplement (Xie and Hsieh, 2003)
Zein/hyaluronic acid/PVA Ethanol/water – Material for medical and packaging (Yao et al., 2007)
applications
Nucleic acid DNA Ethanol/water Nucleosides Delivery of genetic material (Fang and Reneker, 1997)

Synthetic and semisynthetic polymers


Polyurethanes DMF Organic units joined by carbamate Protective clothing (Schreuder-Gibson et al., 2002)
links
Polycarbonate Dichloromethane Organic units joined by carbonate Sensor and filter (Bognitzki et al., 2001b)
links
Polyacrylonitrile DMF Acrylonitrile Carbon nanotubes (Dzenis and Wen, 2001)
Polystyrene THF Styrene As catalyst (Torres, 2001)
Poly vinyl phenol THF Phenol moieties connected with Antimicrobial agent (Kenawy and Abdel-Fattah, 2002)
vinyl group
Polylactic acid Dichloromethane Lactic acid and lactide Drug delivery system (Caruso et al., 2001)
(continued on next page)
European Journal of Pharmaceutical Sciences 107 (2017) 148–167
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

obtained. In proceeding sections our main emphasis will be on elec-


trospinning process.
The electrospinning process utilizes high electrostatic forces for
nanofiber generation; generally from a polymeric solution or melt. The
main assembly of electrospinning unit comprises of electrical supply
Reference

(for generation of high electrical voltage), a syringe (contains solution/


melt/suspension to be electrospun), metallic needle (as a capillary) and
a grounded conductive collector (rotatable drum or static plate type) all
enclosed within a chamber (Schiffman and Schauer, 2008). The process
involves an application of the high electric field to the tip of the needle
which contains the solution fluid held in the form of a droplet, by
means of surface tension. The flow rate of solution/melt is controlled by
a syringe pump. As the electric field is increased the hemispherical
shape of solution droplet at the tip starts to elongate and forms a conical
shape known as Taylor cone. Further increase in electric field reaches a
(Bognitzki et al., 2001a)

point where repulsive forces overcome surface tension and a charged jet
is ejected from the apex of a conical meniscus. This charged jet after
(Li et al., 2002)

ejection from the tip is associated with elongation and various fluid
Applications

instability processes, simultaneously solvent start evaporating; finally,


fibers in the form of a solid non-woven mat are obtained on grounded
collector (Huang et al., 2003). Fluid instability also termed as ‘splaying’
is considered the process of splitting of the single jet into multiple fi-
laments due to repulsive forces generated by charge (Shin et al.,
2001).Various CPPs known to affect nanofiber properties are; applied
voltage, the distance between tip and collector and temperature inside
the chamber. Besides these CPPs, some parameters related to material
Scaffold for tissue engineering

have their impact on nanofiber properties and are known as CMAs.


These CMAs include the concentration of polymer, the viscosity of the
solution, the surface tension of solution and volumetric charge density.
Table 3 gives a cursory glimpse of these mechanistic aspects of nano-
fiber processing and its effect as reported in the literature. Numerous
Sensor, filter

commercial machines are available for electrospinning of feed to na-


Monomer

nofibers from various organizations as Elmarco, Katotech and others


(Angammana and Jayaram, 2016). Mass productivity always remains a
major concern, when it comes to industrial scale production, as in-
dustries engaged in large-scale production of nanofibers, require op-
diones) of glycolic acid and lactic

timum productivity. Despite the fact that lab scale and pilot scale
cyclic dimers (1,4-dioxane-2,5-

production is easier for electrospinning machines; challenges in their


scale-up to industrial level cannot be ignored. Improved techniques
with large mass productivities are emerging these days with the com-
bined efforts of academia and industries. One can select commercially
N-vinyl carbazole

available set-ups with good yield without compromising their specifi-


Solvent used

cations. Elmarco, Finetex, Katotech and Xanofi are some organizations


providing instruments with high productivities of 0.171 kg h− 1,
6.5 kg h− 1, 1.8 km h− 1 and 5 kg h− 1 respectively (Luo et al., 2012;
acid

Teo et al., 2011). Table 4 provide detailed information of commercially


available electrospinning machines along with their working principle.

2.2. Fabrication: mechanistic approaches

There are two ways for incorporation of drug/drug nanoparticles


into the nanofibers namely direct method and indirect method. The
Dichloromethane

direct method utilizes the approach of directly dispersing drug/drug


nanoparticles in electrospun solution. For drug/drug nanoparticles
which are not getting easily and uniformly dispersed within the elec-
THF

trospun solution; indirect method of nanoparticle fabrication is used. A


brief description of both of the methods is given in proceeding sections.
Poly(lactic-co-glycolic acid)

2.2.1. Direct method


The direct method is the conventional and most widely used method
Polyvinylcarbazole
Table 2 (continued)

for incorporation of drug/drug nanoparticles into nanofibers. Drug/


drug nanoparticles can be easily incorporated in nanofibers by pre-
paring homogenous dispersion with the polymeric solution used for
Polymer

electrospinning. Properties of nanofibers made of nanoparticles can be


easily modified by changing the amount of nanoparticles added. In case
one solvent is not suitable for incorporation of both the drug/drug

152
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Fig. 2. Few nanofibers based commercial products


produced by electrospinning process.

nanoparticles and polymer to be electrospun; two miscible solvents can randomly distributed in polymeric nanofibers; silver nanoparticles have
be used to individually dissolve/disperse drug/drug nanoparticles and reported forming the chain-like structure in electrospinning solution of
subsequently can be mixed with each other. Occasionally Surfactants PVA polymer after mechanical shaking (He et al., 2009). Some nano-
can be added to modify the surface of drug/drug nanoparticles to make particles are found to be specifically present on the surface and some
them easily dispersed (Zhang and Yu, 2014). Several metal/metal oxide are concentrically arranged depending upon the diameter and proces-
nanoparticles, polymeric nanoparticles, and organic nano-chains have sing conditions. If the diameter of a nanoparticle to be incorporated is
been electrospun in different diameters and with different types of below the diameter of nanofibers; these are known to be randomly
polymers using this direct method of nanoparticle incorporation. Gold arranged. Nanoparticles with a diameter of 260 nm and above are
nanoparticles have been electrospun by direct incorporation method for aligned with the axis of nanofibers and form necklace type structure as
their use as scaffolds in skeletal muscle tissue engineering (McKeon- shown in Fig. 3 (Jin et al., 2009; Zhang and Yu, 2014). The size de-
Fischer and Freeman, 2011). Due to photothermal properties of gold pendent alignment of SiO2 could be attributed to two possibilities as
nanoparticles; gold nanoparticle fabricated nanofibers can provide a either smaller size of particles force them to aggregate for a reduction in
therapeutic strategy for cancer treatment (Abadeer and Murphy, 2016) overall surface area or the diameter of nanofibers which is larger than
as well as can be utilized as a source of localized heat for melting of the size of nanoparticles wants them to pack inside in the form of
polymers and alteration in their nanostructure (Maity et al., 2011). clusters. When a mixture of two or more solvent is used positioning of
Silver nanoparticles which are well known for their antimicrobial nanoparticles depends upon vapor pressure. SiO2 nanoparticles were
property have been electrospun in different diameters of gelatin na- reported to migrate to the surface from the middle of nanofibers when
nofiber mats (Rujitanaroj et al., 2008); as well as in PVA nanofibers the vapor pressure of solvent system used (THF/DMF) was decreased
(Tan et al., 2016). These metallic nanoparticles have found to be (Hsu and Liu, 2010). Nanofibers of quantum dots (inorganic

Table 3
Effect of CPPs and CMAs on properties of nanofibers.

Parameter/attribute Effect Reference

Distance between tip and collector As distance is decreased nanofibers become more sticky and tend to stick to collector as (Huang et al., 2003)
well as to each other, diameter increases with decrease in distance
Viscosity of solution/melt As viscosity is increased nanofibers become thicker in diameter but are more continuous (Baumgarten, 1971; Doshi and Reneker,
in nature; while reduced viscosity is associated with finer and shorter nanofibers 1993; Huang et al., 2003)
Concentration of polymer Higher the concentration; higher will be the nanofiber diameter, also the chances of bead (Huang et al., 2003; Sill and von Recum,
formation are less in case of higher polymer concentration 2008)
Operating temperature Diameter of nanofiber obtained at higher temperature are generally more uniform (Huang et al., 2003; Zampetti et al., 2011)
Applied voltage Increase in voltage results in larger nanofiber diameter with less bead formation (Shao et al., 2015; Sill and von Recum,
(optimum value is required) 2008; Theron et al., 2004)
Surface tension Reduction of surface tension results in avoidance of bead formation, lower fiber diameters (Doshi and Reneker, 1993; Huan et al.,
are obtained 2015)
Volumetric charge density and solvent Increase in charge density and solvent conductivity results in finer nanofibers with fewer (Sill and von Recum, 2008)
conductivity chances of bead formation and finer diameter
Flow rate Increase in flow rate is generally associated with increased fiber diameter (Megelski et al., 2002; Sill and von Recum,
2008)
Solvent volatility Increased solvent volatility results in higher chances of pores formation and increased (Liu et al., 2015b; Megelski et al., 2002)
surface area
Temperature Increase in temperature is associated with decrease in nanofiber diameter (Chen et al., 2016)
Humidity Formation of circular pores takes place on nanofibers with increase humidity (Chen et al., 2016)
Concentration of drug and polymer In case of electro-spraying, reduction in concentration of polymer usually results in (Parhizkar et al., 2016b)
uniformly smooth particle with reduced size, and polydispersity index (PDI)

153
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Table 4
Commercially available electrospinning instruments, their manufacturers, and principle of working.

Name of the provider Area Name of the instrument Working principle Pictorial view

Elmarco Czech Republic NS 1S500U Production Line Smallest nanofiber production equipment, working on the
principle of bath spinning using a rotatory cylinder

Katotech Japan Nanofiber electrospinning Capable of producing 50–800 nm fibers, working on the
unit (NEU) principle of nozzle spinning with 400 needles

Nadetech innovations Spain ND-ES lab electrospinning Nozzle spinning with precise control on all parameters
unit

IME technologies Netherland EC-DIG 4 integrated nozzle technology

Finetex technology South Korea – Nozzle spinning with block nozzles

Fig. 3. SEM and TEM images of electrospun PVA/SiO2 fibers with different SiO2 diameters: (A, B) SEM images of 143 nm, PVA: SiO2 = 500:500; (C, D) SEM images of 265 nm, PVA:
SiO2 = 800:200; (E, F) SEM images of 910 nm, PVA: SiO2 = 500:500; work distance 10 cm, voltage 10 kV, TEM images of (G) TEM images of 143 nm, PVA: SiO2 = 500:500, (H) TEM
images of 265 nm, PVA: SiO2 = 700:300, (I) 910 nm, PVA: SiO2 = 300:810 (after removing part of water in the solutions); work distance 10 cm, voltage 15 kV.
“Reprinted (adapted) with permission from (Jin et al. Fabrication of necklace-like structures via electrospinning. Langmuir 2009, 26, (2), 1186–1190.) Copyright (2010) American
Chemical Society.”

154
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

nanocrystals) have shown to have luminescent property. A Photo- surface; In general, for surface loading of nanoparticles, nanofibers are
luminescence spectrum of CdTe quantum dots in nanofiber form was dipped in colloidal dispersions of nanoparticles, to allow surface ad-
found to be blue shifted due to different quantum confinement effect in sorption of these nanoparticles by means of various types of interac-
different media (Cho et al., 2010). Nanoparticles have already been tions. Electrostatic forces, hydrogen bonding and interactions between
established as a platform for delivery of gene, proteins, and drugs to the various functional groups are some of the interactions responsible for
biological system. The simplest way of delivery of aforementioned xe- this type of adsorption. Mahanta et al. have shown the enhanced effi-
nobiotic to the body is encapsulation into polymeric nanoparticles; cacy of PVA nanofibers to adsorb gold and silver nanoparticles after
which can be mechanically strengthened by means of electrospinning in surface modification of hydroxyl group with thiol and amine group.
nanofiber. Innumerable polymeric nanoparticles have been successfully They have shown 90% increase in the propensity of nanofibres to in-
electrospun in nanofiber sheets; such as poly(Ɛ-caprolactone), poly teract with silver and gold nanoparticles, upon surface modification
(styrene-block-isoprene), polyacrylamide, poly(lactide-co-glycolide), (Mahanta and Valiyaveettil, 2011). In- situ reduction (Chen et al., 2008;
polyurethane, poly(methyl methacrylate) and poly(hexamethyleneadi- Li et al., 2004), hydrothermal assisted process (Hedenquist and
pate)-PEO block copolymers (Zhang and Yu, 2014). Coaxial electro- Lowenstern, 1994; Shi et al., 2013), ultra-sonication and sputtering
spinning and blending represent a part of direct incorporation methods. etching (Hsu et al., 2013; Hsu et al., 2012; Wu et al., 2013) are some of
A unique approach for the direct method of incorporation is elec- the indirect methods used for surface fabrication of nanoparticles to
trospinning of cyclodextrin inclusion complex drug delivery systems. A nanofibers (Zhang and Yu, 2014).
small number of researchers have reported polymer free electrospin- The other way of drug/drug nanoparticle incorporation into nano-
ning of various drug candidates with incorporation into cyclodextrin fibers is the formation of nanoparticles within electrospun nanofibers.
inclusion complexes. Celebioglu et al. have reported the formation of In this technique, metallic or ceramic precursors are first electrospun in
polymer free nanofibers with electrospinning of highly concentrated the form of nanofibers and then other treatments like calcination, gas-
(160% w/v) aqueous solutions of hydroxypropyl-β-cyclodextrin solid reaction, and laser ablation are done to form nanoparticles within
(HPβCD) as such, along with urea and its inclusion complexes with nanofibers (Zhang and Yu, 2014).
triclosan as depicted in Fig. 4 (Celebioglu and Uyar, 2011). Urea is used There are certain limitations of the indirect method as many of these
as it is well known for breaking hydrogen bonds of cyclodextrins and are more suitable for incorporation of metallic nanoparticles. Indirect
prevents the aggregation of these during the formation of nanofibers at fabrication requires polymeric nanofiber to be stable and not to get
an optimum concentration. dissolved in the colloidal solution used for pretreatment. In-situ reduc-
tion method is associated with limitation of re-oxidation, further
2.2.2. Indirect method treatments like vacuum drying at room temperature and use of organic
As described in preceding sections for some drugs/drug nano- stabilizers can help to prevent these problems but at the same time
particles which are not homogeneously dispersed in electrospinning increases the cost of all over processing (Li et al., 2004). Hydrothermal
solution even after the use of dispersing agents; surface treatment be- assisted process is a well-known method for nanoparticle fabrication;
comes a necessary step for incorporation into nanofiber mats. Based on different morphologies (sphere, plate, and rod) of various nanoparticles
different methods for surface treatment nanoparticles can be formed on can be easily synthesized on electrospun nanofiber surfaces. Both the
the surface or within the nanofiber. Surface treatment of electrospun direct and indirect methods have their own advantages and limitations;
nanofibers is very easy way to get nanoparticles attached on the depending upon the requirements of the user, limitations can be

Fig. 4. SEM image of (A) HPβCD nanofibers obtained from a 160% (w/v) HPβCD solution and (B) HPβCD nanofibers with 5% (w/w) urea (C) HPβCD nanofibers with 10% (w/w) urea
and (D) bead structures obtained from 160% (w/v) HPβCD containing 20% (w/w) urea.
“Reprinted (adapted) with permission from (Celebioglu et al. Electrospinning of polymer-free nanofibers from cyclodextrin inclusion complexes. Langmuir 2011, 27, (10), 6218–6226.)
Copyright (2011) American Chemical Society.”

155
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

overcome by subsequent treatment strategies. achieved could be due to one of the two reasons. First being a reduction
Apart from electrospinning; electro-spraying can be utilized for di- in fiber diameter owing to stretching could help in the faster release as
rect production of nanoparticles (desired size and morphology) by fiber diameter is known to affect drug release. The second reason could
controlling properties of polymeric solution along with processing be smaller diameter which can lead to decreased distance to diffuse for
conditions as described in preceding sections. In line to this Eltayeb drug particles situated in the core of nanofibers. In addition to this
et al. have reported production of core-shell lipid nanoparticles using PLGA rich nanofibers have shown surface imperfections due to the re-
ethyl-vanillin as active ingredient and stearic acid as model shell for lease of drug as shown in Fig. 5.
controlled release of actives. After optimization of process parameters,
nanoparticles were shown to have size in range of 60–70 nm with en- 3.2. Drug-related parameters affecting drug release
capsulation efficiency of 70% (Eltayeb et al., 2016). In the same year
Parhizkar et al. have reported production of cisplatin loaded PLGA Drug-related parameters affecting its release form nanofibers are
nanoparticles for controlled drug delivery. Applied voltage, flow rate drug loading, molecular weight, the physical state of drug, solubility,
and the concentration of cisplatin and polymer were found to be key and drug-polymer interactions (Hrib et al., 2015). In general, higher
parameters affecting particle size. Prepared nanoparticles with 70% drug loading is associated with the faster release. The crystalline form
entrapment efficiency demonstrated sustained release of drug of the drug gets deposited on nanofiber surface and provides burst re-
(Parhizkar et al., 2016a). Same group have reported production of core lease; while amorphous form gets deposited deeper inside and get re-
shell nanoparticles of same drug by electrohydrodynamic atomization leased in a sustained manner. Low molecular weight drugs are known
process (Reardon et al., 2017). Studies show the potential of electro- for their fast release rate (Natu et al., 2010). Fig. 6 provides glimpse of
spraying technique for production of various nanoparticles with effec- drug related parameters affecting drug release.
tive control over size. Electrospinning technique is almost fully ex-
plored in terms of drug delivery but there is room for electro-spraying 3.3. Stimuli-responsive drug delivery systems
which can further be explored in various drug delivery processes.
Research in the field of polymer sciences has given new opportunity to
3. Control of drug release from nanofibers formulation scientist by the discovery of stimuli-responsive polymers.
Stimuli-responsive systems are dependent upon the rate by which stimuli
Nanofibers can be used as release rate controlling devices based on get transferred to the corresponding responsive material. As nanofibers are
the choice of inactive used for fiber formation. Drug release from na- the concerns; their high surface area to volume ratio makes them the
nofibers could be due to desorption of drug from the surface, diffusion suitable candidate for faster response as compared to other materials
from pores and/or matrix degradation. These all processes of drug re- (Frenot and Chronakis, 2003; Zhang and Yu, 2014). Various electrospun
lease are likely to get affected by choice of inactive (polymer or other nanofibers are reported for stimuli-responsive delivery upon exposure to
material), porosity, morphology, and geometry of nanofibers (Goonoo external stimuli as pH, temperature, light and others. Demirci et al. have
et al., 2014). Generally smaller the diameter of nanofiber faster the reported pH sensitive nanofibers of polymer poly(4-vinylbenzoic acid-co-
release rate is considered from it based on the fact that smaller diameter (ar-vinylbenzyl)trimethylammonium chloride) [poly(VBA-co-VBTAC)] for
fiber has higher surface area and dissolution rate. Later findings sug- stimuli controlled the release of drug ciprofloxacin. The release profile of
gested that drug release cannot be solely governed by diameter and at drug was checked in acidic, neutral and basic medium to check the pH-
the same time effect of porosity is to be considered. It is often shown responsive behavior of drug-loaded nanofibers (Demirci et al., 2014) as
that thicker nanofibers with very high porosity release drug faster as depicted in Fig. 7. The study has shown the homogeneous dispersion of the
compared to thinner fibers with low porosity. Nanofiber alignment is crystalline drug in an amorphous form supported by the absence of any
another parameter known to affect drug release and in general rando- diffraction peak of drug crystals in XRD analysis. In this study authors have
mized pattern is associated with faster drug release because of in- examined release profile for 720 min (12 h) which doesn't seem to be
creased tendency of water uptake (Cui et al., 2006). enough for determination of mechanism of release (whether diffusion/
permeation controlled or via degradation of the polymeric matrix). Drug
3.1. Polymer parameters affecting drug release ciprofloxacin used here as the model drug is known to be showing pH
dependent solubility of its own; increase in drug release examination time
Various polymers related parameters are known to affect drug re- could have shown the clearer view of release mechanism involved as both
lease from nanofibers including polymer composition, crystallinity and the drug as well as the polymer is reported to have pH dependent beha-
molecular weight of the polymer. The polymer composition is a key vior. In another study conducted by Toncheva et al. the same drug ci-
parameter leading to drug loading and release from nanofibers; reports profloxacin was incorporated in nanofibers of poly(L-lactide-co-D,L-lactide)
says that use of hydrophilic and amphiphilic copolymers results in in- in crystalline form. Results of release study conducted in buffer pH 7.4
creased drug loading and decreased chances of burst release of the drug have shown initial burst release of 50% in initial 400 min; further ex-
(Chou and Woodrow, 2017). Polymers with very high crystalline re- amination for drug release up to 24 h does not lead to any significant
gions are known to have slower release rates as compared to amorphous increase in the amount. This clearly showed the release of drug which was
regions due to very low water uptake in highly ordered three-dimen- diffusion controlled. Authors in this study have reported the increase in %
sional arrays of crystals (Chou et al., 2015). Chou et al. examined the drug release by the addition of hydrophilic polymer PEG which could be
effect of the mechanical property of drug-eluting nanofiber on drug due to easier permeation of release media upon addition of hydrophilic
release by taking tenofovir as a model drug (Chou and Woodrow, polymer (Toncheva et al., 2012).
2017). Authors have conducted release studies in 15% (wt%) nanofi- In line with this Li et al. have reported the formation of hybrid
bers with varying concentrations of PCL and PLGA after mechanical nanofibers which can be reversibly controlled to deliver cells and drug
deformation to show the effect of mechanical properties of polymeric both by virtue of near infra-red (NIR) light. Hybrid nanofibers were
nanofibers on drug release as shown in Fig. 5. It was found that the drug composed of poly (N-isopropyl acrylamide), silica-coated gold nanorods
release was sustained in nature when no mechanical stretching was and polyhedral oligomeric silsesquioxanes. Co-electrospun nanofibers
applied. In contrast all the fibers when stretched to the levels of failure of these showed very large volume change of about 83% on exposure to
there was a shift in drug release from sustained to faster drug release as NIR light; interestingly this change is reported to be reversible with
PCL/PLGA (20:80) showed around 70% of drug release in 96 h after proper retention of nanofiber structures (Li et al., 2016). Doxorubicin
stretching as compared to the almost same drug release in 240 h for un- was used as a model drug and the nanofibers were reported to reduce
stretched sample (Chou and Woodrow, 2017). This faster release cell viability up to 97% in human cervical cancer HeLa cells lines.

156
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Fig. 5. Cumulative release curves of PCL/PLGA blend fibers loaded with 15% TFV (a) prior to mechanical stretching (b) after mechanical stretching to failure. (Open circles) 100PCL,
(open squares) 80PCL/20PLGA, (open triangles) 60PCL/40PLGA, (filled circles) 40PCL/60PLGA, (filled squares) 20PCL/80PLGA, and (filled triangles) 100PLGA. (c) SEM images of PCL/
PLGA blend fibers mechanically stretched followed by a 10-day in vitro release study, scale bar = 2 μm.
“Reprinted (adapted) with permission from (Chou et al. Relationships between mechanical properties and drug release from electrospun fibers of PCL and PLGA blends. J. Mech. Behav.
Biomed. Mater. 2017, 65, 724–733.) Copyright (2016).”

Fig. 6. Effect of nanofiber diameter, porosity, nano-


fiber alignment, polymer crystallinity and molecular
weight on drug release.

Studies on similar lines have shown the potential of nanofibers in en- hyperthermia along with drug could have resulted in better therapeutic
capsulating and delivering NIH3T3 fibroblast cells in their functional effects as well as a reduction in required dose of the drug. Liu et al. have
form. In this study photothermal property of gold nanostructures were reported the formation of nanofibers with thermo-sensitive copolymer
used to alter the swelling behavior of poly (N-isopropyl acrylamide) poly(N-vinyl caprolactam-co-methacrylic acid) (PNVCL-co-MAA);
which in turn was responsible for the release of drug/cells. The increase choosing captopril and ketoprofen as model drug candidates. The de-
in NIR laser density and time is reported to be associated with the in- veloped nanofibers exhibited slow release lasting several hours at 40 °C
crease in temperature. Precaution is required in the selection of NIR while burst release of few seconds was observed at 20 °C; following
exposure intensity and time which could lead to cell death at higher Korsmeyer–Peppas model as a major drug release mechanism (Liu et al.,
temperatures when the objective of the study is cell delivery in the 2016). Authors have clearly mentioned mechanism of release as below
functional state. The further study reported an in-vitro therapeutic effect lower critical solution temperature (LCST) polymer fiber is hydrophilic
of released doxorubicin alone; whereas hyperthermia is well-known and drug release is diffusion controlled. In contrast above LCST polymer
therapy for cancer treatment. Study of combination effect of fiber behaves as hydrophilic one and drug release becomes slower with

157
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Fig. 7. Stimuli-responsive drug release from various nanofibers.

more dependence on polymer degradation. Huang and coworkers effi- 4.1. Nanofibers for management of diseases/disorders related to brain
ciently worked on the preparation of thermal and pH sensitive nano-
fiber scaffolds of poly(NIPAAm-Co-N-methylo acrylamide) (PNN) and Dementia associated with Alzheimer's disease is a major neurode-
chitosan. They have reported thermal and pH sensitive release of drug generative disease in elderly patients. Various drugs used for treatment
doxycycline hyclate from PNN/chitosan nanofibers (Huang et al., are donepezil, rivastigmine, galantamine and others. Gencturk et al.
2014). Most of the researchers have failed to report release mechanism have reported preparation of polyurethane/HPC nanofibers of drug
of drug which could be an important parameter governing the overall donepezil hydrochloride for effective management of dementia asso-
performance of stimuli-responsive polymeric nanofibers. Based on ciated with Alzheimer's disease. The transdermal polymeric mat of the
aforementioned studies one can conclude that nanofibers can be ef- drug showed effective diffusion controlled delivery of drug following
fectively used as good candidates for stimuli-responsive drug delivery. Korsmeyer–Peppas release kinetics. In-vitro cytotoxicity studies by MTT
assay has shown nanofiber mat to be well tolerated and non-irritant for
skin (Gencturk et al., 2016). Aricept, marketed product of drug done-
4. Applications in drug delivery pezil hydrochloride is present in the form of orally disintegrating tablets
(ODT) having 5 and 10 mg of the drug. Incorporation of the drug in
Electrospinning being very versatile technique has been recognized nanofiber mat could provide easy control over dose as well as can result
useful in the formation of nanofibers for drug delivery applications. in better patient compliance in dementia. Traumatic and/or surgical
Recently nanofibers have gained huge attention as new drug delivery brain injury is associated with various pathological changes; giving rise
platforms based on the principles of increase in dissolution rates of the to severe parenchymal damage in the brain. Sulejczak et al. have re-
drug due to a tremendous increase in surface area. Besides this nano- ported delayed/reduces neuro-destructive phenomena after dressing
fibres are also amenable to easy modifications to alter release rate with L-lactide-caprolactone copolymer nanofiber net; whereas un-
controlling properties owing to stimuli-sensitive nature of some poly- dressed rat model of surgical brain injury was having massive neuro-
mers used in electrospinning, which can be suitably tweaked. Selection degeneration and scar formation (Sulejczak et al., 2014). Experimental
of inactive material (polymers) for fiber formation can be based on the results proved the ability of biodegradable polymer-based nanofiber
release properties desired like either only diffusion controlled or dif- mats in reducing scar formation associated with various brain injuries
fusion controlled along with nanofiber degradation as described above. which otherwise could hamper the proper functioning of the brain.
Similarly, various incorporation techniques (direct or indirect) can be Xie et al. have reported formulation of Paclitaxel-loaded PLGA na-
used to load drug onto nanofibers as previously described in preceding nofibers for the treatment of brain tumor as alternative drug delivery
section. Proceeding sections will deal with the use of nanofibers as drug device. The encapsulation efficiency of the drug in nanofibers was
delivery systems for various disease/disorders of the body. found to be more than 90%. DSC analysis was carried out in the study to
see the solid state property of drug, and it was found to be present in a

158
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

solid solution state. Sustained release of paclitaxel was reported along cyclosporine A nanofibers as an alternative mode of therapy. poly(L-
with IC50 value which was comparable to the commercial product in lactic acid) (PLA) was used as a polymer for nanofiber formation with
the cytotoxicity assay (Sulejczak et al., 2014). The main limitation of modified needleless Nanospider™ technology. Authors have checked
the aforementioned study is a requirement of preliminary surgery for the extent of inflammatory reaction and corneal healing by macro-
positioning of drug liberating biodegradable implant in the part having scopic, immunohistochemical and biochemical examinations. As com-
a cancerous growth. Proper studies mentioning any possible dose pared to blank nanofibers and eye drops of drug cyclosporine A; na-
dumping should be conducted when working with anticancer drug- nofibers of the drug were found to be more effective in suppression of
loaded nanofibers which would otherwise be associated with severe inflammatory response and neovascularization. The proposed reason
toxicities. In most of the studies reported till date, issues related to the for eye drops of the same drug to be less effective was lesser contact
homogeneity of drug distribution is not addressed. This becomes even time with an injured eye (Cejkova et al., 2016). Authors have reported
more pertinent, if drug under consideration is extremely potent (like representative photographs of alkali-injured eye after 12 days of injury
anticancer drugs) and not completely soluble in the prepared polymer along with eyes after treatment in various groups. It was shown that
solution, resulting in non-homogenous distribution. Therapeutic agents immediately after the alkali injury, the cornea became opalescent. On
showing an effect at very low concentration must be properly examined day 12 after the injury, the untreated injured corneas were showing
for their homogeneity in the sample, particularly when the drug is to be very high vascularization, and corneal transparency was nearly lost
incorporated in suspended form or in crystalline form. It is advisable while in the case of eye drop/blank nanofiber treated eyes were shown
under such circumstances to collect samples of uniform size (of drug to have partial restoration of corneal transparency. In compared to all
incorporated nanofiber sheet) from different parts of collector plate and these drug-loaded nanofibers treated eyes corneal neovascularization
analyze for the amount of drug to rule out any drug uniformity related was strongly suppressed and corneal transparency was restored along
issues. Fig. 8 displays role of nanofibers in the management of brain with restoration of physiological levels of central corneal thickness as
disorders. Except few, most of the studies have not reported the form of shown in Fig. 9. These findings suggested the effective use of drug
drug (crystalline or amorphous) as well as % loading, which could be an nanofibers for the treatment of various eye diseases/disorders over-
important parameter for drug release properties from nanofibers as coming the major hurdle associated with conventional ocular drug
described in preceding sections. The form of a drug not only gives the delivery. Preclinical studies in rabbit eye have depicted more effica-
idea about release profile but is also essential to find out the site where cious delivery of drug and recovery as compared to conventional eye
probable deposition of the drug will occur (on the surface or deeper drops however applicability of dosage form is still a cause for concern.
inside the nanofiber), making it an important parameter for formulation In this study authors have sutured drug containing nanofiber sheet to
development scientist. conjunctiva of eye, requiring a skilled personnel, a process which might
be cumbersome, if extrapolated in humans. Gagandeep et al. have re-
ported treatment of another disorder related to eye, glaucoma with
4.2. Nanofibers in the management of diseases/disorders related to eye nanofibers of drug timolol maleate and dorzolamide hydrochloride.
They have reported preparation of polycaprolactone (PCL) and PVA
Researchers have reported the efficient use of nanofibers with the nanofibers of these drug candidates and evaluated for the in-vivo effect
drug for treatment of various diseases and disorders related to eye. of nanofiber formulation by inserting it in a cul-de-sac of the glaucoma
Cejkova et al. have shown treatment of alkali-injured cornea with

Fig. 8. Electrospun nanofiber mediated drug therapy for brain diseases/disorders.

159
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Fig. 9. The appearance of injured and treated corneas. Representative photographs of healthy corneas (A), corneas immediately after the injury with 0.25 N NaOH (B), injured corneas
with nanofibers sutured to conjunctiva (C), injured untreated corneas on day twelve after the injury (D), injured corneas treated with CsA drops on day twelve after the injury (E) or
injured corneas treated with nanofibers drug-free (day twelve after the injury) (F) or injured corneas treated with CsA-loaded nanofibers (day twelve after the injury) (G).
“Reprinted (adapted) with permission from (Cejkova et al. Treatment of alkali-injured cornea by cyclosporine A-loaded electrospun nanofibers–An alternative mode of therapy. Exp. eye
Res. 2016, 147, 128–137.) Copyright (2016) Elsevier.”

induced rabbit. Results have shown the potential of formulation for compounds which are now-a-days frequently used for the conditions of
effective use in glaucoma; as intraocular pressure was found to be hypertension as well as angina pectoris. Singh et al. have reported
significantly reduced when compared with commercial eye drop for- preparation of nanofiber formulation of such class of compounds and
mulation (Garg et al., 2014). Hsu et al. have reported ophthalmic de- shown its use for the treatment of conditions like angina as depicted in
livery of drug dexamethasone for various disorders of eye. Many dis- Fig. 12. Nicorandil as drug candidate is associated with problems of low
eases related to eye require instillation of dexamethasone eye drops 4–6 bioavailability, slow onset of action and mucosal ulceration at the site
times a day and long term administration of disease is associated with a of action. Several carrier systems have been tried with drug and have
number of local and systemic side effects. Authors have shown for- shown no improvement for complications of mucosal ulceration and
mulation of PCL nanofibers and microbeads for extended release of the delayed onset of action. Author and coworkers have reported pre-
drug. This benefit of extended release for achieving efficacy with re- paration of polymeric nanofibers of vitamin B 12 and a blend of hya-
duced side effects was proposed due to increased crystallinity (Hsu luronic acid and PVA loaded with nicorandil drug for its sublingual
et al., 2016). Fig. 10 have shown diagrammatic presentation of the delivery. Electrospun nanofibers were found to be uniform, non-beaded
strategies. with the diameters of 200–450 nm. In-vitro drug release has shown
sustained drug release over a long period of time. Histopathological
images revealed the absence of mucosal ulceration at the site of ap-
4.3. Nanofibers in the management of diseases/disorders related to ear
plication. This study shows the potential of nicorandil nanofibers in
overcoming hurdles related to its delivery (Singh et al., 2015). These
Chances of ear infection with Methicillin-resistant Staphylococcus
kinds of approach could be effectively used to deliver drugs having
aureus (MRSA) are getting increased these days in patients with middle
solubility issues and require instant effect such as in the management of
ear surgery. The major problem associated with the treatment of peri-
pain. Besides, therapeutic actives having very high first pass metabo-
prosthetic infections is to treat an established biofilm. These biofilms
lism can be formulated as oral patches having nanofibers consisting of
are often known to be formed on implanted medical device like ossi-
immediate release polymers.
culoplasty prostheses and implantable hearing devices. Once formed
Intracranial aneurism is another disease related to a cerebrovascular
these are hard to disrupt and are associated with endo-cochlear in-
system which ultimately can lead to hemorrhagic stroke, brain damage
flammation, persistent non-traceable infections along with patient dis-
and death. Wang et al. have reported formulation of poly(L-lactide-co-
comfort (Jang et al., 2015). Authors have reported the formation of
caprolactone) nanofibers loaded with a potent anticoagulant, heparin
biocompatible and biodegradable PCL/PEO nanofibers for effective
and vascular endothelial growth factor (VEGF). Fibers were reported to
delivery of vancomycin for prevention of MRSA biofilm on the ossicular
be covered on stent surface; for use in the treatment of intracranial
prostheses and otitis media. A triphasic release profile was followed by
aneurism. Release profiles have shown the long-lasting release of both
a drug from drug-loaded nanofibers. In-vitro studies revealed the ab-
the moieties for more than 30 days without any initial burst release
sence of biofilm on implanted medical devices. Otitis media was found
effect. This study has shown the potential of drug nanofiber coated stent
to be devoid of MSRA after treatment with drug-loaded nanofibers
for treatment of aneurism and induction of endothelialization (Wang
(Jang et al., 2015). This seems to be an interesting and novel way of
et al., 2015b).
avoiding infections in otitis media during implantation of medical de-
For many reasons vascular surgery is associated with prosthetic
vices and can be explored further to deliver various classes of anti-
graft infections. Drug delivery system which can deliver antibiotics
biotics. Diagrammatic representation is shown in Fig. 11.
locally in a pattern of sustained release can provide an effective way of
treatment for these types of infections. Liu et al. have reported pre-
4.4. Nanofibers in the management of cardiac and vascular diseases/ paration of biodegradable nanofiber loaded vascular prosthetic graft for
disorders sustained release and local delivery of vancomycin in the treatment of
vascular infections. Eluting method has shown the higher release of
When heart receives lower volumes of oxygenated blood, it results drug for more than 30 days. Finally in-vivo implantation of nanofiber
in Angina pectoris. Potassium channel openers are the class of

160
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Fig. 10. Nanofibers for treatment of ocular ailments.

loaded grafts in subcutaneous pockets of rabbits has shown the poten- preparation of biosensors (Vellayappan et al., 2016). Injectable are able
tial of these systems for effective of prosthetic graft infections (Liu et al., to provide effective delivery of insulin/anti-diabetic drugs but is asso-
2015a). ciated with poor patient compliance. To develop oral means of ad-
ministration for diabetes treatment is a great challenge for pharma-
ceutical scientist till date. Xu et al. have reported preparation of
4.5. Nanofibers in the management of diabetes gelatin/insulin fibers by co-electrospinning for transbuccal delivery of
insulin. Extended release of insulin was found for 4 h. Released insulin
Diabetes is one of major and life-threatening disease of the 21st was checked for showing activity and was found to successfully trigger
century. Several microvascular and macrovascular complications are intracellular AKT phosphorylation. Results concluded successful trans-
associated with diabetes as effect on retina, kidneys and risks of heart buccal delivery of insulin from gelatin/insulin fibers formulated by co-
diseases. These complications can generally be minimized with the electrospinning of gelatin and insulin (Xu et al., 2015). Another elec-
control of blood glucose level. Electrospinning technique has been trospinning based nanofiber formulation for anti-diabetic effect is re-
utilized in the formation of biosensors as well as in the treatment of ported by Modgill et al. in 2015. They have shown the formation of
disease. Vellayappan et al. have shown an effective comparison of linagliptin loaded PVA nanofibers to form a biodegradable polymeric
conventional biosensors with advanced electrospun biosensors which sheet as shown in Fig. 1. Prepared nanofibers were reported to have
provide insight into the application of electrospinning for the

Fig. 11. Drug-loaded nanofiber therapy for ear infection.

161
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

encapsulation efficiency of 92%. When evaluated for mucoadhesive them for inhibitory effect against Escherichia coli. They have also re-
strength drug-loaded nanofiber patches were found to have higher ported it to be effective for testing of anticancer drugs (Švachová et al.,
mucoadhesion as compared to blank nanofibers; probably due to higher 2016).
surface area and water holding capacity. Results were found to be
comparable with commercial formulation upon evaluation of in-vivo 4.8. Nanofibers in the management of AIDS
activity (Modgill et al., 2015). Choi et al. have reported nanofibers with
basic fibroblast growth factor (bFGF) and epidermal growth factor AIDS is a syndrome due to infection from human immunodeficiency
(EGF) for the treatment of diabetic ulcers and reported an increase in virus (HIV) which adversely affects immunity of the patient making
accumulation of both collagen and cemented matrix of keratin. These him/her more vulnerable to other infections. It generally requires a
findings show promising potential nanofibers mats in the treatment of combination or multidrug therapy with antiretroviral agents. Blakney
diabetic ulcers (Choi et al., 2011). et al. have reported preparation of drug-loaded PVA nanofibers for
combined therapy of tenofovir and levonorgestrel for prevention of HIV
4.6. Nanofibers in the management of rheumatic diseases/inflammatory acquisition. Results show preservation of antiviral activity of drug te-
autoimmune diseases nofovir after release from nanofibers without any toxic effects. The
presence of levonorgestrel was shown not to affect the release and
Inflammatory diseases are associated with pain/discomfort and may pharmacodynamic profile of tenofovir (Blakney et al., 2014). Ball et al.
require acute/chronic treatment with anti-inflammatory drugs. As de- have reported preparation of nanofibers of PEO/PLLA blends loaded
scribed in preceding sections, the composition of nanofibers can be either with maraviroc or acyclovir for activity against HIV infection.
easily modified to have either instant release or controlled or sustained Drug release profile for both the drugs was found to be varied based on
release depending upon the properties required. On these basis nano- the ratio of PEO/PLLA blends. Effective HIV inhibition was reported for
fibers present promising drug delivery technique for the acute/chronic drug-loaded fibers upon in-vitro evaluation (Ball et al., 2012). Zovirex™
therapy of inflammatory diseases. Meng et al. have reported prepara- is the marketed product of acyclovir used for oral administration; these
tion of fenbufen loaded PLGA/gelatin nanofibers for effective treatment acyclovir-loaded nanofibers can provide an easy alternative if bioa-
of rheumatoid arthritis and osteoarthritis. When investigated regarding vailability profiles are similar. Comparison of the prepared formulation
drug release, the release was found to increase with an increase in with marketed product in term of release could have provided for easy
gelatin content and randomness of nanofiber arrangement as shown in comparison.
Fig. 12. Thus effective control over drug release was reported with these
drug-loaded composite nanofibers (Meng et al., 2011). A possible 4.9. Nanofibers in the management of diseases/disorders related to oral
reason for the increase in drug release could be the addition of gelatin. cavity
Addition of gelatin increases the charge density of the solution which
ultimately improves the stretching force and the self-repulsion of the jet Candida albicans is one of the very common organisms present in
leading to decrease in the fiber diameter and subsequently increase in oral and gastrointestinal tract (GIT) mucosa. Oral cavity environment
drug release. Fig. 12 provides glimpse of various nanofiber mats formed change is associated with a change in pathogenic state of the bacteria
by PLGA alone and after addition of gelatin. In line with this Park et al. which can ultimately lead to acute and/or chronic infections like can-
have reported ketoprofen loaded PLA nanoparticles for controlled re- didal mucositis and denture stomatitis. Santos et al. have reported na-
lease of drug. When evaluated to check release pattern nanofibers nocrystal based formulation of a common antifungal ingredient present
showed, increased release rates with an increase in drug content as well in mouthwashes to access the effective concentration of drug along with
as temperature (Park and Lee, 2011). the potential of nanofiber-based systems in the delivery of an antifungal
Saifaka et al. on similar lines reported preparation of teriflunomide agent for these types of oral infections. PVP/PMMA nanofibers were
loaded nanofibers of PLA and poly(butylene adipate) (PBAd) blends. used to incorporate cetylpyridinium chloride as antifungal agent and
Various techniques were used to check miscibility of polymeric blends investigation of antifungal activity has shown the potential of this
and state of drug been incorporated. The drug was reported to be in- system for use in oral infections (Santos et al., 2014).
corporated in amorphous form (Siafaka et al., 2016). In-vitro drug re- Chemotherapy/radiation therapy of cancer is associated with
lease was found to be dependent upon the diameter of nanofibers. For painful inflammation of oral mucosa known as oral mucositis. Tort et al.
analgesic and anti-inflammatory drugs selection of polymer plays an have reported preparation of glutamine loaded sodium alginate nano-
important role as in some chronic diseases sustained release of the anti- fibers with mucoadhesive properties for treatment of oral mucositis.
inflammatory drug is required (Fig. 13). Drug release profile has shown the release of 85% drug from nanofibers
within 4 h and shown the ability of system for effective mucoadhesion
4.7. Nanofibers in the management of lung diseases and drug delivery for prevention of inflammatory disease (Tort and
Acartürk, 2016).
Pneumonia is very common lung infection caused by Streptococcus Root canal infection is a very common problem of professionals
pneumoniae and is leading cause of deaths till date in developing as well working in the field of dentistry. Pankajakshan et al. have reported
as developed countries (Shaddock, 2016). Adibkia et al. have reported triple antibiotic nanofibers for intracanal drug delivery. In this study
nanofiber formulation of drug azithromycin for enhanced antimicrobial polydioxanone nanofibers were prepared with electrospinning tech-
effect against Streptococcus pneumoniae and Staphylococcus aureus. Na- nique and loaded with a combination of triple antibiotics (me-
nofibers made up with eudragit RS 100 was found to be showing en- tronidazole, minocycline and ciprofloxacin) and it's; effect on bacterial
hanced efficacy against Streptococcus pneumoniae as compared to Sta- death was evaluated. Results from confocal laser scanning microscopy
phylococcus aureus which showed lower MIC (minimum inhibitory have shown significant bacterial death upon exposure to antibiotic
concentration) values of 40 μg/ml and ˃120 μg/ml respectively. The containing nanofibers (Pankajakshan et al., 2016).
probable reason given was enhanced adsorption efficiency of nanofi-
bers on capsulated cell wall; which can provide an antibiotic depot on 4.10. Nanofibers in the management of various types of cancers
the bacterial surface. Results made it clear that these antibiotic drug-
loaded nanofibers can be a potential candidate for the eradication of Cancer is known to be disease condition characterized by un-
Pneumonia (Adibkia et al., 2016). controlled growth of cells which are capable of escaping general de-
Švachová et al. have reported formulation of novel gelatin/oxy- struction mechanisms of the body. Statistical reports have suggested an
cellulose nanofibers for suitable lung disease modeling and evaluated increase in new cases along with mortality rates of patients suffering

162
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Fig. 12. SEM image showing the morphology of electrospun FBF-loaded PLGA nanofibers (A, B) and corresponding solvent-cast film (C); FBF-loaded PLGA/gelatin (9/1) nanofibers (d–g)
and corresponding film (h).
“Reprinted (adapted) with permission from (Meng et al. Preparation and characterization of electrospun PLGA/gelatin nanofibers as a potential drug delivery system, Colloids Surf. B
84(1) (2011) 97–102.) Copyright (2010) Elsevier.”

from cancer, indicating serious insufficiency of current treatment stra- vivo evaluation have shown enhanced bioavailability and antitumor
tegies available. Nanofibers of various anticancer drugs have been tried efficacy of curcumin nanofibers prepared with PVP as polymer base
to either increase water solubility or to control the release profile of (Wang et al., 2015a). In another study by Qui et al. composite nanofi-
drug candidate. Lin et al. have reported preparation of magnetic elec- bers of doxorubicin-loaded electrospun PLA/mesoporous silica nano-
trospun chitosan nanofiber composite for treatment of cancer with particles are reported for post-surgical cancer treatment. Studies have
hyperthermia. In vitro cell incubation experiments indicated them to be suggested these composite nanofiber scaffolds to be a potential candi-
non-cytotoxic and effective enough to reduce tumor cell proliferation date for the effective treatment of post-surgical cancer (Qiu et al.,
(Lin et al., 2012). In line to this Wang et al. have reported enhanced 2013). Zong et al. have reported preparation of cisplatin-loaded mu-
bioavailability and anticancer effect of curcumin loaded electrospun coadhesive nanofibers for local chemotherapy of cervical cancer. Cis-
nanofibers. X-ray diffraction (XRD) studies have shown an even dis- platin released from nanofiber mats have shown higher accumulation in
tribution of curcumin in amorphous form in nanofibers. In-vitro and in- the cervical region instead of being accumulated in highly perfused

Fig. 13. A) Nanofibers in management of diabetes, B) nanofibers in the management of rheumatoid arthritis, C) nanofibers in the management of vascular diseases.

163
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Table 5
Patents related to drug-loaded polymeric nanofibers by electrospinning.

Patent title Area of invention/final product description Advantage/applications/claims Reference

Electrospunamorphous Stabilization of solid dispersions of amorphous drugs Patent claims enhancement of solubility of poorly (Ignatious and Sun,
Pharmaceutical compositions in polymeric nanofibers water soluble pharmaceutical actives along with 2006)
formation of amorphous solid dispersions of drugs in
nanofiber form to make them stabilized
Modulation of drug release rate Control of release rate of any additive in nanofibers Patent claims co-electrospinning of one or two (Shastri and Sy,
From electrospun fibers by electrospinning polymers of additives (active pharmaceuticals) to 2008)
modulate release of incorporated drug candidates
Electrospun pharmaceutical Field of invention is nanofibers of drug particles Patent claims enhanced solubility of active (Ignatious and
Compositions pharmaceuticals by a novel and alternative Baldoni, 2003)
technique of solubility enhancement
Biodegradable drug-polymer Field of invention is polymeric or cyclodextrin Biodegradable sustained release nanofibers have (Davis et al., 2005)
Delivery system containing nanofibers loaded with drug been claimed and certain embodiments contain
cyclodextrins as fiber forming content; these fibers
can be used for the treatment of ocular diseases/
disorders
Fiber-based nano drug delivery Invention relates to processes for creating extruded A drug delivery system in the form of homo- (Pourdeyhimi et al.,
Systems (NDDS) fibers containing a mixture of drugs and component, bi-component or multi-component 2009)
pharmaceutically acceptable polymers such that the fibers wherein one of the components comprise a
drugs form nanofibers drug compounded with a polymer carrier
Medical device applications of Invention pertains to medical devices and methods Invention claims formation of various medical (Dubrow et al.,
nanostructured surfaces comprising nanofibers devices based on nanofibers and may include use of 2005)
antimicrobial agent in it
Polymeric nanofibers for tissue engineering Invention is related to medical applications of Polymeric nanofibers either biodegradable or non- (Laurencin et al.,
and drug delivery polymer fibers and particularly the use of polymeric biodegradable can be made with various blends of 2007)
nanofibers in tissue engineering, medical devices polymers
and drug delivery
Nanofiber surfaces for use in The patent is mentions invention related to In some aspects the current invention comprises a (Dubrow and
Enhanced surface area applications nanofibers, and nanofiber structures having substrate comprising at least a first surface, a Daniels, 2009)
enhanced surface areas, as well as to the use of such plurality of nanofibers attached to the first surface,
nanofibers and nanofiber structures in various and, one or more specific moiety attached to one or
pharmaceutical applications more member of the plurality of nanofibers
Antibacterial nanofiber material and Invention belongs to the field of nano-fiber and its Invention claims use of polymers or polymeric (Long et al., 2009)
preparation method thereof preparation, and more particularly to an blends for nanofiber formulation containing
antibacterial nanofiber material and preparation antibacterial drug candidate
method
Method for producing shell-core structure This patent discloses shell - core structure of the drug Patent claims nanofiber formulation by co-axial (Qian Zhou et al.,
medicament nanofibre with coaxial Preparation as nanofiber, particularly to a coaxial electrospinning of drug bovine serum albumin, 2009)
electrostatic spinning technology electrospinning technique for preparing shell - core heparin, doxorubicin, paclitaxel, tetracycline
structure hydrochloride, griseofulvin, vitamin C, vitamin E, or
vitamin B2

peripheral organs like kidney, liver and others. This advantage of ac- temperature cells could easily be attached to and detached from the
cumulation at the local site when compared to intravenous route has fibers. In vivo investigations on rats indicated aforementioned nanofi-
shown the ability of mucoadhesive nanofiber mat to deliver drug at the bers to have much more potent wound healing properties than com-
site of action. Another advantage achieved is a better balance between mercial gauze (Li et al., 2017).
anticancer activity and systemic safety in equal drug dose as compared In the same year Alavarse et al. have reported tetracycline hydro-
to intravenous route (Zong et al., 2015). As described previously chloride-loaded electrospun nanofiber mats based on PVA and chitosan
homogeneity of the drug must be taken into account in case of highly for wound dressing. Burst delivery of drug during the first 2 h was re-
potent drugs like anticancer ones; which is not addressed in most of the ported to allow an effective antibacterial activity on the Gram-negative
reports. Escherichia coli as well as on the Gram-positive Staphylococci epidermidis
and Staphylococcus aureus (Alavarse et al., 2017).
4.11. Nanofibers as wound dressing materials
5. Clinical trials of drug containing nanofibers
Wound dressing materials are the ones designed for creating and
maintaining an environment which is most suitable for wound healing Nanofiber drug delivery systems are at the preliminary stages of
by providing easy gaseous exchange, absorbing exudates from the clinical trials supported by a small number of the literature in this re-
wound site and providing a sterile environment which does not support gard. These have been clinically verified for their use only in the area of
microbial growth (Li et al., 2017). Use of antibiotic-loaded nanofibers dentistry. Chaturvedi et al. have reported a clinical study of doxycy-
as wound dressing material serves several advantages due to porosity, cline-loaded PCL nanofibers for their use in patients with chronic per-
high surface area to volume ratio and biocompatibility. This area of iodontitis. Drug-coated nanofibers have shown to be having first order
drug delivery with nanofibers is very well explored and will be de- release kinetics up to a period of 11 days.7 patients (4 females and 3
scribed only in brief. Recently Li et al. have reported thermosensitive males) aged between 20 and 50 years were divided into 2 treatment
nanofibers loaded with ciprofloxacin as antibacterial wound dressing groups as one group having SRP (Scaling and Root Planing) alone and
materials. Ciprofloxacin (broad spectrum antibiotic) was loaded in another one with SRP + doxycycline nanofibers. Patients are reported
thermoresponsive electrospun fiber mats containing poly(di(ethylene to be evaluated continuously for probing depth, plaque index, and
glycol) methyl ether methacrylate). Post electrospinning drug was gingival index. Results revealed that treatment group with SRP
found to be in amorphous form. By virtue of their thermal sensitivity; + doxycycline nanofibers demonstrated better improvement in probing
fibers could promote the proliferation of fibroblasts, and by varying the depth, plaque index, and gingival index as compared to control group

164
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

(Chaturvedi et al., 2013). The same group has reported a clinical study control numerous properties associated with nanofibers. Drug/drug
of metronidazole loaded PCL nanofibers for their use in patients with nanoparticles can be easily incorporated by direct as well as an indirect
chronic periodontitis. 7 patients (4 females and 3 males) aged between method of fabrication which has surely widened the scope for the use of
20 and 50 years were divided into 2 treatment groups as one group nanofibers in the field of pharmaceuticals and medical devices. Though
having SRP (Scaling and Root Planing) alone and another one with SRP this technique has accomplished considerable success in the field of
+ metronidazole nanofibers. Patients are reported to be evaluated pharmaceuticals including antimicrobial applications, wound healing,
continuously for probing depth, plaque index, and gingival index. Re- cancer treatment, periodontics and tissue engineering still there are
sults revealed that treatment group with SRP + metronidazole nanofi- some challenges likes drug loading, the stability of therapeutic in-
bers demonstrated better improvement in probing depth, plaque index, gredient and control over undesired release profile, which needs to be
and gingival index as compared to control group (Chaturvedi et al., addressed. Advances in electrospinning technology with stringent
2012). control over various parameters can overcome these challenges.
Formulation scientists are expanding its use for diseases/disorders of
6. Patents related to drug containing nanofibers brain, ear, eye, lungs and joints still further investigations are required
in terms of preclinical and clinical trials of these drug-loaded nanofibers
Several patents have been granted in the area of drug delivery of for easy commercialization in the pharmaceutical field.
various pharmaceutically active substances in the form of polymeric
nanofibers. These are mainly based on the pattern and type of drug Conflict of interest
release needed along with the use of nanofibers as solubility enhance-
ment technique for various classes of compounds. Some patents are Authors report no conflicts of interest.
related to the particular form of drug and release profile accordingly as
amorphous or crystalline ones. Table 5 describes some of the patents Acknowledgement
related to drug delivery with polymeric nanofibers.
Shreya would like to thank Department of Pharmaceuticals,
7. Challenges in drug delivery with electrospun nanofibers Ministry of Chemicals and Fertilizers, Government of India for pro-
viding financial assistance in form of Ph.D fellowship.
Although nanofibers have shown great potential for drug delivery
applications; still there are certain challenges which need to be cir- References
cumvented before nanofibres obtained using electrospinning can be
used as drug delivery vehicle. Till date certain major factors, including Abadeer, N.S., Murphy, C.J., 2016. Recent progress in cancer thermal therapy using gold
drug loading, the stability of active ingredients, initial burst release, the nanoparticles. J. Phys. Chem. C 120, 4691–4716.
Adibkia, K., et al., 2016. Anti pneumococcal activity of azithromycin-Eudragit RS100
amount of residual solvents and industrial scale-up are crucial bottle- nano-formulations. Adv. Pharm. Bull. 6, 455.
necks, which needs to be overcome before bringing this technology into Alavarse, A.C., et al., 2017. Tetracycline hydrochloride-loaded electrospun nanofibers
mainstream drug delivery technologies. Though several reports cite mats based on PVA and chitosan for wound dressing. Mater. Sci. Eng. C 77, 271–281.
Angammana, C.J., Jayaram, S.H., 2016. Fundamentals of electrospinning and processing
greater extent of drug loading with certain polymers for extended re- technologies. Part. Sci. Technol. 34, 72–82.
lease, however uneven distribution of the drug in nanofibers often re- [19-09-2016]. Available from http://www.nanofibersolutions.com/products.html.
sults in initial burst effect, which still needs a lot of fine-tuning. [18-09-2016]. Available from http://www.nicast.com/index.aspx?id=2930.
Ball, C., et al., 2012. Drug-eluting fibers for HIV-1 inhibition and contraception. PLoS One
Polymers (either synthetic or natural) which are a common ingredient 7, e49792.
to provide the backbone for nanofibers and are reported to cause Baumgarten, P.K., 1971. Electrostatic spinning of acrylic microfibers. J. Colloid Interface
amorphization of crystalline drug candidates. The presence of an active Sci. 36, 71–79.
Begum, S.R., et al., 2012. Enhancement of dissolution rate of piroxicam by electrospin-
ingredient in suitable form whether amorphous or crystalline will have
ning technique. Adv. Nat. Sci. Nanosci. Nanotechnol. 3, 045012.
a major impact on stability, especially in the case of drugs, where Bhardwaj, N., Kundu, S.C., 2010. Electrospinning: a fascinating fiber fabrication tech-
regulatory norms are very stringent and require that polymorphic form nique. Biotechnol. Adv. 28, 325–347.
of the drug be specified at all crucial stages, including stability. Apart Bhattarai, N., Zhang, M., 2007. Controlled synthesis and structural stability of alginate-
based nanofibers. Nanotechnology 18, 455601.
from this, drug loading in polymer solution may adversely affect its Blakney, A.K., et al., 2014. Delivery of multipurpose prevention drug combinations from
viscosity and surface tension rendering it unsuitable for electrospinning electrospun nanofibers using composite microarchitectures. Int. J. Nanomedicine 9,
(Yu et al., 2009). 2967.
Bognitzki, M., et al., 2001a. Nanostructured fibers via electrospinning. Adv. Mater. 13,
Scale-up of nanofiber production by electrospinning technique is 70–72.
again questionable. Laboratory scale machines are generally dealing Bognitzki, M., et al., 2001b. Preparation of fibers with nanoscaled morphologies: elec-
with small volumes in milliliters and when it comes to liters in in- trospinning of polymer blends. Polym. Eng. Sci. 41, 982–989.
Boland, E.D., et al., 2004. Electrospinning collagen and elastin: preliminary vascular
dustrial scale the thing presents lots of challenges and there is room for tissue engineering. Front. Biosci. 9, e32.
further improvement of process and machinery (Persano et al., 2013). Caruso, R.A., et al., 2001. Titanium dioxide tubes from sol–gel coating of electrospun
In the case of textile industries other applications beyond drug delivery polymer fibers. Adv. Mater. 13, 1577–1579.
Cejkova, J., et al., 2016. Treatment of alkali-injured cornea by cyclosporine A-loaded
even large-scale production is possible by proper optimization of the
electrospun nanofibers–an alternative mode of therapy. Exp. Eye Res. 147, 128–137.
process but for drug delivery nanofiber commercialization by electro- Celebioglu, A., Uyar, T., 2011. Electrospinning of polymer-free nanofibers from cyclo-
spinning presents huge challenges for technology development scien- dextrin inclusion complexes. Langmuir 27, 6218–6226.
Chaturvedi, T.P., et al., 2012. Evaluation of metronidazole nanofibers in patients with
tists. Hence it is desirable that all these challenges must be well un-
chronic periodontitis: a clinical study. Int. J. Pharm. Invest. 2, 213.
derstood and controlled before nanofibres obtained using Chaturvedi, T., et al., 2013. Doxycycline poly e-caprolactone nanofibers in patients with
electrospinning could be used in mainstream drug delivery technologies chronic periodontitis–a clinical evaluation. J. Clin. Diagn. Res. 7, 2339.
offering desired drug release profile and adequate drug loading. Chen, R., et al., 2008. Fabrication of the silver/polypyrrole/polyacrylonitrile composite
nanofibrous mats. Mater. Lett. 62, 4031–4034.
Chen, Z., et al., 2016. Electrospun nanofibers for cancer diagnosis and therapy. Biomater.
8. Conclusion Sci. 4, 922–932.
Cho, K., et al., 2010. Synthesis and characterization of electrospun polymer nanofibers
incorporated with CdTe nanoparticles. Synth. Met. 160, 888–891.
Electrospun nanofibers have shown great potential for drug delivery Choi, J.S., et al., 2011. Coaxial electrospun nanofibers for treatment of diabetic ulcers
as well as control of release pattern. Proper selection of polymers/ with binary release of multiple growth factors. J. Mater. Chem. 21, 5258–5267.
polymeric blends/materials to provide the backbone and effective un- Chou, S.-F., Woodrow, K.A., 2017. Relationships between mechanical properties and drug
release from electrospun fibers of PCL and PLGA blends. J. Mech. Behav. Biomed.
derstanding of various CPPs and CMAs enable formulation scientist to

165
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

Mater. 65, 724–733. spinning and catalytic activity. Chem. Eng. Technol. 37, 1146–1154.
Chou, S.-F., et al., 2015. Current strategies for sustaining drug release from electrospun Kang, Y., et al., 2002. Manufacturing the cellulose web by using electro-spinning and in-
nanofibers. J. Control. Release 220, 584–591. vitro behaviour of oxidized cellulose web. J. Korean Fiber Soc. 39, 14–20.
Cui, W., et al., 2006. Investigation of drug release and matrix degradation of electrospun Kenawy, E.R., Abdel-Fattah, Y.R., 2002. Antimicrobial properties of modified and elec-
poly (DL-lactide) fibers with paracetanol inoculation. Biomacromolecules 7, trospun poly (vinyl phenol). Macromol. Biosci. 2, 261–266.
1623–1629. Kessick, R., et al., 2004. The use of AC potentials in electrospraying and electrospinning
Davis, M.E., et al., 2005. Biodegradable Drug-polymer Delivery System. US 2005/ processes. Polymer 45, 2981–2984.
0276841A1. United States. Konwarh, R., et al., 2013. Electrospun cellulose acetate nanofibers: the present status and
Demir, M.M., et al., 2004. Palladium nanoparticles by electrospinning from poly (acry- gamut of biotechnological applications. Biotechnol. Adv. 31, 421–437.
lonitrile-co-acrylic acid)-PdCl2 solutions. Relations between preparation conditions, Kowalewski, T., et al., 2005. Experiments and modelling of electrospinning process. Tech.
particle size, and catalytic activity. Macromolecules 37, 1787–1792. Sci. 53, 385–394.
Demirci, S., et al., 2014. pH-responsive nanofibers with controlled drug release proper- Laurencin, C.T., et al., 2007. Polymeric Nanofibers for Tissue Engineering and Drug
ties. Polym. Chem. 5, 2050–2056. Delivery. United States.
Doshi, J., Reneker, D.H., 1993. Electrospinning process and applications of electrospun Li, W.J., et al., 2002. Electrospun nanofibrous structure: a novel scaffold for tissue en-
fibers. In: Industry Applications Society Annual Meeting, 1993. Conference Record of gineering. J. Biomed. Mater. Res. 60, 613–621.
the 1993 IEEE. IEEEpp. 1698–1703. Li, D., et al., 2004. Photocatalytic deposition of gold nanoparticles on electrospun na-
Dubrow, R., Daniels, R.H., 2009. Nanofiber Surfaces for use in Enhanced Surface Area nofibers of titania. Chem. Phys. Lett. 394, 387–391.
Applications. United States. Li, Y.F., et al., 2016. Light responsive hybrid nanofibres for on-demand therapeutic drug
Dubrow, R.S., et al., 2005. Medical Device Applications of Nanostructured Surfaces. US and cell delivery. J. Tissue Eng. Regen. Med.
2005/0038498A1. United States. Li, H., et al., 2017. Thermosensitive nanofibers loaded with ciprofloxacin as antibacterial
Dzenis, Y., Wen, Y., 2001. Continuous carbon nanofibers for nanofiber composites. In: wound dressing materials. Int. J. Pharm. 517, 135–147.
MRS Proceedings. Cambridge Univ Press, pp. U5. 4.1. Lin, T.-C., et al., 2012. In vitro feasibility study of the use of a magnetic electrospun
Eltayeb, M., et al., 2016. Electrosprayed nanoparticle delivery system for controlled re- chitosan nanofiber composite for hyperthermia treatment of tumor cells. Acta
lease. Mat. Sci. Eng. C-Bio. S 66, 138–146. Biomater. 8, 2704–2711.
Fang, X., Reneker, D., 1997. DNA fibers by electrospinning. J. Macromol. Sci. Part B Phys. Liu, K.-S., et al., 2015a. Sustained release of vancomycin from novel biodegradable na-
36, 169–173. nofiber-loaded vascular prosthetic grafts: in vitro and in vivo study. Int. J.
Frenot, A., Chronakis, I.S., 2003. Polymer nanofibers assembled by electrospinning. Curr. Nanomedicine 10, 885.
Opin. Colloid. In. 8, 64–75. Liu, W., et al., 2015b. Electrospinning of grooved polystyrene fibers: effect of solvent
Frenot, A., et al., 2007. Electrospinning of cellulose-based nanofibers. J. Appl. Polym. Sci. systems. Nanoscale Res. Lett. 10, 1–10.
103, 1473–1482. Liu, L., et al., 2016. Controlled release from thermo-sensitive PNVCL-co-MAA electrospun
Garg, T., et al., 2014. Development and characterization of nano-fiber patch for the nanofibers: the effects of hydrophilicity/hydrophobicity of a drug. Mater. Sci. Eng. C
treatment of glaucoma. Eur. J. Pharm. Sci. 53, 10–16. 67, 581–589.
Gencturk, A., et al., 2016. Polyurethane/hydroxypropyl cellulose electrospun nanofiber Long, H.C., et al., 2009. Antibacterial Nano Fiber Material and Preparation Method
mats as potential transdermal drug delivery system: characterization studies and in Thereof. China.
vitro assays. Artif. Cell Nanomed. Biotechnol. 1–10. Luo, C., et al., 2012. Electrospinning versus fibre production methods: from specifics to
Goonoo, N., et al., 2014. Drug loading and release from electrospun biodegradable na- technological convergence. Chem. Soc. Rev. 41, 4708–4735.
nofibers. J. Biomed. Nanotechnol. 10, 2173–2199. Mahalingam, S., Edirisinghe, M., 2013. Forming of polymer nanofibers by a pressurised
Haider, A., et al., 2015. A comprehensive review summarizing the effect of electrospin- gyration process. Macromol. Rapid Commun. 34, 1134–1139.
ning parameters and potential applications of nanofibers in biomedical and bio- Mahanta, N., Valiyaveettil, S., 2011. Surface modified electrospun poly (vinyl alcohol)
technology. Arab. J. Chem. membranes for extracting nanoparticles from water. Nano 3, 4625–4631.
He, D., et al., 2009. Large-scale synthesis of flexible free-standing SERS substrates with Maity, S., et al., 2011. Embedded metal nanoparticles as localized heat sources: an al-
high sensitivity: electrospun PVA nanofibers embedded with controlled alignment of ternative processing approach for complex polymeric materials. Polymer 52,
silver nanoparticles. ACS Nano 3, 3993–4002. 1674–1685.
Hedenquist, J.W., Lowenstern, J.B., 1994. The role of magmas in the formation of hy- Matthews, J.A., et al., 2002. Electrospinning of collagen nanofibers. Biomacromolecules
drothermal ore deposits. Nature 370, 519–527. 3, 232–238.
Hrib, J., et al., 2015. Nanofibers for drug delivery–incorporation and release of model McKeon-Fischer, K., Freeman, J., 2011. Characterization of electrospun poly (L-lactide)
molecules, influence of molecular weight and polymer structure. Beilstein J. and gold nanoparticle composite scaffolds for skeletal muscle tissue engineering. J.
Nanotechnol. 6, 1939–1945. Tissue Eng. Regen. Med. 5, 560–568.
Hsu, C.-Y., Liu, Y.-L., 2010. Rhodamine B-anchored silica nanoparticles displaying white- Megelski, S., et al., 2002. Micro-and nanostructured surface morphology on electrospun
light photoluminescence and their uses in preparations of photoluminescent poly- polymer fibers. Macromolecules 35, 8456–8466.
meric films and nanofibers. J. Colloid Interface Sci. 350, 75–82. Meng, Z., et al., 2011. Preparation and characterization of electrospun PLGA/gelatin
Hsu, P.-C., et al., 2012. Passivation coating on electrospun copper nanofibers for stable nanofibers as a potential drug delivery system. Colloids Surf. B 84, 97–102.
transparent electrodes. ACS Nano 6, 5150–5156. Mengistu Lemma, S., et al., 2016. Preparation of pure and stable chitosan nanofibers by
Hsu, P.-C., et al., 2013. Performance enhancement of metal nanowire transparent con- electrospinning in the presence of poly (ethylene oxide). Int. J. Mol. Sci. 17, 1790.
ducting electrodes by mesoscale metal wires. Nat. Commun. 4. Min, B.-M., et al., 2004. Chitin and chitosan nanofibers: electrospinning of chitin and
Hsu, K.-H., et al., 2016. Hybrid electrospun polycaprolactone mats consisting of nanofi- deacetylation of chitin nanofibers. Polymer 45, 7137–7142.
bers and microbeads for extended release of dexamethasone. Pharm. Res. 33, Modgill, V., et al., 2015. Transmucosal delivery of linagliptin for the treatment of type-2
1509–1516. diabetes mellitus by ultra-thin nanofibers. Curr. Drug Deliv. 12, 323–332.
Huan, S., et al., 2015. Effect of experimental parameters on morphological, mechanical Natu, M.V., et al., 2010. Effects of drug solubility, state and loading on controlled release
and hydrophobic properties of electrospun polystyrene fibers. Materials 8, in bicomponent electrospun fibers. Int. J. Pharm. 397, 50–58.
2718–2734. Nayak, R., et al., 2011. Recent advances in nanofibre fabrication techniques. Text. Res. J
Huang, Z.-M., et al., 2003. A review on polymer nanofibers by electrospinning and their 0040517511424524.
applications in nanocomposites. Compos. Sci. Technol. 63, 2223–2253. Neamnark, A., et al., 2006. Electrospinning of hexanoyl chitosan. Carbohydr. Polym. 66,
Huang, C.-H., et al., 2014. Preparation of a thermo-and pH-sensitive nanofibrous scaffold 298–305.
with embedded chitosan-based nanoparticles and its evaluation as a drug carrier. Noh, H.K., et al., 2006. Electrospinning of chitin nanofibers: degradation behavior and
Cellulose 21, 2497–2509. cellular response to normal human keratinocytes and fibroblasts. Biomaterials 27,
Husain, O., et al., 2016. Investigating the particle to fibre transition threshold during 3934–3944.
electrohydrodynamic atomization of a polymer solution. Mat. Sci. Eng. C-Bio. S 65, Ohkawa, K., et al., 2004. Electrospinning of chitosan. Macromol. Rapid Commun. 25,
240–250. 1600–1605.
Ignatious, F., Baldoni, J.M., 2003. Electrospun Pharmaceutical Compositions. US 2003/ Paaver, U., et al., 2015. Electrospun nanofibers as a potential controlled-release solid
0017208 A1. United States. dispersion system for poorly water-soluble drugs. Int. J. Pharm. 479, 252–260.
Ignatious, F., Sun, L., 2006. Electrospunamorphous Pharmaceutical Compositions. US Pankajakshan, D., et al., 2016. Triple antibiotic polymer nanofibers for intracanal drug
2006/0013869 A1. United States. delivery: effects on dual species biofilm and cell function. J. Endod. 42, 1490–1495.
Ikegame, M., et al., 2003. Template synthesis of polypyrrole nanofibers insulated within Parhizkar, M., et al., 2016a. Electrohydrodynamic encapsulation of cisplatin in poly
one-dimensional silicate channels: hexagonal versus lamellar for recombination of (lactic-co-glycolic acid) nanoparticles for controlled drug delivery. Nanomedicine 12,
polarons into bipolarons. Angew. Chem. Int. Ed. 42, 2154–2157. 1919–1929.
Jaeger, R., et al., 1998. Electrospinning of ultra-thin polymer fibers, macromolecular Parhizkar, M., et al., 2016b. Electrohydrodynamic encapsulation of cisplatin in poly
symposia. Wiley Online Library 141–150. (lactic-co-glycolic acid) nanoparticles for controlled drug delivery. Nanomed.
Jang, C.H., et al., 2015. Antibacterial effect of electrospun polycaprolactone/polyethylene Nanotech. Biol. Med. 12, 1919–1929.
oxide/vancomycin nanofiber mat for prevention of periprosthetic infection and Park, J.-Y., Lee, I.-H., 2011. Controlled release of ketoprofen from electrospun porous
biofilm formation. Int. J. Pediatr. Otorhinolaryngol. 79, 1299–1305. polylactic acid (PLA) nanofibers. J. Polym. Res. 18, 1287–1291.
Jiang, H., et al., 2004. Optimization and characterization of dextran membranes prepared Park, W.H., et al., 2004. Effect of chitosan on morphology and conformation of electro-
by electrospinning. Biomacromolecules 5, 326–333. spun silk fibroin nanofibers. Polymer 45, 7151–7157.
Jin, Y., et al., 2009. Fabrication of necklace-like structures via electrospinning. Langmuir Park, K.E., et al., 2006a. Biomimetic nanofibrous scaffolds: preparation and character-
26, 1186–1190. ization of chitin/silk fibroin blend nanofibers. Int. J. Biol. Macromol. 38, 165–173.
Kaiser, S., et al., 2014. Porous perovskite fibers–preparation by wet phase inversion Park, K.E., et al., 2006b. Biomimetic nanofibrous scaffolds: preparation and

166
S. Thakkar, M. Misra European Journal of Pharmaceutical Sciences 107 (2017) 148–167

characterization of PGA/chitin blend nanofibers. Biomacromolecules 7, 635–643. Sulejczak, D., et al., 2014. Electrospun nanofiber mat as a protector against the con-
Park, J.-Y., et al., 2015. Fabrication and characterization of antimicrobial ethyl cellulose sequences of brain injury. Folia Neuropathol. 52, 56–69.
nanofibers using electrospinning techniques. J. Nanosci. Nanotechnol. 15, Švachová, V., et al., 2016. Novel electrospun gelatin/oxycellulose nanofibers as a suitable
5672–5675. platform for lung disease modeling. Mater. Sci. Eng. C 67, 493–501.
Persano, L., et al., 2013. Industrial upscaling of electrospinning and applications of Taghavi, S., Larson, R., 2014. Regularized thin-fiber model for nanofiber formation by
polymer nanofibers: a review. Macromol. Mater. Eng. 298, 504–520. centrifugal spinning. Phys. Rev. E 89, 023011.
Pina, M.F., et al., 2017. The generation of compartmentalized nanoparticles containing Tan, G., et al., 2016. Synthesis and characterization of silver nanoparticles integrated in
siRNA and cisplatin using a multi-needle electrohydrodynamic strategy. Nano 9, polyvinyl alcohol nanofibers for bionanotechnological applications. Turk. J. Biol. 40,
5975–5985. 643–651.
Polat, Y., et al., 2016. Solution blowing of thermoplastic polyurethane nanofibers: a facile Teo, W.-E., et al., 2011. Technological advances in electrospinning of nanofibers. Sci.
method to produce flexible porous materials. J. Appl. Polym. Sci. 133. Technol. Adv. Mater. 12, 013002.
Pourdeyhimi, B., et al., 2009. Fiber-based Nano Drug Delivery Systems (NDDS). US Theron, S., et al., 2004. Experimental investigation of the governing parameters in the
7.491.407 B2. United States. electrospinning of polymer solutions. Polymer 45, 2017–2030.
Qian Zhou, L.X., Yan, Su, Xiumei, Mo, 2009. Method for Producing Shell-core Structure Toncheva, A., et al., 2012. Antibacterial fluoroquinolone antibiotic-containing fibrous
Medicament Nanofibre With Coaxial Electrostatic Spinning Technology. China. materials from poly (l-lactide-co-d, l-lactide) prepared by electrospinning. Eur. J.
Qiu, K., et al., 2013. Doxorubicin-loaded electrospun poly (L-lactic acid)/mesoporous Pharm. Sci. 47, 642–651.
silica nanoparticles composite nanofibers for potential postsurgical cancer treatment. Torres, B., 2001. Ultrafine fibers of polystyrene dissolved in tetrahydrofuran prepared
J. Mater. Chem. B 1, 4601–4611. using the electrospinning method. In: Proceeding of the National Conference on
Reardon, P.J., et al., 2017. Electrohydrodynamic fabrication of core–shell PLGA nano- Undergraduate Research, pp. 15–17.
particles with controlled release of cisplatin for enhanced cancer treatment. Int. J. Tort, S., Acartürk, F., 2016. Preparation and characterization of electrospun nanofibers
Nanomedicine 12, 3913. containing glutamine. Carbohydr. Polym. 152, 802–814.
Rujitanaroj, P.-O., et al., 2008. Wound-dressing materials with antibacterial activity from Vellayappan, M., et al., 2016. Electrospinning applications from diagnosis to treatment of
electrospun gelatin fiber mats containing silver nanoparticles. Polymer 49, diabetes. RSC Adv. 6, 83638–83655.
4723–4732. Wang, J., Nain, A.S., 2014. Suspended micro/nanofiber hierarchical biological scaffolds
Santos, V.A.d., et al., 2014. Antifungal effect of electrospun nanofibers containing ce- fabricated using non-electrospinning STEP technique. Langmuir 30, 13641–13649.
tylpyridinium chloride against Candida albicans. Braz. Oral Res. 28, 1–6. Wang, C., et al., 2015a. Enhanced bioavailability and anticancer effect of curcumin-
Schiffman, J.D., Schauer, C.L., 2008. A review: electrospinning of biopolymer nanofibers loaded electrospun nanofiber: in vitro and in vivo study. Nanoscale Res. Lett. 10, 1.
and their applications. Polym. Rev. 48, 317–352. Wang, J., et al., 2015b. Heparin and vascular endothelial growth factor loaded poly (L-
Schiffman, J.D., et al., 2009. Chitin and chitosan: transformations due to the electro- lactide-co-caprolactone) nanofiber covered stent-graft for aneurysm treatment. J.
spinning process. Polym. Eng. Sci. 49, 1918–1928. Biomed. Nanotechnol. 11, 1947–1960.
Schreuder-Gibson, H., et al., 2002. Protective textile materials based on electrospun na- Wang, X., et al., 2016. Enhancing the electrospinnability of low molecular weight poly-
nofibers. J. Adv. Mater. 34, 44–55. mers using small effective cross-linkers. Macromolecules 49, 891–899.
Shaddock, E.J., 2016. How and when to use common biomarkers in community-acquired Wnek, G.E., et al., 2003. Electrospinning of nanofiber fibrinogen structures. Nano Lett. 3,
pneumonia. Pneumonia 8, 17. 213–216.
Shao, H., et al., 2015. Effect of electrospinning parameters and polymer concentrations on Wu, X., et al., 2005. Effect of solvent on morphology of electrospinning ethyl cellulose
mechanical-to-electrical energy conversion of randomly-oriented electrospun poly fibers. J. Appl. Polym. Sci. 97, 1292–1297.
(vinylidene fluoride) nanofiber mats. RSC Adv. 5, 14345–14350. Wu, H., et al., 2013. A transparent electrode based on a metal nanotrough network. Nat.
Shastri, V.P., Sy, J.C., 2008. Modulation of Drug Release Rate From Electrospun Fibers. Nanotechnol. 8, 421–425.
US 2008/0220054A1. United States. Xie, J., Hsieh, Y.-L., 2003. Ultra-high surface fibrous membranes from electrospinning of
Shi, W., et al., 2013. Hydrothermal synthetic strategies of inorganic semiconducting na- natural proteins: casein and lipase enzyme. J. Mater. Sci. 38, 2125–2133.
nostructures. Chem. Soc. Rev. 42, 5714–5743. Xie, J., et al., 2015. Electrohydrodynamic atomization: a two-decade effort to produce
Shin, Y., et al., 2001. Experimental characterization of electrospinning: the electrically and process micro-/nanoparticulate materials. Chem. Eng. Sci. 125, 32–57.
forced jet and instabilities. Polymer 42, 09955–09967. Xu, L., et al., 2015. Semi-interpenetrating network (sIPN) co-electrospun gelatin/insulin
Shukla, S., et al., 2005. Electrospinning of hydroxypropyl cellulose fibers and their ap- fiber formulation for transbuccal insulin delivery. Pharm. Res. 32, 275–285.
plication in synthesis of nano and submicron tin oxide fibers. Polymer 46, Yao, C., et al., 2007. Electrospinning and crosslinking of zein nanofiber mats. J. Appl.
12130–12145. Polym. Sci. 103, 380–385.
Siafaka, P.I., et al., 2016. Novel electrospun nanofibrous matrices prepared from poly Yu, D.-G., et al., 2009. Electrospun nanofiber-based drug delivery systems. Health 1, 67.
(lactic acid)/poly (butylene adipate) blends for controlled release formulations of an Zampetti, E., et al., 2011. Effects of temperature and humidity on electrospun conductive
anti-rheumatoid agent. Eur. J. Pharm. Sci. 88, 12–25. nanofibers based on polyaniline blends. J. Nanopart. Res. 13, 6193–6200.
Sill, T.J., von Recum, H.A., 2008. Electrospinning: applications in drug delivery and tissue Zhang, C.-L., Yu, S.-H., 2014. Nanoparticles meet electrospinning: recent advances and
engineering. Biomaterials 29, 1989–2006. future prospects. Chem. Soc. Rev. 43, 4423–4448.
Singh, B., et al., 2015. Development, optimization, and characterization of polymeric Zhao, Y., et al., 2006. TiO2 micro/nano-composite structured electrodes for quasi-solid-
electrospun nanofiber: a new attempt in sublingual delivery of nicorandil for the state dye-sensitized solar cells. Nanotechnology 17, 2090.
management of angina pectoris. Artif Cell Nanomed. Biotechnol. 1–10. Zong, S., et al., 2015. The use of cisplatin-loaded mucoadhesive nanofibers for local
Stojanovska, E., et al., 2016. A review on non-electro nanofibre spinning techniques. RSC chemotherapy of cervical cancers in mice. Eur. J. Pharm. Biopharm. 93, 127–135.
Adv. 6, 83783–83801.

167

You might also like