Sei sulla pagina 1di 11

Oral Apomorphine Delivery from Solid Lipid Nanoparticles

with Different Monostearate Emulsifiers: Pharmacokinetic


and Behavioral Evaluations
MING-JUN TSAI,1,2 YAW-BIN HUANG,3 PAO-CHU WU,3 YAW-SYAN FU,4 YAO-REN KAO,3 JIA-YOU FANG,5,6 YI-HUNG TSAI3
1
Food and Drug Administration, Department of Health, Executive Yuan, Taipei, Taiwan
2
Institute of Basic Medical Sciences, National Cheng Kung University, Taiwan
3
School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
4
Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University,
Kaohsiung, Taiwan
5
Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan
6
Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia

Received 16 January 2010; revised 30 March 2010; accepted 8 June 2010


Published online 25 August 2010 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.22285

ABSTRACT: Apomorphine, a dopamine receptor agonist for treating Parkinson’s disease, has
very poor oral bioavailability (<2%) due to the first-pass effect. The aim of this work was to in-
vestigate whether the oral bioavailability and brain regional distribution of apomorphine could
be improved by utilizing solid lipid nanoparticles (SLNs). Glyceryl monostearate (GMS) and
polyethylene glycol monostearate (PMS) were individually incorporated into SLNs as emulsi-
fiers. It was found that variations in the emulsifiers had profound effects on the physicochemical
characteristics. Mean diameters of the GMS and PMS systems were 155 and 63 nm, respec-
tively. More than 90% of the apomorphine was entrapped in the SLNs. The interfacial film was
the likely location for most of apomorphine molecules. The PMS system, when incubated in
simulated intestinal medium, was found to be more stable in terms of particle size and encapsu-
lation efficiency than the GMS system. Using the GMS and PMS systems to orally administer
apomorphine (26 mg/kg) equally enhanced the bioavailability in rats. SLNs showed 12- to 13-
fold higher bioavailability than the reference solution. The drug distribution in the striatum,
the predominant site of therapeutic action, also increased when using the SLNs. The anti-
Parkinsonian activity of apomorphine was evaluated in rats with 6-hydroxydopamine-induced
lesions, a model of Parkinson’s disease. The contralateral rotation behavior was examined after
oral apomorphine delivery. The total number of rotations increased from 20 to 94 and from 20
to 115 when the drug was administered from SLNs containing GMS and PMS, respectively.
The experimental results suggest that SLNs may offer a promising strategy for apomorphine
delivery via oral ingestion. © 2010 Wiley-Liss, Inc. and the American Pharmacists Association
J Pharm Sci 100:547–557, 2011
Keywords: apomorphine; oral absorption; bioavailability; nanoparticles; pharmacokinetics;
drug transport

INTRODUCTION striatum.1 This disease affects about 1.6% of the gen-


eral population older than 65 years, and the num-
Parkinson’s disease is characterized by progressive
ber of patients is predicted to rise as life expectancy
degeneration of dopaminergic neurons of the ni-
increases.2 Apomorphine is a mixed dopamine
grostriatal system and dopamine depletion in the
D1 /D2 receptor agonist that is potentially useful in the
Correspondence to: Jia-You Fang (Telephone: +8863-2118800
management of Parkinson’s disease. It is also used to
ext. 5521; Fax: +8863-2118236; E-mail: fajy@mail.cgu.edu.tw); Yi- treat erectile dysfunction.3 In 2004, apomorphine was
Hung Tsai (Telephone: +8867-3121101 ext. 2261; Fax: +8867- approved by the US Food and Drug Administration
3210683; E-mail: yhtsai@kmu.edu.tw)
as a rescue medication for Parkinson’s disease, par-
Journal of Pharmaceutical Sciences, Vol. 100, 547–557 (2011)
© 2010 Wiley-Liss, Inc. and the American Pharmacists Association
ticularly for treating “off” periods by a subcutaneous

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011 547


548 TSAI ET AL.

injection. The half-life of apomorphine following sub- Taiwan). N-Propylnorapomorphine, Pluronic F68
cutaneous administration is about 40 min, and the (PF68), L-ascorbic acid, pepsin, and pancreatin were
duration of the benefit ranges from 40 to 90 min.4 purchased from Sigma-Aldrich Chemical (St. Louis,
Moreover, this administration route might not be con- Missouri). Tripalmitin was supplied by Tokyo Kasei
venient for all patients. Oral administration is a deliv- (Tokyo, Japan). Hydrogenated soybean phosphatidyl-
ery system that is suitable for widespread clinical ap- choline (HSPC, 90%) was from NOF (Tokyo, Japan).
plication. However, apomorphine administered orally GMS and PMS (C18 H35 O2 (C2 H4 O)55 H) were pur-
was not successful because of the rapid degradation chased from Wako Chemical (Osaka, Japan).
in the gastrointestinal (GI) tract and first-pass effect,
resulting in bioavailability of 1.7%.5 High oral doses Preparation of SLNs
of apomorphine may cause GI complications and have
been associated with nephrotoxicity.6 The lipid and aqueous phases were prepared sepa-
The oral route continues to be a challenge, even rately. The lipid phase consisted of 200 mg of tri-
though it is the most attractive way to adminis- palmitin, 50 mg of HSPC, and 15 mg of apomorphine,
ter drugs. Incorporation of drugs into nanoparticles whereas the aqueous phase with water consisted of
opens the perspective of enhanced and less vari- 30 mg of PF68, and 12 mg of L-ascorbic acid. The emul-
able bioavailability and prolonged plasma levels.7 sifier GMS or PMS (20 mg) was added in the lipid or
Solid lipid nanoparticles (SLNs) represent an alter- aqueous phase. The total volume of the aqueous phase
native drug carrier system to emulsions and poly- was 3 mL. The reason that L-ascorbic acid was added
meric nanoparticles.8 These particles are made of was to prevent apomorphine autoxidation.14 The two
crystalline solid lipids. They can overcome the mem- phases were heated separately to 80◦ C for 20 min. The
brane stability and drug-leaching problems associ- aqueous phase was added to the lipid phase and mixed
ated with emulsions and the toxicity problems of poly- using a probe-type sonicator (UP50H, Hielscher Ul-
meric nanoparticles.9 Distinct advantages of SLNs trasonics, Teltow, Germany) for 20 min at 50 W.
include their ability to protect chemically labile in- The temperature was maintained at 80◦ C during
gredients, the possibility of large-scale production, sonication.
their controlled-release characteristics, and improved
bioavailability.10,11 Particle Size and Zeta Potential
The aim of the present work was to develop SLNs
The mean particle size (z-average) and zeta poten-
for apomorphine via the oral route, which may facil-
tial of the SLNs were measured by photon correla-
itate the oral bioavailability and brain targeting. If
tion spectroscopy (Zetasizer 3000HS; Malvern Instru-
the obtained blood profile after oral ingestion is not
ments Ltd., Worcestershire, UK) using a helium–neon
optimal, it can be modulated by changing the compo-
laser with a wavelength of 633 nm at 25◦ C. The size
sition of the nanoparticles, mainly the emulsifiers.12
values are given as a volume distribution. A 1:150 di-
Glyceryl monostearate (GMS) and polyethylene gly-
lution of the nanoparticulate systems was made using
col monostearate (PMS) were used as emulsifiers in
double-distilled water for the measurements.
SLNs to evaluate the effects of different emulsifiers
on oral apomorphine administration. The feasibility
of using apomorphine-loaded SLNs as an oral sys- Transmission Electron Microscopy
tem was demonstrated through extensive character- The size and morphology of SLNs were observed us-
ization of the size, charge, appearance of the SLNs, ing a Jeol JEM-2000EX electron microscope (Tokyo,
and drug stability. The in vivo pharmacokinetics of Japan). The system of SLNs was diluted sixfold with
apomorphine in SLNs was evaluated to elucidate the double-distilled water. One drop of the diluted SLNs
applicability. The intracerebral injection of the neu- was deposited on the carbon film–covered copper grid
rotoxin 6-hydroxydopamine (6-OHDA) has served as to form a thin-film specimen, which was then stained
an experimental basis to develop anti-Parkinsonian with 0.5% phosphotungstic acid. The sample was ex-
drugs and treatment strategies.13 This lesion model amined and photographed with the microscope.
has been used to investigate the behavioral functions
of the substantia nigra. 6-OHDA lesions in rats were
Apomorphine Entrapment Efficiency
examined after the oral delivery of apomorphine from
SLNs with various emulsifiers. The encapsulation efficiency of apomorphine en-
trapped by SLNs was determined by an ultracen-
MATERIALS AND METHODS trifugation method. The systems were centrifuged
at 120,000 rpm (649,000 × g) at 4◦ C for 2 h in a
Materials Hitachi CS150GXL ultracentrifuge (Tokyo, Japan) to
Apomorphine hydrochloride was kindly provided by separate the incorporated drug from the free form.
Standard Chemical and Pharmaceutical (Tainan, The supernatant was analyzed by high-performance

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011 DOI 10.1002/jps
ORAL APOMORPHINE DELIVERY FROM SLNS WITH MONOSTEARATE EMULSIFIERS 549

liquid chromatography (HPLC) to determine the en- the catheter. An aliquot of a 0.3-mL blood sample
trapment efficiency (%) of the total drug load. The was withdrawn from the jugular vein into a heparin-
HPLC system for apomorphine was the Hitachi 7000 rinsed vial according to a programmed schedule at 5,
series. A 25 cm long, 5 mm inner diameter stainless 10, 15, 30, 60, 120, 240, 360, and 480 min after dosing.
steel RP-18 column (Inertsil-N5 ODS; GL Sciences, Each blood sample was centrifuged at 12,000 rpm for
Torrance, California) was used. The mobile phase was 10 min. The resulting plasma sample (100 :L) was
an acetonitrile/pH 3 sodium phosphate–ethylenedi- vortex-mixed with 50 :L of a 0.05% L-ascorbic acid
aminetetraacetic acid solution (20:80) at a flow rate solution with 0.5 :g/mL of N-propylnorapomorphine.
of 1 mL/min. The UV wavelength was set to 280 nm. This mixture was extracted using 4 mL of ethyl ether
N-Propylnorapomorphine was utilized as the internal with shaking for 20 min. The resulting solution was
standard. centrifuged at 3000 rpm for 10 min, and 3 mL of the
supernatant was evaporated. The residue in the tube
Storage Stability was added to 150 :L of the mobile phase for the HPLC
analysis.
The prepared SLNs in a solution form were stored
at room temperature in a desiccator for 3 weeks. The
relative humidity was kept at 50 ± 5%. An aliquot Data Analysis
of sample was taken at predetermined time intervals Pharmacokinetic calculations were performed on
to investigate the particle size and encapsulation per- each individual set of data, using the pharmacoki-
centage. The mean size was determined by photon netic software WinNonlin Standard Edition Version
correlation spectroscopy as described in the previous 1.1 (Pharsight, Mountain View, California) using a
section. The percentage of apomorphine loading was noncompartmental method. The maximum concen-
determined by ultracentrifugation. tration, Cmax , and the time to reach Cmax (tmax ) were
determined by observing individual animal concen-
Stability in Simulated Gastric and Intestinal Media trations versus time curves. The area under the
plasma concentration curve from the time of adminis-
Stability studies were carried out in a pepsin solution
tration to infinity (AUC0→∞ ) was calculated using the
and pancreatin solution.15 The proteolytic enzyme so-
trapezoidal rule, with extrapolation to infinity. The
lution at 2.7 mL was added to 0.3 mL of apomorphine-
absolute bioavailability (F) of the oral formulations
loaded SLNs with a stirring bar. The stirring rate and
was estimated as the AUC0→∞ ratio of the oral dosage
temperature were kept at 600 rpm and 37◦ C, respec-
form to the parenteral solution after dose calibration.
tively. Samples were withdrawn at 1 h. The particle
The clearance of the oral formulation was calculated
size of SLNs after incubation was determined. The re-
from the elimination rate constant (ke ) multiplied by
maining concentration of apomorphine in the sample
the volume of distribution.
was determined. Apomorphine in samples (0.1 mL)
was extracted using 0.9 mL of methanol. After cen-
trifugation, the organic layer was carefully removed In Vivo Apomorphine Content in Brain Tissues
and subjected to the HPLC analysis.
Animals that received oral administration of 26 mg/
kg of apomorphine were decapitated at 10 min af-
Animals ter gavage. The skull was cut open, and the olfactory
The in vivo experiments were performed with male bulb, cerebellum, hippocampus, brainstem, thalamus,
Wistar albino rats (200–250 g). The animal experi- striatum, and cerebral cortex were carefully isolated
ment protocol was reviewed and approved by the In- and weighed. Subsequently, the regional brain tissue
stitutional Animal Care and Use Committee of Kaoh- was mixed with 2 mL of normal saline for homoge-
siung Medical University. Animals were housed and nization. The homogenate at 200 :L was withdrawn
handled according to institutional guidelines. All ani- and mixed with 50 :L of a 0.05% L-ascorbic acid solu-
mals were starved overnight prior to the experiments. tion with 0.5 :g/mL of N-propylnorapomorphine. The
sample preparation was the same as that described
for the plasma extraction method.
In Vivo Pharmacokinetics
Apomorphine was given orally (26 mg/kg) or the
In Vivo Rotational Behavior in 6-OHDA-Lesioned Rats
femoral vein was exposed for an apomorphine in-
jection (0.5 mg/kg, Apo-Go
R
Pen; Britannia Pharma- Animals were anesthetized with sodium pentobarbi-
ceutical, Newbury, UK). The rats were anesthetized tal (25 mg/kg) via an intraperitoneal injection and
with 25% (w/v) urethane at a dose of 2.5 mL/kg. Oral placed in a stereotaxic frame (Kopf Instruments,
formulations were delivered by an oral intubation Tujunga, California), with the incisor bar 3.3 mm be-
cannula. A parenteral solution was injected through low the interaural line. Two microliters (12 :g) of

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011
550 TSAI ET AL.

Figure 1. Transmission electron microscopic micrographs of (a) apomorphine-loaded solid


lipid nanoparticles (SLNs) with glyceryl monostearate and (b) apomorphine-loaded SLNs with
polyethylene glycol monostearate. Original magnification: 10,000×.

6-OHDA dissolved in double-distilled water was in- RESULTS


jected into the left middle forebrain bundle 4.7 mm
Physicochemical Characterization of the SLNs
posterior to the bregma, 7.8 mm ventral to the skull,
and 1.4 mm left of the midline, using a 0.5 mm The mean diameter, polydispersity index, zeta poten-
diameter Hamilton syringe at a rate of 0.25 :L/min.16 tial, and entrapment efficiency of the resulting SLNs
Our intention was to generate partial, rather than with apomorphine are listed in Table 1. The results
complete, lesions similar to the extent of nigrostriatal showed that the emulsifiers were critical parameters
dopamine depletion in patients with Parkinson’s dis- governing the particle size. The size of SLNs incorpo-
ease. One week after surgery, the animals were tested rating GMS was 155 nm, with a polydispersity index
for contralateral rotation behavior. Apomorphine- of 0.33. It can be seen that the size of SLNs was sig-
induced rotational behavior was measured following nificantly reduced (p < 0.05) to 63 nm by replacing
an oral ingestion of apomorphine (26 mg/kg) in a con- GMS with PMS, with no influence on polydispersity.
trol solution (0.05% L-ascorbic acid solution) or SLNs. To obtain more information about the particle size
Animals that had completed a 360◦ circle toward the and morphology, a transmission electron microscopic
intact (contralateral) side were continuously counted (TEM) analysis was also performed. Figures 1a and
and separately recorded. 1b show images of SLNs with GMS and PMS, respec-
tively. TEM showed that the particles had round, uni-
form shapes. A dense, well-distributed pattern was
Statistical Analysis
observed. The mean diameters of GMS- and PMS-
Statistical analysis of differences between the vari- containing SLNs were in the ranges of approximately
ous treatments was performed using unpaired Stu- 130 to 200 and 50 to 80 nm, respectively. These ranges
dent’s t test. The post hoc test used for checking indi- correlated well with those found using photon corre-
vidual differences between the formulations was the lation spectroscopy.
Newman–Keuls test. A 0.05 level of probability (p < The absolute zeta potentials of apomorphine-
0.05) was taken as the level of significance. Data en- loaded SLNs were 23 and 7 mV for the GMS and PMS
try and analysis were completed using Winks SDA systems, respectively, as shown in Table 1. A positive
6.0 software (Texasoft, Duncanville, TX, USA). surface charge was observed for these two carriers.

Table 1. The Characterization of Apomorphine-Loaded Solid Lipid Nanoparticles with Different Emulsifiers by Mean
Diameter, Polydispersity, Zeta Potential, and Encapsulation∗

Emulsifier Mean Diameter (nm) Polydispersity Zeta Potential (mV) Encapsulation (%)
Glyceryl monostearate 154.97 ± 2.83 0.33 ± 0.02 23.27 ± 0.70 90.38 ± 0.04
Polyethylene glycol monostearate 63.20 ± 0.98 0.31 ± 0.02 7.23 ± 0.25 91.03 ± 0.14

Each value represents the mean ± SD (n = 3).

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011 DOI 10.1002/jps
ORAL APOMORPHINE DELIVERY FROM SLNS WITH MONOSTEARATE EMULSIFIERS 551

Figure 2. Changes in (a) particle size (z-average) and (b) apomorphine entrapment efficiency
(%) of solid lipid nanoparticles with glyceryl monostearate (GMS) and (c) polyethylene glycol
monostearate (PMS) following storage at room temperature for 21 days. Each value is presented
as the mean and SD (n = 4).

The zeta potential of blank SLNs without the drug Stability in Simulated Gastric and Intestinal Media
was also examined. The surface charge was −15.63 ±
Table 2 summarizes the particle sizes of the GMS sys-
0.21 and −4.57 ± 0.21 mV for the GMS and PMS sys-
tem and apomorphine content remaining in carriers
tems, respectively. The encapsulation of apomorphine
after incubation in simulated gastric and intestinal
in GMS- and PMS-containing SLNs was similar (90%
fluids for 1 h. Both the mean size and polydispersity
vs. 91%).
increased (p < 0.05) in the gastric medium. A contrary
result was observed in the intestinal medium, which
Storage Stability
showed a decreasing trend after incubation. Apomor-
The physical stability of SLNs was determined by phine is known to be biologically unstable. The apo-
measuring particle sizes at 0, 7, 14, and 21 days, as morphine content in the aqueous solution (control)
depicted in Figure 2a. After 21 days of storage at room dropped from 100% to 96.88% ± 1.25% and 85.60% ±
temperature, the mean diameter of GMS-containing 1.09% after a 1-h incubation period in gastric and in-
SLNs had gradually increased from 155 to 190 nm testinal media, respectively. No protective effect of the
(p < 0.05). The particle size of PMS-containing SLNs apomorphine amount in gastric medium was found af-
remained relatively stable during this period (p > ter drug incorporation in the GMS system. The resid-
0.05). The change in the drug entrapment efficiency ual apomorphine percentage of GMS-containing car-
within SLNs with GMS and PMS during 21 days riers was 98%, which was similar to that of the free
was also examined as shown in Figures 2b and 2c, form in the aqueous solution. Table 3 gives the stabil-
respectively. Curves of the entrapment efficiency af- ity profiles of the PMS system in gastric and intestinal
ter storage were approximately the same for the two fluids. When the PMS system was kept for 1 h in gas-
systems. The entrapment efficiencies for both GMS tric medium, a slight increase in particle size from
and PMS were reduced by about 12% after 21 days of 63 to 71 nm (p < 0.05) was observed. The polydis-
storage. persity also increased after this incubation. The size

Table 2. The Mean Size, Polydispersity, and Apomorphine Content of Solid Lipid Nanoparticles with
Glyceryl Monostearate After Incubation in Simulated Gastric and Intestinal Media at 1 h∗

Condition Mean Diameter (nm) Polydispersity Drug Content (%)


0h 154.97 ± 2.83 0.33 ± 0.02 100
Gastric medium 174.63 ± 3.19 0.55 ± 0.05 94.74 ± 2.32
Intestinal medium 113.97 ± 1.16 0.26 ± 0.02 98.52 ± 0.69

Each value represents the mean ± SD (n = 3).

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011
552 TSAI ET AL.

Table 3. The Mean Size, Polydispersity, and Apomorphine Content of Solid Lipid Nanoparticles with
Polyethylene Glycol Monostearate After Incubation in Simulated Gastric and Intestinal Media at 1 h∗

Condition Mean Diameter (nm) Polydispersity Drug Content (%)


0h 63.20 ± 0.98 0.31 ± 0.02 100
Gastric medium 71.10 ± 1.00 0.45 ± 0.01 98.16 ± 1.18
Intestinal medium 62.73 ± 1.45 0.46 ± 0.04 96.11 ± 4.48

Each value represents the mean ± SD (n = 3).

receiving apomorphine gavage exhibited a concen-


tration–time profile characterized by an early peak
concentration followed by a continuous decay in the
plasma concentration. At all times, the mean plasma
concentrations were higher in rats treated with SLNs
than in those treated with the free control. Pharma-
cokinetic parameters obtained by a noncompartmen-
tal analysis after the oral administration of apomor-
phine are listed in Table 4. Following oral ingestion of
the control group, the GMS system, and the PMS sys-
tem, the average peak plasma concentrations (Cmax )
were 17.32 ± 1.62, 83.79 ± 5.37, and 116.58 ± 24.02
ng/mL at 29.27, 16.81, and 12.04 min, respectively.
The AUC0→∞ values of apomorphine for the GMS and
PMS systems were 238.61 ± 52.98 and 264.51 ± 70.28
Figure 3. Mean plasma concentration of apomorphine (ng h)/mL, respectively, whereas for the aqueous solu-
versus time curves after intravenous injection of apomor- tion, it was only 20.04 ± 1.64 (ng h)/mL. The absolute
phine solution (0.5 mg/kg) and oral administration of apo- bioavailabilities (F) of the aqueous solution, the GMS
morphine to rats at a dose of 26 mg/kg from a control so- system, and the PMS system were 2.09%, 24.85%,
lution and solid lipid nanoparticles (SLNs) with glyceryl and 27.55%, respectively. It can be seen that the res-
monostearate (GMS) or polyethylene glycol monostearate idence time of apomorphine was significantly longer
(PMS). Each value represents the mean and SD (n = 5).
(p < 0.05) for SLNs than for the control.

of the PMS system remained constant when passing In Vivo Apomorphine Content in Brain Tissues
through the intestinal medium, although the polydis- The apomorphine concentrations in different brain
persity significantly increased (p < 0.05). Similar to regions were determined to assess the distribution of
the GMS system, the PMS system exhibited a protec- the drug when orally administered as a free control
tive effect on the residual apomorphine percentage or SLNs. As shown in Figure 4, apomorphine was de-
against enzymatic attack in the intestinal medium tected in various brain regions upon oral treatment
but not the gastric medium. with the aqueous solution and SLNs for 10 min. The
drug level in brain by intravenous apomorphine injec-
In Vivo Pharmacokinetics
tion was also listed in this figure. Apomorphine con-
The plasma concentration versus time curves of apo- centrations with the SLNs in the cerebellum, brain-
morphine after a single oral dose (26 mg/kg) and in- stem, and striatum were significantly higher (p <
travenous injection (0.5 mg/kg) from the aqueous so- 0.05) than those with the aqueous solution. There
lution and SLNs are illustrated in Figure 3. Animals was no significant difference (p > 0.05) between the

Table 4. Pharmacokinetic Parameters of Apomorphine (26 mg/kg) After Oral Administration of the
Control Solution, GMS System, and PMS System to Rats∗

Parameter Control GMS System PMS System


Cmax (ng/mL) 17.32 ± 1.62 83.79 ± 5.37 116.58 ± 24.02
Tmax (min) 29.27 ± 2.50 16.81 ± 3.76 12.04 ± 1.25
AUC0→∞ [(ng h)/mL] 20.04 ± 1.64 238.61 ± 52.98 264.51 ± 70.28
F (%) 2.09 ± 0.17 24.85 ± 5.52 27.55 ± 7.32
Clearance (mL/min) 243.50 ± 17.10 43.60 ± 11.61 42.51 ± 14.50

Each value represents the mean ± SD (n = 5).
Cmax , maximum plasma concentration; Tmax , time to reach Cmax ; AUC0→∞ , area under the plasma concentration–time
curve; F, absolute bioavailability.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011 DOI 10.1002/jps
ORAL APOMORPHINE DELIVERY FROM SLNS WITH MONOSTEARATE EMULSIFIERS 553

Figure 4. Apomorphine concentrations in different brain regions at 10 min after intravenous


injection of apomorphine solution (0.5 mg/kg) and oral administration of apomorphine to rats
at a dose of 26 mg/kg from a control solution and solid lipid nanoparticles (SLNs) with glyceryl
monostearate (GMS) or polyethylene glycol monostearate (PMS). Each value represents the
mean and SD (n = 4). ∗ , p < 0.05 as compared with the data of oral control solution.

apomorphine amounts from the GMS and PMS carri- Table 5. Effect of Apomorphine Treatment on Rotational
ers. SLNs did not show a benefit for enhanced brain Behavior in 6-Hydroxydopamine-Lesioned Rats by Control
Solution and SLNs with Different Emulsifiers∗
uptake in other regions, including the olfactory bulb,
hippocampus, thalamus, and cerebral cortex. Total Number
Formulation of Rotations Rotations (min−1 )
In Vivo Rotational Behavior in 6-OHDA-Lesioned Rats Solution 20.00 ± 8.60 0.62 ± 0.14
We determined the contralateral rotation behaviors in SLNs with glyceryl 94.25 ± 9.95 0.95 ± 0.13
monostearate
rats following the oral delivery of SLNs and the cor-
SLNs with polyethylene 114.50 ± 9.33 1.08 ± 0.09
responding control. 6-OHDA was used to produce an glycol monostearate
animal model of Parkinson’s disease. Apomorphine ∗
Each value represents the mean ± SD (n = 4).
can induce contralateral turning by lesioned rats. The SLN, solid lipid nanoparticle.
animals that were not challenged with apomorphine
did not exhibit any bias in turning behavior. Oral apo-
morphine delivery from the aqueous solution resulted a subcutaneous injection. Unfortunately, as a result
in a total rotation number of 20 as shown in Table 5. of the short duration of therapeutic effect (<1 h),17
Animals displayed a total of 94 and 115 rotations fol- a high injection frequency (more than 10–15 times
lowing administration of SLNs with GMS and PMS, a day) is often required. The results of this work
respectively. The total number with the PMS system have shown that loading apomorphine into SLNs of-
was significantly greater (p < 0.05) than that with the fers benefits in terms of increased oral absorption and
GMS system. The rotation rate (rotations per minute) sustained drug efficacy. The selection of optimal emul-
of rats treated with SLNs was higher than that of rats sifiers is important to facilitate these advantages.
treated with the free control (p < 0.05). There was no PMS produced a relatively smaller particle size
significant difference (p > 0.05) between the rotation than GMS. The hydrophile–lipophile balances (HLBs)
rates of rats treated with the GMS and PMS systems. of GMS and PMS are 5.5 and 18.0, respectively. This
suggests that the more hydrophilic emulsifier inter-
face contributed to smaller sized particles. PMS used
in this work has stearate groups and a polyethylene
DISCUSSION
glycol (PEG) spacer. The ability of amphiphilic PEG
Poor oral bioavailability in patients and a short half- to act as a hydrophilic shield in drug carriers can
life for apomorphine therapy remain clinical issues. reduce the surface tension of particles, further reduc-
Apomorphine is normally clinically administered by ing the particle size.18 The emulsifier system with

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011
554 TSAI ET AL.

determined HLBs may well stabilize SLNs with spec- system showed better stability over 21 days. Transi-
ified components. One parameter for the emulsifier tion of dispersed lipid from a metastable form to a
film separating the water and lipid domains of the stable form may occur during storage.28,29 Particle
triglyceride-based system is the spontaneous mean collisions and partial destruction of the emulsifier in-
curvature, H0 ,19 which expresses the natural ten- terface can increase the possibility of aggregation of
dency of the monolayer to bend away from a flat geom- the nanoparticles.8 GMS may produce a rapid tran-
etry. H0 is positive for emulsifiers with a large polar sition from a metastable condition to a stable con-
head group and a small nonpolar group and decreases dition for SLNs. A previous study30 suggested that
with an increasing size of the nonpolar group. GMS monoglycerides exhibit lipid degradation and insta-
may be too highly lipophilic to form stable systems. In bility in SLN systems. Replacement of GMS with PMS
the absence of an optimized emulsifier, this may lead improved the stability of the dispersion. The incor-
to instability of dispersions because the interface can- poration of PEG into the lipid matrix increases the
not be fully covered by emulsifier molecules.20 surface hydrophilicity, thus inhibiting nanoparticle
SLNs without apomorphine loading showed a neg- aggregation.18 Although the size of SLNs with GMS
ative zeta potential. PF68 is a nonionic species. The increased after storage, a great fusion did not occur
negative charge shown by SLNs is believed to have in this case. Measurement of the zeta potential allows
resulted in HSPC. HSPC used in this study con- predictions about the storage stability of colloidal dis-
tained 90% phosphatidylcholine, which that is un- persions. A high zeta potential of more than ±30 mV
charged. The other components (10%), such as phos- can provide an electric repulsion that helps to prevent
phatidylethanolamine, phosphatidic acid, and phos- particle aggregation.31 Our system did not fulfill this
phatidylinositol, are negatively charged. Some free criterion. This rule cannot be strictly applied in the
fatty acids derived from the hydrolysis of monoglyc- presence of steric stabilizers such as PF68 and PEG
erides in GMS may have occurred.21 This contributed used in this work.
to the greater negative charge on GMS-containing The chemical nature of lipids is also important be-
SLNs (−16 mV) than on PMS-containing formula- cause lipids that form crystalline particles with a per-
tions (−5 mV). The addition of PEG led to an initial fect lattice lead to drug expulsion during storage.31
shielding of the negative charge. This reduction in the The SLNs developed in this study exhibited a good
case of PEG-coated nanoparticles was due to an ex- ability to reduce apomorphine expulsion. The location
tension in the plane of shear produced by the presence of apomorphine in the interfacial film but not the lipid
of this polymer on the surface.22 matrix may be the predominant factor in the reduced
Apomorphine hydrochloride has a pKa value (disso- possibility of expulsion. Although a slight increase in
ciation constant) of 8.92. Apomorphine exhibited pos- size and a decrease in the entrapment efficiency were
itive charges in the nanoparticulate systems. The in- observed, the values were still sufficient to maintain
clusion of cationic apomorphine in the interface may the stability of the SLNs.
shield the negative charge of the membrane. This The microclimate of the stomach favors particle fu-
indicates that the drug seemed to have a high affin- sion as a result of its acidity and high ionic strength.10
ity for the nanoparticle interface. Most SLNs incor- Both SLNs developed in this study showed aggrega-
porate lipophilic drugs because of the lipidic charac- tion at the same level following incubation in simu-
teristics of the inner phase. SLNs were proposed as lated gastric medium. The SLNs showed increases in
carriers for encapsulating hydrophilic drugs such as the polydispersity. This may confirm the instability of
diminazene and methotrexate.23–25 The loading level these two systems. However, the 12% increase in size
of approximately 90% by these SLNs is considered compared with the nontreatment group can be toler-
very high because apomorphine is a molecule with ated. The incubation of SLNs in simulated intestinal
high hydrophilicity. Therefore, we assumed that apo- fluid did not alter the particle size of the PMS sys-
morphine was localized within the interfacial layer tem. PEG can form hydrophilic and steric barriers
close to the aqueous phase. The crystalline tendency against the attachment of enzymes,32 thus diminish-
of the shell near the inner core decreases with the ing the aggregation process. The particle size of the
addition of phospholipids onto the interface.26 More- GMS system was even reduced after incubation in the
over, the presence of phospholipids at the interface intestinal medium. Lipid digestion and degradation
containing PF68 causes a rearrangement of the mono- can occur by enzymatic attack from pancreatic lipase
layer membrane to a disordered form.27 The resulting and trypsin.33 This can lead to a decrease in the par-
matrix of lipid particles thus produces imperfections ticle size of SLNs with GMS. The question concerning
in the crystal lattice and leaves space to accommo- the influence of the intestinal fluid on SLN degrada-
date drugs near the shell, leading to improved drug tion remains open. Unfortunately, only a few studies
entrapment. have been performed in this regard.10 It is clear that
In terms of the storage stability, the GMS system further research is needed to fully elucidate the mech-
showed physical changes in particle size. But the PMS anisms.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011 DOI 10.1002/jps
ORAL APOMORPHINE DELIVERY FROM SLNS WITH MONOSTEARATE EMULSIFIERS 555

Apomorphine is known to be chemically and biolog- the result of continuous and more sustained delivery
ically unstable. This phenomenon was insignificant of apomorphine to the circulation.
in the acidic environment because the aqueous solu- The results of drug brain distribution indicated a
tion retained 97% of the apomorphine after incuba- detectable apomorphine concentration in different re-
tion in gastric fluid. Hence, the SLNs did not exhibit gions of the brain. Apomorphine from the SLNs tar-
a protective effect on apomorphine in the gastric en- geted the cerebellum, brainstem, and striatum with
vironment. Another possibility is the release of drug a relatively high concentration compared with the
molecules from inner core to outer phase by incubat- aqueous solution. This is important for patients with
ing in the gastric medium. Apomorphine produces dif- Parkinson’s disease because the dorsal striatum is the
ferent derivatives in acidic and alkaline solutions.14 critical site of therapeutic action.38 Many sites of the
According to the experimental results of this work, brain can be considered candidates for supraspinal
SLNs were useful in protecting the drug against at- control of penile erection, including structures in the
tack by intestinal medium. Both the GMS and PMS cortex, the thalamus, and the limbic system, as well
systems showed similar effects. This may be benefi- as several nuclei in the medulla, the pons, and the
cial for prolonging apomorphine’s half-life in the GI hypothalamus.39 Because the midbrain, pons, and
tract after oral administration. medulla are components of the brainstem, SLNs may
The oral bioavailability of apomorphine from the also be beneficial in treating erectile dysfunction.
control solution was 2.09%. The results were similar It is possible that intact nanoparticles of SLNs can
to those obtained in a human study,5 which showed pass through the GI tract.35,40 Bargoni et al.41 demon-
bioavailability of 1.7%. After oral administration of strated that the uptake and transport of SLNs parti-
SLNs to rats, the bioavailability was considerably in- cles in lymph and blood after duodenal administration
creased by 12- to 13-fold compared with the control. were evidenced by TEM and by ( counting of labeled
Both systems showed similar pharmacokinetic pro- particles. Intact particles in circulation may target
files after oral ingestion. The bioavailability of sub- brain tissues for increased therapeutic potency. Many
lingual apomorphine is estimated to be 16% to 18%.34 possible mechanisms of nanoparticle-mediated drug
The oral delivery route by SLNs can surpass this transport to the brain, such as increased retention
level. These data are very encouraging and represent of nanoparticles in the brain blood capillaries, higher
a new model of an oral delivery system for apomor- concentration gradients, and endocytosis of nanopar-
phine devoid of unfavorable pharmacokinetics. The ticles, were reported.42 The most likely mechanism is
ability of SLNs to enhance the transport of the drugs endocytosis by the endothelial cells lining the brain
through the GI tract is attributed to different mecha- blood capillaries. This is triggered by binding of li-
nisms including (a) lymphatic transport, (b) drug pro- pidic SLNs to negatively charged sites on the surface
tection efficiency, and (c) sustained drug release.22,35 of endothelial cells so that they fuse with the cells for
The inclusion of drugs into lipid nanoparticles can transport across the biomembrane.7 SLNs less than
enhance targeting to the lymph system.36 Nanopar- 200 nm in size have an increased blood circulation
ticles are composed of a lipid core that can stimu- by avoiding the reticuloendothelial system and thus
late chylomicron formation, which ultimately pulls an increased time for which the drug remains in con-
the carrier along with the drug following the clas- tact with the blood–brain barrier (BBB) and for the
sical transcellular mechanism of lipid absorption.25 drug to be taken up by the brain.43 Both the GMS and
Apomorphine should be protected from the harsh in- PMS systems were of a size that fits this criterion. The
testinal environments. The advantage of SLNs devel- mechanisms underlying the brain uptake are not yet
oped in this study is the protection of the drug by fully understood. Further investigation is needed and
the carriers from enzymatic degradation, thereby en- is underway to explore the mechanisms of transport.
hancing the occurrence of lymph targeting and delay- 6-OHDA lesions significantly reduced the number
ing metabolism. When sterically stabilizing polymers of dopaminergic cells and the levels of dopamine and
such as PF68 are used, there is a steric hindrance its metabolites.2 Unilateral lesioning of the striatum
of the anchoring of the complex leading to slower with 6-OHDA is an animal model for Parkinson’s dis-
degradation.12 ease, as it reflects both the nigrostriatal dopaminergic
The benefit of the cationic charge of nanoparticles loss and increased oxidative stress.38 Parkinsonian
was demonstrated and was correlated with the inter- rats exhibit reproducible behavioral asymmetry with
action between positively charged particles and the the administration of apomorphine, resulting from
negative charge of the GI tract.37 This may be another postsynaptic dopamine receptor supersensitivity.43
reason for the oral enhancement with apomorphine- An apomorphine solution and apomorphine-loaded
loaded SLNs. SLNs can immobilize the drug and thus SLNs induced contralateral rotation after oral deliv-
serve as sustained-release systems. This allows the ery within a certain duration. Apomorphine-induced
release of apomorphine at a constant controlled rate. turning was seen in only those animals that had
Hence, the enhanced efficacy of SLNs may have been nearly complete dopaminergic damage at the lesioned

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011
556 TSAI ET AL.

site.44 This indicates the successful induction of brain of lipid peroxidation in mouse striatum. Neurochem Res
lesions by 6-OHDA in our study. Higher concentra- 34:1299–1303.
tions of apomorphine from SLNs in circulation and 2. Dalta KP, Zbarsky V, Rai D, Parkar S, Osakabe N, Aruoma
OI, Dexter DT. 2007. Short-term supplementation with plant
the brain may contribute to the higher rotation num- extracts rich in flavonoids protect nigrostriatal dopaminergic
ber and rate than for the aqueous solution. A narrow neurons in a rat model of Parkinson’s disease. J Am Coll Nutr
interindividual variability of the behavioral profiles 26:341–349.
was also observed for the group treated with SLNs. 3. Heaton JPW, Morales A, Adams MA, Johnston B, El-Rashidy
R. 1995. Recovery of erectile function by the oral administra-
It can be seen that the total number of rotations for
tion of apomorphine. Urology 45:200–206.
the PMS system was greater than that for the GMS 4. Stacy M, Silver D. 2008. Apomorphine for the acute treatment
system. Although the pharmacokinetic profiles for of “off” episodes in Parkinson’s disease. Parkinson Relat Disord
both systems were similar, the PMS system showed 14:85–92.
a higher level than the GMS system on the basis 5. Subramony JA. 2006. Apomorphine in dopaminergic therapy.
Mol Pharm 3:380–385.
of a comparison of the mean values. This insignifi-
6. Danhof M, Van der Geest R, Van Laar T, Boddé HE. 1998.
cant discrepancy may have led to a meaningful ef- An integrated pharmacokinetic-pharmacodynamic approach
fect for behavioral rotation. Nanoparticles are PEGy- to optimization of R-apomorphine delivery in Parkinson’s dis-
lated to form a hydrated stealth shell that protects ease. Adv Drug Deliv Rev 33:203–263.
particles from destruction by proteins.45,46 PEG can 7. Kaur IP, Bhandari R, Bhandari S, Kakkar V. 2008. Poten-
tial of solid lipid nanoparticles in brain targeting. J Control
prolong the blood circulation time of a drug. Com-
Release 127:97–109.
pared with classical nanoparticles, PEGylated coun- 8. Freitas C, Müller RH. 1999. Correlation between long-term
terparts show increased half-lives, decreased plasma stability of solid lipid nanoparticles (SLN R
) and crystallinity
clearance rates, and a shift in distribution in favor of the lipid phase. Eur J Pharm Biopharm 47:125–132.
of diseased tissues. A previous study47 also indicated 9. Müller RH, Runge SA, Ravelli V, Thünemann AF, Mehnert
the targeting of PEGylated nanoparticles to the brain W, Souto EB. 2008. Cyclosporin-loaded solid lipid nanopar-
ticles (SLN R
): Drug–lipid physicochemical interactions and
without damage to the BBB. Another possibility is the characterization of drug incorporation. Eur J Pharm Biopharm
smaller size of the PMS system, which had a higher 68:535–544.
total surface area for lymphatic transport. 10. Sawant KK, Dodiya SS. 2008. Recent advances and patents
on solid lipid nanoparticles. Recent Pat Drug Deliv Formul
2:120–135.
CONCLUSIONS 11. Fang JY, Fang CL, Liu CH, Su YH. 2008. Lipid nanoparticles
as vehicles for topical psoralen delivery: Solid lipid nanoparti-
The purpose of this study was to assess the feasi- cles (SLN) versus nanostructured lipid carriers (NLC). Eur J
bility of oral apomorphine delivery using SLNs as Pharm Biopharm 70:633–640.
carriers. The physicochemical properties showed that 12. Müller RH, Runge S, Ravelli V, Thünemann AF,. Souto
EB. 2006. Oral bioavailability of cyclosporine: Solid lipid
each nanoparticulate carrier (GMS and PMS systems) nanoparticles (SLN R
) versus drug nanocrystals. Int J Pharm
had distinguishable characteristics, which may affect 317:82–89.
the in vivo performances of the SLNs. Both systems 13. Schwarting RKW, Huston JP. 1996. The unilateral 6-
increased the oral bioavailability of apomorphine to hydroxydopamine lesion model in behavioral brain research.
similar levels. The clinical relevance of this is that the Analysis of functional deficits, recovery and treatments. Prog
Neurobiol 50:275–331.
dose and frequency of the drug administration can be 14. Abarca B, Ballesteros R, Bielsa P, Moragues J, D’Ocon P,
reduced. The in vivo drug distribution results indi- Garcı́a-Zaragozá E, Noguera MA. 2003. Opposite vascu-
cated that SLNs successfully targeted apomorphine lar activity of (R)-apomorphine and its oxidized derivatives.
to the brain striatum and greatly improved the abil- Endothelium-dependent vasoconstriction induced by the auto-
ity of apomorphine to treat Parkinson’s disease. In a oxidation metabolite. Eur J Med Chem 38:501–511.
15. Wu ZH, Ping QN, Lai JM, Wei Y. 2003. Hypoglycemic effect
rat model of Parkinson’s disease, we found that the of polysaccharide-coated insulin liposomes after oral adminis-
efficacy of the PMS system was superior to that of the tration in mice. Acta Pharm Sin 38:138–142.
GMS system in terms of a behavioral index. Carriers 16. Vorobyov V, Sengpiel F. 2008. Apomorphine-induced differ-
with the PEG moiety also exhibited superior stabil- ences in cortical and striatal EEG and their glutamatergic
ity as nanoparticles. It should be noted that SLNs mediation in 6-hydroxydopamine-treated rats. Exp Brain Res
191:277–287.
with PMS are a promising delivery system for apo- 17. Ikechukwu Ugwoke M, Kaufmann G, Verbeke N, Kinget
morphine after oral administration. The oral inges- R. 2000. Intranasal bioavailability of apomorphine from
tion of apomorphine from SLNs may provide a better carboxymethylcellulose-based drug delivery systems. Int J
alternative to subcutaneous administration. Pharm 202:125–131.
18. Yuan H, Wang LL, Du YZ, You J, Hu FQ, Zeng S. 2007.
Preparation and characteristics of nanostructured lipid carri-
REFERENCES ers for control-releasing progesterone by melt-emulsification.
Colloids Surf B-Biointerf 60:174–179.
1. Kabuto H, Amakawa M, Mankura M, Yamanushi TT, Mori 19. von Corswant C, Thorén P, Engström S. 1998. Triglyceride-
A. 2009. Docosahexaenoic acid ethyl ester enhances 6- based microemulsion for intravenous administration of spar-
hydroxydopamine-induced neuronal damage by induction ingly soluble substances. J Pharm Sci 87:200–208.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011 DOI 10.1002/jps
ORAL APOMORPHINE DELIVERY FROM SLNS WITH MONOSTEARATE EMULSIFIERS 557

20. Helgason T, Awad TS, Kristbergsson K, McClements DJ, 35. Yang S, Zhu J, Lu Y, Liang B, Yang C. 1999. Body distribution
Weiss J. 2009. Effect of surfactant surface coverage on for- of camptothecin solid lipid nanoparticles after oral adminis-
mation of solid lipid nanoparticles (SLN). J Colloid Interface tration. Pharm Res 16:751–757.
Sci 334:75–81. 36. Priano L, Esposti D, Esposti R, Castagna G, De Medici L,
21. Wang JJ, Liu KS, Sung KC, Tsai CY, Fang JY. 2009. Lipid Fraschini F, Gasco MR, Mauro A. 2007. Solid lipid nanopar-
nanoparticles with different oil/fatty ester ratios as carriers of ticles incorporating melatonin as new model for sustained
buprenorphine and its prodrugs for injection. Eur J Pharm Sci oral and transdermal delivery systems. J Nanosci Nanotechnol
38:138–146. 7:3596–3601.
22. Garcia-Fuentes M, Prego C, Torres D, Alonso MJ. 2005. A com- 37. Poullain-Termeau S, Crauste-Manciet S, Brossard D,
parative study of the potential of solid triglyceride nanostruc- Muhamed S, Nicolaos G, Farinotti R, Barthélémy C, Robert
tures coated with chitosan or poly(ethylene glycol) as carriers H, Odou P. 2008. Effect of oil-in-water submicron emulsion
for oral calcitonin delivery. Eur J Pharm Sci 25:133–143. surface charge on oral absorption of a poorly water-soluble
23. Olbrich C, Gessner A, Schröder W, Kayser O, Müller RH. 2004. drug in rats. Drug Deliv 15:503–514.
Lipid–drug conjugate nanoparticles of the hydrophilic drug 38. Kelsey JE, Langelier NA, Oriel BS, Reedy C. 2009. The ef-
diminazene: Cytotoxicity testing and mouse serum adsorption. fects of systemic, intrastriatal, and intrapallidal injections of
J Control Release 96:425–435. caffeine and systemic injections of A2A and A1 antagonists on
24. Ruckmani K, Sivakumar M, Ganeshkumar PA. 2006. forepaw stepping in the unilateral 6-OHDA-lesioned rat. Psy-
Methotrexate loaded solid lipid nanoparticles (SLN) for chopharmacology 201:529–539.
effective treatment of carcinoma. J Nanosci Nanotechnol 39. Pampin O. 2001. Mode of action of a new oral treatment
6:2991–2995. for erectile dysfunction: Apomorphine SL. BJU Int 88(Suppl.
25. Paliwal R, Rai S, Vaidya B, Khatri K, Goyal AK, Mishra N, 3):22–24.
Mehta A, Vyas SP. 2009. Effect of lipid core material on char- 40. Zara GP, Bargoni A, Cavalli R, Fundarò A, Vighetto D,
acteristics of solid lipid nanoparticles designed for oral lym- Gasco MR. 2002. Pharmacokinetics and tissue distribution
phatic delivery. Nanomedicine 5:184–191. of idarubicin-loaded solid lipid nanoparticles after duodenal
26. zur Mühlen A, Schwarz C, Mehnert W. 1998. Solid lipid administration to rats. J Pharm Sci 91:1324–1333.
nanoparticles (SLN) for controlled drug delivery: Drug release 41. Bargoni A, Cavalli R, Caputo O, Fundaró A, Gasco MR, Zara
and release mechanism. Eur J Pharm Biopharm 45:149–155. GP. 1998. Solid lipid nanoparticles in lymph and plasma
27. Fang JY, Hung CF, Liao MH, Chien CC. 2007. A study of after duodenal administration to rats. Pharm Res 15:745–
the formulation design of acoustically active lipospheres as 750.
carriers for drug delivery. Eur J Pharm Biopharm 67:67–75. 42. Kreuter J. 2001. Nanoparticulate systems for brain delivery
28. Zhang X, Pan W, Gan L, Zhu C, Gan Y, Nie S. 2008. Prepa- of drugs. Adv Drug Deliv Rev 47:65–81.
ration of a dispersible PEGylate nanostructured lipid carriers 43. Senthilkumar KS, Saravanan KS, Chandra G, Sindhu KM,
(NLC) loaded with 10-hydroxycamptothecin by spray-drying. Jayakrishnan A, Mohanakumar KP. 2007. Unilateral im-
Chem Pharm Bull 56:1645–1650. plantation of dopamine-loaded biodegradable hydrogel in
29. Venkateswarlu V, Manjunath K. 2004. Preparation, charac- the striatum attenuates motor abnormalities in the 6-
terization and in vitro release kinetics of clozapine solid lipid hydroxydopamine model of hemi-Parkinsonism. Behav Brain
nanoparticles. J Control Release 95:627–638. Res 184:11–18.
30. Radomska-Soukharev A. 2007. Stability of lipid excipients in 44. Camp DM, Loeffler DA, Farrah DM, Borneman JN, LeWitt PA.
solid lipid nanoparticles. Adv Drug Deliv Rev 59:411–418. 2009. Cellular immune response to intrastriatally implanted
31. Müller RH, Mäder K, Gohl S. 2000. Solid lipid nanoparticles allogeneic bone marrow stromal cells in a rat model of Parkin-
(SLN) for controlled drug delivery: A review of the state of the son’s disease. J Neuroinflamm 6:17.
art. Eur J Pharm Biopharm 50:161–177. 45. Harris JM, Chess RB. 2003. Effect of pegylation on pharma-
32. Zhang N, Ping Q, Huang G, Xu W, Cheng Y, Han X. 2006. ceuticals. Nat Rev Drug Discov 2:214–221.
Lectin-modified solid lipid nanoparticles as carriers for oral 46. Zhang JQ, Liu J, Li XL, Jasti BR. 2007. Preparation and
administration of insulin. Int J Pharm 327:153–159. characterization of solid lipid nanoparticles containing silib-
33. Nicolaos G, Crauste-Mancient S, Farinotti R, Brossard D. inin. Drug Deliv 14:381–387.
2003. Improvement of cefpodoxime proxetil oral absorption in 47. Calvo P, Gouritin B, Chacun H, Desmaële D, D’Angelo J,
rats by an oil-in-water submicron emulsion. Int. J. Pharm. Noel J, Georgin D, Fattal E, Andreux JP, Couvreur P. 2001.
263:165–171. Long-circulating PEGylated polycyanoacrylate nanoparticles
34. Argiolas A, Hedlund H. 2001. The pharmacology and clinical as new drug carrier for brain delivery. Pharm Res 18:1157–
pharmacokinetics of apomorphine SL. BJU Int 88:18–21. 1166.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 100, NO. 2, FEBRUARY 2011

Potrebbero piacerti anche