You are on page 1of 15

389

Send Orders for Reprints to reprints@benthamscience.net

Pharmaceutical Nanotechnology, 2019, 7, 389-403


RESEARCH ARTICLE
ISSN: 2211-7385
eISSN: 2211-7393

Effect of Acyclovir Solid Lipid Nanoparticles for the Treatment of


Herpes Simplex Virus (HSV) Infection in an Animal Model of
HSV-1 Infection
BENTHAM
SCIENCE

Ritika Kondel1, Nusrat Shafiq1,*, Indu P. Kaur2, Mini P. Singh3, Avaneesh K. Pandey1,
Radha K. Ratho3 and Samir Malhotra1

1
Department of Pharmacology, PGIMER, Chandigarh, 160012, India; 2UIPS, Punjab University,
Chandigarh, 160014, India; 3Department of Virology, PGIMER, Chandigarh, 160012, India

Abstract: Background: Acyclovir use is limited by a high frequency of administra-


tion of five times a day and low bioavailability. This leads to poor patient compliance.
Objectives: To overcome the problem of frequent dosing, we used nanotechnology
platform to evaluate the proof of concept of substituting multiple daily doses of acy-
clovir with a single dose.
Methods: Acyclovir was formulated as solid lipid nanoparticles (SLN). The nanoparti-
Pharmaceutical Nanotechnology

cles were characterized for particle size, surface charge and morphology and in vitro
ARTICLE HISTORY drug release. The pharmacokinetic and pharmacodynamic of SLN acyclovir were
compared with conventional acyclovir in a mouse model.
Received: June 25, 2019
Revised: July 17, 2019
Accepted: August 16, 2019 Results: SLN showed drug loading of 90.22% with 67.44% encapsulation efficiency.
DOI: Particle size was found to be of 131 ± 41.41 nm. In vitro drug release showed 100%
10.2174/2211738507666190829161737 release in SIF in 7 days. AUC0-∞ (119.43 ± 28.74 µg/ml h), AUMC0-∞ (14469 ±
4261.16 µg/ml h) and MRT (120.10 ± 9.21 h) were significantly higher for ACV SLN
as compared to ACV AUC0-∞ (12.22 ± 2.47 µg/ml h), AUMC0-∞ (28.78 ± 30.16 µg/ml
h) and MRT (2.07 ± 1.77 h), respectively (p<0.05). In mouse model, a single dose of
ACV SLN was found to be equivalent to ACV administered as 400mg TID for 5 days
in respect to lesion score and time of healing.
Conclusion: The proof of concept of sustained-release acyclovir enabling
administration as a single dose was thus demonstrated.

Keywords: Acyclovir, HSV-1, pharmacodynamic, pharmacokinetic, Solid Lipid Nanoparticles (SLN),


sustained release.

1. INTRODUCTION Acyclovir was the first specific antiviral drug to


become widely used in the treatment of HSV and
Infections due to herpes simplex virus (HSV) are
herpes zoster virus (HZV) infections [3, 4]. One of
among the most common viral infections [1, 2].
the major limitations of oral acyclovir is that it
needs to be administered 5 times a day due to its
*Address correspondence to this author at the Department of short half-life and low oral bioavailability [5]. This
Pharmacology, PGIMER, Chandigarh, 160012, India; is inconvenient and can lead to poor patient com-
Tel: 0172-2755254; E-mail: nusrat_shafiq@hotmail.com
pliance.

2211-7393/19 $58.00+.00 © 2019 Bentham Science Publishers


390 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 Kondel et al.

We and others have shown that nanotechnolo- 2.2. Preparation of Acyclovir (ACV) Solid Lipid
gy-based drug delivery platforms are able to pro- Nanoparticles (SLN)
vide sustained release plasma profile for various
Solid lipid nanoparticles of acyclovir were
drugs [6-9]. Acyclovir loaded PLGA microparti-
cles have shown increased efficacy against HSV-1 prepared by microemulsion method [20]. Briefly,
in cell culture [10]. Majority platforms include the lipidic phase (8% lipid) and the aqueous phase
polymer-based systems. However, polymer-based (polysorbate 80, soy lecithin and water) were
systems use organic solvent like dichloromethane heated to 10°C above the lipid melt temperature.
(DCM) and acetone which can cause toxicity. Sol- The proportion of surfactant and the volumes of
id lipid nanoparticles (SLN) provide an alternative two phases were so adjusted that a microemulsion
drug delivery platform. It comprises of solid lipids was formed spontaneously upon mixing the two
as matrix for drug delivery which has biodegrada- phases. Hot microemulsion thus formed, was
ble lipids and omits the use of organic solvents transferred into the same amount of cold water
[11]. Camptothecin [12], piribedil [13] and cyclo- (2°C) under constant stirring (Wise Tis HG-15
sporine [14] have been encapsulated in SLN and D, 10,000 rpm) for 20 minutes to obtain SLN. The
have shown sustained release profile. Various an- prepared SLN was used as such for further studies.
tivirals including those for HIV have been encap- Table 1 describes the composition of acyclovir
sulated in SLN and targeted in vivo [15, 16]. Acy- solid lipid nanoparticles having both lipid and
clovir SLN has been developed in various studies, aqueous phase.
however, they have been developed for dermal and
ocular delivery which is not the desired route for Table 1. Composition of acyclovir solid lipid nano-
infections requiring systemic delivery [17-19]. particles.
Therefore, with this background, the current
study was planned to prepare SLN based oral sus- Aqueous Phase Lipid Phase (0.985 g) (4:1)
tained release formulation of acyclovir, evaluate
the pharmacokinetics of the formulation and its Water (4.75 ml) Compritol (0.788 g)
efficacy in an animal model of cutaneous herpes
infection. Drug (40% lipid) (0.394g) Stearic acid (0.197 g)

Tween 80 (6.64g) -
2. MATERIALS AND METHODS
2.1. Materials Soya lecithin (0.4%) -

Acyclovir was purchased from Sigma Aldrich,


Inc. St Louis, MO, USA. Soy lecithin (Phospho- 2.3. Bioanalytical HPLC Method
lipon 90H) was received as a gift sample from Acyclovir was analyzed using HPLC by modi-
Lipoid GmbH, Germany. Compritol 888 ATO®
fication of the previously described method [21].
was a gift sample from Colorcon Asia Pacific pvt.
(Shimadzu Pump LC-20AD, Prominence liquid
Ltd., India. Stearic acid and Tween 80 (poly-
chromatography) with SPD-20A prominence UV-
sorbate 80) were purchased from Central Drug
Vis detector. C-18 column (Hibar 250×4.6 Puro-
House, Mumbai. Chemicals and solvents used
spher STAR, RP-18e (5µm) was used. Serial dilu-
were analytical or HPLC grade. 3-(4,5-Dimethyl-
thiazol-2-yl)-2,5- diphenyl tetrazolium bromide tions within the range of 0.2-2.56 μg/ml were
(MTT) was purchased from Sigma Aldrich, Inc., made from stock solution and standard plot was
St Louis, MO, USA. Dulbecco’s modified Eagle’s obtained. The mobile phase consisted of potassium
medium (DMEM), fetal bovine serum (FBS) and di-hydrogen phosphate and acetonitrile (KH2PO4:
penicillin-streptomycin (50U/ml) hepes buffer 1M, ACN) (97:3) at pH=2.5 The mobile phase was de-
non-essential amino acids, L-glutamine, phosphate livered at a flow rate of 1.5 ml/ min and the detec-
buffer saline pH 7.2 (PBS) were obtained from tion of acyclovir was carried out at 254 nm. The
Gibco-BRL, Spain. Crystal violet were purchased injection volume was 20 µl and the analysis was
from Hi-Media, India. performed at 30°C (sample 4°C).
Effect of Acyclovir Solid Lipid Nanoparticles for the Treatment Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 391

2.4. Solid Lipid Nanoparticles (SLN) Charac- Calculated amount of drug is the quantity of
terization drug which was determined by HPLC and total
amount of drug is the quantity of the drug which is
SLN formulation was characterised for particle
added to prepare the SLN.
size, PDI and zeta potential using Delsa Nano C,
Beckman Coulter, Inc. Double distilled water Encapsulation efficiency (EE) was determined
(DDW) was used as a dispersant medium for by analyzing the clear supernatant obtained by
diluting the sample to 10-1 concentration. The sizes centrifuging the developed SLN dispersions at
of nanoparticles are expressed in nanometer scale. 8.02 lakh×(g) for 2 h at 4°C using Beckman
The polydispersity index (PDI) is the dimension- Coulter Ultracentrifuge (L100K and 100 Ti rotor).
less number which indicates the width of the The EE was calculated as follows:
distribution, having a value between 0 and 1. It is
self-calculated by the software. Zeta potential EE = TDC-Df / TDC ×100
measures the surface charge of the particles and it where Df = amount of drug in the clear
is also measured by DelsaNano C. It is extremely supernatant fluid
important parameter of colloidal stability and
flocculation properties of the colloidal systems. The drug entrapment, drug loading and the in
vitro drug release were expressed as percentages.
Transmission electron microscopy (TEM) analy-
sis of the prepared SLNs was carried out to study Stability study of ACV SLNs was performed.
sphericity and aggregation. For TEM, samples ACV SLNs kept for 1 year at 4 – 8°C. After 1
were examined by TEM (H 100, Hitachi Ltd., year, particle size, PDI, zeta potential, percent
Japan). Samples were stained with phosphotun- drug loading and encapsulation efficiency were
gstic acid (PTA, 2%) for 5 minutes and excess measured. Effect of temperature was observed on
PTA was removed. These were them spread on a the ACV SLNs. As we have to perform the studies
gold grid and examined. Nanoparticles from five on cell cultures, therefore, autoclaving needs to be
to seven different TEM fields were measured to done. ACV SLNs were autoclaved at 121°C, 15
determine the mean particles size of nanoparticles. per square inch (psi) above atmospheric pressure
for 15 minutes. After autoclaving, particle size,
Fourier Transmission Infrared Spectroscopy (FTIR) PDI, zeta potential, percent drug loading and
was conducted on samples of free acyclovir, blank encapsulation efficiency were measured.
SLN and acyclovir SLN to assess the lipid-drug
interaction obtained using a Perkin Elmer spectro- In vitro release was determined in the following
meter (Perkin Elmer, Boston, MA). The FT-IR simulated fluids: simulated gastric fluid (SGF)
spectrums were obtained by mixing the samples pH=1.2; phosphate buffer saline (PBS) pH=6.8;
with Potassium Bromide and pressed to obtain simulated intestinal fluid (SIF) pH=7.8 by dialysis
pellets which were then scanned in the IR range bag method using dialysis membrane (molecular
from 400 to 4000 cm-1, with a resolution of 8 cm-1. weight cut-off of 12 kDa). The incubation took
place in rotating vials at 37°C. 10 µl of samples
The concentration of acyclovir in solid lipid were withdrawn at pre-set time intervals (0.25 hrs,
nanoparticles was determined by HPLC as pre- 0.5 hrs, 1 hrs, 2 hrs, 3 hrs, 4 hrs, 5 hrs, 8 hrs, 12
viously described. The total amount of drug per hrs, 24 hrs, 48 hrs, 60 hrs, 72 hrs, 96 hrs, 120
unit volume present in the formulation was hrs,144 hrs, 168 hrs) and replaced by an equal
determined by suitably disrupting 0.1 ml of the volume of fresh dissolution medium. The aliquots
SLN dispersion in 5 ml chloroform: methanol were then diluted with mobile phase and filtered
(2:1) volumetrically. The total drug content (TDC) through 0.45 µm filters. The samples were
or percent drug loading was determined by using analyzed by HPLC. The cumulative amount of
the equation given below: acyclovir release from the nanoparticles was

TDC = Calculated amount of drug/ml of SLN dispersion ×100


Total amount of drug added/ml of SLN dispersion
392 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 Kondel et al.

calculated using the following equation: essential amino acids and 1% l-glutamine. The
maintenance medium for Vero cells was described
% Cumulative release= Wt / W0 х 100
above with 2% FBS. Herpes Simplex type 1
Wt is the amount of drug released from the (HSV-1) (KOS strain) obtained from National In-
nanoparticles and present in receiving phase at stitute of Virology, Pune, India, was prepared in
time (t) and Wo is the initial amount of drug aliquots and stored at -80°C until use. Virus titer
present in the nanoparticles. The cumulative was determined by plaque assay in Vero cells and
percentage (%) of drug released was plotted expressed as plaque-forming units (pfu) per ml.
against time. The samples were analysed by HPLC
by the method described above to measure the 2.7. Cell Infection
amount of acyclovir in the samples. The titer of the virus stocks, determined by
plaque reduction assay, was 3.3 x 106 plaques/ml.
2.5. Pharmacokinetics Studies Monolayers of Vero cells grown in 24-well tissue
Swiss albino male mice weighing 20-40 g were culture plates, at 80% confluence were infected by
used. The protocol was approved by the adsorption of HSV-1 (300 µL) supplemented with
Institutional Animal Ethics Committee (IAEC), 2% FBS for 1 hr at 37°C. The inoculum with the
PGIMER, Chandigarh (Ref. No. 73/IAEC/444). non-adsorbed virus was removed and cells were
Animals were divided into two groups (n=9), ACV washed with the medium. Finally, the infected
and ACV SLN. They were dosed with 36 mg/kg of cells were treated according to the assay protocol.
acyclovir, either in a solution form of acyclovir or
as ACV SLN orally using an oral feeding canula. 2.8. MTT Assay
Blood samples were collected at pre-determined 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetra-
time points: 0, 15, 30 mins, 1, 2, 4, 8, 12 and 24 zolium bromide (MTT) is a yellow water-soluble
hrs after drug administration for ACV and for 0, tetrazolium dye that is reduced by live cells, but
15, 30 mins, 1, 2, 4, 8, 12 and 24, 48, 72, 96, 120, not by dead ones, to a purple formazan product
144 and 168 hrs after drug administration for ACV that is insoluble in aqueous solutions. Monolayers
SLN. Plasma was separated out by centrifuging of 1x103 Vero cells were placed in 96 well plates.
the blood samples at 4000 rpm for 10 min at 4°C. Treatment was done on these plates with ACV and
Protein precipitation was carried out by adding ACV SLN over a concentration range of 2.5-250
100 µl of 20% perchloric acid in 500 µl of sample, µg/ml and the plates were incubated for overnight
vortexing it for 1 min and then centrifuging the at 37°C in 5% CO2. MTT (20 µl of 5 mg/ml) was
samples at 10,000 rpm for 15 min [22]. The super- added to each well plate and incubated for 4 hrs.
natant was collected and then analyzed by stand- Media was discarded and DMSO (200 µl) was
ardized HPLC method. Cmax (Peak plasma added. Absorbance in each well plate was meas-
concentration) and Tmax (time to reach peak plasma ured at 490 nm.
concentration) were obtained from actual plasma
concentration versus time data. The area under the 2.9. Plaque Reduction Assay
curve (AUC) was calculated using the trapezoidal
rule, from the plasma vs. time plots. AUC0-∞, total ACV and ACV SLN were tested for antiviral
AUC and mean residence time (MRT) were activity against HSV-1 by plaque reduction assay
calculated from the individual AUCs from the over a concentration range of 0.25µM-8 µM.
plasma-time plots. 1x105 Vero cells were seeded in 24-well plates and
incubated at 37°C and 5% CO2. When the cells
2.6. Cell Culture and Viruses reached 70% confluence, they were infected with
300 µl of 3.3x106 pfu/ml of HSV-1. After
Vero cells (African green monkey kidney cell incubation for 1 hr at 37°C to allow viral
line) were purchased from the National Institute of adsorption, the plates were washed, and the
Virology, Pune, India. This cell line was grown in medium replaced with maintenance medium
DMEM supplemented with 6% FBS, 1% penicil- containing different concentrations of ACV and
lin-streptomycin, 1% Hepes buffer 1M, 1% non- ACV SLN. After 96 hrs incubation, the medium
Effect of Acyclovir Solid Lipid Nanoparticles for the Treatment Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 393

Fig. (1). Calibration curve of standard acyclovir in mice plasma. (A higher resolution / colour version of this figure
is available in the electronic copy of the article).

was removed, and the monolayers were fixed with and results of plaque reduction assay between the
10% formaldehyde (200 µl) for 1 hr and then stai- two groups were analysed by unpaired t-test. For
ned with 300 µl of 1% solution of crystal violet in lesion scoring in animal model, among the control,
30% methanol for 1 hr. Percent plaque reduction ACV and ACV SLN group. Data were analysed by
from control was calculated for both the treatment ANOVA and post-hoc Bonferroni test was used
groups. for between-group comparisons. p value of <0.05
was considered significant.
2.10. Evaluation of Antiviral Activity in Cuta-
neous Model of HSV-1 Infection 3. RESULTS
Female BALB/c mice (6 weeks old) were used. 3.1. HPLC Estimation
The right mid flank of each mouse was depilated.
One day later, the skin was scratched using a 27- ACV was standardized for estimation by
gauge needle and 40 µL of HSV-1 suspension of HPLC. A standard curve was obtained with 0.2-
3.3×106 pfu /ml was applied to the scarified area 25.6 µg/ml concentration range. The correlation
[23-25]. ACV and ACV SLNs were orally admin- coefficient value of 0.99 was obtained. The stand-
istered as an oral gavage 24 h after HSV-1 infec- ard curve was found to be eligible for the PK-PD
tion. ACV (400 mg) was given thrice a day for 5 studies as acyclovir is active against HSV-1 in
concentration range of 0.02-0.9 µg/ ml (Fig. 1).
days whereas ACV SLN was a single dose admin-
Recoveries of acyclovir from plasma samples were
istration (400mg). Beginning 1 day after infection,
found to be 88.11 ± 0.27% which confirms less
the abraded site was evaluated and scored as fol-
sample loss. Inter-day and intra-day variability
lows: 0, no visible change in the abrasion or surr-
were also found to be within range (15%) [26].
ounding tissue; 0.5, the earliest lesion characteri-
sed by slight swelling near the abrasion and mild 3.2. Characterization of ACV Solid Lipid
erythema; 1, papule(s) at or around the abrasion; 2, Nanoparticles
ulcerated papules at or around the abrasion site with
eschar formation; 3, fusion of ulcerated lesions ACV SLN were prepared by microemulsion
into large eschar; 4, large open ulcerated lesion; 5, technique by optimizing the ratio of the lipid phase
death or sacrifice. The animals were evaluated and aqueous phase. ACV SLN had a size of 131 ±
daily for 11 days beginning 1 day after infection. 41.41nm with a polydispersity index (PDI) of 0.30
± 0.014. Zeta potential was found out to be -16.00
2.11. Statistics ± 1.90 mV (Table 2).
PK data were expressed as mean ± standard TEM micrographs showed smooth spherical
deviation (SD). The difference in PK parameters surface morphology and non-aggregated nanopar-
394 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 Kondel et al.

Table 2. Results of parameters used for characterization of nanoparticles PDI, particle size, zeta potential
results are of 3 sets of experiments.

Percent Drug Percent Encapsula-


Particle Size (nm) Zeta Potential (mV) PDI
Loading tion Efficiency
(Mean ± S.D) (Mean ± S.D) (Mean ± S.D)
(Mean ± S.D) (Mean ± S.D)

131 ± 41.44 -16 ± 1.90 0.30 ± 0.014 90.22 ± 1.18% 67.44 ± 6.21%

103.7 nm

Focused view

drug slnzd.tif
Print Mag: 347000x @ 7.0 in 100 nm
2:35:50 p 04/24/15 HV = 80.0kV
Direct Mag: 200000x
AMT Camera System

Fig. (2). Transmission Electron Microscopy (TEM) micrograph of Acyclovir SLN showing spherical nanoparticles.
(A higher resolution / colour version of this figure is available in the electronic copy of the article).

ticles (Fig. 2). The drug loading and encapsulation respectively in the first 24 hours. 87% and 95% of
efficiency were found out to be 90.22 ± 1.18% and entrapped drug were released until 7 days in PBS
67.44 ± 6.21% respectively. In ACV SLNs, FT-IR and SGF. 100% drug release was observed in SIF
spectra peak shifted from 3521 cm-1 to 3404 cm-1 in 7 days (Fig. 4).
in -NH stretch, and shift in 1485 cm-1 to 1472 cm-1
due to C-OH group was also noted (Fig. 3) (Table 3). 3.3. Stability of ACV SLN
In vitro drug release study demonstrated a biphasic Stability of ACV SLN was assessed for drug
release pattern where it was observed that there loading, particle size, polydispersity index and
was an initial burst release of drug followed by a zeta potential. After storage for 1 year, percent
sustained release. An initial burst release of a drug drug loading was 85%. Particle size, PDI and zeta
was 51%, 45% and 52% in SIF, PBS and SGF potential changed to 152 nm, -31.11 and 0.318,
Effect of Acyclovir Solid Lipid Nanoparticles for the Treatment Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 395

1960.63
784.57
8090.53 73.56 755.56
1538.54 1417.42 719.53630.52
3185.46 1601.45 885.4 679.52 532.48
259.4 561.55
3309.40 1001.40 545.44
3430.16 1629.34 1387.37
3404.37 1472.30 1350.29 959.31
1463.29 1297.53
1736.22 1250.29

2870.1 1966.72 719.71 526.71


2850.5 1643.67 847.66
%T 2916.5 1111.6
1298.60 950.58
3437.56 1463.57 1350.57 1249.58
1736.53

2850.39
107.35
2917.33 2102.16 416.18
650.13 506.13
1262.11 867.11 575.9
3521.5 2928.4 1309.6 1217.6 1012.6 901.5 779.6 652.6
3309.1 2856.4 1611.1 1455.2 1539.2 1183.3 1005.2 753.6 627.6
3471.1 3184.1 2790.4 1715.0 1577.1 1345.4 1105.1
3441.1 2716.3 1632.0 1541.3 1049.3

4000.0 3600 3200 2800 2400 2000 1800 1600 1400 1200 1000 800 600 450.0
cm-1
Ritika Kondel-1-sp- 1/11/2016 - ACV
Ritika Kondel-2-sp- 1/11/2016 - BSNS
Ritika Kondel-3-sp- 1/11/2016 - ACV SCNS

Fig. (3). FT-IR spectra of acyclovir SLNs, Blank SLNs and free acyclovir. (A higher resolution / colour version of
this figure is available in the electronic copy of the article).

Fig. (4). In vitro release of Acyclovir SLN in three simulated fluids; SIF (pH=7.8), SGF (pH=1.2), PBS (pH= 6.8).
(A higher resolution / colour version of this figure is available in the electronic copy of the article).

Table 3. Major bands of FT-IR spectra of acyclovir, blank SLNs and acyclovir SLNs.

Band Acyclovir Blank SLNs Acyclovir SLNs Functional Groups (Frequency Range)

1 3521.3 3437.56 3404.37, 3436. 36 -NH stretch (3100-3500 cm-1

2 1485.2 - 1472. 30 -C-OH group (1500-1065 cm-1)

3 1715.0 1736.53 1736.53 -C=O group (1700-1800)

4 3309.1 - 3309.40 -OH stretch (3200-3500 cm-1)


396 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 Kondel et al.

Table 4. Characterisation of formulation before and after 1 year of storage at 4-8°C and effect of sterilisation
on formulation.

Parameters Initial Final After Sterilisation

Particle Size (nm) 131.37 ± 41.44 151.9 ± 3.31 246.6 ± 16.2*


PDI 0.30 ± 0.014 0.315 ± 0.023 0.229 ± .079
*
Zeta potential -16.03 ± 1.63 -30.6 ± 1.89 -12.0 ± 3.57
% Drug loading 90.3 ± 1.18 85 ± 2.44 85 ± 2.18
*
Values are expressed as mean ± S.D; p<0.05.

Fig. (5). Plasma concentration- time profile of orally administered free acyclovir (36 mg/kg) and acyclovir solid
lipid nanoparticles (equivalent 36 mg/kg of acyclovir in SLNs) in mice dosed orally. (n=3) mice were taken at each
time point from 0 hour to 168 hours. Time points were shown as logarithmic scale. (A higher resolution / colour
version of this figure is available in the electronic copy of the article).

respectively (Table 4). Sterilisation did not have for ACV SLNs as compared to ACV (3.59 ± 3.00
any effect on percent drug loading, though it did vs. 10.54 ± 5.46 µg/ ml). The AUC0-∞ (119.43 ±
increase the particle size of SLN from 13 nm to 28.74 µg/ ml h), AUMC 0-∞ (14469 ± 4261.16 µg/
256.6 nm. ml h) and MRT (120.10 ± 9.21 hrs) were signifi-
cantly higher for ACV SLN as compared to ACV
3.4. Comparative Assessment of Pharmacoki- AUC0-∞ (12.22 ± 2.47 µg/ ml hrs), AUMC0-∞
netics of Free and Nanoformulation of Acyclo- (28.78 ± 30.16 µg/ ml hrs) and MRT (2.07 ± 1.77
vir in Mice hrs), respectively (p<0.05, Table 5) (Fig. 5).
ACV SLN demonstrated a prolonged release of
drug for up to 7 days in comparison to ACV (Ta- 3.5. Pharmacokinetic/Pharmacodynamic Index
ble 5). The drug levels observed in both the groups of Acyclovir Solid Lipid Nanoparticles
corresponds to a concentration which is needed for EC50 against HSV-1 isolates ranged from (0.02
antiviral activity (0.02-0.9 µg/ ml) [5]. Plasma to 13.5 µg/ mL) [5, 27, 28]. Time above EC50 was
concentrations were maintained for up to 7 days in found to be significantly higher for ACV SLN
ACV SLN as compared to 2.5 hrs in the ACV equivalent to (36 mg/kg) of acyclovir as compared
group. The Cmax was significantly decreased (p<0.05) to ACV (168 hours v/s 4 hours) (Table 6).
Effect of Acyclovir Solid Lipid Nanoparticles for the Treatment Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 397

Table 5. Pharmacokinetic parameters of acyclovir and acyclovir solid lipid nanoparticles (Cmax, Tmax, AUC,
AUMC, MRT; n=3).

AUC AUMC
Cmax(µg/ml) Tmax(h) AUC (µg/ml*h)(0-24) MRT (h)
Group (µg/ml*h) (0-∞) (µg/ml* h)(0-∞)
(Mean ± S.D) (Mean ± S.D) (Mean ± S.D) (Mean ± S.D)
(Mean ± S.D) (Mean ± S.D)

ACV 10.54 ± 5.46 0.33 ± 0.144 15.11 ± 3.21 10.56 ± 1.30 28.78 ± 30.16 2.07 ± 1.77
ACV SLN 3.59 ± 3.00 1±0 10.14 ± 1.21 119.43 ± 28.74 14469.73 ± 4261 120.10 ± 9.21

Fig. (6). Effect of Acyclovir and Acyclovir SLN on non-infected Vero cells viability at different drug concentration
demonstrating cell toxicity. Each point represents the mean ± S.D (n=3). (A higher resolution / colour version of
this figure is available in the electronic copy of the article).

Table 6. Time above EC50 for free acyclovir and control. At higher concentration of ACV tested,
acyclovir solid lipid nanoparticles (Acyclo- 250 µg/ml, cell viability was 19.1% (Fig. 6). How-
vir SLNs) at an EC50 (0.02-13.5 µg/ml). ever, at 100 µg/ml, cell viability observed was
60.5% (Fig. 6). Similar results were obtained for
ACV SLN and no significant difference was ob-
Drugs Time>EC50 (Hours)
served between the two treatment groups. Cytotox-
Free acyclovir (36 mg/kg) 4 icity was observed at concentration above ≤100
µg/ ml for both the treatment groups.
Acyclovir NPs
168
(equivalent to 36 mg/kg) 3.6.2. Plaque Reduction Assay
The antiviral efficacy of ACV SLN against
3.6. Comparative Antiviral Efficacy of ACV HSV-1 was assessed by plaque reduction assay in
and ACV SLN a monolayer of Vero cells. ACV showed a concen-
tration-dependent response on plaque formation.
3.6.1. Cellular Toxicity
As shown in Fig. (4), ACV decreased the percent
To investigate cytotoxicity, uninfected Vero (%) plaque count from 61% at the lowest (0.25
cells were treated with ACV and ACV SLN and µM) to 2% at the highest concentration (8 µM).
then subjected to cell viability assay. The amount On the other hand, ACV SLN showed better
of surviving cells after incubation with an ACV antiviral efficacy at lowest concentration (0.25
and ACV SLN during a 24 hours incubation was µM) (Fig. 7) as no plaque formation occurred in
estimated by MTT assay. The viability was ex- wells having ACV SLN in concentration range of
pressed in percent by comparison with non-treated (0.5 µM - 8 µM) (p<0.05). The effective concen-
398 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 Kondel et al.

Fig. (7). Plaque reduction assay at different drug concentration of Acyclovir and Acyclovir SLN. Percentage plaque
formation reduction from the control was shown as percentage HSV-1 plaques formed in both the treatment groups.
(A higher resolution / colour version of this figure is available in the electronic copy of the article).

Fig. (8). Comparative pharmacodynamic activity of Acyclovir and Acyclovir SLN was shown. Treatment of HSV-1
infected mice was given after 24h infection. Control mice were given saline. ACV was given 3 times a day treat-
ment for consecutive 5 days. ACV SLNs were given once administration during five days (n=6). (A higher resolu-
tion / colour version of this figure is available in the electronic copy of the article).

tration to achieve 50% protection against virus significant difference observed in lesion scores
replication (EC50) when using ACV SLN and between all the treatment groups. On the subse-
ACV was 0.003 µM and 0.296 µM, respectively. quent days, significant difference was observed in
both the treatment groups when compared to the
3.6.3. Cutaneous Mouse Model of HSV-1 Infec- saline-treated group (p<0.005) (Fig. 8). Time of
tion healing was also found to be significantly dec-
reased in treatment groups as compared to control
For this purpose, infected mice were divided
group (9 days vs. 4 days). When ACV and ACV
into three groups and were treated with saline,
SLNs groups were compared, no significant differ-
ACV and ACV SLNs after 24 hours of infection.
ence was observed in lesion scoring and time of
Lesion development in mice was assessed for 11
healing (Fig. 9).
days. It was observed that as compared to the
saline-treated mice, treatment with ACV and ACV 4. DISCUSSION
SLNs both significantly reduced the lesion score in
infected mice and also reduced the time of healing In the current study, we developed solid lipid
of lesions (p<0.005). At day 1, there was no nanoparticles (SLNs) of acyclovir containing
Effect of Acyclovir Solid Lipid Nanoparticles for the Treatment Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 399

Fig. (9). Representative images depicting HSV lesions in BalB/C mice at day 3. A) Control mice given saline; B)
Free acyclovir group mice given 3 times a day administration for 5 days; C) Acyclovir SLNs group mice given
once for five days; D,E,F focused images of above images above groups. (A higher resolution / colour version of
this figure is available in the electronic copy of the article).

suitable lipids and surfactants which are biodeg- 200 nm is desirable for sustained release action as
radable and biocompatible. they remain inaccessible to the reticuloendothelial
system (RES) and can show prolonged action [34].
4.1. Preparation and Characterisation of Acy- A high total drug capacity holds promise in the
clovir Solid Lipid Nanoparticles drug development process as scaling up the for-
We were able to achieve a drug loading of mulation may not be a wasteful excess [34]. The
67.44%. Previously, Seyfoddin A 2013, have pre- suspension was also found to be stable over one-
pared acyclovir SLNs the microemulsion tech- year showing non-significant change in particle
nique and have used Tween 80 as a surfactant and size and total drug loading. The nanoparticles
have reported encapsulation efficiency of 13.32% exhibited low negative zeta potential of -16 ± 1.90
[29]. They have tried with different lipids and mV which could be due to terminal carboxylic
different drug lipid ratio to get the best encap- group of stearic acid. Under TEM, nanoparticles
sulation efficiency. We have used compritol 888 had smooth surface and were found to be spherical
ATO as a lipid and Tween 80 as a surfactant to in shape. The particle size observed with TEM
prepare microemulsion. The high encapsulation micrographs corresponds well with number distri-
efficiency of 67.44% demonstrates that the lipid bution pattern. FTIR spectra of acyclovir showed
combination used have synergistic interaction with an -NH stretch at 3521.3 cm-1, -C=O stretch at
the drug molecule. By combining the two lipids, it 1715.0 cm-1 and -C-OH stretch at 1485 cm-1. The
resulted in better incorporation of acyclovir into shift in the stretch of these groups indicates
the lipid mix melt into the empty spaces of the interaction with the lipids. In ACV SLNs groups a
imperfect lipid matrix. Most of the other studies higher shift was observed at all these groups which
which have developed acyclovir nanoparticles suggest an interaction of acyclovir with compritol
have developed using different lipids and most of 888 ATO and stearic acid both which resulted in
them have not prepared nanoparticles for oral good encapsulation efficiency of 67.44%. In vitro
delivery [30-33]. drug release of ACV SLN showed an initial phase
of burst release of drug in first 24 hours due to the
Acyclovir solid lipid nanoparticles had an presence of unentrapped drug onto the nanopar-
average size of 131 ± 41.41 nm with total drug ticles. Afterwards, it showed a sustained release of
loading (TDC) of 90.22%. Particle size less than drug by diffusion from the lipid core, wherein the
400 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 Kondel et al.

drug was homogeneously distributed. A slow 4.3. Pharmacodynamics of Acyclovir Solid Lipid
release of drug is may be accounted by the high Nanoparticles
lipophilicity of the compritol 888 ATO. Higher
Efficacy studies were carried out both in cell
carbon chain length of compritol AT0 888
culture by plaque reduction assay and in the
contribute to higher sustained release [35].
animal model of cutaneous HSV-1 infection. 50%
Further, amorphous nature of formulation leads to
inhibition observed with the treatment groups
high solubility [35].
corresponds to the value which is above the
4.2. Pharmacokinetics of Acyclovir Solid Lipid reported EC50 value (0.02-13.5 µg/ml ≈ 0.08-60
Nanoparticles µM). Approximately 100 times improvement in
the efficacy was observed in the ACV SLNs group
Oral administration of acyclovir solid lipid when compared to ACV alone. This improvement
nanoparticles led to a prolonged drug release with in antiviral efficacy could be due to the longer
levels above EC50 for 7 days in comparison to 4 duration of contact of the drug-loaded SLNs with
hours in the free drug group. Several reasons may the infected cells [10]. Jalon et al., 2003 have also
be given for improved oral bioavailability of shown a significant decrease in IC50 value (0.15 ±
acyclovir after administration of acyclovir solid 0.03 µM), in cell plates treated with ACV
lipid nanoparticles. Firstly, smaller particle size microparticles as compared to drug solution alone
increases the surface area of particles itself which (0.27 ± 0.02 µM). The difference in EC50 value of
leads to increased absorption from the gastro- the reported study, when compared to our study,
intestinal tract [36]. Bioadhesion of SLN to the maybe due to the reason that we are using
mucosa of the gastrointestinal tract due to lipid nanoparticles unlike microparticles used in their
moieties may increase the residence time at the gut study. Similar studies also showed increase
wall. This close contact with the epithelial mem- efficacy of drugs incorporated in the SLN through
branes may enhance the drug absorption thus increased cellular uptake in the cells [41-44].
leading to increased oral bioavailability [36]. Efficacy study done in an animal model of
Secondly, absorption can also be facilitated by cutaneous herpes infection showed that acyclovir
presence of fatty acids and mono or diacylgly- given three times a day for consecutive five days
cerols being converted by triglycerides present in was comparable with a single dose of acyclovir
the nanoformulation by gastrointestinal lipase solid lipid nanoparticles. The lesions observed in
[37]. Thirdly, lipids present in SLN increase lipo- control animals are scored up to 3 which corres-
protein formation which stimulates lymphatic lipid ponds to large lesions fused into large eschar,
flux. This increased lymphatic delivery of drug whereas, for the treatment groups maximum lesion
leading to enhanced bioavailability. Lastly, nano- score of 2 was observed. The score of 2 corres-
particles are preferentially taken by the M-cells ponds to small ulcerated papules leading to eschar
present in the Peyer's patches of the gut-associated formation. Time of healing was also found to be
lymphoid tissue (GALT) followed by shuttling decreased in treatment groups as compared to
into the systemic circulation and release of drug in control group (9 days vs. 4 days). However, no
the blood. This specialised uptake mechanism can significant difference was observed in lesion sco-
also improve absorption of drug [38]. Similar ring and time of healing (p<1.000). The pharma-
studies have shown increased bioavailability of cokinetic study observed significant increase in
drug encapsulated in solid lipid nanoparticles [37, MRT in the ACV SLN group when compared to
39, 40]. A significant increase in the MRT of acy- ACV alone (120.10 h vs. 2.07 h). The efficacy
clovir in case of acyclovir solid lipid nanoparticles results observed are in accordance with the phar-
as compared to free acyclovir was due to change in macokinetics results which demonstrate that the
the acyclovir absorption and elimination after drug levels were maintained above EC50 for up to
encapsulating into solid lipid nanoparticles. This 7 days in ACV SLN group as compared to 4 hours
may be also because of slow release of drug from in ACV alone group. This could be a valid reason
the nanoparticles due to higher biodegradation for the comparable effect shown when free was
time of acyclovir solid lipid nanoparticles [40]. given acyclovir given 3 times a day for five days
Effect of Acyclovir Solid Lipid Nanoparticles for the Treatment Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 401

and a single dose of ACV SLN. The animal model CONFLICT OF INTEREST
of cutaneous herpes used in our study is a well-
The authors confirm that this article content has
validated model and has been used to evaluate ef-
no conflict of interest.
ficacy in HSV-1 and HSV-2 infections models in
various studies [45, 46].
ACKNOWLEDGEMENTS
There are limitations to our current study. We
were not able to show the antiviral efficacy of our Declared none.
nanoparticles through a genotypic assessment like
polymerase chain reaction (PCR) and western REFERENCES
blotting. We have shown the efficacy in one ani- [1] Arvin A, Campadelli-Fiume G, Mocarski E, et al.
mal model of herpes infections. Screening needs to Human herpesviruses: biology, therapy, and im-
be done on different models of herpes infections. munoprophylaxis. Cambridge University Press: Cam-
bridge 2007.
[2] Hay CM, Reichman RC. Antiviral drugs. In: Gold-
CONCLUSION smith LA, Katz SI, Gilchrest BA, Paller AS, Leffell
In conclusion, our study showed that the DJ, Wolff K, Eds. Fitzpatrick’s Dermatology in Gen-
developed single dose acyclovir solid lipid eral Medicine, 7th ed. McGraw-Hill: New York 2008;
pp. 2203-11.
nanoparticles have shown comparative efficacy to [3] Chang YC, Madkan VK, Sra K, Carrasco DA, Tyring
the multiple-dose regimen of conventional SK. Systemic antiviral agents In: Wolff K, Ed. Com-
acyclovir. They found to be potential carriers for prehensive Dermatologic Drug Therapy. WB Saun-
oral therapy in the treatment of HSV-1 infection. ders: Philadelphia 2007; pp. 101-24.
In future, we will plan for the ACV SLN toxicity [4] Dollery C. Therapeutic drugs. 2nd ed. Churchill Liv-
studies and its efficacy studies in HSV-2 infection. ingstone: Edinburgh 1999; pp. A39-44.
[5] Goodman L, Gilman A, Brunton L, Lazo J, Parker K.
Goodman & Gilman’s the pharmacological basis of
ETHICS APPROVAL AND CONSENT TO therapeutics, 13th ed. McGraw-Hill: New York 2006;
PARTICIPATE pp. 1106-7.
Ethical approval from the Institutional Animal [6] Arora A, Shafiq N, Jain S, Khuller GK, Sharma S,
Malhotra S. Development of sustained release
Ethics Committee, PGIMER, Chandigarh, India “NANOFDC (fixed dose combination)” for hyperten-
(Ref. No. 73/IAEC/444). sion - an experimental study. PLoS One 2015; 10(6):
e0128208.
HUMAN AND ANIMAL RIGHTS [7] Arora A, Shafiq N, Jain S, et al. Development of sus-
tained release “Nanopolypill” of ischemic heart dis-
No humans were used. We have followed ease drugs - an experimental study. Curr Nanosci
OECD guidelines for the PK-PD experiments and 2014; 10(6): 816-26.
the protocol was approved by the Institute Animal [8] Kumar G, Malhotra S, Shafiq N, Pandhi P, Khuller
Ethics committee. GK, Sharma S. In vitro physicochemical characteriza-
tion and short term in vivo tolerability study of ethi-
CONSENT FOR PUBLICATION onamide loaded PLGA nanoparticles: potentially ef-
fective agent for multidrug resistant tuberculosis. J
Not applicable. Microencapsul 2011; 28(8): 717-28.
[9] Kumar G, Sharma S, Shafiq N, Khuller GK, Malhotra
AVAILABILITY OF DATA AND MATERIALS S. Optimization, in vitro-in vivo evaluation, and short-
term tolerability of novel levofloxacin-loaded PLGA
The raw data that support the finding of the nanoparticle formulation. J Pharm Sci 2012; 101(6):
results of this study are available from the 2165-76.
corresponding author, [Dr. NS], and the first au- [10] de Jalón EG, Blanco-Príeto MJ, Ygartua P, Santoyo
S. Increased efficacy of acyclovir-loaded microparti-
thor [Dr. RK] upon request.
cles against herpes simplex virus type 1 in cell cul-
ture. Eur J Pharm Biopharm 2003; 56(2): 183-7.
FUNDING [11] Müller RH, Mäder K, Gohla S. Solid lipid nanoparti-
The work was funded by Life Science Research cles (SLN) for controlled drug delivery - a review of
the state of the art. Eur J Pharm Biopharm 2000;
Board (LSRB), DRDO, New Delhi, India (No. 50(1): 161-77.
DLS/81/4822/LSRB-257/SH&DD/2012).
402 Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 Kondel et al.

[12] Yang S, Zhu J, Lu Y, Liang B, Yang C. Body distri- [27] Food Drug Administration. Zovirax®. Available at:
bution of camptothecin solid lipid nanoparticles after https://www.accessdata.fda.gov/drugsatfda_docs/label
oral administration. Pharm Res 1999; 16(5): 751-7. /2001/018604s018lbl.pdf
[13] Demirel M, Yazan Y, Müller RH, Kiliç F, Bozan B. [28] O’Brien JJ, Campoli-Richards DM. Acyclovir. An
Formulation and in vitro-in vivo evaluation of updated review of its antiviral activity, pharmacoki-
piribedil solid lipid micro- and nanoparticles. J Mi- netic properties and therapeutic efficacy. Drugs 1989;
croencapsul 2001; 18(3): 359-71. 37(3): 233-309.
[14] Penkler L, MuÈller RH, Runge SA. Pharmaceutical [29] Seyfoddin A, Al-Kassas R. Development of solid
cyclosporin formulation with improved biopharma- lipid nanoparticles and nanostructured lipid carriers
ceutical properties, improved physical quality and for improving ocular delivery of acyclovir. Drug Dev
greater stability, and method for producing said for- Ind Pharm 2013; 39(4): 508-19.
mulation United States Patent US 6551619B1, 1999. [30] Hasanovic A, Zehl M, Reznicek G, Valenta C. Chi-
[15] Abd-Rabou AA, Bharali DJ, Mousa SA. Taribavirin tosan-tripolyphosphate nanoparticles as a possible
and 5-fluorouracil-loaded pegylated-lipid nanoparticle skin drug delivery system for aciclovir with enhanced
synthesis, p38 docking, and antiproliferative effects stability. J Pharm Pharmacol 2009; 61(12): 1609-16.
on MCF-7 breast cancer. Pharm Res 2018; 35(4): 76. [31] Cortesi R, Ravani L, Menegatti E, Drechsler M, Es-
[16] Jain AK, Jai SK. Galactosylated poly (d, l-lactic-co- posito E. Colloidal dispersions for the delivery of
glycolic acid) nanoparticles for liver targeted delivery acyclovir: a comparative study. Indian J Pharm Sci
of acyclovir. J Biomed Pharm Res 2013; 2: 7-14. 2011; 73(6): 687-93.
[17] Bhalekar MR, Upadhaya PG, Madgulkar AR, Kshir- [32] Bhosale U, Kusum DV, Jain N. Formulation and op-
sagar SJ, Dube A, Bartakke US. In vivo bioavailabil- timization of mucoadhesive nanodrug delivery system
ity and lymphatic uptake evaluation of lipid nanopar- of acyclovir. J Young Pharm 2011; 3(4): 275-83.
ticulates of darunavir. Drug Deliv 2016; 23(7): 2581- [33] AK, Jain, J. S. Galactosylated poly (d, l-lactic-co-
6. glycolic acid) nanoparticles for liver targeted delivery
[18] Cavalli R, Donalisio M, Bisazza A, et al. Enhanced of acyclovir. J Biomed Pharm Res 2013; 2: 7-1.
antiviral activity of acyclovir loaded into nanoparti- [34] Rohit B, Pal KI. A method to prepare solid lipid na-
cles. Methods Enzymol 2012; 509: 1-19. noparticles with improved entrapment efficiency of
[19] Seyfoddin A, Al-Kassas R. Development of solid hydrophilic drugs. Curr Nanosci 2013; 9: 29-33.
lipid nanoparticles and nanostructured lipid carriers [35] Aburahma MH, Badr-Eldin SM. Compritol 888 ATO:
for improving ocular delivery of acyclovir. Drug Dev a multifunctional lipid excipient in drug delivery sys-
Ind Pharm 2013; 39(4): 508-19. tems and nanopharmaceuticals. Expert Opin Drug De-
[20] Bhandari R, Kaur IP. Pharmacokinetics, tissue distri- liv 2014; 11(12): 1865-83.
bution and relative bioavailability of isoniazid-solid [36] Qi J, Lu Y, Wu W. Absorption, disposition and
lipid nanoparticles. Int J Pharm 2013; 441(1-2): 202- pharmacokinetics of solid lipid nanoparticles. Curr
12. Drug Metab 2012; 13(4): 418-28.
[21] Emami J, Bazargan N, Ajami A. HPLC determination [37] Hu L, Tang X, Cui F. Solid lipid nanoparticles
of acyclovir in human serum and its application in bi- (SLNs) to improve oral bioavailability of poorly sol-
oavailability studies. Res Pharm Sci 2009; 4: 47-54. uble drugs. J Pharm Pharmacol 2004; 56(12): 1527-
[22] Brown SD, White CA, Chu CK, Bartlett MG. Deter- 35.
mination of acyclovir in maternal plasma, amniotic [38] Italia JL, Yahya MM, Singh D, Ravi Kumar MNV.
fluid, fetal and placental tissues by high-performance Biodegradable nanoparticles improve oral bioavaila-
liquid chromatography. J Chromatogr B Analyt bility of amphotericin B and show reduced ne-
Technol Biomed Life Sci 2002; 772(2): 327-34. phrotoxicity compared to intravenous fungizone.
[23] Lipipun V, Kurokawa M, Suttisri R, et al. Efficacy of Pharm Res 2009; 26(6): 1324-31.
Thai medicinal plant extracts against herpes simplex [39] Zhang Z, Lv H, Zhou J. Novel solid lipid nanoparti-
virus type 1 infection in vitro and in vivo. Antiviral cles as carriers for oral administration of insulin.
Res 2003; 60(3): 175-80. Pharmazie 2009; 64(9): 574-8.
[24] Docherty JJ, Smith JS, Fu MM, Stoner T, Booth T. [40] Müller RH, Runge S, Ravelli V, Mehnert W, Thüne-
Effect of topically applied resveratrol on cutaneous mann AF, Souto EB. Oral bioavailability of cyclo-
herpes simplex virus infections in hairless mice. An- sporine: solid lipid nanoparticles (SLN) versus drug
tiviral Res 2004; 61(1): 19-26. nanocrystals. Int J Pharm 2006; 317(1): 82-9.
[25] Kurokawa M, Ochiai H, Nagasaka K, et al. Antiviral [41] Zhuang CY, Li N, Wang M, et al. Preparation and
traditional medicines against herpes simplex virus characterization of vinpocetine loaded nanostructured
(HSV-1), poliovirus, and measles virus in vitro and lipid carriers (NLC) for improved oral bioavailability.
their therapeutic efficacies for HSV-1 infection in Int J Pharm 2010; 394(1-2): 179-85.
mice. Antiviral Res 1993; 22(2-3): 175-88. [42] Hariharan S, Bhardwaj V, Bala I, Sitterberg J,
[26] Guidelines for the validation of analytical methods Bakowsky U, Ravi Kumar MNV. Design of estradiol
used in residue depletion studies www.ema.europa. loaded PLGA nanoparticulate formulations: a poten-
eu/docs/en_GB/document...guideline/2010/.../WC500 tial oral delivery system for hormone therapy. Pharm
040499.pdf2010. Res 2006; 23(1): 184-95.
Effect of Acyclovir Solid Lipid Nanoparticles for the Treatment Pharmaceutical Nanotechnology, 2019, Vol. 7, No. 5 403

[43] Sharma CP, Kalarikkal N, Sandeep K. Thomas S, [45] Mosallaei N, Jaafari MR, Hanafi-Bojd MY, Golmo-
Pothen LA. Evaluation of in vitro cytotoxicity and hammadzadeh S, Malaekeh-Nikouei B. Docetaxel-
cellular uptake efficiency of zidovudine-loaded solid loaded solid lipid nanoparticles: preparation, charac-
lipid nanoparticles modified with aloe vera in glioma terization, in vitro, and in vivo evaluations. J Pharm
cells. Mater Sci Eng C 2016; 66: 40-50. Sci 2013; 102(6): 1994-2004.
[44] Baek JS, Cho CW. Surface modification of solid lipid [46] Ahmadnia S, Moazeni M, Mohammadi-Samani S,
nanoparticles for oral delivery of curcumin: im- Oryan A. In vivo evaluation of the efficacy of albend-
provement of bioavailability through enhanced cellu- azole sulfoxide and albendazole sulfoxide loaded sol-
lar uptake, and lymphatic uptake. Eur J Pharm Bio- id lipid nanoparticles against hydatid cyst. Exp Para-
pharm 2017; 117: 132-40. sitol 2013; 135(2): 314-9.

You might also like