You are on page 1of 13

RESEARCH ARTICLE – Pharmaceutical Biotechnology

Quercetin-Containing Self-Nanoemulsifying Drug Delivery System


for Improving Oral Bioavailability
THANH HUYEN TRAN,1 YI GUO,2 DONGHUI SONG,1 RICHARD S. BRUNO,2 XIULING LU1
1
Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269
2
Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, Ohio 43210

Received 14 November 2013; revised 23 December 2013; accepted 3 January 2014


Published online 24 January 2014 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.23858

ABSTRACT: Quercetin is a dietary flavonoid with potential chemoprotective effects, but has low bioavailability because of poor aqueous
solubility and low intestinal absorption. A quercetin-containing self-nanoemulsifying drug delivery system (Q-SNEDDS) was developed to
form oil-in-water nanoemulsions in situ for improving quercetin oral bioavailability. On the basis of the quercetin solubility, emulsifying
ability, and stability after dispersion in an aqueous phase, an optimal SNEDDS consisting of castor oil, Tween 80, Cremophor RH 40, and
R R

PEG 400 (20:16:34:30, w/w) was identified. Upon mixing with water, Q-SNEDDS formed a nanoemulsion having a droplet size of 208.8
± 4.5 nm and zeta potential of −26.3 ± 1.2 mV. The presence of Tween 80 and PEG 400 increased quercetin solubility and maintained
R

supersaturated quercetin concentrations (5 mg/mL) for >1 month. The optimized Q-SNEDDS significantly improved quercetin transport
across a human colon carcinoma (Caco-2) cell monolayer. Fluorescence imaging demonstrated rapid absorption of the Q-SNEDDS within
40 min of oral ingestion. Following oral administration of Q-SNEDDS in rats (15 mg/kg), the area under the concentration curve and
maximum concentration of plasma quercetin after 24 h increased by approximately twofold and threefold compared with the quercetin
control suspension. These data suggest that this Q-SNEDDS formulation can enhance the solubility and oral bioavailability of quercetin
for appropriate clinical application.  C 2014 Wiley Periodicals, Inc. and the American Pharmacists Association J Pharm Sci 103:840–852,

2014
Keywords: oral drug delivery; absorption; pharmacokinetics; bioavailability; solubility; nanoparticle

INTRODUCTION formulations requires hazardous solvents (e.g., chloroform, ace-


tone, and ethanol).6,8 Therefore, a critical need exists to develop
Cancer chemoprevention, using natural or synthetic molecules
alternative administrative vehicles to circumvent existing chal-
to prevent, inhibit, or reverse carcinogenesis, is becoming in-
lenges of current formulations for quercetin.
creasingly common, especially at a time when the use of
Self-nanoemulsifying drug delivery systems (SNEDDS) have
complementary and alternative medicine with natural health
attracted increased attention as a mean to improve the oral
products has become more prevalent.1,2 Quercetin (3,3 ,4 ,5,7-
bioavailability of lipophilic compounds because of their abil-
pentahydroxyflavone) is a dietary flavonoid present in veg-
ity to improve drug solubility, membrane transport, and ab-
etables, fruits, seeds, nuts, tea, and red wine. It exerts an-
sorption via the lymphatic system, thus bypassing the liver
tioxidant activity by upregulating endogenous free radical
and avoiding hepatic xenobiotic metabolism.9,10 SNEDDS are
defenses and suppressing oncogenesis and tumor progres-
anhydrous homogeneous liquid mixtures of oil, surfactant,
sion signaling pathways, and has been studied as a poten-
cosurfactant, and lipophilic drug, which spontaneously form
tial chemoprevention agent.3,4 However, clinical applications
transparent nanoemulsions upon aqueous dilution with gen-
of quercetin for chemoprotection are limited due to its hy-
tle agitation.11,12 Unlike thermodynamically unstable disper-
drophobicity, poor gastrointestinal absorption, and extensive
sion systems, such as emulsions and suspensions, SNEDDS
xenobiotic metabolism at intestines and liver, which collec-
are thermodynamically stable and have a high solubilizing ca-
tively contribute to its low oral bioavailability.5 Various ap-
pacity of lipophilic drugs. They also can be filled directly into
proaches have been used to enhance the solubility, dissolution
gelatin capsules, which improves commercial viability and pa-
rate, and hence, bioavailability of quercetin including solid
tient compliance.13 Several marketed self-emulsifying drug de-
dispersions,5 nanosuspensions,6 microemulsions,7 and solid
livery system-based products, such as Sandimmune , NeoralR R

lipid nanoparticles.8 Despite successful preparations of stable


(cyclosporine), Norvir (ritonavir), and Fortovase (saquinavir),
R R

solid dispersions and nanosuspensions, maximal solubility of


have showed effectiveness of this delivery system.12
a quercetin-containing nanosuspension was limited to only 0.4
Challenges exist to develop SNEDDS formulations that
mg/mL.6 Although solid lipid nanoparticles improve quercetin
maintain the targeted dose in solution during gastrointestinal
bioavailability in rats up to five times,8 these nanoparticles fre-
transit. This is attributed to many poorly soluble compounds
quently have stability issues such that poorly soluble drugs are
having high solubility in SNEDDS, but being precipitated fol-
precipitated during storage. Furthermore, preparation of those
lowing aqueous dispersion of the formulation or during intesti-
nal digestion.14 Persimmon leaf and Hippophae rhamnoides L.
Correspondence to: Xiuling Lu (Telephone: +860-486-0517; Fax: +860-486- extracts containing quercetin have been incorporated into sta-
2076; E-mail: xiuling.lu@uconn.edu)
Thanh Huyen Tran and Yi Guo contributed equally to this work. ble SNEDDS formulations that improved quercetin solubility
Journal of Pharmaceutical Sciences, Vol. 103, 840–852 (2014) and quercetin release in vitro with modest increases in bioavail-

C 2014 Wiley Periodicals, Inc. and the American Pharmacists Association ability in vivo.15 Quercetin-containing SNEDDS (Q-SNEDDS)

840 Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014


RESEARCH ARTICLE – Pharmaceutical Biotechnology 841

have also been reported to improve quercetin bioavailability, and observing the homogenous emulsion appearance without
but this formulation requires a large amount of ethanol (20%), phase separation.
which may limit the clinical application.16 Because a need ex-
ists to improve SNEDDS formulations that enhance quercetin Construction of Pseudo-Ternary Phase Diagrams
bioavailability, the objective of this study was to develop and Oil and surfactant/cosurfactant mixtures that could self-
optimize a SNEDDS formulation that maintains supersatu- emulsify under dilution and gentle agitation were identified
rated quercetin concentrations in nanoemulsions to enhance from pseudo-ternary phase diagrams of systems containing oil
its oral bioavailability. We therefore developed an optimized (20%–70%, w/w), surfactant (30%–80%, w/w), and cosurfactant
Q-SNEDDS formulation and validated its effectiveness in en- (0%–30%, w/w). Three hundred milligrams of surfactant, cosur-
hancing quercetin permeability in cells and bioavailability in factant, and oils with combination one (castor oil/Cremophor R

rats. RH 40/Transcutol HP) and combination two (castor oil/Tween


R R

80/Cremophor RH 40/PEG 400) at various concentrations


R

MATERIALS AND METHODS were mixed at 50◦ C. Efficiency of nanoemulsion formation was
assessed by adding 5 mg of each mixture to 2 mL of distilled
Materials water, followed by gentle agitation using a magnetic stirrer.
The nanoemulsion formation was transparent as assessed by
Quercetin (aglycone), isorhamnetin, tamarixetin, morin, $-
visual inspection and particle size of the resulting dispersions
glucuronidase/sulfatase from Helix pomatia (S9626), Hank’s
was determined by dynamic light scattering (DLS) (Malvern
balanced salt solution (HBSS), Transwell inserts, and lucifer R

Zetasizer, Worcestershire, UK). Dispersions having sizes <200


yellow were purchased from Sigma–Aldrich Chemical Com-
nm were deemed acceptable.
pany (St. Louis, Missouri). Ascorbic acid, diethylene triamine
pentaacetic acid (DTPA), diethyl ether, methanol, nile red, Preparation and Characterization of Q-SNEDDS
sodium carboxymethyl cellulose (CMC-Na), and sodium acetate
were purchased from Fisher Scientific Company (Fair Lawn, The selected SNEDDS formulations from phase-diagrams were
New Jersey). Brij 78, Tween 80, castor oil, and PEG 400
R R
used for solubility testing. SNEDDS formulations having
were received as gifts from Croda (Columbus Circle Edison, the highest solubility of quercetin were used to prepare Q-
New Jersey). Miglyol 812 and Capmul MCM were gener-
R R
SNEDDS. Quercetin (30 mg/g) was added to each formulation
ously donated by Abitect Corporation (Janesville, Wisconsin). followed by gently heating at 50◦ C and mixing using a mag-
Capryol , Labrasol , Labrafil M 1944 CS, Labrafil M 2155
R R R R
netic stirrer until a clear mixture was obtained. Formulations
CS, and Transcutol HP were kindly donated by Gattefosse
R
were examined for turbidity or phase separation before self-
(Saint-Priest Cedex, France). Cremophor RH 40 was provided R
emulsification and particle size analysis. Water was then added
by BASF (Ludwigshafen, Germany). Caco-2 cells, Eagle’s mini- to the Q-SNEDDS with gentle stirring to form a nanoemulsion
mum essential medium, and fetal bovine serum (FBS) were pur- (Q-SNEDDS nanoemulsion, 3 mg/mL). Particle size and zeta-
chased from American Type Culture Collection (Manassas, Vir- potential of the nanoemulsion were measured using DLS in-
ginia). All other chemicals and solvents were of reagent grade strument. The particle size was further confirmed by transmis-
and used without further purification. sion electron microscopy (TEM). One drop of the nanoemulsion
was placed on a formvar coated carbon grid for 3–5 min followed
Solubility Assessment by a negative staining with phosphotungstic acid solution (1%,
An excess quantity of quercetin was added to 1 g of various oils w/v, pH 7.4) for 1 min. The dried sample in the grid was imaged
(castor oil, soybean oil, Miglyol 812, Capryol 90, Capmul C8,
R R R
by TEM (FEI Tecnai Biotwin, Eindhoven, Netherlands).
Capmul MCM, Labrafil 1944, and Labrafil 2155), surfactants
R R R

Determination of Quercetin Encapsulation Efficiency


(Brij 020, Brij 78, Brij 721, Tween 20, Tween 60, Tween 80,
R R R R R R

PEG-35-castor oil, Cremophor RH40, and Solutol HS15), and


R R
Free quercetin was separated from Q-SNEDDS nanoemul-
cosurfactants (PEG 400 and Transcutol HP). Mixtures of R
sions by ultrafiltration using a 3000 Da molecular-weight
quercetin and excipients were first vortexed to facilitate mixing cutoff (MWCO) filter. Samples containing free quercetin and
of quercetin with vehicles, and the mixtures were then allowed Q-SNEDDS nanoemulsion were diluted in methanol before
to reach equilibrium at 40◦ C in a shaking water bath. After high-performance liquid chromatography (HPLC) analysis
48 h, mixtures were centrifuged at 10,000g for 10 min to sepa- as described, with minor modifications,17 using a Ultimate
rate undissolved quercetin, followed by filtration through a 0.45 3000 HPLC-electrochemical system (Thermo Fisher Scien-
:m membrane filter. Filtrates were diluted with methanol and tific, Inc., Sunnyvale, California) equipped with a solvent
the concentration of quercetin was determined using a UV/Vis delivery module (LPG-3400BM), a refrigerated autosampler
spectrometer at 372 nm based on a standard curve of known (WPS-3000) maintained at 4◦ C, and a dual-channel coulo-
quercetin concentrations. metric analytical cell (6011 RS ULTRA). Samples were sep-
arated isocratically at 0.45 mL/min on a Kinetex pentafluo-
Screening of Surfactants for Emulsifying Activity
rophenyl column (150 × 3.0 mm2 i.d., 3 :m; Phenomenex,
Surfactants (100 mg each of Labrasol , Cremophor RH 40, R R
Inc., Torrance, California) using a mobile phase consisting of
Solutol , or Tween 80) were added to 100 mg each of the oil
R R
75 mmol/L citric acid, 25 mmol/L ammonium acetate, and
phase (Capryol 90, Capmul MCM, Labrafil , or castor oil).
R R R
0.27 mmol/L EDTA prepared in 50% methanol (vol/vol). De-
The mixtures were heated at 50◦ C with mild stirring to achieve tection was performed at potential settings of −100 and
homogeneity. Each mixture (50 mg) was weighed and diluted 225 mV and quercetin was quantified at 225 mV. The amount
with 15 mL distilled water to yield a fine emulsion. The for- of quercetin encapsulated in SNEDDS was calculated by sub-
mation of nanoemulsions was monitored by size measurement tracting free quercetin from total quercetin in the Q-SNEDDS

DOI 10.1002/jps.23858 Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014


842 RESEARCH ARTICLE – Pharmaceutical Biotechnology

suspension. Encapsulation efficiency (EE) was calculated as: analysis, and an equivalent volume of HBSS was added to the
EE (%) = W × 100%/Wtotal , where W = quantity of quercetin apical side. Media were directly injected onto the aforemen-
loaded into Q-SNEDDS and Wtotal = total quantity of quercetin tioned HPLC-electrochemical system (Thermo Fisher Scien-
in the Q-SNEDDS suspension. tific, Inc.) to determine quercetin concentration as described
above. Integrity of the Caco-2 cell monolayer treated with
In Vitro Quercetin Release Study free quercetin or Q-SNEDDS nanoemulsion was evaluated by
In vitro release of quercetin from the nanoemulsions was per- adding lucifer yellow (100 :g/mL) to the donor chamber at the
formed using a dialysis technique. Briefly, Q-SNEDDS was beginning of the transport study. At the end of the study, sam-
diluted with simulated gastric fluid (pH 1.2) and simulated ples (200 :L) from the receiver chambers were transferred to
intestinal fluid (pH 6.8) (United States Pharmacopeia 23 a 96-well plate. The concentration of Lucifer yellow was mea-
standard) to form Q-SNEDDS nanoemulsions at 3 mg/mL of sured using a microplate reader (Ex /Em , 480 nm/530 nm; Tecan
quercetin. The quercetin nanoemulsions (3 mL) were intro- group Ltd., Männedorf, Switzerland).
duced into 5 mL-dialyzers (MWCO: 10,000 Da) and immersed
in 25 mL of simulated gastric and intestinal fluids contain- Visualization of Intestinal Q-SNEDDS Absorption in Rats
ing 0.5% (w/v) Tween 80 at 37◦ C in a shaking bath at 100
rpm. At selected time intervals, aliquots (1 mL) were removed The protocol for all studies in rats was approved by Institu-
from the dissolution medium and an equivalent volume of fresh tional Animal Care and Use Committee at the University of
medium was added. The concentration of quercetin was deter- Connecticut. Visualization of intestinal quercetin absorption
mined spectrophotometrically at a wavelength of 372 nm. The delivered in rats was accomplished using the hydrophobic fluo-
amount of quercetin released was calculated by comparing with rescent dye nile red, which was encapsulated into Q-SNEDDS
standards of known quercetin concentrations. as described above. Free nile red (20 mg/mL) suspended in
0.5% (w/v) CMC-Na and Q-SNEDDS nanoemulsion contain-
Stability of the Q-SNEDDS ing nile red (20 :g/mL) was administered by oral gavage to
male Sprague–Dawley rats (300 ± 10 g; Charles River, Wilm-
To determine the extent to which quercetin precipitated dur- ington, Massachusetts) at 15 mg/kg BW of quercetin and then
ing dispersion and storage, Q-SNEDDS stability was assessed sacrificed after 40 min. Following a midline incision, the gas-
by storing Q-SNEDDS and Q-SNEDDS nanoemulsions at a trointestinal tract was removed; individual segments (duode-
quercetin concentration of 5 mg/mL in amber vials at room num, jejunum, and ileum) were separated, and subsequently
temperature. Size changes, drug precipitation, and phase sep- placed in cold phosphate-buffered saline (PBS; pH 7.4). Isolated
aration were evaluated at predetermined time point. For quan- gastrointestinal segments were dissected along the mesenteric
tification of quercetin content, Q-SNEDDS and Q-SNEDDS border and rinsed with cold PBS to remove luminal contents. A
nanoemulsions were sampled at predetermined time points 0.5 cm piece of each segment was fixed overnight in 4% neutral
and centrifuged at 10,000g for 5 min to remove any precipi- buffered formalin. The gastrointestinal segments were washed
tated quercetin. An aliquot of the supernatant was dissolved in with PBS and frozen in cryoembedding media (OCT) for subse-
methanol, followed by the dilution to an appropriate quercetin quent cryostat sectioning at 20 :m per slice (CM3050S; Leica,
concentration before assay by UV/Vis spectrophotometry at Buffalo Grove, United State). The sections were applied to glass
372 nm. slides, rinsed with cold PBS, and visualized under an inverted
fluorescence microscope at 10× magnification (Olympus, Tokyo,
Transport of Q-SNEDDS in Caco-2 Cells
Japan).
Caco-2 cells were grown in MEM supplemented with 1%
nonessential amino acids, 1% sodium pyruvate, 1% antibiotics,
Bioavailability of Quercetin in Rats
and 10% FBS at 37◦ C in an atmosphere of 5% CO2 . When
cells were confluent, they were trypsinized and seeded onto Male Sprague–Dawley rats (300 ± 10 g) having a jugular vein
Transwell polycarbonate inserts (12-well plate) at a density
R
cannulation were purchased (Charles River). For 1 week before
of 100,000 cells/well. Medium in the Transwell plate was R
the experimentation, rats had free access to water and a puri-
changed every 2 days and cells were allowed to differentiate fied AIN-93G diet. Rats (n = 5–6/group) were fasted overnight
for 21–28 days. to assess quercetin bioavailability in response to oral adminis-
Before the experiment, cells were washed three times with tration of Q-SNEDDS and quercetin control suspension. Food
prewarmed transport medium (HBSS buffered with 0.35 g/L was provided after 4 h of the study. Control suspension of
NaHCO3 and 25 mM HEPES). Transwells containing trans-
R
quercetin was prepared by suspending quercetin in 0.5% (w/v)
port medium (0.4 mL in the apical side and 1.2 mL in the CMC-Na, followed by sonication for 15 min. The quercetin con-
basolateral side) were then incubated at 37◦ C for 15 min while trol suspension or Q-SNEDDS nanoemulsion were provided by
shaking in an orbital shaking bath. Quercetin stock solution oral gavage (3 mg/mL; 15 mg/kg BW) at a dose consistent with
was prepared by dissolving quercetin in DMSO (3 mg/mL). our clinical bioavailability studies where participants ingested
Both the stock solution and Q-SNEDDS nanoemulsion were 1095 mg quercetin (∼15 mg/kg BW).17 Blood (0.4 mL) was col-
diluted using HBSS to achieve the same quercetin concentra- lected into tubes containing sodium heparin before (0 h) and 1,
tion of 50 :g/mL and filtered using a 0.2 :m sterile filter mem- 2, 4, 6, 8, 12, and 24 h after administration, and then plasma
brane immediately before initiating transport studies. Media was separated by centrifugation (1200g, 10 min, 4◦ C) and stored
from the apical side were removed and quercetin solution or at −80 ◦ C. Liver and small intestine were collected at 24 h. Be-
Q-SNEDDS nanoemulsion (0.4 mL) was subsequently added to fore freezing all tissues in liquid nitrogen and storing at −80◦ C,
the apical side. At 0, 15, 30, 45, and 60 min, samples (0.5 mL) intestinal contents were flushed with ice-cold PBS (pH 7.4) con-
were collected from the receiver chamber, frozen at −80◦ C until taining 2 g/L of albumin and 16.5 mmol/L sodium taurocholate.

Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014 DOI 10.1002/jps.23858


RESEARCH ARTICLE – Pharmaceutical Biotechnology 843

Figure 1. Solubility of quercetin in various oils, surfactants, and cosurfactants.

Plasma throughout the 24 h pharmacokinetics study and tion rate constant (ke ) were calculated using PK Solutions R

tissues collected at 24 h were analyzed for quercetin and its software (Summit Research Services, Inc., Montrose, Colorado).
methylated derivatives isorhamnetin (3 -O-methyl-quercetin) Absorption or appearance rate constants were not calculated
and tamarixetin (4 -O-methyl-quercetin) using the aforemen- because of insufficient time points before Tmax . Data (means
tioned HPLC-electrochemical system. Because of the limited ± SEM) were analyzed using GraphPad Prism 5.0 (GraphPad
plasma availability at each time point, plasma quercetin anal- Software, Inc., La Jolla, California) and were initially evaluated
ysis was limited to total quercetin (i.e., the sum of free quercetin for normality using the Kolmogorov–Smimov test. Treatment
and its glucuronidated and sulfates conjugates). For analy- differences were analyzed using a Student’s unpaired t-test for
sis, 100 :L of plasma was mixed with 10 :L of 27.6 :mol/L data having a normal distribution and a Mann–Whitney U-
morin (internal standard) dissolved in methanol, 977 U $- test was used for data lacking a normal distribution. All anal-
glucuronidase and 33 U of sulfatase prepared in 1 mol/L sodium yses were considered statistically significant at an "-level of
acetate buffer (pH 5.5), and 20 :L of 6% ascorbic acid (w/v) pre- p < 0.05.
pared in 1 mol/L sodium acetate buffer containing 2.5 mmol/L
DTPA. Samples were incubated (37◦ C, 2 h), extracted with di-
ethyl ether, and then dried under nitrogen gas before being RESULTS AND DISCUSSION
reconstituted in methanol and injected onto the HPLC. For the Selection of Components for SNEDDS
analysis of liver and small intestine, tissue (∼1 g) was pulver-
ized in liquid nitrogen and then mixed with 2 mL of 1 mol/L The selection of oil, surfactant, and cosurfactant are critical for
sodium acetate buffer (pH 5.5) containing 28 mmol/L ascorbic improving solubility and achieving high payload in a SNEDDS
acid. Following centrifugation (600g, 15 min, 4◦ C), the super- formulation.18,19 Components of SNEDDS were selected to max-
natant was collected and two separate aliquots (600 :L) were imize quercetin solubility while attaining high miscibility with
mixed with 60 :L of 4.96 :mol/L morin and 1 mL of sodium ac- each component to form a stable nanoemulsion spontaneously
etate buffer containing 28 mmol/L ascorbic acid. Samples were following dilution with water. The solubility of quercetin in
incubated (37◦ C, 2 h) in the presence or absence of 977 U $- different oils, surfactants, and cosurfactants was determined
glucuronidase and 33 U of sulfatase to determine free and total (Fig. 1). Quercetin showed higher solubility in Capryol , R

(i.e., the sum of hydrolyzed conjugates and free) flavonoids, and Capmul MCM, and castor oil compared with the solubility in
R

extracted with diethyl ether, dried under nitrogen gas and re- soybean oil, miglyol 812, and labrafil and these oils were there-
constituted in methanol before HPLC analysis. fore investigated further. Quercetin also exhibited good solubil-
ity in each of the surfactants tested with Labrasol (32.0 ± 1.3
R

mg/g), Cremophor RH 40 (24.5 ± 0.9 mg/g), and Tween 80


R R

Statistical Analysis
(17.9 ± 0.5 mg/g). The selection of surfactant was governed by
Plasma pharmacokinetics of quercetin, isorhamnetin, and their emulsification efficiency rather than their ability to solu-
tamarixetin, including area under the concentration curve bilize quercetin, consistent with the approach of others.9,20 Both
(AUC0–24 h ), maximum concentration (Cmax ), time to maximum Transcutol HP and PEG 400 had great solubilizing capacity
R

concentration (tmax ), elimination half-life (t1/2 ), and elimina- of quercetin, showing values of 73.8 ± 2.2 and 51.8 ± 0.5 mg/g,

DOI 10.1002/jps.23858 Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014


844 RESEARCH ARTICLE – Pharmaceutical Biotechnology

respectively. Thus, Transcutol HP and PEG 400 were chosen


R
oxy 40 hydrogenated castor oil) and castor oil, which could help
as a cosurfactants to improve loading capacity of quercetin. Cremophor RH 40 incorporate the oil droplet uniformly.23 In
R

Various combinations of oils and surfactants were examined addition, Transcutol HP contains a diethylene glycol struc-
R

to determine their ability to form emulsions. Labrasol emulsi- R


ture that likely associates with polyethylene glycol structure
fied the selected oils poorly as evidenced by the appearance of contained in Cremophor RH 40, which would be expected to
R

many oil droplets following dispersion of the Labrasol mixture R


further stabilize the nanoemulsions. Table 1 shows the average
in aqueous solution. Cremophor RH 40 showed the best emul-
R
size and appearance of nanoemulsions in the phase diagram.
sification efficiency, forming transparent solutions with oils af- Particle size and turbidity of the nanoemulsions increased with
ter adding water. Although Tween 80 was able to emulsify
R
increasing oil content. Formulations having ≤50% surfactant
oils, it occurred with lower efficiency than Cremophor RH R
with transparent and bluish nanoemulsions and sizes ≤200
40. The three selected surfactants have HLB values ranged nm were selected for quercetin encapsulation.
from 13 to 16, thus, the differences observed in their emulsify- Unexpectedly, quercetin (3%, w/w) precipitated within 30
ing ability may be attributed to the head group structure and min after each of the selected formulations was diluted with
chain length.11,21 Cremophor RH 40 and Tween 80, which
R R
distilled water regardless of the oil, surfactant, and cosurfac-
yielded clear nanoemulsions, were selected for further study. tant concentrations. However, when PEG 400 was substituted
The systems containing Capryol or Capmul MCM were un-
R R
by Transcutol HP, precipitation of quercetin was inhibited.
R

able to form stable nanoemulsions containing quercetin. Upon Inhibiting quercetin precipitation by PEG 400 might be at-
contact with aqueous media, these systems precipitated imme- tributed to suppression in compound nucleation, the first step in
diately. Although quercetin had lower solubility in castor oil crystallization of compounds intended for nanoemulsification.24
compared with the medium chain length lipids (6–12 carbons), When Tween 80 was combined with Cremophor RH 40
R R

precipitation of quercetin did not occur in formulations con- and Transcutol HP for preparation of Q-SNEDDS, stable na-
R

taining castor oil, suggesting that lipid chain length regulates noemulsions were generated. A combination of Tween 80 withR

nanoemulsion stability. Long chain fatty acid lipids (13–21 car- Cremophor EL has been also used to form stable nanoemulsions
bons) such as castor oil have also been reported to form stable with Sefsol 218 oil and Oleanolic acid as Tween 80 could not
R

nanoemulsions without precipitation from SNEDDS contain- generate stable nanoemulsions when used alone.25 Therefore,
ing indomethacin.19 Therefore, castor oil was deemed as the a system composed of castor oil, Cremophor RH 40:Tween
R R

best oil for Q-SNEDDSpreparation. 80, and PEG 400 was used to construct a new phase diagram
in which the ratio of Tween 80 and Cremophor RH 40 was
R R

Construction of Pseudo-Ternary Phase Diagram and Formulation 2:1. This ratio was chosen based on the ability of the surfac-
Optimization tant mixture to solubilize quercetin. Even though quercetin
had higher solubility in Cremophor RH 40 than Tween 80,
R R

Ternary phase behavior investigations aid in the selection of the


the presence of larger amount of Tween 80 than Cremophor
R R

optimal concentration of oil, surfactant, and cosurfactant in for-


RH 40 in the surfactant mixture resulted in higher solubility of
mulations to produce stable nanoemulsions. Formulations (n =
quercetin (data not shown). The mechanism for this enhanced
18) composed of castor oil/Cremophor RH 40/Transcutol HP
R R

quercetin solubility is not completely clear. The combination of


were prepared using the selected Smix (surfactant:cosurfactant)
Cremophor RH 40 and Tween 80 could emulsify 40% (w/w)
R R

as well as castor oil in the percentage range of 20%–70% (w/w).


of castor oil (Fig. 2b).
Their self-emulsifying properties were evaluated. Figures 2a
The droplet size of the nanoemulsions increased with in-
and 2b show the phase diagrams in which the shaded re-
creasing oil concentration and decreasing surfactant content
gion indicates the nanoemulsion region. The relatively larger
(Table 2). Formulations 1–4 containing 20% (w/w) castor oil
shaded region indicates better self-nanoemulsifying ability.20,22
formed clear bluish nanoemulsions with droplet sizes less than
The phase diagram (Fig. 2a) showed a large nanoemulsifica-
200 nm and were selected for further optimization. Increasing
tion region up to 60% (w/w) of castor oil. This may be due to
the proportion of PEG 400 resulted in an increase in quercetin
the same carbon chain length of both Cremophor RH 40 (poly- R

Figure 2. Pseudo-ternary phase diagrams of formulations composed of castor oil/CremophorR RH 40/TranscutolR HP (a), and castor oil/TweenR
80/CremophorR RH 40/PEG 400 (b).

Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014 DOI 10.1002/jps.23858


RESEARCH ARTICLE – Pharmaceutical Biotechnology 845

Table 1. Formulations, Droplet Size, and Appearance of Nanoemulsions Composed of Castor Oil/CremophorR RH 40/TranscutolR HP

Castor Oil CremophorR TranscutolR Average Droplet Appearance of


Formulation Oil:Smix (%) RH 40 (%) HP (%) Size (nm) Nanoemulsion

1 2:8 20 80 0 28.24 Transparent


2 70 10 26.07 Transparent
3 60 20 27.82 Transparent
4 50 30 40.21 Transparent
5 3:7 30 70 0 35.31 Transparent
6 60 10 38.78 Transparent
7 50 20 75.53 Transparent
8 40 30 197.7 Clear, bluish
9 4:6 40 60 0 57.16 Transparent
10 50 10 83.06 Clear, bluish
11 40 20 209.6 Transparent
12 30 30 248.6 Milky-like
13 5:5 50 50 0 105.7 Clear, bluish
14 40 10 253.6 Translucent
15 30 20 ND Cannot form
16 6:4 60 40 0 188.1 Translucent
17 30 10 ND Cannot form
18 7:3 70 30 0 ND Cannot form

Table 2. Formulations, Droplet Size, and Appearance of Nanoemulsions Composed of Castor Oil/TweenR 80/CremophorR RH 40/PEG 400

Castor Oil TweenR 80: CremophorR Average Droplet Appearance of


Formulation Oil:Smix (%) RH 40 (2:1) (%) PEG 400 (%) Size (nm) Nanoemulsion

1 2:8 20 80 0 159.5 Clear, bluish


2 70 10 157.1 Clear, bluish
3 60 20 188.4 Clear, bluish
4 50 30 200.8 Clear, bluish
5 3:7 30 70 0 266.3 Translucent
6 60 10 181.6 Translucent
7 50 20 245.6 Milky-like
8 40 30 261.6 Milky-like
9 4:6 40 60 0 251.5 Milky-like
10 50 10 273.8 Milky-like
11 40 20 ND Cannot form
12 30 30 ND Cannot form
13 5:5 50 50 0 ND Cannot form
14 40 10 ND Cannot form
15 30 20 ND Cannot form
16 6:4 60 40 0 ND Cannot form
17 30 10 ND Cannot form
18 7:3 70 30 0 ND Cannot form

solubility in SNEDDS. Among these four formulations, those SNEDDS (containing no quercetin) resulted in nanoemulsions
containing 20% oil, 50% surfactant, and 30% cosurfactant had of 200 ± 10 nm. When quercetin was incorporated into the
the greatest solubilizing ability for quercetin (49.2 ± 1.2 mg/g). SNEDDS, the size was slightly increased to 215 ± 8 nm
Furthermore, no phase separation or precipitation of quercetin (Fig. 3). TEM images of optimal blank SNEDDS and Q-
from the nanoemulsions was observed upon 72 h storage at am- SNEDDS 72 h post dilution in distilled water are shown in
bient temperature. Therefore, this formulation was selected as Figure 4. Spherical nanoemulsions were formed without change
the optimal SNEDDS formulation for quercetin. in morphology after quercetin incorporation. Furthermore, no
sign of quercetin precipitation was observed in the TEM im-
age inferring stability of the formed nanoemulsions. Quercetin-
Characterization of the Optimized Q-SNEDDS
loaded nanoemulsions were negatively charged at their sur-
The optimal Q-SNEDDS spontaneously dispersed in aqueous face as reflected by their zeta potential of −26.3 ± 1.2 mV,
media to form nanoemulsions at high EE of quercetin (≥90%). likely due to the presence of castor oil. Zeta potential is an
The aqueous solubility of quercetin increased from 5 :g/mL indicator of the stability of the nanoemulsion. A higher elec-
to 5 mg/mL after incorporation into SNEDDS. Nanoemulsion trical charge (zeta potential > ±30 mV) on the surface of the
droplet size is an important factor in formulating SNEDDS be- nanodroplets prevents aggregation due to the strong repellent
cause it determines dissolution rate and absorption extent of forces among droplets.27 Thus, the optimized formulation with
compound.26 Size distribution and morphology of the result- zeta potential value close to −30 mV is expected to be stable in
ing nanoemulsions were evaluated by DLS and TEM. Blank solution.

DOI 10.1002/jps.23858 Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014


846 RESEARCH ARTICLE – Pharmaceutical Biotechnology

Figure 3. Particle size distribution of optimal nanoemulsions without quercetin (a), and optimal quercetin-loaded nanoemulsions (b).

SNEDDS nanoemulsions exhibited slow quercetin release Stability of Q-SNEDDS


at both pH 1.2 and pH 6.8 with about 8% release within 24 h
To evaluate storage stability of Q-SNEDDS, we monitored
(Fig. 5). Quercetin release from the nanoemulsions at pH 1.2
quercetin content and phase separation of the Q-SNEDDS as
and pH 6.8 was not significantly different in the first 8 h
well as changes in particle size and quercetin content of Q-
(p > 0.05). Quercetin-loaded nanoemulsions in both media
SNEDDS after dilution with water upon storage at ambient
showed no sign of precipitation, cloudiness, or separation
temperature. After 1 month, phase separation with settling
within 24 h. The results indicated that SNEDDS formulation
of the quercetin-containing layer on the bottom was observed
improved quercetin solubility but retained quercetin within the
in the system composed of quercetin-castor oil/ Cremophor R

nanoemulsions for an extended period of time.


RH 40/Transcutol HP. In contrast, there was no change in
R

Figure 4. TEM images of optimal nanoemulsions without quercetin (a), and optimal Q-SNEDDS nanoemulsions at 72 h post-dilution of
Q-SNEDDS (b).

Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014 DOI 10.1002/jps.23858


RESEARCH ARTICLE – Pharmaceutical Biotechnology 847

self-emulsifying nanoemulsion while the integrity value was


about 90% at 15 min and 95% at 2 h time point.31 The test
is time dependent and may only reflect some changes of the
integrity of cell monolayer as the tight junction opening is a
reversible process. Tight junction opening after being treated
with self-emulsifying systems and certain surfactants has been
observed,32 which presents a possible mechanism for the en-
hanced absorption in terms of paracellular transport. During
the transport study, quercetin control suspension precipitated
from solution, which attenuated its transport compared with
quercetin solubilized in Q-SNEDDS. It is also likely that the
presence of Cremophor RH 40 and Tween 80 in Q-SNEDDS
R R

increases the permeability of quercetin by facilitating transcel-


lular absorption.22,33 The enhanced quercetin transport across
the intestinal cell monolayer suggests the potential of bioavail-
ability enhancement from the Q-SNEDDS.

Histological Evaluation
To visualize quercetin absorption in rat intestines in response
to Q-SNEDDS, the hydrophobic fluorescent dye (nile red) was
Figure 5. Release profiles of Q-SNEDDS nanoemulsion in simulated incorporated into Q-SNEDDS and quercetin suspended in
gastric fluid (pH 1.2) and simulated intestinal fluid (pH 6.8) containing CMC-Na. The fluorescent signal may indicate quercetin due
0.5% (w/v) Tween 80 at 100 rpm and 37◦ C.
to its similar physicochemical properties to nile red. Average
particle size and zeta potential of the Q-SNEDDS containing
quercetin content or phase separation from the optimal Q- nile red were unchanged compared with the formulation with-
SNEDDS formulation (inset of Fig. 6a). While there were out the dye (data not shown). Figure 8 shows fluorescence im-
insignificant changes in particle size and quercetin content ages of different segments of rat intestine 40 min after oral
of the optimal Q-SNEDDS nanoemulsion, nanoemulsions of administration of quercetin control suspension (control) and
quercetin-castor oil/Cremophor RH 40/Transcutol HP pre-
R R Q-SNEDDS, and 1 and 2 h post-administration of Q-SNEDDS
cipitated with a dramatic decrease in quercetin content and containing nile red. After 40 min, the red fluorescent signal
increase in particle size within 10 days (Fig. 6). These find- originating from nile red was minimally visible in intestinal
ings indicate that the inclusion of PEG 400 and Tween 80 in R segments of rats treated with the control, whereas a strong
the optimal Q-SNEDDS formulation prevented quercetin pre- red signal appeared in all intestinal segments of rats treated
cipitation and maintained it at supersaturated levels for an with the Q-SNEDDS. The dim signal in controls was likely be-
extended period of time. cause of the existence of nile red in its insoluble form which was
washed out at the end of the study, whereas the solubilized form
Transport of the Q-SNEDDS Across Caco-2 Cell Monolayer of the dye in the nanoemulsions emitted a strong fluorescence
signal. Moreover, stronger red signals were visible outside than
Human Caco-2 cell monolayers are frequently utilized as a
inside the intestine villi, which may be attributed to the fact
model of intestinal absorption.28 Changes in intestinal perme-
that the Q-SNEDDS remained intact and became attached to
ability of quercetin by the SNEDDS formulation were assessed
the intestine villi. The red signal of the Q-SNEDDS outside the
by measuring quercetin transport across epithelial cell layers.
intestine villi decreased and no red signal was visible inside
Quercetin concentration in both DMSO/HBSS solution (control
the villi after 1 and 2 h, indicating that the dye/quercetin were
solution) and SNEDDS was kept the same (50 :g/mL) to elimi-
absorbed rapidly from the intestinal lumen. The intensity of
nate the concentration effect on quercetin transport. Quercetin
the red signal was not significantly different in duodenum, je-
is known to be unstable after exposure to cell culture con-
junum, and ileum, suggesting that these three sites of the rat
ditions longer than 60 min.29 Thus, the transport study was
intestine were all important for Q-SNEDDS absorption. This
performed at 37◦ C for 60 min. Quercetin transport across the
result is supported by prior studies showing that the absorption
Caco-2 cell monolayer increased time-dependently (Fig. 7). At
in duodenum, jejunum, and ileum of microemulsions contain-
45 and 60 min, the transport amount of quercetin adminis-
ing quercetin was not significantly different and ranged from
tered as Q-SNEDDS was significantly greater than that from
34% to 41%.8
quercetin control solution (p < 0.05). By 60 min, the cumulative
amount of transported quercetin delivered from Q-SNEDDS
Pharmacokinetics of Quercetin in Rats
was approximately two times greater than that of quercetin
control solution (159.7 ng/mL for the Q-SNEDDS vs. 85.1 ng/mL Plasma pharmacokinetics of quercetin was determined to ex-
for quercetin control solution). Permeability of lucifer yellow amine the extent to which Q-SNEDDS increased quercetin
though the Caco-2 cell monolayer was utilized as an indicator bioavailability. Plasma quercetin concentrations increased fol-
of tight junction integrity.30 Transport of lucifer yellow after 1 h lowing oral administration of either formulation and de-
was 1.8 ± 0.2% in all samples tested, indicating good integrity creased to concentrations no different from baseline by 24 h
of the Caco-2 cell monolayer at the end of the study. However, regardless of formulation administered (Fig. 9a). However,
it was reported that the width of Caco-2 cell tight junction plasma quercetin Cmax increased by three times and AUC0–24 h
increased largely shown on TEM images after exposure to a increased by two times when quercetin delivered from

DOI 10.1002/jps.23858 Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014


848 RESEARCH ARTICLE – Pharmaceutical Biotechnology

Figure 6. Stability of Q-SNEDDS nanoemulsions from two systems (castor oil/CremophorR RH 40/TranscutolR HP and castor oil/TweenR
80/CremophorR RH 40/PEG 400) stored at room temperature for 1 month (a) average particle size, and (b) quercetin content remained in the
nanoemulsions.

Q-SNEDDS was administered compared with quercetin control which was in agreement with two times greater AUC0–24 h from
suspension (Table 3), and these observations occurred without Q-SNEDDS (Table 3). The Tmax of isorhamnetin and tamarix-
any differences in Tmax or ke between treatments. Collectively, etin was 2.3–3.4 h earlier following Q-SNEDDS administra-
Q-SNEDDS increased quercetin Cmax and AUC0–24 h without tion compared with quercetin control suspension, but this oc-
affecting its elimination kinetics, suggesting that Q-SNEDDS curred without any statistical significance. In addition, no other
improved quercetin bioavailability by enhancing its absorption. pharmacokinetic parameters differed between groups (Table 3).
Similar to quercetin, plasma isorhamnetin and tamarix- These findings indicate that the extent and rate of methylation
etin (methylated metabolites of quercetin) increased after in- of quercetin increased in response to SNEDDS administration.
gestion of either formulation and returned to baseline lev- Isorhamnetin and tamarixetin are positional methy-
els by 24 h (Figs. 9b and 9c). The Cmax of isorhamnetin and lated isomers generated from quercetin in a catechol-O-
tamarixetin increased by three times following Q-SNEDDS methyltransferase (COMT)-dependent manner.34 Tamarixetin
administration compared with quercetin control suspension, has been reported to show greater potency in inhibiting

Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014 DOI 10.1002/jps.23858


RESEARCH ARTICLE – Pharmaceutical Biotechnology 849

Figure 7. The mean cumulative transport versus time of quercetin


solution and Q-SNEDDS nanoemulsions across Caco-2 cell monolayer
at 37◦ C for 1h using initial quercetin concentration of 50 :g/mL. *Sig-
nificantly different from quercetin solution.

platelet aggregation in vitro than quercetin itself.35 There-


fore, Q-SNEDDS increases tamarixetin to a greater extent
than quercetin control suspension, which may have impor-
tant benefits to human health. Despite the similar mecha-
nism of generation of tamarixetin and isorhamnetin, they ex- Figure 8. Fluorescence images of rat intestine treated with Q-
hibited distinct pharmacokinetic responses. Regardless of the SNEDDS nanoemulsions and quercetin control suspension (control)
quercetin formulation administered, plasma pharmacokinetics containing nile red.
of isorhamnetin was biphasic (Fig. 9c), suggesting the possi-
bility of enterohepatic recirculation of quercetin and isorham-
netin, a common route of disposition of quercetin and its methy- in response to Q-SNEDDS might extend their bioactivity, but
lated metabolites.36 Isorhamnetin Cmax was also greater than clearly such work was beyond the scope of this study. Likewise,
that of tamarixetin regardless of the quercetin formulation ad- additional work is needed to assess time-dependent differences
ministered (p < 0.05), which is in agreement with earlier stud- in tissue accumulation of quercetin.
ies showing that isorhamnetin is the predominant methylated In contrast to quercetin, isorhamnetin was detected only as
metabolite of quercetin in rats fed a quercetin-containing diet.37 its conjugated metabolites in small intestine regardless of for-
Consistent with our clinical findings from a quercetin phar- mulation administered, whereas total isorhamnetin increased
macokinetics study,17 plasma isorhamnetin was eliminated by 1.6 times in small intestine in response to Q-SNEDDS ad-
slower (p < 0.05) and its half-life was longer (6.0 ± 1.8 h; ministration. In liver, Q-SNEDDS increased total isorhamnetin
p < 0.05) than that of tamarixetin (Table 3). Thus, isorham- and free isorhamnetin accumulation by three and six times,
netin pharmacokinetics differed from tamarixetin, suggest- respectively, compared with quercetin control suspension. Con-
ing that the positional methylation of quercetin regulates its sistent with plasma pharmacokinetics indicating faster tamar-
disposition. ixetin elimination, tamarixetin was not detectable in small in-
Accumulation of quercetin occurs in the intestines and testine or liver regardless of formulationadministered.
liver at 24 h following an acute, single-dose administration Collectively, these findings demonstrate that Q-SNEDDS
of quercetin-4 -glucoside in rats.38 In the present study, accu- increases quercetin absorption, thereby enhancing its bioavail-
mulation of quercetin was evaluated in small intestines and ability. Additional studies are needed to identify the mech-
liver at 24 h when the pharmacokinetic study was terminated. anisms for the enhanced bioavailability of quercetin by Q-
Small intestinal accumulation of quercetin did not differ be- SNEDDS. According to the available reports, we speculate
tween treatments, whereas concentrations of total isorham- that it may be associated with the following factors. Poor ab-
netin were greater in response to Q-SNEDDS administra- sorption of quercetin is partly attributed to its limited water
tion (Table 4). In contrast, quercetin and isorhamnetin at solubility.39 The optimal SNEDDS formulation used in this
liver, whether free or total quercetin or isorhamnetin, accu- study increases the apparent water solubility of quercetin and
mulated to a greater extent in response to Q-SNEDDS, indicat- maintains quercetin at supersaturated concentrations in gas-
ing that the SNEDDS formulation improved quercetin absorp- trointestinal tract, thereby the large surface area provided by
tion. Greater persistence of free quercetin at liver compared the fine droplets of the nanoemulsion increases dissolution,
with small intestines likely reflects tissue-specific differences in which is one of the rate limiting factors for quercetin absorption.
xenobiotic metabolism. Our findings also suggest that the The presence of surfactants in the SNEDDS formulation may
greater accumulation of quercetin and isorhamnetin at liver increase intestinal epithelial permeability by disturbing the

DOI 10.1002/jps.23858 Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014


850 RESEARCH ARTICLE – Pharmaceutical Biotechnology

Figure 9. Plasma concentration–time profiles of quercetin (a), tamarixetin (b), and isorhamnetin (c) in Spraque–Dawley rats followed by an
oral administration of 15 mg/kg quercetin control suspension or 15mg/kg Q-SNEDDS nanoemulsion.

Table 3. Pharmacokinetic Parameters of Quercetin in Rats after Oral Administration of Quercetin Control Suspension and Q-SNEDDS
(15mg/kg)*

Quercetin Tamarixetin Isorhamnetin

Quercetin Control Quercetin Control Quercetin Control


Suspension Q-SNEDDS Suspension Q-SNEDDS Suspension Q-SNEDDS

Cmax (mg/L) 1.20 ± 0.17 3.75 ± 0.96** 0.04 ± 0.01 0.12 ± 0.01** 0.11 ± 0.03 0.35 ± 0.07**
Tmax (h) 1.8 ± 0.6 1.2 ± 0.2 3.8 ± 1.2 1.5 ± 0.2 5.4 ± 2.0 2.0 ± 0.4
ke (h−1 ) 0.19 ± 0.05 0.15 ± 0.04 0.21 ± 0.05 0.28 ± 0.01 0.08 ± 0.01 0.09 ± 0.02
t1/2 (h) 5.1 ± 1.6 7.4 ± 2.4 4.5 ± 1.4 2.5 ± 0.1 10.4 ± 1.9 8.6 ± 1.1
AUC0–24h (mg/Lh) 6.7 ± 1.4 14.0 ± 2.8** 0.3 ± 0.1 0.6 ± 0.1** 1.4 ± 0.5 3.2 ± 0.7**

*Data are means ± SEM (n=5–6 rats/group).


**Significantly different from quercetin control suspension, p<0.05.
Abbreviations: Cmax , plasma maximum concentration; Tmax , time to maximum concentration; ke , elimination rate constant; t1/2 , elimination half-life; AUC0–24h ,
area under the concentration curve.

Table 4. Tissue Concentrations of Quercetin Metabolites after Oral Administration of Quercetin Control Suspension and Q-SNEDDS to Rats
at Doses of 15 mg/kg*

Small Intestine Liver

Quercetin Control Suspension Q-SNEDDS Quercetin Control Suspension Q-SNEDDS

Free quercetin (ng/g tissue) 0.37 ± 0.14 0.36 ± 0.11 0.49 ± 0.19 2.35 ± 0.68**
Total quercetin (ng/g tissue) 4.04 ± 1.06 5.46 ± 0.52 2.01 ± 0.29 6.29 ± 1.10**
Free isorhamnetin (ng/g tissue) NDa NDa 1.64 ± 0.62 10.7 ± 1.64**
Total isorhamnetin (ng/g tissue) 2.24 ± 0.53 3.64 ± 0.39** 4.13 ± 1.06 11.6 ± 1.73**

*Data are means ± SEM (n = 5–6 rats/group).


a
Not detectable.
**Significantly different from quercetin control suspension, p<0.05.

cell membrane and partitioning into the cell membrane where increasing chylomicron synthesis in rats.42 Tween 80 also en- R

they can form polar defects in the lipid bilayer. SNEDDS formu- hances chylomicron secretion in Caco-2 cells.43 Thus, admin-
lation may also produce an increased reversible effect on the istration of Q-SNEDDS is expected to increase chylomicron-
opening of tight junction so that improve the permeability.31 dependent transport of quercetin into the lymphatic system.
Because quercetin is transported by chylomicrons,40,41 Q- Cremorphor RH 40 was reported to decrease the efflux activ-
R

SNEDDS may increase lymphatic transport of quercetin in ity of multidrug resistance-associated protein 2.44 This suggests
small intestine. It was reported that long-chain fatty acids that SNEDDS formulation may reduce intestinal excretion of
of castor oil enhance lymphatic transport of quercetin by quercetin at the present of Cremorphor RH 40 to favor its R

Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014 DOI 10.1002/jps.23858


RESEARCH ARTICLE – Pharmaceutical Biotechnology 851

absorption. The developed SNEDDS formulation is biocompat- 12. Date AA, Desai N, Dixit R, Nagarsenker M. 2010. Self-
ible and free of alcohol, which potentiated commercialization nanoemulsifying drug delivery systems: Formulation insights, appli-
and clinical application compared with another newly devel- cations and advances. Nanomedicine (Lond) 5:1595–616.
oped self-emulsifying formulation.16 13. Constantinides PP. 1995. Lipid microemulsions for improving drug
dissolution and oral absorption: Physical and biopharmaceutical as-
pects. Pharm Res 12:1561–1572.
14. Mohsin K, Long MA, Pouton CW. 2009. Design of lipid-based for-
CONCLUSIONS mulations for oral administration of poorly water-soluble drugs: Pre-
cipitation of drug after dispersion of formulations in aqueous solution.
In this study, Q-SNEDDS was successfully prepared and opti-
J Pharm Sci 98:3582–3595.
mized. The optimal Q-SNEDDS composed of castor oil, Tween R

15. Zhao G, Duan J, Xie Y, Lin G, Luo H, Li G, Yuan X. 2013. Ef-


80, Cremophor RH 40, and PEG 400 (20:16:34:30) sponta-
R

fect of solid dispersion and self-emulsifying formulation on the solubil-


neously self-emulsified in water to form nanoemulsions that ity, dissolution, permeability and pharmacokinetics of isorhamnetin,
significantly increased quercetin solubility (5 mg/mL) and are quercetin and kaempferol in total flavones of Hippophae rhamnoides
stable for 1 month at ambient temperature. Our findings L. Drug Dev Ind Pharm. 39:1037–1045.
demonstrate that this Q-SNEDDS formulation increases the 16. Jain S, Jain AK, Pohekar M, Thanki K. 2013. Novel self-emulsifying
transport of quercetin across Caco-2 cell monolayer and en- formulation of quercetin for improved in vivo antioxidant potential:
hances absorption and oral bioavailability of quercetin in rats. Implications on drug induced cardiotoxicity and nephrotoxicity. Free
Radical Biol Med 65:117–130.
Further studies are needed to investigate the mechanisms of
17. Guo Y, Mah E, Davis CG, Jalili T, Ferruzzi MG, Chun OK, Bruno
this improvement in bioavailability and whether SNEDDS for-
RS. 2013. Dietary fat increases quercetin bioavailability in overweight
mulation reinforces chemoprotection activity of quercetin. adults. Mol Nutr Food Res 57:896–905.
18. Balakrishnan P, Lee B-J, Oh DH, Kim JO, Hong MJ, Lee J-
P, Kim JA, Yoo BK, Woo JS, Yong CS, Choi H-G. 2009. Enhanced
ACKNOWLEDGMENT oral bioavailability of dexibuprofen by a novel solid self-emulsifying
drug delivery system (SEDDS). Eur J Pharm BioPharm 72:539–
The authors are grateful for financial support from the Diet 545.
and Health Initiative at the University of Connecticut. 19. Prasad D, Chauhan H, Atef E. 2013. Studying the effect of lipid
chain length on the precipitation of a poorly water soluble drug
from self-emulsifying drug delivery system on dispersion into aqueous
medium. J Pharm Pharmacol 65(8):1134–44.
REFERENCES
20. Basalious EB, Shawky N, Badr-Eldin SM. 2010. SNEDDS contain-
1. Gupta SC, Kim SH, Prasad S, Aggarwal BB. 2010. Regulation of ing bioenhancers for improvement of dissolution and oral absorption
survival, proliferation, invasion, angiogenesis, and metastasis of tumor of lacidipine. I: Development and optimization. Int J Pharm 391:203–
cells through modulation of inflammatory pathways by nutraceuticals. 211.
Cancer Metastasis Rev 29(3):405–434. 21. Neslihan Gursoy R, Benita S. 2004. Self-emulsifying drug delivery
2. Kennedy D A, Hart J, Seely D. 2009. Cost effectiveness of natu- systems (SEDDS) for improved oral delivery of lipophilic drugs. Biomed
ral health products: A systematic review of randomized clinical trials. Pharmacother 58(3):173–182.
Evid-Based Complement Altern Med 6(3):297–304. 22. Elnaggar YS, El-Massik MA, Abdallah OY. 2009. Self-
3. Jagtap S, Meganathan K, Wagh V, Winkler J, Hescheler J, Sachini- nanoemulsifying drug delivery systems of tamoxifen citrate: Design
dis A. 2009. Chemoprotective mechanism of the natural compounds, and optimization. Int J Pharm 380:133–141.
epigallocatechin-3-O-gallate, quercetin and curcumin against cancer 23. Li W, Yi S, Wang Z, Chen S, Xin S, Xie J, Zhao C. 2011.
and cardiovascular diseases. Curr Med Chem 16(12):1451–1462. Self-nanoemulsifying drug delivery system of persimmon leaf ex-
4. Gibellini L, Pinti M, Nasi M, Montagna JP, De Biasi S, Roat E, tract: Optimization and bioavailability studies. Int J Pharm 420:161–
Bertoncelli L, Cooper EL, Cossarizza A. 2011. Quercetin and cancer 171.
chemoprevention. Evid Based Complement Altern Med, 2011:591356. 24. Xu S, Dai WG. 2013. Drug precipitation inhibitors in supersat-
5. Kakran M, Sahoo NG, Li L. 2011. Dissolution enhancement of urable formulation. Int J Pharm 453:36–43.
quercetin through nanofabrication, complexation, and solid dispersion. 25. Xi J, Chang Q, Chan CK, Meng ZY, Wang GN, Sun JB, Wang YT,
Colloids Surf B Biointerfaces 88:121–171. Tong HHY, Zheng Y. 2009. Formulation development and bioavailabil-
6. Gao L, Liu G, Wang X, Liu F, Xu Y, Ma J. 2011. Preparation of a ity evaluation of a self-nanoemulsified drug delivery system of oleanolic
chemically stable quercetin formulation using nanosuspension technol- acid. AAPS Pharm Scitech 10 (1):172–182.
ogy. Int J Pharm 404:231–237. 26. Parmar N, Singla N, Amin S, Kohli K. 2011. Study of cosurfactant
7. Gao Y, Wang Y, Ma Y, Yu A, Cai F, Shao W, Zhai G. 2009. Formulation effect on nanoemulsifying area and development of lercanidipine loaded
optimization and in situ absorption in rat intestinal tract of quercetin- (SNEDDS) self nanoemulsifying drug delivery system. Colloids Surf B
loaded microemulsion. Colloids Surf B Biointerfaces 71:306–314. Biointerfaces 86:327–338.
8. Li H, Zhao X, Ma Y, Zhai G, Li L, Lou H. 2009. Enhancement of 27. Mu L, Feng SS. 2002. Vitamin E TPGS used as emulsifier in the
gastrointestinal absorption of quercetin by solid lipid nanoparticles. J solvent evaporation/extraction technique for fabrication of polymeric
Control Release 133:238–244. nanospheres for controlled release of paclitaxel (TaxolR ). J Control Re-
9. Beg S, Jena SS, Patra CN, Rizwan M, Swain S, Sruti J, Rao MEB, lease 80:129–144.
Singh B. 2013. Development of solid self-nanoemulsifying granules (SS- 28. Zhang L, Strong JM, Qiu W, Lesko LJ, Huang SM. 2006. Scientific
NEGs) of ondansetron hydrochloride with enhanced bioavailability po- perspectives on drug transporters and their role in drug interactions.
tential. Colloids Surf B Biointerfaces 101:414–423. Mol Pharma 3:62–69.
10. Wang L, Dong J, Chen J, Eastoe J, Li X. 2009. Design and opti- 29. Walgren RA, Walle UK, Walle T. 1998. Transport of quercetin and
mization of a new self-nanoemulsifying drug delivery system. J Colloid its glucosides across human intestinal epithelial Caco-2 cells. Biochem
Interface Sci 330:443–448. Pharmacol 55:1721–1727.
11. Date AA, Nagarsenker MS. 2007. Design and evaluation of self- 30. Fenyvesi F, Kiss T, Fenyvesi E, Szente L, Veszelka S, Deli MA,
nanoemulsifying drug delivery systems (SNEDDS) for cefpodoxime Váradi J, Fehér P, Ujhelyi Z, Tosaki A, Vecsernyes M, Bácskay I.
proxetil. Int J Pharm 329:166–72. 2011. Randomly methylated b cyclodextrin derivatives enhance taxol

DOI 10.1002/jps.23858 Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014


852 RESEARCH ARTICLE – Pharmaceutical Biotechnology

permeability through human intestinal epithelial Caco-2 cell mono- 38. Mullen W, Rouanet JM, Auger C, Teissedre PL, Caldwell ST, Hart-
layer. J Pharm Sci 100:4734–4744. ley RC, Lean ME, Edwards CA, Crozier A. 2008. Bioavailability of
31. Chen Y, Li G, Huang JG, Wang RH, Liu H, Tang R. 2009. Com- [2-(14)C] quercetin-4 -glucoside in rats. J Agric Food Chem 56:12127–
parison of self-microemulsifying drug delivery system versus solid dis- 12137.
persion technology used in the improvement of dissolution rate and 39. Pool H, Mendoza S, Xiao H, McClements DJ. 2013. Encapsulation
bioavailability of vinpocetine. Acta Pharm Sin 44:658–666. and release of hydrophobic bioactive components in nanoemulsion-
32. Sha X, Yan G, Wu Y, Li J, Fang X. 2005. Effect of self- based delivery systems: Impact of physical form on quercetin bioac-
microemulsifying drug delivery systems containing Labrasol on tight cessibility. Food Funct 4:162–174.
junctions in Caco-2 cells. Eur J Pharm Sci 24:477–486. 40. Murota K, Terao J. 2005. Quercetin appears in the lymph of unanes-
33. Chen Y, Li G, Wu X, Chen Z, Hang J, Qin B, Chen S, Wang R. 2008. thetized rats as its phase II metabolites after administered into the
Self-microemulsifying drug delivery system (SMEDDS) of vinpocetine: stomach. FEBS Lett 579:5343–5346.
Formulation development and in vivo assessment. Biol Pharm Bull 41. Murota K, Hotta A, Ido H, Kawai Y, Moon JH, Sekido K, Hayashi
31:118–125. H, Inakuma T, Terao J. 2007. Antioxidant capacity of albumin-bound
34. Manach C, Morand C, Demigne C, Texier O, Regerat F, Remesy C. quercetin metabolites after onion consumption in humans. J Med In-
1997. Bioavailability of rutin and quercetin in rats. FEBS Lett 409:12– vest 54:370–374.
16. 42. Murota K, Cermak R, Terao J, Wolffram S. 2013. Influence of fatty
35. Wright B, Spencer JP, Lovegrove JA, Gibbins JM. 2013. Flavonoid acid patterns on the intestinal absorption pathway of quercetin in tho-
inhibitory pharmacodynamics on platelet function in physiological en- racic lymph duct-cannulated rats. Br J Nutr 109:2147–2153.
vironments. Food Funct 4(12):1803–1810. 43. Seeballuck F, Lawless E, Ashford MB, O’Driscoll CM. 2004. Stim-
36. Matsukawa N, Matsumoto M, Hara H. 2009. High biliary excretion ulation of triglyceride-rich lipoprotein secretion by polysorbate 80: In
levels of quercetin metabolites after administration of a quercetin gly- vitro and in vivo correlation using Caco-2 cells and a cannulated rat
coside in conscious bile duct cannulated rats. Biosci Biotechnol Biochem intestinal lymphatic model. Pharm Res 21:2320–2326.
73:1863–1865. 44. Hanke U, May K, Rozehnal V, Nagel S, Siegmund W, Weitschies
37. Boer VC, Dihal AA, van der Woude H, Arts IC, Wolffram S, Alink W. 2010. Commonly used nonionic surfactants interact differently with
GM, Rietjens IM, Keijer J, Hollman PC. 2005. Tissue distribution of the human efflux transporters ABCB1 (p-glgycoprotein) and ABCC2
quercetin in rats and pigs. J Nutr 135:1718–1725. (MRP2). Eur J Pharm Biopharm 76:260–268.

Tran et al., JOURNAL OF PHARMACEUTICAL SCIENCES 103:840–852, 2014 DOI 10.1002/jps.23858

You might also like