You are on page 1of 9

Pharmaceutical Research, Vol. 26, No.

5, May 2009 ( # 2008)


DOI: 10.1007/s11095-008-9729-6

Research Paper

Solubility, Stability, Physicochemical Characteristics and In Vitro Ocular Tissue


Permeability of Hesperidin: A Natural Bioflavonoid

Soumyajit Majumdar1,2,3 and Ramesh Srirangam1

Received April 30, 2008; accepted September 11, 2008; published online September 23, 2008
Purpose. Hesperidin holds potential in treating age-related macular degeneration, cataract and diabetic
retinopathy. The aim of this study, constituting the first step towards efficient ocular delivery of
hesperidin, was to determine its physicochemical properties and in vitro ocular tissue permeability.
Methods. pH dependent aqueous solubility and stability were investigated following standard protocols.
Permeability of hesperidin across excised rabbit cornea, sclera, and sclera plus retinal pigmented
epithelium (RPE) was determined using a side-bi-side diffusion apparatus.
Results. Hesperidin demonstrated poor, pH independent, aqueous solubility. Solubility improved
dramatically in the presence of 2-hydroxypropyl-beta-cyclodextrin (HP-β-CD) and the results supported
1:1 complex formation. Solutions were stable in the pH and temperature (25, 40°C) conditions tested,
except for samples stored at pH 9. Transcorneal permeability in the apical-basal and basal-apical
directions was 1.11±0.86×10−6 and 1.16±0.05×10−6 cm/s, respectively. The scleral tissue was more
permeable (10.2±2.1×10−6 cm/s). However, permeability across sclera/choroid/RPE in the sclera to
retina and retina to sclera direction was 0.82 ± 0.69 × 10−6, 1.52 ± 0.78 × 10−6 cm/s, respectively,
demonstrating the barrier properties of the RPE.
Conclusion. Our results suggest that stable ophthalmic solutions of hesperidin can be prepared and that
hesperidin can efficiently permeate across the corneal tissue. Further investigation into its penetration
into the back-of-the eye ocular tissues is warranted.
KEY WORDS: hesperetin; hesperidin; ocular; permeability; solubility.

INTRODUCTION can lead to diabetic macular edema, the major factor


responsible for vision loss (2).
Recently, it has been reported that among non- Hesperidin (hesperetin 7-rutinoside) is a flavanone
institutionalized American adults, 18 years or older, prevalence glycoside, comprising of an aglycone, hesperetin, and an
of visual impairment is 9.3%, including 0.3% with blindness (1). attached disaccharide, rutinose. The structures of hesperidin
The major eye diseases like cataract, glaucoma, macular and hesperetin are illustrated in Fig. 1. Hesperidin is
degeneration and diabetic retinopathy, are supposedly the abundantly found in citrus fruits (family Rutaceae) and
leading causes. Pathophysiology of diabetic retinopathy has also been reported to occur in many plants other than
involves the process of physiological or pathological angiogen- Citrus, such as in genera Fabaceae, Betulaceae, Laminaseae
esis. Oxidative damage to the retinal neuronal cells as a result and Papilionaceae. What makes hesperidin, and hesperetin,
of free radical generation, a complication associated with particularly attractive for the treatment of diabetic retinop-
diabetes, is considered to be a principal factor responsible for athy or macular degeneration is their potential effect on
the initiation and progression of diabetic retinopathy (2). ocular blood flow and vascular permeability, two important
Decreased retinal blood supply, as a consequence of long term factors leading to the initiation and progression of diabetic
diabetes is another major contributor to the progression of this retinopathy. It has been demonstrated that these compounds
disease. In patients with diabetes, proliferative diabetic reti- can support retinal function recovery subsequent to retinal
nopathy is the neovascular result wherein ischemia induced ischemia (3). Hesperidin’s effect on ocular blood vessels has
angiogenesis on the surface of the retina, and into the vitreous, not been studied. However, this compound was initially
is postulated to occur. Increased permeability in these vessels referred to as ‘Vitamin P’ to indicate that it could decrease
capillary permeability and fragility, although subsequently
the term vitamin P has been discontinued (4). Literature
1 reports suggest that hesperidin can prevent microvascular
Department of Pharmaceutics, School of Pharmacy, The University
leakage by virtue of its vasoprotective action through the
of Mississippi, 111 Faser Hall, University, Mississippi 38677, USA.
2
Research Institute of Pharmaceutical Sciences, The University of inhibition of the enzyme hyaluronidase which is reported to
Mississippi, University, Mississippi 38677, USA. regulate the permeability of capillary walls and supporting
3
To whom correspondence should be addressed. (e-mail: majumso@ tissues (5). Additionally, it has been demonstrated that
olemiss.edu) hesperidin can decrease blood cell and platelet aggregation,

1217 0724-8741/09/0500-1217/0 # 2008 Springer Science + Business Media, LLC


1218 Majumdar and Srirangam

proteins would limit intestinal absorption. Additionally,


reports indicate that hesperidin needs to be converted to
hesperetin, by the beta-glycosidases secreted by the intestinal
flora (22), for absorption to occur.
Over and above all the barriers to oral bioavailability, in
order to exert a therapeutic effect in diabetic retinopathy,
hesperidin must penetrate into the neural retina from the
systemic circulation. For this, it needs to permeate across the
blood-retinal barrier (BRB), formed by retinal pigmented
epithelium (RPE) and endothelial cells of retinal blood
vessels. The diffusion limiting capabilities of the BRB has
been well established for both hydrophilic (limited by tight
junctions) as well as lipophilic (through efflux mechanism)
compounds. Thus, following oral administration very little, if
any, amounts of hesperidin can reach the neural retina.
Fig. 1. A and B represents chemical structure of hesperidin The long term goal of this project is to achieve therapeu-
(hesperetin—7-rhamnoglucoside) and hesperetin (3′, 5,7-trihydroxy- tically optimal hesperidin concentrations at the target site, i.e.
4′-methoxy flavanone), respectively. posterior segment of the eye. Considering the barriers encoun-
tered in oral/systemic administration, topical instillation may be
an attractive alternative for producing therapeutic hesperidin
believed to be beneficial in cases of capillary permeability
levels at the active site. Some other possible routes/modes of
and fragility (4).
administration for achieving higher drug concentrations in the
Besides their effect on vascular permeability and ocular
retina include intravitreal injection and implants. However,
blood flow, both hesperidin and hesperetin demonstrate
these are invasive techniques and are associated with side-
strong antioxidant properties (6). This antioxidant activity is
effects such as retinal detachment and endophthalmitis.
through their ability to quench oxidative radical chain
For a topically administered drug, penetration into the
reactions (capable of oxidizing and nitrating cellular proteins,
posterior chamber tissues, such as the RPE, may be through
nucleic acids, and lipids), and can thus help preserve neuronal
corneal or noncorneal routes. Transcorneal absorption rep-
health. Hesperidin also exhibits significant anti-inflammatory
resents the major route of penetration for most therapeutic
activity by modulating the prostaglandin synthesis and COX-
agents. However, several studies demonstrate that the non-
2 gene expression pathways (7). Ayalasomayajula et al. (8)
corneal route is also a significant route, wherein the drug
reported that, in diabetic rat retina, up-regulation of COX-2 is
molecule is supposed to penetrate into the intraocular tissues
responsible for production of prostaglandin E2, a pro-
via diffusion across the conjunctiva and sclera (23).
angiogenic factor implicated in vascular permeability and
Thus, the objective of this project, constituting the first step
leakage. This prostaglandin up-regulation was inhibited by
towards efficient ocular delivery of hesperidin, was to determine
celecoxib (8), a selective COX-2 inhibitor.
hesperidin’s solubility, stability, physicochemical properties and
Hesperidin has been reported to possess analgesic (9),
in vitro permeability across the ocular tissues—cornea, sclera
hypolipidemic (10), anti-hypertensive and diuretic activity
and RPE.
(11). Another potential therapeutic application of hesperidin
is its anticancer activity mediated through the suppression of
MATERIALS AND METHODS
cell proliferation (12,13). Thus, both hesperidin and its
metabolite hesperetin, appear to be capable of modulating
Materials
multiple pathways (Fig. 2) identified in the genesis and
progression of diabetic retinopathy and diabetic macular
Hesperidin, hesperetin and 2-hydroxypropyl beta cyclo-
edema. It can also be used in other ocular diseases because
dextrin (HP-β-CD) (1,300 MW 0.6 substitution) were pur-
of its varied pharmacological actions.
chased from Sigma-Aldrich (St. Louis, MO, USA). [14C]
Currently hesperidin is used as a dietary supplement for
mannitol was purchased from American Radiolabelled
improving blood flow and for its vasoprotective properties,
Chemicals Inc. (St. Louis, MO, USA) and [3H]diazepam
and is available as an oral dosage form. Ameer et al. (14)
reported that following oral administration, hesperidin is
Decreased retinal
absorbed across the gastrointestinal tract, but cumulative Retinal
blood flow * Arachidonate* PGG2 PGH2
Hypoxia
urinary recovery indicates low bioavailability (<25%). Several
factors limit oral bioavailability of hesperidin, including poor Free radical Transcription COX-2 *
water solubility and precipitation in an acidic environment mediated damage factor Prostaglandins
of retinal blood activation e.g
(15). Moreover, hesperidin and hesperetin are substrates of vessels. * NK-κβ * PGE2, PGI2, PGF
VEGF
the intestinal efflux protein, P-glycoprotein (P-pg), (16–18)
and intestinal and hepatic drug metabolizing enzymes Hyperglycemia Angiogenesis,
CYP450 (19). These factors, acting together, severely restrict NO * vascular permeability.

systemic bioavailability of the orally administered drug. Fig. 2. Schematic representation of some of the currently identified
Furthermore, hesperidin possesses poor transmembrane per- pathways involved in the development and progression of diabetic
meability and is believed to be absorbed primarily by the retinopathy and macular degeneration. Asterisk indicates pathways
paracellular pathway (20,21). Thus intestinal tight junction which hesperidin and hesperetin can potentially inhibit.
Physicochemical Characteristics and Ocular Permeability of Hesperidin 1219

was obtained from PerkinElmer Life and Analytical Sciences 1 mg/mL hesperidin stock solution to 5 mL of the respective
(Boston, MA, USA). All other chemicals and solvents were buffer previously equilibrated to either 25°C or 40°C.
obtained from Fisher Scientific, USA. Aliquots (200 μL) were collected at 0, 0.5, 1, 2, 3, 4, 5, 6, 7,
14, 30 and 60 days and were stored at −80°C until further
Ocular Tissues analysis. Samples were analyzed using HPLC technique as
described under the analytical method section.
Ocular tissues were isolated from euthanized adult male
New Zealand albino rabbits weighing between 2 and 2.5 kg Stability in Ocular Tissue Homogenates
(Myrtle’s Rabbitry, Thompson Station, TN, USA). Experi-
ments using rabbits conformed to the tenets of the Associa- Ocular tissues isolated from New Zealand albino rabbits
tion for Research in Vision and Ophthalmology statement on were used for the metabolism studies. Animals were euthan-
the Use of Animals in Ophthalmic and Vision Research. Each ized with an overdose of pentobarbital administered through
experiment was conducted at least in quadruplicates. the marginal ear vein. Eyes were enucleated immediately and
washed with ice-cold Dulbecco’s Phosphate Buffered Saline
Solubility Studies (DPBS pH 7.4) to remove traces of blood. Vitreous humor
was collected through an incision in the sclera. Sclera was
Aqueous solubility of hesperidin was determined following isolated with the retinal pigmented epithelium (RPE) at-
standard shake flask method wherein excess quantity of drug tached. The RPE/choroid section was then carefully separat-
was added to 5 mL of solvent in a tightly capped glass vial. To ed from underlying sclera. This procedure takes
achieve uniform mixing, samples were constantly agitated at approximately 10–15 min. The pooled RPE/choroid tissue
75 rpm at room temperature (25°C) for 24 h in a reciprocating was homogenized (TISSUMISER, Fisher Scientific, USA) in
water bath (Fisher Scientific, USA). At the end of 24 h, samples 2 mL ice-cold Isotonic Phosphate Buffer Saline (IPBS),
were centrifuged (AccuSpin 17R, Fisher Scientific, USA) and pH 7.4, and then centrifuged at 4°C (accuSpin 17R, Fisher
the supernatant was analyzed for drug content. Solubility was Scientific, USA). Supernatant was collected and estimated for
determined in water and in buffers with pH values ranging total protein content by the method of Bradford (Sigma-
between 1.2 and 9. Additionally, effect of surfactants and HP-β- Aldrich, USA) (24). Final protein concentration was adjusted
CD on the solubility of hesperidin, was also evaluated. HP-β- to 1 mg/mL by diluting with IPBS and used for the hydrolysis
CD concentrations used for the phase solubility studies were 1, studies. Vitreous humor was used as such without any dilution
2.5, 5, 7.5, 10, 15 and 20%w/v in water. or homogenization.
Reactions were initiated by adding 20 μL of drug
Differential Scanning Calorimetry (DSC) Studies solution (1 mg/mL) to 2 mL of the supernatant. The reactions
were carried out in a water bath at 37°C. Samples (100 μL)
DSC analysis of hesperidin, HP-β-CD, physical mixture of were withdrawn at predetermined time intervals and the
hesperidin and HP-β-CD (1:1 molar ratio) and the complex enzymatic degradation process was arrested with the addition
were carried out using a Diamond Differential Scanning of an equal volume of ice-cold methanol/acetonitrile mixture
Calorimeter (PerkinElmer Life and Analytical Sciences, Shel- (60:40). Rate of hydrolysis in IPBS pH 7.4 at 37°C was used
ton, CT, USA). Samples, 2–3 mg, were heated in a hermetically as a control. Samples were analyzed using an HPLC
sealed aluminium pans at a heating rate of 10°C/min (25–280°C technique as described under the analytical method section.
range) under a nitrogen atmosphere (flow rate 20 mL/min). Experiments were carried out in triplicate.
An empty aluminium pan was used as the reference.
Corneal Permeation Studies
Fourier Transform Infrared Spectroscopy (FTIR) Studies
Permeation of hesperidin was studied using a side-bi-side
FTIR spectra were obtained using a Perkin Elmer FTIR diffusion apparatus (PermeGear, Inc.). Conreas isolated from
spectrometer (PerkinElmer Life and Analytical Sciences, New Zealand Albino male rabbits were used for these
Shelton, CT, USA). Samples were mixed with dry crystalline studies. Rabbits were euthanized, and eyes were collected as
KBr in a 1:100 (sample: KBr) ratio and pellets were prepared. described earlier. Cornea was isolated with a ring of sclera
A spectrum was collected for each sample within the wave around it, which helps in mounting between diffusion cells.
number region 4,000–400 cm−1. Corneas were washed in DPBS and mounted on the side-
bi-side diffusion apparatus (standard 9 mm cells were used)
Log P (Fig. 3A) with the epithelial side facing the donor cell for
apical to basolateral transport (A–B), and with the endothe-
Predicted value of Log P was obtained using the ACD/I- lial side facing the donor cell for basolateral to apical
Lab web service (ACD/Log P 8.02). transport (B–A). Temperature of the cells was maintained
at 34°C with the help of a circulating water bath. In all cases,
pH Stability 3 mL of transport buffer (DPBS containing 5% HP-β-CD) or
drug solution (150 µM of hesperidin in DPBS with 5% HP-β-
Solution stability of hesperidin was determined at CD) was added to the apical side of the cornea while 3.2 mL
various pH (1.2, 3, 5, 7.4, and 9) and temperature (25°C and was added to the basal side, to maintain the natural
40°C) conditions. Buffers were prepared according to USP. hydrostatic pressure. Aliquots (200 μL) were removed at
Stability studies were initiated with the addition of 25 μL of a predetermined time points (15, 30, 45, 60, 90, 120, 150 and
1220 Majumdar and Srirangam

an analytical method based on reversed phase HPLC. An


HPLC system equipped with Waters 600 pump controller,
2470 dual wavelength UV detector, refrigerated 717 plus
auto-sampler and Agilent 3394B integrator was used. The
detector was operated at 284 nm. Mobile phase consisted of
20 mM monobasic potassium phosphate (pH adjusted to 2.5
with orthophosphoric acid) and acetonitrile in a ratio of 75:25
and flow rate was maintained at 1 mL/min. A Waters
Symmetry Shield C18 column was used.
Analytical method for the determination of hesperetin
content was also developed. The method for hesperetin was
Fig. 3. Illustration depicting mounting of ocular tissue on regular
similar to that for hesperidin, except for the mobile phase,
side-bi-side diffusion cells (A) and on Valia–Chein side-bi-side
diffusion cells (B).
which, in this case, was a 50:50 mixture of acetonitrile and
20 mM monobasic potassium phosphate (pH adjusted to 2.5
with orthophosphoric acid).
180 min) from the receiver side and the volume withdrawn DATA ANALYSIS
was replaced with an equal volume of transport buffer.
Samples were stored at −80°C until further analysis. Cumulative amount (Mn) transported was calculated
Similar procedures were followed to determine trans- using Eq. 1 and steady state (SS) fluxes (μg min−1 cm−2)
corneal permeability of [14C]mannitol (55 mCi/mmol) and were determined from the slope of the cumulative amount of
[ 3 H]diazepam (70 mCi/mmol), both at 0.5 μCi/mL drug transported vs. time graph and expressed as per unit of
concentration. Five milliliters of scintillation cocktail (Fisher surface area as described by Eq. 2.
Scientific, USA) was added to each sample and radioactivity
was measured using Liquid Scintillation Analyzer X
x¼n
(PerkinElmer Life and Analytical Sciences, Model Tri-Carb Mn ¼ Vr CrðnÞ þ VSðx1Þ Crðx1Þ ð1Þ
2900TR, Shelton, CT, USA). x¼1

Permeation Across Sclera and Retinal Pigmented Epithelium


(RPE) ðdM=dt Þ
FluxðJÞ ¼ ð2Þ
A
Transscleral permeation studies were carried out using
the 5 mm diameter Valia–Chein side-bi-side diffusion cells Where, Mn is the cumulative amount of the drug in the
from PermeGear, Inc (Fig. 3B). After carefully isolating receiver chamber at nth sampling time point (n=1, 2, 3, 4, 5, 6,
sclera alone, or sclera with RPE/choroid, the tissues were 7 and 8 corresponding to 15, 30, 45, 60, 90, 120, 150 and
mounted in a manner such that outer surface of the sclera, 180 min sampling points), Vr is the volume of the medium in
known as the episclera, faced the donor side (Fig. 3B; half-cell the receiver chamber, VS is the volume of the sample
a) and the inner side of sclera, or the RPE side, faced the withdrawn at the nth time point, Cr(n) is the concentration of
receiver chamber (Fig. 3B half-cell b) for the transport studies the drug in the receiver chamber medium at nth time point, A
in the sclera to choroid/RPE direction (S to R direction). is the surface area of the tissue exposed to the permeant
Donor solution was added to the RPE/choroid side (Fig. 3B (surface area of cornea and sclera/RPE exposed is 0.636 and
half-cell b) for transport in the RPE/choroid to scleral 0.192 cm2, respectively). Note that when n=1, the second part
direction (R to S direction). Temperature of the cells was of the right hand side of Eq. 1 becomes zero since
maintained at 37°C with the help of a circulating water bath. concentration in the receiver chamber, Cr, is zero at time zero.
150 μM of hesperidin in DPBS with 5% HP-β-CD was placed Membrane permeability (cm/s) values were determined
in donor chamber and DPBS with 5% HP-β-CD was placed in by normalizing the steady state fluxes to the donor concen-
receiver chamber. Aliquots (200 μL) were removed at tration, Cd according to Eq. 3.
predetermined time points (15, 30, 45, 60, 90, 120, 150 and  
180 min) from the receiver side and replaced with an equal Permeability Papp ¼ Flux=Cd ð3Þ
volume of transport buffer. Samples were stored at −80°C
until further analysis.
Similar procedures were followed to determine perme- STATISTICAL ANALYSIS
ability of [14C]mannitol (55 mCi/mmol) and [3H]diazepam
(70 mCi/mmol), both at 0.5 μCi/mL concentration, across All experiments were carried out at least in triplicate.
sclera/choroid/RPE. Five milliliters of scintillation cocktail Unpaired t test and ANOVA was used for statistical analysis.
(Fisher Scientific, USA) was added to each sample and In all the cases a p value less than 0.05 was considered to
radioactivity was measured using liquid scintillation analyzer. denote statistically significant difference.

Analytical Method RESULTS AND DISCUSSION

Hesperidin content in samples collected from the solu- Diabetic retinopathy and macular edema are serious
bility, stability and permeability studies was estimated using sight threatening conditions. While involvement of several
Physicochemical Characteristics and Ocular Permeability of Hesperidin 1221

0.0008
mechanisms in the pathogenesis of this disease has been

Hesperidin Solubility
suggested, a drug candidate capable of blocking multiple 0.0007

pathways remains to be identified, as yet. Current treatment 0.0006

(moles/L)
options, apart from controlling blood glucose and lipid levels, 0.0005
include corticosteroids, anti-vascular endothelial growth fac- 0.0004
tor, laser photocoagulation, and vitrectomy. However, long
0.0003
term treatment with these drugs is associated with toxicity
0.0002
and corticosteroids may lead to cataract formation and
increased intraocular pressure (25). Hesperidin appears to 0.0001

modulate multiple pathways associated with diabetic retinop- 0


0 0.02 0.04 0.06 0.08 0.1 0.12 0.14 0.16
athy and macular edema (Fig. 2) but a suitable ocular
HP-β-CD concentration (moles/L)
delivery mechanism is lacking. In this study, feasibility of
developing an ophthalmic solution dosage form of hesperidin Fig. 4. Phase solubility of hesperidin in the presence of HP-β-CD, at
was investigated 25°C, following 24 h equilibration. Values are expressed as mean±SD
Solubility is an important factor affecting drug perme- (n=3).
ability across biological membranes. Solubility of hesperidin
in different pH buffers and in water is presented in Table I.
The compound was observed to be very poorly soluble in complex was not reached within the concentration range of
water and exhibited an aqueous solubility value of 4.95 μg/mL HP-β-CD used in this study. Stoichiometry, binding constant
(7.5×10−6 M) at 25°C. Hesperidin did not demonstrate pH (K) and aqueous saturation solubility (S0) were determined
dependent solubility within the pH range of 1.2–9. However, from a plot of solubility as a function of HP-β-CD concentra-
solubility of hesperidin at pH values greater than 9 was tion using Eq. 4, according to the method proposed by Higuchi
significantly higher (data not shown) but was accompanied and Connors. Hesperidin binding constant was calculated to be
with degradation. These results are in agreement with the 625±78 M−1 (R2 =0.975±0.023, slope=0.0052±0.0006).
studies reported by Serra et al. wherein the pKa of hesperidin
has been reported to be 10±0.2 for the two phenolic hydroxyl
groups and greater than 11.5 for all of the alcoholic hydroxyl
Slope
groups (21). K¼ ð4Þ
In an attempt to improve hesperidin’s aqueous solubility, S0 ð1  SlopeÞ
effect of ethanol, surfactants and HP-β-CD, was evaluated.
Ethanol and surfactants such as Tween 80 and TPGS (d-
alpha-tocopheryl polyethylene glycol 1000 succinate) were Further evidence of the formation of an inclusion
ineffective. However, HP-β-CD markedly improved hesper- complex was obtained from thermal and spectral analysis.
idin’s solubility. Cyclodextrins are a group of cyclic oligosac- The DSC thermograms are presented in Fig. 5. DSC scans of
charides that have been shown to improve solubility of a pure hesperidin showed an endothermic peak at 259°C,
multitude of poorly soluble compounds, through the forma- corresponding to the melting point of the compound. Physical
tion of inclusion complexes. Among the different cyclo- mixture of the drug and HP-β-CD, in a 1:1 molar ratio, also
dextrins available, HP-β-CD has been reported to be safe exhibited the endothermic peak associated with hesperidin.
for use in ocular preparations (26,27), even when used at However, the peak was not observed in the complex,
concentrations as high as 45% (28). prepared by dissolving both the components in methanol
A linear increase in aqueous solubility of hesperidin was (1:1 molar ratio) followed by evaporation. This suggests
observed with increasing concentrations of HP-β-CD (Fig. 4 and molecular encapsulation of drug inside the cyclodextrin
Table II) suggesting formation of a 1:1 inclusion complex cavity. FTIR scans complemented the DSC data (Fig. 6).
between hesperidin and HP-β-CD. It is interesting to note that Hesperidin spectra demonstrate a strong absorption band at
hesperidin’s solubility was increased approximately 100-fold in 1,644 cm−1, corresponding to its carbonyl stretching vibration.
the presence of 20% HP-β-CD. The solubility limit of the This characteristic carbonyl stretching vibration was shifted to

Table II. Aqueous Solubility of Hesperidin (25°C, 24 h Equilibration)


in the Presence of Various Concentrations of HP-β-CD
Table I. Solubility of Hesperidin as a Function of pH (200 mM Buffer
Concentration) and in Water Concentration of Hesperidin Fold Increase
HP-β-CD (%w/v) solubility (µg/mL) in solubility
pH Saturation solubility (µg/mL)
0 4.95±0.99
1.21 4.15±0.34 1 42.97±4.28 9
3.11 8.44±0.47 2.5 83.34±6.34 18
5.12 5.96±1.92 5 169.90±25.36 37
7.48 5.26±0.31 7.5 219.03±15.62 48
9.11 8.93±0.73 10 252.77±8.81 55
Water 4.93±0.99 15 348.55±5.28 76
20 431.97±0.43 95
The solubility studies were carried out at 25°C for a period of 24 h.
Values are expressed as mean±SD (n=3) Values are expressed as mean±SD (n=3)
1222 Majumdar and Srirangam

Fig. 5. DSC thermograms of the pure hesperidin and HP-β-CD, physical mixture of hesperidin and HP-β-CD (1:1 molar ratio) and the
complex.

1,652 cm−1 in the complex and was much broader, suggesting


the formation of complex. Thus, chemical, thermal and
spectral analysis, taken together, strongly supports the
formation of an inclusion complex.
Phase solubility studies pointed towards the formation
of the complex in a 1:1 molar ratio, which is consistent
with an earlier report by Tommasini et al. (29). However,
the observed intrinsic solubility of hesperidin (7.5×10−6 M)
in the current study was less than the value (3.6×10−5 M)
reported by Tommasini et al. Also, the estimated binding
constant in the previous study was 60 M−1. The observed
differences, between the earlier report and the current
study, with respect to these two parameters, may be
attributed to the experiment protocol. While Tommasini
et al. sonicated the excess drug containing cyclodextrin
solution for a period of 15 min and then allowed
equilibration for a period of 4 days, in the current study
sonication was not a part of the protocol and equilibration
period allowed was only 24 h.
Hesperidin, a flavonoid glycoside, can be converted into
its aglycone, hesperetin through chemical or enzymatic
processes. Stability studies in aqueous solutions did not
demonstrate any decrease in hesperidin content upto
2 months, the final time point tested, in the pH range and
temperature conditions employed. The only exception was
pH 9 samples. In this case, degradation rate constant and
half-lives, at 25°C and 40°C were observed to be 0.03 and
Fig. 6. FTIR spectra of the HP-β-CD, hesperidin and the complex. 0.15 day−1, and, 23 and 4.5 days, respectively (Fig. 7). This
Physicochemical Characteristics and Ocular Permeability of Hesperidin 1223

0.9
entire duration of the diffusion experiments. Since hesperidin
0.8
demonstrates poor aqueous solubility, 5% HP-β-CD was
Log Concentration Remaining

0.7 incorporated in the transport buffer.


40 Deg
0.6 Trans-epithelial electrical resistance (TEER) was moni-
0.5 25 Deg tored (Multimeter, Agilent Technologies, USA) throughout
0.4 the duration of the corneal permeation studies, 3 h, to
0.3
evaluate the integrity of tissue in the presence of 5% HP-β-
CD. TEER values for corneas exposed to transport buffers
0.2
with or without HP-β-CD were found to be 5±0.3 KΩcm2
0.1
throughout, demonstrating lack of any effect of HP-β-CD on
0 ocular tissue integrity at this concentration. The observed
-0.1 0 10 20 30 40 50 60 70 TEER is in agreement with previously reported TEER values
-0.2 for rabbit cornea (33). In addition, diffusion of [14C]mannitol,
Time (Days)
a paracellular marker, and [3H]diazepam, a transcellular
Fig. 7. Stability of hesperidin in pH 9 buffer at 25°C and 40°C. Values marker, were also evaluated. The mean percent cumulative
are expressed as mean±SD (n=3). amount of the [14C]mannitol transported per hour across the
cornea remained the same, for the duration of the experiment
at 1±0.15. In case of diazepam also, mean percent cumulative
amount of [3H]diazepam transported per hour across the
cornea remained steady at 1.62±0.04. These results taken
signifies that hesperidin may undergo alkaline hydrolysis at
together, demonstrate that integrity and viability of the
higher pH and temperature conditions. corneal tissues are maintained during the experimental
Hesperidin undergoes enzymatic degradation by beta-
protocol.
glucosidases to yield hesperetin. Literature reports suggest
Mean cumulative amount of hesperidin transported
that beta-glucosidases are expressed in the ocular tissues,
across ocular tissues as a function of time is depicted in
especially in the RPE (30,31). Ideally, one would like to Fig. 8 and the permeability coefficients have been presented
maintain the drug in the unmetabolized form at the target
in Fig. 9. Steady state diffusion was achieved within 5 min.
tissue for maximizing therapeutic effect. With respect to
hesperidin, however, both hesperidin and its aglycone hes-
peretin, are pharmacologically active. Even then, it is
important to know the rate of generation of hesperetin, from
3.0
hesperidin, in the ocular tissues as the two compounds would
exhibit different pharmacokinetic profiles. For example,
hesperetin is more lipophilic than hesperidin and is known
Mean cumulative amount transported

2.5
to be a much better substrate of the efflux protein P-gp, which
is also known to be expressed on the RPE (32). Consequent-
ly, conversion of hesperidin to hesperetin may impact 2.0
therapeutic activity. Taking this into consideration, stability
of hesperidin in ocular tissue homogenates was determined.
(µg)

Hesperetin generation, or decrease in hesperidin concentra- 1.5


tions, in the homogenates was not observed even after 15 h.
Further studies investigating generation of hesperetin in the
ocular tissues in vivo, and their in vivo pharmacokinetic 1.0
profile following topical and intravitreal administration, are in
progress.
0.5
Oral hesperidin administration for ocular indications
may not be very effective. Local delivery of hesperidin for
ocular diseases may prove to be a much more viable option,
0.0
compared to oral administration. To our knowledge, hitherto, 0 50 100 150 200
permeability of hesperidin across ocular tissues has not been Time (min)
determined. In this study, ocular tissues isolated from New
Zealand albino rabbits, a model widely used for ophthalmic Cornea (A-B) y= 0.0038x + 0.0125
R² = 0.9949
drug delivery studies, were used to evaluate feasibility of
y= 0.0036x -0.0529
penetration of hesperidin into the back-of-the eye following Cornea (B-A)
R² = 0.9761
topical administration. The predicted Log P value of hesper- Sclera
y= 0.0114x -0.0296
idin was determined to be 1.78±0.72 using ACD/I-Lab R² = 0.9971
Sclera with RPE (S-R) y= 0.0006x + 0.016
software, which indicated that the compound is fairly R² = 0.863
lipophilic. Diffusion studies were carried out for 3 h, to y= 0.0015x + 0.028
Sclera with RPE (R-S)
determine in vitro permeability coefficients across cornea, R² = 0.9959
sclera and sclera/choroid/RPE. Stability studies of hesperidin Fig. 8. Mean cumulative amount of hesperidin transported across
in aqueous buffered solutions and tissue homogenates ocular tissues as a function of time. Values are expressed as mean±
demonstrated that drug was stable in these matrices for the SD (n=4).
1224 Majumdar and Srirangam

14
indicating that their membrane permeation characteristics are
12
also comparable. The close approximation of the
Permeability x 106

10 permeability of ACV obtained using the Valia-Chien cells


8 with that of GCV in the Franz diffusion cells, indicate that the
6 RPE structure was intact during the experimental protocol.
4 Moreover, the mean percent cumulative amount of [14C]
2
mannitol and [3H]diazepam transported per hour across the
sclera/choroid/RPE tissues were 0.1±0.04 and 0.08±0.02,
0
Cornea (A-B) Cornea (B-A) Sclera Sclera with Sclera with respectively, similar to the values reported by Kansara et al.
RPE (S-R) RPE (R-S) (35). The above results indicate that sclera/choroid/RPE
Fig. 9. Permeability (cm/s) of hesperidin across various ocular tissue maintains its integrity during the experimental
tissues. A–B denotes permeation in the apical to basolateral protocol.
direction. S–R and R–S denotes permeation in the sclera to RPE Permeability of hesperidin across the sclera alone (10.2±
direction and RPE to sclera direction. Values are expressed as 2.1×10−6 cm/s) was almost tenfold higher than that across
mean ± SD (n= 4). sclera with RPE (Figs. 8 and 9) and that across the cornea.
These results thus substantiate the concept that RPE presents
a significant barrier to the transscleral diffusion process
following systemic as well as topical administration.
However, topical instillation should generate significantly
Less than 2% decrease in hesperidin concentration in higher concentration gradients across the RPE and thus
the donor solution was observed during the total time higher retinal hesperidin concentrations.
course of the experiments signifying maintenance of sink The barrier properties of the RPE may be as a result of
conditions. efflux pump involvement or high lipophilicity of the thera-
Hesperidin’s permeability across the rabbit cornea was peutic agent. The RPE is reported to express P-gp (32), which
1.11±0.86×10−6 cm/s consistent with other compounds with can modulate drug clearance from the vitreous humor. To
similar LogP values (34). Although, P-gp has been reported delineate the role of efflux mechanisms on trans-RPE drug
to play a important role in transcorneal drug absorption delivery, permeability of hesperidin across RPE in both
(32), corneal permeability of hesperidin in the apical to directions i.e. sclera→choroid→RPE (S–R) and RPE→chor-
basolateral (A–B; 1.11±0.86×10−6 cm/s) and basolateral to oid→sclera (R–S) was measured and were found to be 0.82±
apical (B-A; 1.16±0.05×10−6 cm/s) directions did not differ 0.68×10−6 cm/s and 1.52±0.78×10−6 cm/s, respectively (Figs. 8
statistically, indicating lack of involvement of any carrier- and 9). The difference in permeability in the S–R and R–S
mediated process (efflux or influx) in the corneal permeation directions, under the experimental conditions employed, was
process, at the concentration studied. These results are statistically insignificant suggesting lack of any efflux
consistent with an earlier study by Kobayashi et al. (20) with mechanism.
Caco-2 cell monolayers demonstrating similar hesperidin In conclusion, results from this study suggest that
permeability in the A–B and B–A directions. The authors hesperidin solubility can be markedly improved by complex-
concluded that hesperidin permeates through paracellular ation with HP-β-CD and that hesperidin exhibits sufficient
route and absence of active transport process. Moreover, chemical and enzymatic stability in aqueous solutions and
hesperidin behaves as a neutral molecule in the pH range of biological matrices, respectively. Additionally, ocular tissue
1–10 (21) indicating lack of involvement of any anion or permeation characteristics of hesperidin are not limited by
cation transporters. efflux mechanisms. Thus, topical delivery of hesperidin in the
Transport studies across sclera and sclera/choroid/RPE form of ophthalmic solutions/suspensions appears feasible. A
were carried out using Valia–Chein cells (PermeGear, Inc. pH range of 6.5–8.5 is considered to be optimal for
USA). To our knowledge, these cells are being adapted for ophthalmic forumations. Considering the favorable perme-
studying permeability across the RPE for the first time. The ability, solubility and stability profiles, it would be reasonable
scleral tissue tends to fold-up into its natural elliptical shape to formulate hesperidin based ophthalmic solutions at pH 7.4.
while mounting on the regular flat face cells. This creates Further investigation into penetration of hesperidin into the
mounting problems, especially the risk of damaging the RPE back-of-the eye ocular tissues, following topical and systemic
layer. On the other hand, it is very easy to mount the sclera/ administration is warranted.
choroid/RPE tissue on the Valia–Chein cells. To check the
validity of the new experimental set up, acyclovir (ACV)
transport across sclera with RPE was carried out from the S
to R (sclera to RPE) direction. The observed permeability ACKNOWLEDGEMENT
was 2.5×10−6 cm/s. Although there are no prior reports for
trans-RPE permeability of ACV, Kansara et al. (35) reported This project was supported by NIH grant numbers
that the permeability of ganciclovir (GCV) in the S to R P20RR021929, from the National Center for Research
direction was 1.61×10−6 cm/s. GCV and ACV are very similar Resources, and EY018426-01 from the National Eye Institute.
structurally, and with respect to their physicochemical The content is solely the responsibility of the authors and
characteristics. Moreover, the permeability of ACV and does not necessarily represent the official views of the
GCV across the rabbit cornea are reported to be very National Center for Research Resources or the National
similar (3.5×10−6 and 4.1×10−6 cm/s, respectively) (36,37), Eye Institute, National Institutes of Health.
Physicochemical Characteristics and Ocular Permeability of Hesperidin 1225

REFERENCES 19. V. M. Breinholt, E. A. Offord, C. Brouwer, S. E. Nielsen, K.


Brosen, and T. Friedberg. In vitro investigation of cytochrome
P450-mediated metabolism of dietary flavonoids. Food Chem.
1. A. Ryskulova, K. Turczyn, D. M. Makuc, M. F. Cotch, R. J. Toxicol. 40:609–616 (2002) doi:10.1016/S0278-6915(01)00125-9.
Klein, and R. Janiszewski. Self-reported age-related eye diseases 20. S. Kobayashi, S. Tanabe, M. Sugiyama, and Y. Konishi. Trans-
and visual impairment in the United States: results of the 2002 epithelial transport of hesperetin and hesperidin in intestinal
national health interview survey. Am. J. Public Health. 98:454– Caco-2 cell monolayers. Biochim. Biophys. Acta. 1778:33–41
461 (2008) doi:10.2105/AJPH.2006.098202. (2008) doi:10.1016/j.bbamem.2007.08.020.
2. J. L. Wilkinson-Berka. Vasoactive factors and diabetic retinop- 21. H. Serra, T. Mendes, M. R. Bronze, and A. L. Simplicio.
athy: vascular endothelial growth factor, cycoloxygenase-2 and Prediction of intestinal absorption and metabolism of pharma-
nitric oxide. Curr. Pharm. Des. 10:3331–3348 (2004) doi:10.2174/ cologically active flavones and flavanones. Bioorg. Med. Chem.
1381612043383142. 16:4009–4018 (2008).
3. X. R. Chiou GC. Effects of some natural flavonoids on retinal 22. C. Manach, C. Morand, A. Gil-Izquierdo, C. Bouteloup-
function recovery after ischemic insult in the rat. J. Ocular. Demange, and C. Remesy. Bioavailability in humans of the
Pharmacol. Therap. 20:107–113 (2004) doi:10.1089/108076 flavanones hesperidin and narirutin after the ingestion of two
804773710777.
doses of orange juice. Eur. J. Clin. Nutr. 57:235–242 (2003)
4. A. Garg, S. Garg, L. J. Zaneveld, and A. K. Singla. Chemistry
doi:10.1038/sj.ejcn.1601547.
and pharmacology of the citrus bioflavonoid hesperidin. Phyt-
23. A. K. Mitra, S. Macha, and P. M. Hughes. Ophthalmic Drug
other. Res. 15:655–669 (2001) doi:10.1002/ptr.1074.
Delivery Systems. Marcel Dekker, New York, 2003.
5. J. M. Beiler, and G. J. Martin. Inhibition of hyaluronidase action
24. M. M. Bradford. A rapid and sensitive method for the
by derivatives of hesperidin. J. Biol. Chem. 174:31–34 (1948).
quantitation of microgram quantities of protein utilizing the
6. J. Zhang, R. A. Stanley, L. D. Melton, and M. A. Skinner. Inhibition
principle of protein-dye binding. Anal. Biochem. 72:248–254
of lipid oxidation by phenolic antioxidants in relation to their
(1976) doi:10.1016/0003-2697(76)90527-3.
physicochemical properties. Pharmacologyonline. 1:180–189 (2007).
25. N. M. Holekamp, M. A. Thomas, and A. Pearson. The safety
7. A. Hirata, Y. Murakami, M. Shoji, Y. Kadoma, and S. Fujisawa.
Kinetics of radical-scavenging activity of hesperetin and hesper- profile of long-term, high-dose intraocular corticosteroid deliv-
idin and their inhibitory activity on COX-2 expression. Antican- ery. Am. J. Ophthalmol. 139:421–428 (2005) doi:10.1016/j.
cer Res. 25:3367–3374 (2005). ajo.2004.09.046.
8. S. P. Ayalasomayajula, A. C. Amrite, and U. B. Kompella. 26. T. Jansen, B. Xhonneux, J. Mesens, and M. Borgers. Beta-
Inhibition of cyclooxygenase-2, but not cyclooxygenase-1, reduces cyclodextrins as vehicles in eye-drop formulations: an evaluation
of their effects on rabbit corneal epithelium. Lens Eye Toxic. Res.
prostaglandin E2 secretion from diabetic rat retinas. Eur. J.
7:459–468 (1990).
Pharmacol. 498:275–278 (2004) doi:10.1016/j.ejphar.2004.07.046.
9. E. M. Galati, M. T. Monforte, S. Kirjavainen, A. M. Forestieri, 27. P. Saarinen-Savolainen, T. Jarvinen, K. Araki-Sasaki, H. Wata-
A. Trovato, and M. M. Tripodo. Biological effects of hesperidin, nabe, and A. Urtti. Evaluation of cytotoxicity of various
a citrus flavonoid. (Note I): antiinflammatory and analgesic ophthalmic drugs, eye drop excipients and cyclodextrins in an
activity. Farmaco. 40:709–712 (1994). immortalized human corneal epithelial cell line. Pharm. Res.
10. M. T. Monforte, A. Trovato, S. Kirjavainen, A. M. Forestieri, E. 15:1275–1280 (1998) doi:10.1023/A:1011956327987.
M. Galati, and R. B. Lo Curto. Biological effects of hesperidin, a 28. T. Loftssona, and T. Jarvinen. Cyclodextrins in ophthalmic drug
citrus flavonoid. (note II): hypolipidemic activity on experimen- delivery. Adv. Drug Deliv. Rev. 36:59–79 (1999) doi:10.1016/
tal hypercholesterolemia in rat. Farmaco. 50:595–599 (1995). S0169-409X(98)00055-6.
11. E. M. Galati, A. Trovato, S. Kirjavainen, A. M. Forestieri, A. 29. S. Tommasini, M. L. Calabro, R. Stancanelli, P. Donato, C.
Rossitto, and M. T. Monforte. Biological effects of hesperidin, a Costa, S. Catania, V. Villari, P. Ficarra, and R. Ficarra. The
citrus flavonoid. (Note III): antihypertensive and diuretic activity inclusion complexes of hesperetin and its 7-rhamnoglucoside
in rat. Farmaco. 51:219–221 (1996). with (2-hydroxypropyl)-beta-cyclodextrin. J. Pharm. Biomed.
12. T. Tanaka, H. Makita, M. Ohnishi, H. Mori, K. Satoh, A. Hara, Anal. 39:572–580 (2005) doi:10.1016/j.jpba.2005.05.009.
T. Sumida, K. Fukutani, T. Tanaka, and H. Ogawa. Chemo- 30. D. K. Wilcox. Extracellular release of acid hydrolases from
prevention of 4-nitroquinoline 1-oxide-induced oral carcinogen- cultured retinal pigmented epithelium. Invest. Ophthalmol. Vis.
esis in rats by flavonoids diosmin and hesperidin, each alone and Sci. 28:76–82 (1987).
in combination. Cancer Res. 57:246–252 (1997). 31. A. J. Adler. Selective presence of acid hydrolases in the
13. T. Tanaka, H. Makita, K. Kawabata, H. Mori, M. Kakumoto, K. interphotoreceptor matrix. Exp. Eye Res. 49:1067–1077 (1989)
Satoh, A. Hara, T. Sumida, T. Tanaka, and H. Ogawa. Chemo- doi:10.1016/S0014-4835(89)80027-2.
prevention of azoxymethane-induced rat colon carcinogenesis by 32. E. Mannermaa, K. S. Vellonen, and A. Urtti. Drug transport in
the naturally occurring flavonoids, diosmin and hesperidin. corneal epithelium and blood-retina barrier: emerging role of
Carcinogenesis. 18:957–965 (1997) doi:10.1093/carcin/18.5.957. transporters in ocular pharmacokinetics. Adv. Drug Deliv. Rev.
14. B. Ameer, R. A. Weintraub, J. V. Johnson, R. A. Yost, and R. L. 58:1136–1163 (2006) doi:10.1016/j.addr.2006.07.024.
Rouseff. Flavanone absorption after naringin, hesperidin, and 33. W. S. Marshall, and S. D. Klyce. Cellular and paracellular
citrus administration. Clin. Pharmacol. Ther. 60:34–40 (1996) pathway resistances in the “tight” Cl- -secreting epithelium of
doi:10.1016/S0009-9236(96)90164-2. rabbit cornea. J. Membr. Biol. 73:275–282 (1983) doi:10.1007/
15. A. Gil-Izquierdo, M. I. Gil, F. A. Tomas-Barberan, and F. BF01870542.
Ferreres. Influence of industrial processing on orange juice 34. M. R. Prausnitz, and J. S. Noonan. Permeability of cornea, sclera,
flavanone solubility and transformation to chalcones under and conjunctiva: a literature analysis for drug delivery to the eye.
gastrointestinal conditions. J. Agric. Food Chem. 51:3024–3028 J. Pharm. Sci. 87:1479–1488 (1998) doi:10.1021/js9802594.
(2003) doi:10.1021/jf020986r. 35. V. Kansara, Y. Hao, and A. K. Mitra. Dipeptide monoester
16. T. H. Tsai, and M. C. Liu. Determination of extracellular ganciclovir prodrugs for transscleral drug delivery: targeting the
hesperidin in blood and bile of anaesthetized rats by microdialysis oligopeptide transporter on rabbit retina. J. Ocul. Pharmacol.
with high-performance liquid chromatography: a pharmacokinet- Ther. 23:321–334 (2007) doi:10.1089/jop.2006.0150.
ic application. J. Chromatogr. B Analyt. Technol. Biomed. Life 36. S. Majumdar, K. Hippalgaonkar, and M. A. Repka. Effect of
Sci. 806:161–166 (2004) doi:10.1016/j.jchromb.2004.03.047. chitosan, benzalkonium chloride and ethylenediaminetetraacetic
17. Y. Mitsunaga, H. Takanaga, H. Matsuo, M. Naito, T. Tsuruo, H. acid on permeation of acyclovir across isolated rabbit cornea. Int.
Ohtani, and Y. Sawada. Effect of bioflavonoids on vincristine J. Pharm. 348:175–178 (2008) doi:10.1016/j.ijpharm.2007.08.017.
transport across blood–brain barrier. Eur. J. Pharmacol. 395:193– 37. S. Majumdar, Y. E. Nashed, K. Patel, R. Jain, M. Itahashi, D. M.
201 (2000) doi:10.1016/S0014-2999(00)00180-1. Neumann, J. M. Hill, and A. K. Mitra. Dipeptide monoester
18. M. Ofer, S. Wolffram, A. Koggel, H. Spahn-Langguth, and P. ganciclovir prodrugs for treating HSV-1-induced corneal epithe-
Langguth. Modulation of drug transport by selected flavonoids: in- lial and stromal keratitis: in vitro and in vivo evaluations. J. Ocul.
volvement of P-gp and OCT? Eur. J. Pharm. Sci. 25:263–271 (2005). Pharmacol. Ther. 21:463–474 (2005) doi:10.1089/jop.2005.21.463.

You might also like