You are on page 1of 11

Article

Journal of
Biomedical Nanotechnology
Copyright © 2015 American Scientific Publishers
All rights reserved Vol. 11, 351–361, 2015
Printed in the United States of America www.aspbs.com/jbn

Solid Lipid Nanoparticles Formulated for


Transdermal Aconitine Administration and
Evaluated In Vitro and In Vivo
Zhang Yong-Tai† , Han Meng-Qing† , Shen Li-Na, Zhao Ji-Hui, and Feng Nian-Ping∗
Department of Pharmaceutical Sciences, School of Pharmacy, Shanghai University of Traditional Chinese Medicine,
Pudong New District, Shanghai 201203, P. R. China

In this study, solid lipid nanoparticles were formulated for transdermal delivery of aconitine to improve its safety and
permeability. Aconitine-loaded solid lipid nanoparticles were formulated as an oil-in-water microemulsion. Drug encapsu-
lation efficiencies for these formulations were higher than 85%, and correlated positively with levels of surfactant and oil
matrix. The size of the solid lipid nanoparticles was increased with an increase of the oil matrix, and reduction of the
surfactant levels. Compared with an ethanol tincture, all the tested solid lipid nanoparticle formulations achieved improved
transdermal fluxes and drug deposition in skin in vitro. Real-time monitoring of drug distribution in rat dermis using in vivo
microdialysis showed that aconitine concentration was markedly higher following application of solid lipid nanoparticles,
compared to tincture, throughout Delivered
the experimental period. ATechnology
by Publishing regional comparison of rat skin found that application of
to: xing liwei
IP: 166.111.120.71
solid lipid nanoparticles to the scapular On:in Tue,
region resulted 20AUC
higher Jan0−t2015
and 16:48:29
Cmax , compared to those achieved with
application to the abdomen or chest (p Copyright: American
< 005). In contrast, Scientific Publishers
the application to the chest resulted in the lowest AUC0−t
and Cmax . Together with findings of a structural study of the skin, these results indicated that the drug accumulated more
readily in thicker skin regions, and to a lesser extent in well-perfused skin, because of drug transfer to capillaries. The
superior transdermal permeability of aconitine-loaded solid lipid nanoparticles contributed to stronger anti-inflammatory
and analgesic effects on mouse in vivo models of pain than the tincture (p < 005). In vitro and in vivo studies indicated
that smaller particle sizes of solid lipid nanoparticles enhanced the transdermal permeability of aconitine, which can
promote drug efficacy, reduce administration time, and improve medication safety.

KEYWORDS: Encapsulation Efficiency, Microdialysis, Permeability, Topical Delivery, Transdermal.

INTRODUCTION conventional formulations, such as topical tinctures and


Aconitum carmichaelii Debeaux, known as Chuanwu in poultices containing aconitine, in the treatment of vari-
China, is a traditional herb with a long history of com- ous types of pain including those induced by arthritis and
mon use in traditional Chinese medicine. It is usually used cancer.5 Although transdermal administration improves the
to treat intractable pain and has been officially listed in clinical safety and effectiveness of aconitine, these advan-
the Pharmacopoeia of the People’s Republic of China.1 2 tages are reduced in conventional formulations that require
The major active ingredient isolated from this herb is frequent dosing due to poor transdermal permeability.6
aconitine, which has strong analgesic effects, and does not Solid lipid nanoparticles (SLN) provide a novel nanocar-
cause addiction.3 However, the therapeutic range of aconi- rier for drug delivery. Drug loaded in SLN can be
tine is narrow and oral administration has a poor safety released in a sustained manner, effectively improving its
profile.4 Previous studies have investigated the efficacy of bioavailability.7 Over the past decade, many reports have
focused on the advantages of SLN formulated for transder-
mal delivery. The SLN can greatly enhance drug transder-

Author to whom correspondence should be addressed. mal permeability, and effectively increase sustained drug
Email: npfeng@hotmail.com

These two authors contributed equally to this work.
release from the epidermis into deep skin tissue, and
Received: 23 August 2013 onwards into the systemic circulation.8 Previous findings
Revised/Accepted: 4 January 2014 showed that if drug was efficiently entrapped in SLN, they

J. Biomed. Nanotechnol. 2015, Vol. 11, No. 2 1550-7033/2015/11/351/011 doi:10.1166/jbn.2015.1902 351


Solid Lipid Nanoparticles Formulated for Transdermal Aconitine Administration and Evaluated In Vitro and In Vivo Yong-Tai et al.

could accumulate in the epidermis and form a drug reser- by the Animal Ethical Committee, Shanghai University of
voir, resulting in superior sustained drug release.9 10 SLN Traditional Chinese Medicine. Animals were kept in an
produced more effective sustained release than nanostruc- agreeable environment with free access to rodent diet and
tured lipid carriers (NLC).11 This enhanced percutaneous water, and were acclimatized for at least 1 week before
drug absorption from SLN may be due to the nanopar- the start of the study.
ticles tendency to form a dense lipid film adhered to
the skin surface. This promotes hydration of the stratum High Performance Liquid Chromatography
corneum, facilitating its penetration by SLN. The perme- (HPLC) Assay
ability of SLN can be influenced by nanoparticle size dis- The concentrations of aconitine in samples were deter-
tribution and crystallinity. SLN with small particle size mined by HPLC using a LC-2010A HT Liquid Chro-
and high crystallinity can form the lipid film more effec- matograph system (Shimadzu Corporation, Kyoto, Japan).
tively, enhancing adhesion on the skin surface and increas- A diamonsil C18 reverse phase column was used
ing drug permeability.12 Lipophilic drugs entrapped within (5 m, 4.6 mm inner diameter × 25 cm; Welch Mate-
SLN can show sustained release after topical administra- rials, Inc., Shanghai, China) with a mobile phase was
tion, producing a continuous supply of drug to the dermis of methanol:water:trichloromethane:triethylamine (70:30:
and markedly enhancing drug transdermal permeability.13 2:0.1, v/v) and a flow rate of 1 mL/min. The column tem-
SLN systems may provide a viable approach to control- perature was constant at 35  C and the detection wave-
ling dermal drug penetration, which is particularly valu- length was 235 nm. The HPLC method was validated for
able for drugs with narrow therapeutic ranges.14 SLN accuracy, precision, limit of detection and limit of quantita-
were successfully used for transdermal administration of tion for aconitine. Samples from in vitro experiments were
podophyllotoxin and triptolide, demonstrating their suit- filtered through a nylon 0.45 m pore disposable syringe
ability for this application.15 16 These features indicated filter (diameter: 13 mm [Welch Materials, Inc., Shanghai,
that SLN represented a promising transdermal aconitine China]) before automatic injection into the HPLC. Sam-
carrier. ples from in vivo microdialysis were directly assayed in a
The present study aimed to improve aconitine safety and timely manner without any handling.
permeability by establishing a novel transdermal delivery
system using SLN as the vehicle,Delivered
prepared using the hot Technology to: xing liwei
by Publishing Preparation of SLNs and Tincture
microemulsion method.17 In vitroIP:transdermal permeation
166.111.120.71 On: Tue,A20 Jan 2015 16:48:29
microemulsion precursor method was employed for the
studies were carried out to investigateCopyright:
the effect American
of SLN Scientific Publishers 17
preparation of SLNs. A mixture was prepared by melting
composition on permeability, compared with a tincture.
oil (Compritol® 888 ATO, the core matrix of the SLNs),
In vivo dermal microdialysis was also used to provide a
surfactant (Cremophor® EL) and co-surfactant (Transco-
more realistic measure of aconitine release from SLN into
tol P) at 80  C. Aconitine was dissolved in this mixture
the dermis in a range of skin regions.
prior to adding water dropwise at 80  C under mag-
netic stirring (magnetic stirrer, IKA Works GmbH & Co.,
MATERIALS AND METHODS Staufen, Germany) at 300 rpm until a uniform microemul-
Materials sion was formed. The microemulsion (SLN suspension)
Diethylene glycol single ethyl ether (Transcotol P) and was refrigerated quickly at − 20  C for 2 h and then stored
®
glyceryl behenate (Compritol 888 ATO) were pur- at 4  C. The amount of aconitine loaded in all the SLN
chased from Gattefossé (Lyon, France). Polyethylene gly- suspensions was 0.25 mg/mL (0.25‰ w/v), as shown in
col (PEG)-35 castor oil (Cremophor® EL) was obtained Table I.
from BASF (Ludwigshafen, Germany). Ethyl oleate was A tincture was prepared with the same aconitine con-
purchased from the Shanghai Yunhong Chemical Prepa- centration as the SLNs (0.25‰ w/v) in 70% ethanol
ration Auxiliary Technology Co., Ltd. (Shanghai, China). water (v/v).
Aconitine (isolated from aconite), with a purity of not
less than 98.0%, was supplied by Ze-lang BioScience Characterization of SLNs
(Nanjing, China). Votalin (diclofenac diethylamine emul- The mean particle size of the prepared SLNs was mea-
gel, drug content 1% [w/w]) was purchased from Novar- sured using dynamic light scattering (DLS). The SLN sus-
tis AG (Basel, Switzerland). All other chemicals were pensions were diluted with 8 times (v/v) with purified
obtained from the Sinopharm Chemical Reagent Co., Ltd. water before DLS measurements. A computerized Zeta-
(Shanghai, China) and were of HPLC or analytical grade. sizer Nano ZS90 (Malvern Instruments Ltd., Malvern, UK)
was employed to detect particle size and zeta potential of
Animals the SLNs. All measurements were performed in triplicate.
Male Sprague-Dawley rats of clean grade, weighing 300 ± The appearance of the SLN formulation was also exam-
20 g with about 2 months of age, and Konmin mice weigh- ined by transmission electron microscopy (TEM, Philips
ing 20 ± 2 g, were used. The animal study was approved Tecnai 12; Philips, Amsterdam, The Netherlands). TEM

352 J. Biomed. Nanotechnol. 11, 351–361, 2015


Yong-Tai et al. Solid Lipid Nanoparticles Formulated for Transdermal Aconitine Administration and Evaluated In Vitro and In Vivo

Table I. Compositions and drug loading of solid lipid nanoparticle formulations.

Formulation Km S-Cos/oil matrix (w/w) S (%, w/v) Cos (%, w/v) Oil matrix (%, w/v) Aconitine (‰ w/v) Water (%, w/v) EE (%)

SLN1 1:1 9:1 1800 1800 400 0.25 60.00 89.90 ± 5.30
SLN2 1:1 8:2 1600 1600 800 0.25 60.00 89.52 ± 3.44
SLN3 1:1 7:3 1400 1400 1200 0.25 60.00 88.79 ± 6.48
SLN4 1:2 9:1 1200 2400 400 0.25 60.00 88.64 ± 6.00
SLN5 1:2 8:2 1067 2133 800 0.25 60.00 87.92 ± 5.70
SLN6 1:3 9:1 900 2700 400 0.25 60.00 87.87 ± 4.02
SLN7 1:3 8:2 800 2400 800 0.25 60.00 87.81 ± 7.36

Notes: Compared with each other for EE, p > 005. (For EE detection, n = 3); Abbreviations: SLN, solid lipid nanoparticles; Km, ratio of surfactant to cosurfactant
(w/w); S, surfactant (Cremophor® EL); Cos, cosurfactant (Transcotol P); S-Cos, total content of surfactant and cosurfactant; oil matrix (Compritol® 888 ATO);
EE, encapsulation efficiency.

samples were prepared for negative staining by placing aqueous solution magnetically stirred (300 rpm) and main-
copper nets carrying formvar-supporting film (200 mesh, tained at 32 ± 0.5  C to provide sink conditions. Each
Zhong Jing Ke Yi Technology Inc., Beijing, China) onto a experiment was performed in triplicate. At predetermined
stencil plate and dropping SLNs gently onto the film. The time points, 1 mL samples were removed from each recep-
film was allowed to dry for about 20 min before a drop tor compartment and replaced with an equal volume of
of 2% uranyl acetate was added and allowed to dry for receptor fluid equilibrated to 32 ± 0.5  C. The concentra-
10 min prior to imaging the film using TEM. tion of aconitine in the obtained samples was determined
using HPLC, as described above. The permeation profile
Entrapment Efficacy of SLNs for aconitine was obtained by plotting the mean cumulative
Centrifugal ultrafiltration was used to investigate entrap- permeation per cm2 of rat skin against time. Linear regres-
ment efficacy (EE). The aconitine-loaded SLN suspension sion analysis was performed to determine the transdermal
was sucked up precisely with a pipettor (Eppendorf AG, flux of aconitine from SLN preparations, as compared to
Hamburg, Germany), transferred into a centrifugal ultra- tincture.
filtration tube with a molecular-weight cutoff
Delivered (MWCO) Technology to: xing liwei
by Publishing
of 10 kDa (Pall Corporation, New York, USA), and cen-
IP: 166.111.120.71 Determination
On: Tue, of Skin Aconitine Deposition
20 Jan 2015 16:48:29
trifuged at 12 000 rpm (× 11913 g)Copyright:
for 20 minAmerican
in a Scientific
At the endPublishers
of the in vitro skin permeation studies (24 h),
Sigma 2-16K centrifuge (Sigma Laborzentrifugen GmbH, the skins were removed from the diffusion cell, and
Osterode am Harz, Germany). It was demonstrated no the residual aconitine preparations on their surfaces were
free aconitine crystals dispersed in the prepared SLN sus- wiped off carefully with cotton wool balls, then washed
pensions by using scanning calorimetry and polarization 3 times with 3 mL fresh receptor fluid per time, which
microscope in our previous studies. The concentration of assured that the adherent drug on skin surface was com-
aconitine in the ultrafiltrate was determined directly by pletely removed. The skins were cut into small pieces and
HPLC, as described above. The EE was calculated using homogenized in manual glass homogenizers with 1 mL
the following formula (Eq. (1)): fresh receptor fluid before centrifuging at 10,000 rpm
EE% = Wt − Wf /Wt × 100 (1) for 10 min. The supernatants were filtered through a
0.45-m filter membrane, and assayed for aconitine using
where Wt was the amount of aconitine loaded in the SLN HPLC. The skin aconitine deposition was shown as: skin
suspension and Wf was the amount of aconitine in the deposition = Wa /Sd , where Wa means total amount of
ultrafiltrate.17 aconitine deposited in per rat skin, and Sd is the effective
diffusion area (1.77 cm2 ).
In Vitro Skin Permeation Studies
A rat was humanely sacrificed, its abdominal fur was Stability Study
shaved off with a razor, and the skin was excised care- SLN stability was determined by investigating size distri-
fully to keep undamaged, and washed with normal saline. bution and EE after being stored for 0 day and 30 days
The permeation of aconitine delivered by SLN or alcohol at 4  C. The change rates of the particle size or EE (size
tincture was compared in vitro using a Franz diffusion cell change rate, SCR; EE change rate, ECR) were calculated
(Fulansi Electronic Science and Trade Co., Ltd., Tianjin, using Eq. (2):
China) fitted with excised rat skin. Each donor compart-
S − S0  E − E0 
ment had a diffusion area of 1.77 cm2 and 2 mL of SLN SCR = 30 × 100 ECR = 30 × 100 (2)
suspension or tincture was added to each compartment S 0 E0
under non-occlusive conditions. Each receptor compart- where S0 or E0 means particle size or EE determined at
ment had a volume of 16 mL and was filled with freshly the day SLNs were prepared (0 days), and S30 or E30 were
prepared 20% (v/v) polyethylene glycol 400 (PEG 400) the corresponding values after storage for 30 days.

J. Biomed. Nanotechnol. 11, 351–361, 2015 353


Solid Lipid Nanoparticles Formulated for Transdermal Aconitine Administration and Evaluated In Vitro and In Vivo Yong-Tai et al.

Microdialysis System STD solutions were used as perfusates at a flow rate of


The microdialysis system consisted of a WZ-50C6 micro 3.33 L/min. After equilibration for 30 min, a dialysate
infusion pump (Smiths Medical, Norwell, MA) with a sample was collected into a vial for another 30 min. The
20 mL plastic syringe and a linear microdialysis probe. diffusive loss of aconitine from perfusates was determined,
Spectra/Por® microdialysis hollow fibers (Spectrum Labo- and RR (also known as delivery rate) was calculated using
ratories, Inc., Chicago, USA) were prepared using regen- Eq. (4), where Cp was the drug concentration in the per-
erated cellulose (200 m inner diameter, 280 m outer fusate.
diameter, 13 kDa MWCO). The fibers were glued to quartz RR = Cp − Cd /Cp × 100 (4)
capillary tubing (Welch Materials, Inc., Shanghai, China)
In vivo RR was determined using the retrodialysis-by-
with cyanoacrylate adhesive (Mxbon® Super Glue; Cartell
drug method, which relies on the assumption that net drug
Chemical Co. Ltd., Chia-Yi Hsien, Taiwan) in order to cre-
transport from the perfusate into the surrounding tissues
ate linear microdialysis probes, which were connected to
through the microdialysis membrane is equal to the net
the microinjection pump using polyethylene tubing. In all
drug transport from the tissues into the perfusate. A rat
experiments, the length of the membrane was 20 mm,
was anesthetized with urethane aqueous solution (1.3 g/kg,
and the perfusate flow rate was 3.33 L/min. Cannulae
intraperitoneal), and anesthesia was maintained throughout
were used as insertion guides, and vials were used to col-
the experiment. The ambient temperature was kept con-
lect the dialysate samples. Standard solutions of aconitine
stant at 25  C. A linear microdialysis probe was inserted
(STD) were prepared by dissolving pure aconitine in 20%
into the dermis of the rat’s abdominal skin. After perfusing
PEG 400.
with 20% PEG 400 for 1 h, STD aconitine was used as
the perfusate. HPLC assays were carried out to determine
Recovery Validation In Vitro and the loss of aconitine from the STD solutions, and RR was
Correction In Vivo calculated using Eq. (4).
In vitro recovery was estimated prior to microdialysis
studies to ensure that the probes used would provide In Vivo Microdialysis Studies
reproducible and efficient sampling. A linear microdialy-
A rat was anesthetized with urethane aqueous solution
sis probe was placed in a 50 mL beaker containing dif-
prior to administration of aconitine, the fur at the admin-
ferent concentrations of aconitineDelivered by (Table
in sequence Publishing
II), Technology to: xing liwei
and the dialysis membrane portion IP:of
166.111.120.71 On: Tue,istration
the probe was com- 20 Jan sites
2015 on each rat was manually shaved. The
16:48:29
Copyright: American administration
Scientific sites (abdomen, chest, and scapularis) were
Publishers
pletely immersed in the solution at room temperature.
located with the bellybutton, the intermediate point of two
The probe was perfused with 20% PEG 400 at a flow
front paws and the shoulder blade as the centers, respec-
rate of 3.33 L/min. After an equilibration period of
tively. A microdialysis probe was implanted in the der-
30 min, the dialysate was collected into a small vial for
mis, with the active dialysis window placed immediately
30 min and analyzed using HPLC to determine the aconi-
below the site of topical drug administration. The probe
tine concentration. Relative recovery (RR) was calculated
was continuously perfused with 20% PEG 400. The skin
as the slope of the linear regression of drug concentration
was allowed to equilibrate for 1 h before a blank sam-
in the dialysate (Cd ) as a function of drug concentration
ple was taken from the microdialysis probe. At 1.5 h after
in the medium (Cm ; Eq. (3)).
the start of perfusion, the drug was applied. A flat cylin-
RR = Cd /Cm × 100 (3) drical plastic cover about 1 cm high, 1.5 cm in diame-
ter, and with an edge width of 2 mm was glued above
For the retrodialysis studies, the probe was perfused the administration site using cyanoacrylate adhesive. 1 mL
with aconitine STD solutions (Table II). A linear probe of SLN suspension or tincture was applied to the skin
was placed in a 50 mL beaker with the membrane por- within the cover. During the experiment, the administra-
tion completely immersed in 20% PEG 400 at 25  C. tion site and the probe were kept level. Dialysate samples

Table II. Recovery of aconitine for microdialysis probe. (In vitro: n = 3; In vivo: n = 5).

Cm (Cp ) (g/mL) a
Cd (g/mL) a
RR (%) b
Cp − Cd (g/mL) b
RR (%) c
Cp − Cd (g/mL) c
RR (%)

1.00 0.34 ± 0.01 34.28 ± 4.06 0.35 ± 0.04 35.04 ± 3.83 0.33 ± 0.03 32.67 ± 2.65
5.00 1.68 ± 0.25 33.50 ± 2.37 1.72 ± 0.35 34.37 ± 4.03 1.57 ± 0.21 31.42 ± 3.18
10.00 3.23 ± 0.67 32.31 ± 3.50 3.46 ± 0.65 34.61 ± 3.12 3.10 ± 0.45 31.03 ± 3.28
25.00 8.32 ± 0.88 33.27 ± 3.33 8.43 ± 0.74 33.72 ± 2.94 8.14 ± 0.78 32.56 ± 2.02
50.00 17.04 ± 2.45 34.08 ± 4.58 17.33 ± 3.19 34.66 ± 3.00 15.79 ± 1.99 31.58 ± 1.73

Note. (a) Recovery of different aconitine standard solutions in vitro determined using Eq. (3) in text; (b) Recovery of different aconitine standard solutions
with retrodialysis-by-drug method in vitro determined by using Eq. (4) in text; (c) Recovery of different aconitine standard solutions with retrodialysis-by-drug
method in vivo determined by using Eq. (4) in text; a RR compared with b RR, p > 005.

354 J. Biomed. Nanotechnol. 11, 351–361, 2015


Yong-Tai et al. Solid Lipid Nanoparticles Formulated for Transdermal Aconitine Administration and Evaluated In Vitro and In Vivo

were collected into small vials, which were replaced every or high aconitine-loaded aconitine SLN suspension vol-
30 min for a maximum of 10 h. umes application to per mouse (SLN-LDG, with 10 L;
SLN-MDG, with 15 L; and SLN-HDG, with 20 L). The
Detection of Skin Blood Flow test substances were applied evenly on the dorsal region.
A rat was anesthetized and fur was shaved from its After 2 h topical administration, the inflammation and
abdomen, chest and scapularis regions, as described for pain models were established and relevant behaviors were
the in vivo microdialysis study. After being anesthetized immediately recorded. For AAIPM, body twisting within
for 1 h, a surface probe (XP200, low-profile service probe) 15 min was recorded. The latency before beginning to lick
for laser doppler flowmetry (OxyLab LDF™ and OxyFlo™ , the affected paw and the number of licks within 5 min
Oxford Optronix, Oxford, UK) was closely pasted to the (I phase) as well as repeat licking within 1 h (II phase)
skin surface with a special adhesive ring plate. The doppler were recorded for the FIPM. The weight gain of the left
bandwidth was 780 ±10 nm, the blood flow reflection time (control) auricle as compared to the right, after removal
was 0.2 s, and recordings were made using a unipolar from the mice and shaving, was used to assess the DIPM
lead and multi-channel physiology recorder with Chart 5.0 and the time required for mice to lift their tails out of the
software for Windows (Powerlab 8/30, ADInstruments Pty water bath was recorded for the HWPM, as a measure of
Ltd., New South Wales, Australia). Data were expressed pain threshold.
as the mean value for 1 h.
Statistical Analysis
Regional Skin Structure The results are expressed as mean ± standard deviation.
A rat was humanely sacrificed before excising skin sam- Statistical data were analyzed by one-way analysis of vari-
ples of approximately 1 × 1 cm from the abdomen, chest ance (ANOVA), and comparisons were made with least-
and scapularis regions and fixing in 4% paraformaldehyde significant difference (LSD) using SPSS software (v 13.0;
solution (w/v). After 24 h, the skins were embedded in SPSS, Inc., Chicago, USA). P -values less than 5% were
paraffin and sliced transversely. The sections were dehy- considered to be significant. The cutaneous pharmacoki-
drated stepwise with alcohol, and stained with hematoxylin netic parameters of aconitine were calculated using Win-
and eosin (HE). The sections were observed using an opti- NonLin (v 5.2; Pharsight Corporation, Sunnyvale, USA)
Delivered by Publishing
cal microscope (IX70, inverted fluorescence microscope, and noncompartmental
Technology to: xing liweianalysis.
IP:
Olympus Corporation, Hatagaya, Japan).166.111.120.71 On: Tue, 20 Jan 2015 16:48:29
Copyright: American Scientific Publishers
RESULTS AND DISCUSSION
Anti-Inflammatory and Analgesic Effects SLN Preparation and Characterization
Four frequently used mouse models of inflammation To investigate the appropriate concentration range of com-
and pain were employed to evaluate the pharmacolog- ponents for microemulsion formation, the weight ratios
ical effects of aconitine-loaded SLN, compared with of surfactant and co-surfactant (Km) were set at 1:1, 1:2,
the tincture. These were the acetic acid, formaldehyde 1:3, and the weight ratios of oil in a mixture of surfactant
or dimethylbenzene induced inflammatory pain models and co-surfactant were set at 1:9, 2:8, 3:7, 4:6, 5:5, 6:4,
(respectively abbreviated as AAIPM, FIPM, and DIPM), 7:3, 8:2, and 9:1. After equilibration, the mixtures were
and the hot water-induced pain model (HWPM). Ani- visually assessed to determine whether they had formed
mals were administered 20 L of 0.6% acetic acid (v/v) microemulsions or not. Seven formulations that readily
by intraperitoneal injection (AAIPM); 2.5% formaldehyde formed uniform microemulsions were then selected and
(w/v) aqueous solution by subcutaneous injection at the used to form SLNs (SLN1 to SLN7). The components of
back of the right rear paws (FIPM); or pure dimethyl- these SLNs are listed in Table I.
benzene, daubed on the inside and outside surfaces of Previous reports indicated that use of Compritol® 888
the right auricles (DIPM). For the HWPM, half of the ATO as the SLN lipid core could be conducive to form-
mouse’s tail was immersed into hot water at 50  C. ing stable dispersions with smaller particle sizes, and this
Except for the DIPM, 1 × 1 cm of fur was shaved from was borne out by the present study, where particle size
the dorsal region. For every pharmacological experiment, distributions ranged from 34.78 ± 3.75 nm to 131.90 ±
70 mice (50% male, 50% female) were divided randomly 24.65 nm.18–20 The mean particle sizes were increased
into 7 groups as follows: normal saline group (NSG, by reducing the Km in the presence of a constant ratio
with 20 L normal saline application to per mouse); of S-Cos (surfactant [Cremophor® EL] and cosurfactant
positive medicine control group (PCG, with 20 mg of [Transcotol P]) versus oil matrix (Compritol® 888 ATO).
Votalin [Diclofenac Diethylamine Emulgel] application to This indicated that SLN size increased as surfactant con-
per mouse); blank based group (BBG, with 20 L SLN centration decreased. Previous studies have reported a sim-
suspension without aconitine application to per mouse); ilar relationship between mean particle size and surfactant
aconitine-loaded tincture group (TTG, with 20 L appli- concentration.21 22 At a constant Km value, the particle
cation to per mouse); and 3 SLN groups with low, medium sizes decreased with an increasing level of oil matrix

J. Biomed. Nanotechnol. 11, 351–361, 2015 355


Solid Lipid Nanoparticles Formulated for Transdermal Aconitine Administration and Evaluated In Vitro and In Vivo Yong-Tai et al.

content of surfactant (Table I). The augmented entrapment


of aconitine with increased surfactant concentration sug-
gested that aconitine might be embedded in the surfac-
tant layer, rather than incorporated into the lipid matrix.22
As a highly lipophilic drug, aconitine can be efficiently
solubilized in the oil phase (composed of oil, surfactant
and cosurfactant) of the microemulsion system. In addi-
tion, the contents of aconitine were only 0.25‰ (w/v) in
every SLN formulations, so that all SLNs achieved high
EE, with no significant differences between the 7 formu-
lations (p > 005).
Figure 1. Mean particle sizes and zeta potentials of the
aconitine-loaded solid lipid nanoparticle formulations (S-Cos:
In Vitro Permeation Studies
oil matrix [w/w] = 1:9 [], 2:8 [], 3:7 [•]). Abbreviations: SLN,An enhanced transdermal release of aconitine was
solid lipid nanoparticles; Km, ratio of surfactant to cosurfac- achieved by using SLNs as the vehicle for topical adminis-
tant (w/w); S, surfactant (Cremophor® EL); Cos, cosurfactant tration, compared with the tincture. Transdermal fluxes of
(Transcotol P); S-Cos, total content of surfactant and cosur-
aconitine from the SLN formulations ranged from 1.15 ±
factant; oil matrix (Compritol® 888 ATO). (n = 3).
0.20 g/cm2 /h to 11.80 ± 0.76 g/cm2 /h, and were all
(Fig. 1). A higher level of oil matrix may decrease the vis- higher than the tincture (Figs. 3 and 4). Although the high
cosity of the melted droplets, increasing the likelihood that ethanol concentration in tinctures can promote transdermal
23
they will disperse and result in smaller particle sizes. The delivery, the permeability provided is limited and erratic,
zeta potentials of the SLNs were distributed from − 3.42 ± due to evaporation.
0.45 to − 26.97 ±2.31 mv. Zeta potentials were lower with At a constant Km value, aconitine transdermal flux
increased use of oil matrix at a constant Km value. They increased with an increased level of oil matrix (Fig. 4),
were also lower at reduced Km value with a constant ratio while the SLN particle sizes were reduced with increased
of S-Cos to oil matrix, indicating that the level of surfac- oil matrix. Reduced surfactant level led to increased trans-
Delivered by Publishing
tant could affect the zeta potential. These zeta potential dermal
Technology flux,
to:and decreased
xing liwei particle size, at a constant ratio
IP: 166.111.120.71
results suggested that the SLNs had a negatively charged On: Tue,
of 20 Jan
S-Cos 2015
to oil 16:48:29
matrix (w/w). A small particle size can per-
Copyright: American Scientific
mit close Publishers
contact of SLNs with the stratum corneum and
surface, which may be provided by the surfactant molecu-
®
lar chain of Cremophor EL surrounding the edge of the further improve drug permeation into skin. After applica-
nanoparticle. 22–24
Thereby, larger amounts of Cremophor ® tion onto the skin, the small SLNs can form a close lipid
EL led to increased negative charge on the SLN surface, film on the stratum corneum, and drug molecules can be
resulting in higher particle zeta potential. expulsed from SLNs, which can form a higher concen-
SLN7 was found to be spherical in shape when imaged tration gradient towards the skin, further enhanced more
25
using TEM (Fig. 2). The nanoparticles appeared sepa- effectively the transdermal delivery of drug, compared
rately from each other, and showed no change in shape
before and after 8-fold dilution with purified water.

SLN Entrapment Efficacies


The EE of SLN1 to SLN7 varied between 87.81 ± 7.36%
and 89.90 ± 5.30%, and was raised slightly with increasing

(a) (b)

Figure 3. In vitro rat skin permeation profiles of aconitine


from the aconitine-loaded solid lipid nanoparticle formula-
tions and a tincture (S-Cos:Oil matrix [w/w] = 1:9 [], 2:8 [],
3:7 [•]). Abbreviations: SLN, solid lipid nanoparticles; TT, tinc-
ture; Km, ratio of surfactant to cosurfactant (w/w); S, sur-
Figure 2. Transmission electron microscopy of aconitine- factant (Cremophor® EL); Cos, cosurfactant (Transcotol P);
loaded solid lipid nanoparticles (SLN7). ((a) 10,000× magnifi- S-Cos, total content of surfactant and cosurfactant; oil matrix
cation; (b) 100,000× magnification). (Compritol® 888 ATO). (n = 3).

356 J. Biomed. Nanotechnol. 11, 351–361, 2015


Yong-Tai et al. Solid Lipid Nanoparticles Formulated for Transdermal Aconitine Administration and Evaluated In Vitro and In Vivo

Figure 5. Perfusate concentration of aconitine (Cp ) as deter-


Figure 4. In vitro transdermal fluxes and skin deposition of mined by the retrodialysis-by-drug method in vitro and in vivo
aconitine from solid lipid nanoparticle formulations or tinc- (Cp : aconitine concentration in perfusates, Cd : aconitine con-
ture through excised rat skin (S-Cos:oil matrix [w/w] = 1:9 [], centration in dialysate sample).
2:8 [], 3:7 [•]). Abbreviations: SLN, solid lipid nanoparticles;
TT, tincture; Km, ratio of surfactant to cosurfactant (w/w); constant Km value and a constant ratio of S-Cos to oil
S, surfactant (Cremophor® EL); Cos, cosurfactant (Transco-
tol P); S-Cos, total content of surfactant and cosurfactant; oil
matrix, the transdermal flux and skin deposition of aconi-
matrix (Compritol® 888 ATO). Compared with the tincture, † p < tine from SLNs were increased as the particle zeta poten-
005 for skin deposition, # p < 005 for transdermal flux. (n = 3).
tial decreased. The repulsion between negatively charged
cell membranes and nanoparticles was weakened when the
zeta potential value was reduced, promoting SLN perme-
with the SLNs possess larger particle sizes.26–28 In addi- ability and drug deposition in the epidermis.37 38
tion, they may enhance transdermal drug delivery via hair Results for the in vitro permeation studies showed that
follicles.29 30 transdermal flux followed the same trend as skin depo-
SLNs have been reported to increase epidermal drug sition of aconitine from the SLN formulations. Previous
deposition.8 31 In this study, the skin deposition of aconi- studies reported that SLN strongly targeted the epidermis,
tine from SLNs ranged from 18.19 ± 234 g/cm2 (SLN1) leading to ato:high drug concentration gradient between the
Delivered by Publishing Technology xing liwei
to 66.62 ± 5.11 g/cm2 (SLN7, IP: Fig.166.111.120.71
4). Aconitine depo-
On: Tue, 20 Jan 2015
epidermis 16:48:29
and deep skin layers, promoting drug perme-
sition by all the SLNs were 1.5- to Copyright:
5.6-fold thatAmerican
of the Scientific Publishers
ation through skin and contributing to a high transdermal
tincture. Drug was delivered by tincture with poor per- flux. 8 13
meability into the skin directly, and immediately trans- The formulation SLN7 showed the highest transdermal
ferred to the microcirculation under the epidermis. This flux and skin deposition of aconitine and was selected for
may result in the lowest drug deposition from the tinc- further studies.
ture. In contrast, lipid carriers such as SLN can attach to
the skin surface and allow lipid exchange with the out-
Stability of SLN7
ermost layers of the stratum corneum, thus providing a
After being stored for 30 days at 4  C, SLN7 particle
higher drug concentration in the skin.32 In addition, the
size distribution was increased and EE was reduced, but
lipid matrix of the SLN oil core, formed by solid crystal-
neither change was statistically significant (p > 005, data
lization, may transition to a more ordered structure super-
not shown). The SCR and ECR were 6.81 ± 0.63% and
saturated with drug after being applied to the skin surface,
3.95 ± 0.19%, respectively. Using Compritol® 888 ATO as
leading to drug expulsion and accumulation as a drug
reservoir in the upper skin layers and prolonging skin res-
idence time.33 34 These factors may account for the higher
skin deposition of aconitine from SLN, as compared to
tincture.
The amounts of aconitine deposited in skin were
improved with an increased level of oil matrix at a constant
Km value, while they were decreased with increased sur-
factant at a constant ratio of S-Cos to oil matrix (Fig. 4).
Increased oil matrix and decreased surfactant produced
smaller particles, leading to stronger skin hydration and
transdermal permeability of SLNs, which promoted lipid
exchange with epidermal lipids and increased drug skin
Figure 6. Ten-hour time course of aconitine concentration
deposition.35 36 after application of drug-containing solid lipid nanoparticles
The present studies discovered that SLN surface charge (SLN7) or tincture to the abdominal skin of Sprague-Dawley
also affected skin permeability (Figs. 2 and 4). At a rats in vivo. (n = 5).

J. Biomed. Nanotechnol. 11, 351–361, 2015 357


Solid Lipid Nanoparticles Formulated for Transdermal Aconitine Administration and Evaluated In Vitro and In Vivo Yong-Tai et al.

Table III. Pharmacokinetic parameters of aconitine loaded the perfusate (in vitro: Cp − Cd  = 0345Cp − 0003, r 2 =
in SLN and the tincture via microdialysis after applied on 0999; in vivo: Cp − Cd  = 0317 Cp + 0015, r 2 = 0999;
abdomen of rat skin in vivo. (n = 5). Fig. 5). This correlation implied that retrodialysis recovery
Parameter Unit SLN Tincture was independent of perfusate concentration both in vitro
and in vivo, supporting the feasibility of detecting the true
Tmax min 300.00 ± 00.00 60.00 ± 00.00†
Cmax g/mL 1.21 ± 0.11 0.34 ± 0.14†
unbound extracellular level of aconitine using the micro-
AUC0−t min/g/mL 346.31 ± 25.21 152.94 ± 28.95 † dialysis technique.
MRT0−t min 314.92 ± 16.35 313.71 ± 26.23 The aconitine concentrations in rat abdominal skin
after topical application of SLN or tincture are shown
Note: Compared with the pharmacokinetic parameters from the SLN, † p <
005; Abbreviations: SLN, solid lipid nanoparticle; Tmax , time to peak concen- in Figure 6 and Table III. Skin aconitine levels were
tration; Cmax , peak concentration; AUC, area under the concentration-time higher with SLN than with tincture throughout the experi-
curve; MRT, mean residence time.
ments. The peak concentration (Cmax ) and AUC0−t for SLN
were 3.56-fold and 2.26-fold higher than the correspond-
ing measures using tincture, respectively. These findings
demonstrated that use of SLN increased aconitine levels in
the dermis, indicating improved transdermal permeability
compared to tincture.
The drug concentration profile in skin exposed to SLN
rose initially and then decreased. The Cmax occurred
300 min after SLN application. This reflected accumula-
tion of SLN-borne drug molecules in the epidermis prior
to their release and transfer to capillaries. For the ini-
tial 300 min, accumulation of SLN in the lipid epider-
mis meant that more aconitine was released than was
Figure 7. Ten-hour time course of aconitine concentration transferred to the skin microcirculation, and the Cmax was
after application of drug-containing solid lipid nanoparticles reached. Subsequent epidermal saturation with SLN led
(SLN7) to the abdomen, scapularis, and chest regions of
Delivered by Publishing Technology to the decrease
to: xingin liwei
drug concentration as it was trans-
Sprague-Dawley rats in vivo. (n = 5).
IP: 166.111.120.71 On: Tue,ferred 20 Jan 2015
to the 16:48:29
microcirculation faster than it was delivered.
Copyright: American Scientific Publishers
In contrast, the drug concentration after tincture applica-
the SLN lipid core promoted particle size stability, and the tion was relatively moderate at Tmax (time to peak con-
high lipophilicity of aconitine may have contributed to the centration, 60 min). The aconitine in tincture displayed
stability of its encapsulation.17 18 diffusion behavior during percutaneous absorption, con-
forming to Fick’s first law and producing a more simple
In Vivo Dermal Microdialysis Studies skin drug concentration profile over time.
Aconitine absorption after topical application of SLN7 or The regional comparisons of skin aconitine concentra-
tincture was investigated in vivo using skin microdialy- tions over time are presented in Figure 7 and Table IV.
sis. There was no significant difference between RR from The concentration of aconitine from SLN applied to the
dialysis and from retrodialysis (drug delivery) in vitro abdomen and chest initially increased more rapidly than
(p > 005), satisfying the prerequisite for retrodialysis use in the scapular region. However, the scapular drug con-
in the in vivo RR studies.39 The in vitro and in vivo centration continued to rise and achieved the highest Cmax
probe recoveries were consistently above 30% (Table II). of the compared skin regions within the test period at
In the in vitro and in vivo recovery validation studies 510 min, whilst Cmax and AUC0−t were lowest in the chest.
using STD as perfusates, drug concentration in the per- These regional permeability differences may relate to skin
fusate showed a linear correlation with drug loss from structure.40 41 The current study found that rat epidermal

Table IV. Pharmacokinetic parameters of aconitine loaded in SLN via microdialysis after applied on different regions of rat skin
in vivo. (n = 5).

Parameter unit Abdomen Chest Scapular region

Tmax min 300.00 ± 00.00 420.00 ± 00.00† 510.00 ± 00.00†∗


Cmax g/mL 1.21 ± 0.11 0.73 ± 0.06† 1.45 ± 0.32†∗
AUC0−t min/g/mL 346.31 ± 25.21 291.91 ± 15.63 397.64 ± 25.38∗
MRT0−t min 314.92 ± 16.35 337.45 ± 28.70 405.99 ± 18.59†∗

Note: Compared with the pharmacokinetic parameters from the abdomen, † p < 005; compared with the pharmacokinetic parameters from the chest, ∗ p < 005;
Abbreviations: SLN, solid lipid nanoparticle; Tmax , time to peak concentration; Cmax , peak concentration; AUC, area under the concentration-time curve; MRT,
mean residence time.

358 J. Biomed. Nanotechnol. 11, 351–361, 2015


Yong-Tai et al. Solid Lipid Nanoparticles Formulated for Transdermal Aconitine Administration and Evaluated In Vitro and In Vivo

(a)

Figure 9. In vivo skin blood flow of various rat skin regions


over 1 h. (n = 5).

(b)

Figure 10. Writhing times of various mice groups over 15 min


after being induced by 0.6% acetic acid aqueous solution (v/v)
intraperitoneally. Abbreviations: NSG, normal saline group
(20 L per mouse); PCG, positive medicine control group
Delivered by Publishing Technology to: xing liwei
(20 mg of Votalin [Diclofenac Diethylamine Emulgel] per
IP: 166.111.120.71 On: Tue, 20 Jan 2015 16:48:29
mouse); BBG, blank based group (20 L SLN suspension
Copyright: American Scientific Publishers
without aconitine per mouse); TTG, aconitine-loaded tincture
group (aconitine content 0.25‰ [w/v], 20 L per mouse); SLN-
LDG, aconitine-loaded solid lipid nanoparticles (SLN7, aconi-
(c) tine content 0.25‰ [w/v]) groups administered with low dosage
(10 L per mouse); SLN-MDG, middle dosage (15 L per
mouse); SLN-HDG, high dosage (20 L per mouse). Compared
with NSG, † p < 005; compared with TTG, ∗ p < 005. (n = 10).

Figure 11. Pain thresholds of mice groups exposed to hot


Figure 8. Microscopy images of rat skin sections from vari- water-induced pain (HWPM). Abbreviations: NSG, normal
ous regions using paraffin microtomy and hematoxylin eosin saline group (20 L per mouse); PCG, positive medicine con-
(HE) staining. Arrows indicate the thickness of the epidermis trol group (20 mg of Votalin [Diclofenac Diethylamine Emulgel]
((a) abdomen; (b) chest; (c) scapular region; magnification per mouse); BBG, blank based group (20 L SLN suspension
×400). without aconitine per mouse); TTG, aconitine-loaded tincture
group (aconitine content 0.25‰ [w/v], 20 L per mouse); SLN-
LDG, aconitine-loaded solid lipid nanoparticles (SLN7, aconi-
thickness was about 70 m on the abdomen, 40 m on
tine content 0.25‰ [w/v]) groups administered with low dosage
the chest and 110 m on the scapular region (Fig. 8). Skin (10 L per mouse); SLN-MDG, middle dosage (15 L per
blood flow was highest in the chest region, and lowest in mouse); SLN-HDG, high dosage (20 L per mouse). Compared
the scapularis (Fig. 9). These regional data are consistent with NSG, † p < 005; compared with TTG, ∗ p < 005. (n = 10).

J. Biomed. Nanotechnol. 11, 351–361, 2015 359


Solid Lipid Nanoparticles Formulated for Transdermal Aconitine Administration and Evaluated In Vitro and In Vivo Yong-Tai et al.

In Vivo Anti-Inflammatory and Analgesic Effects


In the present studies, the aconitine-loaded SLNs (SLN7)
showed the satisfactory pharmaceutical effects on the
inflammatory and physical models of pain (Figs. 10–13).42
The delivery vehicle (BBG) had no significant influence
(p > 005) on the results (Figs. 10–13). No significant
dose-dependent effects (p > 005) were present in the
experimental groups exposed to SLN. However, aconitine
administered using SLN showed more efficacy in the four
pain models tested than the same amount (0.25‰ w/v)
administered in the tincture (p < 005). This probably
Figure 12. Latent period (latent phase) and times of lick- reflected the improved transdermal permeability of aconi-
ing paws (I phase and II phase) of mice groups exposed to tine delivered by SLN.
formaldehyde-induced pain (FIPM). Abbreviations: NSG, nor-
mal saline group (20 L per mouse); PCG, positive medicine
control group (20 mg of Votalin [Diclofenac Diethylamine CONCLUSION
Emulgel] per mouse); BBG, blank based group (20 L SLN
The in vitro and in vivo study data showed that SLN pro-
suspension without aconitine per mouse); TTG, aconitine-
loaded tincture group (aconitine content 0.25‰ [w/v], 20 L vided effective topical delivery of aconitine. This novel
per mouse); SLN-LDG, aconitine-loaded solid lipid nanopar- aconitine-loaded nanocarrier produced superior transder-
ticles (SLN7, aconitine content 0.25‰ [w/v]) groups adminis- mal permeability and deposition in epidermis, as demon-
tered with low dosage (10 L per mouse); SLN-MDG, middle strated by the in vitro skin permeation and in vivo
dosage (15 L per mouse); SLN-HDG, high dosage (20 L per
microdialysis studies. SLN-delivered aconitine produced
mouse). Compared with NSG, † p < 005; compared with TTG,

p < 005. (n = 10). improved anti-inflammatory and analgesic effects, com-
pared to aconitine tincture.
with the idea that SLN-controlled release of aconitine lev- Acknowledgments: This work was financially sup-
els depend on the balance between drug accumulation in ported by the National Natural Science Fund of
Delivered by Publishing
the epidermis and its removal by microcirculation, indicat- Technology to: xing liwei
IP: 166.111.120.71 On: China
Tue, 20 (81303234),
Jan 2015 Youth Scientific Research Fund
16:48:29
ing that higher sustained levels of drug release in dermis (20134q098) from Shanghai Health Bureau, and Students
Copyright: American Scientific Publishers
could be achieved in regions with thicker skin and lower Research Fund (2012) from Shanghai Municipal Education
blood flow. This can be observed clearly after 390 min in Committee.
Figure 7.
Declaration of Interest
The authors report no declarations of interest.

REFERENCES
1. Chinese Pharmacopoeia Committee (ed.), Pharmacopoeia of the Peo-
ple’s Republic of China, Chinese Chemical Industry Press, Beijing
(2010), Vol. I.
2. J. Singhuber, M. Zhu, S. Prinz, and B. Kopp, Aconitum in tradi-
tional Chinese medicine: A valuable drug or an unpredictable risk?
J. Ethnopharmacol. 126, 18 (2009).
3. A. Ameri, The effects of Aconitum alkaloids on the central nervous
system. ProgNeurobiol. 56, 211 (1998).
4. T. Y. Chan, B. Tomlinson, L. K. Tse, J. C. Chan, W. W. Chan,
Figure 13. Weight increment of right auricles of various mice and J. A. Critchley, Aconitine poisoning due to Chinese herbal
groups with dimethylbenzene-induced pain (DIPM). The left medicines: A review. Vet Hum Toxicol. 36, 452 (1994).
ears acted as controls. Abbreviations: NSG, normal saline 5. L. Kang, S. W. Yue, X. L. Wang, and M. Fang, The mechanism
group (20 L per mouse); PCG, positive medicine control underlying the analgesic effect of the iontophoresis of aconitine on
group (20 mg of Votalin [Diclofenac Diethylamine Emulgel] the adjuvant arthritis in rats. Chin. J. Phys. Med. Rehabil. 27, 733
per mouse); BBG, blank based group (20 L SLN suspen- (2005).
sion without aconitine per mouse); TTG, aconitine-loaded tinc- 6. T. Y. Chan and J. A. Critchley, Usage and adverse effects of Chinese
ture group (aconitine content 0.25‰ [w/v], 20 L per mouse); herbal medicines. Hum. Exp. Toxicol. 15, 5 (1996).
SLN-LDG, aconitine-loaded solid lipid nanoparticles (SLN7, 7. A. Jain, S. K. Singh, Y. Singh, and S. Singh, Development of lipid
aconitine content 0.25‰ [w/v]) groups administered with low nanoparticles of diacerein, an antiosteoarthritic drug for enhance-
dosage (10 L per mouse); SLN-MDG, middle dosage (15 L ment in bioavailability and reduction in its side effects. J. Biomed.
per mouse); SLN-HDG, high dosage (20 L per mouse). Com- Nanotechnol. 9, 891 (2013).
pared with NSG, † p < 005; compared with TTG, ∗ p < 005. 8. D. Liu, Y. Ge, Y. Tang, Y. Yuan, Q. Zhang, R. Li, and Q. Xu, Solid
(n = 10). lipid nanoparticles for transdermal delivery of diclofenac sodium:

360 J. Biomed. Nanotechnol. 11, 351–361, 2015


Yong-Tai et al. Solid Lipid Nanoparticles Formulated for Transdermal Aconitine Administration and Evaluated In Vitro and In Vivo

Preparation, characterization and in vitro studies. J. Microencapsul. 25. M. R. Patel and M. F. San Martin-Gonzalez, Characterization of
27, 726 (2010). ergocalciferol loaded solid lipid nanoparticles. J. Food Sci. 77, N8
9. N. Aggarwal and S. Goindi, Preparation and in vivo evaluation of (2012).
solid lipid nanoparticles of griseofulvin for dermal use. J. Biomed. 26. K. Moser, K. Kriwet, Y. N. Kalia, and R. H. Guy, Enhanced skin
Nanotechnol. 9, 564 (2013). permeation of a lipophilic drug using supersaturated formulations.
10. S. L. Borgia, M. Regehly, R. Sivaramakrishnan, W. Mehnert, H. J. Control Release 73, 245 (2001).
C. Korting, K. Danker, B. Röder, K. D. Kramer, and M. Schäfer- 27. E. B. Souto, A. J. Almeida, and R. H. Müller, Lipid nanoparticles
Korting, Lipid nanoparticles for skin penetration enhancement- (SLN® , NLC® ) for cutaneous drug delivery: Structure, protection
correlation to drug localization within the particle matrix as and skin effects. J. Biomed. Nanotechnol. 3, 317 (2007).
determined by fluorescence and parelectric spectroscopy. J. Control 28. A. Kogan and N. Garti, Microemulsions as transdermal drug delivery
Release 110, 151 (2005). vehicles. Adv. Colloid Interface Sci. 123–126, 369 (2006).
11. J. J. Wang, K. S. Liu, K. C. Sung, C. Y. Tsai, and J. Y. Fang, 29. K. Bhaskar, J. Anbu, V. Ravichandiran, V. Venkateswarlu, and Y. M.
Skin permeation of buprenorphine and its ester prodrugs from lipid Rao, Lipid nanoparticles for transdermal delivery of flurbiprofen:
nanoparticles: Lipid emulsion, nanostructured lipid carriers and solid Formulation, in vitro, ex vivo and in vivo studies. Lipids Health Dis.
lipid nanoparticles. J. Microencapsul. 26, 734 (2009). 8, 6 (2009).
12. S. Wissing and R. Müller, The influence of the crystallinity of lipid 30. J. Shim, H. Seok Kang, W. S. Park, S. H. Han, J. Kim, and I. S.
nanoparticles on their occlusive properties. Int. J. Pharm. 242, 377 Chang, Transdermal delivery of mixnoxidil with block copolymer
(2002). nanoparticles. J. Control Release 97, 477 (2004).
13. V. Jenning, M. Schäfer-Korting, and S. Gohla, Vitamin A-loaded 31. C. Santos Maia, W. Mehnert, M. Schaller, H. C. Korting, A. Gysler,
solid lipid nanoparticles for topical use: Drug release properties. A. Haberland, and M. Schäfer-Korting, Drug targeting by solid
J. Control Release 66, 115 (2000). lipid nanoparticles for dermal use. J. Drug Target 10, 489
14. S.B. Lim, A. Banerjee, and H. Önyüksel, Improvement of drug safety (2002).
by the use of lipid-based nanocarriers. J. Control Release 163, 34 32. M. Schäfer-Korting, W. Mehnert, and H. C. Korting, Lipid nanopar-
(2012). ticles for improved topical application of drugs for skin diseases.
15. H. Chen, X. Chang, D. Du, W. Liu, J. Liu, T. Weng, Y. Yang, H. Xu, Adv. Drug Deliv. Rev. 59, 427 (2007).
and X. Yang, Podophyllotoxin-loaded solid lipid nanoparticles for 33. M. M. Abdel-Mottaleb, D. Neumann, and A. Lamprecht, Lipid
epidermal targeting. J. Control Release 110, 296 (2006). nanocapsules for dermal application: A comparative study of lipid-
16. F. L. Xiong, H. B. Chen, X. L. Chang, Y. J. Yang, H. B. Xu, based versus polymer-based nanocarriers. Eur. J. Pharm. Biopharm.
and X. L. Yang, Research progress of triptolide-loaded nanoparticles 79, 36 (2011).
delivery systems. Conf. Proc. IEEE. Eng. Med. Biol. Soc. (2005), 34. S. Jain, S. Jain, P. Khare, A. Gulbake, D. Bansal, and S. K. Jain,
Vol. 5, pp. 4966–4969. Design and development of solid lipid nanoparticles for topical deliv-
17. K. Sutthanut, X. Lu, M. Jay, and B.Delivered
Sripanidkulchai,
by Publishing ery of an to:
Solid lipid Technology anti-fungal agent. Drug Deliv. 17, 443 (2010).
xing liwei
of Kaempferia parviflora 35.
IP: 166.111.120.71 On: Tue, 20 Jan 2015 16:48:29 New microemulsion vehicle facilitates
nanoparticles for topical administration A. C. Sintov and L. Shapiro,
extracts, J. Biomed. Nanotechnol. 5, 224 (2009).
Copyright: American Scientific percutaneous penetration in vitro and cutaneous drug bioavailability
Publishers
18. I. P. Kaur, R. Bhandari, S. Bhandari, and V. Kakkar, Potential of solid in vivo. J. Control Release 95, 173 (2004).
lipid nanoparticles in brain targeting. J. Control Release 127, 97 36. B. Baroli, M. A. López-Quintela, M. B. Delgado-Charro, A. M.
(2008). Fadda, and J. Blanco-Méndez, Microemulsions for topical delivery
19. S. Dhawan, R. Kapil, and B. Singh, Formulation development and of 8-methoxsalen. J. Control Release 69, 209 (2000).
systematic optimization of solid lipid nanoparticles of quercetin for 37. M. Trotta, E. Ugazio, E. Peira, and C. Pulitano, Influence of ion
improved brain delivery. J. Pharm. Pharmacol. 63, 342 (2011). pairing on topical delivery of retinoic acid from microemulsions.
20. V. Kakkar, S. Singh, D. Singla, and I. P. Kaur, Exploring solid lipid J. Control Release 86, 315 (2003).
nanoparticles to enhance the oral bioavailability of curcumin. Mol. 38. E. Peira, M. E. Carlotti, C. Trotta, R. Cavalli, and M. Trotta, Posi-
Nutr. Food Res. 55, 495 (2011). tively charged microemulsions for topical application. Int. J. Pharm.
21. W. Weyenberg, P. Filev, D. Van den Plas, J. Vandervoort, 346, 119 (2008).
K. De Smet, P. Sollie, and A. Ludwig, Cytotoxicity of submicron 39. Q. L. Zhang, J. H. Hu, Q. G. Zhu, F. Q. Li, J. Y. Liu, and D. Wang,
emulsions and solid lipid nanoparticles for dermal application. Int. Development of a novel HPLC-MS/MS method for the determina-
J. Pharm. 337, 291 (2007). tion of aconitine and its application to in vitro and rat microdialysis
22. G. A. Castro, A. L. Coelho, C. A. Oliveira, G. A. Mahecha, R. L. samples. Biomed. Chromatogr. 23, 692 (2009).
Oréfice, and L. A. Ferreira, Formation of ion pairing as an alternative 40. P. C. Mills and S. E. Cross, Regional differences in the in vitro pen-
to improve encapsulation and stability and to reduce skin irritation etration of hydrocortisone through equine skin. J. Vet. Pharmacol.
of retinoic acid loaded in solid lipid nanoparticles. Int. J. Pharm. Ther. 29, 25 (2006).
381, 77 (2009). 41. P. C. Mills, B. M. Magnusson, and S. E. Cross, Investigation of in
23. K. Mitri, R. Shegokar, S. Gohla, C. Anselmi, and R. H. Müller, Lipid vitro transdermal absorption of fentanyl from patches placed on skin
nanocarriers for dermal delivery of lutein: Preparation, characteriza- samples obtained from various anatomic regions of dogs. Am. J. Vet.
tion, stability and performance. Int. J. Pharm. 414, 267 (2011). Res. 65, 1697 (2004).
24. C. Schwarz and W. Mehnert, Solid lipid nanoparticles (SLN) for 42. F. V. Abbott, K. B. Franklin, and R. F. Westbrook, The formalin
controlled drug delivery. II. Drug incorporation and physicochemical test: Scoring properties of the first and second phases of the pain
characterization. J. Microencapsul. 16, 205 (1999). response in rats. Pain 60, 91 (1995).

J. Biomed. Nanotechnol. 11, 351–361, 2015 361

You might also like