You are on page 1of 16

European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

Contents lists available at ScienceDirect

European Journal of Pharmaceutics and Biopharmaceutics


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

Research paper

Poly (vinyl alcohol) microneedles: Fabrication, characterization, and T


application for transdermal drug delivery of doxorubicin
Hiep X. Nguyena, Behnam Dasht Bozorga, Yujin Kima, Alena Wieberb, Gudrun Birkb,

Dieter Lubdab, Ajay K. Bangaa,
a
Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, United States
b
MilliporeSigma a Business of Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany

A R T I C LE I N FO A B S T R A C T

Keywords: Poly (vinyl alcohol) microneedles were fabricated, characterized, and applied to enhance in vitro transdermal
Fabrication delivery of doxorubicin. The microneedles were fabricated using the micromolding technique with the drug load
Characterization in different locations within the needle array. The polymer solution was assessed for rheological properties, drug
Doxorubicin dissolution, and chemical structurestudies. Microneedles (unloaded) and drug-loaded microneedles were char-
Microneedles
acterized by optical microscopy, fluorescent microscopy, scanning electron microscopy, and drug release ki-
Polyvinyl alcohol
Transdermal delivery
netics. Successful microporation of dermatomed human cadaver skin was demonstrated by dye binding, pore
uniformity, histology, confocal laser microscopy, and skin integrity studies. The microneedles-mediated trans-
dermal delivery of doxorubicin was investigated using vertical Franz diffusion cells.
The fabricated microneedles were sharp, strong, and uniform. In vitro permeation studies showed that the
microneedle-treated skin (4351.55 ± 560.87 ng/sq.cm) provided a significantly greater drug permeability than
the untreated group (0.00 ± 0.00 ng/sq.cm, n = 4, p < 0.01). The drug location within the needle array was
found to affect the drug release profile as well as its permeation into and across human skin.
Skin microporation achieved by poly (vinyl alcohol) microneedles was found to enhance transdermal delivery
of doxorubicin in vitro.

1. Introduction needs to be released from the formulation, dissolved in a small volume


of interstitial fluid, and diffused into skin layers [6].
Transdermal delivery helps drugs to bypass hepatic first-pass me- To address these drawbacks, dissolving, biodegradable, and drug-
tabolism, avoid intestinal degradation, and be delivered at controlled loaded microneedles have been developed using water-soluble poly-
rates [1]. However, the application of transdermal delivery has been mers [7]. Dissolving microneedles can be used in a single-step process
limited to only small, potent, and moderately lipophilic molecules due of insertion into the skin. Once the needles are embedded into the skin,
to the lipophilic, inert, and tight structure of the stratum corneum layer the needles dissolve rapidly and release the drug load into skin layers
of skin [1–3]. Skin permeability has been enhanced by using physical which result in a rapid onset of drug delivery. Therefore, dissolvable
enhancement techniques such as ultrasound, skin abrasion [4], and microneedles have been suggested as a promising technology to en-
thermal ablation [5]. In general, these techniques are employed to hance transdermal drug delivery. In addition, dissolving microneedles
disrupt the structure of skin layers: stratum corneum, epidermis, and are minimally invasive, safely disposable, and clinically efficient [1].
dermis to create a pathway for drug penetration. However, these These needles could be easily manufactured with an appropriate drug
methods require a two-step process: skin treatment (different physical load and low cost [8].
forces are applied to disturb the skin barrier function) and formulation Microneedles penetrate the stratum corneum, epidermis, and su-
application (drug formulation is applied onto the treated area of skin). perficial dermis; create microchannels in skin; rapidly dissolve in the
This has some drawbacks when drug formulation is applied on areas interstitial fluid of skin; release the encapsulated drug load within
other than the treated site or the drug delivery is delayed since the drug minutes; and are able to administer a broad range of therapeutic agents

Abbreviations: PPI, Pore Permeability Index; MN, Microneedles; TEWL, Transepidermal Water Loss; DOX, Doxorubicin; SEM, Scanning Electron Microscopy; PBS, Phosphate Buffered
Saline; SD, Standard Deviation; HPLC, High Performance Liquid Chromatography; PVA, Poly (Vinyl Alcohol); PDMS, Poly (dimethylsiloxane); DI water, Deionized water; FTIR, Fourier
Transform Infrared Spectroscopy

Corresponding author.
E-mail address: Banga_ak@mercer.edu (A.K. Banga).

https://doi.org/10.1016/j.ejpb.2018.05.017
Received 22 January 2018; Received in revised form 4 April 2018; Accepted 15 May 2018
Available online 22 May 2018
0939-6411/ © 2018 Elsevier B.V. All rights reserved.
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

(small molecules, macromolecules, and biologics) into the skin layers scanning electron microscopy, mechanical properties, drug release from
for either local or systemic delivery [7,9,10]. the needles, and in-situ dissolution; characterization of microchannels:
Microneedle fabrication technology allows to produce microneedles skin integrity measurement, dye binding, pore uniformity, histology,
with higher amount of drug to increase the applied dose and to load the skin penetration of PVA microneedles, and confocal laser microscopy;
drug at the tip of microneedles to minimize the waste of drug which can and in vitro permeation study to measure the drug delivery into and
remain in the needles or on the skin surface after removal of the needles across dermatomed human cadaver skin. For the first time, 100% PVA
[1,7]. When the drug load is localized at the needle tip, an incomplete was used to fabricate dissolving microneedles with sufficient sharpness
insertion of the microneedles (due to the user-to-user variation of insertion and strength. A simple fabrication process using centrifugation was
force or skin indentation due to its viscoelastic property, or the physical employed to make the needles. In this experiment, DOX was loaded in
properties of the needles) might not significantly affect the drug delivery if different areas in the array to study the effect of drug location on drug
the tip of the needles penetrates the skin layers. Dissolving microneedles release and delivery profile. This study can serve as a basis for future in
have been fabricated using a wide range of polymers, limited to those that vivo and clinical studies on PVA microneedles and transdermal delivery
are mechanically robust for skin insertion, biocompatible, capable of dis- of DOX.
solution in the skin, and provide a stable matrix for encapsulated drugs
[6]. Polyvinylalcohol (PVA) has been used to fabricate coated micro- 2. Materials and methods
needles [11], mold to fabricate microneedles [12,13] and dissolving mi-
croneedles [1,6,14,15]. PVA, a water-soluble biodegradable polymer, has 2.1. Materials
been shown to be promising for microneedle fabrication [1,6]. PVA mi-
croneedles appeared superior than other dissolving microneedles such as EMPROVE® exp Ph Eur, USP, JPE polyvinyl alcohol 26–88
trehalose, raffinose, polyvinylpyrrolidone, carboxymethylcellulose, hy- (Viscosity 40 g/L in water: 22.1–29.9 mPa-s), 4–88 (Viscosity 40 g/L in
droxypropylmethylcellulose, and sodium alginate with respect to poration water: 3.4–4.6 mPa-s) were kindly provided by Millipore Sigma, a
of stratum corneum and the epidermis [14]. Interestingly, PVA has been Business of Merck KGaA, Darmstadt, Germany. Doxorubicin hydro-
used in combination with PVP K17 (3:1) [1], PVP K17 (ratio 1:1) [6], chloride was obtained from Medchemexpress (NJ, USA). Fluoresoft®
sucrose (mass ratio 1:1) [6], trehalose [14], dextran and carbox- (0.35%) was from Holles Laboratories Inc. (Cohasset, MA, USA), me-
ymethylcellulose [15] to fabricate drug-loaded dissolving microneedles. thylene blue dye from Eastman Kodak Co (Rochester, NY, USA), and
Dissolving microneedles appeared suitable for drugs with a low dose (< 1 Phosphate buffered saline (0.1 M PBS pH 7.4) from Fisher Scientific
mg) and low dermal irritation and sensitization potential [6,7]. (Fair Lawn, NJ, USA) while all the other chemicals were obtained from
Doxorubicin (DOX) (molecular weight 543.52 g/mol) is a non-se- Sigma-Aldrich (St. Louis, MO, USA). Dermatomed human cadaver skin
lective class I anthracycline antibiotic compound [16], which is among (thickness 0.45 ± 0.11 mm, n = 28) was obtained from New York Fire
the most widely used Food and Drug Administration-approved anti- Fighter (NY, USA). Stainless steel microneedles (master structures
tumor drugs to treat various types of cancers especially solid tumors consisted of 10x10 pyramidal-shaped 500 µm-long needles, the needle-
such as breast cancer, neuroblastoma, and ovarian cancer [17,18]. DOX to-needle distance of 500 µm, and 150 µm × 150 µm square base) were
has been proved to restrain rapidly dividing cells and slow the disease procured from Micropoint Technologies Pte, Ltd. (CleanTech Loop,
progression [16]. The common route of administration of DOX is a Singapore) [30].
venous or arterial injection [18]. However, due to non-targeted de-
livery, the systemic administration of a therapeutically effective dose of 2.2. Fabrication of PVA microneedles
DOX caused toxic adverse effects such as non-selective cardiotoxicity,
myelosuppression, and mucositis [19,20]. Topical chemotherapy could Unloaded and DOX-loaded PVA microneedles were fabricated using
be an effective alternative to reduce the systemic toxicity of DOX the micromoulding technique. PVA (26–88, 4 g) was dissolved in 8 mL
[20,21]. pre-heated deionized (DI) water at 90 °C for 24 h. Unloaded micro-
DOX has a favorable pharmacokinetic profile for transdermal route needles were later used in Poke&Solution group while DOX-en-
such as short half-life, low bioavailability, high dose, and dose-depen- capsulated microneedles were fabricated with the drug load in different
dent toxicity. However, skin permeability of DOX is limited by the su- locations within the needle array. For unloaded microneedles, the PVA
perficial lipophilic stratum corneum layer due to its physical properties solution (300 µL) was placed on a poly (dimethylsiloxane) (PDMS)
such as its hydrophilicity, charge, and high molecular weight [21]. mold, then centrifuged at 4000 rpm and 40 °C for 20 min (Eppendorf
Furthermore, the anthracycline structure of DOX interacts with anionic Centrifuge 5810 R, Eppendorf AG, Hamburg, Germany) to pull the so-
lipids inside the stratum corneum to prevent it from diffusing into the lution into the mold cavities and eliminate any entrapped air bubble.
underlying skin layers [21]. Owing to these reasons, the skin permea- For DOX-loaded microneedles G1 (Fig. 1), DOX solution in ethanol (100
tion of DOX needs to be improved to increase the tumor bioavailability. µL, 4 mg/mL) was filled into the mold cavities and dried under a va-
An enhanced delivery of DOX has been achieved by using monoolein- cuum application at 25 °C for 30 min. Five adhesive tapes (3M) and one
containing propylene glycol formulations [21], nanoparticles [17,22], water-soaked cotton swab were used to remove the drug on the mold
liposomes [23,24], hydrogels [25], iontophoresis with chitosan gel surface with minimal interference in the drug inside the mold cavities.
[20], and combination of iontophoresis and liposomes [26]. DOX has For DOX-loaded microneedles G2, PDMS mold was covered with DOX
also been loaded in near-infrared-light-triggered biodegradable poly- solution in ethanol (200 µL, 4 mg/mL) and dried in a vacuum oven at
caprolactone microneedles [27], encapsulated into poly (lactic-co-gly- 25 °C for 30 min to have the drug deposited on both cavities and surface
colic) acid nanoparticles and coated on metal microneedles for intra- of the mold. The drug-contained mold (G1 and G2) was then covered
tumoral delivery [28]. The fluorescent property (the excitation wave- with PVA solution (300 µL) and centrifuged at 4000 rpm and 40 °C for
length of 490 nm and the emission wavelength of 560–590 nm) of DOX 20 min. For drug-loaded microneedles G3 and G4 (Fig. 1), DOX was
has been used to detect and track its permeation, distribution, and dissolved in the above-prepared PVA solution (4 mg/mL). The drug-
transport pathway through the skin [26,27,29]. contained PVA solution was filled into the mold cavities under a cen-
The aim of this study was to fabricate, characterize, and apply PVA trifugation at 4000 rpm for 30 min at room temperature. For G3, re-
microneedles to enhance in vitro transdermal delivery of DOX. sidual polymer solution on the mold surface was discarded using a
Comprehensive experiments were performed for characterization of water-dipped spatula and water-soaked cotton swab, leaving the drug-
PVA formulation: rheological properties, drug dissolution in PVA ma- loaded PVA solution in the cavities only while the backing layer was
trix, Fourier Transform Infrared spectroscopy (FTIR), and stability unloaded PVA solution. For G4, the residual drug-contained PVA so-
studies; characterization of PVA microneedles: fluorescent microscopy, lution was kept in place to fabricate microneedles with the drug load in

89
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

Fig. 1. PVA microneedles (A) G1 DOX in the channels of PDMS mold, (B) G2 DOX on the surface and in the channels of PDMS mold, (C) G3 DOX in PVA solution in
the channels of PDMS mold, (D) G4 DOX in microneedles and base of the array.

both microneedles and backing layer. The microneedles (G1, G2, G3, For microneedles G1 and G2, the amount of drug removed by tapes,
and G4) were dried in an oven at 60 °C for three days, manually sepa- remained on the mold post-fabrication, and lost due to the evaporation
rated from the mold, and stored in a desiccator until used. of ethanol were included into the calculation of the drug thermal sta-
bility. For microneedles G4, the percentage of drug degradation was
2.3. Preparation of skin samples contributed from both dissolution in PVA solution (90 °C for one day)
and drying step (60 °C for three days).
Dermatomed human cadaver skin was prepared and treated
manually by PVA microneedles for two minutes using a thumb [9,31]. 2.5. Characterization of PVA microneedles
For the unloaded microneedles, the needles were removed after two-
min insertion and before the application of drug solution while the 2.5.1. Optical microscopy
drug-encapsulated microneedles were left in the skin for 24 h after the After fabrication, array base, unloaded, and DOX-loaded PVA mi-
insertion. Then, the treated skin was used in characterization and in croneedles (G1, G2, G3, and G4) were visualized under a ProScope HR
vitro permeation studies. digital USB microscope (Bodelin Technologies, OR, USA) to investigate
the distribution pattern of microneedles on the array.
2.4. Characterization of formulation
2.5.2. Fluorescent microscopy studies
2.4.1. Rheological properties of PVA matrix
Due to the intrinsic fluorescent properties, DOX could be detected
A rheometer (Rheoplus/32 V3.62. Anton Paar Germany GmbH,
under an Olympus CX41 phase contrast and polarized light microscope
Germany) was used to analyze the rheological properties of PVA solu-
in tandem with 510–560 nm excitation, 590 nm barrier filters, and
tion including an oscillatory amplitude sweep (angular frequency
cellSens standard software 1.3 (Tokyo, Japan). Untreated, 1 h-treated,
10 rad/s and strain 0.001–100%), oscillatory frequency sweep (shear
and 24 h-treated PVA microneedles were visualized to study the drug
strain 0.05% and angular frequency 0.1–100 rad/s), viscosity study
distribution within the array and the deformation of the needles with
(different temperatures 25, 40, and 60 °C), oscillatory 3-step thixotropy
time.
test (the first and third intervals: strain 1%, angular frequency of
10 s−1; the second interval: shear rate 3000 s−1) [32].
2.5.3. Scanning electron microscopy studies
PVA microneedles were coated with gold and visualized under
2.4.2. DOX dissolution in PVA matrix
Phenom™ SEM system to measure the needle dimensions before and
DOX-dissolved PVA solution (4 mg/mL) was spread thinly on a
after the skin insertion [31,33].
microscopic slide and observed under polarized light (Leica DM750,
over the entire area for presence of DOX crystals) to study the drug
dissolution in the polymer matrix. 2.5.4. Mechanical properties of PVA microneedles
Layers of parafilm (Parafilm “M” Laboratory film, Neenah, WI, USA)
2.4.3. Fourier transform infrared spectrophotometer studies was treated by PVA microneedles and observed under an optical mi-
The presence of PVA and DOX in the polymer matrix and fabricated croscope to study the mechanical properties of the needles [31]. The
microneedles was validated using a Fourier Transform Infrared mechanical uniformity of PVA microneedles was estimated from the
Spectrophotometer (FTIR) equipped with LabSolution IR software dimensions of the pores in the first layer (n = 20) and the number of
(IRAffinity-1S, Shimadzu Corporation, Kyoto, Japan). The samples were pores in the last layer.
scanned 20 times in a range of wave number of 400–4500 cm−1 with
the spectra resolution of 4 cm−1 at room temperature to obtain the 2.5.5. Release kinetics of DOX from microneedles
FTIR spectra. Release profiles of DOX from different microneedles groups (G1, G2,
G3, and G4) were reported to study the effect of the drug location
2.4.4. Stability studies within the microneedle array on the drug release kinetics. PVA micro-
We investigated the effect of light exposure on the stability of DOX needle arrays of different groups were gradually dissolved in 10 mL PBS
in an aqueous solution or solid PVA microneedles. DOX solution in DI (10 mM, pH 7.4 to simulate the body fluid) under a constant shaking at
water (50 µL, 4 mg/mL) or DOX-loaded PVA microneedles G4 100 rpm and room temperature. Samples (300 μL) were withdrawn
(29.38 ± 1.97 mg, n = 4) was kept in a transparent glass vial under a from the vials at pre-determined intervals and replaced with the same
controlled fluorescent light (4100 K cool white, 40 W, 2600 lm bright- volume of fresh 10 mM PBS. The average amount of DOX released from
ness, Philips) for 24 h at room temperature. The drug levels in the the microneedles into the medium was plotted against time (n = 4).
formulations before and after the exposure were analyzed to calculate Then, the release flux was calculated from the slope of the linear por-
the drug photo-stability. tion of the release profile.

90
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

2.6. Characterization of microchannels 2.8. Skin distribution studies

2.6.1. Dye binding studies After 24 h of permeation studies, DOX formulation in the donor was
Microchannels created by unloaded PVA microneedles were stained discarded using Kimwipes and three receptor-soaked cotton swabs. Two
using methylene blue solution (1% w/v in deionized water) and vi- tapes (D-Squame stripping discs D101, CuDerm, Dallas, TX, USA) were
sualized under ProScope HR Digital USB Microscope to prove the skin pressed on the permeation area of skin using D-Squame pressure in-
microporation [31,33]. strument (225 g/sq.cm pressure, 22.24 mm pressure area, CuDerm,
Dallas, TX, USA). The tapes, containing the residual drug on the skin
2.6.2. Pore uniformity studies surface, were removed quickly using forceps. Then, the epidermis and
Fluorescent images of stained microchannels (Fluoresoft® 0.35%) dermis layers of skin were manually separated. The skin layers were
were captured and used to calculate Pore Permeability Index (PPI) minced with surgical scissors and collected individually in six-well
(Fluoropore analysis tool) to build a histogram of fluorescent intensity plates (Becton Dickinson, Franklin Lakes, NJ, USA). Ethanol (2 mL) was
of individual pore and indicate the uniformity of the channels [31,33]. added to each well. The wells were then covered with aluminum foil
and placed on a shaker at 100 rpm, room temperature, for 24 h. The
extracts were filtered through a 0.2 µm filter and analyzed using the
2.6.3. Histology studies
HPLC method.
Microneedles-treated skin was vertically sectioned (10 µm-thick
sections) to study the skin layers that microneedles could reach [31,33].
2.9. Quantitative analysis

2.6.4. Confocal laser microscopy studies


Drug analysis was performed using a Waters Alliance HPLC system
The microchannels in skin were stained using Fluoresoft® 0.35%
(2795 Separating Module) equipped with a fluorescent detector (Waters
and scanned under a computerized Leica SP8 confocal laser microscope
2475), an autosampler, a column heater, and Empower™ software
to measure the depth of the channels (n = 10) [31,33]. Due to the
version 2 (Waters Co., Milford, MA, USA). Samples were carried
fluorescent properties of DOX, its distribution within the needles and
through a column (Kinetex C18, 5 µm, 150 × 4.6 mm, Phenomenex,
deposition within the skin tissue was detected using z-stack confocal
Torrance, CA, USA) using a mixture of acetonitrile and DI water (0.1%
scanning (5 µm step size, n = 10) [8].
Triethylamine, pH was adjusted to 3.0 by phosphoric acid) (45:55 v/v).
The flow rate was set at 1.0 mL/min, the injection volume of 20 µL, and
2.6.5. Skin integrity measurement the detection excitation and emission wavelengths at 480 and 560 nm,
The barrier integrity of skin tissues before and after unloaded PVA respectively. The column temperature was maintained at 35 °C. The
microneedles insertion (as Poke&Solution group) was evaluated rapidly sample run time was 10 min while the drug retention time was ap-
and noninvasively using transepidermal water loss (TEWL, VapoMeter) proximately 5.4 min. This reversed-phase HPLC method provided a
and skin electrical resistance measurement (n = 4) (passing a constant linear range of 5–50,000 ng/mL (R2 = 0.999), LOD of 28.89 ng/mL,
current of 100 mV, 10 Hz, and duty cycle 50% through skin) [31,32]. and LOQ of 87.54 ng/mL.

2.7. In vitro permeation studies by vertical Franz diffusion cells 2.10. Data analysis

We investigated the permeation of DOX into and across derma- All results were reported as the mean with standard deviation (SD)
tomed human cadaver skin (New York Firefighters, NY, USA) using (n ≥ 4). Statistical calculations were performed in Microsoft Excel and
jacketed PermeGear V6 station vertical Franz diffusion cells (9 mm SPSS software package version 21.0 (IBM, USA). The Student’s t-test
orifice and 0.64 sq.cm diffusion area, Hellertown, PA, USA). Donor and One-Way ANOVA followed by Tukey HSD post-hoc test was applied
chamber contained 200 µL DOX solution in DI water 4.0 mg/mL to compare the results of different groups. Statistically, a significant
(Untreated, Poke&Solution, and Base-treated groups) or an array of difference was denoted by p value < 0.05.
DOX-loaded PVA microneedles (G1, G2, G3, and G4). The needles were
secured on the skin with an adhesive tape, and the donor was kept open 3. Results
to mimic in vivo conditions. The receptor compartment was filled with
10 mM PBS solution (5 mL, pH 7.4), continuously stirred at 600 rpm, 3.1. Characterization of formulation
and maintained at 37 °C by the built-in water circulation on the lower
part of the Franz cells. Skin tissues were treated by unloaded PVA mi- 3.1.1. Rheological properties of PVA matrix
croneedles (Poke&Solution) or array base (Base-treated) or DOX-loaded Rheological properties of PVA solution might affect its ability to fill
microneedles (G1, G2, G3, and G4). The intact skin, which was not the PDMS mold, the drug diffusion within the needle array, and the
treated by the microneedles, was assigned as control (Untreated group). conditions required to dry the microneedles. PVA solution in DI water
The skin was then clamped between the donor and receptor chambers displayed an elastic solid-like behavior as the storage modulus (G′) was
with the stratum corneum facing upward. Aliquots of 300 µL receptor always greater than the loss modulus (G″) at any point within the strain
fluid were taken at 0, 1, 2, 4, 6, 8, 22, and 24 h, followed by the re- range 0.01–100% (Fig. 2A). G′ and G″ were heading together at strain
placement with an equal volume of fresh 10 mM PBS. Then, the samples 25.10% but the convergence was absent from the curves. The elastic
were analyzed by the HPLC method. The cumulative delivery of DOX structure of PVA formulation was also revealed in the oscillatory fre-
through a diffusion unit area of skin was plotted against time (n = 4). quency sweep. A decrease in the formulation’s viscosity was caused by
The steady-state flux and lag time of DOX permeability was calculated an increase in the angular frequency (Fig. 2B). A reverse correlation
from the slope and x-intercept of the linear portion of the in vitro per- between temperature and viscosity of PVA solution was observed in
meation profile, respectively. The diffusion coefficient of DOX was which an increase in the temperature from 25 to 40 to 60 °C led to a
determined using Eq. (1) [34]: decrease in the viscosity (Fig. 2C). Thixotropy oscillatory study sug-
gested a stable structure of PVA solution under a high shear stress
h2
D= (shear rate 3000 s−1, 84.87% recovery) (Fig. 2D).
6t (1)

where D = diffusion coefficient (sq.cm/h), h = skin thickness (cm), and 3.1.2. DOX dissolution in PVA matrix
t = lag time (h). A polarized light microscope was used to investigate the drug

91
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

Fig. 2. Rheological properties of PVA solution (A) amplitude sweep, (B) frequency sweep, (C) temperature dependence of viscosity, (D) thixotropy oscillatory test.

dissolution in PVA solution. We observed a complete dissolution of PVA When PVA was removed from the spectrum as the background, the
in DI water (PVA 4–88, 50% w/w and PVA 26–88, 33.33% w/w) as well presence of DOX in the drug-loaded microneedles was validated with
as DOX in PVA solution (4 mg/mL). No traces of DOX crystal or un- peaks at 3324 cm−1 of NeH amine group, 1726 cm−1 of stretching C]
dissolved PVA debris were found in the microscopic images (data not O group, 1227 cm−1 of stretching CeO (strong alkyl aryl ether) group,
shown). These results favored a controlled uniformity of the drug dis- and 1110–1148 cm−1 of stretching CeN amine group (Fig. 3B). Thus,
tribution within the microneedles and between the needle arrays. the fabrication method successfully encapsulated DOX in PVA micro-
needles.

3.1.3. Fourier transform infrared spectrophotometer studies


The presence of the components (DOX and PVA) of the micro- 3.1.4. Stability studies
needles were investigated using a Fourier transform infrared spectro- The stability of DOX in solid PVA microneedles was studied under
photometer (FTIR). The FTIR spectrum of DOX-loaded PVA micro- the photo and thermal stress. Light stability studies revealed that under
needles indicated that PVA contributes the major portion of the a controlled light exposure, doxorubicin was markedly more stable in
microneedles. The PVA spectrum was represented by the peak at solid PVA microneedles (86.75 ± 4.08%) than in an aqueous solution
3425 cm−1 indicating stretching of hydroxyl groups, 2916 cm−1 of (26.37 ± 1.95%, n = 4, p < 0.01) (Fig. 4). This result suggested that
stretching CeH of alkane, 1369 cm−1 of OeH bending group of alcohol, incorporation of drug into solid microneedles improved the drug’s
and 1093 cm−1 of stretching C-O group (a secondary alcohol) (Fig. 3A). photo stability. Also, the effect of high temperature (60 and 90 °C

Fig. 3. FTIR spectrum of (A) unloaded PVA and DOX-loaded PVA microneedles, (B) DOX-loaded PVA microneedles using PVA as the background.

92
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

% Drug remained ± SD 100 # microneedles G1 and G3, most of the drug load-in the needle tips-was
dissolved within 1 h while the drug was constantly released from PVA
microneedles G2 and G4 (DOX was in both the needles and the base).
80
3.2.3. Scanning electron microscopy studies
60
SEM has been widely used to study the morphology and dimensions
of microneedles before and after skin treatment. Different grades of
40 PVA polymer (PVA 4-88 and PVA 26-88) and concentrations of PVA
solution (8 g/8 mL and 4 g/8 mL DI water) were examined for the ca-
20 pacity to fabricate the desired microneedles (Table 1). We used SEM
images to visualize freshly fabricated, untreated microneedles and two-
0 minute treated PVA microneedles (Fig. 7A–L). The results revealed that
PVA microneedles PVA solution sharp and strong microneedles could be fabricated from the above-
Groups mentioned materials and conditions. The needles had square pyramid
shape, sharp tips, and appropriate needle-to-needle distance. The di-
Fig. 4. DOX stability in PVA microneedles and PVA solution after 24-h light
mensions of PVA microneedles were elaborated in Table 2. The change
exposure (# indicated a significant difference between the two groups,
mean ± SD, n = 4, p < 0.001). in PVA grades and concentrations significantly alter the needle tip
diameter and needle length, however, the needle-to-needle distance and
base side were unaffected. The desired microneedle was chosen-PVA
during the fabrication process) on DOX stability in microneedles G1, 26–88 4 g/8mL-based on its sharp tip (tip diameter 1.93 ± 0.49 µm,
G2, and G4 was investigated (Fig. 1). For PVA microneedles G1, the n = 10), needle length (451.02 ± 8.07 µm, n = 10), and rapid in-skin
amount of drug adhered to tapes and evaporated due to vacuum ap- dissolution (needle length post-treatment 288.83 ± 119.01 µm,
plication were 349.21 ± 36.91 µg and 391.40 ± 16.57 µg (n = 4), n = 10). The dimensions of DOX-loaded microneedles (G1, G2, G3, G4)
respectively. The amount of DOX remained on PDMS mold after re- were also measured by SEM (Fig. 7M–P). Untreated DOX-loaded PVA
moval of the microneedles was 875.23 ng and the drug level in the microneedles had the average length ranging from 453.84 ± 20.16 µm
finished microneedles G1 was 8542.16 ng. We calculated the percen- (G4) to 465.33 ± 14.15 µm (G3), which was comparable to untreated
tage of DOX in microneedles G1 after the fabrication process to be microneedle (451.02 ± 8.07 µm, n = 10, p > 0.05). This result sug-
20.68%. For PVA microneedles G2, the amount of DOX evaporated in gested that the microneedle dimensions remained unchanged after the
the vacuum oven was 537.78 ± 43.63 µg (n = 4) while the average drug encapsulation. Upon skin insertion, the length of PVA micro-
drug levels adhered to PDMS mold and in the finished microneedles G2 needles in four groups continuously decreased with time, however, the
were 557.17 and 169017.41 ng, respectively. Thus, the percentage of needle-to-needle distance and base side did not. Also, we observed
drug remained in microneedles G2 post-fabrication was 31.46%. In PVA different rates of in-skin dissolution of microneedles in different groups
microneedles G4, DOX was exposed to 90 °C for one day (drug dis- (G1, G2, G3, and G4) (Table 3). PVA microneedles G4 displayed the
solution in PVA solution) in addition to the drying step (60 °C as G1 and fastest speed of dissolution from 453.84 ± 20.16 µm (0 h) to
G2), thus, might have a higher level of degradation than G1 and G2. 192.20 ± 35.84 µm (1 h) to 105.00 ± 23.50 µm (24 h).
The percentage of drug remained after the dissolution step was
35.08 ± 1.48% (n = 4); the amount of DOX in the finished micro- 3.2.4. Mechanical properties of PVA microneedles
needles G4 was 38.48 µg. The percentage of drug in microneedles G4 After the insertion of PVA microneedles on a stack of parafilm layers
after the fabrication process was calculated to be 17.46 ± 1.57% by either thumb (1.5 N) or an applicator, each layer was separated and
(n = 4). visualized under optical microscopes to study the mechanical uni-
formity of PVA microneedles. One hundred square-shaped pores, fol-
3.2. Characterization of PVA microneedles lowing the geometry of the square pyramidal microneedles, were cre-
ated on the parafilm layers. The side length of the square pores created
3.2.1. Optical microscopy on the first parafilm layer was 33.74 ± 1.79 µm (n = 20); the minimal
An optical microscope was employed to visualize the microneedle standard deviation of the side length suggested the uniformity of the
array base (Fig. 5A), unloaded PVA microneedles (Fig. 5B), and DOX- pore as well as the mechanical uniformity of the microneedles (Fig. 8).
encapsulated microneedles G1, G2, G3, and G4 (Fig. 5C–F). The array Furthermore, one hundred pores on the second parafilm layer revealed
base appeared clean and smooth without any microneedles while PVA the uniformity of the needle length. Also, microneedle insertion using
microneedles array was found to consist of 100 uniformly distributed thumb was performed to simulate the practical use of microneedles in
microneedles. Moreover, the color pattern of the drug-loaded micro- clinical settings. The treatment resulted in 100 square-shaped pores on
needles was in accordance with the location of the red-color drug load. parafilm layers. The side length of the pores (55.68 ± 1.65 µm,
Also, the dissolution and diffusion of DOX in PVA matrix was observed n = 20) was significantly larger than that by the applicator. Further-
in microneedles G1, G2, G3 and G4. more, the pressure from thumb moved the needles to the fourth layer of
the parafilm stack as compared to two layers disrupted by the needle
3.2.2. Fluorescent microscopy studies pressed by the applicator. These results indicated that thumb provided
We visualized the location of the drug load in untreated (G1, G2, a higher pressure on the microneedles than the applicator. In addition,
G3, and G4) (Fig. 6A–D) and treated PVA microneedles (1 h or 24 h the use of thumb also created uniform pores with a low standard de-
after the insertion into dermatomed human cadaver skin). As expected viation of the pores’ side length (1.65).
by the design (Fig. 1), PVA microneedles G1 and G3 had the drug lo-
cated at the tips of the needles (Fig. 6A and C). A uniform distribution 3.2.5. Release kinetics of DOX from microneedles
of DOX was observed in the needle tips (G3) or the entire array (G4) DOX-loaded PVA microneedles dissolved gradually in a limited
since DOX was completely dissolved in PVA solution before being volume of skin fluid to release the drug into skin layers. There was a
casted on PDMS mold. In microneedles G1 and G2, the drug diffusion marked difference in the release flux among the microneedles groups:
created a gradient of the drug concentration in the needles. We also G2 (8452.50 ± 79.86 ng/h), G1 (429.81 ± 9.80 ng/h), G4
observed a gradual dissolution of PVA microneedles in skin with a (395.72 ± 73.60 ng/h), and G3 (37.62 ± 0.33 ng/h) (n = 4,
decrease in the needle length after 1 h or 24 h post-treatment. In p < 0.001). DOX was released faster from PVA microneedles G1 and

93
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

Fig. 5. Microscopic images of (A) array base, (B) unloaded PVA microneedles, PVA microneedles group (C) G1, (D) G2, (E) G3, (F) G4.

G2 than microneedles G3 and G4. This observation suggested that the microchannels, Pore Permeability Index (PPI) reveals the relative flux
drug location significantly affect its release rate from PVA micro- of each pore to indicate the transdermal drug delivery. In this study, the
needles. The total amount of DOX in microneedles G1, G2, G3, and G4 bell-shaped distribution of PPI histogram (16.6 ± 8.17, n = 100,
was 8542.16 ± 47.21, 169017.41 ± 199.21, 3563.87 ± 178.96, and nzero = 0) indicated the uniformity of the microchannels created by
38483.29 ± 514.89 ng (n = 4), respectively (Fig. 9). unloaded PVA microneedles (Fig. 11). This result, in accordance with
the study of mechanical uniformity of PVA microneedles on the array,
3.3. Characterization of microchannels suggested a uniform diffusion of DOX solution through each micro-
channel.
3.3.1. Dye binding studies
Dye binding studies have been performed to confirm the success of 3.3.3. Histology studies
skin poration by microneedles. After the insertion of unloaded PVA The histological images of the vertical section of microneedles-cre-
microneedles, the skin was stained with methylene blue (a hydrophilic ated channels provide an accurate and reliable observation into the
dye) that could merely diffuse into the disrupted areas of skin (the fluid- actual skin layers that microneedles disrupt. Untreated dermatomed
filled microchannels), not the intact and lipophilic vicinity. The pattern human cadaver skin had an intact layer of stratum corneum (Fig. 12A
of 100 microchannels in skin followed the distribution of 100 PVA and C) while the insertion of unloaded PVA microneedles failed the skin
microneedles on the array (Fig. 10). This observation indicated a 100% barrier function, passed the stratum corneum and epidermis layers, and
penetration efficiency of PVA microneedles into dermatomed human reached the superficial layer of skin dermis (Fig. 12B and D).
skin.
3.3.4. Confocal laser microscopy studies
3.3.2. Pore uniformity studies Confocal laser microscopy in tandem with z-stack scanning has been used
The uniformity of microchannels in skin depends on various factors to measure the actual depth of microneedle-created channels in skin. The
that include mechanical uniformity, the sharpness of microneedles, and two-minute insertion, followed by the removal of the needles and application
properties of skin tissues. As drug permeated preferably through of Fluoresoft® (Poke&Solution), resulted in microchannels of

94
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

144.60 ± 40.43 kΩ/sq.cm (n = 28) while the insertion by PVA mi-


croneedles caused a sudden drop in the resistance from
173.79 ± 30.61 to 5.36 ± 2.34 kΩ/sq.cm (n = 4, p < 0.001), de-
noting successful poration. Interestingly, the insertion of array base
resulted in an insignificant alteration to the skin resistance from
159.99 ± 27.01 to 131.73 ± 43.36 kΩ/sq.cm (n = 4, p > 0.05)
(Fig. 14). These results suggested that dermatomed human skin was
disrupted by PVA microneedles, not the array base. Also, the skin in-
tegrity was investigated noninvasively by transepidermal water loss
measurement using a Vapometer. The base value of TEWL of untreated
dermatomed human skin was 38.41 ± 5.04 g/m2h (n = 8). The in-
sertion of either PVA microneedles or array base was found to cause an
insignificant change in TEWL value (n = 4, p > 0.05) (Fig. 14). The
skin disruption was represented by an increase in both ion transport
and water evaporation from the skin surface.

3.4. In vitro permeation studies by vertical Franz diffusion cells

In vitro permeation study has been commonly used to study the


transdermal drug delivery into and across the skin. In this study, we
investigated the effect of microneedle treatment and drug encapsula-
tion on the drug permeability. The drug’s high molecular weight
(543.52 g/mol), high melting point (229–231 °C), and hydrophilicity
Fig. 6. Fluorescent microscopic images of untreated PVA microneedles (A) G1, minimized its passive diffusion through the intact dermatomed human
(B) G2, (C) G3, (D) G4. skin (Untreated group, 0.00 ± 0.00 ng/sq.cm). Even though skin in-
tegrity studies (TEWL and skin electrical resistance measurement)
suggested that the insertion of array base caused an insubstantial dis-
Table 1
ruption to the skin barrier, the base treatment markedly enhanced the
PVA microneedles fabricated from different PVA grades.
drug delivery to 2393.95 ± 125.64 ng/sq.cm (n = 4). In Poke&
Symbol PVA Grade mPVA (g) VWater (mL) Solution group, PVA microneedles successfully porated the skin, cre-
ated 100 microchannels, and facilitated a substantial increase in the
PVA MN (4-88;4-8) 4-88 4 8
PVA MN (4-88;8-8) 4-88 8 8 drug cumulative delivery (4351.55 ± 560.87 ng/sq.cm, n = 4,
PVA MN (26-88;4-8) 26-88 4 8 p < 0.05), as compared to other groups. No drug was found in the
receptor fluid in G1, G3, and G4 groups. However, a significant amount
of DOX was released from microneedles G2, transported through the
121.50 ± 12.48 µm (n = 10) in depth. Meanwhile, the insertion of DOX- skin, and entered the receptor compartment (2305.05 ± 65.98 ng/
encapsulated microneedles without the following removal created micro- sq.cm) (Fig. 15A). We also calculated the steady-state flux of DOX from
channels in the skin with different depths: 112.00 ± 14.18 µm (G1), the slope of the linear portion of the permeation curves. The insertion of
154.50 ± 15.71 µm (G2), 125.50 ± 13.01 µm (G3), and unloaded PVA microneedles (Poke&Solution), DOX-loaded micro-
158.50 ± 17.96 µm (G4). One hour after the insertion, DOX was released needles (G2), and array base (Base-treated) resulted in a significantly
from PVA microneedles G2, deposited into skin tissues, and detected by higher flux than the untreated group (p < 0.05) (Table 4). Further-
confocal microscopy with 100 fluorescent-stained microchannels (Fig. 13A). more, the drug location within the microneedles array was found to
A rapid diffusion of DOX from its solution into fluid-filled microchannels in affect the drug release kinetics from the microneedles, thus, the drug
the skin was observed in Poke&Solution group (Fig. 13B). Untreated PVA delivery profile. The application of DOX solution on PVA microneedles-
microneedles G1, G2, G3, and G4 were also scanned by z-stack in confocal treated skin (Poke&Solution) provided the highest flux, diffusion coef-
microscopy to study the drug distribution within the array. DOX was uni- ficient, and rapid onset of drug delivery as compared to other groups
formly distributed in the entire array in microneedles G4 (Fig. 13C) while the (p < 0.05). No marked difference was found between the base-treated
drug was located only in tips of the microneedles in G1 and G3. Furthermore, and microneedles G2 groups regarding flux, lag time, and diffusion
the length of the drug location within the microneedles was also measured: coefficient (p > 0.05). The flux of DOX permeability after the insertion
422.00 ± 18.74 µm (G1), 451.00 ± 13.70 µm (G2), 431.00 ± 22.34 µm of PVA microneedles array (100 microneedles in an area of 0.2025
(G3), and 456.00 ± 23.19 µm (G4) (Fig. 13C and D), which was shorter sq.cm) was 225.71 ± 44.22 (Poke&Solution) and 15.29 ± 1.81 ng/
than the actual length of microneedles G1 and G3, measured by SEM images. sq.cm/h (G2) (Table 4). We extrapolated this result to predict that 494
The exposure to skin fluid allowed PVA microneedles to dissolve gradually to microchannels would be created in an area of 1 sq.cm of skin [31],
result in a significant decrease in the needle length to 245.00 ± 27.18 µm resulting in a flux of 1.11 (Poke&Solution) and 0.08 µg/sq.cm/h (G2).
(G1), 268.00 ± 37.65 µm (G2), 256.00 ± 34.38 µm (G3), and Then, the steady-state plasma concentration was predicted using Eq. (2)
202.00 ± 36.15 µm (G4) at 1 h after the treatment. [35]:
A∗Jss
3.3.5. Skin integrity measurement Css =
Cl (2)
We evaluated the intactness of the stratum corneummultilayers of
lipid and corneocyte membraneby measuring the ion transport through where Css = steady-state plasma concentration (µg/mL), A = permea-
skin (skin electrical resistance) and water evaporation from the skin tion area of skin (0.64 sq.cm), Jss = steady-state flux (µg/sq.cm/h), and
surface (TEWL) [31,32]. In this study, skin electrical resistance was Cl = clearance of DOX from the body.
measured before the microneedle treatment to guarantee the skin in- The clearance of DOX from human subjects was reported to be
tegrity before the in vitro permeation studies; and after the insertion of 10.5 mL/h (630 ± 150 mL/min) [36]. Thus, the steady-state plasma
PVA microneedles to validate the skin disruption. An uncompromised concentration of DOX was calculated to be 0.07 µg/mL (Poke&Solution)
barrier function of untreated skin provided a mean resistance value of and 0.005 µg/mL (G2). In clinical studies, Berg et al obtained a steady-

95
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

Fig. 7. SEM images of PVA microneedles (4-88; 4-8) (A, B) untreated microneedle, (C, D) treated microneedles; PVA microneedles (4-88; 8-8) (E, F) untreated
microneedles, (G, H) treated microneedles; PVA microneedles (26-88; 4-8) (I, J) untreated microneedles, (K, L) treated microneedles; untreated microneedles of (M)
G1, (N) G2, (O) G3, (P) G4.

Table 2
Microneedle dimensions (n = 10).
Microneedles Needle length (µm) Needle-to-needle distance (µm) Base side (µm) Tip Diameter (µm)

Untreated PVA MN (4-88;4-8) 420.97 ± 12.81 511.80 ± 3.04 161.45 ± 8.49 20.83 ± 2.22
Untreated PVA MN (4-88;8-8) 443.30 ± 30.46 12.74 ± 3.24
Untreated PVA MN (26-88;4-8) 451.02 ± 8.07 1.93 ± 0.49
Treated PVA MN (4-88;4-8) 343.19 ± 50.27 32.34 ± 16.68
Treated PVA MN (4-88;8-8) 336.96 ± 59.39 36.89 ± 12.93
Treated PVA MN (26-88;4-8) 288.83 ± 119.01 86.96 ± 33.41

PVA MN: Polyvinyl alcohol microneedles.


Treated PVA MN: PVA microneedles after two-minute insertion in dermatomed human skin.

state plasma concentration of DOX (0.02 µg/mL) after 72-h infusion in could be appropriate for mass production as well as clinical use since
49-year-old women with metastatic breast cancer [36]. To achieve this fentanyl transdermal patch has been fabricated with a larger size
desired drug level, we might use a larger size of PVA microneedle array: (5.25–42 sq.cm) [14].
0.32 sq.cm (Poke&Solution) and 4.72 sq.cm (G2). These dimensions

96
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

Cum Amt Release (ng) ± SD


Table 3 200000 A MN G1
Length of microneedle at different time after skin insertion (µm, n = 10).
MN G2
MN groups Untreated MN 1-h treated MN 24-h treated MN
150000 MN G3
G1 461.22 ± 12.47 288.12 ± 19.56 134.18 ± 20.85 MN G4
G2 458.58 ± 12.51 276.83 ± 25.13 109.58 ± 36.12
G3 465.33 ± 14.15 315.84 ± 40.50 142.15 ± 28.82
100000
G4 453.84 ± 20.16 192.20 ± 35.84 105.00 ± 23.50

50000
MN: PVA microneedles.
G1: DOX in the channels of PDMS mold.
G2: DOX on the surface and in the channels of PDMS mold. 0
G3: DOX in PVA solution in the channels of PDMS mold. 0 10 20 30 40 50
G4: DOX in microneedles and base of the array. Time (h)

Percentage drug release ± SD (%)


MN G1
3.5. Skin distribution studies 150 B
MN G2
The drug levels in skin layers were studied by skin separation and MN G3
extraction techniques. The application of DOX solution on unloaded MN G4
100
PVA microneedles-treated skin (Poke&Solution, 3020.09 ± 499.84 ng/
sq.cm) delivered a significantly higher amount of drug into skin tissue
than other treatment groups (n = 4, p < 0.05). A negligible amount of
drug was found in skin layers after the insertion of microneedles G3 and 50
G4 (Fig. 1). The encapsulation of DOX into both microneedles and array
base (Microneedles G2, 1609.86 ± 16.41 ng/sq.cm) insignificantly
increased the drug levels in the epidermis and dermis layers than the 0
array with drug contained in microneedles only (Microneedles G1, 0 1 2 20 30 40 50
985.90 ± 395.29 ng/sq.cm, n = 4, p > 0.05). For the untreated Time (h)
group, DOX was found in both the epidermis and dermis layers with a
Fig. 9. Release kinetic profile of DOX from PVA microneedles.
comparable level of the base-treated group (p > 0.05) (Fig. 15).

strength of polymeric microneedles: change in the components of


4. Discussion polymer matrix or dimensions of microneedles. For microneedles’ di-
mensions, widening the needle base can increase the needle strength,
Microneedles technology has been widely investigated to disrupt however, could result in a poor efficiency of skin penetration [1,6]. For
the stratum corneum layer, the primary barrier to drug diffusion, to alteration in the polymer matrix, PVA matrix had a weak elastic nature,
enhance transdermal delivery of therapeutic agents. In this study, we thus, had been used in combination with other materials (PVP, sucrose,
fabricated doxorubicin-encapsulated PVA microneedles with the drug dextran, CMC) to improve the mechanical strength of the needles [1].
load in different locations within the array. We deposited DOX into the The choice of materials significantly affects the reliability and con-
needles by discarding the drug on the mold surface, which left the drug sistency of needles penetration into skin tissues [14]. Also, the strength
load inside the mold cavities only. Also, we used highly viscous PVA of PVA microneedles could be significantly enhanced by increasing the
solution to minimize the drug diffusion, which would alter the locali- dextran concentration in the polymer matrix and the number of freeze-
zation, release, and delivery profile of the drug (Fig. 2). Chu et al re- thaw cycles [15], or using the atomized spray-filling method to make
ported that the low viscosity of the casting solution might facilitate the needle tips out of polymers with high mechanical strength [14]. This
diffusion of sulfohodamine B from the mold cavity to the microneedle work, for the first time, fabricated 100% (pure) PVA microneedles with
matrix during the microneedle drying step [1]. However, the high sufficient mechanical strength to penetrate skin tissues. We employed
viscosity of the polymer matrix created a challenge to fill the mold by the method described in the literature to fabricate microneedles G3 and
vacuum application [15,31] or atomized spray technique [14]. Thus, G4 [1]. However, this method posed a challenge to small molecules
centrifugation method was used for the fabrication process [37], and with high dose due to the low solubility of drugs in the viscous poly-
may also be simple and cost effective for mass production. meric solution that resulted in a small drug amount loaded in the
Investigators have tested the methods to improve the mechanical needles. Meanwhile, the technique used to make microneedles G1 and

Fig. 8. Microscopic images of parafilm treated by PVA microneedles by an applicator (A) first layer, (B) second layer by a microscope.

97
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

Fig. 10. Microchannels created in dermatomed human skin (A) untreated skin, (B) PVA microneedle-treated skin.

Fig. 11. The microchannels (A) with fluorescent intensity (B) and pore permeability index (PPI) histogram (C) of PVA microneedles.

Fig. 12. Histological sectioning images of (A, B) untreated dermatomed human skin and (C, D) PVA microneedle-treated dermatomed human skin.

98
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

(A) (B) (C)

0 μm 10 μm 20 μm 30 μm 40 μm

50 μm 60 μm 70 μm 80 μm 90 μm

100 μm 110 μm 120 μm 130 μm 140 μm


(D)
Fig. 13. Confocal images of (A) dermatomed human skin one hour after treatment by PVA MN G2, (B) microchannels in Poke&Solution group, (C) PVA MN G4, (D) z-
stack of microchannels at 1 h after PVA MN G2 insertion.

et al also filled the mold with a solution of sulforhodamine B in deio-


Skin Resistance ± SD (kOhm/sq.cm)

nized water, dried the drug solution, and covered the mold with
200 200 polymer solution [1]. The drug load located within the needle tips (G1
* *
TEWL Value ±SD (g/m2h)

and G3) could be advantageous to minimize the drug wastage, reduce


150 150 the production cost for expensive drugs, mitigate the variation in pe-
netration efficiency of microneedles in different user groups, and con-
trol the delivery of potent compounds with narrow therapeutic window
100 100 [1].
The in-situ dissolution of microneedles provided a practical in-
vestigation into the dissolution kinetics of the needles in skin tissue
50 50
where the needle tips dissolved first, followed by the needle base. In
this study, PVA microneedles dissolved gradually in skin moisture to
0 0 leave a portion of the needle base unchanged after 24 h in the skin
d d ion d d ion while the backside of the microneedle substrate remained intact. This
ate ate t ate ate t
n tre e-tre Solu n tre e-tre Solu result could be explained by the slow dissolution kinetics of PVA matrix
U as & U s &
B ke Ba oke as well as the indentation of skin surface to reduce the contact area
Po P between the needle base and the skin fluid. The needles must be em-
Groups bedded in the skin for a longer time to facilitate the dissolution of the
Fig. 14. Measurement of transepidermal water loss and skin electrical re- needle base outside the skin. Also, the rate of dissolution of PVA mi-
sistance values (* indicated statistical difference from Poke&Solution group, croneedles could be increased by using the combination of PVA and
mean ± SD, n = 4, p < 0.05). PVP or sucrose in the polymer matrix [6].
We also studied the drug release profile from PVA microneedles,
G2 (evaporation of ethanol to leave solid DOX in the mold) allowed a which has been found to be highly correlated with the drug locations
markedly higher drug load since the volume and concentration of the within microneedle array and the drug delivery into and across the skin
drug solution could be adjusted to a desired level [1,14]. Similarly, Chu [1]. The design of microneedles G1 and G3 might be favorable to

99
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

(A) dissolving microneedles remains challenging. The loading capacity


could be enhanced to certain magnitudes by increasing (i) the drug
Avg Amt/sq.cm (ng/sq.cm) ± SD

6000 concentration in the polymer matrix which could weaken the me-
Untreated
chanical strength of the needles, (ii) the needle length which might
Base-treated
cause a risk of pain, bleeding, and patients’ incompliance, (iii) the
4000 * Poke&Solution
MN G1 needle density which might lead to “bed of nail” effect, (iv) the volume
MN G2 of microneedles by expanding the microneedle base which decreased
2000 ** MN G3 the penetration efficiency, (v) the treatment duration of microneedles
MN G4 to facilitate the dissolution of the needles in skin, and (vi) the area of
needle array which might be difficult to design the applicator or insert
0 the needles on the uneven surface of skin.
0 2 4 6 8 22 24 A complete dissolution of drugs in the solvent (G1 and G2) or
Time (h) polymer matrix (G3 and G4) appears crucial for the uniform distribu-
tion of the drug inside the fabricated microneedles as well as the con-
(B) tent uniformity among the arrays. The drug distribution within mi-
Avg Amt/sq.cm (ng/sq.cm) ± SD

croneedles has been estimated using a dye: sulforhodamine B [1] or red


4000
Untreated FluoSpheres™ [14]. In this study, the fluorescent properties of DOX
* Base-treated allowed visualizing the actual distribution pattern of the drug within
3000
Poke&Solution the microneedle array. Due to the poor solubility, insulin was just
MN G1 suspended in polymer mixture of PVA, dextran, and CMC before being
2000 * * MN G2 cast on the mold [15]. On one hand, this approach might allow an in-
MN G3 crease in the drug load in the needles, thus, enhance the drug delivery
1000 MN G4 into the skin. On the other hand, the use of drug suspension might lead
to a needle-to-needle and array-to-array variation in the drug levels.
0 Thus, an increase in the drug solubility in the polymer mixture or sol-
Total Skin Epidermis Dermis
vent vehicle is favorable for an enhanced encapsulation of drug in the
Skin Layers
microneedles.
Fig. 15. (A) In vitro permeation studies on dermatomed cadaver human skin, The rheological properties of PVA solution were studied to evaluate
(B) DOX distribution in skin layers (* indicated statistical difference from the its physical characteristics and possibility to fabricate PVA micro-
other groups, mean ± SD, n = 4, p < 0.05). needles. PVA matrix showed an elastic behavior while its temperature-
dependent viscosity was comparable to that of a topical hydrogel [32].
release the drug loads (from the needle tips) in a fast and complete The distinguished effect of temperature on the viscosity was found in
manner. In microneedles G4, DOX could be released only when the PVA the low range of shear rate. This observation was used to explain the
needles dissolved. This mechanism slowed down the rate of drug re- use of high temperature (to reduce the viscosity of the polymer matrix)
lease in microneedles G4. Yang et al reported a fast release of insulin to fill the mold cavities with PVA solution (40 °C) [14] and dry the
from PVA microneedles where the drug is loaded inside the needles microneedles (60 °C). In contrast, using atomized spray technique,
tips: 30% of the drug load was released within 30 min and an additional McGrath et al found viscosity an inessential factor in the fabrication
26% released within 4 h [15]. Chu et al fabricated PVA/PVP micro- process. Materials with various viscosities could be spray-filled the
needles with the encapsulation of sulforhodamine B in the needle tips to mold using the same set of processing parameters [14]. The structural
obtain an initial burst release within the first 30 s and approximately stability of PVA solution, as illustrated in thixotropy oscillatory study,
80% of the drug load released from the needle within 10 min. Mean- suggested that the polymer structure most likely resisted the cen-
while, the microneedles with sulforhodamine B located in the needle trifugation force (4000 rpm for 30 min during the microneedle fabri-
matrix but not in the base achieved a smaller burst release, followed by cation).
a gradual increase in the drug release to approximately 70%. The en- Parafilm M® film has been shown as a simple and effective model to
capsulation of the drug into both needle and base demonstrated a low study the insertion behavior, length uniformity, and mechanical uni-
release efficiency (about 20% of the drug load) [1]. However, the en- formity of microneedles [31]. In this study, we used Parafilm M® film to
capsulation of drugs within the tips of the microneedles could sig- evaluate the mechanical properties of PVA microneedles. The com-
nificantly reduce the drug load in the needles [6]. parable dimensions of pores created in parafilm layers indicated the
In general, crossing the high dose threshold of drug encapsulation in uniformity of the needles while the indentation area suggested that only

Table 4
Flux, lag time, and diffusion coefficient of DOX transdermal delivery (n = 4).
Group Treatment Drug amount applied (µg) Drug deliverya (ng/sq.cm) Fluxb (ng/sq.cm/h) Lag timec (h) Diffusion coefficientd (sq.cm/h) × 10−5

Untreated No 800 0.00 ± 0.00 0 NAe 0


Base-treated Array base 800 2393.95 ± 125.64 17.55 ± 3.21 113.52 ± 21.41 0.30
Poke&solution Unloaded MN 800 4351.55 ± 560.87 225.71 ± 44.22 7.77 ± 1.71 4.34
G1 Loaded MN 8.54 ± 0.05 0.00 ± 0.00 0 NAe 0
G2 Loaded MN 169.02 ± 0.20 2305.05 ± 65.98 15.29 ± 1.81 125.50 ± 14.86 0.27
G3 Loaded MN 3.56 ± 0.18 0.00 ± 0.00 0 NAe 0
G4 Loaded MN 38.58 ± 0.51 0.00 ± 0.00 0 NAe 0

a
Cumulative amount (Q24) of MTX permeated through unit diffusion area in 24 h.
b
Steady-state flux (Jss) - calculated from the linear slope of the permeation curve.
c
Lag time (Tlag)- calculated as the x-intercept of the linear portion of the permeation curve (R2 > 0.95).
d
Diffusion coefficient (D) – calculated using an equation: D = h2/6t, where D is diffusion coefficient (sq.cm/h), h is skin thickness (cm), and t is lag time (h).
e
Not applicable.

100
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

certain portion of microneedles could penetrate the viscoelastic mate- insertion [14]. Yang et al have used histological images of skin sections
rial. Furthermore, the insertion behavior of microneedles depended on to measure the depth of microchannels, created by polymer micro-
the needle density, geometry, material, needle length, sharpness, and needles (PVA, dextran, and CMC) to be approximately 50% of the
base dimensions [31]. Larraneta et al reported that hydrogel-forming needles’ length [15]. However, measuring the depth of the channels
microneedles could reach 100% penetration ratio in the parafilm layer using histological sectioning might be challenging and flawed as phy-
and less than 50% in the second layer [38]. Yang et al have found that a sical distortion could alter the dimensions of the channels. Also, we
modification of polymer formulation or processing parameters of found a significant decrease in skin electrical resistance and increase in
drying step could considerably alter the strength and mechanical TEWL values after microneedles insertion to indicate the disruption of
properties of fabricated microneedles [15]. Regarding the method for the skin barrier [32,33].
inserting microneedles, a simple and standardized applicator might be Microneedles have displayed their effectiveness in enhancing
preferable to thumb pressure to provide a consistent force, speed, and transdermal delivery of various therapeutic agents [6,31,33]. The
vibration, especially to be used by untrained users [6]. magnitude of delivery enhancement varied, depending on the needle
We also observed a significant difference in the microneedle length dimensions, needle materials, skin viscoelasticity, and physicochemical
(untreated, 1-h treated, and 24-h treated microneedles G1 and G3) properties of the compounds [33]. In this study, in vitro permeation
when measured by SEM and confocal laser microscopy (p < 0.05) studies were performed using Franz diffusion cells to indicate that the
since SEM provided the actual dimensions of PVA microneedles while transdermal drug delivery was highly associated with the drug release
confocal microscopy only revealed the location of the fluorescent drug profile, the depth of microneedle penetration, and the drug localization
load. This difference was absent from microneedles G2 and G4 where within the microneedle array [1]. The results of permeation studies
the drug was distributed in both the microneedles and array substrate. were in accordance with the characterization studies of both micro-
Successful skin microporation was affected by several factors such as needles and microchannels that validated the successful microporation
microneedles length, design, geometry, density, tensile strength, and of the dermatomed human skin by PVA microneedles. The hydro-
the component materials [39,40]. The dimensions of PVA micro- philicity, charge, and high molecular weight of DOX limited its diffu-
needles: sharpness (tip diameter), length, and density (needle-to-needle sion through the lipophilic layer of stratum corneum [20,21]. Fur-
distance), measured in scanning electron microscopy, indicated the thermore, the anthracycline structure of DOX interacted with anionic
likeliness of PVA microneedles to avoid the “bed of nail” effect, bypass lipids in the stratum corneum to prevent the drug penetration into the
the skin indentation, and penetrate the skin tissue [14]. Experiments of underlying skin layers [21]. These characteristics of DOX contributed to
skin insertion were conducted to validate this suggestion: dye binding, almost no delivery of DOX by passive diffusion in the untreated group.
pore uniformity, histology, confocal laser microscopy, and skin in- Thus, we employed PVA microneedles to disrupt the stratum corneum
tegrity studies. The results of these studies were in an agreement to to facilitate the drug diffusion through the created hydrophilic, fluid-
indicate the successful microporation of 100 PVA microneedles to filled channels to reach the hydrophilic layer of skin dermis.
create 100 microchannels in dermatomed human skin. An insignificant decrease in the electrical resistance of skin in base
Using a rapid, accurate, and non-invasive techniqueconfocal laser treatment (159.99 ± 27.01 to 131.73 ± 43.36 kΩ/sq.cm, Fig. 14),
scanning microscopywe could measure the actual depth of the channels denoting a partial failure of the skin barrier, was highly correlated to an
(approximately 27% the length of PVA microneedles) in the skin enhanced delivery in the base-treated group (2393.95 ± 125.64 ng/
without physical artifacts [31,33]. This incomplete insertion of PVA sq.cm, n = 4). This result could be possibly explained by the sharp
microneedles could be explained by the indentation of skin upon the periphery of the base after the drying step, which might be able to
needle insertion [1,31,41] and the immediate resealing of skin after the disrupt the skin integrity to a certain level. The difference in the drug
needle removal [42]. The efficiency and depth of penetration of mi- permeability in microneedles groups could be attributed to the differ-
croneedles depended on several factors such as the pressure and ence in the amount of drug load as well as the drug localization within
duration of application, composition of the needles [14], dimensions the needle array. In this study, we loaded 169.02 ± 0.20 µg DOX into
(length, sharpness, and geometry) of the needles, and the viscoelastic PVA microneedles G2 to deliver 2.31 ± 0.07 µg drug into dermatomed
properties of the skin samples [6,14,43]. The insertion ratio of micro- human skin in vitro while a smaller drug load in the needles (G1:
needles into human skin in vivo could increase noticeably due to the 8.54 ± 0.05 µg; G3: 3.56 ± 0.18 µg, and G4: 38.58 ± 0.51 µg) led to
decrease in skin deformation under the body tension [6,44]. Based on a negligible transdermal drug permeability (Table 4). The drug on the
the depth of the channels (112.00–158.50 µm), we could conclude that surface of the array substrate (G2) might be able to dissolve within 24 h
PVA microneedles bypassed the stratum corneum (10–15 µm thick) and study to provide an additional amount of drug on skin while the drug,
the underlying epidermis layer (50–100 µm thick) to reach the super- widely scattered in the solid array substrate (G4), was least likely re-
ficial layer of the skin dermis [2,13]. Furthermore, the protocol of leased out of the polymer matrix, thus be considered as a waste [1].
confocal laser microscopy studies simulated the design of in vitro per- Using an atomized spray process, McGrath et al have fabricated keto-
meation studies of the Poke&Solution group, thus, indicated the skin profen-encapsulated dissolving microneedles from 5% w/v PVA aqu-
layer that the drug formulation could reach initially. Interestingly, the eous solution. They could load 26.97 ± 8.56 µg ketoprofen into the
fluorescent property of DOX allowed for visualization of the permeation needles to deliver 5.6 ± 1.9 µg into porcine skin ex vivo after 24 h
pathway and distribution pattern of the drug in skin tissue under a permeation [14]. Thus, the drug delivery was dependent on the mi-
confocal laser microscope [26,29]. croneedles design, the fabrication technique, and the skin models.
In histology studies, PVA microneedles bypassed the stratum cor- A crucial factor in transdermal drug delivery is the lag time that has
neum and epidermis layers and reached the superficial dermis layer. been defined as the duration that a drug takes to bypass the skin layers
This result appeared ideal for microneedle design. In addition to to enter the blood circulation [31]. A shorter lag time is preferable to
stratum corneum layer, the epidermis was found to limit the drug de- achieve a rapid onset of drug delivery as well as therapeutic effect. The
livery [45]. Thus, breaching the entire epidermis layer allowed to place insertion of PVA microneedles significantly decreased the lag time of
the drug into the dermis layer and maximize the transdermal drug DOX delivery through the human skin. The shortest lag time of Poke&
delivery into the blood circulation. Meanwhile, the depth of the chan- Solution group (7.77 ± 1.71 h) might be due to the physical insertion
nels was sufficiently shallow to avoid the intrusion into pain receptor or of the microneedles, which penetrated the stratum corneum and epi-
blood capillaries in the dermis. The minimal invasiveness has been dermis; and allowed the drug to diffuse into the dermis layer, thus
reported to be the major advantage of microneedles [46,47]. Also, significantly shortened the traveling distance of the drug. Even though
McGrath et al have studied the rate of epidermis compression, epi- microneedles G2 successfully pierced the skin tissue, the needles had to
dermis breach and stratum corneum rupture upon microneedle slowly dissolve in a limited volume of skin interstitial fluid, release the

101
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

drug load, and deliver it into the skin layers. This observation explained Conflict of interest
the longer lag time (125.50 ± 14.86 h) and delayed drug delivery in
microneedles G2. An alteration to the polymer matrix and components The authors declare no conflict of interest.
could be made to enhance the dissolution rate and decrease the lag time
of the drug delivery. We have reported that microneedles treatment Appendix A. Supplementary material
resulted in a reduction in the lag time with several compounds such as
nicardipine hydrochloride in vitro and in vivo [48] and low molecular Supplementary data associated with this article can be found, in the
weight heparin in vitro [49]. Differently, the insertion of maltose mi- online version, at https://doi.org/10.1016/j.ejpb.2018.05.017.
croneedles in vitro did not cause any significant change in the lag time
of transdermal delivery of glycopyrrolate [50]. References
The skin treatment by PVA microneedles in this study provided a
higher DOX delivery (4351.55 ± 560.87 ng/sq.cm), shorter lag time [1] L.Y. Chu, S.-O. Choi, M.R. Prausnitz, Fabrication of dissolving polymer micro-
(7.77 ± 1.71 h), and higher flux (225.71 ± 44.22 ng/sq.cm/h, n = 4) needles for controlled drug encapsulation and delivery: bubble and pedestal mi-
croneedle designs, J. Pharm. Sci. 99 (2010) 4228–4238.
as compared to the physical disruption of dermatomed human cadaver [2] A.K. Banga, Transdermal and Intradermal Delivery of Therapeutic Agents:
skin (0.73 ± 0.10 mm thickness, n = 12) by maltose microneedles Application of Physical Technologies, CRC Press, 2011.
with the drug permeability of 1513.68 ± 91.24 ng/sq.cm, lag time of [3] A. Sivaraman, S.S. Ganti, H.X. Nguyen, G. Birk, A. Wieber, D. Lubda, A.K. Banga,
Development and evaluation of a polyvinyl alcohol based topical gel, J. Drug Deliv.
8.14 ± 5.27 h, and flux of 91.45 ± 36.63 ng/sq.cm/h (n = 4) [31]. Sci. Technol. 39 (2017) 210–216.
This difference might be attributed to the use of thinner skin in the [4] G.M. Glenn, D.C. Flyer, L.R. Ellingsworth, S.A. Frech, D.M. Frerichs, R.C. Seid,
present study (0.45 ± 0.11 mm thickness, n = 28) as well as the mi- J. Yu, Transcutaneous immunization with heat-labile enterotoxin: development of a
needle-free vaccine patch, Expert Rev. Vaccines. 6 (2007) 809–819.
croneedle structure. Thicker skin samples could cause a “cushion” effect
[5] A.K. Banga, Microporation applications for enhancing drug delivery, Expert Opin.
to reduce the penetration depth of the needles and form a longer dis- Drug Deliv. 6 (2009) 343–354, http://dx.doi.org/10.1517/17425240902841935.
tance for the drug to travel and reach the receptor fluid. Similarly, [6] L.Y. Chu, M.R. Prausnitz, Separable arrowhead microneedles, J. Control. Release
Off. J. Control. Release Soc. 149 (2011) 242–249, http://dx.doi.org/10.1016/j.
McGrath et al reported that the use of full thickness porcine skin led to a
jconrel.2010.10.033.
decrease in the delivery of ketoprofen as compared to dermatomed skin [7] C. Tas, J.C. Joyce, H.X. Nguyen, P. Eangoor, J.S. Knaack, A.K. Banga,
[14]. Maltose microneedles could rapidly dissolve in skin tissue M.R. Prausnitz, Dihydroergotamine mesylate-loaded dissolving microneedle patch
whereas PVA microneedles took a significantly longer period to swell made of polyvinylpyrrolidone for management of acute migraine therapy, J.
Controlled Release 268 (2017) 159–165.
and dissolve. Thus, PVA microneedles most likely delayed the partial [8] M.-C. Chen, M.-H. Ling, K.-W. Wang, Z.-W. Lin, B.-H. Lai, D.-H. Chen, Near-infrared
resealing of the channels to maintain a larger volume of the opened light-responsive composite microneedles for on-demand transdermal drug delivery,
channels to facilitate the drug diffusion. Biomacromolecules 16 (2015) 1598–1607, http://dx.doi.org/10.1021/acs.biomac.
5b00185.
The application of microneedles in future in vivo and clinical ex- [9] H.X. Nguyen, A.K. Banga, Microneedle-mediated delivery of vismodegib across
periments have been elaborated previously. Also, the possible differ- skin, J. Invest. Dermatol. (2015) S58–S58.
ence in the drug delivery in in vivo and in vitro studies has been dis- [10] A. Puri, H.X. Nguyen, A.K. Banga, Microneedle-mediated intradermal delivery of
epigallocatechin-3-gallate, Int. J. Cosmet. Sci. 38 (2016) 512–523.
cussed [31]. Due to the comparable barrierlayers of dead cells in the [11] H.S. Gill, M.R. Prausnitz, Pocketed microneedles for drug delivery to the skin, J.
stratum corneumbetween cadaver and living skin, the results of this Phys. Chem. Solids 69 (2008) 1537–1541.
study were applicable and relevant to human studies. We envision a [12] M. Matteucci, M. Fanetti, M. Casella, F. Gramatica, L. Gavioli, M. Tormen,
G. Grenci, F. De Angelis, E. Di Fabrizio, Poly vinyl alcohol re-usable masters for
more likely scenario that users could purchase microneedles and self- microneedle replication, Microelectron. Eng. 86 (2009) 752–756, http://dx.doi.
administer them without any special training [6]. org/10.1016/j.mee.2009.01.068.
[13] F. Pérennès, B. Marmiroli, M. Matteucci, M. Tormen, L. Vaccari, E.D. Fabrizio,
Sharp beveled tip hollow microneedle arrays fabricated by LIGA and 3D soft li-
5. Conclusion
thography with polyvinyl alcohol, J. Micromech. Microeng. 16 (2006) 473, http://
dx.doi.org/10.1088/0960-1317/16/3/001.
Poly (vinyl alcohol) microneedles were fabricated by micromolding [14] M.G. McGrath, S. Vucen, A. Vrdoljak, A. Kelly, C. O’Mahony, A.M. Crean, A. Moore,
technique and characterized by optical microscopy, fluorescent micro- Production of dissolvable microneedles using an atomised spray process: effect of
microneedle composition on skin penetration, Eur. J. Pharm. Biopharm. 86 (2014)
scopy, scanning electron microscopy, mechanical properties, and drug 200–211.
release. The microneedles successfully porated dermatomed human [15] S. Yang, Y. Feng, L. Zhang, N. Chen, W. Yuan, T. Jin, A scalable fabrication process
cadaver skin to create uniform microchannels (dye binding, pore uni- of polymer microneedles, Int. J. Nanomed. 7 (2012) 1415–1422.
[16] O. Tacar, P. Sriamornsak, C.R. Dass, Doxorubicin: an update on anticancer mole-
formity, histology, confocal laser microscopy, and skin integrity stu- cular action, toxicity and novel drug delivery systems, J. Pharm. Pharmacol. 65
dies). The insertion of microneedles (4351.55 ± 560.87 ng/sq.cm) (2013) 157–170.
resulted in a significantly greater delivery of doxorubicin into and [17] K.A. Janes, M.P. Fresneau, A. Marazuela, A. Fabra, M.J. Alonso, Chitosan nano-
particles as delivery systems for doxorubicin, J. Controlled Release 73 (2001)
across human skin, as compared to passive diffusion (0.00 ± 0.00 ng/ 255–267.
sq.cm, n = 4, p = 0.00). A change in the drug localization within the [18] J. Wu, S. Xu, W. Jiang, Y. Shen, M. Pu, Facile preparation of a pH-sensitive nano-
microneedle array was found to markedly alter the release and delivery magnetic targeted system to deliver doxorubicin to tumor tissues, Biotechnol. Lett.
37 (2015) 585–591.
profile of doxorubicin in vitro. [19] J.O. Kim, A.V. Kabanov, T.K. Bronich, Polymer micelles with cross-linked polyanion
core for delivery of a cationic drug doxorubicin, J. Controlled Release 138 (2009)
Acknowledgements 197–204.
[20] S.F. Taveira, A. Nomizo, R.F. Lopez, Effect of the iontophoresis of a chitosan gel on
doxorubicin skin penetration and cytotoxicity, J. Controlled Release 134 (2009)
This work was supported by a grant from Millipore Sigma, a 35–40.
Business of Merck KGaA, Darmstadt, Germany. [21] H. Herai, T. Gratieri, J.A. Thomazine, M.V.L.B. Bentley, R.F.V. Lopez, Doxorubicin
skin penetration from monoolein-containing propylene glycol formulations, Int. J.
Pharm. 329 (2007) 88–93.
Author contributions [22] M.L. Tan, A.M. Friedhuber, D.E. Dunstan, P.F. Choong, C.R. Dass, The performance
of doxorubicin encapsulated in chitosan–dextran sulphate microparticles in an os-
H.X.N. carried out most experimental studies, analyzed data, and teosarcoma model, Biomaterials 31 (2010) 541–551.
[23] A. Gabizon, H. Shmeeda, Y. Barenholz, Pharmacokinetics of pegylated liposomal
wrote the manuscript; B.D.B and Y.K. assisted in developing the mi- doxorubicin, Clin. Pharmacokinet. 42 (2003) 419–436.
croneedle fabrication method; H.X.N., A.W., G.B., D.L., and A.K.B. de- [24] G.L. Plosker, Pegylated liposomal doxorubicin, Drugs 68 (2008) 2535–2551.
signed the study; B.D.B developed and run HPLC analytical method; [25] H.T. Ta, C.R. Dass, I. Larson, P.F. Choong, D.E. Dunstan, A chitosan–dipotassium
orthophosphate hydrogel for the delivery of Doxorubicin in the treatment of os-
B.D.B, Y.K., A.W., G.B., D.L., and A.K.B. revised the manuscript; and teosarcoma, Biomaterials 30 (2009) 3605–3613.
A.W., G.B., D.L., and A.K.B. supervised the project.

102
H.X. Nguyen et al. European Journal of Pharmaceutics and Biopharmaceutics 129 (2018) 88–103

[26] I. Han, M. Kim, J. Kim, Enhanced transfollicular delivery of adriamycin with a li- [39] R.F. Donnelly, R. Majithiya, T.R.R. Singh, D.I.J. Morrow, M.J. Garland, Y.K. Demir,
posome and iontophoresis, Exp. Dermatol. 13 (2004) 86–92. K. Migalska, E. Ryan, D. Gillen, C.J. Scott, A.D. Woolfson, Design, optimization and
[27] M.-C. Chen, K.-W. Wang, D.-H. Chen, M.-H. Ling, C.-Y. Liu, Remotely triggered characterisation of polymeric microneedle arrays prepared by a novel laser-based
release of small molecules from LaB6@SiO2-loaded polycaprolactone microneedles, micromoulding technique, Pharm. Res. 28 (2011) 41–57, http://dx.doi.org/10.
Acta Biomater 13 (2015) 344–353, http://dx.doi.org/10.1016/j.actbio.2014.11. 1007/s11095-010-0169-8.
040. [40] J.-H. Park, Y.-K. Yoon, S.-O. Choi, M.R. Prausnitz, M.G. Allen, Tapered conical
[28] Y. Ma, S.E. Boese, Z. Luo, N. Nitin, H.S. Gill, Drug coated microneedles for mini- polymer microneedles fabricated using an integrated lens technique for transdermal
mally-invasive treatment of oral carcinomas: development and in vitro evaluation, drug delivery, IEEE Trans. Biomed. Eng. 54 (2007) 903–913.
Biomed. Microdev. 17 (2015) 44, http://dx.doi.org/10.1007/s10544-015-9944-y. [41] W. Martanto, J.S. Moore, T. Couse, M.R. Prausnitz, Mechanism of fluid infusion
[29] I. Mansoor, J. Lai, S. Ranamukhaarachchi, V. Schmitt, D. Lambert, J. Dutz, during microneedle insertion and retraction, J. Control. Release Off. J. Control.
U.O. Häfeli, B. Stoeber, A microneedle-based method for the characterization of Release Soc. 112 (2006) 357–361, http://dx.doi.org/10.1016/j.jconrel.2006.02.
diffusion in skin tissue using doxorubicin as a model drug, Biomed. Microdev. 17 017.
(2015) 9967, http://dx.doi.org/10.1007/s10544-015-9967-4. [42] H. Kalluri, A.K. Banga, Formation and closure of microchannels in skin following
[30] H.X. Nguyen, A.K. Banga, Delivery of methotrexate and characterization of skin microporation, Pharm. Res. 28 (2011) 82–94, http://dx.doi.org/10.1007/s11095-
treated by fabricated PLGA microneedles and fractional ablative laser, Pharm. Res. 010-0122-x.
35 (2018) 68, http://dx.doi.org/10.1007/s11095-018-2369-6. [43] D. Duan, C. Moeckly, J. Gysbers, C. Novak, G. Prochnow, K. Siebenaler, L. Albers,
[31] H.X. Nguyen, A.K. Banga, Fabrication, characterization and application of sugar K. Hansen, Enhanced delivery of topically-applied formulations following skin pre-
microneedles for transdermal drug delivery, Ther. Deliv. 8 (2017) 249–264. treatment with a hand-applied, plastic microneedle array, Curr. Drug Deliv. 8
[32] H.X. Nguyen, A. Puri, A.K. Banga, Methods to simulate rubbing of topical for- (2011) 557–565.
mulation for in vitro skin permeation studies, Int. J. Pharm. 519 (2017) 22–33, [44] J. Enfield, M.-L. O’Connell, K. Lawlor, E. Jonathan, C. O’Mahony, M. Leahy, In-vivo
http://dx.doi.org/10.1016/j.ijpharm.2017.01.007. dynamic characterization of microneedle skin penetration using optical coherence
[33] H.X. Nguyen, A.K. Banga, Enhanced skin delivery of vismodegib by microneedle tomography, J. Biomed. Opt. 15 (2010) 46001, http://dx.doi.org/10.1117/1.
treatment, Drug Deliv. Transl. Res. 5 (2015) 407–423, http://dx.doi.org/10.1007/ 3463002.
s13346-015-0241-3. [45] S.N. Andrews, E. Jeong, M.R. Prausnitz, Transdermal delivery of molecules is lim-
[34] R. Eliaz, N. Grossman, S. Katz, R. Zur-Mashiach, M.H. Grunwald, H. Reuveni, ited by full epidermis, not just stratum corneum, Pharm. Res. 30 (2013) 1099–1109.
L. Rosenberg, J. Kost, In vitro analysis of bromine chemical burns with use of full- [46] H.S. Gill, D.D. Denson, B.A. Burris, M.R. Prausnitz, Effect of microneedle design on
thickness human skin, J. Burn Care Res. 19 (1998) 18–24. pain in human volunteers, Clin. J. Pain. 24 (2008) 585–594, http://dx.doi.org/10.
[35] R.H. Guy, J. Hadgraft, Rate control in transdermal drug delivery? Int. J. Pharm. 82 1097/AJP.0b013e31816778f9.
(1992) R1–R6. [47] M.I. Haq, E. Smith, D.N. John, M. Kalavala, C. Edwards, A. Anstey, A. Morrissey,
[36] S.L. Berg, K.H. Cowan, F.M. Balis, J.S. Fisherman, A.M. Denicoff, M. Hillig, J.C. Birchall, Clinical administration of microneedles: skin puncture, pain and
D.G. Poplack, J.A. O’Shaughnessy, Pharmacokinetics of taxol and doxorubicin ad- sensation, Biomed. Microdev. 11 (2009) 35–47.
ministered alone and in combination by continuous 72-hour infusion, J. Natl. [48] C.S. Kolli, A.K. Banga, Characterization of solid maltose microneedles and their use
Cancer Inst. 86 (1994) 143–145. for transdermal delivery, Pharm. Res. 25 (2008) 104–113.
[37] J.W. Lee, J.-H. Park, M.R. Prausnitz, Dissolving microneedles for transdermal drug [49] S.S.S. Lanke, C.S. Kolli, J.G. Strom, A.K. Banga, Enhanced transdermal delivery of
delivery, Biomaterials 29 (2008) 2113–2124. low molecular weight heparin by barrier perturbation, Int. J. Pharm. 365 (2009)
[38] E. Larrañeta, R.E.M. Lutton, A.J. Brady, E.M. Vicente-Pérez, A.D. Woolfson, 26–33, http://dx.doi.org/10.1016/j.ijpharm.2008.08.028.
R.R.S. Thakur, R.F. Donnelly, Microwave-assisted preparation of hydrogel-forming [50] M. Gujjar, A.K. Banga, Iontophoretic and microneedle mediated transdermal de-
microneedle arrays for transdermal drug delivery applications, Macromol. Mater. livery of glycopyrrolate, Pharmaceutics 6 (2014) 663–671, http://dx.doi.org/10.
Eng. 300 (2015) 586–595, http://dx.doi.org/10.1002/mame.201500016. 3390/pharmaceutics6040663.

103

You might also like