Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

http://informahealthcare.

com/phd
ISSN: 1083-7450 (print), 1097-9867 (electronic)

Pharm Dev Technol, Early Online: 1–8


! 2013 Informa Healthcare USA, Inc. DOI: 10.3109/10837450.2013.829092

ORIGINAL RESEARCH ARTICLE

Development and characterization of lutein-loaded SNEDDS for


enhanced absorption in Caco-2 cells
Pattravee Niamprem, Soravoot Rujivipat, and Waree Tiyaboonchai
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by University Library Utrecht on 09/01/13

Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand

Abstract Keywords
A self-nanoemulsifying drug delivery system (SNEDDS) has been developed for enhanced oral Absorption, Caco-2 cells, dissolution, lutein,
bioavailability of lutein. Its permeation enhancement has been evaluated using monolayers self-nanoemulsifying drug delivery system,
of Caco-2 cells. SNEDDS is composed of a mixture of LexolÕ and EmulmetikÕ 900, LabrasolÕ , solubility
and Tween 80 as oil, surfactant and co-surfactant, respectively. Upon dilution of lutein-
loaded SNEDDS with water, a nanoemulsion was obtained in 510 s with spherical droplets of History
40–150 nm in diameter. The zeta potential was in the range of 19 to 32 mV. Increasing the
ratio of surfactant to co-surfactant decreased the mean droplet size. Dissolution studies showed Received 12 March 2013
that lutein was released rapidly (55 min) from SNEDDS into 0.1 N HCl and pH 6.8 phosphate Revised 6 June 2013
buffer solution without any aggregation. In vitro studies using Caco-2 cells revealed that lutein- Accepted 11 July 2013
loaded SNEDDS showed shorter lag time and greater (2-fold) cellular accumulation compared Published online 28 August 2013
with the lutein dispersion.
For personal use only.

Introduction delivery systems16–18, the effects of different formulation factors,


such as ratio of oil phase to surfactant phase and ratio of surfactant
Age-related macular degeneration (AMD) is one of the leading
to co-surfactant on the in vitro release, transport, and accumu-
causes of blindness in people over the age of 60 in industrialized
lation in Caco-2 cells, are not known.
countries1. AMD affects 50% of people at age 75  5 years
The objectives of this study, therefore, were to characterize the
and is rarely found in people 550 years of age2. The etiology
physicochemical properties of the lutein-loaded SNEDDS under
of AMD is not completely understood, but oxidative stress is
various formulation factors with measurement of emulsification
known to play a role.
time, droplet size and zeta potential analysis, morphology, and
Lutein, a well-known antioxidant, is highly concentrated in
in vitro release of SNEDDS formulation. Moreover, the transport
the macula and lens of the eyes. It plays an important role in the
and accumulation of lutein have been investigated using mono-
prevention of AMD by blocking the high-energy wavelengths of
layers of Caco-2 cells.
blue light3–5. This effect is thought to reduce drusen formation4,6.
In addition, clinical and epidemiological studies have indicated
Materials and methods
that increased intake of lutein (6 mg/day) can reduce the risk of
AMD2,7,8. Materials
Unfortunately, lutein exhibits poor water solubility and as a
Lutein was purchased from KEB Biotechnology Co., Ltd.
result, it has a low oral bioavailability. To overcome this problem,
(Beijing, China). Standard lutein was purchased from Sigma
in recent years, much attention has been focused on developing
chemical (Lot No.070M4056, MO). LabrasolÕ (caprylocaproyl
lipid-based formulations for enhancing the oral bioavailability
macrogol-8-glycerides, LS) and TranscutolÕ HP (diethylene
of poorly water-soluble drugs9,10. Self-nanoemulsifying drug
glycol monoethyl ether) were purchased from Gattefosse
delivery system (SNEDDS) is one of the most popular approaches
(Cedex, France). EmulmetikÕ 900 (Soybean phospholipids,
that can increase interfacial area of the drugs to improve
E 900) was purchased from Lucas Meyer Cosmetic (Champlan,
solubilization as well as absorption across the mucosa11,12.
France). Tween 80 [Polyoxyethylene (20) sorbitan monooleate,
SNEDDS is an isotropic mixture of natural/synthetic oils,
T80] was purchased from Ajex Finechem Pty Ltd. (Seven Hills,
surfactants/co-surfactants and drug. Upon mild agitation followed
Australia). LexolÕ (medium chain triglyceride), propylene glycol,
by dilution with aqueous media, oil-in-water nanoemulsions
polyethylene glycol 400 (PEG 400) were purchased from
are instantaneously formed9,10,13–15. Although several studies
Namsiang Co., Ltd. (Bangkok, Thailand). Caco-2 cells were
have developed and characterized lutein in self-emulsifying drug
purchased from the American Type Culture Collection (VA).
Dulbecco’s modified Eagle’s medium (DMEM F-12) and all the
materials for cell culture were purchased from Sigma chemical
(MO). Cell culture plate and TranswellÕ plate (12 mm diameter,
Address for correspondence: Waree Tiyaboonchai, Department of
0.4 mm-pore-size polycarbonate membrane) were purchased
Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, from Corning Costar (MA). 3-[4,5-dimethylthiazol-2-yl]-2,5-
Naresuan University, Phitsanulok, Thailand. Tel: +66 55 261873. Fax: diphenyl-tetrazolium bromide (MTT) and dimethy sulfoxide
+66 55 261057. E-mail: wareet@nu.ac.th were purchased from AmrescoÕ (OH).
2 P. Niamprem et al. Pharm Dev Technol, Early Online: 1–8

Development of lutein-loaded SNEDDS Micro/nano-emulsions was obtained by introducing 50 ml of


formulation in 50 ml deionized water and mixed for 10 min.
Solubility study
On carbon-coated 300 mesh copper grid, 20 ml of the micro/
The solubility of lutein in various vehicles, including oil, nano-emulsions were directly deposited and stained with 10 ml
surfactant and co-surfactant, were determined using shake flask of uranyl acetate solution (0.5% w/v). Excessive solvent was
method19. An excess amount of lutein was added to 1 ml of each removed by Whatman filter paper (11 mm). The dried sample was
vehicle. The mixture was shaken for 48 h at ambient temperature. kept in desiccator overnight before observation by TEM.
After equilibrium, the mixture was centrifuged at 14 000 rpm for The mean droplet size and size distribution of SNEDDS were
10 min (Mikro 120, Hettich, Burladingen, Germany). The super- determined by dynamic light scattering with a ZetaPALSÕ
natants were diluted appropriately with a mobile phase, a mixture analyzer (ZetaPALS, Brookhaven Corp., NY). The micro/nano-
of 2-propanol/dichloromethane/methanol (20:10:70, v/v), before emulsions were transferred to polystyrene cuvette. Measurements
applied to the high performance liquid chromatography (HPLC). were then carried out at 90 to the incident light, the wavelength
The dissolved lutein was quantified with a calibration curve of 659 nm in 10 mm diameter cell. The measurement was
of standard lutein in a concentration range of 0.01–5 mg/ml. repeated 10 times. The mean droplet size and size distribution
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by University Library Utrecht on 09/01/13

An HPLC apparatus (Model LD10A, Shimadzu, Kyoto, Japan) were obtained from the method of cumulants. In addition, the zeta
equipped with a Vertisep bio C30 column (5 mm, 4.6  100 mm) potential of micro/nano-emulsions were determined by measuring
was used. A flow rate of 1.0 ml/min and detection wavelength of the droplet electrophoretic mobility using ZetaPALSÕ . The
446 nm were employed. measurement was repeated 5 times. The zeta potential was
calculated based on the Smoluchowski equation23.
Construction of pseudo-ternary phase diagram Emulsification time was determined by adding 0.5 ml
SNEDDS formulation in a paddle apparatus (USP XXXII)
Pseudo-ternary phase diagrams were constructed (using CHEMIX
(VankelÕ VK 700, Vankel Industries, Edison, NJ) [50 rpm,
School software, version 3.51) in the absence of lutein by
37  C, 900 ml, 0.1 N HCl and pH 6.8 phosphate buffer solution,
water titration method at ambient temperature20–22. Based on the
n ¼ 3]18. Time for the appearance of micro/nano-emulsions was
solubility study, three SNEDDS formulation systems were
visually monitored.
investigated. The mixture of LexolÕ and EmulmetikÕ 900 at 9:1
was used as oil phase in all systems. Surfactants and co-
In vitro dissolution studies
surfactants (LabrasolÕ and Tween 80; LabrasolÕ and TranscutolÕ
HP; Tween 80 and TranscutolÕ HP) were used in system I, II and Dissolution studies were performed using a basket apparatus (USP
III, respectively. Surfactant and co-surfactant were mixed in XXXII) (UDT-804, Logan Instrument Corp., NJ) [100 rpm, 37  C,
For personal use only.

different volume ratios (7:3, 8:2 and 9:1). For each pseudo-ternary 900 ml, 0.1 N HCl and pH 6.8 phosphate buffer solution, n ¼ 3].
phase diagram at specific surfactant/co-surfactant ratio, oil and Lutein-loaded SNEDDS or lutein powder equivalent to 5 mg
surfactant/co-surfactant mixtures were mixed thoroughly in of lutein was filled into hard gelation capsule No. 0 prior to
different volume ratios (1:9,1.5:8.5, 2:8, 2.5:7.5, 3:7, 3.5:6.5, introduction into the dissolution tester. An aliquot of 5 ml was
4:6, 5:5, 6:4, 7:3, 8:2 and 9:1). Each mixture was titrated with withdrawn at 0, 5, 10, 15 and 30 min and then replaced with fresh
distilled water and visually examined for transparency after medium. Samples were filtered and subjected to lutein analysis
equilibrium. Then, the mixture was further titrated with aliquots using UV spectrophotometer at 446 nm.
of distilled water until turbidity has been reached. Clear and
isotropic samples were deemed to be within the microemulsions Determination of lutein SNEDDS absorption in
regions corresponding to the selected optimum ratios of combin- Caco-2 cells
ation vehicles for developing lutein-loaded SNEDDS.
Cell culture
Preparation of lutein-loaded SNEDDS formulations and Caco-2 cells were maintained in DMEM/F-12 supplemented with
content determination 10% fetal bovine serum (heat-inactivated at a temperature of
56  C for 30 min) and 1% penicillin/streptomycin. Cells were
From pseudo-ternary phase diagram, lutein-loaded SNEDDS,
grown at 37  C, 5% CO2, and cultured for 14–21 days to obtain a
0.5% lutein, were prepared using the mixture of LexolÕ and
differentiated cell monolayer prior to use. All studies were
EmulmetikÕ 900 at 9:1 as oil phase and LabrasolÕ and Tween 80
performed between passage 35 and 45. The integrity of the
as surfactant phase. All components were mixed using a magnetic
monolayer was confirmed by measuring the transepithelial
stirrer at 80  C for 10 min or until a clear solution was obtained.
electrical resistance (TEER) using a volt-ohm meter (MillicellÕ
Then, the formulation was equilibrated at ambient temperature
ERS-2, Millipore Corporation, MA). Only cell monolayer with
for 24 h to examine for turbidity or phase separation before
TEER value of 500  100
cm2 were used22,24.
characterization. All samples were prepared in triplicate. The
varying formulation factors were the ratios of oil phase to
Cell viability
surfactant phase at 3:7, 2.5:7.5 and 2:8 and the ratios of LabrasolÕ
to Tween 80 at 9:1, 8:2 and 7:3. Cell viability of lutein-loaded SNEDDS on the Caco-2 cells was
The lutein content in SNEDDS was determined. Each formu- evaluated by MTT assay. Cells were seeded in 96-well plate at a
lations (20 ml) were diluted with 980 ml of 95% ethanol. density of 1  104 cells/well in 100 ml of cell culture medium.
The mixture was then vortexed vigorously for 2 min and diluted Immediately prior to each experiment, the medium was removed.
100-fold with the mobile phase before the content of lutein was Then, the cells were incubated with the 100 ml of lutein-loaded
determined by HPLC as describe earlier. SNEDDS dilutions for 4 h. After that, 10 ml of MTT (5 mg/ml in
phosphate buffer saline) was added and incubated at 37  C, 5%
CO2, for 2 h. The formazan crystals were dissolved in 100 ml of a
Physicochemical characterization of lutein-loaded
mixture of 0.1 M glycine:DMSO (1:9). The absorbance of
SNEDDS formulations
formazan at 595 nm was measured using microplate reader (DTX
The morphology of lutein-loaded SNEDDS was investigated by 880, Multimode Detector, Beckmancoulter, CA). Cell viability
transmission electron microscope (TEM, Tecnai 12, Philips, OR). was expressed as a percentage of the control group.
DOI: 10.3109/10837450.2013.829092 Absorption of lutein SNEDDS in Caco-2 cells 3

Lutein transport and cellular accumulation 5:5 produced turbid preparations, and hence were not employed.
Õ 5 Nonionic surfactants, LabrasolÕ (HLB 14) and Tween 80 (HLB
Cells were seeded in Transwell plate at density of 1  10 cells/
15), provided good solubility of lutein (4.76  1.25 and
well for 14–21 days. The culture medium was changed every 2
4.41  0.48 mg/ml, respectively). Co-surfactant TranscutolÕ HP
days. After differentiation, cells were incubated with serum-free,
was found to be an effective solubilizer for lutein with solubility
non-phenol red medium overnight before use. At the beginning of
of 7.24  1.52 mg/ml, therefore it was used in the SNEDDS
each experiment, 0.5 ml of lutein-enrich medium (containing
development for drug loading improvement.
lutein-loaded SNEDDS with serum free medium) was added
to the apical chamber, and 1.5 ml of serum free medium was
Construction of the pseudo-ternary phase diagram
added to the basolateral chamber. The samples were incubated
at 37  C, 5% CO2. In this experiment, the content of lutein Based on the results of solubility studies, pseudo-ternary
in three parts, apical chamber, basolateral chamber and cells, phase diagrams in the absence of lutein were constructed to
were collected at 1, 2, 3, 4, 5 and 6 h. Medium from apical identify microemulsion and thus to determine the optimum
and basolateral chamber were harvested. Cell monolayer was ratio of oil, surfactant and co-surfactant. The phase diagrams
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by University Library Utrecht on 09/01/13

washed twice with 0.5 ml of ice-cold pH 7.4 phosphate buffer were constructed for the following three systems; LabrasolÕ and
saline to eliminate lutein on monolayer surface. Then, the Tween 80, LabrasolÕ and TranscutolÕ HP, and Tween 80 and
cell monolayer was scraped and collected in 1 ml of phosphate TranscutolÕ HP.
buffer saline. All the samples were stored at 80  C under The phase diagrams of the system containing LabrasolÕ
nitrogen for a maximum 2 days before lutein extraction and and Tween 80 showed the largest region of microemulsion
quantitation. The transport of lutein from the apical to the (Figure 1A) compared to preparations containing Tween 80 and
basolateral chamber was expressed as permeation amount of TranscutolÕ HP (Figure 1C) or LabrasolÕ and TranscutolÕ
lutein versus time. The accumulation of lutein in Caco-2 cells HP (Figure 1B). TranscutolÕ HP enhanced the highest solubil-
monolayer was expressed as accumulation amount of lutein versus ity of lutein but produced a narrower microemulsion region.
time. To investigate the effect of different SNEDDS formulations, Therefore, the system containing LabrasolÕ and Tween 80, which
the content of lutein in three parts was examined after incubated provide the largest microemulasion region, was chosen for further
for 6 h. investigations.
For lutein extraction, 100 ml of aqueous sample was mixed The effect of the ratios of LabrasolÕ to Tween 80 (7:3, 8:2 and
with 500 ml of dichloromethane and vortexed vigorously for 2 min. 9:1) on the microemulsion region was investigated. The system
The sample was then centrifuged at 10 000 rpm for 10 min, with LabrasolÕ to Tween 80 at a ratio of 7:3 showed the largest
and the supernatant was collected. Lutein was extracted twice, microemulsion region followed by those containing a ratio of
For personal use only.

and then the resultant supernatant was evaporated under nitrogen. 8:2 and 9:1, respectively (data not shown). Increasing Tween 80
The dried extract was dissolved in 500 ml of mobile before resulted in increased microemulsion region.
HPLC was used to determine the lutein content. In addition, the
cell samples were centrifuged at 3000 rpm for 5 min to remove Physicochemical properties
supernatant from cells. Then, lutein in cell samples was extracted
using the above method. Upon mixing lutein-loaded SNEDDS with water, oil-in-water
nanoemulsions formed rapidly without any precipitation even
Statistical analysis after 1 day of storage. However, the system with LabrasolÕ and
Tween 80 at a ratio of 6:4 produced turbidity after 1 day of
All data were expressed as mean  SD from at least three different storage. Regardless of formulation factors, TEM micrographs
experiments. The data were analyzed by one-way analysis of of the nanoemulsions showed spherical droplets (Figure 2).
variance (ANOVA). Differences were considered to be significant However, formulation factors affected the mean particle size.
when the p value was 0.05. It decreased from 154 to 111 nm upon an increase of surfactant
phase (Table 2). Increasing Tween 80 in LabrasolÕ and Tween 80
Results system decreased the droplet size from 139 to 36 nm (Table 2).
Development of lutein-loaded SNEDDS The zeta potential of all formulations was in the range of 19 and
32 mV.
Solubility studies Emulsification time of lutein-loaded SNEDDS was evaluated
As shown in Table 1, among various oils examined, a mixing ratio as an important index for the assessment of the efficiency of self-
of LexolÕ to EmulmetikÕ 900 at 9:1 provided the highest emulsification. Upon mixing with both medium (0.1 N HCl
solubility of lutein, 2.12  0.08 mg/ml, and was chosen for further and pH 6.8 phosphate buffer solution), all formulations exhibited
investigations. In preliminary studies, ratios of 8:2, 7:3, 6:4 and rapid self-nanoemulsification with emulsification time ranging
from 6 to 12 s (Table 2). No phase separation or aggregation was
observed in any of these formulations.
Table 1. Solubility of lutein in various vehicles.
In vitro dissolution studies
Solubility
Vehicle Component (mg/ml) The cumulative percent release of lutein form SNEDDS at
Õ varying time intervals were examined. Lutein was rapidly released
Labrasol Surfactant 4.76  1.25
Tween 80 4.41  0.48 from SNEDDS, 90% within 5 min, and the release profile was
TranscutolÕ HP Co-surfactant 7.24  1.52 independent of formulation parameters. Moreover, similar drug
PEG 400 1.79  0.19 release pattern was observed for both dissolution mediums; 0.1 N
Propylene glycol 0.22  0.02 HCl and pH 6.8 phosphate buffer solution (Figure 3A and B).
LexolÕ : EmulmetikÕ 900 (9:1) Oil 2.12  0.08 On the other hand, lutein powder exhibited extremely poor
LexolÕ 1.82  0.24 dissolution, 51% of lutein dissolved in 30 min in both medium.
Corn oil 0.93  0.09
Olive oil 0.75  0.11
As expected, the dissolution rate of SNEDDS was significantly
higher than that of lutein powder.
4 P. Niamprem et al. Pharm Dev Technol, Early Online: 1–8
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by University Library Utrecht on 09/01/13

Figure 2. TEM micrograph of lutein-loaded SNEDDS OS8 at a


magnification of 120 000. Bar corresponds to 200 nm.

480% was considered as non-toxic25. At high lutein concentration


(30 mg/ml), the cell viability of SNEDDS LT1 and OS8 decreased
to 40% and 70%, respectively. On the contrary, SNEDDS tested
For personal use only.

at lower lutein concentrations (10, 5, 3 and 1 mg/ml) possessed


cell viability 480% indicating no cytotoxicity. Thus, 3 mg/ml
of lutein-loaded SNEDDS was selected for transport studies (data
not shown).
The cellular accumulation of lutein-loaded SNEDDS OS8
was investigated comparing to lutein dispersion. As shown in
Figure 4, the cellular accumulations of lutein from SNEDDS and
lutein dispersion after 1-h incubation were 0.02 and 0.01 mg/
cm2, respectively; the difference was statistically significant
(p ¼ 0.002). Lag times of 3 and 4 h were observed with the
SNEDDS and lutein dispersion, respectively (Figure 5). However,
the permeation amount of lutein from SNEDDS was similar to
that of lutein dispersion (p ¼ 0.59). Therefore, the 6-h incubation
was chosen for further investigation of the effect of formulation
excipients on the transport. The lutein transport of different lutein-
loaded SNEDDS formulation was investigated using differentiated
Caco-2 cells. The lutein distribution of SNEDDS in apical
chamber, basolateral chamber and cell monolayer after 6-h
incubation was shown in Table 3. The percentage of lutein
recovery was 80% of the initial level. There were no significant
difference in percentage of lutein in apical and basolateral
chamber with varying formulation factors. However, the percent-
age of lutein in cell monolayer tended to increase with higher
ratio of surfactant phase and LabrasolÕ (Table 3). The TEER
value of cell monolayer before and after experiment was similar,
suggesting that the integrity of cell monolayer was maintained
Figure 1. Pseudo-ternary phase diagrams of SNEDDS formulations. (A) during the in vitro absorption studies (data not shown).
SNEDDS system I, (B) SNEDDS system II and (C) SNEDDS system III.
The gray area represents microemulsion regions.
Discussion
Lutein exhibits poor water solubility with a log octanol/water
Cytotoxicity and in vitro transport in Caco-2 cells
partition coefficient (log Poct) of 14.8 resulting in low oral
The surfactant in the SNEDDS formulations could damage the bioavailability26. To overcome this problem, SNEDDS was
intestinal epithelial barrier. Therefore, the potential cytotoxicity chosen as a delivery system as it could improve bioavailability
of SNEDDS formulation on differentiated Caco-2 cells was of lutein through surfactant-induced enhancement in its dissol-
assessed by MTT assay. Cultures were incubated with varying ution. SNEDDS is an isotropic mixture of natural/synthesis oils,
concentration of lutein-loaded SNEDDS for 6 h. Cell viability surfactants/co-surfactants and drug. Upon mild agitation as the
DOI: 10.3109/10837450.2013.829092 Absorption of lutein SNEDDS in Caco-2 cells 5
Table 2. Effect of formulation factors on physicochemical characterization of lutein-loaded SNEDDS.

Emulsification time (s) (mean  SD)


Mean droplet size Polydispersity index Zeta potential Phosphate buffer
Formulation Formulation factor (nm) (mean  SD) (mean  SD) (mV) (mean  SD) solution 0.1 N HCl
Ratio of oil phase: surfactant phase
OS7 3:7 154  14 0.173  0.015 32.94  2.94 12  2 71
OS7.5 2.5:7.5 138  4 0.186  0.019 30.41  1.93 91 61
OS8 2:8 111  8 0.263  0.012 25.01  1.78 71 71
Ratio of LabrasolÕ :Tween 80
LT1 9:1 139  9 0.228  0.036 31.26  1.89 10  2 71
LT2 8:2 111  8 0.263  0.012 25.01  1.78 71 71
LT3 7:3 36  6 0.291  0.022 19.01  7.35 81 61
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by University Library Utrecht on 09/01/13
For personal use only.

Figure 3. Cumulative percentage released of lutein from lutein powder and lutein-loaded SNEDDS in (A) 0.1 N HCl and (B) pH 6.8 phosphate buffer
solution. Each value represents means  SD (n ¼ 3).

digestive motility and dilution with aqueous medium, oil-in-water ternary phase diagram studies. Oil is a necessary ingredient
micro/nano-emulsion could be instantaneously formed and spread for SNEDDS because it can solubilize the lipophilic drug in
rapidly in gastrointestinal tract9,10,13,14,18. a specific amount, facilitate self-emulsification, and increase
The components of developed SNEDDS (oil, surfactant and enterocyte and intestinal lymphatic uptake, thereby increasing
co-surfactant) were selected based on the solubility and pseudo- lipophilic drug absorption from the gastrointestinal tract.
6 P. Niamprem et al. Pharm Dev Technol, Early Online: 1–8
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by University Library Utrecht on 09/01/13

Figure 4. Accumulative amount of lutein in Caco-2 cells monolayer. Each value represents means  SD (n ¼ 3).

Figure 5. Permeation amount of lutein across


Caco-2 cells monolayer. Each value repre-
sents means  SD (n ¼ 3).
For personal use only.

Table 3. The percentage of lutein in apical chamber, basolateral chamber and cell monolayer.

Lutein (%)
Formulation Formulation factor Apical Basolateral Cell
Lutein dispersion 64.92  2.51 11.70  0.33 0.94  0.10
Ratio of oil phase: surfactant phase
OS7 3:7 65.60  4.23 11.64  0.68 1.77  0.50
OS7.5 2.5:7.5 65.69  2.81 12.00  0.09 1.81  0.24
OS8 2:8 69.72  3.79 11.26  0.68 1.93  0.41
Ratio of LabrasolÕ :Tween 80
LT1 9:1 68.12  5.18 12.16  0.22 2.08  0.20
LT2 8:2 69.72  3.79 11.26  0.68 1.93  0.41
LT3 7:3 66.41  1.32 12.21  0.23 1.66  0.34

From the solubility study, the mixture of LexolÕ and the interfacial energy to improve the thermodynamic stability of
EmulmetikÕ 900 (9:1) was selected as an oil phase due to the microemulsions27. Upon dilution in aqueous solution, they assist
highest solubilizing effect (2.12  0.08 mg/ml). EmulmetikÕ 900 the immediate formation of oil in water droplets and rapid
consists of 50% w/w phosphatidylcholine that could help spreading of the formulation in the medium. Based on pseudo-
facilitate micelle formation16. In general, surfactant and co- ternary phase diagram studies, LabrasolÕ and Tween 80 were
surfactant are preferentially absorbed at the interface, reducing selected as a surfactant phase. They are nonionic surfactants
DOI: 10.3109/10837450.2013.829092 Absorption of lutein SNEDDS in Caco-2 cells 7

with HLB value of 14 and 15, respectively, which render them a improve lutein bioavailability compared to free drug in vivo
good affiance for self-emulsification. Even though, TranscutolÕ studies17.
HP, as a co-surfactant, showed the best solubilizing enhancement
effect for lutein, it provided comparatively less effective
Conclusion
emulsification at the narrowest microemulsion region than the
emulsification by the combined surfactants, LabrasolÕ and In conclusion, the present study has clearly demonstrated the
Tween 80. The combined surfactant might have provided greater potential of SNEDDS as a promising delivery system for lutein
HLB value and hence, the microemulsion region was greatly to improve aqueous solubility and oral bioavailability by lutein
increased in the phase diagram. A similar observation was dissolution and absorption. The obtained lutein-loaded SNEDDS
reported28,29. This result suggested that the selection of surfac- performed rapid and complete self-nanoemulsification within
tant should consider for both their emulsification and solubil- 10 s, and lutein was completely dissolved in dissolution
ization efficiency. In addition, the microemulsion region medium within 5 min. Upon dilution in water, they possessed a
increased as the amount of Tween 80 was increased. This mean droplet size of 40–150 nm with a narrow distribution. The
might be explained by a greater HLB value of Tween 80 than improvement of lutein solubilization could be attributed to the
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by University Library Utrecht on 09/01/13

LabrasolÕ , thus leading to a greater ability to produce water high HLB value of surfactant used in the formulation.
soluble micelle. Additionally, the in vitro transport studies using Caco-2 cells
The droplet size after micro/nano emulsification is one of the demonstrated that SNEDDS showed a shorter lag time and greater
most important property of SNEDDS formulation30. The smaller two-fold cellular accumulation compared to lutein dispersion.
droplet size (30 nm) leads to larger interfacial surface area for Therefore, lutein-loaded SNEDDS shows faster and greater
drug absorption and provides a faster release rate of drug than uptake than lutein dispersion, implying an increase in the oral
larger droplet size (150 nm)21. The mean droplet size prepared bioavailability of lutein. However, these assumptions may further
here was in the range of 40–150 nm with a narrow distribution. be confirmed by animal experimental followed by extensive
The ratio of oil phase to surfactant phase and the ratio of clinical evaluation.
LabrasolÕ to Tween 80 were found to have a major impact on the
mean droplet size. Increasing surfactant phase in SNEDDS Acknowledgements
resulted in a decrease of mean droplet size from 154 to 111 nm. The author would like to thank Associate Professor Srinivas SP for his
This was attributed to a decrease of interfacial energy on the help in the manuscript preparation and discussion of the results.
surface of emulsion droplets12,18. However, when varying the ratio
of LabrasolÕ and Tween 80, a dramatic decrease in the mean Declaration of interest
droplet size, from 139 to 40 nm, was observed with increasing
For personal use only.

amount of Tween 80. Although, LabrasolÕ has a lower molecular This study was supported by Agricultural Research Development Agency
(Public organization) and Faculty of Pharmaceutical Sciences, Naresuan
weight than Tween 80, the latter showed higher HLB value that University.
may facilitate greater ability to reduce interfacial energy, resulting
in a smaller droplet size31. The zeta potential of the micro/nano- References
emulsions of all formulations was in a range of j19j to j32j
mV, which are expected to inhibit aggregation because of charge 1. Prasad PS, Schwartz SD, Hubschman J-P. Age-related macular
repulsion. In addition, lutein-loaded SNEDDS dispersed rapidly degeneration: current and novel therapies. Maturitas 2010;66:46–50.
2. Losso JN, Khachatryan A, Ogawa M, et al. Random centroid
and completely in 0.1 N HCl and pH 6.8 phosphate buffer solution optimization of phosphatidylglycerol stabilized lutein-enriched
within 5 min, suggesting fast emulsification in the gastrointestinal oil-in-water emulsions at acidic pH. Food Chem 2005;92:737–744.
tract after intake of these formulations. The improvement of 3. Beatty S, Koh H-H, Phil M, et al. The role of oxidative stress in the
lutein solubilization could be attributed to the high HLB value pathogenesis of age-related macular degeneration. Surv Ophthalmol
of surfactant. 2000;45:115–134.
Even though SNEDDS appeared to be an effective approach 4. Semba RD, Dagnelie G. Are lutein and zeaxanthin conditionally
essential nutrients for eye health? Med Hypotheses 2003;61:
to increase lutein solubility as confirmed by the dissolution study, 465–472.
the permeation study using Caco-2 cells model showed insignifi- 5. Roberts RL, Green J, Lewis B. Lutein and zeaxanthin in eye and skin
cant permeation profile between lutein dispersion and SNEDDS. health. Clin Dermatol 2009;27:195–201.
This could be attributed to two critical factors. Firstly, the 6. Neelam K, Nolan J, Chakravarthy U, Beatty S. Psychophysical
lipophilicity of lutein, shown by a high octanol/water partition function in age-related maculopathy. Surv Ophthalmol 2009;54:
coefficient value of 14.8, resulted in high affinity of lutein to 167–210.
7. Alves-Rodrigues A, Shao A. The science behind lutein. Toxicol Lett
the cell26. Even though lutein was able to rapidly dissolve
2004;150:57–83.
from SNEDDS and readily absorbed by the cell, it was slowly 8. Seddon JM, Ajani UA, Sperduto RD, et al. Dietary carotenoids,
partitioned to the medium, as evident from a lag time32. The vitamins A, C, and E, and advanced age-related macular degener-
limitation of Caco-2 cells for very lipophilic drug has been ation. Eye Disease Case-Control Study Group. J Am Med Assoc
previously published32,33. Secondly, a limited number of recep- 1994;272:1413–1420.
tors/transporters capable of handling lutein may be responsible. 9. Kohli K, Chopra S, Dhar D, et al. Self-emulsifying drug delivery
The transport process of lutein has been reported as passive systems: an approach to enhance oral bioavailability. Drug Discov
Today 2010;15:958–965.
diffusion and facilitated process by a scavenger receptor class B 10. Neslihan Gursoy R, Benita S. Self-emulsifying drug delivery
type I (SR-BI)26,34,35. Nevertheless, SNEDDS showed a shorter systems (SEDDS) for improved oral delivery of lipophilic drugs.
lag time and a greater two-fold cellular accumulation than lutein Biomed Pharmacother 2004;58:173–182.
dispersion. Considering that, in human, there is a larger surface 11. Shahnaz G, Hartl M, Barthelmes J, et al. Uptake of phenothiazines
area for absorption than in vitro study using Caco-2 cells, the by the harvested chylomicrons ex vivo model: influence of self-
uptake of lutein SNEDDS would be faster and greater than lutein nanoemulsifying formulation design. Eur J Pharm Biopharm 2011;
79:171–180.
dispersion32,36. Therefore, this study showed that the lag time and 12. Zhao Y, Wang C, Chow AHL, et al. Self-nanoemulsifying drug
cellular accumulation could be used as parameters to determine delivery system (SNEDDS) for oral delivery of Zedoary essential
the efficacy of lipophilic drug transport across Caco-2 cells. oil: formulation and bioavailability studies. Int J Pharm 2010;383:
In addition, Shanmugam et al. reported that SNEDDS could 170–177.
8 P. Niamprem et al. Pharm Dev Technol, Early Online: 1–8

13. Basalious EB, Shawky N, Badr-Eldin SM. SNEDDS containing cells by using vesicle-mediated transcytosis. J Control Release 2009;
bioenhancers for improvement of dissolution and oral absorption of 140:174–181.
lacidipine. I: development and optimization. Int J Pharm 2010;391: 25. Raiman J, Tormalehto S, Yritys K, et al. Effects of various
203–211. absorption enhancers on transport of clodronate through Caco-2
14. Wang L, Dong J, Chen J, et al. Design and optimization of a new cells. Int J Pharm 2003;261:129–136.
self-nanoemulsifying drug delivery system. J Colloid Interface Sci 26. Reboul E, Abou L, Mikail C, et al. Lutein transport by Caco-2 TC-7
2009;330:443–448. cells occurs partly by a facilitated process involving the scavenger
15. Singh SK, Verma PRP, Razdan B. Development and characterization receptor class B type I (SR-BI). Biochem J 2005;387:455–461.
of a lovastatin-loaded self-microemulsifying drug delivery system. 27. Singh A, Chaurasiya A, Singh M, et al. Exemestane loaded self-
Pharm Dev Technol 2010;15:469–483. microemulsifying drug delivery system (SMEDDS): development
16. Shanmugam S, Baskaran R, Balakrishnan P, et al. Solid self- and optimization. AAPS J 2008;9:628–634.
nanoemulsifying drug delivery system (S-SNEDDS) containing 28. Borhade V, Pathak S, Sharma S, Patravale V. Clotrimazole
phosphatidylcholine for enhanced bioavailability of highly lipophilic nanoemulsion for malaria chemotherapy. Part I: preformulation
bioactive carotenoid lutein. Eur J Pharm Biopharm 2011;79: studies, formulation design and physicochemical evaluation.
250–257. Int J Pharm 2012;431:138–148.
29. Beg S, Jena SS, Patra CN, et al. Development of solid self-
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by University Library Utrecht on 09/01/13

17. Shanmugam S, Park J-H, Kim KS, et al. Enhanced bioavailability


and retinal accumulation of lutein from self-emulsifying phospho- nanoemulsifying granules (SSNEGs) of ondansetron hydrochloride
lipid suspension (SEPS). Int J Pharm 2011;412:99–105. with enhanced bioavailability potential. Colloids Surf B 2013;101:
18. Yoo J, Shanmugam S, Thapa P, et al. Novel self-nanoemulsifying 414–423.
drug delivery system for enhanced solubility and dissolution of 30. Li X, Yuan Q, Huang Y, et al. Development of silymarin
lutein. Arch Pharmacal Res 2010;33:417–426. self-microemulsifying drug delivery system with enhanced oral
19. Setthacheewakul S, Mahattanadul S, Phadoongsombut N, et al. bioavailability. AAPS J 2010;11:672–678.
Development and evaluation of self-microemulsifying liquid and 31. Ali H, Nazzal M, Zaghloul A-AA, Nazzal S. Comparison
between lipolysis and compendial dissolution as alternative tech-
pellet formulations of curcumin, and absorption studies in rats.
niques for the in vitro characterization of a-tocopherol self-
Eur J Pharm Biopharm 2010;76:475–485.
emulsified drug delivery systems (SEDDS). Int J Pharm 2008;352:
20. Cui J, Yu B, Zhao Y, et al. Enhancement of oral absorption
104–114.
of curcumin by self-microemulsifying drug delivery systems. Int J
32. Artursson P, Palm K, Luthman K. Caco-2 monolayers in experi-
Pharm 2009;371:148–155.
mental and theoretical predictions of drug transport. Adv Drug Deliv
21. Kang BK, Lee JS, Chon SK, et al. Development of self-
Rev 2001;46:27–43.
microemulsifying drug delivery systems (SMEDDS) for oral
33. Fossati L, Dechaume R, Hardillier E, et al. Use of simulated
bioavailability enhancement of simvastatin in beagle dogs. Int J intestinal fluid for Caco-2 permeability assay of lipophilic drugs. Int
Pharm 2004;274:65–73. J Pharm 2008;360:148–155.
22. Yin Y-M, Cui F-D, Mu C-F, et al. Docetaxel microemulsion for 34. Mamatha BS, Baskaran V. Effect of micellar lipids, dietary fiber and
enhanced oral bioavailability: preparation and in vitro and in vivo
For personal use only.

[beta]-carotene on lutein bioavailability in aged rats with lutein


evaluation. J Control Release 2009;140:86–94. deficiency. Nutrition 2011;27:960–966.
23. Souto EB, Wissing SA, Barbosa CM, Muller RH. Development of a 35. Kijlstra A, Tian Y, Kelly ER, Berendschot TTJM. Lutein: more than
controlled release formulation based on SLN and NLC for topical just a filter for blue light. Prog Retin Eye Res 2012;31:303–315.
clotrimazole delivery. Int J Pharm 2004;278:71–77. 36. Shah P, Jogani V, Bagchi T, Misra A. Role of Caco-2 cell
24. Roger E, Lagarce F, Garcion E, Benoit JP. Lipid nanocarriers monolayers in prediction of intestinal drug absorption. Biotechnol
improve paclitaxel transport throughout human intestinal epithelial Prog 2006;22:186–198.

You might also like