You are on page 1of 16

Research Article

www.acsami.org

Double-Walled Microparticles-Embedded Self-Cross-Linked,


Injectable, and Antibacterial Hydrogel for Controlled and Sustained
Release of Chemotherapeutic Agents
Pooya Davoodi,† Wei Cheng Ng,‡ Wei Cheng Yan,† Madapusi P. Srinivasan,†,§ and Chi-Hwa Wang*,†

Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585

NUS Environmental Research Institute, National University of Singapore, 1 Create Way, Create Tower #15-02, Singapore 138602
§
Civil, Environmental and Chemical Engineering, RMIT University, GPO Box 2476, Melbourne Victoria 3001, Australia
*
S Supporting Information

ABSTRACT: First-line cancer chemotherapy has been


prescribed for patients suffered from cancers for many years.
However, conventional chemotherapy provides a high
parenteral dosage of anticancer drugs over a short period,
which may cause serious toxicities and detrimental side effects
in healthy tissues. This study aims to develop a new drug
delivery system (DDS) composed of double-walled micro-
particles and an injectable hydrogel for localized dual-agent
drug delivery to tumors. The uniform double-walled micro-
particles loaded with cisplatin (Cis-DDP) and paclitaxel
(PTX) were fabricated via coaxial electrohydrodynamic
atomization (CEHDA) technique and subsequently were embedded into injectable alginate-branched polyethylenimine. The
findings show the uniqueness of CEHDA technique for simply swapping the place of drugs to achieve a parallel or a sequential
release profile. This study also presents the simulation of CEHDA technique using computational fluid dynamics (CFD) that will
help in the optimization of CEHDA’s operating conditions prior to large-scale production of microparticles. The new synthetic
hydrogel provides an additional diffusion barrier against Cis-DDP and confines premature release of drugs. In addition, the
hydrogel can provide a versatile tool for retaining particles in the tumor resected cavity during the injection after debulking
surgery and preventing surgical site infection due to its inherent antibacterial properties. Three-dimensional MDA-MB-231
(breast cancer) spheroid studies demonstrate a superior efficacy and a greater reduction in spheroid growth for drugs released
from the proposed composite formulation over a prolonged period, as compared with free drug treatment. Overall, the new
core−shell microparticles embedded into injectable hydrogel can serve as a flexible controlled release platform for modulating the
release profiles of anticancer drugs and subsequently providing a superior anticancer response.
KEYWORDS: injectable hydrogels, core−shell microparticles, antibacterial surfaces, dual-agent drug delivery, 3D tumor spheroids

1. INTRODUCTION Recently, injectable in situ-gelling hydrogels have gained


more attention due their superior advantages compared with
Hydrogels are an important class of materials with unique
the implantable counterparts.9 The macroscopic dimension of
features for biomedical applications exhibiting an exceptional
preformed hydrogels is the main obstacle in the use of hydrogel
biocompatibility given their physiochemical properties and high implants in a noninvasive surgery. However, injectable
capacity for water absorption.1−3 In addition, hydrogels can hydrogels can facilitate the administration and protection of
provide a 3-dimentional microstructure, which closely mimics bioactive molecules or cells via simple and noninvasive
natural extracellular matrix of cells for tissue regeneration.4−6 approaches.10−12 The use of injectable hydrogels not only
The mechanical and physiochemical characteristics of hydrogels enhances patient compliance but also reduces recovery time,
can be readily modified for clinical practices. For instance, the risk of infection,13,14 and treatment costs.15 Although the highly
permeability of hydrogels can be readily manipulated through porous structure and hydrophilic nature of hydrogels benefit
the control of pore size and degradation rate, where an optimal their widespread applications for tissue engineering, premature
porous structure benefits the transport of medications and release of entrapped molecules (e.g., anticancer drugs) from
nutrients inside the gel network.7 Leveraging these fascinating hydrogel confines their utilization as drug delivery vectors. Bulk
properties, researchers have developed numerous hydrogel
systems for various clinical applications ranged from drug Received: March 11, 2016
delivery and tissue engineering to diagnostics and coating of Accepted: August 17, 2016
medical implants.8 Published: August 17, 2016

© 2016 American Chemical Society 22785 DOI: 10.1021/acsami.6b03041


ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

modification of hydrogels (e.g., increasing cross-link density) degree of swelling and degradation rate, antibacterial effects,
can partially diminish this limitation, while it may have adverse cellular cytotoxicity, and its impact on the release kinetics of
effects on biocompatibility and mechanical properties of the both agents were investigated and compared with the drug
hydrogel.16−18 On the other hand, a composite hydrogel system release kinetics achieved by microparticles alone.
with the immobilized hydrophobic depots inside a hydrophilic
gel matrix may overcome the physiochemical limitations of 2. MATERIALS AND METHODS
hydrogels for the encapsulation and controlled release of small
2.1. Materials. Poly(lactic-co-glycolic acid) (PLGA 50:50, inherent
molecules.19,20 viscosity in hexafluoroisopropanol 0.55−0.75 dL g−1) and poly(D,L-
Over the past two decades, polymeric microparticles loaded lactide) (PDLLA, inherent viscosity in hexafluoroisopropanol 0.55−
with different medications for local drug delivery have 0.75 dL g−1) were obtained from LACTEL Absorbable Polymers
demonstrated versatile features in the treatment of chronic (DURECT Corporation, USA). Sodium alginate (medium viscosity of
disease (e.g., cancer).21−24 Microparticles can locally release 3500 cps for a 2% in H2O), branched polyethylenimine (25 kDa),
their payload at tumor site, which significantly reduces sodium metaperiodate (NaIO4, ≥99.0%), and picrylsulfonic acid (5%
detrimental systemic side-effects and improves patients’ in H2O) were purchased from Sigma-Aldrich (Singapore). Paclitaxel
satisfaction and compliance. However, the use of single agent was supplied courtesy of Bristol-Meyers Squibb (New Brunswick, NJ)
therapy often fails to achieve complete cancer remission due to and cis-diamminedichloroplatinum(II) (Cis-DDP, ≥99.99%) was
obtained from Sigma-Aldrich (Singapore). HPLC-grade dichloro-
rapid development of drug resistance in tumor cells. Recently, methane (DCM) (TEDIA, Fairfield, OH, USA) and phosphate buffer
combination chemotherapy has opened a new avenue for the saline (10 × PBS) were used for the fabrication of microparticles and
treatment of drug-resistant tumor cells.25,26 The use of multiple in vitro release tests, respectively. Dulbecco’s modified eagle medium
drugs with different mechanisms of action has exhibited less (DMEM), fetal bovine serum (FBS), penicillin-streptomycin, and
cancer adaptation process (i.e., development of drug resistance trypsin-EDTA (0.25%) were purchased from Life Technology
through abnormal altered drug transport) and remarkable (Singapore). All other materials and reagents used were of analytical
therapeutic outcome through synergistic effects against cancer grade and used without further purification.
cells. Therefore, in addition to encapsulating chemotherapeutic 2.2. Fabrication of Double-Layered Microparticles. The
fabrication of double-layered microparticles comprising of PDLLA
agents, microparticles should be engineered for the controlled
(core) and PLGA (shell) compartments, and loaded with drugs, were
release of multiple agents at target tissues with optimal release accomplished using CEHDA technique, as reported earlier.32 Briefly,
rates.27−29 PDLLA and PLGA (8 wt.%, in DCM) solutions were prepared
Microparticle-based local drug delivery is a promising overnight and mixed properly with Cis-DDP drug solution, where Cis-
technique to reduce the risk of recurrence for cancer survivors DDP was dissolved in water or dimethyl sulfoxide (DMSO) or
after a segmental resection. Drug loaded microparticles can dimethylformamide (DMF). PTX was directly dissolved in polymer
provide a high local concentration gradient of drugs within 2 solution at a predetermined weight ratio. The polymer/drug solutions
cm of a resected tumor and target tumor cells remained after a were subsequently pumped through a cocentric metal nozzle at
surgery. However, retaining the particles (in suspension) in the predetermined flow rates for the core (0.5 mL h−1) and shell (2.5 mL
h−1) streams, respectively. The solutions formed a liquid cone with a
surgical cavity during the injection and treatment period is still thin emerging jet at the tip of the nozzle due to increase of potential
a major challenge confining the wide use of such a delivery difference between the nozzle and grounded substrate using a high
system.19,20 Therefore, a 3D biodegradable matrix, which can voltage generator (Glassman High Voltage Inc., NJ, USA). The
efficiently entrap and retain microparticles in the cavity is polymer jet breakup occurred owing to accumulation of positive
strongly needed,30 participate in regulating of release profiles,31 charges at liquid−air interface and formed monodispersed micro-
and fill the cavity after tumor resection. An in situ-gelling droplets. Solid polymeric particles were collected on the negatively
hydrogel would be the best candidate that can play all the charged collectors after quick evaporation of DCM from droplets.
aforementioned roles successfully. The pre-encapsulation of Lastly, microparticles were freeze-dried for 2 days to fully eliminate
residual DCM from samples and stored at −20 °C for characterization
therapeutic medications into microparticulate structures and
and in vitro experiments.
the subsequent entrapment of the depots inside a hydrogel To have a better understanding of the phenomena that occurred
system can elevate drug loading and delivery duration in a during particle fabrication, computational fluid dynamic (CFD) study
controlled manner, while immobilizing delivery vectors at a was performed in the area near the nozzle tip. The equations used in
target site. CFD simulation were introduced in Supporting Information (Table
We have reported that double-walled microparticles S1).
consisting of a polymeric core surrounded by a shell layer are 2.3. Aldehyde Functionalized Sodium Alginate. The aldehyde
able to concurrently encapsulate hydrophilic and hydrophobic functionalization of sodium alginate was performed as previously
drugs and release them over prolonged period in a controlled described elsewhere.34 Briefly, sodium alginate (5.0 g) was dispersed in
25 mL of ethanol. Sodium metaperiodate (2.5 g) was dissolved in 25
manner.32,33 In this study, a novel drug delivery system mL of ultrapure water and was added dropwise to the suspension of
composed of microparticles and an injectable hydrogel is alginate/ethanol. The reaction mixture was then stirred magnetically
introduced to address the shortcomings mentioned above. We for 6, 12, and 18 h at room temperature in dark. The solution was then
employed coaxial electrohydrodynamic atomization (CEHDA) transferred into tubular dialysis membrane (MWCO = 1000 Da,
for the fabrication of double-walled microparticles loaded with Spectrum Laboratories, Inc.), where it was extensively dialyzed against
Cis-DDP and PTX in the core and shell compartments, deionized-water (5 L). A minimum of five dialysis cycles were
respectively. Particles were examined for drug loading, performed prior to lyophilization of the solution.
encapsulation efficiency, and their ability for controlled release 2.4. Synthesis of in Situ Gelling Hydrogels and the
Microcomposite. To prepare Alg-Ald (3 w/v %) and bPEI (1%,
of the therapeutic agents. The interior geometry of the particles 2.5%, 5%, and 10 w/v %) solutions, the chemicals were separately
was also examined using laser scanning confocal microscopy. dissolved in ultrapure water and the pH of the solutions was adjusted
Next, these particles were entrapped inside an in situ-gelling at ∼7.5 using hydrochloric acid (HCl, 32.5%, VWR International).
carbohydrate-based hydrogel to form a microcomposite Hydrogels were formed by mixing an equal volume (150 μL) of Alg-
structure. The hydrogel physiochemical properties such as Ald and bPEI solutions. The hydrogel was allowed to reach to stable

22786 DOI: 10.1021/acsami.6b03041


ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

form for 30 min at 37 °C. For microparticles loaded hydrogels, Waters 2414 Reflective Index detector at constant temperature (35
microparticles at the final 25%, 50%, and 75% of dried-gel weight were °C). Water was used as mobile phase at a flow rate of 1.0 mL min−1.
suspended in Alg-Ald solution and quickly mixed with bPEI solution Sample injection was set at 20 μL. A set of five polyethylene glycol
to form hydrogel composites. The hydrogel is formed upon mixing (PEG) solution with standard molecular weights was used for
bPEI (1%) and Alg-Ald (3%) solutions (short gelation time, ∼2−3 s). molecular weight calculation.
However, the gelation time becomes longer as bPEI concentration 2.5.6. Elemental Analysis. The percentage estimation of three key
increases, where mixing bPEI (10%) and Alg-Ald (3%) solutions does elements (i.e., carbon (C), hydrogen (H), and nitrogen (N)) in the
not produce obvious hydrogel pellet (the viscosity of the mixture hydrogel samples were measured using Elementar vario MICRO cube
increases, but no obvious hydrogel is formed). analyzer. All samples were freeze-dried for 48 h prior to analysis.
2.5. Physicochemical Characterization. 2.5.1. Morphology, 2.5.7. Thermogravimetric Analysis (TGA). TGA was performed to
Particle Size, and Uniformity. The surface morphology and size investigate the thermal profile of intermediates and final hydrogel
distribution of microparticles were examined via Scanning Electron products, where bare sodium alginate was used as control sample.
Microscopy (SEM) (JEOL JSM 5600LV, Tokyo, Japan) with an Samples of 5−15 mg were prepared and freeze-dried overnight and
accelerating voltage of 10 kV. The microparticles were collected on the subsequently loaded in thermogravimetric analyzer (DTG-60 AH,
surface of a double-side adhesive carbon tape and subsequently coated Simultaneous DTA-TG apparatus, Shimadzu) furnace equipped with
with a thin layer of platinum using a JFC-1300 Platinum Coater TA-60WS thermal analyzer device. The samples were heated up to 600
(JEOL, Tokyo, Japan) at 30 mA for 50 s prior to analysis. °C under dry nitrogen gas at a flow rate of 10 mL min−1 and the
The inner morphology of microparticles was observed using measurements were conducted at the scanning speed of 10 °C min−1.
Confocal Laser Scanning Microscopy (CLSM) (Olympus FluoView The weight loss pattern of each sample was recorded simultaneously
FV1000, Olympus America Inc., USA), where coumarin 6 (Sigma- during the analysis.
Aldrich, Singapore), a green fluorescence dye, was mixed with the 2.5.8. Differential Scanning Calorimetry. The thermal properties
polymer solution of the inner nozzle prior to fabrication of of intermediates Alg-Ald and final hydrogel products and the physical
microparticles. state of the drug inside microparticles were determined using
2.5.2. Drug Loading and Encapsulation Efficiency. The drug METTLER TOLEDO 822e (Greifensee, Switzerland) equipped with
loading and encapsulation efficiency were determined by dissolving 15 a gas controller (TSO800GC1) and controlled by STARe Default DB
mg of microparticles in 2 mL DCM and were allowed to stand until V9.10-STARe software. Approximately 5−10 mg of the microparticles
complete disappearance of the microparticles. Then, 5 mL 1 × PBS were sealed in covered aluminum pans. A standard empty pan was
was added to the previous solution and the mixture was incubated at used as a reference for all analyses. Samples were tested over the
37 °C overnight. The concentrations of Cis-DDP and PTX were temperature range from −10 to 110 °C at a heating rate of 10 °C
determined using inductively coupled plasma optical emission min−1. The equipment was purged with nitrogen gas at a rate of 50
spectrometry (ICP-OES) and high-performance liquid chromatog- mL·min−1 and a liquid nitrogen stream was employed as coolant.
raphy (HPLC), respectively. The HPLC apparatus was equipped with Blank microparticles were used as control in all thermal analysis tests.
a C18 reverse-phase (RP) column and UV-2487 UV-detector where 2.5.9. Determination of Swelling Ratio. The samples were
acetonitrile: H2PO4 buffer (50:50 v/v) at 1.0 mL min−1 was utilized as prepared as described above and were freeze-dried overnight. A
mobile phase. known weight of dry gels was soaked in ultrapure water at room
2.5.3. Determination of the Degree of Cross-Linking. The cross- temperature. At predetermined intervals, the excessive surface water
linking degree of the hydrogel (Ald-Alg: bPEI of 3:1 w/w %) at was removed using filter paper. After weighting the samples, the
different pH was determined using picrylsulfonic acid (2,4,6- swelling ratio was calculated using the equation
trinitrobenzenesulfonic acid (TNBSA)) reagent, known as a rapid
and sensitive technique for quantitating unreacted primary amines. SR(%) = (Wi , st /Wi , s0) × 100 (1)
After the formation of hydrogels, the samples were centrifuged and 10
μL of each supernatant was diluted with 90 μL of 0.1 M sodium
bicarbonate (pH ∼8.5) before being transferred (25 μL) into 96-well where Wi,st, and Wi,s0 were the weight of sample i at time t and the dry
plate. Next, 50 μL of freshly prepared TNBSA (0.01 w/v %) reagent sample, respectively.
was introduced to the diluted samples and incubated at 37 °C for 2 h, 2.5.10. Bulk Degradation of Hydrogels. The hydrogels were
required for the completion of the reaction. The reaction was freshly prepared and placed in Eppendorf test-tubes filled with 1.5 mL
terminated after adding 25 μL of 10% sodium dodecyl sulfate (SDS) 1 × PBS solution. The samples were incubated at 37 °C and 240 rpm
and 12.5 μL of 1 N HCl to each sample. The absorbance of the and the degradation rates were determined at predetermined time
mixtures was measured at a wavelength of 335 nm using Infinite 200 intervals. The samples were lyophilized at indicated time-points and
PRO microplate reader (TeCAN, Switzerland). The amount of accurately weighted to calculate the weight loss for each sample.
unreacted bPEI was calculated after comparing the results with a 2.5.11. Rheological Measurement. The rheological properties of
standard curve of serially diluted pure bPEI solutions. bPEI (1%)/Alg-Ald (3%) hydrogel was investigated using AR-G2
2.5.4. Fourier Transform Infrared Spectroscopy (FTIR) and Solid- Stress Controlled Rheometer (TA Instruments, U.S.A.). Stress sweep
State 13C NMR. Fourier transform infrared spectrum of Alg-Alds and and frequency sweep measurements were carried out at 37 °C using a
final hydrogel products were recorded using Bio-Rad FTS-3500ARX at parallel plate configuration (diameter, 20 mm) with a gap of 500 μm.
transmittance mode in the range of 400 cm−1 to 4000 cm−1. Potassium The storage (elastic) modulus (G′) and the loss of modulus (G″) of
bromide (KBr) pellets were used for the preparation of samples. A the hydrogel were measured as functions of oscillatory pressure (0.5−
bare KBr pellet was considered as background, where its 1000 Pa, 1.0 Hz) and angular frequency (∼0.6−600 rad s−1, 0.1 kPa).
corresponding spectrum was subtracted from that of samples. All All samples were tested ∼1 h after preparation.
samples were purged by nitrogen atmosphere for 15 min before 2.6. Bacteria Culture. The antibacterial property of the synthetic
recording their absorbance. The chemical structure of the hydrogel hydrogels was examined against Escherichia coli (E. coli, Gram
and the formation of Schiff’s base chemical bonds were determined negative) and Bacillus pumilus (B. pumilus, Gram positive) bacteria.
using solid-state 13C-nuclear magnetic resonance (13C NMR) Prior to the experiments, the bacteria were cultivated in a streaked
spectroscopy (BRUKER 400 MHz, equipped with a 4 mm MAS agar-plate to form distinct colonies overnight. A single colony was
BB-BB probe) at room temperature (296.1 K). The frequency and the harvested and subsequently inoculated in autoclaved tryptic soy broth
number of scans were set at 100 MHz and 16380, respectively. (TSB). The bacteria were allowed to grow overnight at 37 °C, shaking
2.5.5. Gel Permeation Chromatography (GPC). Relative molecular at 120 rpm to ensure that they grew at the exponential growth-phase.
weight and weight distribution of the native and oxidized alginates The bacteria concentration was monitored photometrically 600 nm
were measured by gel permeation chromatography system equipped using NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific
with a PL aquagel−OH Mixed 300 column (300 × 7.5 mm, 8u) and Inc., U.S.A). The initial optical density (O.D.) of the bacteria solution

22787 DOI: 10.1021/acsami.6b03041


ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

Figure 1. SEM micrographs of electrosprayed microparticles depicting the surface morphology and the size distribution of core-sell microparticles:
core flow rate = 0.5 mL h−1and shell flow rate = (a, a′, a″) 1.0 mL h−1, (b, b′, b″) 1.5 mL h−1, (c, c′, c″) 2.0 mL h−1, (d, d′, d″) 2.5 mL h−1.

was adjusted to ∼0.1, corresponded to concentrations ∼108 CFU was introduced on top of the gels in the transwell inserts. A transwell
ml−1. insert and bacteria solution without hydrogel were employed as
2.6.1. Concentration-Dependent Antibacterial Activity. Hydrogels negative controls. All samples were incubated at 37 °C with
used for the antibacterial experiments were freshly prepared by mixing continuous shaking at 120 rpm for 18 h. The number of colony-
an equal volume of bPEI solutions (100 μL) at different concentration forming units (CFUs) was determined and compared with control
with 100 μL of Alg-Ald (3 wt %, MMw, 6 h oxidation) solution. The after 12 h incubation at 37 °C.
hydrogels were then transferred to a 96-well plate and incubated at 37 2.6.4. Fluorescent Imaging of Bacteria-Hydrogel Contact on
°C for 1 h before washing gently with sterilized 1 × PBS. Next, the Agar Gel. Luria broth (LB) agar gels were prepared in six-well plates
stock bacterial samples were further diluted to 104 CFU ml−1 in 1 × according to the manufacturer’s instructions. A circular piece of the
PBS, followed by introducing 100 μL to the surface of the gels. agar gel was removed by 1 mL blue pipet tip to make a cylindrical
Bacteria samples without hydrogels were employed as control. The cavity inside the gel. A freshly prepared hydrogel was transferred into
samples were then incubated at 37 °C with constant shaking at 120 the cavity and incubated at 37 °C for 30 min, after which the surface of
rpm for 18 h, after which 10 μL of the supernatant was serially diluted. sample and agar gel were washed with 1 × PBS twice. A volume of
The number of colony-forming units (CFUs) (on agar-plate) was diluted bacteria solution (400 μL) in 1 × PBS (E. coli, 104 CFU ml−1)
determined and compared with control after incubation at 37 °C for was added to each well and rocked to uniformly cover the surfaces. An
12 h. agar gel with cavity and without hydrogel was used as control. The
To investigate the effect of bacteria concentration on the plate was incubated at 37 °C for 18 h, followed by staining bacteria
antibacterial behavior of the hydrogel, sample 1 (S1) was incubated with green fluorescent SYTO 9 dye (Life Technologies, Singapore).
with a 90 μL of bacteria solution prepared after 1:10 serial dilutions of After 15 min, the plate was imaged using Confocal Laser Scanning
a concentrated stock of bacteria (108 CFU ml−1) with TSB. The O.D. Microscopy.
and the number of colony-forming units (CFUs) (on agar-plate) were 2.7. In Vitro Cytotoxicity of Hydrogels. The cytotoxicity of the
determined and compared with control as previously explained. hydrogels was evaluated using hepatocellular carcinoma (HepG2) liver
2.6.2. Time-Dependent Antibacterial Activity. The killing kinetic cancer and MDA-MB-231 triple negative breast cancer cell lines, Hela,
of the hydrogel was determined via monitoring the concentration of MRC5, NIH3T3 fibroblast, and smooth muscle cell. The cells were
bacteria solutions at indicated intervals. Briefly, the freshly prepared harvested from 25 cm2 T-flask culture dish and seeded at a density of
hydrogels were incubated with 100 μL of bacteria solution (104 CFU ∼4 × 105 cell per well onto a 24-well plate and allowed to attach
mL−1) at 37 °C, shaking at 120 rpm. At the specific time-points, 10 μL overnight. The cells were incubated with freshly prepared hydrogels in
of the supernatant was removed, serially diluted in 1 × PBS, and 400 μL of DMEM supplemented with 10% FBS and 1% antibiotics at
placed on an agar plate surface at 37 °C. The number CFUs was 37 °C and 5% CO2. After 24 h, the cells were rinsed with 1 × PBS
counted after 12 h of incubation. twice and resuspended in 400 μL fresh growth medium supplemented
2.6.3. Assessment of Bacteria-Hydrogel Contact on Antibacterial with 80 μL of CellTiter 96 AQueous One Solution Cell Proliferation
Behavior. Fresh hydrogels (400 μL of bPEI 1.0 wt. % + 400 μL of Alg- Assay (Promega Corporation, U.S.A.). After 4 h, the absorbance of the
Ald 3 wt. %) were washed with 1 × PBS once, and transferred into solution was measured at 492 nm using TeCAN microplate reader.
transwell cell culture (Corning Transwell) inserted in a 24-well plate. Cells without hydrogels were used as control.
A volume of 400 μL of 104 CFU ml−1 bacteria (E. coli) solution in TSB To examine the long-term cytotoxic effect of hydrogels, cells
was loaded to each well and an additional 100 μL of bacteria-free TSB (MDA-MB-231) were incubated with hydrogels (bPEI(1%)/Alg-Ald

22788 DOI: 10.1021/acsami.6b03041


ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

Figure 2. Confocal micrographs illustrating coumarin 6 (green) loaded core−shell microparticles. The radially averaged fluorescence intensity
profiles depicting the spatial location of the core compartment inside microparticles (scanning intervals = 32).

(3%)) for 1, 2, and 3 days. Then, cells were washed with 1× PBS and 3. RESULTS AND DISCUSSION
stained with fluorescence-based LIVE/DEAD Cell Viability Assays
3.1. Particle Fabrication and Characterization. Coaxial
according to manufacturer’s instructions. The samples were imaged electrohydrodynamic atomization (CEHDA) technique was
using confocal laser scanning microscopy. employed in this study for the fabrication of core−shell
2.8. Three-Dimensional Spheroids for Cell Viability Assay. microparticulate structures. As seen in Figure 1, the size of
The MDA-MB-231 tumor spheroids were prepared according to microparticles increased with the increase of shell flow rate,
method reported by Friedrich et al.35 For homospheroid formation, while the morphology of the particles slightly changed from
cell suspension was diluted to a density of 5000 cells mL−1 in high- irregular-shaped microparticles to well-formed microspheres.
glucose Dulbecco’s modified Eagle’s medium (DMEM) supplemented Our previous studies demonstrated that dichloromethane
with 10% fetal bovine serum (FBS). Two hundred microliters of (DCM) was a suitable candidate for dissolution of PLGA and
diluted cells was added to wells of 96-well plate precoated with 50 μL PDLLA and fabrication of microparticles using CEHDA
agarose gel (1 wt. %). The spheroids formed after 5 days and technique. In this study, a mixture of DCM and dimethyl
sulfoxide (DMSO) (5:1 v/v) was used for the core solution due
subsequently were incubated with freshly prepared microparticles/
to the higher solubility of Cis-DDP in DMSO compared to
hydrogel delivery systems (day 0). The size and morphology of the 3D
aqueous solution. The presence of DMSO in the core solution
tumor spheroids were monitored at predetermined intervals and led to the formation of microparticles with smoother surfaces.
compared with control groups. However, it adversely affected the morphology of the particles,
2.9. Statistical Analysis. All experiments have been conducted in particularly at a lower shell flow rate (1.0 mL h−1).
triplicate or more per condition. The statistical analysis (student t-test) DCM has a higher vapor pressure compared with common
was performed, where p < 0.05 and p < 0.01 were considered organic solvents such as DMSO and dimethylformamide
statistically significant. (DMF). Therefore, microdroplets can quickly form and solidify
22789 DOI: 10.1021/acsami.6b03041
ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

Figure 3. CFD simulation results: (a) Taylor cone-jet and droplet formation processes (core, PLA solution; shell, PLGA solution; surrounding fluid,
air), (b) electric potential distribution, electric field strength distribution, charge density distribution, and (c) velocity vector. Operating conditions:
Voltage (5600 V), shell:core flow rate (2.5:0.5 mL h−1).

Figure 4. Schematic representation of the preparation of injectable hydrogel composed of alginate aldehyde (Alg-Ald) and PEI-25k.

after breaking-up of the polymer jet at the tip of the nozzle and morphology of particles produced. Solvent evaporation rate
flying toward the collecting substrate. Although the fast affects polymer diffusion, which plays a determinant role in
evaporation of organic solvents is crucial to fabricate micro- morphology of final products. A greater particle size and higher
particles with uniform size distribution, it can affect the surface surface roughness are the major impacts of using a highly
22790 DOI: 10.1021/acsami.6b03041
ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

volatile solvent. The fast solvent evaporation usually confines was preferably preformed in a heterogeneous medium
the interaction and rearrangement of polymer chains inside a composed of ethanol:water (1:1 v/v). As reported by
droplet flying toward the oppositely charged substrate. In Balakrishnan and Jayakrishnan,34 the oxidation degree of
addition, the fast evaporation of the solvent may cause a highly alginate is associated with the quantity of periodate in the
porous surface, which can influence the release rate at a later reaction medium and the duration of the reaction. It means that
stage. The presence of DMSO reduces the volatility of DCM a longer reaction produces more aldehyde groups ready for
and allows polymer chains to rearrange and form a less porous reacting with cross-linkers at a later stage. However, an
surface during the solidification step. However, the micro- extensive depolymerization happens during the oxidation
particles may reach the substrate before being fully dried, where reaction (Table 1), which may adversely affect the formation
the particle’s shape may further change at this stage.
As seen in Figure 1, the size of the particles increased as the Table 1. Gel Permeation Chromatography (GPC) Results of
shell flow rate changed from 1.0 to 2.5 mL h−1. However, the Sodium Alginate and Alg-Ald
formation of secondary droplets at high flow rate (i.e., 2.5 mL
Mw (g mol−1)
h−1) adversely affected the size distribution of the particles. The item Mn PDI
size of microparticles is correlated with the flow rates of the sodium alginate (MMw) 207 200 223 000 1.08
core and shell solutions. Therefore, the flow rates must be sodium alginate (HMw) 259 500 270 300 1.04
carefully chosen to allow complete solvent evaporation from MMw Alg-Ald −6h 134 000 162 600 1.21
droplets before reaching the collector. HMw Alg-Ald −12 h 141 100 166 300 1.18
The core−shell structure of microparticles was confirmed HMw Alg-Ald −18 h 126 100 156 600 1.24
using laser scanning confocal images, where the fluorescent dye
(coumarin 6) was mixed with the core polymer solution prior and stability of hydrogels. Depolymerization reaction can be
to fabrication process. From the confocal image, the core attributed to the formation of 1-hydroxyethyl radicals produced
compartment was completely surrounded by the shell matrix, as a side product during the desired reaction.36
where the fluorescence intensity profiles showed that the shell The hydrogel was formed upon mixing of Alg-Ald solution
layer was uniformly distributed around the core (Figure 2). with PEI-25k solution in deionized water pH ∼7.5. PEI-25k
However, due to the diffusion and dissolution of coumarin 6 chains act as a cross-linker where their ε-amino groups reacted
into the shell solution containing DCM, the estimation of shell with aldehyde groups of oxidized alginate via Schiff’s base
thickness (∼8−10 μm) using the confocal image may not be so formation at physiological temperature. The concentration of
accurate. Alg-Ald solution was kept constant at 3 wt. % throughout the
3.2. CFD Simulation of CEHDA Process. To observe the experiments, while the PEI concentration was changed from 1
Taylor cone-jet mode and droplet formation, which dominate to 10 wt. %. The time required for hydrogel formation showed
the formation of core−shell microparticles, a CFD simulation a PEI concentration dependent trend. The time was quite short
was carried out near the tip of nozzle. Figure 3 illustrates the when the PEI concentration was 1 wt. %, while no obvious gel
CFD simulation results involving Taylor cone-jet and droplet network was formed at PEI concentrations above 5 wt. %, even
formation processes, electric potential distribution, electric field after 15 min. This was due to the change of primary amines
strength distributions, charge density, and velocity vector. Two concentrations available to react with aldehyde (RCHO)
fluid meniscus were set at the tip outlets of the nozzle as initial groups. At low PEI concentrations (>5 wt. %), primary amines
conditions for both streams. As time proceeded, the fluids were of one PEI molecule react with a number of RCHO groups
elongated along the axis and formed a cone-jet due to the force from different alginate chains. Therefore, it can make a dense
balance among the gravity, surface tension, electric stress, and cross-linked network. However, as PEI concentration increases,
viscous stress. Finally, the jet broke into droplets and formed the probability of the reactions between amines of one PEI
core−shell droplets with the size of ∼30 μm. The final particle molecule and RCHO decreases. It can be attributed to the
size (∼13 μm) can be predicted from droplet size by eq S8, physical mixing ability of the high concentration of bPEI with
which is correlated well with the experimental result. From the Alg-Ald solution. These solutions cannot be mixed homoge-
velocity vector distributions, the motions of fluids inside the neously, where the access of RCHO groups to primary amines
Taylor-cone can be obtained. The liquids moved down along suddenly reduces. Thus, a weak polymer network with a
two vortex edges located near liquid−gas interface. At the liquid nonporous morphology is formed (Figure S1).
cone apex, the fluids moved into the jet flow, followed by jet 3.4. Chemical Characterization of Hydrogel. FTIR
breaking up and droplet formation. The results also showed spectra of sodium alginate, oxidized alginate, and composite
that the highest potential appeared at the tip and liquid cone, hydrogel are shown in Figure 5a. The spectrum of sodium
while the highest electric field strength was located at the gas− alginate showed characteristic absorption bands around 945
liquid interface near the cone apex. This phenomenon can be (C−O stretching), 1029 (C−O−C stretching), 1130 (C−C
attributed to the presence of induced charges at the surface of stretching), and 1321 cm−1 (C−O stretching). The strong
liquid cone, especially near the cone apex as confirmed by the bands at 1612 and 1413 cm−1 are associated with asymmetric
results of charge density distribution. and symmetric stretching bands of the carboxylate salt groups.
3.3. Alginate-Aldehyde and Hydrogel Formation. The The appearance of aldehyde symmetric band at 1724 cm−1
synthetic procedure of injectable hydrogel was illustrated in confirmed the formation of alginate-aldehyde due to reaction
Figure 4. Three kinds of alginate-aldehyde of different with sodium meta-periodate. However, hemiacetal formation of
molecular weight were synthesized using sodium metaperiodate free aldehydes, in some cases, may reduce the intensity of this
for various duration. Periodate molecules target the α-glycol band. As seen in Figure 5b, the aldehyde symmetric band at
groups of polysaccharides and form their dialdehyde derivatives. 1724 cm−1 disappeared upon the mixing of alginate-aldehyde
As the aqueous solution of alginate forms a highly viscous and PEI and formation of hydrogel. Therefore, the absence of
solution even at low concentrations, the oxidation of alginate the band in hydrogel spectrum clearly indicated the formation
22791 DOI: 10.1021/acsami.6b03041
ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

Figure 5. Chemical characterization of synthetic hydrogels. (a and b) FTIR spectra of the hydrogel formed after the mixing of alginate-aldehyde with
PEI at different concentrations. (c) Solid-state 13C NMR spectrum of hydrogel. (d) Thermogravimetric analysis of bare sodium alginate, oxidized
alginate, and synthetic hydrogel. (e) DSC curves of bare sodium alginate, oxidized alginate, and synthetic hydrogel.

of Schiff’s base (i.e., CN chemical bond) between −CHO degradation steps, where the first one occurred ∼100 °C. The
and −NH2 groups. Figure 5c presents the solid-state 13C NMR first step of mass loss corresponded to the elimination of free
spectrum of the hydrogel upon mixing alginate aldehyde and water and bound water from the sample. The second step
PEI-25k solutions at room temperature. Compared to bare occurred ∼220 °C that can be attributed to the decomposition
sodium alginate spectrum shown in Figure S2, the peaks of alginate at C−H and C−O−C bonds in the main
appeared at ∼35−60 ppm and 160−163 ppm are assigned to polysaccharide chain and vaporization and elimination of
PEI carbons and Schiff base carbon (−HCNH−), volatile products. The third step was associated with the
respectively. The presence of −HCNH− peak in the formation of Na2CO3 and other carbonized materials. The
hydrogel 13C NMR spectrum is in a good agreement with the thermogram of alginate-aldehyde, however, showed a few
results of FTIR analysis, where the aldehyde peak appeared and differences in the peak area and position as compared with bare
disappeared upon the oxidation of alginate molecules with alginate. The first degradation step of alginate-aldehyde was
NaIO4 and formation of the hydrogel, respectively. faster than that of alginate. The variation on the peak area and
The elemental analysis (EA) results are presented in Table position during the first step of mass loss was corresponded to
S2, where the molar ratio of carbon/nitrogen (nC/nN) is water molecule interactions with polymer chains and the lower
calculated from the weight percentage of both components. capacity of alginate-aldehyde for holding of water. In addition,
Because of constant carbon:nitrogen ratio in each PEI unit the second degradation step of alginate-aldehyde exhibited a
(2C:1N) and alginate unit (6C:0N), the grafting rate can maximum peak at ∼200 °C, which was slightly lower than the
subsequently be calculated using eq S9. Using this equation, α maximum peak (∼220 °C) of alginate thermogram. As the
= 0.003 for PEI (1%)/Alg-Ald (3%) is the maximum grafting stability of the samples is directly correlated with their
ratio. decomposition behavior, the results demonstrated a significant
The data provided by TGA analysis of sodium alginate, decrease in the thermal stability of alginate-aldehyde compared
alginate aldehyde, and the hydrogel were presented in Figure to pure alginate. In the case of hydrogel, the results
5d and Figure S3. The thermograms showed different thermal demonstrated a two-step mass loss similar to that of alginate-
behavior of the samples. Sodium alginate exhibited three aldehyde, followed by a gradual mass loss from ∼220 °C
22792 DOI: 10.1021/acsami.6b03041
ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

Figure 6. Swelling ratios of different hydrogels as a function of time: (a) hydrogels prepared at different Ald-Alg: PEI weight ratios and (b) hydrogel
prepared at Ald-Alg: PEI of 3:1 w/w %, where high molecular weight alginate was oxidized by NaIO4 for 12 and 18 h. Medium molecular weight
alginate was oxidized for 6 h using the same process. (c) The swelling ratios of hydrogels at different pH values. (d) PEI release percentage during 5
days at different pH values (hydrogel: Ald-Alg: bPEI of 3:1 w/w %). All data are expressed as the average ± standard deviation of three independent
experiments. (The deswelling behavior of the hydrogel is reported in Supporting Information).

onward. The new peak appeared at ∼440 °C might be due to the particles could not be uniformly dispersed at high (∼75 w/
the decomposition of the materials and was in agreement with w %) concentration.
results (i.e., a less thermal stability for biopolymeric Schiff based 3.6. Physical Characterization of Hydrogel. The
materials) of previous study.37 swelling ratio (SR) of hydrogels with different compositions
The data provided by DSC curves (Figure 5e) were in was measured in 1× PBS buffer at different pH values and 37
agreement with TGA results. The DSC curves showed that °C (Figure 6). The hydrogels generally reached to a state of
dehydration process of pure alginate occurred at a lower swelling equilibrium within half an hour, although average
temperature as compared with other samples. The exothermic quantities were slightly fluctuating around the equilibrium
peaks ∼200 and 220 °C can be attributed to decomposition of ratios. As illustrated in Figure 6a, the SR of the hydrogels
the samples, where a broad peak in this area was associated with increased significantly with the decrease of PEI concentration
a gradual decomposition of hydrogel due to temperature used for hydrogel preparation. However, the SR of hydrogels
increase. cross-linked by PEI at concentrations of 1.0 and 5.0 wt. %
3.5. Preparation of Microparticles Embedded Hydro- fluctuated around 1800% of their initial dry weights and
gel. Figure S4 shows the morphological images of core−shell remained almost constant for 12 h.
microparticles embedded in Alg-Ald/PEI hydrogel networks. The water absorption in the hydrogels is a function many of
Microparticles were dispersed in Alg-Ald (3 wt. %) solution and variables such as cross-link density, hydrogel microstructure
(porous or poreless), network parameters, nature of the
PEI-25k (1 wt. %) solution was subsequently added to form the
materials, etc. For instance, the cross-link density determines
hydrogel matrix. Figure S1a shows the morphology of the
the distance between two cross-link nodes on a single polymer
hydrogel prior to incorporation of microparticles. The hydrogel
chain. Therefore, a longer distance results in a lower cross-link
had a highly porous structure, which can significantly help to density. In addition, the swelling process can be controlled by
absorb large amount of water and increase its biocompatibility. diffusion and relaxation processes. The diffusion process,
The addition of microparticles (25, 50, 75 w/w %) did not identified as the rate-determining step at the beginning of the
change the morphology of hydrogel and microparticles (Figure swelling process, depends on solvent molecular weight, porosity
S4b−d). However, because of hydrophobicity of microparticles, of hydrogel, and temperature. The chain relaxation step occurs
the homogeneity of final product is affected by the increase of slower than diffusion process at low cross-link density.
particle weight percentage. This conclusion was further proved Therefore, as explained by Omidian and Park,38 the chain
using higher magnification SEM micrographs for the particles relaxation step is considerably slower than diffusion process at a
embedded hydrogels at 50 and 75 w/w % (Figure S4e and f). high cross-link density, which results in a sharp increase in SR,
The dispersion of microparticles (Figure S4g) within the followed by an equilibrium step.
hydrogel was further examined using fluorescence dye-loaded The effect of alginate molecular weight on SR was
microparticles and corresponding fluorescence intensity profiles investigated as shown in Figure 6b. The concentration of PEI
(Figure S4h). Although the intensity profiles correlated well solutions was kept constant at 1 wt. % for all experiments. As
with the concentration of microparticles, a low light intensity can be seen, the SR quickly reached an equilibrium condition
was observed near the gel surface in tube (iv), which means that for hydrogel prepared using medium molecular weight alginate
22793 DOI: 10.1021/acsami.6b03041
ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

(MMw 6), while it was gradually increasing for high molecular the cross-linked hydrogels is shown in Figure 8a. Over the
weight alginate oxidized for 12 and 18 h. However, the results linear range, the applied stress results in strain. However,
showed that SR could slightly decrease when the oxidation beyond LVR, G′ (elastic modulus) sharply decreases due to the
process was longer (i.e., 18 h). The longer the oxidation time, structure breakdown upon increasing the deformation imposed.
the more aldehyde groups for cross-linking process. As a result, Therefore, the pressure (∼200 Pa) at which the hydrogel
the higher cross-link density confined the polymer relaxation network begins its nonlinear viscoelastic behavior was
steps and reduced SR. On the other hand, longer oxidation determined as critical shear stress. As the linear part happened
period caused polymer decomposition and reduced molecular over the range of 0.5 to 200 Pa (Figure 8a), the frequency
weight. The lower the molecular weight, the more the sweep experiment was subsequently conducted at 10 Pa to
compacted gel network is and consequently the lower SR. investigate the stability of three-dimensional cross-linked gel
The pH of the aqueous medium had a marginal effect on SR networks over a frequency range from ∼0.6 to 600 rad s−1. As
and became effect-less after ∼1 h (Figure 6c). pH can partially shown in Figure 8b, there is a large viscoelastic plateau between
increase the protonation of amine groups available in the ∼0.6 and 200 rad s−1, which is an indication of a stable hydrogel
solution, which may delay the cross-linking process and provide structure. No cross-point between G′ and G″ was also
a slightly longer relaxation period for polymer chains inside the observed, hence representing the stability of the hydrogel
gel network. These results were ratified by PEI release test at structure. In addition, the frequency independent behavior of
different pH within 5 days (Figure 6d), where the amount of elastic modulus indicates the solid-like behavior of the hydrogel.
PEI release in the first day was negligible for all pH. However, At low frequency, the polymer chains between the cross-link
the carbonyl-amine bonds showed greater sensitivity to pH less points can rearrange themselves due to comparable relaxation
than 6 in the following days. time scales, while at higher frequencies, they are impotent to
The degradation of hydrogel samples was investigated in rearrange themselves within a short period of imposed motion
vitro and the results were presented in Figure 7. Among (chain stiffening) and show solid-like behavior.
3.7. Antibacterial Activity of Hydrogels. To evaluate the
antibacterial property of the hydrogels, the surface of the
hydrogels was challenged with model Gram-negative (E. coli)
and Gram-positive (B. pumilus) bacteria. Figure 9a and 9c
demonstrate the results of antibacterial tests in which the
surface of hydrogels prepared with varying amount of PEI was
incubated to 104 CFU ml−1 of bacteria. The empty tissue
culture wells and the same amount of PEI used for the
preparation of hydrogels were considered as negative and
positive controls, respectively. The antibacterial activity of
formulations as well as controls were evaluated by the colony
Figure 7. Hydrogel degradation results. All data are expressed as the formation ability within 18 h postexperiment times. The results
average ± standard deviation of three independent experiments. showed that antibacterial activity of hydrogels increases with
the increase of PEI concentration in the formulations. On the
different hydrogels, bPEI (1%)+Alg-Ald (3%) was the most other hand, a bare 3 wt. % alginate solution show no
stable sample over 60 days, while others lost their weight antibacterial activity against both E. coli and B. pumilus, similar
considerably faster over the same period. As seen in Figure 7, to empty wells (negative controls). For instance, at 1 wt. % PEI
the initial concentration of bPEI solution strongly affects the the hydrogel showed ∼100 times greater antibacterial activity
stability of the hydrogels and degradation rates, where bPEI compared with bare alginate. The inhibitory effect of hydrogels
(10%) + Alg-Ald (3%) that did not form obvious gel was significantly higher at PEI concentration ≥2.5 wt. %, where
completely disappeared before day 30. no colony was formed after 18 h incubation on agar plates.
The dynamic mechanical behavior of bPEI (1%)/Alg-Ald These results suggest that the protonated amines in PEI
(3%) hydrogel was investigated by oscillatory rheology at 37 molecules can directly influence its antibacterial behavior.
°C. First, the stress sweep at a constant frequency was Figure 9b and 9d presented the colony formation ability of
conducted to identify the linear viscoelastic region (LVR) bacteria as a function of increasing initial CFU for a hydrogel
profiles of the hydrogel. The effect of the stress amplitude on prepared from 3 wt. % alginate+1 wt. % PEI. The data showed

Figure 8. Rheological analysis of bPEI (1%)/Alg-Ald (3%) hydrogel. (a) Stress sweep and (b) frequency sweep.

22794 DOI: 10.1021/acsami.6b03041


ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

Figure 9. Antibacterial activity of hydrogels: Alg-Ald cross-linked with different concentration of PEI-25K were incubated with 104 CFU mL−1 (a) E.
coli or (c) B. pumilus for 18 h. Hydrogel surface (Alg-Ald: PEI 3:1 w/w) was challenged with serially diluted (b) E. coli or (d) B. pumilus in PBS for
18 h. (e) The time course of bacterial (E. coli) killing experiment. (f) Contact-dependent antibacterial effect. The control experiment (purple bars)
was performed by incubating bacteria (104 CFU mL−1) without hydrogels for the corresponding time points.

that the hydrogel is highly active at low bacteria concentrations. All data presented above suggests a contact-dependent
In addition, a significant reduction in bacteria activity (∼1000 mechanism of action for antibacterial activity of the hydrogels.
times) was observed at extremely high bacteria concentration To assess this hypothesis a set of quantitative and qualitative
(108 CFU ml−1). This high concentration was ∼10 million experiments was performed in vitro. The hydrogel prepared
times greater than that is expected in a contaminated operating was loaded into a transwell tissue culture insert and the inset
was placed in the culture plate containing bacteria in 1 × PBS,
theater.39,40
where the porous membrane of the inset separated the
Figure 9e shows the time-dependent antibacterial test of the
hydrogel and bacteria. After 18 h, the effect of PEI molecules
hydrogel prepared from 1 wt. % PEI. The colony formation leached from the hydrogel on the activity of bacteria was
ability of bacteria incubated with the hydrogel at various time- determined. The results of bacteria-hydrogel contact test were
points was evaluated and the data showed a time-dependent presented in Figure 9f. As can be seen, the number of colonies
activity where the number of colonies gradually decreased as formed after the experiment was very close to that of control
the incubation period extended. groups. The data suggest that most of PEI molecules reacted
22795 DOI: 10.1021/acsami.6b03041
ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

Figure 10. Viability of bacteria (E. coli) on the surface of hydrogel. E. coli viability was imaged using confocal microscopy after 18 h of incubation on
agar gel/hydrogel interface. (a) The bright field, (b) the fluorescence micrograph of bacteria stained with SYTO 9, and (c) the overlay image of
bacteria and gels. The white dash-line indicates the interface of agar gel/hydrogel. Scale bars: 100 μm.

Figure 11. Results of in vitro biocompatibility tests for cells cocultured with hydrogels. (a) Cell viability percentage after 24 h incubation with
hydrogels at various PEI concentrations. Note: Smooth muscle cells were cultivated in smooth muscle cell medium (SMCM) supplemented with 2%
of FBS, 1% of cell growth supplement, and 1% of penicillin-streptomycin antibiotic solution. The viability percentage of (b) HepG2 and (c) MDA-
MB-231 cells after 24 h treatment with hydrogels at various PEI concentrations. (d) Live/dead cell viability staining images of MDA-MB-231 cells
after three consecutive days cocultured with hydrogel (Alg-ald:PEI 3:1 w/w %). The green color demonstrates live cells and the red color
demonstrates dead cells. Dead cells were indicated by red dash-circles. All data represent as mean ± SD of n = 4 independent experiments. Scale
bars: 200 μm.

22796 DOI: 10.1021/acsami.6b03041


ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

with Alg-ald molecules and did not leach from the hydrogel as In addition, for formulation A1 (Figure S6a), both PTX and
previously indicated in Figure 6d (PEI concentration in VRP release curves overlapped with each other in the first 10
supernatant). This mechanism was qualitatively investigated days, after which VRP was released slightly faster, while the gap
by bacteria-hydrogel contact experiment on agar plate. A between the two release curves was significantly larger in
cylindrical portion of preformed agar gel was removed and the formulation B1 (Figure S6b). Moreover, the slope of VRP
hole was filled with the hydrogels. Next, the surfaces of curve suddenly increased after 30 days, causing an even greater
hydrogel and agar gel were exposed to bacteria solution at 104 deviation from PTX curve. Therefore, VRP was released
CFU mL−1 for 18 h. Figure 10 shows both the bright field and significantly faster and in a larger dosage, making it seemed to
fluorescent image of bacteria stained with SYTO-9 fluorescent be released first while PTX was released second. These results
dye. The lack of green fluorescence observed on the area indicated that highly versatile drug carriers could be designed
occupied by the hydrogel indicated that there were no viable by simply swapping the drugs in a one-step CEHDA fabrication
bacteria on the hydrogel surface. However, bacteria could technique to present a parallel or a sequential release of drugs.
proliferate on the surrounding agar surface. On the basis of This flexibility is an advantage of the CEHDA fabrication
these results, the hydrogel is a contacted-based bactericidal technique compared to other techniques such as the emulsion/
surface, which kills adhering bacteria. solvent evaporation method.41,42 Emulsion/solvent evaporation
3.8. Biocompatibility of Hydrogels. Since biocompati- method involves a rapid stirring step that adversely promotes
bility is a crucial factor for their successful applications of the disintegration of the drugs and confines loading capacity of
synthetic biomaterials, the cytocompatibility of the hydrogels hydrophilic drugs due to prolonged contact with aqueous
was evaluated by MTS assay using six different cell lines. The environment.
cell lines were selected from different tissues, where the drug In the case of Cis-DDP and PTX loaded microparticles
delivery system proposed in this article may be used. Among (Table S4), the release behavior was considerably different
four different hydrogels tested for cytocompatibility, the from what was previously mentioned above. For the case of
hydrogel with the lowest PEI concentration (i.e., Alg-ald:PEI Cis-DDP dissolved in water, the release of Cis-DDP from both
3:1 wt. %) showed minimum toxicity and maximum cell formulation B2 and C2 (Figure 12) displayed an initial burst of
viability (Figure 11a). However, the number of live cells was approximately 40%, followed by a sustained release throughout
gradually decreased with increasing PEI concentration from 2.5 a period of 45 days in a near zero-order rate. This initial burst
to 10 wt. %. Notably, cell proliferation assay revealed that the release can be attributed to the premature release of drug
hydrogel prepared with 1 wt. % of PEI should be more stable molecules from the surface of particles or the diffusion of drugs
placed close to the surface. Furthermore, the hydrophilic nature
than other hydrogels due to formation of a well-interconnected
of Cis-DDP may contribute to higher premature release, as it is
network, which prevents the premature release of non-cross-
more readily available for release into surrounding medium. In
linked toxic PEI. A similar trend was found for the cell lines
contrast, the release of PTX from both formulation A2 and C2
incubated with medium molecular weight and high molecular
did not show significant initial burst, but just a progressive and
weight hydrogel at various PEI concentrations (Figure 11b and
sustained release throughout the study. The steady release of
c). Therefore, it can be concluded that molecular weight of the
PTX could be explained by the drug diffusion within the
oxidized alginate has no detrimental effect on cell growth and microspheres, hence giving a predictable trend. As time
proliferation. The extended cell viability test was performed for proceeded, the continuous penetration of aqueous medium
3 days. Cells were stained using LIVE/DEAD assay and caused polymer degradation, pore formation, thinning of the
observed using laser scanning confocal microscopy (Figure shell layer,43 and consequently faster release of drug into
11d). The results showed a few dead cells (red color), and cells surrounding medium.
maintained normal cell morphology for 3 days. These results As the limited dissolution of Cis-DDP in water (<2.5 mg
demonstrated that the hydrogel (i.e., Alg-ald:PEI 3:1 w/w %) is mL−1) considerably reduced drug loading efficiency, we used
an appropriate candidate for long-term drug delivery dimethyl sulfoxide (DMSO) known as a suitable solvent for
applications. Cis-DDP (>25 mg mL−1). In this case, the drug loading
3.9. In Vitro Drug Release: Microparticles and Micro- content was significantly improved (Table S4). However, a
particles-Embedded Hydrogels. The molecular weight of huge initial burst release of Cis-DDP (∼82% for formulation B3
an anticancer agent and its affinity for the carrier’s molecules and ∼78% for formulation C3) was observed within the first
are important parameters contributing to the design of a day of experiments (Figure 12d). It was due to the miscibility of
successful controlled release system. Thus, the ability of DMSO and DCM that led to the undesirable diffusion of core
microparticles for the sustained release of verapamil (VRP), solution into shell counterpart during particle fabrication, where
paclitaxel (PTX), and cisplatin (Cis-DDP) as model drugs was the effective thickness of the shell compartment markedly
examined in vitro. Table S3 presents the results of drug loading decreased. Therefore, Cis-DDP could readily pass the shell
content (DLC) and encapsulation efficiency (EE) of the barrier and dissolve into the surrounding medium.
particles. The release profiles of both formulations showed a To control the initial burst of Cis-DDP, the previously
small initial burst of ∼11% VRP and ∼12% PTX for introduced hydrogel was employed to entrap the drug-loaded
formulation A1, and ∼13% VRP and ∼4% PTX for formulation microspheres. Thus, the initial burst was significantly reduced
B1 (Figure S6). However, after an initial premature release, the to ∼40% (Figure 12d) becaue of a more complex diffusion
drugs were gradually released in a sustained manner over a pathway to be passed. The hydrogel matrix functioned as an
period of 60 days, indicating the progressive degradation of additional barrier to restrict direct exposure of microparticles to
polymeric microspheres for a sustained and long-term release. PBS medium, thereby, reducing its high initial burst as well as
In formulation A1, both drugs exhibited a near zero order making its release more sustainable over a longer period.
release rate, however, for VRP in formulation B1 (Figure S6b), As shown in Figure 12e, after preparation of composite
it showed a sharp increase in release profile after 30 days. formulation, 75 w/w % microspheres-loaded hydrogel exhibited
22797 DOI: 10.1021/acsami.6b03041
ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

a slightly lower initial burst and cumulative release as compared


to 25 w/w % and 50 w/w % composite formulations, though all
three release curves had very similar shape and gradient. The
composite formulation C3 showed similar trend for Cis-DDP,
while release rate of PTX was undoubtedly reduced (Figure
12f). Regardless of the initial burst release of Cis-DDP, the new
system showed a simultaneous release of both agents in a
controlled and sustained manner, here is highly promising for
the synergistic treatment of tumors such as triple-negative
breast cancer (TNBC).
3.10. Drug Release Effect on 3D MDA-MB-231
Spheroids. Although the traditional 2D cell-culture is suitable
for mechanism study and rigorously control the experimental
and treatment conditions, it usually provides overestimated or
underestimated results that may not be reproduced in vivo.
Therefore, a 3D tumor spheroid model is employed in the
current study as it demonstrates dose−response relationships
that may better predict tumor responses in vivo in future
studies. MDA-MB-231 spheroids in a prolonged study of 10
days were used to evaluate the efficacy of combined
formulation released from microparticles/hydrogel composite.
As shown in Figure 13, the composite formulation exerted a
greater inhibitory effect on the growth of tumor spheroids, as
compared with control and free drug groups. The free drug
treatment temporarily inhibited the growth of spheroids and
the size of the spheroids remained constant or increased within
Figure 12. Release profiles of Cis-DDP and PTX from core−shell 10 days. In contrast, the spheroids slowly responded to the
microparticles: (a) the scheme of microparticles loaded with Cis-DDP treatment of composite formulation at the early stage, while the
and PTX (formulation A2, PTX (shell), formulation B2, Cis-DDP responses were pronounced over 10 days and significantly
(core), formulation C2, PTX (shell) and Cis-DDP (core)); (b) the inhibited the growth of spheroids (Figure 13b and 13c). The
release profiles of formulation A2 and B2 (Cis-DDP was dissolved in
water); (c) the release profile of formulation C2 (Cis-DDP was
fast but short period response of spheroids to free drug
dissolved in water); (d) the initial step of Cis-DDP release profile from treatment was due to exposure with high concentration of
formulation B3 and C3 (Cis-DDP was dissolved in DMSO) with and drugs. However, to mimic the in vivo conditions, concentration
without hydrogel; (e) the release profile of formulation B3 (Cis-DDP of drugs was gradually reduced over 48 h. Therefore, the
was dissolved in DMSO) and hydrogel at 25, 50, and 75 w/w % spheroid could restore their proliferation conditions after ∼8
loading ratios; (f) the release profile of formulation C3 (Cis-DDP was days. Despite the free drug results, higher drug efficacy
dissolved in DMSO) and hydrogel at 50 w/w % loading ratio. pH of observed for spheroids incubated with the composite
PEI-25k and Alg-Ald solutions were adjusted at physiological pH (pH formulation could be attributed to the controlled and sustained
∼7.0) using hydrochloric acid (32.5%) prior to hydrogel formation. release of Cis-DDP and PTX from the new delivery systems.
Data represents average ± standard deviation of three independent
measurements, n = 3.
Although the initial drug concentrations were not sufficient to
cause significant cell death, the gradual accumulation of drug

Figure 13. (a) Bright-field images of tumor spheroids treated with combination of Cis-DDP and PTX: free drug (second column) and composite
formulation (third column) (scale bar = 200 μm). (b) Growth of tumor cell spheroids upon exposure to microparticle formulations over 10 days. (c)
Cell viability of tumor spheroids upon exposure to microparticle formulations at day 10. Data represents average ± standard deviation of three (n =
3) independent measurements.

22798 DOI: 10.1021/acsami.6b03041


ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

within the spheroid killed cells in the outer rim of the spheroid,
where it promoted penetration of drugs into the primed tumor
■ REFERENCES
(1) Park, H.; Park, K. Biocompatibility Issues of Implantable Drug
and caused a substantial cell death. Delivery Systems. Pharm. Res. 1996, 13, 1770−1776.
(2) Park, J. H.; Bae, Y. H. Hydrogels Based on Poly(ethylene oxide)
4. CONCLUSIONS and Poly(tetramethylene oxide) or Poly(dimethyl siloxane): Synthesis,
Cis-DDP and PTX loaded core−shell microparticles were Characterization, In vitro Protein Adsorption and Platelet Adhesion.
fabricated using the CEHDA techniques. A CFD simulation Biomaterials 2002, 23, 1797−1808.
(3) Slaughter, B. V.; Khurshid, S. S.; Fisher, O. Z.; Khademhosseini,
model was developed and utilized for the optimization of A.; Peppas, N. A. Hydrogels In Regenerative Medicine. Adv. Mater.
CEHDA operating parameters prior to fabrication of micro- 2009, 21, 3307−3329.
particles. The Alg-ald:PEI hydrogel was successfully synthesized (4) Zamanian, B.; Masaeli, M.; Nichol, J. W.; Khabiry, M.; Hancock,
and evaluated for entrapment of microparticles inside a porous M. J.; Bae, H.; Khademhosseini, A. Interface-directed Self-assembly of
matrix, modulating release profiles, and antibacterial activity Cell-laden Microgels. Small 2010, 6, 937−944.
against Gram-positive and Gram-negative bacterial via a (5) Zelikin, A. N.; Price, A. D.; Stadler, B. Poly(methacrylic acid)
contact-dependent mechanism. Moreover, three-dimensional Polymer Hydrogel Capsules: Drug Carriers, Sub-compartmentalized
MDA-MB-231 spheroid studies demonstrated that the Microreactors, Artificial Organelles. Small 2010, 6, 2201−2207.
combination of Cis-DDP and PTX released from the proposed (6) Thiele, J.; Ma, Y.; Bruekers, S. M.; Ma, S.; Huck, W. T. 25th
microparticles/hydrogel formulations could provide greater Anniversary Article: Designer Hydrogels for Cell Cultures: A Materials
Selection Guide. Adv. Mater. 2014, 26, 125−147.
efficacy and gradually reduce the size of spheroids. In contrast, (7) Jeon, O.; Bouhadir, K. H.; Mansour, J. M.; Alsberg, E.
the free drug treatment was impotent to effectively reduce cell Photocrosslinked Alginate Hydrogels with Tunable Biodegradation
viability in tumor spheroid in vitro. The new composite Rates and Mechanical Properties. Biomaterials 2009, 30, 2724−2734.
formulation may hold great promise for practical applications in (8) Li, H.; Kong, N.; Laver, B.; Liu, J. Hydrogels Constructed from
cancer chemotherapy through localized delivery of multiple Engineered Proteins. Small 2016, 12, 973−987.
agents in a controlled and sustained manner. (9) Bae, K. H.; Wang, L.-S.; Kurisawa, M. Injectable Biodegradable


Hydrogels: Progress and Challenges. J. Mater. Chem. B 2013, 1, 5371−
ASSOCIATED CONTENT 5388.
(10) Li, Y.; Rodrigues, J.; Tomas, H. Injectable and Biodegradable
*
S Supporting Information
Hydrogels: Gelation, Biodegradation and Biomedical Applications.
The Supporting Information is available free of charge on the Chem. Soc. Rev. 2012, 41, 2193−2221.
ACS Publications website at DOI: 10.1021/acsami.6b03041. (11) Lü, S.; Gao, C.; Xu, X.; Bai, X.; Duan, H.; Gao, N.; Feng, C.;
Tables representing the CFD model description and Xiong, Y.; Liu, M. Injectable and Self-Healing Carbohydrate-Based
governing equations, drug loading content and encapsu- Hydrogel for Cell Encapsulation. ACS Appl. Mater. Interfaces 2015, 7,
13029−13037.
lation efficiency of VRP and PTX loaded microparticles, (12) Liu, J.; Qi, C.; Tao, K.; Zhang, J.; Zhang, J.; Xu, L.; Jiang, X.;
and drug loading content and encapsulation efficiency of Zhang, Y.; Huang, L.; Li, Q.; Xie, H.; Gao, J.; Shuai, X.; Wang, G.;
Cis-DDP and PTX loaded microparticles and figures Wang, Z.; Wang, L. A Sericin/ Dextran Injectable Hydrogel as an
showign the morphology of injectable hydrogel com- Optically Trackable Drug Delivery System for Malignant Melanoma
posed of Alg-Ald and PEI-25k at various concentrations, Treatment. ACS Appl. Mater. Interfaces 2016, 8, 6411.
solid-state 13C NMR spectrum of bare sodium alginate, (13) Wang, B.-l.; Ren, K.-f.; Chang, H.; Wang, J.-l.; Ji, J. Construction
TGA and DTG curves of the hydrogel, the deswelling of Degradable Multilayer Films for Enhanced Antibacterial Properties.
kinetics of the hydrogel; SEM micrographs of hydrogel ACS Appl. Mater. Interfaces 2013, 5, 4136−4143.
morphologies before and after loading with core−shell (14) Poon, Y. F.; Cao, Y.; Liu, Y.; Chan, V.; Chan-Park, M. B.
Hydrogels Based on Dual Curable Chitosan-graft-Polyethylene Glycol-
microparticles, TGA and DTG curves of the hydrogel at graft-Methacrylate: Application To Layer-by-Layer Cell Encapsulation.
different heating rates, weight changes of the hydrogel ACS Appl. Mater. Interfaces 2010, 2, 2012−2025.
over time, and in vitro release profiles of PTX and VRP (15) Overstreet, D. J.; Dutta, D.; Stabenfeldt, S. E.; Vernon, B. L.
from double-walled microparticles (PDF) Injectable Hydrogels. J. Polym. Sci., Part B: Polym. Phys. 2012, 50, 881−


903.
AUTHOR INFORMATION (16) Siemoneit, U.; Schmitt, C.; Alvarez-Lorenzo, C.; Luzardo, A.;
Otero-Espinar, F.; Concheiro, A.; Blanco-Méndez, J. Acrylic/cyclo-
Corresponding Author dextrin Hydrogels with Enhanced Drug Loading And Sustained
*Tel.: +65-65165079. Fax: +65-67791936. E-mail: chewch@ Release Capability. Int. J. Pharm. 2006, 312, 66−74.
nus.edu.sg. (17) Liu, Y.-Y.; Fan, X.-D. Synthesis, Properties and Controlled
Notes Release Behaviors of Hydrogel Networks Using Cyclodextrin As
Pendant Groups. Biomaterials 2005, 26, 6367−6374.
The authors declare no competing financial interest.


(18) Dulong, V.; Mocanu, G.; Le Cerf, D. A Novel Amphiphilic pH-
sensitive Hydrogel Based On Pullulan. Colloid Polym. Sci. 2007, 285,
ACKNOWLEDGMENTS 1085−1091.
This work is financially supported by A*STAR and National (19) Ranganath, S. H.; Kee, I.; Krantz, W. B.; Chow, P. K.; Wang, C.
University of Singapore under the project/grant numbers H. Hydrogel Matrix Entrapping PLGA-paclitaxel Microspheres: Drug
APG2013/40A (A*STAR BMRC Strategic Positioning Fund, Delivery with Near Zero-order Release and Implantability Advantages
for Malignant Brain Tumour Chemotherapy. Pharm. Res. 2009, 26,
A*STAR-P&G Collaboration, R279-000-487-305) and R261-
2101−2114.
509-001-646 (3D Printing Initiatives), respectively. Pooya (20) Sivakumaran, D.; Maitland, D.; Hoare, T. Injectable Microgel-
Davoodi greatly appreciates the National University of Hydrogel Composites for Prolonged Small-Molecule Drug Delivery.
Singapore for PhD graduate research scholarship. We thank Biomacromolecules 2011, 12, 4112−4120.
Dr. Marc V Garland and Mrs. Li Li Ong for their help with (21) Xie, J.; Marijnissen, J. C. M.; Wang, C. H. Microparticles
solid-state 13C NMR experiments. Developed by Electrohydrodynamic Atomization For The Local

22799 DOI: 10.1021/acsami.6b03041


ACS Appl. Mater. Interfaces 2016, 8, 22785−22800
ACS Applied Materials & Interfaces Research Article

Delivery of Anticancer Drug To Treat C6 Glioma In vitro. Biomaterials Okano, T., Eds.; Springer New York: New York, NY, 2010; Chapter 1,
2006, 27, 3321−3332. pp 1−16.
(22) Cleek, R. L.; Ting, K. C.; Eskin, S. G.; Mikos, A. G. (39) Giano, M. C.; Ibrahim, Z.; Medina, S. H.; Sarhane, K. A.;
Microparticles of Poly(dl-lactic-co-glycolic acid)/poly(ethylene glycol) Christensen, J. M.; Yamada, Y.; Brandacher, G.; Schneider, J. P.
Blends For Controlled Drug Delivery. J. Controlled Release 1997, 48, Injectable Bioadhesive Hydrogels with Innate Antibacterial Properties.
259−268. Nat. Commun. 2014, 5, No. 4095, DOI: 10.1038/ncomms5095.
(23) Hu, H.; Lin, Z.; He, B.; Dai, W.; Wang, X.; Wang, J.; Zhang, X.; (40) Pasquarella, C.; Pitzurra, O.; Savino, A. The Index of Microbial
Zhang, H.; Zhang, Q. A Novel Localized Co-delivery System with Air Contamination. J. Hosp. Infec. 2000, 46, 241−256.
Lapatinib Microparticles and Paclitaxel Nanoparticles in a Peritumor- (41) Xie, J.; Jiang, J.; Davoodi, P.; Srinivasan, M. P.; Wang, C. H.
ally Injectable In situ Hydrogel. J. Controlled Release 2015, 220, 189− Electrohydrodynamic Atomization: A Two-decade Effort To Produce
200. and Process Micro-/nanoparticulate Materials. Chem. Eng. Sci. 2015,
(24) Davoodi, P.; Srinivasan, M. P.; Wang, C. H. Synthesis of 125, 32−57.
Intracellular Reduction-sensitive Amphiphilic Polyethyleneimine and (42) Lee, W. L.; Widjaja, E.; Loo, S. C. One-step Fabrication of
Poly(ε-caprolactone) Graft Copolymer for On-demand Release of Triple-layered Polymeric Microparticles with Layer Localization of
Doxorubicin and p53 Plasmid DNA. Acta Biomater. 2016, 39, 79−93. Drugs as A Novel Drug-delivery System. Small 2010, 6, 1003−1011.
(25) Miller, K.; Wang, M.; Gralow, J.; Dickler, M.; Cobleigh, M.; (43) Xu, Q.; Chin, S. E.; Wang, C. H.; Pack, D. W. Mechanism of
Perez, E. A.; Shenkier, T.; Cella, D.; Davidson, N. E. Paclitaxel Plus Drug Release from Double-walled PDLLA(PLGA) Microspheres.
Bevacizumab versus Paclitaxel Alone for Metastatic Breast Cancer. N. Biomaterials 2013, 34, 3902−3911.
Engl. J. Med. 2007, 357, 2666−2676.
(26) Sovak, M. A.; Dupont, J.; Hensley, M. L.; Ishill, N.; Gerst, S.;
Abu-Rustum, N.; Anderson, S.; Barakat, R.; Konner, J.; Poyner, E.;
Sabbatini, P.; Spriggs, D. R.; Aghajanian, C. Paclitaxel and Carboplatin
in The Treatment of Advanced or Recurrent Endometrial Cancer: A
Large Retrospective Study. Int. J. Gynecol. Cancer. 2007, 17, 197−203.
(27) Lai, W.-F.; Susha, A. S.; Rogach, A. L. Multicompartment
Microgel Beads for Co-Delivery of Multiple Drugs at Individual
Release Rates. ACS Appl. Mater. Interfaces 2016, 8, 871−880.
(28) Lee, W. L.; Guo, W. M.; Ho, V. H. B.; Saha, A.; Chong, H. C.;
Tan, N. S.; Tan, E. Y.; Loo, S. C. J. Delivery of Doxorubicin and
Paclitaxel From Double-Layered Microparticles: The Effects of Layer
Thickness and Dual-drug vs. Single-drug Loading. Acta Biomater.
2015, 27, 53−65.
(29) Ma, H.; He, C.; Cheng, Y.; Yang, Z.; Zang, J.; Liu, J.; Chen, X.
Localized Co-delivery of Doxorubicin, Cisplatin, and Methotrexate by
Thermosensitive Hydrogels for Enhanced Osteosarcoma Treatment.
ACS Appl. Mater. Interfaces 2015, 7, 27040−27048.
(30) Panayiotou, M.; Pöhner, C.; Vandevyver, C.; Wandrey, C.;
Hilbrig, F.; Freitag, R. Synthesis and Characterisation of Thermo-
responsive Poly(N,N′-diethylacrylamide) Microgels. React. Funct.
Polym. 2007, 67, 807−819.
(31) Chen, P. C.; Kohane, D. S.; Park, Y. J.; Bartlett, R. H.; Langer,
R.; Yang, V. C. Injectable Microparticle−gel System for Prolonged and
Localized Lidocaine Release. II. In vivo Anesthetic Effects. J. Biomed.
Mater. Res. 2004, 70A, 459−466.
(32) Yan, W. C.; Davoodi, P.; Tong, Y. W.; Wang, C. H.
Computational Study of Core-shell Droplet Formation In Coaxial
Electrohydrodynamic Atomization Process. AIChE J. 2016,
DOI: 10.1002/aic.15361.
(33) Davoodi, P.; Feng, F.; Xu, Q.; Yan, W. C.; Tong, Y. W.;
Srinivasan, M. P.; Sharma, V. K.; Wang, C. H. Coaxial Electro-
hydrodynamic Atomization: Microparticles for Drug Delivery
Applications. J. Controlled Release 2015, 205, 70−82.
(34) Balakrishnan, B.; Lesieur, S.; Labarre, D.; Jayakrishnan, A.
Periodate oxidation of sodium Alginate in Water and in Ethanol-water
Mixture: A Comparative Study. Carbohydr. Res. 2005, 340, 1425−
1429.
(35) Friedrich, J.; Seidel, C.; Ebner, R.; Kunz-Schughart, L. A.
Spheroid-based Drug Screen: Considerations and Practical Approach.
Nat. Protoc. 2009, 4, 309−324.
(36) Oldfield, F. F.; Cowan, D. L.; Sun, A. Y. The Involvement of
Ethanol in The Free Radical Reaction of 6-hydroxydopamine.
Neurochem. Res. 1991, 16, 83−87.
(37) Fajardo, A. R.; Silva, M. B.; Lopes, L. C.; Piai, J. F.; Rubira, A. F.;
Muniz, E. C. Hydrogel Based on an Alginate-Ca2+/chondroitin Sulfate
Matrix As A Potential Colon-specific Drug Delivery System. RSC Adv.
2012, 2, 11095−11103.
(38) Omidian, H.; Park, K. Introduction to Hydrogels. In Biomedical
Applications of Hydrogels Handbook; Ottenbrite, M. R., Park, K.,

22800 DOI: 10.1021/acsami.6b03041


ACS Appl. Mater. Interfaces 2016, 8, 22785−22800

You might also like