You are on page 1of 13

Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

Contents lists available at ScienceDirect

Colloids and Surfaces B: Biointerfaces


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

Meloxicam emulgel potently suppressed cartilage degradation in knee


osteoarthritis: Optimization, formulation, industrial scalability and
pharmacodynamic analysis
Vaskuri GS Sainaga Jyothi a, Harithasree Veerabomma a, Rahul Kumar b,
Dharmendra Kumar Khatri b, Shashi Bala Singh b, Jitender Madan a, *
a
Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
b
Department of Biological Sciences, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India

A R T I C L E I N F O A B S T R A C T

Keywords: Background and objective: Meloxicam (MLX) is prescribed for the management of pain and inflammation allied
Emulgel with osteoarthritis (OA). However, MLX causes intestinal damage in long term administration. Hence, meloxicam
Meloxicam loaded emulgel (MLX-emulgel) was optimized, formulated and examined under stringent parameters in
Osteoarthritis
monosodium-iodoacetate (MIA) induced knee OA in Wistar rats.
Western blotting
Methods and results: Nanoemulsion of MLX was fabricated by ultrasonication and microfluidization method with
Inflammatory cytokines
a droplet size of 66.81 ± 5.31-nm and zeta potential of − 24.6 ± 0.72-mV. Further, MLX nanoemulsion was
optimized with centrifugation, heating-cooling cycles and transmittance parameters in addition to scale-up
feasibility with microfluidizer. Post optimization, MLX-nanoemulsion was tailored as emulgel with Carbopol
Ultrez 10 NF and assessed for pH, rheology, textural properties, assay and stability features. The in-vitro release
study revealed the Korsmeyer-Peppas release kinetics and ex-vivo skin permeation was improved by 6.71-folds.
The skin distribution of MLX-emulgel evinced the transfollicular mode of permeation. In-vivo study indicated the
protective action of MLX-emulegl expressed in terms of inflammatory cyctokines level, X-ray analysis of knee
joints of rats, histopathology and OARSI (Osteoarthritis Research Society International) scoring. MLX-emulgel
treated group displayed lower (P < 0.001) level of COX-2 intensity as compared to positive control group.
However, it was comparable (P > 0.05) to the normal control group, MLX oral dispersion, i.v. solution and
etoricoxib gel groups. MLX-emulgel showcased an alternative to the long term usage of analgesics for relieving
the symptoms of knee OA.
Conclusion: MLX-emulgel may be a potential candidate for translating in to a clinically viable dosage form in the
management of knee OA.

1. Introduction metalloproteinases (MMPs) [8,9], contributing to cartilage destruction


[10,11]. Conversely, TNF-α interferes with the chondrocytes’ produc­
Osteoarthritis (OA), is a degenerative joint disease, that mainly af­ tion of type II collagen, proteins that bind proteoglycans, and compo­
fects hip and knee joints [1] with worldwide prevalence >7 % [2,3]. OA nents of proteoglycans [12,13] and IL-6 inhibits the formation of type II
is triggered by both internal and external factors including ageing, collagen and enhances MMPs’ production [14]. Additionally, IL-1β and
obesity, joint trauma, increased mechanical loading, and hereditary TNF-α stimulates the expression of cyclooxygenase-2 (COX-2) enzyme
predisposition [4]. Pathophysiologically, OA alters the equilibrium of [15,16] leading to PGE2 (Prostaglanding) production which plays a
catabolic and anabolic process of articular cartilage where the catabolic major role in pain and inflammation.
process predominates causing breakdown of cartilage leading to syno­ International and national guidelines recommended oral non-
vial inflammation by inflammatory cytokines namely, TNF-α, IL-1β and steroidal anti-inflammatory drugs (NSAIDs) for managing pain in OA,
IL-6 [5–7]. IL-1β upregulates matrix-degrading enzymes called matrix which inhibit COX-2 enzyme for those who do not respond to

* Corresponding author.
E-mail address: jitenderpharmacy@gmail.com (J. Madan).

https://doi.org/10.1016/j.colsurfb.2023.113399
Received 18 March 2023; Received in revised form 7 June 2023; Accepted 8 June 2023
Available online 10 June 2023
0927-7765/© 2023 Elsevier B.V. All rights reserved.
V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

paracetamol [17–21]. Hence, NSAIDs like diclofenac, naproxen and Next, the screened surfactant was combined separately with different
meloxicam (MLX) are prescribed for oral administration to manage OA cosurfactants, namely span 80 (HLB~4.3), span 20 (HLB~8.6) and
[22]. However, MLX has anti-inflammatory effects similar to or better transcutol HP (HLB~4) at 7.5 % w/w concentration in 2.5 mL of water
than those of other NSAIDs in animal models, with a greater therapeutic and noticed for turbidity of the emulsion.
ratio [23,24]. MLX is an oxicam derivative recommended by US-FDA for
pain and inflammation management in OA. It is a selective COX-2 in­ 2.2.3. Preparation of nanoemulsion and construction of ternary phase
hibitor and commercially available as an oral dosage form (7.5–15 mg) diagram
[25,26]. However, long term oral use of MLX is associated with certain High energy emulsification method was adopted for the preparation
side-effects like gastrointestinal ulceration, perforation, bleeding, and of nanoemulsion using probe sonicator [44,45]. The viscous nature of
colitis, contraindicated in patients having asthma [27] and increases the castor oil hampered the preparation of self-emulsifying nanoemulsion,
risk of myocardial infarction by 38 % due to COX-2 inhibition activity leading to gelation of the nanoemulsion. Hence, the fraction of castor oil
[28]. Hence, topical NSAIDs are generally recommended over oral was kept constant to 10 % w/w [46] while the surfactant ratio was
NSAIDs for pain management in OA [29]. varied from 15 % to 20 % w/w. Surfactant ratio less than 15 % w/w did
Nevertheless, potential of topical application of MLX in coping pain not yield emulsion formation and higher than 20 % w/w yielded gela­
and inflammation in OA is still not explored. Recently, our group re­ tion of nanoemulsion [47]. The surfactant and cosurfactant were taken
ported MLX loaded solid lipid nanoparticles topical gel in the manage­ as Smix ratio in the range of 1:9–9:1 and weighed amount of water was
ment of OA [30]. MLX owing to low molecular weight (Mw~351.403 added drop-wise under continuous stirring at 700-rpm for 30 min.
Da), BCS (Biopharmaceutical Classification System) Class-II, and Log P Subsequently, the preformed emulsion was probe sonicated and the
of 3.02 is a suitable drug entity for topical delivery [31] in managing OA. sonication time was optimised (Table 1 and Fig. 1) for gaining desirable
Lately, emulgel dosage form has been widely explored for topical droplet size [44]. Ternary phase diagram was constructed using Tri-plot
delivery of therapeutic entities in several pain disorders like sunburn v4.1.2 by varying the concentration of surfactant (1.5–18 % w/w),
[32], anal fissure [33], post hemorrhoidectomy pain [34], and vulvar cosurfactant (1.5–18 % w/w) and water (70–75 % w/w).
pain [35]. Emulgel, an amalgamation of emulsion and gel [36], dem­
onstrates several desirable features like spreadable, thixotropic, 2.2.4. Preparation of meloxicam loaded nanoemulsion
greaseless, pleasing appearance, emollient and long shelf-life. In addi­
tion, emulgel has exhibited great clinical success in delivery of topical 2.2.4.1. Ultrasonication method. Post optimization, nanoemulsion of
therapeutics [37–39]. MLX was prepared by dissolving the drug (1 % w/w) and meglumine in
Therefore, in present investigation, MLX was loaded in emulgel 1:1.25 ratio in addition to surfactant, cosurfactant (Table 1) and oil
(MLX-emulgel) for demonstrating a high degree of analgesic and anti- mixture under continuous stirring at 70 ◦ C followed by cooling at room
inflammatory activity in managing knee OA. The therapeutic potential temperature. Water was added drop-wise with continuous stirring at
of MLX-emulgel was evaluated in monosodium iodoacetate (MIA) 700-rpm to obtain an emulsion and ultrasonicated to reduce the droplet
induced OA in Wistar rats under a set of stringent in vivo parameters size and stored for further characterization.
[40].
2.2.4.2. Microfluidizer method. Microfluidizer was used for MLX-
2. Materials and methods nanoemulsion fabrication as industrial scalable technique [48]. In
brief, 100 mL of coarse emulsion was processed through microfluidizer
2.1. Materials (LM20, Microfluidics, USA), operated at different pressures of
20000–30000 psi and the samples were passed for 1–6 cycles and
MLX (99.5 %) was received as a gift sample from Swati Spentose assessed for droplet size [49].
Private Limited, Gujarat, India; Castor oil was purchased from Sisco
Research Laboratories, Mumbai, India; Transcutol HP was obtained as a
gift sample from Gattefosse, France; Tween 80 was purchased from Avra 2.3. Characterization of nanoemulsion
Laboratories Private Limited, Hyderabad, India. Carbopol®Ultrez 10 NF
was provided by Lubrizol, Mumbai, India. Meglumine was procured 2.3.1. Droplet size and zeta potential
from Sigma Aldrich, Bangalore, India. All the solvents and chemicals The droplet size of MLX-nanoemulsion (F1-F8; Table 1) was deter­
used were of higher analytical grade. Water used was of Milli-Q purified. mined by diluting 1 mL of nanoemulsion to 10-folds with distilled water
and the droplet size, polydispersity index (PDI) and zeta potential were
2.2. Preparation and optimization of meloxicam nanoemulsion measured using a Zetasizer (Zeta Sizer Nano NS, Malvern, UK) at 25 ◦ C
over a 90◦ angle.
2.2.1. Selection and solubility of drug in oil, surfactants and cosurfactants
Castor oil was selected as oil phase owing to its analgesic properties 2.3.2. Centrifugation and thermodynamic stability study
in OA [41]. Preliminary solubility of MLX was determined in castor oil, MLX-nanoemulsion (F1-F8; Table 1) was centrifuged at 25,000-rpm
where 0.1 g of MLX was suspended in 1 g of castor oil in presence and for 20 min and observed for cracking, creaming and phase separation.
absence of 0.125 g of meglumine and shaken vigorously for 2 h by vortex The stable samples were subjected to thermodynamic stability studies
mixer, centrifuged and supernatant was collected, diluted appropriately [50] where MLX-nanoemulsion (F3-F6; Table 1) were subjected to
with ethanol and filtered through 0.22 µm membrane filter to determine heating-cooling cycles, placed alternatively at 4 ℃ and 45 ℃ for a
the drug content by HPLC method [42]. Similarly, the solubility of MLX period of 48 h for six cycles and observed for cracking, creaming and
in selected surfactants and cosurfactants was also determined (Suppl. phase separation [51].
Table 1).
2.3.3. Percentage transmittance
2.2.2. Nanoemulsification capability screening of surfactants and Percentage transmittance of the stable MLX-nanoemulsion (F3-F6;
cosurfactants Table 1) was determined using UV–VIS spectrophotometer (V-650
Surfactants labrasol (HLB~14), tween 20 (HLB~16.7), and tween 80 double beam UV-Visible spectrophotometer, Jasco Inc., Germany) by
(HLB~15) were screened where 2.5 mL of 15 % w/w of each surfactant diluting the sample 100-folds with distilled water and analyzed at
solution was tested for its emulsifying ability against castor oil, added in 680 nm. The optimized ratio was selected based on percentage trans­
a drop-wise fashion till the solution turns cloudy under vortexing [43]. mittance [52].

2
V.G.S. Jyothi et al.
Table 1
Ternary phase composition, characterization and optimizing parameters of meloxicam loaded nanoemulsion.
Formulation Cosurfactant to Cosurfactant Surfactant Water (% Region/ Droplet Polydispersity Zeta Observation
Code surfactant ratio (Transcutol, % w/w) (Tween 80; % w/ w/w) observation size (nm) index potential
Centrifugation Thermodynamic Percent
w) (mV)
stability transmittance

F1 01:09 1.5 13.5 75 Δ/transparent 60.71 ± 0.303 ± 0.08 -25 ± 3.25 Turbid _ _
8.23
F2 01:09 2 18 70 Δ/transparent 68.36 ± 0.222 ± 0.05 -25.9 ± 3.61 Turbid _ _
0.91
F3 01:07 1.875 13.125 75 Δ/transparent 61.84 ± 0.3 ± 0.05 -28.9 ± 3.76 Clear and Clear and 94.75 ± 0.92
3.86 transparent transparent
F4 01:07 2.5 17.5 70 Δ/transparent 66.81 ± 0.112 ± 0.03 -24.6 ± 0.72 Clear and Clear and 96.06 ± 0.63
5.31 transparent transparent
F5 01:05 2.5 12.5 75 Δ/transparent 65.06 ± 0.321 ± 0.05 -31.1 ± 1.82 Clear and Clear and 89.38 ± 1.36
3.33 transparent transparent
F6 01:05 3.33333 16.6667 70 Δ/transparent 55.79 ± 0.208 ± 0.01 -25.3 ± 3.26 Clear and Clear and 94.04 ± 1.6
3

1.38 transparent transparent


F7 01:03 3.75 11.25 75 Δ/transparent 75.6 ± 0.316 ± 0.1 -24.5 ± 2.2 Turbid _ _
14.16
F8 01:03 5 15 70 Δ/transparent 70.1 ± 0.084 ± 0.09 -24.5 ± 1.65 Turbid _ _
12.54
F9 01:01 7.5 7.5 75 +/ cracking _ _ _ _ _ _
F10 01:01 10 10 70 +/ cracking _ _ _ _ _ _
F11 03:01 11.25 3.75 75 +/ cracking _ _ _ _ _ _
F12 03:01 15 5 70 +/ cracking _ _ _ _ _ _
F13 05:01 12.5 2.5 75 +/ cracking _ _ _ _ _ _
F14 05:01 16.6667 3.33333 70 +/ cracking _ _ _ _ _ _
F15 07:01 13.125 1.875 75 +/ cracking _ _ _ _ _ _

Colloids and Surfaces B: Biointerfaces 228 (2023) 113399


F16 07:01 17.5 2.5 70 +/ cracking _ _ _ _ _ _
F17 09:01 13.5 1.5 75 +/ cracking _ _ _ _ _ _
F18 09:01 18 2 70 +/ cracking _ _ _ _ _ _

Note: All measurements were noted in triplicate (n = 3)


V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

Fig. 1. A) Optimization of sonication time of nanoemulsion and B) Ternary phase diagram of nanoemulsion (Δ/ transparent, +/cracking). Data repre­
sented ***p < 0.001.

2.3.4. Drug loading efficiency and capacity 2.5.3. Assay


The drug loading efficiency (DLE) and drug loading capacity (DLC) Assay of the MLX-emulgel was performed by dispersing in 5 mL of
of MLX-nanoemulsion (F4; Table 1) was performed by diluting the water and acetonitrile (1:1 ratio) mixture and suitably diluted to analyse
sample with water and acetonitrile (1:1 ratio) mixture and analysed by RP-HPLC method [30,42].
using SHIMADZU HPLC over C18 column (250 × 4.6 mm, 5 µm) sup­
Weight of drug recovered
plied with Lab solutions software. The mobile phase was composed of %Assay = × 100 (3)
Weight of drug added
water (pH 3 adjusted with trifluoroacetic acid) and acetonitrile
(35:65 v/v) [30,42].
2.5.4. Accelerated and real time stability studies
Weight of drug recovered MLX-emulgel was subjected to stability studies as per ICH guidelines
%DLE = × 100 (1)
Weight of drug added at accelerated stability conditions of 40 ± 2 ◦ C with 75 ± 5 % relative
humidity (RH) in stability chamber (Osworld Scientific Equipment Pri­
%DLC =
Weight of drug recovered
× 100 (2) vate Limited, India) [55] for 6 months and at room temperature for 12
Weight of nanoemulsion months and assessed for pH and assay at the time intervals of 1, 3, 6
and/or 12 months.
2.3.5. Transmission electron microscopy (TEM)
The optimized MLX-nanoemusion (F4; Table 1) was stained with 2 % 2.5.5. In vitro drug release
phosphotungstic acid on copper grid and allowed to air dry for 1 min The release of drug from the MLX-emulgel was assessed by Franz
and observed under TEM (Jeol India Private Limited, JEM 1400HR) with diffusion cell apparatus [56]. Dialysis membrane-110 (Mw
an accelerating voltage of 120 kV. 2000–14000 Da) was embedded between the donor and receptor
compartment. MLX-emulgel (~2.5 mg of MLX) and MLX-suspension
(2.5 mg) were separately dispersed in donor compartment while the
2.4. Preparation of meloxicam loaded emulgel receiver compartment was filled with 28 mL of phosphate buffer saline
(pH~7.4) containing 30 % w/v polyethylene glycol 400 [57]. The buffer
Carbopol gel was used as gelling agent where 4 g of Carbopol® in the receptor compartment was maintained at 32 ± 1 ◦ C with
Ultrez 10 NF was suspended in 100 mL of water with stirring at 1000- continuous stirring at 200-rpm. 1 mL of sample was withdrawn at in­
rpm overnight and pH adjusted with triethanolamine to 6–6.5. MLX- tervals of 0.16, 0.33, 0.5, 0.75, 1, 2, 3, 4, 5, 6, 7 and 8 h and replaced
nanoemulsion was mixed with carbopol gel in 1:3 ratio with gentle with equal volume of buffer. Samples were passed through 0.22-µm
stirring at 1000-rpm under overhead stirrer for 3 h to obtain MLX- syringe filter and analysed by RP-HPLC. Release model kinetics were
emulgel [53]. applied using the DDSolver® software to assess zero order, first order,
Higuchi, Korsmeyer-Peppas and Hixson-Crowell models.

2.5. Characterization of emulgel 2.5.6. Ex-vivo skin permeation studies


Ex-vivo skin permeation studies of MLX-emulgel were performed in
2.5.1. Visual inspection, pH determination male Wistar rats which were approved (NIP/05/2021/PE/411) for study
The MLX-emulgel was inspected visually for its appearance, homo­ by Institutional Animal Ethics Committee (IAEC). Animals were sacri­
geneity and colour. Further, 1 g of emulgel was dispersed in 20 mL of ficed and skin of the dorsal abdominal region was shaved, excised and
water and assessed by pH meter (Mettler Toledo, UK) [54]. the subcutaneous fatty tissue was removed, washed and stored at − 80 ◦ C
until further use [58]. The skin was mounted between the donor and
2.5.2. Rheology and texture analysis receptor compartment of Franz diffusion cell apparatus with the stratum
Rheological behaviour was measured by modular compact rheom­ corneum facing towards the donor compartment and experiment was
eter (Anton Paar Private Limited, Germany) at 25 ◦ C. MLX-emulgel was performed as discussed in the release study session [57]. The slope of the
placed on the plate and measured by increasing the shear rate from 0.1 linear portion of the curve was contemplated as flux (Jss, μg/cm2/h) and
to 100 s− 1 and compared with the marketed gel. The spreadability was the enhancement ratio (ER) was calculated [59].
evaluated by texture analyser (T.A. XT Plus, UK) where MLX-emulgel
Flux of drug from emulgel
was placed on the female cone and a graph was obtained by pene­ ER = (4)
Flux of drug from drug suspension
trating the male cone in female cone cavity and vice-versa.

4
V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

2.6.3. Western blotting


2.5.7. Skin retention studies
Animals were sacrificed on 14th day of induction and the synovial
For skin retention study, the skin was removed post ex-vivo skin
tissues were collected and stored at − 80 ◦ C till use. The synovial tissues
permeation study [60], washed to get rid of remaining content, minced,
were lysed using RIPA (Radioimmunoprecipitation assay) buffer con­
homogenized with 10 mL of acetonitrile, vortexed, incubated overnight
taining both protease and phosphatase inhibitors and homogenised at
and sonicated for 1 h. The sample was then centrifuged at 12,000-rpm
4 ◦ C. The lysates were centrifuged (13,000 rpm) for 15 min at 4 ◦ C and
for 15 min and the supernatant was filtered through 0.22-µm syringe
the supernatant was separated. The synovial tissues were assessed for
filter and analyzed by RP-HPLC [30,42].
protein estimation by Bicinchoninic Acid (BCA) protein assay kit and
tissue samples of 10-µg equivalent amount of protein were fractioned on
2.5.8. Confocal laser scanning microscopy (CLSM)
10 % SDS-PAGE gel electrophoresis and electroblotted on nitrocellulose
Visualization of emulgel permeation pathways in skin was achieved
membrane, treated with COX-2 primary monoclonal antibody (Santa
by loading coumarin-6 dye (0.4 % w/w). Coumarin-6 dye loaded
Cruz Biotechnology, Inc., Texas, USA) and incubated again with bio­
emulgel incubated with skin in Franz diffusion cell apparatus for 2 h and
tinylated secondary IgG antibody conjugated with horseradish-
compared with the coumarin-6 dye suspension [53]. The skin was
streptavidin (Santa Cruz Biotechnology, Inc., Texas, USA) for 1 h. The
sectioned to 20-µm by cryotome at − 25 ◦ C, stained with DAPI (4′ ,
membrane was envisaged by chemiluminescene reagent under chemdoc
6-diamidino-2-phenylindole) and observed under CLSM (Leica TCS SP8
and bands were quantified with Image-J software [40,63].
Laser Scanning Spectral Confocal).
2.6.4. Inflammatory cytokines
2.6. Therapeutic efficacy analysis
The level of inflammatory cytokines in synovial tissue homogenates
was estimated by commercially available ELISA kits [40] for TNF-α,
The therapeutic efficacy of MLX-emulgel was evaluated in MIA
IL-1β, IL-6 and IL-10. Briefly, 96-well plates were incubated with the
induced knee OA in male Wistar rats [61]. Studies were performed as per
corresponding capture antibody overnight and incubated with standard
Committee for the Purpose of Control and Supervision of Experiments on
or synovial tissue homogenates for 2 h, followed by incubation with
Animals (CPCSEA) guidelines, Ministry of Fisheries, Animal Husbandry,
detection antibody and streptavidin-horsereddish peroxidase B solution
and Dairying, Department of Animal Husbandry, and Dairying, Gov­
and colour reagents, followed by the addition of stop solution
ernment of India, New Delhi, India. The study was approved by the
(2 N H2SO4). The wells were immediately analysed by microplate reader
Institutional Animal Ethics Committee (IAEC) vide protocol #
to determine the optical density at 450 nm.
NIP/05/2021/PE/411. All animals were acclimatized for one week in
animal house and divided into 8 groups with 6 animals in each group.
2.6.5. Histopathology
Group A was retained as normal control group with no induction of OA
Histopathology of knee joints was assessed by fixing the knee joints
and injected with 50-µL of sterile normal saline to the right knee joint.
in 10 % v/v formalin solution and decalcified for 21 days in 20 % v/v
All the remaining groups were induced OA by injecting a single dose of
Ethylenediamine tetra-acetic acid and implanted in paraffin wax [64].
3-mg of MIA dispersed in 50-µL normal saline via intra-articular route to
The joints were sectioned sagittally, stained with toluidine blue and
the right knee joint [40]. Group B was assigned as disease or positive
visualised under microscope (Leica Microsystems DM750 microscope,
control group and given no treatment. Group C and D were treated with
Germany). OARSI (Osteoarthritis Research Society International)
oral dispersion of MLX (1-mg/kg) and intravenous (i.v.) injection of MLX
scoring ranging from 0 to 24 was given to the joint sections [65].
(0.2-mg/kg) once-a-day, respectively. Group E, F, G and H were treated
with 1 % w/w MLX-emulgel, commercial 1 % w/w etoricoxib (COX-2
2.7. Statistical analysis
inhibitor) gel, 1 % w/w commercial diclofenac (non-selective COX in­
hibitor) gel and blank emulgel twice-a-day, respectively. The treatment
All measurements were noted in triplicate (n = 3). Using GraphPad
was given for 14 days.
Prism version 5 software (GraphPad software, San Diego, California,
US), the data was statistically evaluated. "Bonferroni’s Multiple Com­
2.6.1. Thickness of knee joint, motor incoordination and thermal
parison Test" was used for a follow-up study, and results were deemed
hyperalgesia test
statistically significant at P < 0.05.
The swelling of knee joint following the administration of MIA was
assessed using a calibrated Vernier calliper (Mitutoyo ABSOLUTE digital
3. Results and discussion
caliper, USA) by measuring the thickness of right knee joint while the
left knee joint thickness served as control [61].
3.1. Ultrasonication and microfluidization assisted in scale-up of a stable
Motor incoordination was assessed by rota-rod device which reveals
MLX-emulgel
the ability of knee joint to coordinate moment. Rats were placed on
rotating cylinders with an acceleration speed of 5–35 rpm over 1 min
According to Association of the Scientific Medical Societies in Ger­
and the time taken for rats to fall-off the rod was noted with a cut-off
many guideline for knee OA, reducing risk should be the primary
time of 3 min [62].
approach to improve the quality of life while treating with analgesic and
Thermal hyperalgesia response of rats to heat was assessed by hot
anti-inflammatory drugs [66]. Hence, MLX-emulgel dosage form [67]
plate test. Rats were individually exposed to the hot plate heated to a
was explored as a vehicle for delivering therapeutic entity in OA topi­
temperature of 55 ◦ C and the pain threshold was measured as a response
cally to reduce systemic exposure [68]. Thus, MLX-nanoemulsion with
of jumping or paw licking [62]. All tests were performed at time in­
castor oil as an oil phase was customized owing to its clinical utility in
tervals of 1, 3, 7 and 14 days.
OA [41]. However, the solubility of MLX in castor oil was low (0.66
± 0.06 mg/mL) (Suppl. Table 1) and therefore meglumine was incor­
2.6.2. Radiographic analysis of knee joints
porated in the oil phase to enhance its solubility [63]. The 1.8-folds hike
The radiographic images of rat knee joints were taken on 14th day of
in solubility of MLX in presence of meglumine in castor oil may be
induction in antero-posterior and lateral view, captured at Versha Di­
attributed to the formation of molecular complex between MLX and
agnostics (Wipro GE - ELPRO X-ray machine), Hyderabad, India at a
meglumine (pH~11.0 and log P~− 2.509) in castor oil (Hydroxyl value:
voltage of 50-kV and a current of 100-mA with 10 s as exposure time.
≥160; Acidity value~1.5; pH~5.7–6.3) owing to the presence of alka­
line (-NH2) group in meglumine and -OH of MLX (Suppl. Fig. 1) [69].
However, the increased solubility was not sufficient to load MLX in

5
V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

nanoemulsion. Hence, the solubility of MLX in the selected surfactants 77]. F1-F8 nanoemulsions (Table 1) showed zeta-potential in the range
and cosurfactants was also determined (Suppl. Table 1). of − 24.5 ± 1.65 mV to − 31.1 ± 1.82 mV. The negative zeta potential of
Next, the critical parameter in nanoemulsion formulation was the tailored nanoemulsions may be attributed to the hydroxide ions in
proper selection of surfactants and their concentration [70,71]. water, which reacted with ricinoleic acid, present in the castor oil and
Non-ionic surfactants are relatively less toxic than their ionic counter­ carried to the interface and provided a negative charge at the hydro­
parts [72]. As the intended formulation is o/w nanoemulsion, surfac­ phobic/water interface [78].
tants with high HLB values and cosurfactants with low HLB values were Centrifugation was performed to evaluate the effect of physical stress
selected for screening [73]. Based on the emulsifying ability, tween 80 on nanoemulsion stability. F3-F6 nanoemulsions (Table 1) were found to
and transcutol HP was selected as surfactant and cosurfactant respec­ be stable post centrifugation with no sign of phase separation and sub­
tively, based on the emulsifying ability. The solubility of MLX in sur­ jected to thermodynamic stability analysis. F3-F6 nanoemulsions were
factant and cosurfactant was found to be 115.91 ± 0.52 mg/g and found to be stable and clear with no sign of phase separation or creaming
79.77 ± 0.54 mg/g (Suppl. Table 1) respectively, which were adequate (Table 1).
to load MLX in the nanoemulsion. Percentage transmittance of F3-F6 was estimated between 89 % and
Nanoemulsion was prepared by ultrasonication and the droplet size 96 % (Table 1). A value of percentage transmittance closer to 100 %
was decreased as a function of sonication time (Fig. 1A). Nevertheless, indicates that the nanoemulsion is clear and transparent, hence; F4
there was no significant (One-way ANOVA test, P > 0.05) difference nanoemulsion with 1:7 ratio (20 % w/w) of cosurfactant to surfactant
between the droplet size at 6 and 9 min sonication time, hence; 6 min was selected for the further characterization. The droplet size, PDI and
was taken as optimized time (Fig. 1A). Ternary phase curve was plotted zeta-potential of F4 nanoemulsion was found to be 66.81 ± 5.31-nm,
by varying the concentration of surfactant, cosurfactant and water and 0.112 ± 0.03 % and − 24.6 ± 0.72-mV, respectively (Fig. 2 A-D and
keeping the oil quantity as constant. The cosurfactant to surfactant ra­ Table 1). TEM was used to analyse the size and shape of F4 nano­
tios were evaluated from 1:9–9:1 where a total of 18 (F1-F18, Table 1) emulsion, where the droplets were spherical (60–90 nm range) and well
formulations were prepared of which only F1-F8 were found to be stable separated with absence of aggregation or coalescence (Fig. 2A-D) [79,
and the rest were unstable (Fig. 1B). 80]. The DLE of F4 nanoemulsion was found to be 102.7 ± 1.8 % and
The droplet size of nanoemulsion < 150 nm was preferred for the DLC was found to be 1.37 ± 0.02 %, calculated from HPLC method [30,
topical delivery of therapeutic entity [74]. Lower PDI value (<0.3) in­ 42]. The retention time (RT) of MLX was found to be 6 min with 0.999 as
dicates the uniformity of droplets in nanoemulsion [75]. The droplet size R2 value (Suppl. Fig. 2).
of F1 to F8 nanoemulsion formulations was found in the range of 60.71 Microfluidizer [49], as industry scalable technology was explored for
± 8.22-nm to 75.6 ± 14.16-nm (Table 1) with a PDI between 0.321 tailoring nanoemulsion. The coarse emulsion was subjected to pressures
± 0.05–0.084 ± 0.09. A high zeta-potential value of ≥ ± 25 mV confers ranging from 20,000 to 30,000 psi for 6 cycles (Suppl. Fig. 3). The
stability preventing the agglomeration of droplets of nanoemulsion [76, droplet size of nanoemulsion was reduced significantly (two-way

Fig. 2. A) Average droplet size (66.81 ± 5.3 nm) and droplet size distribution (0.112 ± 0.03) of nanoemulsion by Zeta sizer; B) average zeta potential (− 24.6
± 0.72 mV) of optimised nanoemulsion; C & D) TEM images of nanoemulsion showed spherical droplets with a size range of 60–90 nm at the scale bar of 500 nm
and 200 nm.

6
V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

ANOVA test, P < 0.001) as a function of pressure and number of cycles. (Suppl. Table 2) with no significant (Unpaired t-test, p > 0.05) differ­
At 20,000 psi, the average droplet size was noticed in the range of 203.8 ence at selected time periods.
± 2.4-nm to 141.5 ± 0.9-nm from cycle 1–6 and 166.2 ± 1.0-nm to Release of drug from MLX-emulgel evaluated by Franz diffusion cell
136.2 ± 1.9-nm for 1–6 cycles at 25,000 psi. At 30,000 psi, the droplet apparatus was compared with MLX suspension where MLX-emulgel
size reduced to 68.3 ± 0.57-nm at 4th cycle which was similar (unpaired demonstrated 3.6-folds improvement as compared to MLX suspension
t-test, P > 0.05) to the droplet size tailored by ultrasonication. More­ at 8 h (Fig. 3A). MLX-emulgel showed 75.78 ± 2.8 % drug release at 8 h
over, the droplet size noticed at 30,000 psi with cycle 5 was reduced with significant (two-way ANOVA test, P < 0.001) improvement in
further to 54.45 ± 0.75-nm with PDI of 0.226 ± 0.01 (Suppl. Fig. 3). comparison to MLX suspension exhibiting 21 ± 1.44 % release. Release
Hence, 30,000 psi and cycle 5 was selected as the optimized process kinetic models were assessed where R2 for Korsmeyer-Peppas model
parameters. In addition, DLE and DLC of microfluidizer based nano­ with the value of 0.9874 was selected as best fit (Suppl. Table 3),
emulsion tailored with optimized process parameters were estimated to following diffusion and erosion kinetics [83]. Non-Fickian (anomalous)
be 101.4 ± 1.3 % and 1.35 ± 0.04 %, respectively. However, no sig­ diffusion mechanism of drug permeation was revealed by n value
nificant (Unpaired t test, P > 0.05) differences were noticed between the (0.621) as both diffusion through the gelling polymer and swelling of
DLE and DLC of F4 nanoemulsion formulated by ultrasonication method gelling polymer [84].
and microfluidizer based nanoemulsion. Permeation of drug across the rat skin was evaluated for both the
Next, F4 nanoemulsion prepared by ultrasonication was fabricated as MLX-emulgel and MLX suspension where emulgel displayed improved
MLX-emulgel and found to be smooth and yellow in colour with absence permeation as compared to plain drug (Fig. 3B). Nevertheless, perme­
of grittiness with 6.4 ± 0.2 pH. Rheological parameters were assessed ation of drug from MLX-emulgel across the skin was improved (two-way
by rheometer and the plot of shear rate vis-a-vis shear stress for MLX- ANOVA test, P < 0.001) post 2 h. The ER was calculated to be 6.71-folds
emulgel showed no linear correlation indicating non-Newtonian higher for MLX-emulgel (9.23 ± 0.1 μg/cm2/h) as compared to the MLX
behaviour (Suppl. Fig. 4). Similarly, in viscosity vis-a-vis shear rate suspension (1.38 ± 0.7 μg/cm2/h). This may be attributed to the
curve, the viscosity of MLX-emulgel was decreased as a function of the nanodroplet size of MLX embedded in emulgel which augmented the
shear rate (Suppl. Fig. 4) indicating pseudoplastic behaviour which is absorption of drug across skin [85]. In addition, retention of MLX in skin
anticipated for topical dosage forms [81]. Both the graph patterns were layers was 1.97-folds higher for MLX-emulgel (21.7 ± 8.6 µg/cm2) as
similar to the commercial gel. compared to MLX suspension (11.0 ± 3.5 µg/cm2). Further, higher
Textural properties were determined by texture analyzer and gel retention of MLX within skin layers may act as a reservoir for controlled
strength, work of shear, stickiness and force of extrusion estimated to be release of drug entity required to maintain a continuous therapeutic
259.68 ± 5.07 g, 141.74 ± 6.25 g.sec, − 89.78 ± 1.17 g.sec and concentration at the target site [53].
− 208.92 ± 13.4 g, respectively (Suppl. Fig. 5). Gel strength, work of In order to visualize the permeation pathway, coumarin-6 dye loaded
shear, stickiness and force of extrusion of marketed gel was noted to be emulgel was formulated and penetration pathway was envisioned using
98.28 ± 5.81 g, 31.45 ± 3.39 g.sec, − 16.90 ± 1.74 g.sec, and − 87.97 CLSM [86]. The fluorescence intensity of coumarin-6 dye under CLSM
± 3.99 g, respectively. Gel strength and work shear indicate the was quite low and can be seen in the epidermal layer only. On the other
spreadability of the emulgel [82]. Compared to marketed gel, hand, the fluorescence intensity of coumarin-6 dye loaded emulgel was
MLX-emulgel exhibited low spreadability and therefore requires high noticed in epidermis and dermis layer of skin with high intensity at
pressure for extrusion. The drug content of the MLX-emulgel was esti­ appendages of hair follicles (Fig. 3C). The results revealed transfollicular
mated to be 98.4 ± 0.12 %. Moreover, MLX-emulgel was stable at both route of drug permeation for emulgel [53,85].
room temperature (12 months) and accelerated stability conditions (6
months) in terms of appearance, pH in addition to assay values > 95 %

Fig. 3. A) In vitro drug release study of MLX from emulgel and MLX dispersion# with 3.6- folds improvement in drug release from the emulgel as compared to the
plain drug at 8 h; B) ex-vivo skin permeation profile of MLX from emulgel and MLX dispersion# with a flux of 9.23 ± 0.1 µg/cm2/h and 1.38 ± 0.7 µg/cm2/h
respectively, and C) skin distribution studies of coumarin-6 suspension# showed high fluorescence intensity in epidermis and appendages of hair follicles than
coumarin-6 dye loaded in emulgel where it is found in epidermis only (green arrow: epidermal layer; red arrow: appendages of hair follicles).#Adopted from [30]
with permission.

7
V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

3.2. MLX-emulgel suppressed pain sensation and cartilage degradation in no joint distension (Fig. 5A, Anterior-Posterior view). Positive control
MIA induced knee OA group pointed-out reduced joint space (Fig. 5B, red arrow, Anterior-
Posterior view) with erosions on joint surface [90]. Treatment with
MIA is widely implemented for the OA model development as it in­ MLX oral dispersion, MLX-i.v. injection, MLX-emulgel and etoricoxib gel
hibits glyceraldehyde-3-phosphate dehydrogenase which causes the displayed no joint distension (Fig. 5C-F, Anterior-Posterior view),
death of chondrocytes [87]. This also leads to the formation of osteo­ whereas, blank emulgel and diclofenac gel treated groups demonstrated
phyte and degradation of articular cartilage [88]. Injection of MIA narrowing of joint space (Fig. 5G-H, Anterior-Posterior view). The
resulted in inflammation and swelling of knee joint [61]. Thickness of joint distension was lesser in MLX-emulgel group as compared to the
knee joints was measured and difference between the thickness of right positive control group. MLX oral, i.v. injection, MLX-emulgel and etor­
and left knee joint was noted as change in thickness due to inflammation icoxib gel treated groups displayed mild erosion on the surface of
(Fig. 4A). The positive control group showed significant difference femoral condyles and proximal tibia (Fig. 5C-F, Lateral view). Blank
(two-way ANOVA test, P < 0.001) in swelling of knee joint as compared emulgel and diclofenac gel showed erosion at tibia and femur regions
to normal control group. MLX-emulgel significantly decreased the (Fig. 5G-H, Lateral view). Hence, MLX-emulgel was quite effective and
swelling of knee joint as compared to positive control (two-way ANOVA comparable to the MLX oral dispersion, i.v. injection and etoricoxib gel.
test, P < 0.001) group on all days and normal control group (two-way Blank emulgel was better than positive control in terms of joint space
ANOVA test, P < 0.001) on day 1. However, MLX-emulgel treated group and erosion which could be attributed to the anti-inflammatory property
showed no significant (two-way ANOVA test, P > 0.05) difference with of castor oil in blank emulgel.
normal control group on day 3, 7 and 14. MLX oral dispersion and i.v. Intra-articular injection of MIA resulted in cartilage damage leading
solution treated groups were comparable (two-way ANOVA test, to the release of proinflammatory cytokines, thereby elevated PGE2
P > 0.05) to MLX-emulgel treated group. This showed that the production by upregulating COX-2 expression [91]. Inhibition of COX-2
MLX-emulgel was efficient in reducing swelling of the knee joint. enzyme activity alleviates discomfort and pain of knee joint. Positive
Motor incoordination test was performed to determine incapability control group showed elevated COX-2 levels indicating OA disease
of rats to sustain on rolling rods and measured by marking fall off time model development (Fig. 6). MLX-emulgel treated group displayed
from the rotating rod [89]. MLX-emulgel showed significant (two-way lowered (one-way ANOVA test, P < 0.001) COX-2 intensity as compared
ANOVA test, P > 0.05) difference with the positive control group to positive control group and comparable (one-way ANOVA test,
(Fig. 4B). However, it demonstrated significant (two-way ANOVA test, P > 0.05) to the normal control group, MLX oral dispersion, i.v. solution
P < 0.001) difference in fall off time with normal control for 1st and 3rd and etoricoxib gel groups. Nevertheless, it was significantly different
day and comparable (two-way ANOVA test, P > 0.05) from day 7 on­ from diclofenac gel (one-way ANOVA test, P < 0.05) and blank emulgel
wards. This indicates that MLX-emulgel was effective post 3–7 days of its (one-way ANOVA test, P < 0.01) groups (Fig. 6). Thus MLX-emulgel
application in alleviating the discomfort in the moment of joints. alleviated pain, inflammation and discomfort associated with knee OA
However, MLX-emulgel demonstrated comparable (two-way ANOVA by decreasing COX-2 expression.
test, P > 0.05) results with that of MLX oral dispersion and i.v. solution Degradation of articular cartilage leads to the release of proin­
treated groups. flammatory cytokines [91–94]. MLX-emulgel showed insignificant
Intra-articular injection of MIA also resulted in change in pain (one-way ANOVA test, P > 0.05) difference in IL-1β level as compared to
threshold [61]. The pain threshold was measured by the latency to normal control, MLX oral dispersion, i.v. solution, etoricoxib and
withdrawal from heat where MLX-emulgel treated group was signifi­ diclofenac gels. However, IL-1β level in MLX-emulgel treated group was
cantly (two-way ANOVA test, P < 0.001) different from normal control significantly (one-way ANOVA test, P < 0.01) different from positive
group at day 1 and 3 and comparable (two-way ANOVA test, P > 0.05) control group with 1.32-folds reduction (Fig. 6). Moreover, high level of
since day 7. Nevertheless, it was significantly different from positive TNF-α and IL-6 also modulates MMPs expression stimulating cartilage
control group (two-way ANOVA test, P < 0.05) (Fig. 4C). The results degradation [95]. MLX-emulgel portrayed 2-folds and 2.23-folds
were comparable (two-way ANOVA test, P > 0.05) to MLX oral disper­ decrease in TNF-α and IL-6 levels, respectively as compared to positive
sion and i.v. solution treated groups demonstrating noteworthy effect of control group (Fig. 6). The level of TNF-α and IL-6 in MLX-emulgel
MLX-emulgel in reducing latency period. treated group was similar (one-way ANOVA test, P > 0.05) to MLX
Radiographic analysis of knee joints was performed at 14th day. oral dispersion, etoricoxib and diclofenac marketed gels. However, it
Normal control group showed intactness of the surface of cartilage and showed significant variation from positive control group (one-way

Fig. 4. Therapeutic efficacy analysis measured in terms of A) thickness of knee joint, B) motor incoordination and C) heat hyperalgesia for normal control group#,
positive control group# depicting monosodium iodoacetate induced OA, MLX emulgel treated group, MLX-oral treated group#, MLX i.v. treated group#, etoricoxib
marketed gel treated group#, diclofenac marketed gel# treated group, and blank gel treated group. #Adopted from [30] with permission.

8
V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

Fig. 5. Radiographic analysis of knee joint lateral view and anterior- posterior view: A) normal control# B) positive control# C) MLX oral dispersion#, D) MLX i.v.
solution#, E) MLX emulgel, F) etoricoxib (COX-2 inhibitor) marketed gel#, G) diclofenac (Non-selective COX inhibitor) marketed gel# and H) blank emulgel (N:
normal E: erosion and ME: mild erosion of femoral condyles and proximal tibia; Orange arrow: normal joint space, red arrow: reduced joint space with erosion, green
arrow: joint space similar to normal control and yellow arrow: narrow joint space).#Adopted from [30] with permission.

ANOVA test, P < 0.001), normal control group (one-way ANOVA test, reducing proinflammatory cytokines [97].
P < 0.01 and P < 0.05) and MLX i.v. solution group (one-way ANOVA Anti-inflammatory cytokine, IL-10 is involved in reversing the effect
test, P < 0.01 and P < 0.05). These results showed that MLX- emulgel of TNF- α and thereby reduces the production of prostaglandin [5,98].
was able to reduce the TNF-α and IL-6 levels preventing further pro­ This was reflected in elevated levels of IL-10 in all MIA induced groups
gression of disease by attenuating the expression of MMPs [96]. The role except normal control group (Fig. 6). These results were in-line with a
of castor oil in reducing inflammatory markers was evaluated as blank reported clinical study which justified that IL-10 expression was higher
emulgel. MLX-emulgel showed insignificant (one-way ANOVA test, in severe OA damage patients [99]. Thus, MLX-emulgel showed effec­
P > 0.05) difference in IL-1β level and significant (one-way ANOVA test, tiveness in decreasing COX-2 expression and proinflammatory levels.
P < 0.001 and P < 0.01) difference in TNF-α and IL-6 levels as compared Consequently, histopathology of rat knee joints was performed to
to the blank emulgel. Hence, castor oil had little to weak impact in evaluate the cartilage surface. Normal control group patterned intense

9
V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

Fig. 6. Measurement of COX-2 expression in addition to level of IL-1β, IL-6, TNF-α and IL-10 in normal control group#, positive control group#, MLX oral
dispersion#, MLX i.v. solution#, MLX-emulgel, etoricoxib (COX-2 inhibitor) marketed gel#, diclofenac (non-selective COX- inhibitor) marketed gel# and blank
emulgel. Data represented * **p < 0.001, * *p < 0.01, *p < 0.05 vs MLX emulgel group.#Adopted from [30] with permission.

blue staining of cartilage surface with intact articular cartilage (Fig. 7A, ELISA (Fig. 6) assay, respectively. In addition, protective action of
orange arrow). The positive control group displayed either a very weak MLX-emulgel was demonstrated by X-ray radiographic analysis and
or absence of staining due to deformation of cartilage (Fig. 7A, red histopathology (Fig. 7A-B). Thus, evidences support and advocate the
arrow). This showed the induction of disease condition in the rat’s right topical application of MLX-emulgel for OA management as compared to
knee joint. MLX oral dispersion and i.v. solution showed staining of to­ MLX oral and i.v. administration which eliminates the side-effects
luidine blue with profound discontinuity of the cartilage (Fig. 7A, yel­ associated with the long term systemic exposure of drug.
low arrow). This indicated that both MLX oral dispersion and i.v.
solution were able to protect the cartilage in OA disease condition, 4. Conclusion
consistent to the previous report [100]. MLX-emulgel group showed
incorporation of stain in cartilage and the subchondral bone (Fig. 7A, In conclusion, MLX-emulgel can be prepared by tailoring nano­
orange arrow) with erosion of > 10 % cartilage surface. Etoricoxib and emulsion using ultasonication and microfludizer methods suitable for
diclofenac gels displayed either absence or very weak staining of tolui­ scalability at industry level. Post examining the stringent pharmaceu­
dine blue dye due to denudation of cartilage (Fig. 7, black arrow) tical features, MLX-emulgel showed an improvement in permeation flux
indicating minimal protection against OA disease progression which of 6.71-folds as compared to plain drug and assessed for its potential in
were in harmony with the reported literature [101,102]. Blank emulgel alleviating pain and inflammation associated with knee OA. X-ray
also showed negligible staining and therefore, deformation of cartilage radiography of knee joint of rats revealed protective action of MLX-
(Fig. 7, blue arrow) was noticed. Castor oil present in the blank emulgel emulgel against OA that was further supported by measured cytokines
highlighted no remarkable effect on decreasing the OA severity. level, western blotting, histopathology and OARSI scoring. Interestingly,
Accordingly, MLX-emulgel was found to be superior to etoricoxib and the results were comparable to MLX oral and i.v. groups and showed
diclofenac gels and comparable to MLX oral and i.v. groups in treating superiority in terms of topical application to the knee joint exempting
and providing protection against OA. the side-effects associated with systemic absorption. MLX-emulgel
OARSI scoring was depicted in Fig. 7B. The highest score was noticed showcased an alternative to the long term usage of NSAIDs for
in the positive control group followed by blank emulgel. MLX oral and i. relieving symptoms of knee OA. Therefore, MLX-emulgel may be a po­
v. groups showed scoring equality and lower than MLX-emulgel. How­ tential candidate for translating into a clinically viable dosage form in
ever, score of MLX-emulgel was lesser than blank emulgel, etoricoxib managing knee OA.
and diclofenac gels [64,88]. MLX-emulgel proclaimed effective action
against knee OA by preventing the degradation of cartilage. CRediT authorship contribution statement
Hence, evidences indicated a mechanistic pathway of cartilage pro­
tection in OA by MLX-emulgel. MLX-nanoemulsion droplets embedded Vaskuri G S Sainaga Jyothi: Conceptualizartion, Experimentation
in emulgel dosage form might be diffused through the skin by following and manuscript writing and editing; Harithasree Veerabomma, Rahul
Korsmeyer-Peppas model (Fig. 3A and Suppl. Table 3) post topical Kumar: Experimentation; Dharmendra Kumar Khatri, Shashi Bala Singh,
application in knee OA that subsequently penetrated into the skin layers Jitender Madan: Supervision.
via trans-follicular route as reflected in CLSM imaging (Fig. 3A-C) and
reached to the inflamed tissue surrounding the knee joint [103]. Upon Declaration of Competing Interest
reaching to the inflamed synovial tissue in a therapeutic concentration,
MLX-emulgel decreased COX-2 expression and inflammatory cytokine The authors declare that they have no known competing financial
levels (IL-1β, TNF-α and IL-6), as supported by western blotting and interests or personal relationships that could have appeared to influence

10
V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

Fig. 7. A) Histopathology of representative Wistar rats from normal control group#, positive control group#, MLX oral dispersion#, MLX i.v. solution#, MLX emulgel,
etoricoxib (COX-2 inhibitor) marketed gel#, diclofenac (non-selective COX inhibitor) marketed gel# and blank emulgel. Orange arrow: Intact cartilage, red arrow:
deformation of cartilage, yellow arrow: discontinuity of cartilage, brown arrow: erosion of cartilage, black arrow: denudation of cartilage and blue arrow: defor­
mation of cartilage. B) Osteoarthritis Research Society International (OARSI) scores. #Note: Adopted from [30] with permission.

11
V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

the work reported in this paper. [23] S. Noble, J.A. Balfour, Drugs 51 (1996) 424–430, https://doi.org/10.2165/
00003495-199651030-00007.
[24] J. Hosie, M. Distel, E. Bluhmki, Clin. Drug Investig. 13 (2012) 175–184, https://
Data availability doi.org/10.2165/00044011-199713040-00001.
[25] D. Yocum, R. Fleischmann, P. Dalgin, J. Caldwell, D. Hall, P. Roszko, Arch. Intern
Data will be made available on request. Med 160 (2000) 2947–2954, https://doi.org/10.1001/ARCHINTE.160.19.2947.
[26] N.Y. Khalil, K.F. Aldosari, Meloxicam, Profiles Drug Subst. Excip. Relat. Method.
45 (2020) 159–197, https://doi.org/10.1016/BS.PODRM.2019.10.006.
Acknowledgements [27] V. Domingo, C. Marchuet, D. Culla, S. Joanpere, M. Guadaño, J. Invest. Allergol.
Clin. Immunol. 16 (2006) 364–366.
[28] D. Dalal, M. Dubreuil, C. Peloquin, T. Neogi, Y. Zhang, H. Choi, D. Felson,
Authors are highly thankful to Department of Pharmaceuticals, Rheumatol. Int. 37 (2017) 2071–2078, https://doi.org/10.1007/S00296-017-
Ministry of Chemicals and Fertilizers, Government of India, New Delhi, 3835-X.
India for providing financial assistance to carry out the research work. [29] W. Zhang, G. Nuki, R.W. Moskowitz, S. Abramson, R.D. Altman, N.K. Arden,
S. Bierma-Zeinstra, K.D. Brandt, P. Croft, M. Doherty, M. Dougados, M. Hochberg,
D.J. Hunter, K. Kwoh, L.S. Lohmander, P. Tugwell, Osteoarthr. Cartil. 18 (2010)
Appendix A. Supporting information 476–499, https://doi.org/10.1016/J.JOCA.2010.01.013.
[30] V.G.S.S. Jyothi, C.B. Katta, S. Singothu, K. Preeti, V. Bhandari, S.B. Singh,
J. Madan, J. Drug Deliv. Sci. Technol. 77 (2022), 103914 https://doi.org/
Supplementary data associated with this article can be found in the 10.1016/J.JDDST.2022.103914.
online version at doi:10.1016/j.colsurfb.2023.113399. [31] R.M. Khalil, A. Abd-Elbary, M.A. Kassem, M.M. Ghorab, M. Basha, Pharm. Dev.
Technol. 19 (2013) 304–314, https://doi.org/10.3109/10837450.2013.778872.
[32] J.L. Kienzler, J. Magnette, C. Queille-Roussel, A. Sanchez-Ponton, J.P. Ortonne,
References
Ski. Pharm. Physiol. 18 (2005) 144–152, https://doi.org/10.1159/000084912.
[33] R. Digennaro, G. Pecorella, S. La Manna, A. Alderisio, A. Alderisio, B. De Pascalis,
[1] A. Bortoluzzi, F. Furini, C.A.S. Affiliations, C.A. Scirè, Elsevier. 17(11) (2018), D. Pennisi, G. Santangelo, F. Pezzolla, A. Racalbuto, G. Serra, A. Pulvirenti
1097–1104. https://doi.org/10.1016/j.autrev.2018.06.002. D′ Urso, D.F. Altomare, Tech. Coloproctol. 19 (2015) 287–292, https://doi.org/
[2] D. Primorac, V. Molnar, E. Rod, Ž. Jeleč, F. Čukelj, V. Matišić, T. Vrdoljak, 10.1007/S10151-015-1289-2/FIGURES/4.
D. Hudetz, H. Hajsok, I. Borić, Genes 11 (2020) 854, https://doi.org/10.3390/ [34] S. Ala, M. Alvandipour, M. Saeedi, M. Hamidian, A. Shiva, N. Rahmani,
GENES11080854. F. Faramarzi, World J. Surg. 41 (2016) 596–602, https://doi.org/10.1007/
[3] K. McCulloch, G.J. Litherland, T.S. Rai, Aging Cell 16 (2017) 210–218, https:// S00268-016-3749-X.
doi.org/10.1111/ACEL.12562. [35] F. De Seta, G. Stabile, G. Antoci, G. Zito, R.E. Nappi, Healthcare 10 (2022) 830,
[4] Osteoarthritis | National Institute on Aging, https://www.nia.nih.gov/health https://doi.org/10.3390/HEALTHCARE10050830.
/osteoarthritis. [36] M. Talat, M. Zaman, R. Khan, M. Jamshaid, M. Akhtar, A.Z. Mirza, Drug Dev. Ind.
[5] P. Wojdasiewicz, A. Poniatowski, D. Szukiewicz, Mediat. Inflamm. (2014), Pharm. 47 (8) (2021) 1193–1199, https://doi.org/10.1080/
https://doi.org/10.1155/2014/561459. 03639045.2021.1993889.
[6] W. Lin, J. Klein, Adv. Mater. 33 (2021) 2005513, https://doi.org/10.1002/ [37] N.M. Badawi, M.A. Elkafrawy, R.M. Yehia, D.A. Attia, Drug Deliv. 28 (2021)
ADMA.202005513. 814–825, https://doi.org/10.1080/10717544.2021.1912211.
[7] M.B. Mueller, R.S. Tuan, PM&R. 3 (2011) S3–S11, https://doi.org/10.1016/J. [38] A. Golshah, S. Mirzaeei, N. Nikkerdar, F. Ghorbani, Int. J. Dent. (2021), https://
PMRJ.2011.05.009. doi.org/10.1155/2021/6615900.
[8] J.A. Mengshol, M.P. Vincenti, C.I. Coon, A. Barchowsky, C.E. Brinckerhoff, [39] S. Mohammadi, J. Shokri, M. Ranjkesh, S. Akbari Hamed, F. Monajjemzadeh,
Arthritis Rheum. 43 (2000) 801–811, https://doi.org/10.1002/1529-0131. J. Cosmet. Dermatol. 20 (2021) 174–180, https://doi.org/10.1111/JOCD.13471.
[9] E. Meszaros, C.J. Malemud, Ther. Adv. Chronic Dis. 3 (2012) 219, https://doi. [40] Z.M. Wang, Y.C. Chen, D.P. Wang, Biomed. Pharm. 83 (2016) 763–770, https://
org/10.1177/2040622312454157. doi.org/10.1016/J.BIOPHA.2016.06.050.
[10] R.F. Loeser, Arthritis Rheum. 54 (2006) 1357, https://doi.org/10.1002/ [41] B. Medhi, K. Kishore, U. Singh, S.D. Seth, Phytother. Res. 23 (2009) 1469–1473,
ART.21813. https://doi.org/10.1002/PTR.2804.
[11] R.A. Khalil, Matrix metalloproteinses and tissue remodeling in health and disease, [42] N.K. Sahoo, M. Sahu, P.S. Rao, N. Sandhya Rani, J.N.V. Indira Devi, G. Ghosh,
Target Tissues Ther. Vol.148 (2017) 446. Springer 3 (2014), https://doi.org/10.1186/2193-1801-3-95.
[12] C.A. Séguin, S.M. Bernier, J. Cell Physiol. 197 (2003) 356–369, https://doi.org/ [43] A. Azeem, M. Rizwan, F.J. Ahmad, Z. Iqbal, R.K. Khar, M. Aqil, S. Talegaonkar,
10.1002/JCP.10371. AAPS PharmSciTech 10 (2009) 69, https://doi.org/10.1208/S12249-008-9178-
[13] V. Lefebvre, C. Peeters-Joris, G. Vaes, Biochim Biophys. Acta 1052 (1990) X.
366–378, https://doi.org/10.1016/0167-4889(90)90145-4. [44] Y. Shi, H. Li, J. Li, D. Zhi, X. Zhang, H. Liu, H. Wang, H. Li, J. Drug Deliv. Sci.
[14] B. Porée, M. Kypriotou, C. Chadjichristos, G. Beauchef, E. Renard, F. Legendre, Technol. 27 (2015) 46–55, https://doi.org/10.1016/J.JDDST.2015.04.003.
M. Melin, S. Gueret, D.J. Hartmann, F. Malléin-Gerin, J.P. Pujol, K. Boumediene, [45] R. Kumar, S. Uppal, K. Mansi, J. Das, S.K. Pandey, K. Kaur, S.K. Mehta, J. Mol.
P. Galéra, J. Biol. Chem. 283 (2008) 4850–4865, https://doi.org/10.1074/JBC. Liq. 349 (2022), 118130, https://doi.org/10.1016/J.MOLLIQ.2021.118130.
M706387200. [46] V. Erramreddy, S. Ghosh, Emulsions (2016) 257–292, https://doi.org/10.1016/
[15] F.E. El Mansouri, N. Chabane, N. Zayed, M. Kapoor, M. Benderdour, J. Martel- B978-0-12-804306-6.00008-8.
Pelletier, J.P. Pelletier, N. Duval, H. Fahmi, Arthritis Rheum. 63 (2011) 168–179, [47] V.V. Erramreddy, S. Ghosh, Langmuir 30 (2014) 11062–11074, https://doi.org/
https://doi.org/10.1002/ART.27762. 10.1021/LA502733V/ASSET/IMAGES/MEDIUM/LA-2014-02733V_0010.GIF.
[16] M.M. Hardy, K. Seibert, P.T. Manning, M.G. Currie, B.M. Woerner, D. Edwards, [48] R. Paliwal, R.J. Babu, S. Palakurthi, Ageing Int 15 (2014) 1527–1534, https://doi.
A. Koki, C.S. Tripp, Arthritis Rheum. 46 (2002) 1789–1803, https://doi.org/ org/10.1208/S12249-014-0177-9/FIGURES/3.
10.1002/ART.10356. [49] O. Alliod, E. Almouazen, G. Nemer, H. Fessi, C. Charcosset, J. Pharm. Sci. 108
[17] O. Bruyère, C. Cooper, J.P. Pelletier, J. Branco, M. Luisa Brandi, F. Guillemin, M. (2019) 2708–2717, https://doi.org/10.1016/j.xphs.2019.03.026ï.
C. Hochberg, J.A. Kanis, T.K. Kvien, J. Martel-Pelletier, R. Rizzoli, S. Silverman, J. [50] S. Khani, F. Keyhanfar, A. Amani, Drug Deliv. 23 (2016) 2035–2043, https://doi.
Y. Reginster, Semin Arthritis Rheum. 44 (2014) 253–263, https://doi.org/ org/10.3109/10717544.2015.1088597.
10.1016/J.SEMARTHRIT.2014.05.014. [51] S. Shafiq-un-Nabi, F. Shakeel, S. Talegaonkar, J. Ali, S. Baboota, A. Ahuja, R.
[18] R.R. Bannuru, M.C. Osani, E.E. Vaysbrot, N.K. Arden, K. Bennell, S.M.A. Bierma- K. Khar, M. Ali, AAPS PharmSciTech 8 (2007), E12, https://doi.org/10.1208/
Zeinstra, V.B. Kraus, L.S. Lohmander, J.H. Abbott, M. Bhandari, F.J. Blanco, PT0802028.
R. Espinosa, I.K. Haugen, J. Lin, L.A. Mandl, E. Moilanen, N. Nakamura, [52] S. Kotta, A.W. Khan, S.H. Ansari, R.K. Sharma, J. Ali, Drug Deliv. 22 (2015)
L. Snyder-Mackler, T. Trojian, M. Underwood, T.E. McAlindon, Osteoarthr. Cartil. 455–466, https://doi.org/10.3109/10717544.2013.866992.
27 (2019) 1578–1589, https://doi.org/10.1016/J.JOCA.2019.06.011. [53] Q. Ma, J. Zhang, B. Lu, H. Lin, R. Sarkar, T. Wu, X. Li, AAPS PharmSciTech 22
[19] M.C. Hochberg, R.D. Altman, K.T. April, M. Benkhalti, G. Guyatt, J. McGowan, (2021), https://doi.org/10.1208/S12249-021-02035-5.
T. Towheed, V. Welch, G. Wells, P. Tugwell, Arthritis Care Res. (Hoboken) 64 [54] W. Sakran, R.S. Abdel-Rashid, F. Saleh, R. Abdel-Monem, Drug Deliv. 29 (2022)
(2012) 465–474, https://doi.org/10.1002/ACR.21596. 1477–1491, https://doi.org/10.1080/10717544.2022.2072542.
[20] K.M. Jordan, N.K. Arden, M. Doherty, B. Bannwarth, J.W.J. Bijlsma, P. Dieppe, [55] I.C.H. Q1A (R2), INTERNATIONAL CONFERENCE ON HARMONISATION OF
K. Gunther, H. Hauselmann, G. Herrero-Beaumont, P. Kaklamanis, S. Lohmander, TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS
B. Leeb, M. Lequesne, B. Mazieres, E. Martin-Mola, K. Pavelka, A. Pendleton, FOR HUMAN USE ICH HARMONISED TRIPARTITE GUIDELINE STABILITY
L. Punzi, U. Serni, B. Swoboda, G. Verbruggen, I. Zimmerman-Gorska, TESTING OF NEW DRUG SUBSTANCES AND PRODUCTS Q1A(R2), 2003.
M. Dougados, Ann. Rheum. Dis. 62 (2003) 1145–1155, https://doi.org/10.1136/ [56] S. Khurana, P.M.S. Bedi, N.K. Jain, Chem. Phys. Lipids 175–176 (2013) 65–72,
ARD.2003.011742. https://doi.org/10.1016/J.CHEMPHYSLIP.2013.07.010.
[21] Recommendations | Osteoarthritis: care and management | Guidance | NICE, [57] Y. Yuan, S. ming Li, F. kui Mo, D. fang Zhong, Int J. Pharm. 321 (2006) 117–123,
(2014). https://doi.org/10.1016/J.IJPHARM.2006.06.021.
[22] C. Cooper, R. Chapurlat, N. Al-Daghri, G. Herrero-Beaumont, O. Bruyère, [58] S.H. Auda, D. Fathalla, G. Fetih, M. El-Badry, F. Shakeel, Polym. Bull. 73 (5)
F. Rannou, R. Roth, D. Uebelhart, J.Y. Reginster, Drugs Aging 36 (2019) 15–24, (2015) 1229–1245, https://doi.org/10.1007/S00289-015-1544-8.
https://doi.org/10.1007/S40266-019-00660-1/TABLES/3.

12
V.G.S. Jyothi et al. Colloids and Surfaces B: Biointerfaces 228 (2023) 113399

[59] V. Palanisamy, P. Sanphui, V.G.S.S. Jyothi, N.R. Shastri, G. Bolla, K. Palanisamy, [82] N.K. Garg, R.K. Tyagi, B. Singh, G. Sharma, P. Nirbhavane, V. Kushwah, S. Jain,
M. Prakash, V.R. Vangala, Cryst. Growth Des. 21 (2021) 1548–1561, https://doi. O.P. Katare, Int J. Pharm. 499 (2016) 301–320, https://doi.org/10.1016/J.
org/10.1021/ACS.CGD.0C01344/SUPPL_FILE/CG0C01344_SI_001.PDF. IJPHARM.2015.12.061.
[60] A.O. Abioye, S. Issah, A.T. Kola-Mustapha, Int. J. Pharm. 490 (2015) 112–130, [83] C. Mircioiu, V. Voicu, V. Anuta, A. Tudose, C. Celia, D. Paolino, M. Fresta,
https://doi.org/10.1016/J.IJPHARM.2015.05.030. R. Sandulovici, I. Mircioiu, Pharmaceutics 11 (2019), https://doi.org/10.3390/
[61] A. Ziaei, S. Sahranavard, M.J. Gharagozlou, M. Faizi, Daru J. Pharm. Sci. 24 PHARMACEUTICS11030140.
(2016), https://doi.org/10.1186/S40199-016-0152-Y. [84] B. Das, A.K. Nayak, U. Nanda, Int. J. Biol. Macromol. 62 (2013) 514–517, https://
[62] M.V.K. Patil, A.D. Kandhare, S.D. Bhise, Biomed. Aging Pathol. 2 (2012) 6–15, doi.org/10.1016/J.IJBIOMAC.2013.09.049.
https://doi.org/10.1016/J.BIOMAG.2012.01.002. [85] L.H. Peng, W. Wei, Y.H. Shan, Y.S. Chong, L. Yu, J.Q. Gao, Drug Dev. Ind. Pharm.
[63] V.G.S.S. Jyothi, J. Pawar, V. Fernandes, R. Kumar, C. Singh, S.B. Singh, J. Madan, 43 (2017) 55–66, https://doi.org/10.1080/03639045.2016.1220563.
D.K. Khatri, J. Drug Deliv. Sci. Technol. 70 (2022), 103195, https://doi.org/ [86] Y. Inokuchi, K. Hironaka, T. Fujisawa, Y. Tozuka, K. Tsuruma, M. Shimazawa,
10.1016/J.JDDST.2022.103195. H. Takeuchi, H. Hara, Invest Ophthalmol. Vis. Sci. 51 (2010) 3162–3170, https://
[64] M. Udo, T. Muneta, K. Tsuji, N. Ozeki, Y. Nakagawa, T. Ohara, R. Saito, doi.org/10.1167/IOVS.09-4697.
K. Yanagisawa, H. Koga, I. Sekiya, Osteoarthr. Cartil. 24 (2016) 1284–1291, [87] R.E. Guzman, M.G. Evans, S. Bove, B. Morenko, K. Kilgore, Toxicol. Pathol. 31
https://doi.org/10.1016/J.JOCA.2016.02.005. (2003) 619–624, https://doi.org/10.1080/01926230390241800.
[65] K.P.H. Pritzker, S. Gay, S.A. Jimenez, K. Ostergaard, J.P. Pelletier, K. Revell, [88] E.L. Kuyinu, G. Narayanan, L.S. Nair, C.T. Laurencin, J. Orthop. Surg. Res. 11
D. Salter, W.B. van den Berg, Osteoarthr. Cartil. 14 (2006) 13–29, https://doi. (2016) 1–27, https://doi.org/10.1186/S13018-016-0346-5.
org/10.1016/J.JOCA.2005.07.014. [89] C. Otis, J. Gervais, M. Guillot, J.A. Gervais, D. Gauvin, C. Péthel, S. Authier, M.
[66] J. Steinmeyer, F. Bock, J. Stöve, J. Jerosch, J. Flechtenmacher, Orthop. Rev. A. Dansereau, P. Sarret, J. Martel-Pelletier, J.P. Pelletier, F. Beaudry, E. Troncy,
(Pavia) 10 (2018), https://doi.org/10.4081/OR.2018.7782. Arthritis Res. Ther. 18 (2016), https://doi.org/10.1186/S13075-016-1047-5.
[67] A. Sioufi, F. Pommier, F. Boschet, J. Godbillon, D. Lavoignat, D. Salliere, [90] P. Anchi, V. Swamy, C. Godugu, Life Sci. 266 (2021), https://doi.org/10.1016/J.
Biopharm. Drug Dispos. 15 (1994) 441–449, https://doi.org/10.1002/ LFS.2020.118911.
BDD.2510150602. [91] M.B. Goldring, M. Otero, Curr. Opin. Rheuma 23 (2011) 471–478, https://doi.
[68] S.A. Yousef, Y.H. Mohammed, S. Namjoshi, J.E. Grice, H.A.E. Benson, W. Sakran, org/10.1097/BOR.0B013E328349C2B1.
M.S. Roberts, Pharmaceutics 11 (2019) 639, https://doi.org/10.3390/ [92] J. Bondeson, A.B. Blom, S. Wainwright, C. Hughes, B. Caterson, W.B. Van Den
PHARMACEUTICS11120639. Berg, Arthritis Rheum. 62 (2010) 647–657, https://doi.org/10.1002/ART.27290.
[69] Z. Jun, Z. Jun-shou, J. -China Pharm. Univ. 34 (5) (2003) 423–425. [93] A. Mathiessen, P.G. Conaghan, Arthritis Res. Ther. 19 (2017) 1–9, https://doi.
[70] C.L. Froebe, F.A. Simion, L.D. Rhein, R.H. Cagan, A. Kligman, Dermatology 181 org/10.1186/S13075-017-1229-9/FIGURES/2.
(1990) 277–283, https://doi.org/10.1159/000247822. [94] J. Bondeson, Curr. Drug Targets 11 (2010) 576–585, https://doi.org/10.2174/
[71] A. Seweryn, Adv. Colloid Interface Sci. 256 (2018) 242–255, https://doi.org/ 138945010791011965.
10.1016/J.CIS.2018.04.002. [95] D. Stefik, V. Vranic, N. Ivkovic, D. Abazovic, D. Maric, D. Vojvodic, G. Supic,
[72] E. Lémery, S. Briançon, Y. Chevalier, C. Bordes, T. Oddos, A. Gohier, M. Bosn. J. Basic Med Sci. 21 (2021) 155–162, https://doi.org/10.17305/
A. Bolzinger, Colloids Surf. A Physicochem Eng. Asp. 469 (2015) 166–179, BJBMS.2020.4735.
https://doi.org/10.1016/J.COLSURFA.2015.01.019. [96] K. Asano, M. Sakai, T. Matsuda, H. Tanaka, K. Fujii, T. Hisamitsu, J. Pharm.
[73] P. Sinha, N. Srivastava, V.K. Rai, R. Mishra, P.V. Ajayakumar, N.P. Yadav, J. Drug Pharm. 58 (2006) 359–366, https://doi.org/10.1211/JPP.58.3.0010.
Deliv. Sci. Technol. 52 (2019) 870–884, https://doi.org/10.1016/J. [97] T. Piamphongsant, Dermatol. Surg. 32 (2006) 611–617, https://doi.org/
JDDST.2019.06.007. 10.1111/J.1524-4725.2006.32131.X.
[74] V.K. Rai, N. Mishra, K.S. Yadav, N.P. Yadav, J. Control. Release 270 (2018) [98] G. Schulze-Tanzil, H. Zreiqat, R. Sabat, B. Kohl, A. Halder, R. Muller, T. John,
203–225, https://doi.org/10.1016/J.JCONREL.2017.11.049. Curr. Gene Ther. 9 (2009) 306–315, https://doi.org/10.2174/
[75] M. Danaei, M. Dehghankhold, S. Ataei, F. Hasanzadeh Davarani, R. Javanmard, 156652309788921044.
A. Dokhani, S. Khorasani, M.R. Mozafari, Pharmaceutics 10 (2018), https://doi. [99] F. Iannone, C. De Bari, F. Dell Accio, M. Covelli, F.P. Cantatore, V. Patella, G.
org/10.3390/PHARMACEUTICS10020057. L. Bianco, G. Lapadula, Clin. Exp. Rheuma 19 (2001) 139–145.
[76] C.J. Chirayil, J. Abraham, R.K. Mishra, S.C. George, S. Thomas, Elsevier 3 (2017) [100] E. Nagy, E. Vajda, C. Vari, Ś. Sipka, A.M. Frr, E. Horvth, PeerJ 5 (2017), https://
1–36. https://doi.org/10.1016/B978–0-323–46139-9.00001–3. doi.org/10.7717/PEERJ.3185.
[77] N. Seetapan, P. Bejrapha, W. Srinuanchai, U.R. Ruktanonchai, Micron 41 (2010) [101] B. Liu, C. Ji, Y. Shao, T. Liang, J. He, H. Jiang, G. Chen, Z. Luo, Biomed.
51–58, https://doi.org/10.1016/J.MICRON.2009.08.003. Pharmacother. 127 (2020), 110144, https://doi.org/10.1016/J.
[78] K. Roger, B. Cabane, Angew. Chem. Int. Ed. Engl. 51 (2012) 5625. BIOPHA.2020.110144.
[79] A. Lewińska, M. Domżał-Kędzia, A. Jaromin, M. Łukaszewicz, Pharmaceutics [102] B. Gençosmanoğlu, M. Eryavuz, S. Dervışoğlu, Research in experimental
2020 12 (2020) 953, https://doi.org/10.3390/PHARMACEUTICS12100953. medicine, Zeitschrift fur die gesamte experimentelle Medizin einschliesslich
[80] H. Yan, C. Bao, X. Chen, C. Yu, D. Kong, J. Shi, Q. Lin, RSC Adv. 9 (2019) experimenteller Chirurgie 200 (3) (2001) 215–226.
11649–11658, https://doi.org/10.1039/C9RA00591A. [103] M. Hagen, M. Baker, Curr. Med Res Opin. 33 (2017) 1623–1634, https://doi.org/
[81] C.H. Lee, V. Moturi, Y. Lee, J. Control. Release 136 (2009) 88–98, https://doi. 10.1080/03007995.2017.1352497/SUPPL_FILE/ICMO_A_1352497_SM5675.
org/10.1016/J.JCONREL.2009.02.013. DOCX.

13

You might also like