Anda di halaman 1dari 9

European Journal of Pharmaceutical Sciences 105 (2017) 55–63

Contents lists available at ScienceDirect

European Journal of Pharmaceutical Sciences


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

Preparation of liposomes containing small gold nanoparticles using MARK


electrostatic interactions
Gennaro A. Dichelloa, Takahiro Fukudad, Toru Maekawad, Raymond L.D. Whitbya,b,
Sergey V. Mikhalovskya,b, Mohammed Alavijehc, Ananth S. Pannalaa,⁎, Dipak K. Sarkera
a
Biomaterials & Drug Delivery Research Group, School of Pharmacy & Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, United Kingdom
b
Department of Chemical Engineering, Faculty of Engineering, Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana, Kazakhstan
c
Pharmidex, 14 Hanover Street, Mayfair, London W1S 1YH, United Kingdom
d
Bio-Nano Electronics Research Centre, Toyo University, Kawagoe, Japan

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

Keywords: The development of liposome-nanoparticle colloid systems offers a versatile approach towards the manufacture
Nanoparticle of multifunctional therapeutic platforms. A strategy to encapsulate small metallic nanoparticles (< 4 nm) within
Liposome multilamellar vesicles, effected by exploiting electrostatic interactions was investigated. Two liposome-gold
Encapsulation nanoparticle (lipo-GNP) systems were prepared by the reverse-phase evaporation method employing cationic or
Electrostatic interaction
anionic surface functionalised particles in combination with oppositely charged lipid compositions with
Loading content
subsequent post-formulation PEGylation. Structural characterisation using electron microscopy and elemental
analysis revealed a regular distribution of GNPs between adjacent lipid bilayers of intact liposomes. Nanoparticle
encapsulation efficacy of the two lipo-GNP systems was revealed to be significantly different (p = 0.03),
evaluated by comparing the ratio of measured lipid to gold concentration (loading content) determined by a
colorimetric assay and atomic emission spectroscopy, respectively. It was concluded that the developed synthetic
strategy is an effective approach for the preparation of liposome-nanoparticle colloids with potential to control
the relative concentration of encapsulated particles to lipids by providing favourable electrostatic interactions.

1. Introduction and shape-dependent physical and chemical properties (Link and El-
Sayed, 2000). Although frequently reported as intriguing candidates for
The design and application of functional nanosystems has become a range of applications, few options are available to avoid their
well established in recent years encompassing fields of biology, associated toxicity and side-effects without significantly altering surface
medicine, photonics, catalysis and materials science (Roco, 2004). chemistry (Lewinski et al., 2008). Overcoming this, strategies of
The driving force behind this development can be ascribed to a common incorporating metallic nanoparticles within traditional “soft matter”
goal: to create new materials and devices with superior properties, materials such as polymers and lipids in a non-covalent manner are
functions, efficiency and safety. As increasingly sensitive and complex being investigated (Puri et al., 2009). This approach is hypothesised to
nanosystems are created the demand to find convenient approaches of combine advantageous features whilst enabling individual nanoparti-
protecting and preserving the integrity of functional components has cles to retain their native characteristics (Cattaneo et al., 2010;
arisen (Albet-Torres and Månsson, 2011). In addition, the ability to Longmire et al., 2011). Moreover, combining multiple components into
isolate individual functional materials presents a new strategy of nanosystems facilitates the implementation of multi-functionality, such
controlling in situ responsive systems and potentially, self-assembling as in vivo targeting capabilities combined with controlled drug release
materials. or responsive in situ activity (Mura et al., 2013; Muthu et al., 2014).
A diverse array of nanomaterials are currently under investigation Sharing this notion, nanoparticles have been incorporated into
and as a result an increase in human and environmental contact appears conventional “delivery vehicles” such as liposomes, which are artifi-
inevitable with potentially hazardous consequences (Faraji and Wipf, cially prepared closed vesicles that are composed of lamellae of lipid
2009; Hristozov and Malsch, 2009). Metallic nanoparticles in particular bilayers surrounding an aqueous core (Chang et al., 2012). The
receive considerable attention, owing to their unique exploitable size vesicular structure provides a non-associated surface passivation layer


Corresponding author at: School of Pharmacy & Biomolecular Sciences, University of Brighton, Lewes Road, Brighton BN2 4GJ, United Kingdom.
E-mail address: a.s.v.pannala@brighton.ac.uk (A.S. Pannala).

http://dx.doi.org/10.1016/j.ejps.2017.05.001
Received 18 January 2017; Received in revised form 23 March 2017; Accepted 2 May 2017
Available online 03 May 2017
0928-0987/ © 2017 Elsevier B.V. All rights reserved.
G.A. Dichello et al. European Journal of Pharmaceutical Sciences 105 (2017) 55–63

that protects, preserves and isolates the nanomaterials (Nam et al., TPP-GNPs (20 mg) were resuspended in dichloromethane (4 ml)
2013), whilst also providing the framework in which multiple func- and added to an aqueous solution containing either MES (20 mg,
tional or therapeutic components can be combined into a single system 0.12 mmol in 2 ml) or DMET (40 mg, 0.28 mmol in 3 ml). An argon
(Okuda and Kidoaki, 2012). In addition, the liposomal exterior surface atmosphere was introduced and the biphasic mixtures were vigorously
provides a platform for further modification whereby predictable in situ stirred to ensure sufficient contact between the two phases. Complete
or in vivo behaviour can be attained (Nag and Awasthi, 2013). Two transfer of the coloured nanoparticles into the aqueous phase occurred
general classes of nanoparticle-in-liposome configurations have pre- within 4 h at which point it was extracted. The nanoparticle suspen-
viously been reported with particles distributed within lipid bilayers sions were then washed (×3) with dichloromethane and GNPs were
(Park et al., 2006) or the aqueous core (Hong et al., 1983). However, isolated by ultracentrifugation (200,000 × g). Recovered products were
incorporating nanoparticles into bilayers requires particles with hydro- then re-suspended in ethanol (20 ml) and precipitated upon addition of
phobic surfaces, whilst entrapment within the aqueous core relies on hexane (20 ml) using centrifugation (4000 × g) to remove excess
spontaneous encapsulation resulting in few particles per liposome ligands. Resultant GNP powders were then dried under vacuum over-
(Chen et al., 2006). night before resuspending in deionised water.
In the current study, electrostatic interactions between negatively
charged lipids and positively charged GNPs (NCL-PCG) and vice versa 2.3. Formulation of gold nanoparticle encapsulated liposomes
(PCL-NCG) were investigated as an alternative driving-force to facilitate
entrapment of nanoparticles within liposomes. This concept is based on Two individual formulations were prepared via a three-step strategy
previous reports, such as lipofection systems, where ionic macromole- following previously published protocols with modifications. Initially,
cules have been encapsulated within liposomes containing oppositely crude lipo-GNPs were prepared by reverse-phase-evaporation method
charged ionic lipid compositions (Balazs and Godbey, 2010). GNPs (Szoka and Papahadjopoulos, 1978) before undergoing size-extrusion
were selected as a proof-of-concept to encapsulate within liposomes (Olson et al., 1979) and subsequent post-formulation lipid-PEG mod-
primarily because of their inherent chemical robustness and availability ification (Awasthi et al., 2004). Liposome suspensions were then
of established techniques for surface engineering. Physicochemical separated from unencapsulated GNPs and excess reagents using size-
characterisation and morphological assessment of the synthesised exclusion chromatography (Chen et al., 2006).
lipo-GNP systems was investigated. Short-term (48 h) colloidal stability Lipid compositions DPPC:DPPG (molar ratio 6.1:0.7) and
was evaluated and quantification of internalised gold and lipid DPPC:DPTAP (molar ratio 6.1:0.8) were prepared by combining
concentration was also determined. lyophilised lipid powders and dissolving in chloroform:methanol
(2:1), providing a 2.5 mg/ml total lipid content. Aqueous GNP suspen-
2. Materials and methods sions with a concentration of 0.5 mg/ml were then combined with the
appropriate lipid solutions (1:1 v/v) and subsequently sonicated to
2.1. Materials afford a white/brown emulsion-like mixture. The biphasic suspension
was then transferred onto a rotary evaporator for gradual removal of
Lyophilised lipids: 1,2-dipalmitoyl-sn-glycero-3-phosphocholine the organic phase. Complete removal of organic solvents was achieved
(DPPC); 2-dipalmitoyl-sn-glycero-3-phospho-(1′-rac-glycerol) (DPPG); within 1 h and resultant crude liposome suspensions were diluted in
1,2-dipalmitoyl-3-trimethylammonium-propane (DPTAP); 1,2-dipalmi- PBS (pH 7.4) to provide a total lipid content of 1 mg/ml. Aliquots (1 ml)
toyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene gly- were then briefly sonicated before repeatedly (×15) passing through
col)-5000] (DPPE-PEG5000); 200 nm porous (13 mm diameter) poly- 200 nm porous polycarbonate membranes utilising an “Avanti mini
carbonate filter membranes and “Avanti mini-extruder system” were extruder system.” The temperature was maintained slightly above the
purchased from Avanti Polar Lipids (Alabaster, Alabama, USA). melting transition of the main lipid component, DPPC, (Tm = 41 °C)
Platinum-blue (dye) was obtained from Nisshin EM Corporation and reduced to room temperature during the extrusion process.
(Shinjuku, Tokyo, Japan). Hydrogen tetrachloroaurate, triphenylpho- Aliquots were then either set-aside for purification or added immedi-
sphine (TPP), 2-dimethylaminoethanthiol (DMET), 2-mercaptoethane- ately to 0.2 ml portions of a preheated (50 °C) aqueous solution of
sulfonate (MES), tetraoctylammonium bromide (TOAB), Sodium bor- DPPE-PEG5000 (0.01% w/v) in glass vials. The temperature was
ohydride, bromothymol blue, Sephadex-G50, phosphate buffered saline maintained for 30 min before allowing the liposomal suspensions to
(PBS) and all other reagents were purchased from Sigma-Aldrich naturally reach room temperature.
Chemical Company (Gillingham, Dorset, UK). To separate liposomes from unencapsulated GNPs and excess
reagents, size-exclusion chromatography was performed using
2.2. Synthesis of gold nanoparticles Sephadex-G50 gel as the stationary phase and PBS (pH 7.4) as eluent.
Fractions were collected (1 ml) and the first five fractions identified to
GNPs bearing cationic or anionic ligands on their surfaces were contain lipids by spot staining with bromothymol blue (pH 6) in 10% v/
prepared according to a previously reported two-step approach. v aqueous ethanol solution, were combined and stored at room
Triphenylphosphine (TPP) stabilised GNPs were first prepared via a temperature.
modified Brust method (Weare et al., 2000) and subsequently utilised in
interfacial-ligand exchange reactions with DMET or method (Warner 2.4. Hydrodynamic particle size analysis of liposomes
et al., 2000).
TPP-GNPs were prepared by dissolving hydrogen tetrachloroaurate Lipo-GNPs hydrodynamic size distributions and dispersity values
(0.25 g, 0.64 mmol) and TOAB (0.4 g, 0.73 mmol) in nitrogen sparged were determined using a Zetasizer, (Malvern, NANO ZS90 series),
water/toluene (13 ml/17 ml) followed by addition of triphenylpho- operating in DLS mode at 25 °C with measurements recorded at a 90-
sphine (0.58 g, 2.21 mmol) under vigorous stirring. Freshly prepared degree detection angle. Representative samples from each batch
aqueous sodium borohydride (0.35 g, 9.4 mmol, dissolved in 10 ml) (n = 6) were diluted in PBS buffer (0.45 μm sterile filtered), gently
was then added at once by injecting directly into the organic phase, shaken and rested for 10 min before analysis.
triggering a colour change to dark purple. The resulting mixture was
stirred for 3 h under a nitrogen atmosphere before the organic phase 2.5. Zeta potential analysis of liposomes
was isolated and washed (× 3) with distilled water. The product was
further washed with a series of hexane and methanol:water (2:3) to The same lipo-GNPs specimens prepared for size analysis were
remove the phase-transfer catalyst and unreacted starting materials. studied using a Zetasizer, (Malvern, NANO ZS90 series), operating in

56
G.A. Dichello et al. European Journal of Pharmaceutical Sciences 105 (2017) 55–63

zeta mode at 25 °C using disposable folded capillary cells (DTS1070, application of the Stewart Assay (Stewart, 1980). An ammonium
Malvern Instruments Ltd., UK). Measurements were also recorded for ferrothiocyanate (AF) standard solution was prepared by dissolving
liposome samples that had not undergone PEG modification. ferric chloride hexahydrate (27.03 g, 0.1 mol) and ammonium thiocya-
nate (30.4 g, 0.4 mol) in 1 l of distilled water. Lyophilised DPPC was
2.6. Study of gold encapsulation using TEM dissolved in chloroform and serially diluted to prepare a range of
calibration standards (0–80 μg/ml). Aliquots of each standard (1 ml)
The morphology of liposomes and encapsulation of GNPs was were then combined with AF (2 ml) and vortexed before isolating the
studied using a high-resolution transmission electron microscope organic phase by centrifugation (300 × g, 5 min). Prior to analysis, the
(TEM - JEOL JEM-2200) operating at an accelerating voltage of volume was corrected with chloroform (2 ml) to account for any
200 kV. Samples were diluted 10-fold with distilled water and pipetted evaporation. For the analysis of lipo-GNP specimens, 100 μl of a sample
onto hydrophilised, carbon-formvar coated 200-mesh copper grids suspension was sonicated for 5 min before the addition of the AF
(Agar Scientific, UK) before wick drying excess solution with filter reagent. In validation, a stock solution containing DPPC (200 μg/ml),
paper. Negative staining of specimens was achieved by exposing grids DPPG (20 μg/ml) and DPTAP (20 μg/ml) was prepared in PBS buffer
preloaded with sample to platinum-blue (dye) for 30 s prior to wick and serially spiked (0, 1, 2, 3, 4 ml) with a solution of DPPC (200 μg/
drying; all specimen grids were allowed to rest for 30 min before ml) in 50 ml volumetric flasks before treating with AF reagent. The
analysis. The same unstained-specimens were used for energy disper- absorbance of specimens and standards was measured using a Perki-
sive X-ray spectroscopy (EDS) operating in scanning-transmission nElmer LAMBDA-25 ultraviolet-visible spectrometer (λ = 485 nm).
(STEM) mode, utilising an integrated JEOL-JED-2300 EDS detector.

2.7. Topographical assessment of lipo-GNPs using SEM 3. Results

Lipo-GNPs surfaces were studied using a field-emission scanning 3.1. Preparation of gold nanoparticles
electron microscope (SEM - Zeiss – Sigma 300) operating at an
accelerating voltage of 5 kV using a secondary electron detector (Cho The GNPs utilised in this investigation were prepared by a two-stage
et al., 2015). Samples were diluted 10-fold with distilled water and wet synthesis where TPP-GNPs are first synthesised before undergoing
10 μl was deposited onto silicon wafer mounted on aluminium speci- interfacial-exchange reactions with thiol-containing ionic ligands. The
men stubs. Specimens were snap-frozen by submerging in liquid first step ensures the formation of uniform, spherical GNPs of the
nitrogen and subsequently freeze-dried (Christ®-ALPHA 1–4) before desired size, considered a critical parameter for encapsulation within
sputter coating (5 nm) with platinum (Quorum Q150T ES Turbo- liposomes. The second stage introduces the desired surface ligands
Pumped coater device). whilst crucially retaining the original particle size (Warner et al., 2000).
Characterisation via TEM gives an average gold core size of
2.8. Quantification of gold encapsulation using AES 2.8 ± 1.2 nm for DMET-GNPs and 2.6 ± 1.2 nm for MES-GNPs
(n = 100), comparable to previous reports using this approach
Quantification of encapsulated gold was determined using an (Warner et al., 2000). As expected, zeta potential analysis reveals GNPs
Agilent 4100 microwave-plasma atomic emission spectrometer (AES) display a positive surface potential of + 15.1 ± 1.2 mV and a negative
according to a previously reported protocol with modifications surface potential of −17.5 ± 1.4 mV when functionalised with either
(Chithrani et al., 2006). TPP-GNPs were digested in aqua regia cationic DMET or anionic MES ligands, respectively. The utilised
(HCl:HNO3) for 24 h before serially diluting to prepare a range of approach was found to be a simple and highly reproducible method
calibration standards (0–20 μg/ml). For the analysis of lipo-GNP affording sufficient yields and recovery of GNPs for the purpose of this
samples, 200 μl aliquots were mixed with chloroform (1 ml), vortexed investigation.
for 30 s and the two phases separated by centrifugation (300 × g,
5 min). The aqueous phase was then isolated, dried under mild heating
and residues were digested in aqua regia (0.2 ml) for 24 h. Specimens 3.2. Zeta potential and PEGylation of lipo-GNP formulations
were then accurately made up to a total volume of 2 ml with distilled
water. To attain fitting baselines, blank solutions were prepared by To verify the successful insertion of PEG into the exterior lamella of
digesting 0.1 mmol of the appropriate ligand (TPP, DMET or MES) in prepared lipo-GNPs, zeta potential measurements (Table 1) were
aqua regia and diluting with distilled water. In validation, stock performed before and after post-formulation PEG modification for each
solutions of DMET and MES functionalised GNPs (6.25 μg/ml) were batch (n = 6). For NCL-PCG a reduction in magnitude was observed
serially spiked (0, 2, 4, 6, 8 ml) with further digested GNP solutions from − 20.1 ± 0.2 mV to − 5.11 ± 0.59 mV, whilst PCL-NCG shifted
(25 μg/ml) in 25 ml volumetric flasks. Specimens were introduced to from a positive potential of + 2.9 ± 0.3 mV to negative
the nebuliser at a pump rate of 80 rpm with a 15 s stabilisation time − 3.61 ± 0.32 mV. As revealed, a characteristic charge-shielding
before measurements were recorded (λ = 267.595 nm). effect takes place upon DPPE-PEG insertion, in agreement with previous
reports of liposomal modification with PEG (Nakamura et al., 2012).
2.9. Quantification of DPPC lipid content in formulations The low magnitude negative surface potential displayed by both
systems can be ascribed to PEG itself (Nakamura et al., 2012).
Quantification of the main lipid component (DPPC) was achieved by

Table 1
A summary of data obtained from dynamic light scattering and zeta potential analysis of lipo-GNPs after their preparation (T0), after 48 h of storage (T48), with and without PEGylation
(mean ± standard deviation, n = 6).

Initial DLS measurements (T0) DLS measurements after 48 h (T48) Zeta potential measurements (mV)

Size (d·nm) Dispersity Size (d·nm) Dispersity Without PEGylation After PEGylation

NCL-PCG 183 ± 12 0.08 ± 0.02 148 ± 2 0.08 ± 0.02 − 20.1 ± 0.20 − 5.11 ± 0.59
PCL-NCG 176 ± 8 0.13 ± 0.05 147 ± 1 0.08 ± 0.05 + 2.94 ± 0.30 − 3.61 ± 0.32

57
G.A. Dichello et al. European Journal of Pharmaceutical Sciences 105 (2017) 55–63

3.3. Hydrodynamic size and colloidal stability of liposomes and PCL-NCG are presented in Fig. 1 and Fig. 2 respectively. From SEM
micrographs (Fig. 1A and Fig. 2A), lipo-GNPs are revealed to display
Colloidal particle size data obtained for six individual batches uniform surface morphologies with smooth exteriors and no features
(n = 6) of NCL-PCG and PCL-NCG using dynamic light scattering is that can be ascribed to surface-exposed GNPs. Structures appear to be
summarised in Table 1 with values expressed as mean ± standard well-dispersed, spherical entities consistent with stable colloidal parti-
deviation. Average hydrodynamic diameters of 183.0 ± 12.3 nm for cles. TEM micrographs of liposomes negatively stained with an
NCL-PCG and 175.9 ± 8.1 nm for PCL-NCG were recorded with electron-dense platinum-based dye (Fig. 1B, C and Fig. 2B, C) reveal
dispersity values of 0.08 ± 0.02 and 0.17 ± 0.06 respectively. Aver- intact, spherical, vesicular structures with well-defined boundaries even
age liposome sizes below 200 nm are observed, reflecting the pore size when in close proximity to one another. Features characteristic of GNPs
utilised in the extrusion process. Low dispersity values and size are revealed within the confinements of these structures and appear as
variability for both systems suggest a high degree of colloidal homo- densely contrasted regions relative to the unstained background. In
geneity. both systems, such features are observed in high numbers frequently
As an estimate of colloidal stability specimens used in size measure- appearing concentrated among the bilayer region of vesicles. This is
ments were reanalysed under identical conditions after 48 h. Values of further highlighted in Fig. 1B and Fig. 2B where repeating “rings” of
147.6 ± 2.3 nm for NCL-PCG and 147.4 ± 1.2 nm for PCL-NCG features ascribed to GNPs are observed. To confirm these observations,
reveal a reduction in hydrodynamic particle size for both systems. representative structures were selected for further elemental analysis.
Statistical comparison (paired two-tail student t-test) reveals the STEM micrographs and corresponding elemental maps of gold can be
decrease in particle size to be significant from initial measurements seen in Fig. 1D, E and Fig. 2D, E with interpreted spectra provided in
for both NCL-PCG (p = 0.0014) and PCL-NCG (p = 0.0005). Dispersity Fig. 3. These observations confirm the previous assumptions, with
values (Table 1) remained comparably low for both systems (p > 0.5) features appearing concentrated within liposomal structures ascribed to
indicative of uniform colloidal particles, suggesting size reductions are GNPs correlating with the detection of gold in STEM micrographs and
a universal trend. EDS maps. Other elements revealed from spectra include carbon,
oxygen and phosphorus which can be assigned to the presence of
lipids, sulphur which is indicative of ligands on GNP surfaces and
3.4. Morphological assessment and elemental analysis of lipo-GNPs copper from TEM specimen grids used in the analysis.

SEM was utilised to study the morphological features of lipo-GNPs


evidencing whether GNPs were present on the exterior surfaces of 3.5. Quantifying liposomal gold encapsulation
liposomes. TEM enabled visualisation of internalised nanoparticles as a
result of the electron density difference between gold and lipids. The concentration of internalised gold was determined by atomic
Negative staining of specimens with an electron-dense dye was then emission spectroscopy using a protocol previously applied to quantify
employed to reveal whether liposomal structures were intact. This was GNP uptake in cellular studies (Chithrani et al., 2006). A calibration
evidenced by the internal liposomal structure remaining unstained plot was prepared between 0 and 20.0 μg/ml of digested TPP-GNPs
relative to the stained background (De Carlo and Harris, 2011). STEM- (Fig. 4A) and the equation of least squares regression line was derived
EDS provided elemental maps and comprehensive spectra enabling (y = 8507.5x + 204.02, R2 = 0.998). Corresponding concentrations of
evaluation of gold distribution and identification of other elements gold in lipo-GNPs specimens (n = 6) were then calculated with NCL-
present. Micrographs of SEM, TEM and corresponding EDS for NCL-PCG PCG revealed to contain 14.9 ± 6.1 μg/ml and PCL-NCG containing

Fig. 1. Representative images of NCL-PCG (A): SEM micrograph (B): STEM micrograph of an individual vesicular structure with (C): corresponding EDS map for gold. (D, E): TEM
micrographs of negatively stained specimens.

58
G.A. Dichello et al. European Journal of Pharmaceutical Sciences 105 (2017) 55–63

Fig. 2. Representative images of PCL-NCG (A): SEM micrograph (B): STEM micrograph of an individual vesicular structure with (C): corresponding EDS map for gold. (D, E): TEM
micrographs of negatively stained specimens.

Fig. 3. Interpreted EDS spectra corresponding to STEM images displayed in Figs. 3B and 4B (A): NCL-PCG and (B): PCL-NCG.

6.2 ± 2.9 μg/ml. The procedure was validated using the method of 202.4 ± 39.7 μg/ml and PCL-NCG 148.0 ± 30.4 μg/ml. The proce-
standard additions in a representative matrix, recovery (n = 3) of dure was validated using the method of standard additions in a
DMET-GNPs was calculated to be 93.6 ± 4.6% and MES-GNPs representative matrix, recovery of DPPC was calculated to be
98.8 ± 7.4% with statistical comparison (unpaired student t-test) 103.7 ± 7.3%, (red marker, Fig. 5B).
revealing no significant difference (p > 0.05) between recoveries
(Fig. 4B).
4. Discussion

3.6. Quantifying DPPC lipid content in the lipo-GNP formulations Two distinct lipo-GNP systems were formulated by exploiting
electrostatic interactions between GNP surfaces and lipids that consti-
Measurements of DPPC lipid concentration were determined by the tute the liposomes. This interaction between opposing charges of the
colorimetric Stewart assay, a well-established technique that is suitable two components facilitates their amalgamation as the lipids self-
for the detection of sub-micromolar concentrations of phosphatidylcho- organise to form vesicular structures. The proposed configuration
line lipids in the presence of phosphate buffers utilised in this (Fig. 6) shows a regular nanoparticle distribution within multilamellar
investigation (Stewart, 1980). A calibration plot ranging from 0 to liposomes located at the peripheral region of the central aqueous core
40.0 μg/ml of DPPC lipid was prepared (Fig. 5A) and the equation of and at the interface of adjacent lipid bilayers and separating layers of
least squares regression line was derived (y = 0.0103x + 0.0291, water (Shaheen et al., 2006).
R2 = 0.992). Corresponding concentrations of DPPC in lipo-GNP speci- Adopting the reverse-phase evaporation method, interactions be-
mens (n = 6) were then calculated with NCL-PCG revealed to contain tween lipid “head groups” and GNPs at the organic-aqueous interface

59
G.A. Dichello et al. European Journal of Pharmaceutical Sciences 105 (2017) 55–63

Fig. 4. Quantification of gold encapsulation by AES (A): Calibration plot of digested TPP-GNPs with (insert) highlighting lower concentrations. (B): Bar graph comparing the relative (%)
recovery of DMET-GNPs and MES-GNPs. Values are reported as mean with error bars representing standard deviation. C): Concentration of gold determined for NCL-PCG and PCL-NCG
(mean ± standard deviation, n = 6).

Fig. 5. Quantification of DPPC lipid using the Stewart Assay (A): Calibration plot for DPPC lipid (B): plot of standard additions with extrapolated value (red marker) corresponding to
recovery of DPPC lipid in a representative sample matrix (C): Concentration of DPPC determined for NCL-PCG and PCL-NCG (mean ± standard deviation, n = 6). (For interpretation of
the references to colour in this figure, the reader is referred to the web version of this article.)

60
G.A. Dichello et al. European Journal of Pharmaceutical Sciences 105 (2017) 55–63

Fig. 6. Proposed structure of lipo-GNPs with the underpinning lipid-nanoparticle interaction at the aqueous-bilayer interface highlighted (insert). Lipid structures utilised in the
preparation of NCL-PCG and PCL-NCG are presented (A): DPPC, (B): DPTAP, (C): DPPG.

Fig. 7. Simplified representation of the processes involved in the preparation of lipo-GNPs. (A): Lipid molecules interacting with GNPs (white arrow) at the aqueous-organic interface (B):
GNP-liposome formation, (C): size extrusion, (D): post-formulation PEG insertion (E): PEG-grafted GNP-liposomes after size exclusion chromatography.

bring the two components into close proximity (white arrows, (Fig. 7A, supports insertion into the outermost lamella of liposomes. Removal of
B). As the organic solvent is depleted lipid molecules transfer into the excess GNPs and reagents was then achieved by size-exclusion gel
aqueous phase and self-organise to form closed lipid-bilayers (Shailesh chromatography, where the larger particles (i.e. liposomes) elute from
et al., 2009). Associated GNPs subsequently become entrapped the column quicker than the smaller particles and molecules (i.e. excess
throughout the multilamellar vesicular structures. Crude lipo-GNPs nanoparticles and unincorporated PEG) as they pass through a porous
are then resized via size extrusion, where liposomes are repeatedly matrix (Bozzuto and Molinari, 2015). Although visually a band of
forced through 200 nm porous filter membranes. Starting extrusion at nanoparticles could be seen retained within the chromatography
an elevated temperature and working downward facilitated the process column, as a precautionary measure recovery of liposomes was
and ensured that agglomerates were not formed before the next stage. restricted to a total of five 1 ml aliquots. This step was taken to ensure
The resultant suspensions were then immediately subjected to post- that subsequent characterisation and quantification was representative
formulation PEGylation using a PEG grafted lipid (DPPE-PEG-5000). of lipo-GNPs and encapsulated gold exclusively.
This was performed whilst passing through the melting-transition Microscopy and elemental analysis suggests that the strategy
temperature (Tm) of the main lipid component (DPPC, 41 °C), which employed is a promising route for the preparation of liposomes with

61
G.A. Dichello et al. European Journal of Pharmaceutical Sciences 105 (2017) 55–63

internalised small (< 4 nm) metallic nanoparticles. Although compar- reorganisation and a decrease in particle size (Wang et al., 2008). This
able strategies exploiting electrostatic attractions have previously been was suggested to occur as a consequence of alteration to the tilt-angle of
utilised to encapsulate a range of materials within liposomes, according lipid head groups, resulting in an increase in lipid-tail density and
to available literature this approach has not been applied for the tighter packing, typical of solid-phase bilayer behaviour (Somerharju
internalisation of metal nanoparticles. Furthermore, as a result of et al., 1999). Further investigation is warranted to determine whether
electrostatic interactions between lipid “head groups” and particle the mechanism of particle size reduction observed in this investigation
surfaces, GNPs appear regularly distributed at the liposomal bilayer- can be explained by a similar mechanism and whether such properties
aqueous interface. This leads to the proposal that nanoparticle arrange- could be advantageous.
ment is dissimilar from previous reports of either encapsulation of Currently the evaluation of nanoparticle encapsulation within
hydrophobic nanoparticles within bilayers or hydrophilic nanoparticles liposomes primarily involves qualitative and visual elucidation alone,
within the aqueous core of liposomes. The concept of using electrostatic which can be subjective (Mady et al., 2012). Even in instances
interactions has previously been demonstrated for entrapment of investigating how the concentration of nanoparticles in liposomal
enzymes and DNA within liposomes (Colletier et al., 2002; Ma et al., systems influence resultant performance appears to provide no direct
2007). quantification in support (Adhikari et al., 2015). As a matter of safety, it
Surface charge of cationic DMET-GNPs and anionic MES-GNPs as is vital that nanosystems for therapeutic applications are prepared with
determined by measuring the zeta potentials were initially revealed to absolute precision. Further, as highlighted above, the encapsulation and
be of a comparable magnitude. After subsequent encapsulation within distribution of nanoparticles in liposomes can determine their resultant
liposomes consisting of lipid compositions at identical mole ratios of physical and chemical properties making the gold to lipid ratio a critical
zwitterionic (DPPC) and opposing monoionic lipids (DPTAP or DPPG), design parameter. Although the influence of this ratio on liposomal
zeta potentials of a dissimilar magnitude were observed (Table 1). The bilayer properties has not been investigated in this study, previous
parameters that were considered to be potentially contributing towards reports have demonstrated a range of effects. Among which the
the observations in this study include the quantity of GNPs associated formation of nano-pores within lipid bilayers that is dependent on lipid
per liposome (Mady et al., 2012), ligand density on GNP surfaces composition, nanoparticle functionality and concentration and ensuing
(Laaksonen et al., 2006) and charge distribution of the dissimilar lipid electrostatic interactions (Lu et al., 2015), which may or may not be
structures (Langner and Kubica, 1999). Consequently, after PEGylation, desirable for specific applications. In an attempt to standardise the
both systems again display comparable zeta potential values as a result evaluation of nanoparticle encapsulation and facilitate the development
of characteristic charge-shielding effects of PEG. This phenomenon of analogous systems, the concentration of gold and the main lipid
arises as a result of sheltering the potential of the electrical double- component DPPC were determined in parallel on the same samples.
layer, which in turn effectively diminishes zeta potential (MacLachlan, Corresponding loading content (%) was calculated to reveal the ratio of
2007). These results imply that although NCL-PCG and PCL-NCG gold to lipid, providing a quantitative metric that can be compared to
appear comparable at the surface, their internal properties are different. assess encapsulation efficacy. This criterion has previously been applied
Further investigation is required to probe the “layer by layer” or sub- to evaluate the encapsulation of drugs (Xing et al., 2004) and was used
surface properties beneath the outer PEG stratum and adjacent lamella here to highlight the relationship between lipid compositions and GNPs
to determine how these characteristics vary and what if any implica- of different functionality. From the ratio of encapsulated gold to DPPC
tions that this may entail. Introducing PEG to the surface of prepared lipid measured (n = 6), loading content (%) values of 7.3 ± 2.6% for
lipo-GNPs follows recent trends in literature that stem from a catalogue NCL-PCG and 3.1 ± 1.5% for PCL-NCG were calculated (mean ±
of known advantageous biological and technological properties standard deviation). Statistical comparison (unpaired student t-test)
(Alcantar et al., 2000; Nag and Awasthi, 2013). Incorporating PEG revealed the ratio of gold to lipid to be significantly different for the two
exclusively into the exterior lamellar of liposomes using a post- systems investigated (p = 0.03). Considering the two developed lipo-
formulation approach avoids reducing the internal aqueous volume GNP systems differ only by inclusion of monoionic lipid (DPPG or
(Szoka and Papahadjopoulos, 1978), which could potentially hinder DPTAP) and GNP functionality. It is hypothesised that ensuing variable
encapsulation of GNPs. In addition, post-formulation modification electrostatic interactions between the lipids and gold facilitates the
avoids the use of excessive quantities of lipopolymers such as PEG that entrapment process and is responsible for the differences in observed
can make size-extrusion processing difficult (Nakamura et al., 2012). encapsulation efficacy. Parameters considered likely to be contributing
Particles in suspension must remain dispersed at equilibrium to towards this electrostatic process include the ligand density on GNP
avoid common instability phenomena such as aggregation or phase surfaces and the relative charge distribution of both the lipid and ligand
separation, which is often achieved by electrostatic repulsion or steric molecules (Laaksonen et al., 2006; Langner and Kubica, 1999).
stabilisation. In this investigation steric hindrance was successfully Supporting these findings, electrostatic attractions have previously
implemented by PEGylation of lipo-GNP surfaces with the PEG-grafted been recognised as an essential mechanism for the encapsulation of
lipid DPPE-PEG5000. Previous reports of using PEG with an average macromolecules in liposomes (Colletier et al., 2002), including lipofec-
molecular weight of ~5000 g/mol was reported to provide liposomes tion systems on which the current synthetic strategy was based (Ma
with a 5 nm thick “coating” that acts as a barrier, shielding the surface et al., 2007). Further studies have highlighted that electrostatic
from the external environment (Woodle et al., 1994). This approach attraction between lipids and macromolecules are the primary factor
was taken in an attempt to prevent potential inter-particle interactions determining the encapsulation potential (Hwang et al., 2012), with
between exterior-surface bound GNPs and neighbouring liposomes. To experimental conditions that diminish the interaction significantly
confirm this, lipo-GNP hydrodynamic sizes were monitored upon reducing efficacy (Shariat et al., 2014). Additional parameters identi-
storage, a common approach enabling early identification of instability fied from these studies that have not been investigated in the current
(Mengual et al., 1999). Liposomal suspensions retained a high degree of study but could be examined in future to provide optimal conditions to
uniformity and narrow size distributions after 48 h (Table 1) with no increase the loading content of analogous GNP-lipo systems include
aggregate formation or phase separation observed. Both liposomal buffer composition, pH, ionic strength and liposomal size (Hwang et al.,
systems however did display statistically significant decreases in 2012). On this premise, it is suggested that by selecting GNPs and lipids
particle sizes, although dispersity values, which provide a measure of to provide favourable electrostatic interactions, the reported approach
colloidal homogeneity, remained consistently low indicating this to be a could be used to obtain the desired quantity of nanoparticles per
universal trend. These observations agree with previously reported liposome.
findings where interactions between phospholipids and ionic nanopar-
ticles on the exterior surface of liposomes were responsible for lipid

62
G.A. Dichello et al. European Journal of Pharmaceutical Sciences 105 (2017) 55–63

5. Conclusion operating parameters on the efficiency of liposomal encapsulation of enzymes.


Colloids Surf. B Biointerfaces 94, 296–303.
Laaksonen, T., Ahonen, P., Johans, C., Kontturi, K., 2006. Stability and electrostatics of
In summary, a novel strategy of encapsulating GNPs within multi- mercaptoundecanoic acid-capped gold nanoparticles with varying counterion size.
lamellar liposomes has been developed exploiting electrostatic interac- ChemPhysChem 7, 2143–2149.
Langner, M., Kubica, K., 1999. The electrostatics of lipid surfaces. Chem. Phys. Lipids 101,
tions between ionic lipid compositions and particle surfaces. Evaluation 3–35.
of lipo-GNP morphology reveals particle distribution at the bilayer- Lewinski, N., Colvin, V., Drezek, R., 2008. Cytotoxicity of nanoparticles. Small 4, 26–49.
aqueous interface, differing from previous reports of nanoparticles in Link, S., El-Sayed, M.A., 2000. Shape and size dependence of radiative, non-radiative and
photothermal properties of gold nanocrystals. Int. Rev. Phys. Chem. 19, 409–453.
bilayers or within the aqueous core of liposomes. The prepared Longmire, M.R., Ogawa, M., Choyke, P.L., Kobayashi, H., 2011. Biologically optimized
colloidal particles remain as dispersed and uniform populations for at nanosized molecules and particles: more than just size. Bioconjug. Chem. 22,
least 48 h but appear to experience a universal reduction in particle 993–1000.
Lu, B., Smith, T., Schmidt, J.J., 2015. Nanoparticle-lipid bilayer interactions studied with
size. Determining the ratio of gold to hydrated lipid provides a simple
lipid bilayer arrays. Nano 7, 7858–7866.
approach to assess encapsulation efficacy demonstrating standardised Ma, B., Zhang, S., Jiang, H., Zhao, B., Lv, H., 2007. Lipoplex morphologies and their
evaluation. Using a combination of variable lipid compositions and influences on transfection efficiency in gene delivery. J. Control. Release 123,
GNP functionalities appears to influence the encapsulation process and 184–194.
MacLachlan, I., 2007. Liposomal formulations for nucleic acid delivery. In: Crooke, S.T.
could be exploited for further control and optimisation. The reported (Ed.), Antisense Drug Technology: Principles, Strategies, and Applications, second ed.
strategy of utilising electrostatic interactions for the encapsulation of CRC Press, UK, pp. 237–262.
metallic nanoparticles is envisaged as a platform for the development Mady, M.M., Fathy, M.M., Youssef, T., Khalil, W.M., 2012. Biophysical characterization of
gold nanoparticles-loaded liposomes. Phys. Med. 28, 288–295.
and study of analogous nanosystems. Mengual, O., Meunier, G., Cayre, I., Puech, K., Snabre, P., 1999. Characterisation of
instability of concentrated dispersions by a new optical analyser: the TURBISCAN MA
Acknowledgments 1000. Colloids Surf. A Physicochem. Eng. Asp. 152, 111–123.
Mura, S., Nicolas, J., Couvreur, P., 2013. Stimuli-responsive nanocarriers for drug
delivery. Nat. Mater. 12, 991–1003.
Funding support was provided by the Medical Research Council Muthu, M.S., Leong, D.T., Mei, L., Feng, S.S., 2014. Nanotheranostics application and
(MR/J006661/1) as part of an Industrial CASE studentship award in further development of nanomedicine strategies for advanced theranostics.
Theranostics 4, 660–677.
collaboration with Pharmidex Pharmaceutical Services. Authors would
Nag, O.K., Awasthi, V., 2013. Surface engineering of liposomes for stealth behavior.
like to further thank associates from the Bio-Nano Electronics Research Pharmaceutics 5, 542–569.
Centre (Japan), for access and support during electron microscopy Nakamura, K., Yamashita, K., Itoh, Y., Yoshino, K., Nozawa, S., Kasukawa, H., 2012.
Comparative studies of polyethylene glycol-modified liposomes prepared using
studies.
different PEG-modification methods. Biochim. Biophys. Acta Biomembr. 1818,
2801–2807.
References Nam, J., Ha, Y.S., Hwang, S., Lee, W., Song, J., Yoo, J., Kim, S., 2013. pH-responsive gold
nanoparticles-in-liposome hybrid nanostructures for enhanced systemic tumor
delivery. Nano 5, 10175–10178.
Adhikari, C., Das, A., Chakraborty, A., 2015. Controlled release of a sparingly water- Okuda, T., Kidoaki, S., 2012. Multidrug delivery systems with single formulation-current
soluble anticancer drug through pH-responsive functionalized gold-nanoparticle- status and future perspective. J. Biomater. Nanobitechnol. 3, 50–60.
decorated liposomes. ChemPhysChem 16, 866–871. Olson, F., Hunt, C.A., Szoka, F.C., Vail, W.J., Papahadjopoulos, D., 1979. Preparation of
Albet-Torres, N., Månsson, A., 2011. Long-term storage of surface-adsorbed protein liposomes of defined size distribution by extrusion through polycarbonate
machines. Langmuir 27, 7108–7112. membranes. Biochim. Biophys. Acta Biomembr. 557, 9–23.
Alcantar, N.A., Aydil, E.S., Israelachvili, J.N., 2000. Polyethylene glycol-coated Park, S.H., Oh, S.G., Mun, J.Y., Han, S.S., 2006. Loading of gold nanoparticles inside the
biocompatible surfaces. J. Biomed. Mater. Res. 51, 343–351. DPPC bilayers of liposome and their effects on membrane fluidities. Colloids Surf. B.
Awasthi, V.D., Garcia, D., Klipper, R., Goins, B.A., Phillips, W.T., 2004. Neutral and Biointerfaces 48, 112–118.
anionic liposome-encapsulated hemoglobin: effect of postinserted poly(ethylene Puri, A., Loomis, K., Smith, B., Lee, J.H., Yavlovich, A., Heldman, E., Blumenthal, R.,
glycol)-distearoylphosphatidylethanolamine on distribution and circulation kinetics. 2009. Lipid-based nanoparticles as pharmaceutical drug carriers: from concepts to
J. Pharmacol. Exp. Ther. 309, 241–248. clinic. Crit. Rev. Ther. Drug Carrier Syst. 26, 523–580.
Balazs, D.A., Godbey, W.T., 2010. Liposomes for use in gene delivery. J. Drug. Deliv. Roco, M.C., 2004. Nanoscale science and engineering: unifying and transforming tools.
2011. AICHE J. 50, 890–897.
Bozzuto, G., Molinari, A., 2015. Liposomes as nanomedical devices. Int. J. Nanomedicine Shaheen, S.M., Ahmed, F.R.S., Hossen, M.N., Ahmed, M., Amran, M.S., Anwar-Ul-Islam,
10, 975–999. M., 2006. Liposome as a carrier for advanced drug delivery. Pak. J. Biol. Sci. 9,
Cattaneo, A.G., Gornati, R., Sabbioni, E., Chiriva-Internati, M., Cobos, E., Jenkins, M.R., 1181–1191.
Bernardini, G., 2010. Nanotechnology and human health: risks and benefits. J. Appl. Shailesh, S., Neelam, S., Sandeep, K., 2009. Liposomes: a review. J. Pharm. Res. 2,
Toxicol. 30, 730–744. 1163–1167.
Chang, H.I., Cheng, M.Y., Yeh, M.K., 2012. Clinically-proven liposome-based drug Shariat, S., Badiee, A., Jaafari, M.R., Mortazavi, S.A., 2014. Optimization of a method to
delivery: formulation, characterization and therapeutic efficacy. Open. Access. Sci. prepare liposomes containing HER2/Neu-derived peptide as a vaccine delivery
Rep. 1, 1–8. system for breast cancer. Iran. J. Pharm. Res. 13, 15–25.
Chen, C.S., Yao, J., Durst, R.A., 2006. Liposome encapsulation of fluorescent Somerharju, P., Virtanen, J.A., Cheng, K.H., 1999. Lateral organisation of membrane
nanoparticles: quantum dots and silica nanoparticles. J. Nanopart. Res. 8, lipids: the superlattice view. Biochim. Biophys. Acta. – Mol. Cell Biol. L. 1440, 32–48.
1033–1038. Stewart, J.C.M., 1980. Colorimetric determination of phospholipids with ammonium
Chithrani, B.D., Ghazani, A.A., Chan, W.C.W., 2006. Determining the size and shape ferrothiocyanate. Anal. Biochem. 104, 10–14.
dependence of gold nanoparticle uptake into mammalian cells. Nano Lett. 6, Szoka, F.D., Papahadjopoulos, D., 1978. Procedure for preparation of liposomes with
662–668. large internal aqueous space and high capture by reverse-phase evaporation. Proc.
Cho, H.Y., Lee, C.K., Lee, Y.B., 2015. Preparation and evaluation of PEGylated and folate- Natl. Acad. Sci. U. S. A. 75, 4194–4198.
PEGylated liposomes containing paclitaxel for lymphatic delivery. J. Nanomater. Wang, B., Zhang, L., Bae, S.C., Granick, S., 2008. Nanoparticle-induced surface
16, 36. reconstruction of phospholipid membranes. Proc. Natl. Acad. Sci. U. S. A. 105,
Colletier, J.P., Chaize, B., Winterhalter, M., Fournier, D., 2002. Protein encapsulation in 18171–18175.
liposomes: efficiency depends on interactions between protein and phospholipid Warner, M.G., Reed, S.M., Hutchison, J.E., 2000. Small, water-soluble, ligand-stabilized
bilayer. BMC Biotechnol. 2, 9. gold nanoparticles synthesized by interfacial ligand exchange reactions. Chem.
De Carlo, S., Harris, J.R., 2011. Negative staining and Cryo-negative staining of Mater. 12, 3316–3320.
macromolecules and viruses for TEM. Micron 42, 117–131. Weare, W.W., Reed, S.M., Warner, M.G., Hutchison, J.E., 2000. Improved synthesis of
Faraji, A.H., Wipf, P., 2009. Nanoparticles in cellular drug delivery. Bioorg. Med. Chem. small (dCORE ≈ 1.5 nm) phosphine-stabilized gold nanoparticles. J. Am. Chem. Soc.
17, 2950–2962. 122, 12890–12891.
Hong, K., Friend, D.S., Glabe, C.G., Papahadjopoulos, D., 1983. Liposomes containing Woodle, M.C., Newman, M.S., Cohen, J.A., 1994. Sterically stabilized liposomes: physical
colloidal gold are a useful probe of liposome-cell interactions. Biochim. Biophys. Acta and biological properties. J. Drug Target. 2, 397–403.
Biomembr. 732, 320–323. Xing, F., Cheng, G., Yang, B., Ma, L., 2004. Microencapsulation of capsaicin by the
Hristozov, D., Malsch, I., 2009. Hazards and risks of engineered nanoparticles for the complex coacervation of gelatin, acacia and tannins. J. Appl. Polym. Sci. 91,
environment and human health. Sustainability 1, 1161–1194. 2669–2675.
Hwang, S.Y., Kim, H.K., Choo, J., Seong, G.H., Hien, T.B.D., Lee, E.K., 2012. Effects of

63

Anda mungkin juga menyukai