You are on page 1of 11

Biomedicine & Pharmacotherapy 112 (2019) 108622

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Quercetin loaded nanoemulsion-based gel for rheumatoid arthritis: In vivo T


and in vitro studies
Jayanti P. Gokhalea, , Hitendra S. Mahajana, Sanjay J. Suranab

a
Department of Pharmaceutics, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India
b
Department of Pharmacognosy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Maharashtra, India

ARTICLE INFO ABSTRACT

Keywords: Current research reports the development, optimization and evaluation of Quercetin (QCT) loaded nanoemul-
Quercetin sion (NE)-based gel for the effective rheumatoid arthritis (RA) management. The formulation of QCT- NE was
Nanoemulsion developed using spontaneous emulsification techniques using the Box- Behnken experimental design. The cy-
Rheumatoid arthritis totoxicity study and effect on TNF-α production were evaluated respectively on HIG-82 and RAW 264.7 cells.
Gel
The study showed that QCT- NE has no toxic effect on synoviocytes and a strong inhibitory effect on LPS-induced
TNF-α production. QCT- NE gel has confirmed adequate rheological behavior with a good texture profile and
improved drug permeation compared to free QCT gel. In addition, the gel was found to be non-irritating and
showed the inhibition of paw edema in rats induced by CFA over 24 h contrary to free QCT gel. In conclusion, the
formulation of QCT- NE gel is an efficient topical treatment strategy for rheumatoid arthritis.

1. Introduction antiviral, antidiabetic, anti - inflammatory neuro-protective and anti-


cancer. QCT inhibits, pro-inflammatory cytokines for example, TNF-α,
Arthritic disorders are among the most common diseases and are an nitric oxide, IL-6, IL-1β, chemokines, etc. Consequently, it helps in the
important cause of joint dysfunction and disability in adults [1]. The management of chronic oxidative stress-related diseases such as ar-
most prevalent type of arthritis is rheumatoid arthritis, which is an thritis, inflammation and diabetes, as reported in several research stu-
autoimmune disorder caused by activation of pro-inflammatory med- dies [6]. QCT is administered orally in three daily doses of 250–500 mg;
iators such as TNF-α, IL-1β and it is characterized by peripheral joint afterwards, it is absorbed into the small intestine and colon, where QCT
pain, stiffness and swelling. Rheumatoid arthritis treatment includes and where QCT and its glycoside derivatives are combined with glu-
analgesics, steroidal and non-steroidal anti - inflammatory drugs, dis- curonic acid. It then binds to albumin and is transported to the liver,
ease - modifying anti - rheumatic drugs (DMARDs), monoclonal anti- which means that a very small fraction of QCT is freely available in the
bodies (MAbs) that only provide symptomatic relief [2]. Nevertheless, bloodstream and a greater proportion appears in the blood as QCT
these medications have a wide variety of harmful side effects that re- metabolites [7]. Furthermore, QCT has poor gastrointestinal retention,
semble stomach upset, nephrotoxicity, protein loss, toxicity, lack of low skin penetration, insufficient water solubility and rapid excretion.
target specificity, immunosuppressive effects that ultimately lead to Consequently, the oral delivery of QCT is not applicable and its clinical
poor patient consistency [3,4]. As a result, the importance of various applications are poorly efficient. It degrades rapidly not only in alkaline
anti-inflammatory drugs has decreased and plant – based agents asso- conditions but through heat exposure during product processing and
ciated with the least side effects are in the process of treating rheu- shelf life [6–13]. These obstacles regarding QCT increase the necessity
matoid arthritis [5]. of novel formulation that stabilizes QCT and exploits the therapeutic
In recent years, flavonoids have attained great interest because of benefits of QCT.
the wide range of biological properties such as antioxidants, anti-pro- In recent decades, several QCT nanoformulations, such as micro-
liferative and anti - inflammatory activities. QCT is a leading flavonol emulsion, microspheres, nanostructured lipid carriers, solid lipid na-
collectively distributed in a variety of edible plants and one of the noparticles and polymer nanoparticles, have been investigated to im-
potent plant origin antioxidants. It is a safe and often ingested dietary prove bioavailability by solubilization or encapsulation and to achieve
flavonoid along with numerous therapeutic advantages, such as better therapeutic effects [14–18]. Despite its great biological potential,


Corresponding author.
E-mail address: jayanti.gokhale@yahoo.co.in (J.P. Gokhale).

https://doi.org/10.1016/j.biopha.2019.108622
Received 28 July 2018; Received in revised form 20 January 2019; Accepted 23 January 2019
0753-3322/ © 2019 Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/).
J.P. Gokhale, et al. Biomedicine & Pharmacotherapy 112 (2019) 108622

QCT remains unexplored with regard to the development of an in- for particle size, PDI, and zeta potential [20].
novative topical formulation for the treatment of rheumatism. In this For the development and optimization of the QCT loaded nanoe-
study, we hypothesized that, formulation of nanoemulsion with QCT for mulsion, three factors, three-level Box-Behnken experimental design
topical delivery could lead to significant uptake through skin rendering (BBD) was selected.
its therapeutic potential. The Box-Behnken is an excellent design model for response surface
The colloidal delivery system has proven to be competent to solve methodology as it allows: (i) evaluation of the quadratic model para-
the practical problems associated with solubility, modified release, and meters; (ii) construction of subsequent designs; (iii) recognition of lack
target specificity; nanoemulsion is one of them. The topical drug de- of fit of the model; (iv) make use of blocks v) avoid experimentation
livery based on nanoemulsion has excellence such as small droplet size under extreme conditions, for which unsatisfactory results might occur;
(50–1000 nm), a larger surface area and free energy, high drug loading vi) fewer number of experimental runs where number of factors are
capacity, protection of drug candidates, controlled or sustained release three.
of drugs, excellent drug permeation. However, due to the low viscosity Three factors for this study were designated as A (concentration of
of nanoemulsion, it has low retention at the application site and is also oil), B (concentration of surfactant), and C (concentration of co-sur-
inconvenient to use; it is often included in a gel system [19,21–23]. factant) and arranged into three levels, coded as +1, 0, -1 for high,
Therefore, for the treatment of RA, the nanoemulsion- based gel intermediate and low values respectively. Selection of oils, surfactants,
system containing QCT is proposed in the present study to improve the and co-surfactants were carried out by solubility and their emulsifica-
therapeutic efficacy of QCT which is evaluated on a CFA- induced tion capacity. Independent variables or factors selected as, (A) con-
Wistar rat arthritis model. centration of oil (10–20 %), (B) concentration of surfactant (3–9 %) and
(C) concentration of co-surfactant (3–9 %). Dependent variables or re-
2. Materials sponses selected as mean globule size (nm) and (%) entrapment effi-
ciency (% EE). A total of 17 experiments with 5 center points (to allow
QCT was purchased from Otto Chemie (Mumbai, India). Oleic acid, the estimation of pure error) were designed by the software and ex-
arachis oil, castor oil, sesame oil, coconut oil, soy oil, olive oil, sun- periments were performed in random order. Table 1 shows the in-
flower oil, isopropyl myristate (IPM), tween 20 (Polysorbate-20), dependent coded variables. For the 17 batches generated, formulations
Polyethylene glycol-400 (PEG-400), propylene glycol and Transcutol P were developed and further analyzed.
were obtained from Loba Chemie Pvt. Ltd. (Mumbai, India). Cremophor
RH 40 was obtained from BASF India Ltd. (Mumbai, India). Capryol 90 3.3. Optimization of formula
was obtained from Gattefosse (France). Carbopol 940 was obtained
from Noveon India Ltd. (Mumbai, India). Diclofenac sodium was re- The basic intension of BBD was to optimize the oil, surfactant and
ceived as a gift sample from Novartis Pharmaceuticals Ltd. (Mumbai, co-surfactant ratio. Out of 17 runs generated from design, formulations
India). All the solvents required for the experimental purpose were consisting of least amount of surfactant and co-surfactant were selected.
received from Merck Ltd. (Mumbai, India) and were of analytical grade The selected formulations were subjected to thermodynamic stability
and GRAS category. testing such as heating-cooling cycles, centrifugation, and freeze-thaw
cycles. During the testing, parameters such as drug precipitation, opa-
3. Methods lescence, creaming, and phase separation were observed. The particular
formulations were further examined for globule size and drug entrap-
3.1. Solubility studies ment efficiency and optimized for further processing. The optimized
batch was selected on the basis of relative transparency, clarity, globule
The solubility studies have been carried out to select the most ap- size and % entrapment efficiency (% EE).
propriate oil phase for the development of the formulation. The solu-
bility of QCT was determined in a variety of oils such as oleic acid, 3.3.1. Thermodynamic stability studies
coconut oil, castor oil, olive oil, arachis oil, sesame oil, sunflower oil, 3.3.1.1. Heating–cooling cycles. The intension behind to perform
soy oil, IPM. Similarly, surfactants (tween 20, Cremophor RH 40, and heating- cooling cycles was to observe the effect of temperature
tween 80) and co-surfactants (PEG 400, propylene glycol, and variations on the nanoemulsion stability. Six cycles between 4 and
Transcutol P) were tested. Solubility was estimated by dissolving an 40 °C were performed with storage at not less than 48 h at each
excess quantity (1gm) of QCT in 2 mL of each of the selected oils, temperature. The formulations that were found stable at these
surfactants and co-surfactants. Combinations of oils were also utilized temperatures were subjected to centrifugation test.
for the estimation of solubility. The mixtures were subjected to vortex
mixing followed by orbital shaking at 37 ± 1.0 °C for 72 h. The equi- 3.3.1.2. Centrifugation test. The formulations were centrifuged for
librated samples were centrifuged at 3000 rpm; the supernatant was 30 min at 2000 rpm and evaluations were carried out for phase
filtered, and the filtrates were diluted with appropriate solvent. The separation, creaming or cracking. Formulations that were found
solubility of QCT was measured at 258 nm by using validated UV stable were selected and subjected for freeze- thaw stress test.
spectrophotometric method (UV 1700, Shimadzu, Japan). All estima-
tions were done in triplicates [24]. 3.3.1.3. Freeze–thaw cycle (accelerated ageing). The formulations were
subjected to three freeze–thaw cycles at temperatures between −21
3.2. Formulation of nanoemulsion and +25 °C and were stored at not less than 48 h at each temperature.
For further studies, formulations that survived thermodynamic stability
Nanoemulsion was developed using spontaneous emulsification tests were selected [25].
technique. In brief, accurately weighed quantity of QCT (10 mg) was
added to the oil phase. Subsequently, surfactant and co-surfactant 3.3.2. Preparation of optimized batch of QCT-NE
(Smix) were then added and the mixture was vortexed for 15 min at Accurately weighed amount of oil phase- arachis oil and oleic acid
300 rpm. The prepared Smix then gradually added to the magnetically (15%), surfactant- tween 20 (6%) and co-surfactant- PEG-400 (6%)
stirred aqueous phase. Nanoemulsion formed rapidly. The complete were incorporated into a screw capped bottle on a vortex mixer at an
mixture was further vortex mixed for 20 min. optimal speed. In this Smix, QCT (10 mg) was dissolved and gradually
The resulting nanoemulsion was transparent and readily flowable added to the slightly magnetically stirred aqueous phase. Nanoemulsion
which was allowed to stand for 2 h for equilibration and characterized was rapidly formed. Then complete mixture was vortex mixed for

2
J.P. Gokhale, et al. Biomedicine & Pharmacotherapy 112 (2019) 108622

20 min. and further analysis was done.


Response Y2 (%EE) ( ± SD)

85.82 ± 0.03 3.4. Characterization of QCT loaded nanoemulsion


93.73 ± 0.31

91.12 ± 1.00
86.16 ± 0.61
91.86 ± 2.05
94.12 ± 0.87
95.76 ± 0.52
93.97 ± 0.15
87.18 ± 1.65
93.34 ± 1.83
92.55 ± 0.50
88.52 ± 1.74
90.47 ± 2.30
86.83 ± 0.39
94.65 ± 0.21
92.81 ± 0.51
3.4.1. Identification tests for optimized nanoemulsion
95.4 ± 1.09

3.4.1.1. Staining test. The oil soluble dye- Sudan Red IV, was added to
the drop of nanoemulsion, and observed under the optical microscope.

3.4.1.2. Dilution test. For the development of stable nanoemulsion,


accurate blend of surfactant and co-surfactant is essential. In order to
Response Y1 Globule size (nm) ( ± SD)

detect the stability, the nanoemulsion was diluted with 1:10 and 1: 100
ratios with double distilled water and phosphate buffer of pH 5.8 and
visually observed for any phase separation or cracking and clarity/
turbidity.

3.4.1.3. Electrical conductivity. Nanoemulsion conductivity (σ) was


1.21
0.12
1.32
0.43
0.56
1.48
2.14
0.29
0.81
1.39
1.78
2.51
0.02
0.92
0.59
0.72
1.82

estimated to confirm the type of nanoemulsion, whether it is O/W or


W/O nanoemulsion. For evaluation purposes, the conductivity meter
±
±
±
±
±
±
±
±
±
±
±
±
±
±
±
±
±
125.6
137.3
142.7
135.3
125.4
145.3
144.7
143.2
137.4
139.6
137.9
141.8
130.3
139.6
129.5
136.5
137.4

(306, Systronics, Ltd, India) consisting of Pt / platinized electrodes was


used.
All the estimates for the above mentioned parameters were per-
formed in triplicate.
Factor C Concentration of Co-surfactant (%)

3.4.2. Mean globule size, zeta potential, and morphology


The mean globule size, polydispersity index, and zeta potential were
characterized by photon correlation spectroscopy (PCS) using a
Malvern Zetasizer (Nano ZS 90, Malvern Ltd., UK). Prior to the mea-
surements, all the samples were diluted with double-distilled water,
mixed with vigorous shaking to produce appropriate scattering in-
tensity. The measurements were made at an angle of 90° at 25 °C
[26,27].
9.00 (+1)

9.00 (+1)

9.00 (+1)
9.00 (+1)
3.00 (-1)

3.00 (-1)
3.00 (-1)

3.00 (-1)
6.00 (0)

6.00(0)

6.00(0)
6.00(0)
6.00(0)

6.00(0)

6.00(0)
6.00(0)
6.00(0)

The morphology of nanoemulsion was studied by using a trans-


mission electron microscope (Jeol/JEM 2100, USA). A drop of nanoe-
mulsion was diluted with distilled water appropriately and applied to a
carbon- coated copper grid. The negative stain was used with 2%
Factor B Concentration of surfactant (%)

phosphotungstic acid and kept for 30 s. The grid was analyzed using the
transmission electron microscope with an accelerating voltage of
60–80 kV [28].

3.4.3. Viscosity, pH, refractive index


The viscosity of the nanoemulsion was estimated using Brookfield
viscometer (DV-E Brookfield Engineering Labs Inc., Middleboro, MA,
USA) with a spindle no.00 at 25 ± 0.5 °C [25]. An apparent pH of the
9.00 (+1)

9.00 (+1)

9.00 (+1)
9.00 (+1)
3.00 (-1)
3.00 (-1)

3.00 (-1)

3.00 (-1)

formulations was estimated at 25 ± 0.5 °C by a pH meter (μ pH system,


6.00(0)
6.00(0)
6.00(0)

6.00(0)

6.00(0)

6.00(0)

6.00(0)

6.00(0)
6.00(0)

362, Systronic, Ltd., India). The refractive index of the formulation was
estimated using the Abbe’s type of refractometer. All estimates for the
above parameters were made in triplicate.
Factor A Concentration of oil (%)
Factor level and response data for Box–Behnken study.

3.4.4. Degree of transparency, drug content, % EE


QCT- NE formulation was inspected by measuring percentage
Results are expressed as mean ± SD (n = 3).

transmission (% T) for optical transparency. One mL of the formulation


was diluted with methanol and spectrophotometrically analyzed at
258 nm [29].
(+1)

(+1)
(+1)
(+1)

In the formulation, QCT content was estimated using HPLC analysis


(-1)

(-1)

(-1)

(-1)
(0)

(0)

(0)
(0)
(0)
(0)

(0)

(0)
(0)

method. Briefly, 5 mL of nanoemulsion was dissolved in 10 mL of me-


10.00
15.00
20.00
15.00
10.00
20.00
20.00
20.00
15.00
15.00
15.00
15.00
10.00
15.00
10.00
15.00
15.00

thanol, centrifuged at 3500 rpm for 15 min. (REMI Instruments,


Mumbai, India) and analyzed by HPLC [30]. HPLC system (Younglin S.
K., Korea) consisted of a dual wavelength UV visible detector (UV 730 D
QCT-NE10
QCT-NE11
QCT-NE12
QCT-NE13
QCT-NE14
QCT-NE15
QCT-NE16
QCT-NE17
QCT-NE1
QCT-NE2
QCT-NE3
QCT-NE4
QCT-NE5
QCT-NE6
QCT-NE7
QCT-NE8
QCT-NE9

Shimadzu, Japan) and Autochro 3000 software (Kyungki-do, Korea)


was used. Chromolith® C18 reverse phase column (250 mm × 4.6 mm
Code

i.d., 5μ) was used with methanol: water (60:40 v/v, pH 7.0) as a mobile
phase, at a flow rate of 0.8 ml/min. All the measurements were carried
Table 1

out in triplicate.
Run

F10
F11
F12
F13
F14
F15
F16
F17
F1
F2
F3
F4
F5
F6
F7
F8
F9

Further, % EE was determined using the following formula [31].

3
J.P. Gokhale, et al. Biomedicine & Pharmacotherapy 112 (2019) 108622

%EE = ( Amount of drug content


Total amount of drug )× 100
(1)
matrix. About 0.01% w/v QCT-NE was dispersed in the gel with stirring
for 10 min at 500 rpm. The pH of the formulation was adjusted to 5–6
using triethanolamine and gel formulation was homogenized until de-
sired consistency [30].
3.4.5. In vitro drug release from QCT-NE
in vitro diffusion study of nanoemulsion (F16) was conducted by
3.6. Evaluation of QCT-NE gel formulation
Franz diffusion cell having a capacity of 10 mL. Dialysis membrane
(Avg. Mol wt. 12000 Da Himedia, India) was used as diffusion mem-
3.6.1. pH and rheological measurements
brane and was soaked in phosphate buffer saline (PBS) having pH 5.8,
The topical gel should be non-irritating and skin- pH compatible to
for 24 h prior to experiment. Diffusion cell was filled with PBS pH 5.8
prevent any allergic reactions or irritation. Thus, measurement of the
and dialysis membrane was mounted on cell. The temperature was
pH of the gel formulation is essential. Gel pH was measured using pH
maintained at 37 ± 0.5 °C and magnetically stirred at 100 rpm. After a
meter, (μ pH system, 362, Systronics, Ltd., India) calibrated using pH
pre-incubation time of 20 min., QCT-NE was placed in the donor
4.0, 7.0 and 9.0 standard buffers prior to use. The rheological study was
chamber. The samples (0.5 mL) were withdrawn at regular pre-
conducted with the Brookfield Viscometer (DV-E Brookfield
determined time intervals over a period of 4 h (0, 1, 2, 3, and 4 h) and
Engineering Labs Inc., Middleboro, MA, USA). The viscosity of QCT-NE
replaced with the same amount of fresh PBS (pH 5.8) to maintain sink
gel was evaluated at rotational speeds of 5, 10, 20, 30, 60 and 100 rpm
condition and analyzed by HPLC at 258 nm [7]. The similar procedure
using spindle ≠ 7.
was carried out with free QCT suspension.

3.6.2. Texture profile analysis (TPA)


3.4.6. Stability studies
The texture profile analysis assists to estimate the mechanical
The optimized batch of the nanoemulsion was prepared in tripli-
properties of the gel such as consistency, firmness, cohesiveness, work
cates and kept for three months at 40 ± 2 °C/ 75 ± 5%RH. Samples
of adhesion, gumminess, deformation at hardness and springiness. The
were withdrawn after specific time intervals (0, 30, 60, and 90 days)
measurements were carried out by using a Brookfield CT3 Texture
and visually examined for any physical changes in the formulation. The
analyzer (Brookfield Engineering Labs Inc, Middleboro, USA). A conical
selected criteria for the stability assessment were phase separation, pH,
shape sample holder was filled evenly with the gel sample. The probe
globular size, polydispersity index, zeta potential and drug content.
(TA3/100) was programmed to move down into the sample at a speed
Secondary parameters were also determined, such as refractive index
of 0.5 mm/s with a target value 20 mm and then go up back at the same
and viscosity [32].
speed to its original position. The TPA settings were as follows: pre-test
speed- 2.0 mm/s, test speed- 0.5 mm/s, return speed- 0.5 mm/s, target
3.4.7. Cytotoxicity study mode distance- 10 mm, trigger load- 3 g trigger type – auto, data rate-
In this study, synoviocytes HIG-82 cell lines were used to investigate 20 points per second. The force generated by the probe to break away
the cytotoxicity of the QCT- NE. The cell growth medium comprised of from the gel when starting to ascend (the point of maximum force) was
90% Ham’s F12 medium with 10% fetal bovine serum (FBS). To ex- measured [35,36]. Adhesiveness, cohesiveness, hardness, gumminess,
amine cell proliferation process, MTT [(3-(4, 5-dimethyl thiazolyl-2)-2, springiness were evaluated. The measurements were carried out in
5-diphenyltetrazolium bromide)] assay technique was used. The triplicate. The calculations were performed with the software provided
number of viable cells present in the cell suspension was calculated by along with the instrument and the results were expressed as mean ±
trypan blue exclusion method using hemocytometer. For the assay, SD.
about 5 × 104synoviocytes were seeded and loaded in each well of 6-
well culture plate and kept for 24 h in CO2 incubator at 37 °C with sa- 3.6.3. Ex vivo permeation studies
turated humidity 5% CO2. On a subsequent day, medium was replaced Ex vivo skin permeation studies were performed with a Franz dif-
with free QCT, QCT-NE, and DCS (diclofenac sodium) suspension pre- fusion cell (cell volume 10 mL) having diffusion area of 1.00 cm2,
pared using 10 mL Ham’s F12 medium. Each suspension was added at diameter 11.28 mm using Wistar rat abdominal skin. The skin was ex-
various concentrations of 20,40,60,80 and 100 μM. After 48 h, the cised, washed and stabilized using phosphate buffer in both the com-
medium containing suspensions were discarded, washed with PBS and partments with magnetic stirring for 30 min. After 30 min, solutions
then MTT assay was carried out. To measure the cell viability, the ab- from both the compartments were withdrawn and replaced with fresh
sorbance was measured at the wavelength of 562 nm using ELISA mi- phosphate buffer having pH 5.8. A weighed quantity of QCT-NE gel and
croplate reader (ELx 800, Biotek). The experiments were carried out in free QCT gel (equivalent to 10 mg of QCT) was applied to skin mem-
triplicates, and the results were expressed as mean ± SD [18,33]. brane placed on donor compartment. About 0.5 mL samples were
withdrawn at predetermined time intervals over a period of 24 h (0, 2,
3.4.8. Effect on production of TNF-α 4, 6, 8, 10, 12 and 24 h) from receptor compartment, filtered through
In this study, the effect of QCT-NE on the production of TNF-α was 0.45 μm membrane filter and analyzed for QCT content by using HPLC
investigated on RAW264.7 cells (macrophage cells isolated from mouse at 258 nm. Replacement with fresh medium was ensured to maintain a
blood). The cells were cultured with DMEM (Dulbecco’s Modified constant volume and sink condition [37]. The permeation profile was
Eagle’s Medium - high glucose) + 2 mM Glutamine + 10% Foetal constructed by plotting amount of drug permeated per unit skin surface
Bovine Serum (FBS). RAW264.7 cells seeded at the density of 1 × 105/ area (mcg/cm2) Vs. Time (h). The steady state flux (Jss, mcg/cm2h) was
well were preincubated with QCT, QCT-NE, and DCS for 2 h. calculated from the slope of the linear portion of the plot using linear
Lipopolysaccharide (LPS) was added to the cells for 20 h. The level of regression analysis. All measurements were carried out in triplicate.
TNF-α secreted was measured in the cell supernatants using a sandwich
ELISA kit by following the manufacturer’s experimental protocols. 3.6.4. Antiarthritic activity [Complete Freund’s adjuvant (CFA) model]
Absorbance was read by an ELISA microplate reader (ELx 800, Biotek) All animal experiments were approved and performed following the
at 450 nm. The experiments were carried out in triplicates, and the guidelines by Institutional Animal Ethical Committee of R.C. Patel
results were expressed as mean ± SD (n = 3) [34]. Institute of Pharmaceutical Education and Research, Shirpur, registered
under CPCSEA, India, registration number 651/PO/ReBi/S/02/CPCSEA
3.5. Preparation of QCT-NE based gel formulation and protocol number IAEC/ RCPIPER/ 2014-15/04. Male Wistar rats
(six to seven weeks old), with average body weight of 140 ± 15 g,
A gel was formulated using Carbopol 940 (1.0% w/v) as the gel were randomly divided into four groups of six animals each. The

4
J.P. Gokhale, et al. Biomedicine & Pharmacotherapy 112 (2019) 108622

animals in each group were injected with 0.1 mL of CFA into the sub- performed using Student’s t-test and one-way ANOVA. In all cases, a
plantar region of the left hind paw. This day was considered as day ‘0′. value of p < 0.05 was considered as statistically significant.
Group I served as untreated arthritis- CFA control, Groups II, III, and IV
animals were treated with free QCT gel, QCT-NE gel (equivalent to 4. Results and discussion
10 mg of QCT), and standard diclofenac gel; respectively. Dosing was
done twice a day and continued for 28 days, including the day of in- 4.1. Solubility studies
oculation. The animals were housed in a controlled environment (at
temperature 20 °C ± 1 °C and 52% ± 15% RH) and had free access Usually, the oil phase was selected on the basis of highest solubility
with standard pellet diet and water ad libitum [38,39]. for QCT and suitability for the topical application. In the present study,
various types of oils were selected to estimate the solubility of QCT.
3.6.5. Evaluation of the severity of Arthritis QCT showed the highest solubility in arachis oil (11.14 mg/mL) among
3.6.5.1. Measurements of paw volume. The volume of the left hind paw the oils tested and therefore was selected as an oil phase. Secondly, the
was measured using plethysmometer at regular interval of 0, 7th, 14th, highest solubility was in oleic acid (9.10 mg/mL), which was selected as
21st and 28th day. Changes in the paw volume were determined by the solubility and permeation enhancer for QCT in an oil phase.
measuring the paw volume (mm) at the initial day (Vo) and a particular Solubility efficiency and HLB value were considered for the selec-
day (Vt) [28]. tion of surfactant and co- surfactant. Tween 20, having HLB value 16.7
and QCT solubility 9.28 mg/mL was selected as the surfactant. PEG-
3.6.5.2. Measurement of Arthritis index and joint stiffness. Arthritic index 400, having HLB value 11.4 and QCT solubility 13.59 mg/mL, was se-
(AI) scores were allotted to ankle joints of the rats of each group. lected as the co-surfactant [38,45].
Scoring was performed on a 0–4 scale as follows: 0- no swelling or
erythema, 1- slight swelling and/or erythema, 2- low to moderate 4.2. An experimental design using Box–Behnken model
edema, 3- pronounced edema with limited joint use and 4- excess
edema with joint stiffness. For the development and optimization of the formulation, a three-
The scoring of the joint stiffness was carried out in accordance with factor, three-level BBD was applied using Design-Expert® 7.0.0 software
the assessment scale reported by Butler et al. [41]. The body of the rats (Stat-Easc, Inc., Minneapolis, MN, USA). The independent variables
was held from the back, bending and extension (once in each direction) (factors) were selected as - (A) concentration of oil (%), (B) con-
of the ankle within its limit was observed. Scoring was given as per centration of surfactant (%), (C) concentration of co-surfactant (%). The
scale- Score 2: when there was a restriction of full range of movement of dependent variables (responses) were selected as- (Y1) mean globule
the ankle in both bending and extension. Score 1: there was a restriction size (nm) and (Y2) (%) EE. Table 1 shows the factor levels and response
of full range of movement of the ankle in either bending or extension. data for BBD.
Score 0: no restriction [39,41]. The analysis of experimental results was carried out by using
Design-Expert® 7.0.0 software. After compilation of the data, F- values,
3.6.5.3. Measurement of hematological parameters. The blood markers p-values, and model F-value for the mean globule size and % EE were
provide additional information for the therapeutic evaluation of drugs obtained from ANOVA. The value of responses Y1 (mean globule size)
in RA. Therefore, the parameters such as RBCs, WBCs, erythrocyte and Y2 (% EE) ranges from 125.4 to 145.3 nm and 85.82 to 95.76%
sedimentation rate (ESR), hemoglobin (Hb), C- reactive protein (CRP) respectively. The ratio of the maximum to the minimum for responses
and rheumatoid factor (RF) were evaluated [38]. Y1 and Y2 were 1.1586 and 1.1158 respectively. Therefore power
For the estimation of RBCs, WBCs, Hb and ESR, the blood sample transformation of values is not required for mean globule size and % EE.
was collected through vein puncture and stored in a test tube with the Transformation of response is an essential component of data analysis.
appropriate anticoagulant. The cell counting was performed with the Transformation is required if the error (residuals) is a function of the
help of Automated Hematology Analyzer (Medonic M32B cell counter). magnitude of response (predicted values). In simpler terms, power
CRP and RF were evaluated on the basis of latex agglutination method transformation of responses is required when the ratio of maximum to
as per the test kit manufacturer’s instructions (Microsidd India Pvt. Ltd., minimum response is higher than 10. For ratios less than 3, the trans-
Bengaluru, India). formation has little effect. The selection of the model for analyzing the
responses was made based on the sequential model sum of squares, lack
3.6.6. Skin irritation studies of fit and model summary statistics. The Prob > F value of
The possibility of skin irritation on the application of QCT gel and p < 0.0001, low standard deviation, high R2 and lower predicted re-
the QCT-NE gel was evaluated by carrying out a skin irritation test on sidual error sum of square (PRESS) value suggests to select the quad-
Wistar rats. The rats were acclimatized to the conditions for seven days ratic model for both responses. ANOVA of the data confirms that the
prior to the commencement of the study. The dorsal surface of the rats model was significant (Model Prob > F < 0.05). The Model F value
was made hairless without damaging the skin surface, 4 h before the for response Y1 and Y2 was 921.97 and 18.63 respectively, which im-
experiment. The rats were divided into four groups each containing plies model is significant. ANOVA identifies the significant factors that
three rats: Group I was treated with 0.8% v/v aqueous solution of affect the responses. For mean globule size, oil, surfactant, and co-
formalin, group II was treated with a placebo gel, group III was treated surfactant were identified as significant model terms whereas, for % EE,
with free QCT gel, and group IV was treated with QCT-NE gel. The oil was found to be the significant model term. Lack of fit F-value for Y1
formulations (gel containing 10 mg equivalent amount of QCT) were was found to be 1.65 which implies lack of fit was not significant re-
topically applied to the hairless skin area (1cm2) and were inspected at lative to the pure error, and for Y2 it was 10.64 which imply lack of fit
24, 48 and 72 h for dermal reactions such as erythema and edema. The was significant relative to the pure error. The multiple regression terms
mean scores for erythema and edema were recorded on the basis of were also analyzed. The predicted R2 values for response Y1 and Y2
their degree of severity caused by application of formulations: 0-no were 0.9919 and 0.9032 respectively. Adjusted R2 values for response
erythema/edema, 1- slight erythema/ edema, 2- moderate erythema/ Y1 and Y2 were 0.9981 and 0.9084 respectively. The predicted R2 value
edema and 3- severe erythema/ edema [42–44]. was found to be in reasonable agreement with adjusted R2 value, which
indicates that the model has predicted the responses well. Adeq preci-
3.6.7. Data analysis sion for response Y1 and Y2 were 96.139 and 13.867 respectively. Adeq
All experiments were performed in triplicate (n = 3), and the data precision measures the signal to noise ratio. A ratio higher than four is
were expressed as mean value ± SD. Statistical data analyses were desirable. The ratio of 96.139 and 13.867 indicates an adequate signal.

5
J.P. Gokhale, et al. Biomedicine & Pharmacotherapy 112 (2019) 108622

Fig. 1. (a) Effect of conc. of oil and conc. of surfactant on globule size, (b) effect of conc. of oil and concentration of co-surfactant on globule size, (c) effect of conc. of
surfactant and co-surfactant on globule size. In case of mean globule size, it was found to increase with increase in the (%) concentration of oil, surfactant, and co-
surfactant.

This model can be used to navigate the design space. Final equations in of drug release but also the absorption.
term of coded factors for responses Y1 and Y2 were as follows: Perturbation graphs were plotted to locate the factors that affect the
response [see Fig. 3(a), (b)]. A steep slope or curvature in a factor
(Y 1) Mean globule size = + 137.52 + 10.11 * A + 1.10 * B + 1.21 * C shows that response is sensitive to change in that factor; whereas re-
0.88 * A * B 0.90 * A * C 0.52 * B * C latively flat line shows insensitivity to the factor. If there are more than
0.46 * A2 + 0.16 * B2 + 1.39 * C (2) two factors, a perturbation plot could be beneficial to locate the factors
having maximum influence on the response. For response Y1, factor A
indicates noticeable bend or steep curvature. Factor B and C indicate a
(Y 2) % EE = + 93.47 + 3.73 * A + 0.47 * B 0.34 * C 0.58 * A * B
slight bend, which implies that the concentration of oil effects mean
1.56 * A * C + 1.50 * B * C 1.34 * A 1.41 * B 2 globule size. For response Y2, factor A, B, and C show a noticeable bend
1.73 * C 2 (3) or steep curvature. It specifies that the concentration of oil, surfactant,
and co-surfactant shows significant changes in % EE.
Where A- Concentration of oil (%), B- Concentration of surfactant (%)
Optimization of nanoemulsion was carried out to find the level of
and C- Concentration of co-surfactant (%)
factors A, B, and C, which gives Y1 in the range of 125.4–145.4 nm and
In case of response Y1, i.e. globule size, positive coefficients of A, B,
Y2 in the range of 85.82–95.76 %. The model predicted Y1 and Y2 in
and C indicate mean globule size was found to increase with increase in
the required range at A, B and C values of 12.47%, 8.79%, and 8.66%
the concentration of oil, surfactant, and co-surfactant [see
respectively. The predicted value of responses Y1 and Y2 were
(Fig. 1(a)–(c)]. For response Y2, i.e. % EE, positive coefficients of A and
137.1 nm and 91.01%. By using these values of factors, three different
B indicates % EE was found to increase with increase in the con-
batches of nanoemulsion were prepared. The actual values of Y1 and Y2
centration of oil and surfactant and the negative coefficient of C in-
were found to be 136.8 ± 1.2 nm and 93.7 ± 1.9%, which is in close
dicates % EE decreases with increase in co-surfactant concentration
agreement with the predicted values.
[see Fig. 2(a)–(c)]. Very high concentrations of oil and Smix resulted in
All the batches conducted as per BBD, were subjected to thermo-
the formation of turbidity or microemulsion rather than nanosized
dynamic stress tests. Amongst all the batches F2, F3, F5, F7, F9, F11,
globules. The mean globule size of the nanoemulsion is a critical factor
F16 and F17 passed the thermodynamic tests. It was observed that F3
in emulsification development as it affects not only the rate and extent

Fig. 2. (a) Effect of conc. of oil and concentration of surfactant on % EE, (b) effect of conc. of oil and conc. of co-surfactant on % EE, (c) effect of conc. of surfactant
and co-surfactant on % EE. In case of %EE, it was found to increase with increase in the (%) concentration of oil and surfactant and decreases with increase in (%)
concentration of co-surfactant.

6
J.P. Gokhale, et al. Biomedicine & Pharmacotherapy 112 (2019) 108622

Fig. 3. Perturbation graph for effect of individual factor (a) on response Y1 (globule size) and (b) on response Y2 (% EE).

passes the conductivity test due to water in continuous phase while,


w/o nano-emulsion vice-versa. The conductivity (σ) of QCT-NE
formulation was found to be 489.53 ± 1.08 μS/cm. This indicates
that the formulation was o/w type of nanoemulsion (n = 3).

4.3.2. Mean globule size, zeta potential, and morphology


The mean globule size of an optimized nanoemulsion was found to
be 136.8 ± 1.2 nm, and polydispersity index was 0.265 ± 0.3. The
mean globule size below 200 nm, having a polydispersity index below
one is considered ideal for topical delivery, as it would provide a large
surface area in addition, appropriate for rapid pore transport. The ne-
Fig. 4. Morphology of QCT loaded nanoemulsion globules visualized under gative zeta potential value -25.4 ± 1.7 mV was considered satisfactory
Transmission electron microscopy (TEM) voltage of 60–80 kV (Magnification in order to prevent coalescence and maintain the interfacial boundary
×10,000, scale of 200 nm). of the nanoemulsion droplets resulting in better colloidal system sta-
bility [13].
had greater amount of oil and larger globule size (i.e. 142.70 nm) as In the morphological studies (Fig. 4), the globules were found to be
distributed evenly with uniform sizes. Also, were spherical with a
compared to F5 having lowest % EE (86.16%). Even having higher %EE
(94.12%), F7 has largest globule size (144.7 nm). So, by comparing the smooth, flexible boundary, non-aggregated showing their stability
against Oswald ripening due to globular collapse.
selected batches F16 batch was selected for further processing which
having lower globule size (137.6 nm) and higher %EE (94.49%). Hence,
the best suited composition of the optimized formulation was selected 4.3.3. Viscosity, pH, refractive index
as oil phase (15%), surfactant (6%) and co-surfactant (6%) i.e. (F16). The viscosity of the nanoemulsion was found to be 89.5 ± 1.41 cP.
Optimization was done on the basis of globule size and %EE. The pH was observed to be 5.61 ± 0.07 which is entirely compatible
The results of ANOVA for the dependent variables demonstrated with the pH of the skin, which would therefore be non-irritant.
that the model was significant for the mean globule size and the %EE. In Refractive index is the net value of the components of nanoemulsion
case of mean globule size, the model F-value of 921.97 implies the and indicates the isotropic nature of the formulation. RI value of na-
model is significant. The value p < 0.05 indicates model terms are noemulsion was found to be 1.379 ± 0.041, which reveals transpar-
significant. In this case, A, B, C, AB, AC, BC, A2, B2, C2 are significant ency of nanoemulsion formulation.
model terms. Values higher than 0.1000 indicates the model terms are
not significant. In the case of %EE, The model F-value of 18.63 implies 4.3.4. The degree of transparency, drug content, %EE
the model is significant. The value p < 0.05 indicates model terms are Percentage transmittance of nanoemulsion was found to be
significant. In this case, A, AC, BC, A2, B2, C2 are significant model 97.68 ± 0.05%. The value closer to 100% indicates clarity and trans-
terms. parency of formulation.
The drug content in nanoemulsion was found to be 95.65 ± 0.14%;
4.3. Characterization of optimized QCT-NE formulation the batch was therefore selected for the development of gel formula-
tion.
4.3.1. Identification tests for nanoemulsion Entrapment efficiency of the optimized nanoemulsion was found out
4.3.1.1. Staining test. The optimized formulation (F16) was found to be 94.65 ± 0.14%.
o/w type nanoemulsion. For staining, oil soluble dye was used. The
background was colorless and the oil globules were found red in color. 4.3.5. In vitro drug release
The nanoemulsion is an excellent tool for enhancing the solubility of
4.3.1.2. Dilution test. Visually, QCT-NE was observed clear and hydrophobic drugs thereby the bioavailability of the drug owing to
transparent with bluish tinge. Furthermore, the diluted formulation small-scale globule size. In order to achieve better drug release and
showed no sign of phase separation or cracking. therapeutic effect, drug was distributed into the nanoemulsion globules.
Therefore, the release of QCT from the nanoemulsion globules as
4.3.1.3. Electrical conductivity. Electrical conductivity is directly compared to free QCT suspension was observed. At the end of 4 h,
proportional to the percentage of water. The o/w nano-emulsion 28.23 ± 1.72% of drug was released from free QCT suspension which

7
J.P. Gokhale, et al. Biomedicine & Pharmacotherapy 112 (2019) 108622

Table 2
Stability study of QCT-NE on storage.
Parameter 0 days 60 days 120 days 180 days

pH 5.81 ± 0.03 5.88 ± 0.17 5.97 ± 1.20 6.08 ± 1.53


Globule size (nm) 137.6 ± 1.38 138.66 ± 0.77 142.32 ± 1.98 146.2 ± 2.12
PDI 0.273 ± 1.52 0.286 ± 0.03 0.289 ± 1.051 0.331 ± 0.73
Zeta potential (mV) −25.18 ± 1.97 −25.32 ± 0.44 −26.32 ± 0.67 −27.3 ± 0.23
Refractive Index 1.379 ± 0.041 1.390 ± 0.007 1.461 ± 0.003 1.553 ± 0.123
Viscosity (cP) 89.5 ± 1.41 86.62 ± 1.51 86.43 ± 0.96 86.97 ± 1.21
% Drug content 95.65 ± 0.14 95.40 ± 0.52 94.92 ± 1.02 94.78 ± 1.36

Data is expressed as mean ± SD (n = 3), (p > 0.05).

was considerably lower (p = 0.002) than the drug was released from 4.4. Characterization of QCT-NE gel
QCT- NE (84.12 ± 0.51%). Higher release from nanoemulsion can be
attributed to smaller globule size, which increases the surface area for 4.4.1. pH and rheological measurements
diffusion. Hence it can be stated that, QCT-NE not only improved the The apparent pH of QCT-NE gel was found to be 5.81 ± 0.03. The
solubility of QCT but also the diffusion rate owing to its lipoidal nature pH is slightly acidic and is physiologically compatible with the pH of
and small globule size. the skin, hence would be non-irritant. The viscosity of QCT-NE gel was
found to be 71,210 ± 1.12 cP at 5 rpm. Further the viscosity of QCT-
NE gel was found out decreased with the increase in rpm. In fact, with
4.3.6. Stability studies the increase in rpm, the internal structure of the gels began to disrupt as
The stability of QCT-NE in terms of globule size, PDI, zeta potential, the contact points broke. The particles were aligned until they started to
viscosity and refractive index was estimated at 0, 60, 120 and 180 days flow and their consistency gradually decreased with rpm. The gel ex-
on storage at specific temperatures. These parameters shown varying hibited pseudoplastic flow behavior. However, no evidence of thixo-
trend with respect to time, nevertheless the changes in the studied tropy was observed. To calculate the flow index, the power law model
parameters were not statistically significant (p > 0.05). The results was applied to the rheological properties of the gel formulation. The
were expressed as mean ± SD (n = 3) (Table 2). flow behavior index of QCT- NE gel was found to be 0.415 ± 0.35
confirming the shear thinning behavior of the gel.

4.3.7. Cytotoxicity study 4.4.2. Texture profile analysis


The relative growth rate of cells at 48 h in free QCT and QCT-NE Formulations intended for topical application, must show accep-
was found to be 104 ± 3.2% and 130 ± 0.7%, respectively. The re- table mechanical characteristics, e.g., ease of application, stickiness,
sults indicate that the presence of QCT-NE does not inhibit the growth firmness or hardness, etc. These mechanical properties of QCT-NE gel
of synoviocytes. were studied by texture profile analysis. Results of the texture profile
analysis were generated by Texture PRO CT 1.4 build 17 version soft-
ware.
4.3.8. Effect on production of TNF-α Adhesiveness value of QCT- NE gel was found to be 0.3 ± 0.61 mJ.
In the pathogenesis of RA, TNF-α plays a significant role in the The value near to zero implies that QCT- NE gel is more adhesive and
proliferation of fibroblast, stimulation of proinflammatory mediators has a soft texture.
like PGE2, IL-1β. Thereby it promotes synovitis and stimulates cartilage The cohesiveness (consistency) indicates the strength of internal
and bone breakdown [38]. bonds that make up the gel sample and the extent to which the gel
LPS induced TNF-α secretion from RAW 264.7 cells was sig- sample can be deformed before the application. The cohesiveness of the
nificantly reduced post QCT-NE treatment as compared to free QCT sample is shown by the maximum negative force. Cohesiveness value
suspension (Fig. 5) (p = 0.041).This finding shows that the anti-ar- was found to be 0.79 ± 1.98.
thritic effects of QCT-NE complex may be mediated by the inhibition of Gumminess is the product of hardness and cohesiveness. It is the
inflammatory cytokines like TNF- α. The inhibition of TNF-α produc- critical parameter of gel system with the low hardness and the high
tion is linked with suppression of inflammation and cartilage destruc- cohesiveness extent. The higher gumminess value implies more
tion which can slow down the arthritis progression. sample hardness. Gumminess of the QCT-NE gel was found to be
15 ± 1.34 mg.
The hardness is related to the strength of the compressed gel
structure and is the maximum force during the first compression cycle.
In sensory aspects, it is the maximum force required to apply the gel
system on the skin surface. Higher the value of hardness implies thicker
consistency of the gel system. The hardness value of QCT- NE gel was
found to be 19 ± 0.12 g.
Springiness is the rate at which a deformed sample reverses to its
undeformed state after the removal of deforming force. The springiness
value of QCT-NE gel was found to be 2.73 ± 1.32 mm. The high value
of springiness indicates that additional force is required for the appli-
cation of the gel system. [40].

4.4.3. Ex vivo permeation studies


Fig. 5. Effect of QCT concentration in formulations on TNF-α level Results are Ex vivo permeation study showed that the drug from QCT- NE gel
expressed as mean ± SD (n = 3).Results were found statistically significant permeated rapidly than free QCT gel. At the end of 24 h,
(p = 0.041) as compared to free QCT suspension. 62.51 ± 0.34% and 35.87 ± 0.21% of the drug was permeated from

8
J.P. Gokhale, et al. Biomedicine & Pharmacotherapy 112 (2019) 108622

Fig. 6. Effect of QCT-NE gel on rats following CFA induced arthritis (a) effect on arthritic index, (b) effect on stiffness score (c) effect on paw circumference. [Results
were expressed as mean ± SD (n = 3). Results were found statistically significant (p < 0.05) as compared to CFA-control group].

Table 3
Alterations in hematological parameters, CRP and RF in CFA-induced arthritis in rats.
Group RBC (×106/mm3) WBC (×103/mm3) ESR (mm/h) Hb (mg%) CRP (mg/dL) RF (IU/mL)

Group I-CFA control 7.4 ± 0.5 12 ± 0.9 15 ± 0.5 11.5 ± 1.2 8.7 ± 0.2 71 ± 1.3
Group II- QCT- gel 8.2 ± 0.6 7.9 ± 0.5 10.5 ± 0.5 13.7 ± 0.2 3.6 ± 0.8 34 ± 1.9
Group III- QCT-NE gel 8.8 ± 0.4 5.6 ± 1.5 9.9 ± 1.13 15.0 ± 0.1 2.1 ± 0.3 23 ± 0.2
Group IV- Std. DCS gel 8.9 ± 1.7 5.7 ± 0.13 9.2 ± 1.4 14.3 ± 0.5 1.9 ± 0.9 22 ± 1.6

Data is expressed as (mean ± SD, n = 3) (p < 0.05) when compared to CFA- control group.

QCT- NE gel and free QCT gel respectively. Permeability coefficient of arthritic index and stiffness score of the CFA-control group were
free QCT gel and the QCT-NE gel was found to be 1.6 cm2/ min and 3.7 ± 1.61 and 2.1 ± 1.53 respectively, which were reduced by
2.7 cm2/ min respectively. This study showed a significant improve- QCT-NE gel treatment i.e.1.6 ± 1.27 and 0.73 ± 0.42 respectively.
ment (p = 0.04) of cumulative permeation of QCT from nanoemulsion 4th–14th days mark the proliferative phase which is characteristic of
gels compared to the gel containing free QCT. The nanoemulsion due to inflammation, hyperplasia and macrophage activities in the synovial
its small droplet size (< 200 nm), significantly increases the rate of membrane. The results of arthritic index and stiffness score were
permeation since the nanosized droplets can transfer the drug through found statistically significant in case of QCT-NE gel as compared to
the skin barrier and easily move into the stratum corneum. In addition, CFA-control group (p = 0.003 and 0.02 respectively). Results of the
the Smix increases the solubilization capacity and permeability coeffi- 21st and 28th day evaluation showed that the QCT-NE gel treatment
cient of the skin as the interfacial barrier decreases and partitioning significantly reduced CFA-induced arthritic lesions such as increased
improves due to small droplet size. Water in the gel system hydrates the paw circumference, erythema, swelling, joint stiffness and obstruction
skin and causes the cells to swell in the stratum corneum, widening in the movement, as compared to the CFA- control group. Results of
drug channels, leading to improved cumulative permeation [26,28]. the arthritic index, stiffness score and paw circumference of group I to
IV are shown in Fig. 6.
4.4.4. Antiarthritic activity
4.4.4.1. Measurements of paw volume. After two weeks of CFA 4.4.4.3. Measurement of hematological parameters. In the CFA-control
administration, rheumatoid arthritis was evident in rats. A significant rats, the levels of RBCs and Hb were found to be decreased initially
increase in paw circumference, erythema, swelling, joint stiffness and which is one of the clinical manifestations of RA. The QCT-NE gel
hindrance in the movement was observed. Paw volume was recorded by formulation treated group showed significant increase in RBCs (8.8 ±
using digital plethysmometer on 7th, 14th, 21st and 28th day after CFA 0.4 × 106/mm3)) and Hb level (15.0 ± 0.1 mg %) as compared to
injection. The paw circumference of CFA control group was RBCs (7.4 ± 0.5 × 106/mm3) and Hb level (11.5 ± 1.2 mg %) of CFA-
71.21 ± 0.33 mm, which was found to be decreased up to control group (p = 0.007 and 0.005 respectively).
51.13 ± 1.35 mm with the QCT-NE gel. The inhibition of paw In the ESR test, the rapid sedimentation rate is an indication of the
volume by QCT-NE gel was found statistically significant as compared inflammation; which was found to be decreased from 15 ± 0.5 mm/h
to CFA-control group (p = 0.006). to 9.9 ± 1.13 mm/h with the QCT- NE gel treatment. An elevated WBC
count is also one of the characteristic arthritis diagnoses. After the
4.4.4.2. Arthritis index and stiffness score. The arthritic index is used to treatment with QCT-NE gel formulation, WBCs level was effectively
define the severity of joint inflammation based on a calculation of reduced from 12 ± 0.9×103/mm3 to 5.6 ± 1.5 × 103/mm3. Results
scores of the paws after arthritis induction [20]. Arthritis indices were were found to be statistically significant (p = 0.007) as compared to
recorded on the 7th, 14th, 21st and 28th day after CFA injection. The CFA-control group.

9
J.P. Gokhale, et al. Biomedicine & Pharmacotherapy 112 (2019) 108622

In recent years, CRP has been recognized as significant predictors for [10] M.K. Rao, B. Ghosh, Quercetin inhibits LPS-induced nitric oxide and tumor necrosis
RA diagnosis. CRP is a member of the pentraxin family of proteins, pro- factor-α production in murine macrophages, Int. J. Immunopharmacol. 21 (1999)
435–443.
duced by the liver in response to inflammation. It plays a significant role in [11] X. Cai, J. Dou, A. Yu, G. Zhai, Bioavailability of quercetin: problems and promises,
bone destruction process, as it is found in lining areas of RA synovium. RF Curr. Med. Chem. 20 (2013) 2572–2582.
is considered to be the main serological marker in the inflammatory ar- [12] Y. Gao, Y. Wang, Y. Ma, A. Yu, F. Cai, W. Shao, G. Zhai, Formulation optimization
and in situ absorption in rat intestinal tract of quercetin-loaded microemulsion,
thritis found in 80% of arthritis conditions. RF is an auto antibody directed Colloids Surf. B 71 (2009) 306–314.
against the Fc portion of IgG antibodies. RF and IgG join in forming im- [13] D.V. Ratnam, D.D. Ankola, V. Bhardwaj, D.K. Sahana, M.N.V. Kumar, Role of an-
mune complexes that contribute to the progress of RA [46]. Elevated levels tioxidants in prophylaxis and therapy: a pharmaceutical perspective, J. Control.
Release 113 (2006) 189–207.
of CRP and RF show systemic inflammation and antibody production [14] G.C. Yu, Y.C. Fen, L.Q. Lu, T. Qi, X.Y. Wei, L.W. Na, Z.G. Xi, Development of a
against the injected adjuvant, which has also been found to be significantly Quercetin-loaded nanostructured lipid carrier formulation for topical delivery, Int.
reduced by QCT-NE gel, as p = 0.032 and 0.004 respectively as compared J. Pharm. 430 (2012) 292–298.
[15] M.Y. Wong, N.C.G. Chiu, Liposome formulation of co-encapsulated vincristine and
to CFA-control group. All the results are summarized in Table 3.
quercetin enhanced antitumor activity in a trastuzumab-insensitive breast tumor
xenograft model, Nanomed.: Nanotechnol. Biol. Med. 7 (2011) 834–840.
4.4.5. Skin irritation studies [16] S. Bose, Y. Du, P. Takhistov, B.M. Kohn, Formulation optimization and topical
The topical application should be free of skin irritation and er- delivery of quercetin from solid lipid based nanosystems, Int. J. Pharm. 441 (2013)
56–66.
ythematic reactions. Conventional therapy has been linked to notice- [17] P. Alexandre, R.C.L. Dora, E.L. Andrade, J.S. Chaves, L.F.C. Silva, E.L. Senna,
able skin irritation, which actively restricts its applicability and patient J.B. Calixto, Anti-inflammatory effect of quercetin-loaded microemulsion in the
acceptability. In this study, the results revealed no severe skin irritation airways allergic inflammatory model in mice, Pharmacol. Res. 61 (2010) 288–297.
[18] V. Natarajan, P. Saravanakumar, B. Madhan, Collagen adsorption on quercetin
symptoms such as erythema (redness) and/or edema (swelling) within loaded polycaprolactone microspheres: approach for “stealth” implant, Int. J. Bio.
72 h of QCT-NE gel application. The developed formulation can there- Macromol. 50 (2012) 1091–1094.
fore be considered as non-sensitizing, non-irritating and safe for topical [19] T.P.U. Ravi, T. Padma, Nanoemulsions for drug delivery through different routes,
Res. Bio-technol. 2 (2011) 1–13.
use on the rat skin. [20] N. Anton, T. Vandamme, The universality of low- energy nano-emulsification, Int. J.
Pharm. 377 (2009) 142–147.
5. Conclusion [21] M.J. Lawrence, G.D. Rees, Microemulsion-based media as novel drug delivery
system, Adv. Drug Deliv. Rev. 45 (2000) 89–121.
[22] H. Chen, X. Chang, T. Weng, et al., A study of microemulsion systems for trans-
In conclusion, present research work emphasizes the development dermal delivery of triptolide, J. Control. Release 98 (2004) 427–436.
of a novel formulation through topical administration in order to [23] D. Gulsen, A. Chauhan, Dispersion of microemulsion drops in HEMA hydrogel: a
potential ophthalmic drug delivery vehicle, Int. J. Pharm. 292 (2005) 95–117.
overcome the drawbacks of QCT administration. QCT is extensively
[24] S. Baboota, F. Shakeel, A. Ahuja, J. Ali, S. Shafiq, Design, development and eva-
used phytoconstituent having a similar activity to the NSAIDs. Even luation of novel nanoemulsion formulations for transdermal potential of celecoxib,
though, it has limited applicability via topical route due to low skin Acta PharmaceuticaSinica B 57 (2007) 315–332.
permeability. Incorporation of oleic acid: arachis oil, tween 20 and [25] S. Shafiq, F. Shakeel, S. Talegaonkar, J. Ali, S. Baboota, A. Ahuja, R.K. Khar, M. Ali,
Formulation development and optimization using nanoemulsion technique: a
PEG-400 (15:6:6) resulted in the spontaneous formation of nanoemul- technical note, AAPS Pharm Sci Tech. 8 (2007) E1–E6.
sion and successful carrier system for QCT. The optimized formulation [26] R. Parveen, S. Baboota, J. Ali, A. Ahuja, S.S. Vasudev, S. Ahmad, Oil based nano-
showed improved physicochemical stability, acceptable mechanical carrier for improved oral delivery of silymarin: in vitro and in vivo studies, Int. J.
Pharm. 413 (2011) 245–253.
properties and enhanced skin permeability thus increasing the ther- [27] V. Bali, M. Ali, J. Ali, Study of surfactant combinations and development of a novel
apeutic efficacy, as revealed by its antiarthritic activity. Hence the nanoemulsion for minimising variations in bioavailability of ezetimibe, Colloids
studies have confirmed that QCT-NE gel will be a promising alternative Surf. B: Biointerface 76 (2010) 410–420.
[28] Z. Naz, F.J. Ahmad, Curcumin-loaded colloidal carrier system: formulation opti-
in rheumatic complications via topical application. mization, mechanistic insight, ex vivo and in vivo evaluation, Int. J. Nanomed.
Nanosurg. 10 (2015) 4293–4307.
Declaration of interest [29] N. Morsi, M. Ibrahim, H. Refai, H.E. Sorogy, Nanoemulsion-based electrolyte trig-
gered in situ gel for ocular delivery of acetazolamide, Eur. J. Pharm. Sci. 104 (2017)
302–314.
The authors declare that there are no conflicts of interest. [30] H. Rachmawati, D.K. Budiputra, R. Mauludin, Curcumin nanoemulsion for trans-
dermal application: formulation and evaluation, Drug Dev. Ind. Pharm. 41 (2015)
560–566.
References
[31] A. Sarkar, I.J. Shimu, M.R.H. Tuhin, A.A. Raju, Nanoemulsion: an excellent mode
for delivery of poorly soluble drug through different routes, J. Chem. Pharm. Res. 7
[1] G. Ramadan, O. El-Menshawy, Protective effects of ginger-turmeric rhizomes mix- (2015) 966–976.
ture on joint inflammation, atherogenesis, kidney dysfunction and other compli- [32] V. Bali, M. Ali, J. Ali, Study of surfactant combinations and development of a novel
cations in a rat model of human rheumatoid arthritis, Int. J. Rheum. Dis. 16 (2013) nanoemulsion for minimizing variations in bioavailability of ezetimibe, Colloids
219–229. Surf. B: Biointerfaces 76 (2010) 410–420.
[2] N. Gerwin, C. Hops, A. Lucke, Intraarticular drug delivery in osteoarthritis, Adv. [33] J.K. Jackson, T. Higo, W.L. Hunter, H.M. Burt, The antioxidants curcumin and
Drug Deliv. Rev. 58 (2006) 226–242. quercetin inhibit inflammatory processes associated with arthritis, Inflamm. Res. 55
[3] M. Simadibrata, Gastrointestinal tract disorder as a side effect of non-steroidal anti- (2006) 168–175.
inflammatory drugs (NSAIDs), Acta Med. Indones. 36 (2004) 195–196. [34] B.R. Jeoung, K.D. Lee, C.S. Na, Y.E. Kim, B. Kim, Y.R. Kim, Ganghwaljetongyeum,
[4] P. Sooriakumaran, COX-2 inhibitors and the heart: are all coxibs the same? an anti-arthritic remedy, attenuates synoviocyte proliferation and reduces the
Postgrad. Med. J. 82 (2006) 242–245. production of proinflammatory mediators in macrophages: the therapeutic effect of
[5] J.L. Funk, J.B. Frye, J.N. Oyarzo, et al., Efficacy and mechanism of action of tur- GHJTY on rheumatoid arthritis, BMC Complement. Altern. Med. 13 (2013) 1–6.
meric supplements in the treatment of experimental arthritis, Arthritis Rheum. 54 [35] L. Hongtao, G.U. Xiaochen, Correlation between drug dissolution and polymer
(2006) 3452–3464. hydration: a study using texture analysis, Int. J. Pharm. 342 (2007) 18–25.
[6] T.N. Kaul, E. Middleton, P.L. Ogra, Antiviral effect of flavonoids on human viruses, [36] E.A. Foegeding, C.R. Daubert, M.A. Drake, G. Essick, M. Trulsson, C.J. Vinyard,
J. Med. Virol. 15 (1985) 71–79. D.F. Van, A comprehensive approach to understanding textural properties of semi
[7] S. Jain, A.K. Jain, M. Pohekar, K. Thanki, Novel self-emulsifying formulation of and soft solid foods, J. Texture Stud. 42 (2011) 103–129.
quercetin for improved in vivo antioxidant potential: implications for drug-induced [37] S. Khurana, N.K. Jain, P.M.S. Bedi, Nano-emulsion based gel for transdermal de-
cardiotoxicity and nephrotoxicity, Free Rad. Biol. Med. 65 (2013) 117–130. livery of meloxicam: physico-chemical, mechanistic investigation, Life Sci. 92
[8] V.G. Mediavilla, I. Crespo, P.S. Collado, A. Esteller, S.S. Campos, M.J. Tunon, (2013) 383–392.
J.G. Gallego, The anti-inflammatory flavones quercetin and kaempferol cause in- [38] K.R. Patil, C.R. Patil, R.B. Jadhav, V.K. Mahajan, P.R. Patil, P.S. Gaikwad, Anti-
hibition of inducible nitric oxide synthase, cyclooxygenase-2 and reactive C- pro- arthritic activity of bartogenic acid isolated from fruits of Barringtonia racemosa
tein, and down-regulation of the nuclear factor kappa- B pathway in Chang Liver Roxb. (Lecythidaceae), Evid. Complement. Alternat. Med. (2011) 1–7.
cells, Eur. J. Pharmacol. (2007) 221–229. [39] G. Kaur, S. Sultana, Evaluation of antiarthritic activity of iso-eugenol in adjuvant
[9] E.S. Yu, H.J. Min, S.Y. An, H.Y. Won, J.H. Hong, E.S. Hwang, Regulatory me- induced arthritis in murine model, Food Chem. Toxicol. 50 (2012) 2689–2695.
chanisms of IL-2 and IFNg suppression by quercetin in T helper cells, Biochem. [40] M.V. Chandra, B.A. Shamsundar, Texture profile analysis and functional properties
Pharmacol. 76 (2008) 70–78. of gelatin from the skin of three species of fresh water fish, Int. J. Food. Prop. 18

10
J.P. Gokhale, et al. Biomedicine & Pharmacotherapy 112 (2019) 108622

(2015) 572–584. containing solid lipid nanoparticles of vitamin A, AAPS PharmSciTech 7 (2006)
[41] S.H. Butler, F. Godefroy, J.M. Besson, J. Weil-Fugazza, A limited arthritic model for E63–E69.
chronic pain studies in the rat, Pain 48 (1992) 73–81. [45] A.A. Date, M.S. Nagarsenker, Parenteral microemulsions: an overview, Int. J.
[42] A. Hussain, A. Samad, S.K. Singh, M.N. Ahsan, M.W. Haque, A. Faruk, F.Z. Ahmed, Pharm. 355 (2008) 19–30.
Nano-emulsion gel-based topical delivery of an antifungal drug: in vitro activity and [46] R. Jeyadevi, T. Sivasudha, A. Rameshkumara, D.A. Ananth, G.S.B. Aseervatham,
in vivo evaluation, Drug Deliv. (2014) 642–657. K. Kumaresan, L. Dinesh Kumar, S. Jagadeeswari, R. Renganathan, Enhancement of
[43] J. Draize, G. Woodard, H. Calvery, Methods for the study of irritation and toxicity of anti arthritic effect of quercetin using thioglycolic acid-capped cadmium telluride
substances topically applied to skin and mucous membranes, J. Pharmacol. Exp. quantum dots as nanocarrier in adjuvant induced arthritic Wistar rats, Colloids Surf.
Ther. 82 (1944) 377–390. B: Biointerfaces 112 (2013) 255–263.
[44] P.V. Pople, K.K. Singh, Development and evaluation of topical formulation

11

You might also like